Next Article in Journal
Abdominal Aortic Aneurysm Diameter versus Volume: A Systematic Review
Next Article in Special Issue
Pharmacokinetics of Biopharmaceuticals: Their Critical Role in Molecular Design
Previous Article in Journal
Natural Course of COVID-19 and Independent Predictors of Mortality
Previous Article in Special Issue
Gene Polymorphism of Biotransformation Enzymes and Ciprofloxacin Pharmacokinetics in Pediatric Patients with Cystic Fibrosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Developmental Pharmacokinetics of Antibiotics Used in Neonatal ICU: Focus on Preterm Infants

by
Olga I. Butranova
1,*,
Elena A. Ushkalova
1,
Sergey K. Zyryanov
1,2 and
Mikhail S. Chenkurov
1
1
Department of General and Clinical Pharmacology, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklaya St. 6, 117198 Moscow, Russia
2
State Budgetary Institution of Healthcare of the City of Moscow “City Clinical Hospital No. 24 of the Moscow City Health Department”, Pistzovaya Srt. 10, 127015 Moscow, Russia
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(3), 940; https://doi.org/10.3390/biomedicines11030940
Submission received: 14 February 2023 / Revised: 6 March 2023 / Accepted: 7 March 2023 / Published: 17 March 2023

Abstract

:
Neonatal Infections are among the most common reasons for admission to the intensive care unit. Neonatal sepsis (NS) significantly contributes to mortality rates. Empiric antibiotic therapy of NS recommended by current international guidelines includes benzylpenicillin, ampicillin/amoxicillin, and aminoglycosides (gentamicin). The rise of antibacterial resistance precipitates the growth of the use of antibiotics of the Watch (second, third, and fourth generations of cephalosporines, carbapenems, macrolides, glycopeptides, rifamycins, fluoroquinolones) and Reserve groups (fifth generation of cephalosporines, oxazolidinones, lipoglycopeptides, fosfomycin), which are associated with a less clinical experience and higher risks of toxic reactions. A proper dosing regimen is essential for effective and safe antibiotic therapy, but its choice in neonates is complicated with high variability in the maturation of organ systems affecting drug absorption, distribution, metabolism, and excretion. Changes in antibiotic pharmacokinetic parameters result in altered efficacy and safety. Population pharmacokinetics can help to prognosis outcomes of antibiotic therapy, but it should be considered that the neonatal population is heterogeneous, and this heterogeneity is mainly determined by gestational and postnatal age. Preterm neonates are common in clinical practice, and due to the different physiology compared to the full terms, constitute a specific neonatal subpopulation. The objective of this review is to summarize the evidence about the developmental changes (specific for preterm and full-term infants, separately) of pharmacokinetic parameters of antibiotics used in neonatal intensive care units.

1. Introduction

Bacterial infections are an important cause of mortality and morbidity in newborns especially in low and middle-income countries [1,2] hence antibiotics are among the most used drugs in this population. About a quarter of the top 100 drugs used in the neonatal intensive care unit (NICU) are antibiotics [3,4]. The most common reason for antibiotic therapy in NICU is neonatal sepsis (NS) [5]. In the cross-sectional study of late-preterm and full-term neonates (n—757,979, 11 countries of Europe, North America, and Australia) it was shown that the antibiotic exposure was 135 days per 1000 live births. The incidence of early-onset sepsis (EOS) was 0.49 cases per 1000 live births (range, 0.18–1.45 cases per 1000 live births), and the EOS-associated mortality rate was 3.20% [6]. A systematic review and meta-analysis of 26 publications (2,797,879 live births and 29,608 sepsis cases in 14 countries) revealed NS incidence equal to 2824 (95% credit interval, CI 1892 to 4194) cases per 100,000 live births with mortality of 17.6% (95% CI 10.3% to 28.6%) [7]. Estimation of global sepsis incidence using data from the Global Burden of Disease Study (2017) revealed a peak in early childhood with 2.9 million (95% uncertainty interval, UI 2.6–3.2) sepsis-related deaths among children less than 5 years old [8]. A systematic review and meta-analysis from data of 30 years (period from 1990 to 2019, 29 studies, 164,750 neonates with sepsis) highlighted the etiology of NS. Main causative agents included coagulase-negative staphylococci (CoNS)—33% (95% CI 24–43), Escherichia coli—17% (95% CI 13–20), and Klebsiella spp.—14% (95% CI 11–17) [9]. The etiology of NS in developing regions was studied in the systematic review and meta-analysis (included 152,217 infants): the main pathogens were Klebsiella spp. (26.36%), Staphylococcus aureus (23.22%), CoNS (23.22%), and Escherichia coli (15.30%) [10]. Studies, based on data from the Burden of Antibiotic Resistance in Neonates from Developing Societies (BARNARDS) network also proved the leading role of Klebsiella spp., especially Klebsiella pneumoniae in NS [11,12,13]. In very preterm infants with EOS, in the framework of the BARNERDS network, the main pathogen was E. coli [14].
Empirical antibiotics recommended for the management of NS by different guidelines include benzylpenicillin, ampicillin or amoxicillin, and aminoglycosides (mainly gentamicin) [15,16,17]. Pediatric survey data from 56 countries support that gentamicin and ampicillin are the most prescribed antibiotics in hospitalized neonates in Africa, the Americas, the Eastern Mediterranean, Europe, and South-East Asia, while in the Western Pacific region most common are amoxicillin in combination with β-lactamase inhibitor, ceftizoxime, and meropenem [18]. The global survey of antimicrobial use in NICUs revealed the leading prescriptions: ampicillin (40%), gentamicin (35%), amikacin (19%), vancomycin (15%), and meropenem (9%) for “rule-out” sepsis and “culture-negative” sepsis. Vancomycin (26%), amikacin (20%), and meropenem (16%) were the most prescribed agents in the definitive treatment of presumed/confirmed infection [5]. Depending on bacterial susceptibility profile and involvement of different organs and systems treatment of NS may also include linezolid, dalbavancin, ceftaroline fosamil, and ceftolozane tazobactam [19].
The rise of antibiotic resistance mediates the involvement of additional antibiotics in NS management. The BARNARDS observational cohort study revealed 390 Gram-negative isolates of which 379 (97.2%) were resistant to ampicillin and 274 (70.3%)—to gentamicin [12]. Among causative agents of EOS in very preterm infants 60.6% of E. coli isolates and 42.9% of Klebsiella spp. were multidrug resistant [14]. Increased rates of antibiotic resistance lead to the use of antibiotics by Watch and Reserve groups (World Health Organization classification), which may result in decreased safety profile. The study of antibiotics prescription data in 5267 neonates from 56 countries defined that the use of Watch antibiotics reached 64.5% in China, and maximum rates of Reserve antibiotics use were seen in Russia (13.9%) and Mexico (12.8%) [18]. There was an attempt in the review by Darlow CA et al. (2021) to propose additional five antibiotics to be used in novel empiric regimens for NS [20]. Among these antibiotics, only one belongs to the Access group—amikacin, while four—to the Watch group (tobramycin, flomoxef, and cefepime) and one to the Reserve group (fosfomycin) [21].
A high rate of mortality in NS is associated with a relatively high level of treatment failure: in the review of 90 randomized controlled trials (RCT), it was reported in 15 studies [22]. The current list of antibiotics used in NICU is broad and reasons for NS treatment failure may be drug-related, with administration and/or dosing errors [23,24,25,26], which can be attributed to poor physicians’ awareness about antibiotics pharmacokinetics and, thus, pharmacodynamics in full-term and especially premature newborns. Extrapolating adult antibiotic regimens to the neonatal population may result in increased toxicity or altered efficacy of treatment due to different degrees of organ and system maturation. The problem is compounded by the high heterogeneity of the neonatal population and the dependence of pharmacokinetic properties on postnatal age, gestational age, and postconceptional age. There is a lack of RCTs including neonates making the choice of a proper antibiotic regimen challenging, especially for preterm infants. Optimal antibiotic treatment of NS is proposed to be based on the next key components: correct treatment phase, correct antibiotic dose, proper antibiotic exposure, and microbiological response determined by minimum inhibitory concentration (MIC) of antibiotic and by the type of drug action, time- or concentration-dependent [27]. Achievement of a proper value of MIC, and/or needed time of exposure to antibiotics is based on the pharmacokinetic (PK) parameters of a drug which are highly variable in the neonatal population.
This review highlights the developmental changes in preterm and full-term neonates and their impact on PK parameters of antibiotics of various groups used in NICU practice.

2. General Considerations on Neonate’s Pharmacokinetics

Different rates of maturation of organs and systems in neonates, the different capacities of enzymatic systems together with pathological changes due to critical illness, inflammatory status, and therapeutic interventions underlie variability of PK parameters and, thus, variability of clinical response in the treatment of infectious diseases. Pharmacokinetics is directly connected with pharmacodynamics through the parameter of plasma drug concentration. Considering antibiotics, the value of plasma concentration is especially important and the next pharmacokinetic/pharmacodynamic (PK/PD) indices are used to describe antibiotic efficacy: ratio of the area under the concentration-time curve (AUC) from zero to 24 h (AUC0–24) to the minimum inhibitory concentration (MIC), the ratio of the maximum plasma concentration (Cmax) to the MIC and % of the time during which the free plasma concentration exceeds the MIC (% fT > MIC). Antibiotic concentration is also an important factor in the prevention of antibacterial resistance (Figure 1). Next parameters are considered from this point: the mutant prevention concentration (MPC), defined as the MIC of the least-susceptible, single-step mutant [28], the ratio of minimum concentration to MIC (Cmin/MIC), the ratio of Cmax to MIC (Cmax/MIC), the ratio of AUC0–24 to MPC (AUC0–24/MPC) or to MIC (AUC0–24/MIC). In the systematic review by Sumi CD et al. (2019) values of the listed above ratios suppressing the emergence of antibiotic resistance were established for different groups of antibacterials. For β-lactam antibiotics, Cmin/MIC should be equal or exceed 4; for aminoglycosides, Cmax/MIC should be equal or exceed 20; for fluoroquinolones, AUC0–24/MPC should be ≥35; for tetracyclines, AUC0–24/MIC should be ≥50; for polymyxin, B AUC0–24/MIC should be ≥808; for fosfomycin, AUC0–24/MIC should be ≥3136 [29]. The value of plasma antibiotic concentration changes through the main pharmacokinetic phases: absorption, distribution, metabolism, and excretion. Figure 1 represents PK/PD targets for optimizing efficacy and minimizing antibacterial resistance for time-dependent, concentration-dependent, and concentration-dependent with time-dependent antibiotics.

2.1. Absorption and Bioavailability

Drug absorption is the process of drug transportation from the site of administration to the systemic circulation and the fraction of unmetabolized drug that reaches the systemic blood flow is bioavailability. There are two main measures of the extent and rate of drug absorption—AUC and Cmax. A systematic review and meta-analysis of 31 studies including neonates revealed decreased bioavailability of oral antibiotics with the delayed achievement of Cmax [30].
Absorption and bioavailability both depend on drug-related factors (formulation, rate of solubility, physicochemical properties, etc.) and patient-related factors. Patient-dependent factors affecting absorption and thus bioavailability in the oral route of drug administration include the value of pH in the stomach and intestine, the rate of motility of the gastrointestinal tract (GIT), GIT maturation rate, gastric emptying time, expression of transport proteins in GIT, the quality and quantity of gastric juices, the rate of bile production, pancreatic function, first-pass metabolism, gastric volume, and mucin production. The main sites of absorption of orally administered drugs in neonates include the stomach, small intestine, and colon. Absorption in the stomach is governed first by gastric pH and gastric emptying time. It is considered, that in neonates (both term and preterm) the colon can play a significant role in the absorption of some drugs and nutrients, which is not true for adults [31].
Neonates are characterized by different degrees of the structural and functional maturation of the GIT depending on the term or preterm birth mainly. The anatomical differentiation of the intestine happens within 20 weeks of gestation, while functional maturation demands more time and appears in general after 32–34 weeks of gestation [32]. Preterm newborns are characterized by a reduced intestinal surface area due to limited villi maturation (full maturation of villi is observed around 20 weeks of gestation), which may lead to impaired absorption of orally administered drugs compared to full-term neonates [33].
Gastric acid secretion emerges in the second trimester of pregnancy and is less in preterm babies compared with full-term; from the moment of birth in two months there is a doubling of the value of secretion [33]. Pepsinogen secretion starts after 17–18 weeks of gestation, and enterokinase—after 24 weeks (25% of the level of older infants). Lactase activity is decreased in preterm newborns, while activities of sucrase, maltase, and isomaltase have no differences with full-terms. Pancreatic lipase is decreased in preterm neonates, as well as bile production and bile ileal reabsorption. Concentrations of bile acids and bile salts in the intestinal lumen at birth are relatively low, and insufficient functioning of transporter-mediated uptake and enterohepatic bile circulation is observed, though passive bile reuptake in the intestine and active transport in the distal ileum are presented [31].
An important factor in drug solubility and thus, absorption, is gastric volume. According to the Biopharmaceutics classification system (BCS), drugs can change their solubility class at different ages with the change in gastric volume. For example, antibacterials including chloramphenicol, erythromycin, and cefalexin alter from a low solubility classification in 6-month-old children to high solubility in adults [34]. Gastric motility is low in preterm newborns of 28–32 gestational weeks, while after 32 weeks, the value is close to full-terms. Gastric emptying is considered to be slower in infants than in older children and adults, with discussible factors affecting certain values [35]. A meta-analysis including 49 published studies of 1457 individuals proved the absence of effects of postnatal age or gestational age on the mean gastric emptying time. A significant effect of meal type was established. The fastest emptying time (mean simulated gastric residence time of 45 min) was determined for aqueous solutions, and the slowest (98 min) for solid food [36]. Published data state that the time of intestinal transit in preterm newborns is four times longer than in adults [37].
The value of gastric pH in neonates, both full-term and preterm, is still a discussible question. Some works stated a nearly neutral pH at birth (6–8) with further change to acidic values [38]. In a study of 40 preterm infants (24–33 weeks of gestation) median gastric pH was found to be between 4.5 and 5.5 [39]. A study of 29 preterm neonates ≤28 weeks of gestational age with body weight ≤ 1000 g showed an acidic value of gastric pH on day 1 with nearly no change up to 4 weeks. For neonates ≥26 weeks, mean pH ± standard deviation on the first day was 5.25 ± 3.00, and on the 28th day, it was 4.56 ± 1.34. For neonates <26 weeks, the values were 5.33 ± 2.17 and 4.87 ± 1.06, correspondingly [40]. Research conducted in the earlier period with a smaller number of neonates revealed variable results with mean pH ranging from 1.3 up to 6.9 [41,42]. Postprandial pH in the infant stomach was shown to be on a level above 4.5 for about two hours [35]. In the intestinal lumen of infants, the range of pH was reported to vary between 5.8 and 7.0, based on limited data [31].
The rate of expression of transport proteins provides a great impact on drug absorption since it is generally low in neonates. There was proved a strong relationship between the age of neonate (mainly postmenstrual age) and expression of such transport proteins, as breast cancer resistant protein (BCRP), bile salt efflux pump (BSEP), glucose transporter 1 (GLUT1), P-glycoprotein (P-gp), multidrug-resistance like protein 1 (MRP1), 2 (MRP2), and 3 (MRP3), Na+-taurocholate cotransporting polypeptide (NTCP), organic anion transporter polypeptides 1B1 (OATP1B1), and organic cation transporter 1 (OCT1) [43]. In preterm newborns, it is important to consider a high probability of insufficient levels of some transport proteins administering drugs orally. In the intestine, efflux transporters limiting drug absorption are P-gp, MRP2, and BCRP. Drug intestinal absorption is mainly performed with OATP1A2, OATP2B1, and peptide transporter 1 (PEPT1) [44].
Characteristics of the main transport proteins affecting drug absorption in the intestine are shown in Table 1.
The oral route of drug administration is preferred in children including neonates whenever it is possible since this route is non-invasive and painless. The summary of neonatal physiological factors affecting the absorption and bioavailability of orally administered drugs is given in Table 2.
PK parameters of orally administered antibiotics in neonates were studied mainly for beta-lactams, and for many of them, plasma concentration values high enough to exceed MIC were demonstrated.

2.1.1. Absorption of Oral Penicillins

Ampicillin is the most prescribed antibiotic used in NICU [4,5]. Considering other penicillins in NICU, in the work by Hsieh EM et al. (2014) five drugs were reported (amoxicillin, oxacillin, nafcillin, penicillin G, and piperacillin tazobactam), in the work by Prusakov P et al. (2019)—nine (amoxicillin, and amoxicillin clavulanate, ampicillin sulbactam, penicillin G, cloxacillin, oxacillin, nafcillin, piperacillin, and piperacillin tazobactam) [4,5].
From the middle of XX century PK parameters of many penicillins were studied in neonates, and the results revealed differences between oral and parenteral routs of administration and age-dependent changes [30]. Oral penicillin was characterized by a lower Cmax value compared with penicillin administered intramuscularly [68]. Studies revealed a better absorption of oral flucloxacillin in infants 0–1 month old compared with older children [69], especially for such formulations as mixtures [70], plasma concentrations after both intravenous (i.v.) and oral administration were well above the MIC-values generally reported for Staphylococcus aureus [71]. A review of the works dedicated to oral flucloxacillin in neonates revealed a Tmax of 2 h in a majority of studies [30]. Studies of oral ampicillin and amoxicillin in neonates revealed a delay of Tmax up to 4 h compared with 30 min in intramuscular administration with bioavailability values close to adults, Cmax in preterm neonates was the highest compared with full-terms and older children [30,72].
Amoxicillin clavulanate is one of the most prescribed oral antibiotics for both adult and pediatric populations used in case of proved or strictly suspected beta-lactamase production. Evaluation of PK parameters of oral clavulanic acid in term neonates (n—15, dose of amoxicillin clavulanate 25/6.25 mg/kg, thrice a day administration) resulted in great variance in plasma concentrations (median: 1.4 mg/L; range: 0.20–4.82 mg/L) and extrapolation led to AUC of at least 8.4 mg·h/L, which is comparable to those of adults [73]. The conclusion of high variability of PK parameters of clavulanic acid in the pediatric population is supported by a systemic literature review of 18 studies, which did also state that Cmax, for oral administration, was below that of the intravenous route [73], which is like it is in adults [74].
PK studies of penicillins in neonates with sepsis are limited. The results of the PK study of oral amoxicillin used together with parenteral gentamicin in 60 0–2 month infants with sepsis (final analysis included blood samples from 44 infants) revealed that amoxicillin concentrations exceed the susceptibility breakpoint for amoxicillin (2.0 mg/L) against resistant S. pneumoniae strains for >50% of a 12-h dosing interval [75].

2.1.2. Absorption of Oral Cephalosporines

In the list of most used drugs in neonatal ICU (for the period 2005–2010 years) among cephalosporins there are seven positions, and oral formulation is available only for the one—for cephalexin [4]. In the global point-prevalence survey of antimicrobial use in neonatal ICUs, there were eight cephalosporins with two available in oral form—cefadroxil and cephalexin [5].
The pharmacokinetics of oral cephalosporins are described in several studies including pediatric population. In children with osteoarticular infection, the achievement of optimal plasma exposure was demonstrated for oral cephalexin (median dose 40 mg/kg/dose every 8 h) [76]. Data on PK of oral cephalexin in newborns revealed that the serum levels achieved after a dosage of 15 mg/kg 8 hourly were lower than the average MIC for most of the Gram-negative organisms causing infections in neonates, while the levels achieved after 50 mg/kg 12-hourly were close to those in adults receiving 1 g. Tmax was at about 2 h, demonstrating slower absorption than in adults [77]. PK studies performed in 70 s of XX century with cefadroxil and cephradine included infants and children with no inclusion of neonates. The results reported a decrease in Cmax in the fed state, which can be true also for the neonatal population [78,79]. PK of cefaclor was studied in 10 newborns, a mean peak serum concentration of 7.7 μg/mL was achieved at 1 h after an oral dose of 7.5 mg/kg [80]. For plasma concentrations of cefaclor, the same effects of fed and fasting states were detected as those for cefadroxil and cephradine [81]. On the opposite side, for cefprozil, there were no food effects on PK [82]. PK of oral cefuroxime axetil was studied in the pediatric population (minimum age of participants—3 months). This study reported achievement of concentrations exceeding the MICs for common respiratory tract pathogens, including beta-lactamase-producing strains of Haemophilus influenzae and Moraxella catarrhalis. Administration of 10 or 15 mg/kg doses resulted in serum cefuroxime concentrations similar to adult values following a 250 mg cefuroxime axetil tablet [83]. For oral cefixime pediatric data, supposed GIT absorption from 40 to 50% is more rapid and complete for oral suspension [84]. Oral ceftibuten (single oral dose of either 4.5 or 9.0 mg/kg) PK parameters were studied in infants and children. Rapid absorption was shown with mean Tmax—140 min and Cmax from 5.0 to 19.0 mg/L with no differences between dosing regimens [85]. Pediatric PK studies of cefpodoxime revealed Tmax prolongation in the fed state (fed = 2.79 ± 1.10 h vs. fasted = 1.93 ± 0.54 h), but the extent of absorption was not affected by food [86]. Though this result was derived from the pediatric population, it can be also considered actual for neonates.

2.1.3. Absorption of Oral Carbapenems

The first carbapenem with the oral route of administration approved for pediatric practice in Japan is tebipenem. Though there are no data on neonates, it is interesting to consider PK parameters derived from pediatric studies (n—217, minimum age of participant—6 months, dosing regimen—twice a day at 4 mg/kg or 6 mg/kg for 8 days). Tmax values were 0.74 ± 0.26 and 0.69 ± 0.22 h (average + standard deviation) in 4 mg/kg and 6 mg/kg, Cmax values were 3.48 ± 1.67 and 5.20 ± 2.84 mg/mL, and the AUC0–24 h values were 11.00 ± 1.84 and 16.07 ± 3.35 mg·h/mL in the 4 and 6 mg/kg dosage groups, respectively [87]. Tebipenem pivoxil is characterized by remarkably high absorption in GIT due to its ability to be transported by OATP1A2 and OATP2B1 along with simple diffusion [63]. Interestingly to note, tebipenem pivoxil is the first beta-lactam demonstrated OATP-mediated transport in the process of intestinal absorption. Since there are some studies stating an increased intestinal OATP2B1 expression in neonates [55], there is a potential for variability in intestinal tebipenem pivoxil absorption in this population.

2.1.4. Absorption of Oral Macrolides

Macrolides are antibiotics used mainly in ambulatory practice, but in the list of 100 drugs most used in NICU erythromycin was ranked 24th being the 6th among most used antibiotics and the only macrolide [4]. In the global survey by Prusakov P et al. (2021), there were three macrolides used in neonatal ICU, erythromycin, azithromycin, and clarithromycin [5]. Erythromycin was the first macrolide studied in children. Comparison of absorption of erythromycin suspension in three age groups (0–1 month, 1–6 months, and 6 months to 6 years) revealed the lowest value for infants less than 1 month of age [88]. The value of absolute bioavailability of rectally administered erythromycin was 28% in neonates, 36% in infants, and 54% in children greater than 1 year [89].
PK parameters of orally administered azithromycin were studied in children after a single oral dose of 10 mg per kg of body weight on day 1 followed by single daily doses of 5 mg/kg on days 2 to 5. Mean values of PK parameters (±standard deviation) were estimated for Cmax, Tmax, and AUC0–24: 383 + 142 ng/mL, 2.4 + 1.1 h, and 3109 + 1033 ng × h/mL, respectively. Concentrations in serum at 0 h (predose) and at 24, 48, and 72 h after the final dose were 67 + 31, 64 + 24, 41 + 17, and 29 + 14 ng/mL, respectively [90]. In the study of single (n—14) and multiple oral doses (n—9) of 12 mg/kg in 23 children, Cmax and Tmax of azithromycin were 318.2 + 174.5 μg/L, and 2.4 + 1.1 h, respectively, with no differences estimated between these doses groups [91]. Comparison between 30 mg/kg immediate-release (IR) and 60 mg/kg extended-release (ER) forms of azithromycin oral suspension in children revealed similar or greater systemic exposure in the case of ER form [92].
PK studies including clarithromycin revealed high rates of oral absorption both in children and adults not affected by food. In infants and children (6 months to 10 years), a brief delay in the onset of absorption was demonstrated for clarithromycin (suspension, 7.5 mg/kg). The mean Cmax for clarithromycin was reached within about 3 h both under fasting and fed conditions with corresponding values of 3.59 and 4.58 micrograms/mL in a single-dose regimen. Cmax values for 14-(R)-hydroxylated metabolite for fasting and fed conditions were 1.19 and 1.26 micrograms/mL, respectively [93]. Other studies reported a similar profiles of clarithromycin pharmacokinetics in the pediatric population [94].

2.1.5. Absorption of Oral Oxazolidinones

Linezolid is used in neonates including preterm ones. Its bioavailability is extremely high in the oral route, reaching 100% with minimum decrease under the fed condition, Tmax is about 1–2 h [95]. The PK study of oral linezolid suspension, 10 mg/kg used in 4-month-old infants (birth at 25 weeks) revealed that Cmax and AUC were lower than in full-term infants who used linezolid intravenously [96]. Possible factors affecting the PK of linezolid in neonates may include incomplete absorption, faster clearance, or a smaller actual volume of distribution. PK study including extremely premature infants was performed for oral linezolid (dose 10 mg/kg every 8 h) and continuous intravenous infusion (dose 30 mg/kg). Analysis of 7 serum samples for oral linezolid revealed a mean Cmax equal to 9.04 (0.69–32.9) mg/L, and 17 serum samples for intravenous linezolid revealed a mean Cmax equal to 1723 (2.6–30.4) mg/L. Reported Cmax values for both routes of administration were ≥MIC for causative microflora [97].
Another oxazolidinone, tedizolide, is also used in pediatric practice. The study of oral and intravenous tedizolid in infants (age—1 day to 24 months) started in the 2017 year (NCT03217565) and still has a recruitment status [98]. PK parameters of oral tedizolide (dose 3–6 mg/kg) were studied in children (2 to <12 years old), and results revealed high bioavailability value, compared with that of the intravenous route, median Tmax was 2–3 h compared with 1–2 h after initiation of the 1 h intravenous infusion. Cmax values were higher compared with adults (4.19 vs. ~2.5 mg/L) [99]. A study of oral and intravenous tedizolid in adolescents revealed no significant differences in PK parameters and the received results demonstrated similarity with adult PK parameters of tedizolid [100].

2.1.6. Absorption of Oral Fluoroquinolones

Fluoroquinolones are among the antibacterials prescribed for neonates with infections, including ICU practice [5]. Ciprofloxacin is used in the majority of clinical situations intravenously, though in some situations, oral route is available. Population pharmacokinetics of ciprofloxacin including oral form was derived using data from a mixed population, including 3 newborns, 17 infants and toddlers, 27 children, and 8 adolescents and young adults. For i.v. ciprofloxacin, sampling at a single point (12 h after the start of infusion) allowed the precise and accurate estimation of clearance (CL) and the elimination half-life, as well as the ciprofloxacin concentration at the end of the infusion, for oral ciprofloxacin the presence of a lag time after administration suggests a schedule based on two sampling times of 1 and 12 h [101]. Another population PK study was performed with data from 60 newborn infants who used intravenous ciprofloxacin (postmenstrual age [PMA] range, 24.9 to 47.9 weeks). The main covariates affecting ciprofloxacin pharmacokinetics were gestational age, postnatal age, current weight, serum creatinine concentration, and use of inotropes. Monte Carlo simulation demonstrated that 90% of hypothetical newborns with a PMA of <34 weeks treated with 7.5 mg/kg twice daily and 84% of newborns with a PMA ≥34 weeks and young infants receiving 12.5 mg/kg twice daily would reach the AUC/MIC target of 125, using the standard EUCAST MIC susceptibility breakpoint of 0.5 mg/L [102].
Levofloxacin also may be administered orally in pediatric practice. In the study 85 children (6 months to <2 years, 2 to <5 years, 5 to <10 years, 10 to <12 years, and 12 to 16 years) received a single 7 mg/kg dose of levofloxacin (intravenously or orally) absorption demonstrated no age-dependence and was close to adult values [103].
The rate of oral absorption of other fluoroquinolones, moxifloxacin, and gatifloxacin, were not studied in infants and only extrapolation of the results of PK studies made for an older category of patients is possible. For oral moxifloxacin data are available for the pediatric population suffering from multidrug-resistant tuberculosis (n—23, the median age—11.1 years, 6 out of 23 were human immunodeficiency virus (HIV)-infected). The median Cmax was 3.08 (IQR, 2.85–3.82) µg/mL, AUC from 0–8 h (AUC0–8)—17.24 (interquartile range, IQR, 14.47–21.99) µg × h/mL, Tmax—2.0 (IQR, 1.0–8.0) h. In HIV-infected children, there was a decrease in AUC0–8 and Cmax. Tmax was shorter with crushed vs. whole tablets [104]. PK parameters of oral gatifloxacin were studied for a single dose of suspension (doses—5, 10, or 15 mg/kg of body weight, 600 mg maximum; 76 patients with average age 6.7 ± 5.0 years) and for tablets (dose 10 mg/kg, 2 children >6 years of age). Cmax and AUC increased in a manner approximately proportional to the dose, and at the 10 mg/kg dose, the bioavailability was similar between the suspension and tablet formulation [105].

2.1.7. Absorption of Other Oral Antibacterial Agents

Vancomycin is a glycopeptide administered intravenously to treat neonatal sepsis caused by resistant Gram-positive microflora. Oral vancomycin demonstrated effectiveness in the prophylaxis of necrotizing enterocolitis in preterm, very low birthweight infants, and no serious adverse effects were detected, with negligible serum drug concentrations suggesting the absence of systemic absorption [106]. The same results were revealed for older patients with colitis caused by Clostridium dificile (n—8, age from 2 to 18 years)—serum vancomycin levels were undetectable [107]. Published works reported increased rates of oral absorption of vancomycin in pediatric and adult patients with cancer and associated chemotherapy [108,109].
Trimethoprim sulfamethoxazole (TMPX) being a synthetic antimicrobial agent is found among drugs used in NICU in infants ≥3 days old [5]. Simulation of TMPX exposure (oral route of administration) revealed that for all dosing regimens steady-state area under the concentration-versus-time curve from time zero to τ (AUC0–τ,ss; where τ denotes the dosing interval) in subjects 0 to <2 years was similar with the group of 2 to <6 years of age (within 20%), but 29% less than in older children (6 to <21 years of age). Values of simulated AUC0–τ,ss both for infants and children were generally lower than that for 70-kg adults [110].
Clindamycin is used in neonatal ICU including infants <3 days old [5]. Clindamycin exists in oral form, though due to negative organoleptic properties, it is not a common choice for the pediatric population. Since clindamycin is a highly lipophilic agent, its absorption is thought to be unaffected by age-associated changes specific to the pediatric population including neonates [111].
Fosfomycin (C3H7O4P) is a phosphonic acid derivative representing an epoxide class of antibiotics [112] proposed to treat NS in resistance to first-line antibiotics [113]. Fosomycin is hydrolyzed in the stomach and absorbed in the small intestine; factors affecting its bioavailability include gastric pH and gastric emptying rate [114]. In the PK study of fosfomycin in neonates with suspected sepsis (The NeoFosfo study (NCT03453177)) oral bioavailability was estimated to be 0.48 (dose was 100 mg/kg) [114] that is close to adult values [114].
Rifampin is a macrocyclic antibiotic effective mainly against resistant Staphylococcus aureus and Mycobacterium tuberculosis. Oral forms may contain only rifampicin, or its combination of isoniazid and pyrazinamide. The study included children with tuberculosis (age 3 months to 13 years, dosing for initial treatment, intensive phase—rifampicin 60 mg, isoniazid 30 mg, and pyrazinamide 150 mg; for continuation phase—rifampicin 60 mg and isoniazid 30 mg) the mean AUC0–6 on enrolment was 14.88 and 18.07 μg/hour/mL (p = 0.25) in HIV-infected and HIV-uninfected children, respectively, and after 4 months of treatment 16.52 and 17.94 μg/hour/mL (p = 0.59). The AUC0–6 of all 55 children was 16.81 (+10.82) μg/hour/mL on enrolment and 17.39 (+9.74) μg/hour/mL after 4 months of treatment [115]. Previous PK studies of oral rifampicin in children revealed Cmax three times less than that for the intravenous route [116]. Using PK data from preterm and term infants dosing simulation was performed based on weight and postnatal age, simulated regimens resulted in comparable exposures to adults receiving therapeutic doses of rifampin against staphylococcal infections and TB [117].

2.1.8. Effect of Food on Antibiotics Absorption

An important factor that may alter the rate of absorption of oral drugs is the presence of food in GIT. PK studies described above provide some data in this respect, and Table 3 represents available information on food effects on the absorption of oral antibiotics.

2.1.9. Absorption in Non-Oral Routes of Administration

The data on age-associated change of absorption for non-oral routes of drug administration are given in Table 4.

2.2. Distribution

The process of drug distribution to various organs and systems is essential for the further interaction of drug molecules with their targets, demonstrating a link to pharmacodynamics. Distribution depends both on drug-associated factors and patient-associated factors. Physicochemical properties of drugs, such as lipophilicity and hydrophilicity, can dramatically affect passage through biological membranes since there is a direct relation between permeability and lipophilicity. Hydrophilic antibiotics include beta-lactams, aminoglycosides, glycopeptides, oxazolidinones (tedizolid), and antibacterials of different groups, such as colistimethate sodium, dalbavancin, fosfomycin, telavancin. General features of hydrophilic antibiotics: small size of molecules, relatively low molecular weight, distribution into the extracellular fluid, and low values of the volume of distribution (Vd), reflecting their propensity to remain in the plasma. Lipophilic antibacterials are fluoroquinolones, macrolides and ketolides, metronidazole, rifampicin, trimethoprim/sulfamethoxazole, linezolid (moderate lipophilic). For these agents, high Vd values are specific because of their ability to cross biological membranes with passive diffusion and reach extravascular spaces. Distribution of the same drug may vary depending on health state: in critically ill patients hydrophilic antibiotics (β-lactams, aminoglycosides, glycopeptides) tend to have an increased Vd and a reduced clearance, PK parameters of those, which are lipophilic, are less affected.
Table 5 includes data on the distribution of different groups of antibiotics.
Patient-associated factors affecting Vd include water and fat content in the organism, proportion of extracellular fluids and total water, rate of functioning of the cardiovascular system, the quantity of plasma proteins, structure and function of blood-tissue barriers. Through the first year of life, and organism undergoes significant changes. There is an increase in weight by 50% in the first 6 weeks of life, doubling in the first 3–4 months and tripling at the end of the year. Through the infant period body length increases by 50% and body surface area doubles [123].
Total body water in neonates is higher than in older children and adults. Increased water proportion in the organism of neonate results in increased values of Vd of hydrophilic drugs and thus, leads to the need to use higher doses per kg [124]. Recent systematic review and meta-analysis (22 articles included in the final analysis) revealed that the mean total body water for full-term newborns was 73.8% (95% CI 72.47% to 75.06%, 15 studies, 433 infants). Meta-regression demonstrated that it was higher in preterm infants (up to 90% at 26 weeks gestation, dropping to 75% at 36 weeks corrected gestation) and had a negative correlation with gestation at birth, falling 1.44% per week (95% CI 0.63% to 2.24%, 9 studies, 179 infants) [125]. INTERGROWTH-21st Project was aimed at the estimation of composition patterns of the body of neonates (1019 newborns). The mean body fat percentage (±standard deviation) at birth was 9.6 ± 4.0 for boys and 10.7 ± 4.0 for girls. The fat mass demonstrated a moderate increase with gestational age with very large variability, 36 g/week (95% confidence interval (CI) 28–43 g) for girls and 33 g/week (95% CI 25–42 g) for boys. The mean value of fat-free mass (±standard deviation) was 2965 ± 422 g for boys and 2739 ± 320 g for girls [126]. Investigation of the association between preterm birth and body composition at 6, 18, and 30 years of age suggested that in boys preterm birth was associated with decreased body fat and fat-free mass in early age with subsequent increase in fat mass in adult age [127]. Published data reported a smaller percentage of fat mass in preterm newborns (3% in a 1.5 kg premature neonate) compared with full-terms (12%) [128].
The plasma protein binding (PPB) rate correlates with the amount of pharmacologically active, unbound fraction of a drug, which is responsible for the realization of the main drug action in the organism since it is the unbound fraction, which penetrates the extravascular space. Decreased amount of plasma proteins results in the increase in the unbound fraction and possible increase in toxicity. On the opposite side, an increased binding rate will result in the reduction of free, active drug concentrations, which may lead to the decreased antimicrobial activity of antibacterial agents in the site of infection, which is extravascular typically. It was also shown that the rate of plasma protein binding of antibiotics can affect the extent of their antimicrobial activity [129].
The main plasma proteins binding drugs are human serum albumin (HSA) and α-1-acid glycoprotein (AAG). The mean value of HSA for premature neonates was estimated at the level of 30.6 ± 4.7 g/L in the study including 199 subjects with a mean birth weight of 1272 ± 390 g, mean gestational age of 29.2 ± 2.2 weeks [130], less values were reported in another study: 25 ± 0.6 g/L in the population of 24 premature infants (mean gestational age 28.4 ± 0.4 weeks, mean birth weight 1080 ± 75 g) [131]. Reported values for HSA in neonates are less than in the adult population, resulting in a decrease in plasma protein binding rates. HSA is typically associated with binding acidic exogenous compounds.
AAG is an acute-phase reactant, and its levels are different in healthy subjects and in those with injury, inflammation, or infection. AAG displays a high affinity towards basic lipophilic compounds. In healthy subjects, plasma concentrations of AAG range from ≈50–130 mg/dL [132]. In neonates, AAG levels are less than those in adults. The ontogeny of plasma AAG in healthy subjects (average postnatal age from 0 days to 79 years) was studied using data from 26 separate studies. Maximum plasma AAG concentration (AAGmax) in healthy adults was 93.17 mg/dL and the model estimates median AAG concentrations to be approximately 3.8-fold lower (i.e., 24.67 mg/dL) during the first day of life [133]. The ontogeny of AAG in subjects diagnosed or suspected of infection was studied using 214 individual AAG concentrations (postnatal age range: from 5 days to 20.5 years). For adults with suspected or diagnosed infection, AAGmax was 254.71 mg/dL, and increased AAG concentrations were specific for all ages: at 5 days old, median AAG concentrations were 89.41 mg/dL in infected individuals compared to 41.51 mg/dL in healthy subjects [133]. Factors affecting AAG levels also include gestational age and mode of delivery. AAG levels were found to be higher in infants born vaginally compared with those who were delivered by Caesarean section, and increased gestational age was associated with higher AAG concentrations [134].
An increase or decrease in the PPB rate may produce a significant change in the clinical response while using highly protein-bound drugs (≥90%). In the review by Meesters K et al. (2023), it was summarized that among penicillins relatively high PPB rates were reported for cloxacillin (≈94%), dicloxacillin (96–97%), flucloxacillin (95–96%), and oxacillin (92–96%) [135].
Among antibiotics with high rates of PPB, significant differences were demonstrated for different age groups and for the different health statuses of patients. For oxacillin, cloxacillin, and flucloxacillin an increase in the unbound fraction was demonstrated in hypoproteinemic patients resulting in overdose and risks of toxicity [136,137,138]. Neonatal plasma concentrations of HAS and AAG are less than in adults, resulting in a potential increase in the unbound fraction. In a study of 56 infants aged 3 to 87 days (gestational age, 25–41 weeks) flucloxacillin mean protein binding was 74.5% ± 13.1% with high variability (34.3% to 89.7%) [139].
The results of the systemic review of studies dedicated to PPB of different cephalosporines revealed a high rate of variability of binding patterns considering different fluids and different categories of patients [140]. The highest rates of protein binding in plasma and serum of adults were detected for cefonicid (mean 82–98%, four studies), and cefoperazone (mean 89–92%, four studies). The lowest protein binding rates were demonstrated for cefuroxime (mean 16–33%, five studies), cefotaxime (mean 8–41%, four studies), and ceftazidime (mean 0–21%, six studies) [140]. Estimating concentration ranges authors revealed a linear pattern of protein binding for the next cephalosporines: ceforanide, cefamandole, cefmenoxime, cefotaxime, cefluprenam, cefotetan, and cefoxitin. A typical non-linear pattern was found for cefazolin, cefonicid, cefpiramide, and ceftriaxone [140].
Several studies describe the PPB of cephalosporines in neonates and children. Age-dependent differences were detected in the PK study of cefoperazone. The unbound fraction of cefoperazone in newborns (n—17, mean age 35.7 (30.1–42.3) gestation weeks) was 24.6 (11.3–48.0)%, in infants (n—10, mean age 0.46 (0.1–2.0) years)—16.9 (9.1–27.4)%, in children (n—19, mean age 6.4 (2.2–9.0) years)—11.7 (8.1–18.6)% [141]. In healthy adults, ceftriaxone PPB was 95% [142]. Prediction of unbound ceftriaxone concentration in children resulted in high variability from 4.75% to 39.97% [143]. In critically ill children (0 to 18 years of age) median unbound fraction of ceftriaxone was 13.6 (7.6–70.3)% [144]. In critically ill adult patients unbound fraction of ceftriaxone was estimated at the level of 33.0 (20.2–44.5)% [145].
The highest rate of PPB among carbapenems is for ertapenem. Depending on drug concentration it varies from ~95% at concentrations of <50 μg/mL to ~92% at concentrations of 150 μg/mL (concentration at the end of a 30-min infusion following the 1-g dose) [146]. In critically ill patients, protein binding of ertapenem was also shown to be concentration-dependent and varied from 84% to 96% [147]. The unbound fraction of ertapenem in elderly subjects (~5 to 11%) was generally greater than that in young adults (~5 to 8%) [148].
High rates of PPB are also specific for glycopeptide teicoplanin and lipoglycopeptides. Teicoplanin has plasma protein binding of 90 to 95% in adults [149]. In neonates, this value was demonstrated to be lower—from 80.5% to 71.9% [150]. Though vancomycin has a moderate protein binding rate, PK studies revealed the same tendency: the unbound vancomycin fraction in neonates was higher than in children and adults, and total vancomycin concentration and albumin were the most important covariates of unbound vancomycin concentration [151]. The median fraction unbound of vancomycin was reported to be 0.9 in neonates, and 0.6 in adults [151].
Among lipoglycopeptides, the highest rate of PPB was reported for dalbavancin, 99% [152], for telavancin >90% [153], and for oritavancin ≈ 85% [154]. Dalbavancin clearance change in critically ill patients may propose the effect of hypoalbuminemia on PK parameters [155].
Some macrolides are characterized by relatively high PPB: for erythromycin, it varies from 80 to 93% [156], and for clarithromycin it is about 70% [157]. Azithromycin PPB is about 30% in adults, and the percent binding in newborns, especially preterm, is discussable [158], and for PK simulation in neonates, a PPB of 30% is used [159].
Among oxazolidinones, linezolid has low PPB: its unbound fraction was 85.4 ± 3.7% in plasma samples from adult surgical patients and 92.1 ± 6.2% in adult ICU patients [160]. In the pediatric population (n—15, 0–13 years old), median unbound linezolid concentration was 7.5 mg/L while the median total concentration was 9.4 mg/L, which results in a 79.8% unbound fraction [161]. PPB for tedizolid is from 70 to 90% [162]. It is thought that in critically ill patients, in a hypoalbuminemia state, the PPB of tedizolid will be changed resulting in increased clearance [163].
Lincosamides are characterized by variable values of PPB. Clindamycin has high rates of PPB, from 78 to 94%, and it binds mainly AAG. Decreased concentrations of AAG specific for neonates are expected to affect clindamycin’s distribution into tissues [164]. Another lincosamide, lincomycin, possesses concentration-dependent PPB, varying from 28 to 86% and decreasing with drug plasma concentration increase [165].
A lipopeptide antibiotic daptomycin has a high PPB and binds to several proteins, first to HSA and α-1-acid-glycoprotein [166]. In adults PPB is between 90–94%, it does not depend on concentration, but is decreased with decreasing renal function [167]. The clinical significance of this fact is discussible, in the study of physiologically based pharmacokinetic modeling of daptomycin dose optimization in pediatric patients with renal impairment no dose adjustment was proposed for patients with mild-to-moderate renal impairment, and only for severe cases was a dose decrease recommended [168].
Fluoroquinolone levofloxacin binds both to albumin and globulin, PPB is from 24 to 38% [169] and changes in PK parameters in the hypoalbuminemia state are not expected [170]. PPB for moxifloxacin is about 50% and does not depend on drug concentration [171]. No studies reporting PPB changes in term and preterm neonates are available for these drugs.
Fosfomycin has a negligible PPB rate and change in plasma protein concentration is not an important factor here [172]. Other antibiotics with very low PPB rates include aminoglycosides, so change in PPB is not expected in neonates [20].
Rifampin PPB rate is from 72 to 92% in adults with tuberculosis [173] and is not dependent on drug concentration in plasma [174]. In PK studies of neonates, no certain values of PPB were estimated for rifampin, though clearance increase was reported with postnatal age increase [117].
An important characteristic of antibiotics with high PPB rates is their ability to displace bilirubin from its binding with albumin, which may lead to bilirubin-induced neurotoxicity in neonates with hyperbilirubinemia. In Table 6, examples of antibacterials with different bilirubin displacing activity are given.
Bilirubin displacing activity of drugs, listed above, was mainly reported using data from in vitro studies. Sulfonamides are among the drugs commonly thought to be responsible for kernicterus in neonates. Currently, one of the most used sulfonamides is the combined drug, sulfamethoxazole-trimethoprim. For sulfamethoxazole, there is a 70% PPB [176], and for trimethoprim, PPB is about 44% [177]. The review of clinical and animal studies with cotrimoxazole revealed no strict evidence of kernicterus caused by sulfamethoxazole-trimethoprim in neonates. Authors made a conclusion, that oral administration to neonates for 7–10 days is unlikely to cause kernicterus [178].
Ceftriaxone is another drug considered to have a high potential to displace bilirubin. Clinical studies reported that in neonates (at least 15 days old) a short-term course of ceftriaxone did not have a higher likelihood of developing hyperbilirubinemia compared with a short-term course of cefotaxime [179]. Another recent study also demonstrated the absence of ceftriaxone association with a bilirubin-displacing effect in infants with a mild unconjugated hyperbilirubinemia (n—27, born at term, <7 days old, diagnosed sepsis) [180].

2.3. Metabolism

The primary site for drug metabolism is the liver; others are the kidney, intestine, lungs, and plasma. Biotransformation of drugs is realized in two phases with the assistance of drug-metabolizing enzymes (DMEs). DMEs of phase I are responsible for oxidation, reduction, and hydrolysis reactions. DMEs of phase II are responsible for glucuronidation, sulfation, methylation, and acetylation reactions.
Biotransformation of drugs may lead to: (1) inactivation (e.g., chloramphenicol), which is terminating drug action to excretion, and (2) active metabolite formation from a prodrug. Considering antibiotics prodrugs are available for several pharmacological groups. Among the penicillin group, there are several prodrugs of ampicillin aimed at the improvement of its bioavailability (pivampicillin, talampicillin, bacampicillin, and hetacillin), or the improvement of its pharmacodynamic properties (sultamicillin, a prodrug that links ampicillin and sulbactam by a methylene group) [181]. Highly effective cephalosporins in the form of prodrugs are ceftaroline fosamil [182] and ceftobiprole medocaril [183]. New carbapenem tebipenem became orally available in the form of a prodrug, tebipenem pivoxil [184]. Another prodrug among synthetic antibacterials is metronidazole [185].
The most important enzymes responsible for phase I reactions are cytochrome P450 (CYP450) enzymes with the most significant role of CYP3A4 isoenzyme, which is responsible for the biotransformation of many drugs including antibiotics (erythromycin, linezolid, clindamycin, etc.).
Some antibiotics are metabolized involving not only the most common isoforms but are also less frequent. For example, it was recently demonstrated, that linezolid metabolism is performed with the assistance of several isoenzymes in next order CYP2J2 ≫ CYP4F2 > CYP2C8 > CYP1B1 ≈ CYP2D6 ≈ CYP3A4 > CYP1A1 > CYP3A5 [186].
Developmental changes in the metabolic activity of CYP450 isoenzymes may result in changes in antibiotics pharmacodynamics. In general, expression of DME is typically low at birth and gradually increases with age [187,188]. The level of CYP-dependent metabolism at birth is proposed to be 50–70% of the adult value. The mean total CYP450 protein expression measured with western blot analysis was 390.3 pmol/mg in an infant-neonate group (2 fetuses, 2 newborns, 6 infants), and 644.59 pmol/mg in the >1 year group (6 children, 4 adults, age 2–72 years) [189,190]. Unlike children and adults with dominating CYP3A4, the main fetal hepatic CYP450 isoenzyme is CYP3A7, which accounts for up to 50% of the total fetal hepatic CYP content and up to 87–100% of total fetal hepatic CYP3A content [191].
Three different developmental patterns (classes) were defined for hepatic DME by Hines RN et al. (2013) [192]:
  • First pattern (Class 1)—DME high in fetal life and low or absent after birth;
  • Second pattern (Class 2)—DME stable throughout development;
  • Third pattern (Class 3)—DME low in fetal life to increasing and high after birth.
Considering only CYP450, in the Class 1 there is only CYP3A7, in the Class 2—CYP2B6, CYP2C19, CYP3A5, and in the Class 3—CYP1A2, CYP2C9, CYP2D6, CYP2E1, CYP3A4 [192].
In the work by Fanni D et al. (2014), the activity of CYP1A2, CYP2C9, and CYP3A4 isoforms in 2 to 3 years old children was reported to be higher than in adults, while the activity of CYP2C19, CYP2D6 was similar to that in adults [187].
In Table 7 data on CYP450 isoenzyme ontogeny are given with information about antibiotics-substrates.
Drug interactions can significantly affect the treatment outcome. Drug interactions on the level of metabolism include interaction between CYP450 isoenzyme substrate and isoenzyme inhibitor or inducer, though substrate-substrate interactions are also possible. Among antibiotics, the most significant effect on CYP450 isoenzymes is produced by fourteen-membered ring macrolides (inhibition of CYP3A4 and 3A5) [205] and by rifampicin and its relatives (induction of CYP 3A4) [206].
Studies of fluoroquinolones revealed ciprofloxacin as a relatively strong inhibitor (≥5-fold increase in AUC or >80% decrease in clearance) of CYP1A2 [207]. Levofloxacin demonstrated inhibition of CYP2C9, and negligible inhibition of CYP1A2 and CYP2C9 was reported for caderofloxacin, antofloxacin, moxifloxacin, and gatifloxacin [208]. On the opposite side, novel fluoroquinolone compounds containing cyclopropylamine in the pyrrolidine ring of the 7′ position of fluoroquinolone molecule were demonstrated to be irreversible inhibitors of CYP3A [209,210].
Piperacillin was demonstrated to inhibit CYP2C8 [211], while other penicillins can induce isoenzymes. Flucloxacillin revealed a potential to induce expression of CYP3A4 and P-glycoprotein, most likely through the activation of the nuclear hormone receptor PXR [212], and dicloxacillin was shown to be an inducer of CYP2C19, CYP2C9, and CYP3A4 [53]. For carbapenem meropenem, there was demonstrated a substantial concentration-dependent interaction with voriconazole in a patient treated by these two drugs, and in vitro study confirmed the inhibitor action of meropenem on CYP3A4 and CYP2C19 [213].
Oritavancin produces a nonspecific weak inhibitory effect on CYP2C9 and CYP2C19, CYP1A2, CYP2B6, CYP2D6, CYP3A4, and simultaneously it is an inducer of CYP3A4 and CYP2D6 [214].
Antibiotics that can induce or inhibit main CYP450 isoenzymes are given in Table 8.
Other important DMEs of phase 1 are flavin-containing mono-oxygenase 3 (FMO3), hepatic carboxylesterases 1 (CES1) and -2 (CES2), and alcohol and aldehyde dehydrogenases (ADHs and ALDH, respectively) [189,194].
Studies revealed a lower expression of non-CYP I phase enzymes in neonates and pediatrics than in adults (ADH1A, ADH1B, ADH1C, ALDH1A1, CES1, CES2, and FMO3) [196]. CES1 abundance in hepatic microsomes revealed a 5-fold increase from neonates to adults (315.2 pmol/mg vs. 1664.4 pmol/mg), for CES2—a 3-fold increase (59.8 pmol/mg vs. 174.1 pmol/mg, respectively) [220].
FMO1 expression was reported to be the highest in the embryo (8–15 weeks of gestation; 7.8 ± 5.3 pmol/mg protein) with subsequent decrease, while FMO3 levels were low in embryos with an increase in the fetus [221]. FMO3 abundance in human liver microsomes demonstrated a 2.2-fold increase (13.0 ± 11.4 pmol/mg protein vs. 28.0 ± 11.8 pmol/mg protein) from neonates to adults with achievement of adult values after 6 years of age [222].
In neonates, the levels of protein expression for alcohol and aldehyde dehydrogenases were detected to be lower than in adults: 3-fold for ADH1A, 8-fold for ADH1B, 146-fold for ADH1C, and 3-fold for ALDH1A1. An increase in the levels was seen during the first year of life, and adult values were reached between 1 to 6 years. An exception was detected for ADH1A, its protein abundance in adults was ~40% lower than in the early childhood group [223].
Phase II reactions are catalyzed by uridine diphosphate glucuronosyltransferase (UGT), sulfotransferase (SUT), glutathione-S-transferase (GST), and N-acetyltransferase (NAT) families.
Sulfation of xenobiotics represents one of the most common phase II reactions. It aims to increase the hydrophilicity of drugs, enabling their biliary excretion and subsequent renal clearance. Sulfotransferase age-dependent activity was estimated in several studies. The activity of the cytosolic SUT measured with 2-naphthol revealed a mean value of 0.18 nmol/min/mg protein in fetal samples and 0.63 nmol/min/mg protein in adults [224]. In the study by Ladumor MK et al. (2019) significant age-dependent protein abundance was demonstrated for SULT1A1, SULT1B1, and SULT2A1, whereas SULT1A3 was stable across 0–70 years. In neonatal samples, mean abundances of SULT1A1, SULT1B1, and SULT2A1 accounted for 24%, 19%, and 38% of the adult values. The highest abundance during early childhood (1 to <6 years) was defined for SULT1A1 and SULT2A1 [225].
Glucuronidation is aimed at the completion of sulfation, and conjugation with glucuronic acid occurs at the same molecular moieties as sulfation. As a result of glucuronidation, inactive forms are produced with subsequent excretion of bile or urine. The phenomenon of neonatal jaundice suggested that UGT activity may be absent in the first days of life resulting in severe unconjugated bilirubinemia. Miyagi SJ et al. (2011) demonstrated minimum activity levels of UGT1A1 at birth with an increase of up to 90% of maximum values at 3.8 months of age. For UGT1A6 90% maximum activity was seen at 14 months of age. And for both enzymes estimation of protein expression revealed no age dependence [226].
Glutathione conjugation is specific for potent electrophiles, substrates include electrophilic carbon, nitrogen, oxygen, and sulfur atoms. Reaction products are inactive substances. GST isoenzymes demonstrated some age-dependent changes. Protein levels of GSTA1 were reported to be the lowest in the fetus (10–42 weeks) with a peak in the perinatal period (2–85 weeks) and a secondary decrease in adults. GSTA2 levels demonstrated a gradual increase from minimum in fetal samples to maximum in adults. GSTM levels were the lowest in the fetus with an increase of nearly up to adult values in the perinatal period. And opposite age distribution was revealed for GSTP—maximum in the fetal period with a decline to minimum in adults [227]. High levels of serum GST class Pi were detected in premature infants with severe perinatal asphyxia, compared to those with moderate asphyxia. Estimated high levels of serum GST Pi in the first 6 h after birth were associated with increased mortality and development of acute kidney failure in preterm neonates [228].
Considering antibacterial agents, it is important to discuss acetylation reactions, which involve compounds with amino or hydrazine residues, like sulfonamides or isoniazid. N-acetyltransferase age-dependent changes may result in variable efficacy and safety of these drugs. Insufficient or slow acetylation by NAT2 of sulfamethoxazole may result in increased toxicity risk including idiosyncratic adverse drug reactions [178]. NAT2 is also an enzyme catalyzing acetylation of the first-line antituberculosis agent isoniazid. Age was estimated as a critical factor for activity and affinity of NAT2, and the maximal adult type of activity was encountered after 5.3 years old [229].

2.4. Excretion

Drugs are excreted mainly with urine or bile, and total drug clearance is a sum of renal and hepatics clearances. Excretion with bile is specific for parenterally administered hydrophilic drugs extensively bound to plasma proteins, for orally administered drugs highly bound to hepatic transport proteins, and, in general, for polar large molecules [230]. Antibiotics excreted predominantly with bile include macrolides, rifampicin, fluoroquinolones, oxazolidinones, and lincosamides. Developmental changes in bile excretion were demonstrated in the work by Johnson TN et al. (2016) [231]. For neonates (26-week gestational age) a fraction of adult BE activity of 15% was necessary to predict the clearance of azithromycin, and 100% activity was apparent by 7 months. In premature neonates, (30 weeks gestational age) decreased biliary excretion was demonstrated for digoxin (10% of adults value). The ontogeny of biliary excretion reported for azithromycin, ceftriaxone, digoxin, and buprenorphine revealed rapid achievement of adult values, from days to several months after birth [231].
Antibiotics with high rates of urinary excretion include penicillins, cephalosporines, carbapenems, monobactam aztreonam, aminoglycosides, glycopeptides, cyclic lipopeptide daptomycin, phosphonic antibiotic fosfomycin. The value of renal clearance is based on the sum of the next renal functions: glomerular filtration, tubular secretion, and tubular reabsorption. Developmental changes are seen both in the structure and function of kidneys [232]. Nephrogenesis starts from the 6th week of gestation and its completion happens in the 36th week [233]. In preterm infants, nephrogenesis may continue for up to 40 days after birth. In preterms, kidneys are smaller than in term ones and a doubling of the mean total kidney volume happens from 28 to 37 weeks (from 10.3 cm3 at 28 weeks postmenstrual age to 19.2 cm3 at 37 weeks), though the estimated glomerular filtration rate (eGFR) between premature neonates and term neonates revealed no significant differences (43.5 [39.7–48.9] vs. 42.0 [38.2–50.0] mL/min/1.73 m2) [234]. Further study reported that infants born preterm were characterized by a smaller total kidney volume even at 24 months of age compared with term ones (56.1 (9.4) vs. 64.8 (10.2) mL), and by the same values of eGFR [235]. The meta-analysis by Smeets NJL et al. (2022) included 978 measured GFR values from 881 healthy, term-born neonates. Results revealed a doubling of GFR in the first 5 days of life (from 19.6 (95% CI, 14.7 to 24.6) to 40.6 (95% CI, 36.7 to 44.5) mL/min per 1.73 m2) followed by a gradual increase with the achievement of 59.4 (95% CI, 45.9 to 72.9) mL/min per 1.73 m2 at 4 weeks of age [236].
Salem F et al. (2021) developed an ontogeny model for GFR emphasizing the potent impact of birth on GFR value: longer period outside the uterus (greater postnatal age) resulted in higher GFR values in neonates with the same postmenstrual age. The authors stated that the difference in GFR between pre-term and full-term neonates with the same postmenstrual age is eliminated from beyond 1.25 years [237]. The evaluation of molecules related to glomerular function in preterm neonates (n—40, mean gestational age 30 ± 1 weeks) revealed a pronounced increase in GFR from 72 h until 3 weeks of life [238].
Generally, GFR is increased depending on gestational and post-menstrual age achieving adult values by 1-year post-natal age, and during this maturation period, the effective filtration pressure is driven by the interaction between multiple endogenous intra-renal vasoconstrictive and vasodilator factors [239]. A pathological shift of GFR in neonates may be mediated by a number of pathological states, including hemodynamic disturbances, very low-birth weight, and external factors (use of drugs resulting in vasoconstriction and nephrotoxicity), as it is shown on the Figure 2.
  • ACEi—angiotensin-converting enzyme inhibitors
  • ANP—atrial natriuretic peptide
  • AT I, AT II—angiotensin I and II
  • NO—nitric oxide
  • PGI2, PGE2—prostaglandins I2 and E2.
The next categories of neonates in NICU are at high risk of acute kidney injury [240]:
-
Premature and low birth weight neonates;
-
Neonates with congenital heart disease and cardiac surgery;
-
Neonates with hypoxic ischemic encephalopathy;
-
Neonates with necrotizing enterocolitis;
-
Neonates on extracorporeal life support;
-
Neonates using nephrotoxic drugs.
Antibacterials with possible negative effects on renal function may be involved in the development of tubulointerstitial nephritis (penicillins, cephalosporins, fluoroquinolones, sulfonamides) or acute tubular necrosis (aminoglycosides and amphotericin B) in neonates. The work by Rhone ET et al. (2014) revealed that for very low birth weight infants exposure to ≥1 nephrotoxic drug was reported in 87% (gentamicin—86%, indomethacin—43%, vancomycin—25%) [241].
The accurate estimation of GFR in neonates is essential for ICU treatment, increasing its effectiveness and decreasing the risk of negative outcomes. Methods of GFR determining include measurement of clearance of creatinine, inulin, iohexol, 51Cr-EDTA (ethylene diamine tetra-acetic acid), and 99mTc—DMSA (dimercaptosuccinic acid). The GFR estimate in children is mainly performed with updated versions of the Schwartz equation, though the serum cystatin C level may be used together with the serum creatinine level to estimate GFR [242]. Population pharmacokinetics modeling developed to predict clearance of renally eliminated drugs in neonates with normal renal function should be based on the body weight and postmenstrual age [243].
Developmental changes in renal function can affect the excretion of antibiotics, altering the value of clearance (CL) and, therefore, antibiotic exposure.
Table 9 demonstrates comparative data on PPB, Vd, and CL of antibiotics in neonates and adults.

3. Antibiotic Dosing Regimens in Neonates

Lack of pharmacokinetic trials and RCT including neonates results in challenges in antibiotics dosing; underdosing and underdosing are common factors of pharmacotherapy failure. Available data on antibiotics PK parameters together with simulation studies help to determine proper regimens for antibiotics used in neonates. Information about the proposed regimens of antibiotic dosing in neonates is given in Table 10.

4. Conclusions

Bacterial infections in neonates are considered to be among the main causes of NICU admission and may be life-threatening, especially to preterm newborns. NS highly contributes to neonatal mortality [320] emphasizing the importance of the proper antibiotic choice and dosing. Optimal dosing in neonates is essential to achieve the eradication of bacteria, prevent antibiotic resistance, and reduce the risk of possible toxic effects. There is a significant difference between preterm and full-term newborns, resulting in the inability to apply the same treatment schemes. Different rates of maturation of organs and tissues, transport proteins, and enzymes lead to the variability of PK parameters in heterogenous neonatal populations. Gastric emptying and renal, hepatic, and intestinal functions are potent factors affecting the absorption, distribution, metabolism, and excretion of antibiotics. Current data actualize and transform the postulate “children are not small adults” into “preterm neonates are not small full-term ones”. The extrapolation of PK data from more mature human subjects to neonates, and from term neonates on preterm neonates is challenging, making actual the need for additional pharmacokinetic studies, pharmacogenetic studies (distinguishing genetic and non-genetic factors affecting PK parameters of antibiotics in neonates [321,322]) and RCT in this vulnerable category of patients.

Author Contributions

Conceptualization, O.I.B., S.K.Z. and E.A.U.; resources, O.I.B. and S.K.Z.; data curation, S.K.Z. and E.A.U.; search of data sources—O.I.B. and M.S.C.; writing—original draft preparation, O.I.B.; writing—review and editing, O.I.B. and E.A.U.; supervision, S.K.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Sources of information used in this review are listed in the References.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Birrie, E.; Sisay, E.; Tibebu, N.S.; Tefera, B.D.; Zeleke, M.; Tefera, Z. Neonatal Sepsis and Associated Factors among Newborns in Woldia and Dessie Comprehensive Specialized Hospitals, North-East Ethiopia, 2021. Infect. Drug Resist. 2022, 15, 4169–4179. [Google Scholar] [CrossRef]
  2. Roble, A.K.; Ayehubizu, L.M.; Olad, H.M. Neonatal Sepsis and Associated Factors among Neonates Admitted to Neonatal Intensive Care Unit in General Hospitals, Eastern Ethiopia 2020. Clin. Med. Insights Pediatr. 2022, 16, 11795565221098346. [Google Scholar] [CrossRef] [PubMed]
  3. Van Den Anker, J.; Allegaert, K. Rational Use of Antibiotics in Neonates: Still in Search of Tailored Tools. Healthcare 2019, 7, 28. [Google Scholar] [CrossRef] [Green Version]
  4. Hsieh, E.M.; Hornik, C.P.; Clark, R.H.; Laughon, M.M.; Benjamin, D.K., Jr.; Smith, P.B. Best Pharmaceuticals for Children Act-Pediatric Trials Network. Medication Use in the Neonatal Intensive Care Unit. Am. J. Perinatol. 2013, 31, 811–822. [Google Scholar] [CrossRef] [Green Version]
  5. Prusakov, P.; Goff, D.A.; Wozniak, P.S.; Cassim, A.; Scipion, C.E.; Urzúa, S.; Ronchi, A.; Zeng, L.; Ladipo-Ajayi, O.; Aviles-Otero, N.; et al. A global point prevalence survey of antimicrobial use in neonatal intensive care units: The no-more-antibiotics and resistance (NO-MAS-R) study. EClinicalMedicine 2021, 32, 100727. [Google Scholar] [CrossRef]
  6. Giannoni, E.; Dimopoulou, V.; Klingenberg, C.; Navér, L.; Nordberg, V.; Berardi, A.; el Helou, S.; Fusch, G.; Bliss, J.M.; Lehnick, D.; et al. Analysis of Antibiotic Exposure and Early-Onset Neonatal Sepsis in Europe, North America, and Australia. JAMA Netw. Open 2022, 5, e2243691. [Google Scholar] [CrossRef] [PubMed]
  7. Fleischmann, C.; Reichert, F.; Cassini, A.; Horner, R.; Harder, T.; Markwart, R.; Tröndle, M.; Savova, Y.; Kissoon, N.; Schlattmann, P.; et al. Global incidence and mortality of neonatal sepsis: A systematic review and meta-analysis. Arch. Dis. Child. 2021, 106, 745–752. [Google Scholar] [CrossRef] [PubMed]
  8. Rudd, K.E.; Johnson, S.C.; Agesa, K.M.; Shackelford, K.A.; Tsoi, D.; Kievlan, D.R.; Colombara, D.V.; Ikuta, K.S.; Kissoon, N.; Finfer, S.; et al. Global, regional, and national sepsis incidence and mortality, 1990–2017: Analysis for the Global Burden of Disease Study. Lancet 2020, 395, 200–211. [Google Scholar] [CrossRef] [Green Version]
  9. Yu, Y.-Q.; He, X.-R.; Wan, L.-J.; Yang, Y.-H.; Chen, P.-Y. Etiology, antimicrobial resistance, and risk factors of neonatal sepsis in China: A systematic review and meta-analysis from data of 30 years. J. Matern. Fetal Neonatal Med. 2021, 35, 7541–7550. [Google Scholar] [CrossRef]
  10. Zelellw, D.A.; Dessie, G.; Mengesha, E.W.; Shiferaw, M.B.; Merhaba, M.M.; Emishaw, S. A Systemic Review and Meta-analysis of the Leading Pathogens Causing Neonatal Sepsis in Developing Countries. BioMed Res. Int. 2021, 2021, 6626983. [Google Scholar] [CrossRef]
  11. Sands, K.; Carvalho, M.J.; Portal, E.; Thomson, K.; Dyer, C.; Akpulu, C.; Andrews, R.; Ferreira, A.; Gillespie, D.; Hender, T.; et al. Characterization of antimicrobial-resistant Gram-negative bacteria that cause neonatal sepsis in seven low- and middle-income countries. Nat. Microbiol. 2021, 6, 512–523. [Google Scholar] [CrossRef] [PubMed]
  12. Thomson, K.M.; Dyer, C.; Liu, F.; Sands, K.; Portal, E.; Carvalho, M.J.; Barrell, M.; Boostrom, I.; Dunachie, S.; Farzana, R.; et al. Effects of antibiotic resistance, drug target attainment, bacterial pathogenicity and virulence, and antibiotic access and affordability on outcomes in neonatal sepsis: An international microbiology and drug evaluation prospective substudy (BARNARDS). Lancet Infect. Dis. 2021, 21, 1677–1688. [Google Scholar] [CrossRef]
  13. Solomon, S.; Akeju, O.; Odumade, O.A.; Ambachew, R.; Gebreyohannes, Z.; Van Wickle, K.; Abayneh, M.; Metaferia, G.; Carvalho, M.J.; Thomson, K.; et al. Prevalence and risk factors for antimicrobial resistance among newborns with gram-negative sepsis. PLoS ONE 2021, 16, e0255410. [Google Scholar] [CrossRef] [PubMed]
  14. Ji, H.; Yu, Y.; Huang, L.; Kou, Y.; Liu, X.; Li, S.; Zhang, Y.; Li, Z.; Sun, X.; Wang, J.; et al. Pathogen Distribution and Antimicrobial Resistance of Early Onset Sepsis in Very Premature Infants: A Real-World Study. Infect. Dis. Ther. 2022, 11, 1935–1947. [Google Scholar] [CrossRef]
  15. Fleiss, N.; Schwabenbauer, K.; Randis, T.M.; Polin, R.A. What’s new in the management of neonatal early-onset sepsis? Arch. Dis. Child. Fetal Neonatal Ed. 2023, 108, 10–14. [Google Scholar] [CrossRef] [PubMed]
  16. Paul, S.P.; Khattak, H.; Kini, P.K.; Heaton, P.A.; Goel, N. NICE guideline review: Neonatal infection: Antibiotics for prevention and treatment (NG195). Arch. Dis. Child. Educ. Pract. Ed. 2022, 107, 292–297. [Google Scholar] [CrossRef]
  17. Amer, Y.S.; Shaiba, L.A.; Hadid, A.; Anabrees, J.; Almehery, A.; Aassiri, M.; Alnemri, A.; Al Darwish, A.R.; Baqawi, B.; Aboshaiqah, A.; et al. Quality assessment of clinical practice guidelines for neonatal sepsis using the Appraisal of Guidelines for Research and Evaluation (AGREE) II Instrument: A systematic review of neonatal guidelines. Front. Pediatr. 2022, 10, 891572. [Google Scholar] [CrossRef] [PubMed]
  18. Hsia, Y.; Lee, B.R.; Versporten, A.; Yang, Y.; Bielicki, J.; Jackson, C.; Newland, J.; Goossens, H.; Magrini, N.; Sharland, M.; et al. Use of the WHO Access, Watch, and Reserve classification to define patterns of hospital antibiotic use (AWaRe): An analysis of paediatric survey data from 56 countries. Lancet Glob. Health 2019, 7, e861–e871. [Google Scholar] [CrossRef] [Green Version]
  19. Williams, P.; Qazi, S.; Agarwal, R.; Velaphi, S.; Bielicki, J.; Nambiar, S.; Giaquinto, C.; Bradley, J.; Noel, G.; Ellis, S.; et al. Antibiotics needed to treat multidrug-resistant infections in neonates. Bull. World Health Organ. 2022, 100, 797–807. [Google Scholar] [CrossRef]
  20. Darlow, C.A.; da Costa, R.M.A.; Ellis, S.; Franceschi, F.; Sharland, M.; Piddock, L.; Das, S.; Hope, W. Potential Antibiotics for the Treatment of Neonatal Sepsis Caused by Multidrug-Resistant Bacteria. Pediatr. Drugs 2021, 23, 465–484. [Google Scholar] [CrossRef]
  21. World Health Organization. The 2019 WHO AWaRe Classification of Antibiotics for Evaluation and Monitoring of Use. World Health Organization. 2019. Available online: https://apps.who.int/iris/handle/10665/327957 (accessed on 10 January 2023).
  22. Henry, C.J.; Semova, G.; Barnes, E.; Cotter, I.; Devers, T.; Rafaee, A.; Slavescu, A.; Cathain, N.O.; McCollum, D.; Roche, E.; et al. Neonatal sepsis: A systematic review of core outcomes from randomised clinical trials. Pediatr. Res. 2022, 91, 735–742. [Google Scholar] [CrossRef]
  23. Alghamdi, A.A.; Keers, R.N.; Sutherland, A.; Ashcroft, D.M. Prevalence and Nature of Medication Errors and Preventable Adverse Drug Events in Paediatric and Neonatal Intensive Care Settings: A Systematic Review. Drug Saf. 2019, 42, 1423–1436. [Google Scholar] [CrossRef] [Green Version]
  24. Basil, J.H.; Premakumar, C.M.; Ali, A.M.; Tahir, N.A.M.; Shah, N.M. Prevalence, Causes and Severity of Medication Administration Errors in the Neonatal Intensive Care Unit: A Systematic Review and Meta-Analysis. Drug Saf. 2022, 45, 1457–1476. [Google Scholar] [CrossRef] [PubMed]
  25. Shawahna, R.; Jaber, M.; Said, R.; Mohammad, K.; Aker, Y. Medication errors in neonatal intensive care units: A multicenter qualitative study in the Palestinian practice. BMC Pediatr. 2022, 22, 317. [Google Scholar] [CrossRef] [PubMed]
  26. Pawluk, S.; Jaam, M.; Hazi, F.; Al Hail, M.S.; El Kassem, W.; Khalifa, H.; Thomas, B.; Rouf, P.A. A description of medication errors reported by pharmacists in a neonatal intensive care unit. Int. J. Clin. Pharm. 2017, 39, 88–94. [Google Scholar] [CrossRef] [PubMed]
  27. Van Donge, T.; Bielicki, J.A.; Anker, J.V.D.; Pfister, M. Key Components for Antibiotic Dose Optimization of Sepsis in Neonates and Infants. Front. Pediatr. 2018, 6, 325. [Google Scholar] [CrossRef] [Green Version]
  28. Gianvecchio, C.; Lozano, N.A.; Henderson, C.; Kalhori, P.; Bullivant, A.; Valencia, A.; Su, L.; Bello, G.; Wong, M.; Cook, E.; et al. Variation in Mutant Prevention Concentrations. Front. Microbiol. 2019, 10, 42. [Google Scholar] [CrossRef] [Green Version]
  29. Sumi, C.D.; Heffernan, A.J.; Lipman, J.; Roberts, J.A.; Sime, F.B. What Antibiotic Exposures Are Required to Suppress the Emergence of Resistance for Gram-Negative Bacteria? A Systematic Review. Clin. Pharmacokinet. 2019, 58, 1407–1443. [Google Scholar] [CrossRef]
  30. Keij, F.M.; Kornelisse, R.F.; Hartwig, N.G.; Reiss, I.K.M.; Allegaert, K.; A Tramper-Stranders, G. Oral antibiotics for neonatal infections: A systematic review and meta-analysis. J. Antimicrob. Chemother. 2019, 74, 3150–3161. [Google Scholar] [CrossRef] [Green Version]
  31. Neal-Kluever, A.; Fisher, J.; Grylack, L.; Kakiuchi-Kiyota, S.; Halpern, W. Physiology of the Neonatal Gastrointestinal System Relevant to the Disposition of Orally Administered Medications. Drug Metab. Dispos. 2019, 47, 296–313. [Google Scholar] [CrossRef] [Green Version]
  32. 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. [Google Scholar] [CrossRef] [PubMed]
  33. Simeoli, R.; Cairoli, S.; Decembrino, N.; Campi, F.; Vici, C.D.; Corona, A.; Goffredo, B.M. Use of Antibiotics in Preterm Newborns. Antibiotics 2022, 11, 1142. [Google Scholar] [CrossRef] [PubMed]
  34. Shawahna, R. Pediatric Biopharmaceutical Classification System: Using Age-Appropriate Initial Gastric Volume. AAPS J. 2016, 18, 728–736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Gan, J.; Bornhorst, G.M.; Henrick, B.; German, J.B. Protein Digestion of Baby Foods: Study Approaches and Implications for Infant Health. Mol. Nutr. Food Res. 2018, 62, 1700231. [Google Scholar] [CrossRef] [PubMed]
  36. Bonner, J.J.; Vajjah, P.; Abduljalil, K.; Jamei, M.; Rostami-Hodjegan, A.; Tucker, G.T.; Johnson, T.N. Does age affect gastric emptying time? A model-based meta-analysis of data from premature neonates through to adults. Biopharm. Drug Dispos. 2015, 36, 245–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Bourlieu, C.; Ménard, 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. [Google Scholar] [CrossRef]
  38. Bartelink, I.H.; Rademaker, C.M.A.; Schobben, A.F.A.M.; van den Anker, J.N. Guidelines on Paediatric Dosing on the Basis of Developmental Physiology and Pharmacokinetic Considerations. Clin. Pharmacokinet. 2006, 45, 1077–1097. [Google Scholar] [CrossRef]
  39. Henderickx, J.G.E.; Zwittink, R.D.; Renes, I.B.; van Lingen, R.A.; van Zoeren-Grobben, D.; Jebbink, L.J.G.; Boeren, S.; van Elburg, R.M.; Knol, J.; Belzer, C. Maturation of the preterm gastrointestinal tract can be defined by host and microbial markers for digestion and barrier defense. Sci. Rep. 2021, 11, 12808. [Google Scholar] [CrossRef]
  40. Palla, M.R.; Harohalli, S.; Crawford, T.N.; Desai, N. Progression of Gastric Acid Production in Preterm Neonates: Utilization of In-vitro Method. Front. Pediatr. 2018, 6, 211. [Google Scholar] [CrossRef] [Green Version]
  41. Kelly, E.; Newell, S.; Brownlee, K.; Primrose, J.; Dear, P. Gastric acid secretion in preterm infants. Early Hum. Dev. 1993, 35, 215–220. [Google Scholar] [CrossRef]
  42. Omari, T.I.; Davidson, G.F. Multipoint measurement of intragastric pH in healthy preterm infants. Arch. Dis. Child. Fetal Neonatal Ed. 2003, 88, F517–F520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. van Groen, B.D.; van de Steeg, E.; Mooij, M.G.; van Lipzig, M.M.; de Koning, B.A.; 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. [Google Scholar] [CrossRef] [PubMed]
  44. Karpen, H.; Karpen, S.J. 95-Bile Acid Metabolism During Development. In Fetal and Neonatal Physiology, 5th ed.; Polin, R.A., Abman, S.H., Rowitch, D.H., Benitz, W.E., Fox, W.W., Eds.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 913–929. ISBN 9780323352147. [Google Scholar] [CrossRef]
  45. Konieczna, A.; Erdösová, B.; Lichnovská, R.; Jandl, M.; Čížková, K.; Ehrmann, J. Differential expression of ABC transporters (MDR1, MRP1, BCRP) in developing human embryos. J. Mol. Histol. 2011, 42, 567–574. [Google Scholar] [CrossRef] [PubMed]
  46. Prasad, B.; Gaedigk, A.; Vrana, M.; Gaedigk, R.; Leeder, J.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Qian, X.; Cheng, Y.-H.; Mruk, D.D.; Cheng, C.Y. Breast cancer resistance protein (Bcrp) and the testis—An unexpected turn of events. Asian J. Androl. 2013, 15, 455–460. [Google Scholar] [CrossRef] [Green Version]
  48. BCRP/ABCG2 Substrates. Available online: https://go.drugbank.com/categories/DBCAT002663 (accessed on 20 January 2023).
  49. Klaassen, C.D.; Aleksunes, L.M. Xenobiotic, Bile Acid, and Cholesterol Transporters: Function and Regulation. Pharmacol. Rev. 2010, 62, 1–96. [Google Scholar] [CrossRef] [Green Version]
  50. Sugie, M.; Asakura, E.; Zhao, Y.L.; Torita, S.; Nadai, M.; Baba, K.; Kitaichi, K.; Takagi, K.; Takagi, K.; Hasegawa, T. Possible Involvement of the Drug Transporters P Glycoprotein and Multidrug Resistance-Associated Protein Mrp2 in Disposition of Azithromycin. Antimicrob. Agents Chemother. 2004, 48, 809–814. [Google Scholar] [CrossRef] [Green Version]
  51. Sakaeda, T.; Nakamura, T.; Okumura, K. MDR1 Genotype-Related Pharmacokinetics and Pharmacodynamics. Biol. Pharm. Bull. 2002, 25, 1391–1400. [Google Scholar] [CrossRef] [Green Version]
  52. Putnam, W.S.; Woo, J.M.; Huang, Y.; Benet, L.Z. Effect of theMDR1C3435T Variant and P-Glycoprotein Induction on Dicloxacillin Pharmacokinetics. J. Clin. Pharmacol. 2005, 45, 411–421. [Google Scholar] [CrossRef]
  53. Stage, T.B.; Graff, M.; Wong, S.; Rasmussen, L.; Nielsen, F.; Pottegård, A.; Brøsen, K.; Kroetz, D.L.; Khojasteh, S.C.; Damkier, P. Dicloxacillin induces CYP2C19, CYP2C9 and CYP3A4 in vivo and in vitro. Br. J. Clin. Pharmacol. 2018, 84, 510–519. [Google Scholar] [CrossRef] [Green Version]
  54. Čížková, D.; Mokrý, J.; Mičuda, S.; Österreicher, J.; Martínková, J. Expression of MRP2 and MDR1 transporters and other hepatic markers in rat and human liver and in WRL 68 cell line. Physiol. Res. 2005, 54, 419–428. [Google Scholar] [CrossRef] [PubMed]
  55. Mooij, M.G.; Schwarz, U.I.; De Koning, B.A.E.; Leeder, J.S.; Gaedigk, R.; Samsom, J.N.; Spaans, E.; van Goudoever, J.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Human Transporters MRP2. Available online: https://www.solvobiotech.com/transporters/mrp2 (accessed on 20 January 2023).
  57. Maeda, T.; Takahashi, K.; Ohtsu, N.; Oguma, T.; Ohnishi, T.; Atsumi, R.; Tamai, I. Identification of Influx Transporter for the Quinolone Antibacterial Agent Levofloxacin. Mol. Pharm. 2007, 4, 85–94. [Google Scholar] [CrossRef]
  58. Franke, R.; Baker, S.; Mathijssen, R.; Schuetz, E.; Sparreboom, A. Influence of Solute Carriers on the Pharmacokinetics of CYP3A4 Probes. Clin. Pharmacol. Ther. 2008, 84, 704–709. [Google Scholar] [CrossRef] [PubMed]
  59. Mahalingam, A.; Shenoy, B. Tebipenem: A Novel Oral Carbapenem. Pediatr. Infect. Dis. 2020, 2, 25–28. [Google Scholar] [CrossRef]
  60. Nakakariya, M.; Shimada, T.; Irokawa, M.; Maeda, T.; Tamai, I. Identification and Species Similarity of OATP Transporters Responsible for Hepatic Uptake of β-Lactam Antibiotics. Drug Metab. Pharmacokinet. 2008, 23, 347–355. [Google Scholar] [CrossRef]
  61. Thomson, M.M.S.; Hines, R.N.; Schuetz, E.G.; Meibohm, B. Expression Patterns of Organic Anion Transporting Polypeptides 1B1 and 1B3 Protein in Human Pediatric Liver. Drug Metab. Dispos. 2016, 44, 999–1004. [Google Scholar] [CrossRef] [Green Version]
  62. Tamai, I.; Nezu, J.-I.; Uchino, H.; Sai, Y.; Oku, A.; Shimane, M.; Tsuji, A. Molecular Identification and Characterization of Novel Members of the Human Organic Anion Transporter (OATP) Family. Biochem. Biophys. Res. Commun. 2000, 273, 251–260. [Google Scholar] [CrossRef]
  63. Kato, K.; Shirasaka, Y.; Kuraoka, E.; Kikuchi, A.; Iguchi, M.; Suzuki, H.; Shibasaki, S.; Kurosawa, T.; Tamai, I. Intestinal absorption mechanism of tebipenem pivoxil, a novel oral carbapenem: Involvement of human OATP family in apical membrane transport. Mol. Pharm. 2010, 7, 1747–1756. [Google Scholar] [CrossRef]
  64. 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. [Google Scholar] [CrossRef] [Green Version]
  65. van Elburg, R.M.; Fetter, W.P.F.; Bunkers, C.M.; Heymans, H.S.A. Intestinal permeability in relation to birth weight and gestational and postnatal age. Arch. Dis. Child. Fetal Neonatal Ed. 2003, 88, F52–F55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Flanagan, S.D.; Takahashi, L.H.; Liu, X.; Benet, L.Z. Contributions of saturable active secretion, passive transcellular, and paracellular diffusion to the overall transport of furosemide across adenocarcinoma (Caco-2) cells. J. Pharm. Sci. 2002, 91, 1169–1177. [Google Scholar] [CrossRef]
  67. Johnson, T.N.; Tanner, M.S.; Taylor, C.J.; Tucker, G.T. Enterocytic CYP3A4 in a paediatric population: Developmental changes and the effect of coeliac disease and cystic fibrosis. Br. J. Clin. Pharmacol. 2001, 51, 451–460. [Google Scholar] [CrossRef] [Green Version]
  68. Huang, N.N.; High, R.H. Comparison of serum levels following the administration of oral and parenteral preparations of penicillin to infants and children of various age groups. J. Pediatr. 1953, 42, 657–668. [Google Scholar] [CrossRef] [PubMed]
  69. Bergdahl, S.; Eriksson, M.; Finkel, Y. Plasma concentration following oral administration of di-and flucloxacillin in infants and children. Pharmacol. Toxicol. 1987, 60, 233–234. [Google Scholar] [CrossRef]
  70. Bergdahl, S.; Eriksson, M.; Finkel, Y.; Lännergren, K. Oral absorption of flucloxacillin in infants and young children. Acta Pharmacol. Toxicol. 1996, 58, 255–258. [Google Scholar] [CrossRef] [PubMed]
  71. Herngren, L.; Ehrnebo, M.; Broberger, U. Pharmacokinetics of free and total flucloxacilin in newborn infants. Eur. J. Clin. Pharmacol. 1987, 32, 403–409. [Google Scholar] [CrossRef] [PubMed]
  72. Cohen, M.D.; Raeburn, J.A.; Devine, J.; Kirkwood, J.; Elliott, B.; Cockburn, F.; Forfar, J.O. Pharmacology of some oral penicillins in the newborn infant. Arch. Dis. Child. 1975, 50, 230–234. [Google Scholar] [CrossRef] [Green Version]
  73. Keij, F.M.; Tramper-Stranders, G.A.; Koch, B.C.P.; Reiss, I.K.M.; Muller, A.E.; Kornelisse, R.F.; Allegaert, K. Pharmacokinetics of Clavulanic Acid in the Pediatric Population: A Systematic Literature Review. Clin. Pharmacokinet. 2022, 61, 637–653. [Google Scholar] [CrossRef]
  74. Zhang, J.; Wang, Y.; Xie, H.; Wang, R.; Jia, Z.; Men, X.; Xu, L.; Zhang, Q. Pharmacokinetics study of amoxycillin and clavulanic acid (8:1)-A new combination in healthy Chinese adult male volunteers using the LC–MS/MS method. Cell Biochem. Biophys. 2013, 65, 363–372. [Google Scholar] [CrossRef]
  75. Mir, F.; Pearce, R.E.; Baig-Ansari, N.; Qazi, S.; Barrett, J.S.; Abdel-Rahman, S.; Kearns, G.; Zaidi, A.K. Serum amoxicillin levels in young infants (0–59 days) with sepsis treated with oral amoxicillin. Arch. Dis. Child. 2020, 105, 1208–1214. [Google Scholar] [CrossRef]
  76. Autmizguine, J.; Watt, K.M.; Théorêt, Y.; Kassir, N.; Laferrière, C.; Parent, S.; Tapiéro, B.; Ovetchkine, P. Pharmacokinetics and pharmacodynamics of oral cephalexin in children with osteoarticular infections. Pediatr. Infect. Dis. J. 2013, 32, 1340–1344. [Google Scholar] [CrossRef]
  77. Boothman, R.; Kerr, M.M.; Marshall, M.J.; Burland, W.L. Absorption and excretion of cephalexin by the newborn infant. Arch. Dis. Child. 1973, 48, 147–150. [Google Scholar] [CrossRef] [Green Version]
  78. Ginsburg, C.M.; McCracken, G.H. Pharmacokinetics of Cephradine Suspension in Infants and Children. Antimicrob. Agents Chemother. 1979, 16, 74–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Ginsburg, C.M.; McCracken, G.H.; Clahsen, J.C.; Thomas, M.L. Clinical Pharmacology of Cefadroxil in Infants and Children. Antimicrob. Agents Chemother. 1978, 13, 845–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Chin, K.C.; Kerr, M.M.; Cockburn, F.; McAllister, T.A. A pharmacological study of cefaclor in the newborn infant. Curr. Med. Res. Opin. 1981, 7, 168–170. [Google Scholar] [CrossRef]
  81. McCracken, G.H.; Ginsburg, C.M.; Clahsen, J.C.; Thomas, M.L. Pharmacokinetics of cefaclor in infants and children. J. Antimicrob. Chemother. 1978, 4, 515–521. [Google Scholar] [CrossRef] [PubMed]
  82. Sáez-Llorens, X.; Shyu, W.C.; Shelton, S.; Kumiesz, H.; Nelson, J. Pharmacokinetics of cefprozil in infants and children. Antimicrob. Agents Chemother. 1990, 34, 2152–2155. [Google Scholar] [CrossRef] [Green Version]
  83. Powell, D.A.; James, N.C.; Ossi, M.J.; Nahata, M.C.; Donn, K.H. Pharmacokinetics of cefuroxime axetil suspension in infants and children. Antimicrob. Agents Chemother. 1991, 35, 2042–2045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Tan, B.J. Cefixime use in children: When and why. Can. J. Infect. Dis. 1995, 6, 204–205. [Google Scholar] [CrossRef] [Green Version]
  85. Kearns, G.L.; Reed, M.D.; Jacobs, R.F.; Ardite, M.; Yogev, R.D.; Blumer, J.L. Single-dose pharmacokinetics of ceftibuten (SCH 39720) in infants and children. Antimicrob. Agents Chemother. 1991, 35, 2078–2084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Kearns, G.L.; Abdel-Rahman, S.M.; Jacobs, R.F.; Wells, T.G.; Borin, M.T. Cefpodoxime pharmacokinetics in children: Effect of food. Pediatr. Infect. Dis. J. 1998, 17, 799–804. [Google Scholar] [CrossRef] [PubMed]
  87. Sato, N.; Kijima, K.; Koresawa, T.; Mitomi, N.; Morita, J.; Suzuki, H.; Hayashi, H.; Shibasaki, S.; Kurosawa, T.; Totsuka, K. Population pharmacokinetics of tebipenem pivoxil (ME1211), a novel oral carbapenem antibiotic, in pediatric patients with otolaryngological infection or pneumonia. Drug Metab. Pharmacokinet. 2008, 23, 434–446. [Google Scholar] [CrossRef] [Green Version]
  88. Eriksson, M.; Bolme, P.; Blennow, M. Absorption of erythromycin from pediatric suspension in infants and children. Scand. J. Infect. Dis. 1981, 13, 211–215. [Google Scholar] [CrossRef]
  89. Stratchunsky, L.S.; Nazarov, A.D.; Firsov, A.A.; Petrachenkova, N.A. Age dependence of erythromycin rectal bioavailability in children. Eur. J. Drug Metab. Pharmacokinet. 1991, 3, 321–323. [Google Scholar]
  90. Nahata, M.C.; Koranyi, K.I.; Gadgil, S.D.; Hilligoss, D.M.; Fouda, H.G.; Gardner, M.J. Pharmacokinetics of azithromycin in pediatric patients after oral administration of multiple doses of suspension. Antimicrob. Agents Chemother. 1993, 37, 314–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Stevens, R.C.; Reed, M.D.; Shenep, J.L.; Baker, D.K.; Foulds, G.; Luke, D.R.; Blumer, J.L.; Rodman, J.H. Pharmacokinetics of azithromycin after single- and multiple-doses in children. Pharmacotherapy 1997, 17, 574–880. [Google Scholar]
  92. Liu, P.; Fang, A.F.; LaBadie, R.R.; Crownover, P.H.; Arguedas, A.G. Comparison of azithromycin pharmacokinetics following single oral doses of extended-release and immediate-release formulations in children with acute otitis media. Antimicrob. Agents Chemother. 2011, 55, 5022–5026. [Google Scholar] [CrossRef] [Green Version]
  93. Gan, V.N.; Chu, S.Y.; Kusmiesz, H.T.; Craft, J.C. Pharmacokinetics of a clarithromycin suspension in infants and children. Antimicrob. Agents Chemother. 1992, 36, 2478–2480. [Google Scholar] [CrossRef] [Green Version]
  94. Guay, D.R.P.; Craft, C.J. Overview of the pharmacology of clarithromycin suspension in children and a comparison with that in adults. Pediatr. Infect. Dis. J. 1993, 12 (Suppl. S3), S106–S111. [Google Scholar] [CrossRef]
  95. Minotti, C.; Bonadies, L.; Liberati, C.; De Pieri, M.; Giaquinto, C.; Baraldi, E.; Donà, D. Enteral Linezolid as an Effective Option to Treat an Extremely Preterm Infant with Bacillus cereus Sepsis. Children 2022, 9, 415. [Google Scholar] [CrossRef]
  96. Banniettis, N.; Sharma, R.; Hand, I.; Kohlhoff, S.; Peloquin, C.A.; Hammerschlag, M.R. Steady-state pharmacokinetics of oral linezolid suspension in a premature infant with osteomyelitis. J. Antimicrob. Chemother. 2016, 71, 1738. [Google Scholar] [CrossRef] [Green Version]
  97. Sicard, M.; Launay, E.; Caillon, J.; Jacqueline, C.; Legrand, A.; Deslandes, G.; Navas, D.; Rozé, J.-C.; Guen, C.G.-L. Pharmacokinetics of linezolid treatment using intravenous and oral administrations in extremely premature infants. Eur. J. Clin. Pharmacol. 2015, 71, 611–615. [Google Scholar] [CrossRef] [PubMed]
  98. A Pharmacokinetic Study of Tedizolid Phosphate in Pediatric Participants with Gram-Positive Infections (MK-1986-014). Available online: https://clinicaltrials.gov/ct2/show/NCT03217565 (accessed on 28 January 2023).
  99. Arrieta, A.C.; Ang, J.Y.; Espinosa, C.; Fofanov, O.; Tøndel, C.; Chou, M.Z.; De Anda, C.S.; Kim, J.Y.; Li, D.; Sabato, P.; et al. Pharmacokinetics and Safety of Single-dose Tedizolid Phosphate in Children 2 to <12 Years of Age. Pediatr. Infect. Dis. J. 2021, 40, 317–323. [Google Scholar] [CrossRef]
  100. Bradley, J.S.; Flanagan, S.D.; Arrieta, A.C.; Jacobs, R.; Capparelli, E.; Prokocimer, P. Pharmacokinetics, Safety and Tolerability of Single Oral or Intravenous Administration of 200 mg Tedizolid Phosphate in Adolescents. Pediatr. Infect. Dis. J. 2016, 35, 628–633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Payen, S.; Serreau, R.; Munck, A.; Aujard, Y.; Aigrain, Y.; Bressolle, F.; Jacqz-Aigrain, E. Population pharmacokinetics of ciprofloxacin in pediatric and adolescent patients with acute infections. Antimicrob. Agents Chemother. 2003, 47, 3170–3178. [Google Scholar] [CrossRef] [Green Version]
  102. Zhao, W.; Hill, H.; Le Guellec, C.; Neal, T.; Mahoney, S.; Paulus, S.; Castellan, C.; Kassai, B.; Anker, J.N.V.D.; Kearns, G.L.; et al. Population pharmacokinetics of ciprofloxacin in neonates and young infants less than three months of age. Antimicrob. Agents Chemother. 2014, 58, 6572–6580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Chien, S.; Wells, T.G.; Blumer, J.L.; Kearns, G.L.; Bradley, J.S.; Bocchini, J.A., Jr.; Natarajan, J.; Maldonado, S.; Noel, G.J. Levofloxacin pharmacokinetics in children. J. Clin. Pharmacol. 2005, 45, 153–160. [Google Scholar] [CrossRef]
  104. Thee, S.; Garcia-Prats, A.J.; Draper, H.R.; McIlleron, H.M.; Wiesner, L.; Castel, S.; Schaaf, H.S.; Hesseling, A.C. Pharmacokinetics and safety of moxifloxacin in children with multidrug-resistant tuberculosis. Clin. Infect. Dis. 2014, 60, 549–556. [Google Scholar] [CrossRef] [Green Version]
  105. Capparelli, E.V.; Reed, M.D.; Bradley, J.S.; Kearns, G.L.; Jacobs, R.F.; Damle, B.D.; Blumer, J.L.; Grasela, D.M. Pharmacokinetics of gatifloxacin in infants and children. Antimicrob. Agents Chemother. 2005, 49, 1106–1112. [Google Scholar] [CrossRef] [Green Version]
  106. Siu, Y.K.; Ng, P.C.; Fung, S.C.K.; Lee, C.H.; Wong, M.Y.; Fok, T.F.; So, K.W.; Cheung, K.L.; Wong, W.; Cheng, A.F.B. Double blind, randomised, placebo controlled study of oral vancomycin in prevention of necrotising enterocolitis in preterm, very low birthweight infants. Arch. Dis. Child. Fetal Neonatal Ed. 1998, 79, F105–F109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Antoon, J.W.; Hall, M.; Metropulos, D.; Steiner, M.J.; Jhaveri, R.; Lohr, J.A. A Prospective Pilot Study on the Systemic Absorption of Oral Vancomycin in Children with Colitis. J. Pediatr. Pharmacol. Ther. 2016, 21, 426–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Bergeron, L.; Boucher, F.D. Possible red-man syndrome associated with systemic absorption of oral vancomycin in a child with normal renal function. Ann. Pharmacother. 1994, 28, 581–584. [Google Scholar] [CrossRef] [PubMed]
  109. Yamazaki, S.; Suzuki, T.; Suzuki, T.; Takatsuka, H.; Ishikawa, M.; Hattori, N.; Fujishiro, T.; Miyauchi, H.; Oami, T.; Ariyoshi, N.; et al. An extremely high bioavailability of orally administered vancomycin in a patient with severe colitis and renal insufficiency. J. Infect. Chemother. 2017, 23, 848–851. [Google Scholar] [CrossRef]
  110. Autmizguine, J.; Melloni, C.; Hornik, C.P.; Dallefeld, S.; Harper, B.; Yogev, R.; Sullivan, J.E.; Atz, A.M.; Al-Uzri, A.; Mendley, S.; et al. Population Pharmacokinetics of Trimethoprim-Sulfamethoxazole in Infants and Children. Antimicrob. Agents Chemother. 2017, 62, e01813-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Álvarez, L.A.; Van de Sijpe, G.; Desmet, S.; Metsemakers, W.-J.; Spriet, I.; Allegaert, K.; Rozenski, J. Ways to Improve Insights into Clindamycin Pharmacology and Pharmacokinetics Tailored to Practice. Antibiotics 2022, 11, 701. [Google Scholar] [CrossRef]
  112. Hashemian, S.M.; Farhadi, Z.; Farhadi, T. Fosfomycin: The characteristics, activity, and use in critical care. Ther. Clin. Risk Manag. 2019, 15, 525–530. [Google Scholar] [CrossRef] [Green Version]
  113. Kane, Z.; Gastine, S.; Obiero, C.; Williams, P.; Murunga, S.; Thitiri, J.; Ellis, S.; Correia, E.; Nyaoke, B.; Kipper, K.; et al. IV and oral fosfomycin pharmacokinetics in neonates with suspected clinical sepsis. J. Antimicrob. Chemother. 2021, 76, 1855–1864. [Google Scholar] [CrossRef]
  114. Dijkmans, A.C.; Zacarías, N.V.O.; Burggraaf, J.; Mouton, J.W.; Wilms, E.B.; van Nieuwkoop, C.; Touw, D.J.; Stevens, J.; Kamerling, I.M.C. Fosfomycin: Pharmacological, Clinical and Future Perspectives. Antibiotics 2017, 6, 24. [Google Scholar] [CrossRef] [Green Version]
  115. Schaaf, H.S.; Willemse, M.; Cilliers, K.; Labadarios, D.; Maritz, J.S.; Hussey, G.D.; McIlleron, H.; Smith, P.; Donald, P.R. Rifampin pharmacokinetics in children, with and without human immunodeficiency virus infection, hospitalized for the management of severe forms of tuberculosis. BMC Med. 2009, 7, 19. [Google Scholar] [CrossRef] [Green Version]
  116. Koup, J.R.; Williams-Warren, J.; Viswanathan, C.T.; Weber, A.; Smith, A.L. Pharmacokinetics of Rifampin in Children II. Oral Bioavailability. Ther. Drug Monit. 1986, 8, 17–22. [Google Scholar] [CrossRef] [PubMed]
  117. Smith, P.B.; Cotten, C.M.; Hudak, M.L.; Sullivan, J.E.; Poindexter, B.B.; Cohen-Wolkowiez, M.; Boakye-Agyeman, F.; Lewandowski, A.; Anand, R.; Benjamin, D.K.; et al. Rifampin Pharmacokinetics and Safety in Preterm and Term Infants. Antimicrob. Agents Chemother. 2019, 63, e00284-19. [Google Scholar] [CrossRef] [Green Version]
  118. Benedetti, M.S.; Whomsley, R.; Baltes, E.L. Differences in absorption, distribution, metabolism and excretion of xenobiotics between the paediatric and adult populations. Expert Opin. Drug Metab. Toxicol. 2005, 1, 447–471. [Google Scholar] [CrossRef] [PubMed]
  119. Turner, C.; Thein, N.A.M.; Turner, P.; Nosten, F.; White, N.J. Rectal pH in well and unwell infants. J. Trop. Pediatr. 2012, 58, 311–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Rathi, R.; Kumar, A.; Vishvakarma, V.; Huanbutta, K.; Singh, I.; Sangnim, T. Advancements in Rectal Drug Delivery Systems: Clinical Trials, and Patents Perspective. Pharmaceutics 2022, 14, 2210. [Google Scholar] [CrossRef] [PubMed]
  121. Linakis, M.W.; Roberts, J.K.; Lala, A.C.; Spigarelli, M.G.; Medlicott, N.; Reith, D.M.; Ward, R.M.; Sherwin, C.M.T. Challenges Associated with Route of Administration in Neonatal Drug Delivery. Clin. Pharmacokinet. 2016, 55, 185–196. [Google Scholar] [CrossRef]
  122. 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. [Google Scholar] [CrossRef] [Green Version]
  123. Hansen, T.G. Developmental paediatric anaesthetic pharmacology. Anaesth. Intensiv. Care Med. 2018, 19, 437–443. [Google Scholar] [CrossRef]
  124. Meesters, K.; Alemayehu, T.; Benou, S.; Buonsenso, D.; Decloedt, E.H.; Lorente, V.P.-F.; Downes, K.J.; Allegaert, K. Pharmacokinetics of Antimicrobials in Children with Emphasis on Challenges Faced by Low and Middle Income Countries, a Clinical Review. Antibiotics 2022, 12, 17. [Google Scholar] [CrossRef]
  125. 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. [Google Scholar] [CrossRef]
  126. Villar, J.; Puglia, F.A.; Fenton, T.R.; Ismail, L.C.; Staines-Urias, E.; Giuliani, F.; Ohuma, E.O.; Victora, C.G.; Sullivan, P.; Barros, F.C.; et al. Body composition at birth and its relationship with neonatal anthropometric ratios: The newborn body composition study of the INTERGROWTH-21st project. Pediatr. Res. 2017, 82, 305–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Bortolotto, C.C.; Santos, I.S.; Vaz, J.D.S.; Matijasevich, A.; Barros, A.J.D.; Barros, F.C.; Santos, L.P.; Munhoz, T.N. Prematurity and body composition at 6, 18, and 30 years of age: Pelotas (Brazil) 2004, 1993, and 1982 birth cohorts. BMC Public Health 2021, 21, 321. [Google Scholar] [CrossRef] [PubMed]
  128. Anderson, B.J. Neonatal pharmacology. Anaesth. Intensiv. Care Med. 2016, 18, 68–74. [Google Scholar] [CrossRef]
  129. Ahmed, H.; Bergmann, F.; Zeitlinger, M. Protein Binding in Translational Antimicrobial Development-Focus on Interspecies Differences. Antibiotics 2022, 11, 923. [Google Scholar] [CrossRef] [PubMed]
  130. Torer, B.; Hanta, D.; Yapakci, E.; Gokmen, Z.; Parlakgumus, A.; Gulcan, H.; Tarcan, A. Association of Serum Albumin Level and Mortality in Premature Infants. J. Clin. Lab. Anal. 2016, 30, 867–872. [Google Scholar] [CrossRef] [Green Version]
  131. Bunt, J.E.H.; Rietveld, T.; Schierbeek, H.; Wattimena, J.L.D.; Zimmermann, L.J.I.; van Goudoever, J.B. Albumin synthesis in preterm infants on the first day of life studied with [1-13C]leucine. Am. J. Physiol. Gastrointest. Liver Physiol. 2007, 292, G1157–G1161. [Google Scholar] [CrossRef] [Green Version]
  132. Trainor, G.L. The importance of plasma protein binding in drug discovery. Expert Opin. Drug Discov. 2007, 2, 51–64. [Google Scholar] [CrossRef]
  133. Maharaj, A.R.; Gonzalez, D.; Cohen-Wolkowiez, M.; Hornik, C.P.; Edginton, A.N. Improving Pediatric Protein Binding Estimates: An Evaluation of α1-Acid Glycoprotein Maturation in Healthy and Infected Subjects. Clin. Pharmacokinet. 2018, 57, 577–589. [Google Scholar] [CrossRef]
  134. Anell-Olofsson, M.; Ahmadi, S.; Lönnqvist, P.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Liu, X.; Wright, M.; Hop, C.E.C.A. Rational use of plasma protein and tissue binding data in drug design. J. Med. Chem. 2014, 57, 8238–8248. [Google Scholar] [CrossRef] [PubMed]
  136. Neuville, M.; El-Helali, N.; Magalhaes, E.; Radjou, A.; Smonig, R.; Soubirou, J.-F.; Voiriot, G.; Le Monnier, A.; Ruckly, S.; Bouadma, L.; et al. Systematic overdosing of oxa- and cloxacillin in severe infections treated in ICU: Risk factors and side effects. Ann. Intensiv. Care 2017, 7, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Ulldemolins, M.; Roberts, J.A.; Wallis, S.C.; Rello, J.; Lipman, J. Flucloxacillin dosing in critically ill patients with hypoalbuminaemia: Special emphasis on unbound pharmacokinetics. J. Antimicrob. Chemother. 2010, 65, 1771–1778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Wallenburg, E.; ter Heine, R.; de Lange, D.W.; van Leeuwen, H.; Schouten, J.A.; Oever, J.T.; Kolwijck, E.; Burger, D.M.; Pickkers, P.; Gieling, E.M.; et al. High unbound flucloxacillin fraction in critically ill patients. J. Antimicrob. Chemother. 2021, 76, 3220–3228. [Google Scholar] [CrossRef] [PubMed]
  139. Pullen, J.; Stolk, L.M.L.; Degraeuwe, P.L.J.; Van Tiel, F.H.; Neef, C.; Zimmermann, L.J.I. Protein binding of flucloxacillin in neonates. Ther. Drug Monit. 2007, 29, 279–283. [Google Scholar] [CrossRef] [PubMed]
  140. Jongmans, C.; Muller, A.E.; Broek, P.V.D.; Almeida, B.D.M.C.D.; Berg, C.V.D.; Van Oldenrijk, J.; Bos, P.K.; Koch, B.C.P. An Overview of the Protein Binding of Cephalosporins in Human Body Fluids: A Systematic Review. Front. Pharmacol. 2022, 13, 900551. [Google Scholar] [CrossRef]
  141. Kan, M.; Wu, Y.-E.; Li, X.; Dong, Y.-N.; Du, B.; Guo, Z.-X.; Shi, H.-Y.; Huang, X.; Su, L.-Q.; Wang, W.-Q.; et al. An adapted LC-MS/MS method for the determination of free plasma concentration of cefoperazone in children: Age-dependent protein binding. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2020, 1144, 122081. [Google Scholar] [CrossRef]
  142. DrugBank Online. Ceftriaxone. Available online: https://go.drugbank.com/drugs/DB01212 (accessed on 1 February 2023).
  143. Kan, M.; Shi, H.-Y.; Han, B.; Wu, Y.-E.; Li, Q.; Guo, Z.-X.; Li, X.; Hao, G.-X.; Zheng, Y.; Su, L.-Q.; et al. Prediction of Unbound Ceftriaxone Concentration in Children: Simple Bioanalysis Method and Basic Mathematical Equation. Antimicrob. Agents Chemother. 2020, 65, e00779-20. [Google Scholar] [CrossRef]
  144. Hartman, S.J.F.; Upadhyay, P.J.; Hagedoorn, N.N.; Mathôt, R.A.A.; Moll, H.A.; van der Flier, M.; Schreuder, M.F.; Brüggemann, R.J.; Knibbe, C.A.; de Wildt, S.N. Current Ceftriaxone Dose Recommendations are Adequate for Most Critically Ill Children: Results of a Population Pharmacokinetic Modeling and Simulation Study. Clin. Pharmacokinet. 2021, 60, 1361–1372. [Google Scholar] [CrossRef] [PubMed]
  145. Schleibinger, M.; Steinbach, C.L.; Töpper, C.; Kratzer, A.; Liebchen, U.; Kees, F.; Salzberger, B.; Kees, M.G. Protein binding characteristics and pharmacokinetics of ceftriaxone in intensive care unit patients. Br. J. Clin. Pharmacol. 2015, 80, 525–533. [Google Scholar] [CrossRef] [Green Version]
  146. Majumdar, A.K.; Musson, D.G.; Birk, K.L.; Kitchen, C.J.; Holland, S.; McCrea, J.; Mistry, G.; Hesney, M.; Xi, L.; Li, S.X.; et al. Pharmacokinetics of Ertapenem in Healthy Young Volunteers. Antimicrob. Agents Chemother. 2002, 46, 3506–3511. [Google Scholar] [CrossRef] [Green Version]
  147. Burkhardt, O.; Kumar, V.; Katterwe, D.; Majcher-Peszynska, J.; Drewelow, B.; Derendorf, H.; Welte, T. Ertapenem in critically ill patients with early-onset ventilator-associated pneumonia: Pharmacokinetics with special consideration of free-drug concentration. J. Antimicrob. Chemother. 2007, 59, 277–284. [Google Scholar] [CrossRef] [PubMed]
  148. Musson, D.G.; Majumdar, A.; Holland, S.; Birk, K.; Xi, L.; Mistry, G.; Sciberras, D.; Muckow, J.; Deutsch, P.; Rogers, J.D. Pharmacokinetics of total and unbound ertapenem in healthy elderly subjects. Antimicrob. Agents Chemother. 2004, 48, 521–524. [Google Scholar] [CrossRef] [Green Version]
  149. DrugBank Online. Teicoplanin. Available online: https://go.drugbank.com/drugs/DB06149 (accessed on 1 February 2023).
  150. Sando, M.; Sato, Y.; Iwata, S.; Akita, H.; Sunakawa, K. In vitro protein binding of teicoplanin to neonatal serum. J. Infect. Chemother. 2004, 10, 280–283. [Google Scholar] [CrossRef]
  151. Smits, A.; Pauwels, S.; Oyaert, M.; Peersman, N.; Spriet, I.; Saegeman, V.; Allegaert, K. Factors impacting unbound vancomycin concentrations in neonates and young infants. Eur. J. Clin. Microbiol. Infect. Dis. 2018, 37, 1503–1510. [Google Scholar] [CrossRef] [PubMed]
  152. Turner, N.A.; Xu, A.; Zaharoff, S.; Holland, T.L.; Lodise, T.P. Determination of plasma protein binding of dalbavancin. J. Antimicrob. Chemother. 2022, 77, 1899–1902. [Google Scholar] [CrossRef] [PubMed]
  153. DrugBank Online. Telavancin. Available online: https://go.drugbank.com/drugs/DB06402 (accessed on 1 February 2023).
  154. DrugBank Online. Oritavancin. Available online: https://go.drugbank.com/drugs/DB04911 (accessed on 1 February 2023).
  155. Corona, A.; Agarossi, A.; Veronese, A.; Cattaneo, D.; D’Avolio, A. Therapeutic Drug Monitoring of Dalbavancin Treatment in Severe Necrotizing Fasciitis in 3 Critically Ill Patients: A Grand Round. Ther. Drug Monit. 2020, 42, 165–168. [Google Scholar] [CrossRef]
  156. DrugBank Online. Erythromycin. Available online: https://go.drugbank.com/drugs/DB00199 (accessed on 1 February 2023).
  157. DrugBank Online. Clarithromycin. Available online: https://go.drugbank.com/drugs/DB01211 (accessed on 1 February 2023).
  158. Viscardi, R.M.; Othman, A.A.; Hassan, H.E.; Eddington, N.D.; Abebe, E.; Terrin, M.L.; Kaufman, D.A.; Waites, K.B. Azithromycin to prevent bronchopulmonary dysplasia in ureaplasma-infected preterm infants: Pharmacokinetics, safety, microbial response, and clinical outcomes with a 20-milligram-per-kilogram single intravenous dose. Antimicrob. Agents Chemother. 2013, 57, 2127–2133. [Google Scholar] [CrossRef] [Green Version]
  159. Hassan, H.E.; Othman, A.A.; Eddington, N.D.; Duffy, L.; Xiao, L.; Waites, K.B.; Kaufman, D.A.; Fairchild, K.D.; Terrin, M.L.; Viscardi, R.M. Pharmacokinetics, safety, and biologic effects of azithromycin in extremely preterm infants at risk for ureaplasma colonization and bronchopulmonary dysplasia. J. Clin. Pharmacol. 2011, 51, 1264–1275. [Google Scholar] [CrossRef] [Green Version]
  160. Kratzer, A.; Kees, F.; Dorn, C. Unbound fraction of fluconazole and linezolid in human plasma as determined by ultrafiltration: Impact of membrane type. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2016, 1039, 74–78. [Google Scholar] [CrossRef]
  161. Ogami, C.; Tsuji, Y.; To, H.; Yamamoto, Y. Pharmacokinetics, toxicity and clinical efficacy of linezolid in Japanese pediatric patients. J. Infect. Chemother. 2019, 25, 979–986. [Google Scholar] [CrossRef]
  162. DrugBank Online. Tedizolid. Available online: https://go.drugbank.com/drugs/DB14569 (accessed on 1 February 2023).
  163. Iqbal, K.; Milioudi, A.; Wicha, S.G. Pharmacokinetics and Pharmacodynamics of Tedizolid. Clin. Pharmacokinet. 2022, 61, 489–503. [Google Scholar] [CrossRef] [PubMed]
  164. Gonzalez, D.; Delmore, P.; Bloom, B.T.; Cotten, C.M.; Poindexter, B.B.; McGowan, E.; Shattuck, K.; Bradford, K.K.; Smith, P.B.; Cohen-Wolkowiez, M.; et al. Clindamycin Pharmacokinetics and Safety in Preterm and Term Infants. Antimicrob. Agents Chemother. 2016, 60, 2888–2894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. DrugBank Online. Lincomycin. Available online: https://go.drugbank.com/drugs/DB01627 (accessed on 1 February 2023).
  166. Schneider, E.K.; Huang, J.X.; Carbone, V.; Han, M.; Zhu, Y.; Nang, S.; Khoo, K.K.; Mak, J.; Cooper, M.A.; Li, J.; et al. Plasma Protein Binding Structure–Activity Relationships Related to the N-Terminus of Daptomycin. ACS Infect. Dis. 2017, 3, 249–258. [Google Scholar] [CrossRef] [PubMed]
  167. DrugBank Online. Daptomycin. Available online: https://go.drugbank.com/drugs/DB00080 (accessed on 1 February 2023).
  168. Ye, L.; You, X.; Zhou, J.; Wu, C.; Ke, M.; Wu, W.; Huang, P.; Lin, C. Physiologically based pharmacokinetic modeling of daptomycin dose optimization in pediatric patients with renal impairment. Front. Pharmacol. 2022, 13, 838599. [Google Scholar] [CrossRef]
  169. DrugBank Online. Levofloxacin. Available online: https://go.drugbank.com/drugs/DB01137 (accessed on 1 February 2023).
  170. Mitsuboshi, S.; Yamada, H.; Nagai, K.; Ueno, K. No effect of protein binding ratio of levofloxacin in hemodialysis patients. Ren. Replace. Ther. 2016, 2, 28. [Google Scholar] [CrossRef] [Green Version]
  171. DrugBank Online. Moxifloxacin. Available online: https://go.drugbank.com/drugs/DB00218 (accessed on 1 February 2023).
  172. Obiero, C.W.; Williams, P.; Murunga, S.; Thitiri, J.; Omollo, R.; Walker, A.S.; Egondi, T.; Nyaoke, B.; Correia, E.; Kane, Z.; et al. Randomised controlled trial of fosfomycin in neonatal sepsis: Pharmacokinetics and safety in relation to sodium overload. Arch. Dis. Child. 2022, 107, 802–810. [Google Scholar] [CrossRef] [PubMed]
  173. Verbeeck, R.K.; Singu, B.S.; Kibuule, D. Clinical Significance of the Plasma Protein Binding of Rifampicin in the Treatment of Tuberculosis Patients. Clin. Pharmacokinet. 2019, 58, 1511–1515. [Google Scholar] [CrossRef]
  174. Litjens, C.H.C.; Aarnoutse, R.E.; Kolmer, E.W.J.V.E.-B.; Svensson, E.M.; Colbers, A.; Burger, D.M.; Boeree, M.J.; Brake, L.H.M.T.; Heinrich, N.; Diacon, A.; et al. Protein binding of rifampicin is not saturated when using high-dose rifampicin. J. Antimicrob. Chemother. 2019, 74, 986–990. [Google Scholar] [CrossRef]
  175. Amin, S.B. Bilirubin Binding Capacity in the Preterm Neonate. Clin. Perinatol. 2016, 43, 241–257. [Google Scholar] [CrossRef] [Green Version]
  176. DrugBank Online. Sulfamethoxazole. Available online: https://go.drugbank.com/drugs/DB01015 (accessed on 1 February 2023).
  177. DrugBank Online. Trimethoprim. Available online: https://go.drugbank.com/drugs/DB00440 (accessed on 1 February 2023).
  178. Thyagarajan, B.; Deshpande, S.S. Cotrimoxazole and neonatal kernicterus: A review. Drug Chem. Toxicol. 2014, 37, 121–129. [Google Scholar] [CrossRef] [Green Version]
  179. Hile, G.B.; Musick, K.L.; Dugan, A.J.; Bailey, A.M.; Howington, G.T. Occurrence of Hyperbilirubinemia in Neonates Given a Short-term Course of Ceftriaxone versus Cefotaxime for Sepsis. J. Pediatr. Pharmacol. Ther. 2021, 26, 99–103. [Google Scholar] [CrossRef] [PubMed]
  180. Amin, S.B. Bilirubin-Displacing Effect of Ceftriaxone in Infants with Unconjugated Hyperbilirubinemia Born at Term. J. Pediatr. 2022, 254, 91–95. [Google Scholar] [CrossRef]
  181. Jubeh, B.; Breijyeh, Z.; Karaman, R. Antibacterial Prodrugs to Overcome Bacterial Resistance. Molecules 2020, 25, 1543. [Google Scholar] [CrossRef] [Green Version]
  182. Frampton, J.E. Ceftaroline Fosamil: A Review of its Use in the Treatment of Complicated Skin and Soft Tissue Infections and Community-Acquired Pneumonia. Drugs 2013, 73, 1067–1094. [Google Scholar] [CrossRef] [PubMed]
  183. Eichenbaum, G.; Skibbe, J.; Parkinson, A.; Johnson, M.D.; Baumgardner, D.; Ogilvie, B.; Usuki, E.; Tonelli, F.; Holsapple, J.; Schmitt-Hoffmann, A. Use of enzyme inhibitors to evaluate the conversion pathways of ester and amide prodrugs: A case study example with the prodrug ceftobiprole medocaril. J. Pharm. Sci. 2012, 101, 1242–1252. [Google Scholar] [CrossRef] [PubMed]
  184. McEntee, L.; Johnson, A.; Farrington, N.; Unsworth, J.; Dane, A.; Jain, A.; Cotroneo, N.; Critchley, I.; Melnick, D.; Parr, T.; et al. Pharmacodynamics of Tebipenem: New Options for Oral Treatment of Multidrug-Resistant Gram-Negative Infections. Antimicrob. Agents Chemother. 2019, 63, e00603. [Google Scholar] [CrossRef] [Green Version]
  185. Dingsdag, S.A.; Hunter, N. Metronidazole: An update on metabolism, structure–cytotoxicity and resistance mechanisms. J. Antimicrob. Chemother. 2018, 73, 265–279. [Google Scholar] [CrossRef] [Green Version]
  186. Obach, R.S. Linezolid Metabolism Is Catalyzed by Cytochrome P450 2J2, 4F2, and 1B1. Drug Metab. Dispos. 2022, 50, 413–421. [Google Scholar] [CrossRef]
  187. Fanni, D.; Ambu, R.; Gerosa, C.; Nemolato, S.; Castagnola, M.; Van Eyken, P.; Faa, G.; Fanos, V. Cytochrome P450 genetic polymorphism in neonatal drug metabolism: Role and practical consequences towards a new drug culture in neonatology. Int. J. Immunopathol. Pharmacol. 2014, 27, 5–13. [Google Scholar] [CrossRef]
  188. Ruggiero, A.; Ariano, A.; Triarico, S.; Capozza, M.A.; Ferrara, P.; Attinà, G. Neonatal pharmacology and clinical implications. Drugs Context 2019, 8, 212608. [Google Scholar] [CrossRef] [Green Version]
  189. Allegaert, K.; Anker, J.D. Ontogeny of Phase I Metabolism of Drugs. J. Clin. Pharmacol. 2019, 59 (Suppl. S1), S33–S41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. 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. [Google Scholar] [CrossRef] [PubMed]
  191. 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. [Google Scholar] [CrossRef] [PubMed]
  192. Hines, R. Developmental expression of drug metabolizing enzymes: Impact on disposition in neonates and young children. Int. J. Pharm. 2013, 452, 3–7. [Google Scholar] [CrossRef]
  193. Ku, L.C.; Smith, P.B. Dosing in neonates: Special considerations in physiology and trial design. Pediatr. Res. 2014, 77, 2–9. [Google Scholar] [CrossRef] [Green Version]
  194. O’Hara, K. Pharmacokinetic changes with growth and development between birth and adulthood. J. Pharm. Pract. Res. 2017, 47, 313–318. [Google Scholar] [CrossRef]
  195. Song, G.; Sun, X.; Hines, R.N.; McCarver, D.G.; Lake, B.G.; Osimitz, T.G.; Creek, M.R.; Clewell, H.J.; Yoon, M. Determination of Human Hepatic CYP2C8 and CYP1A2 Age-Dependent Expression to Support Human Health Risk Assessment for Early Ages. Drug Metab. Dispos. 2017, 45, 468–475. [Google Scholar] [CrossRef] [Green Version]
  196. van Groen, B.D.; Nicolaï, J.; Kuik, A.C.; Van Cruchten, S.; van Peer, E.; Smits, A.; Schmidt, S.; de Wildt, S.N.; Allegaert, K.; De Schaepdrijver, L.; et al. Ontogeny of Hepatic Transporters and Drug-Metabolizing Enzymes in Humans and in Nonclinical Species. Pharmacol. Rev. 2021, 73, 597–678. [Google Scholar] [CrossRef]
  197. Choudhary, D.; Jansson, I.; Schenkman, J.; Sarfarazi, M.; Stoilov, I. Comparative expression profiling of 40 mouse cytochrome P450 genes in embryonic and adult tissues. Arch. Biochem. Biophys. 2003, 414, 91–100. [Google Scholar] [CrossRef]
  198. van den Anker, J.; Reed, M.D.; Allegaert, K.; Kearns, G.L. Developmental Changes in Pharmacokinetics and Pharmacodynamics. J. Clin. Pharmacol. 2018, 58 (Suppl. S10), S10–S25. [Google Scholar] [CrossRef] [Green Version]
  199. Koukouritaki, S.B.; Manro, J.R.; Marsh, S.A.; Stevens, J.C.; Rettie, A.E.; McCarver, D.G.; Hines, R. Developmental expression of human hepatic CYP2C9 and CYP2C19. Experiment 2003, 308, 965–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Stevens, J.C.; Marsh, S.A.; Zaya, M.J.; Regina, K.J.; Divakaran, K.; Le, M.; Hines, R.N. Developmental changes in human liver CYP2D6 expression. Drug Metab. Dispos. 2008, 36, 1587–1593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. 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. [Google Scholar] [CrossRef]
  202. Wynalda, M.A.; Hutzler, J.M.; Koets, M.D.; Podoll, T.; Wienkers, L.C. In Vitro Metabolism of Clindamycin in Human Liver and Intestinal Microsomes. Drug Metab. Dispos. 2003, 31, 878–887. [Google Scholar] [CrossRef] [PubMed]
  203. 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. [Google Scholar] [CrossRef] [PubMed]
  204. Williams, J.A.; Ring, B.J.; Cantrell, V.E.; Jones, D.R.; Eckstein, J.; Ruterbories, K.; Hamman, M.A.; Hall, S.D.; Wrighton, S.A. Comparative metabolic capabilities of CYP3A4, CYP3A5, and CYP3A7. Drug Metab. Dispos. 2002, 30, 883–891. [Google Scholar] [CrossRef]
  205. Zhang, L.; Xu, X.; Badawy, S.; Ihsan, A.; Liu, Z.; Xie, C.; Wang, X.; Tao, Y. A Review: Effects of Macrolides on CYP450 Enzymes. Curr. Drug Metab. 2020, 21, 928–937. [Google Scholar] [CrossRef]
  206. Srinivas, N.R. Pharmacokinetic Interaction of Rifampicin with Oral versus Intravenous Anticancer Drugs: Challenges, Dilemmas and Paradoxical Effects Due to Multiple Mechanisms. Drugs R D 2016, 16, 141–148. [Google Scholar] [CrossRef] [Green Version]
  207. Derungs, A.; Donzelli, M.; Berger, B.; Noppen, C.; Krähenbühl, S.; Haschke, M. Effects of Cytochrome P450 Inhibition and Induction on the Phenotyping Metrics of the Basel Cocktail: A Randomized Crossover Study. Clin. Pharmacokinet. 2016, 55, 79–91. [Google Scholar] [CrossRef] [Green Version]
  208. Zhang, L.; Wei, M.-J.; Zhao, C.-Y.; Qi, H.-M. Determination of the inhibitory potential of 6 fluoroquinolones on CYP1A2 and CYP2C9 in human liver microsomes. Acta Pharmacol. Sin. 2008, 29, 1507–1514. [Google Scholar] [CrossRef]
  209. Watanabe, A.; Takakusa, H.; Kimura, T.; Inoue, S.-I.; Kusuhara, H.; Ando, O. Analysis of Mechanism-Based Inhibition of CYP 3A4 by a Series of Fluoroquinolone Antibacterial Agents. Drug Metab. Dispos. 2016, 44, 1608–1616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Watanabe, A.; Takakusa, H.; Kimura, T.; Inoue, S.-I.; Kusuhara, H.; Ando, O. Difference in Mechanism-Based Inhibition of Cytochrome P450 3A4 and 3A5 by a Series of Fluoroquinolone Antibacterial Agents. Drug Metab. Dispos. 2017, 45, 336–341. [Google Scholar] [CrossRef] [Green Version]
  211. Niwa, T.; Morimoto, M.; Hirai, T.; Hata, T.; Hayashi, M.; Imagawa, Y. Effect of penicillin-based antibiotics, amoxicillin, ampicillin, and piperacillin, on drug-metabolizing activities of human hepatic cytochromes P450. J. Toxicol. Sci. 2016, 41, 143–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Huwyler, J.; Wright, M.; Gutmann, H.; Drewe, J. Induction of Cytochrome P450 3A4 and P-Glycoprotein by the Isoxazolyl- Penicillin Antibiotic Flucloxacillin. Curr. Drug Metab. 2006, 7, 119–126. [Google Scholar] [CrossRef] [PubMed]
  213. Mahmoudi, M.; Brenner, T.; Hatiboglu, G.; Burhenne, J.; Weiss, J.; Weigand, M.A.; Haefeli, W.E. Substantial Impairment of Voriconazole Clearance by High-Dose Meropenem in a Patient with Renal Failure. Clin. Infect. Dis. 2017, 65, 1033–1036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Cada, D.J.; Baker, D.E. Oritavancin Diphosphate. Hosp. Pharm. 2014, 49, 1049–1060. [Google Scholar] [CrossRef] [Green Version]
  215. Cattaneo, D.; Gervasoni, C.; Corona, A. The Issue of Pharmacokinetic-Driven Drug-Drug Interactions of Antibiotics: A Narrative Review. Antibiotics 2022, 11, 1410. [Google Scholar] [CrossRef]
  216. Wen, X.; Wang, J.-S.; Backman, J.T.; Laitila, J.; Neuvonen, P.J. Trimethoprim and sulfamethoxazole are selective inhibitors of CYP2C8 and CYP2C9, respectively. Drug Metab. Dispos. 2002, 30, 631–635. [Google Scholar] [CrossRef] [Green Version]
  217. Türk, D.; Hanke, N.; Lehr, T. A Physiologically-Based Pharmacokinetic Model of Trimethoprim for MATE1, OCT1, OCT2, and CYP2C8 Drug–Drug–Gene Interaction Predictions. Pharmaceutics 2020, 12, 1074. [Google Scholar] [CrossRef]
  218. Li, H.; Lampe, J.N. Neonatal cytochrome P450 CYP3A7: A comprehensive review of its role in development, disease, and xenobiotic metabolism. Arch. Biochem. Biophys. 2019, 673, 108078. [Google Scholar] [CrossRef]
  219. Usui, T.; Saitoh, Y.; Komada, F. Induction of CYP3As in HepG2 cells by several drugs. Association between induction of CYP3A4 and expression of glucocorticoid receptor. Biol. Pharm. Bull. 2003, 26, 510–517. [Google Scholar] [CrossRef] [Green Version]
  220. Boberg, M.; Vrana, M.; Mehrotra, A.; Pearce, R.E.; Gaedigk, A.; Bhatt, D.K.; Leeder, J.S.; Prasad, B. Age-Dependent Absolute Abundance of Hepatic Carboxylesterases (CES1 and CES2) by LC-MS/MS Proteomics: Application to PBPK Modeling of Oseltamivir In Vivo Pharmacokinetics in Infants. Drug Metab. Dispos. 2017, 45, 216–223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Xu, M.; Bhatt, D.K.; Yeung, C.K.; Claw, K.G.; Chaudhry, A.S.; Gaedigk, A.; Pearce, R.E.; Broeckel, U.; Gaedigk, R.; Nickerson, D.A.; et al. Genetic and Nongenetic Factors Associated with Protein Abundance of Flavin-Containing Monooxygenase 3 in Human Liver. J. Pharmacol. Exp. Ther. 2017, 363, 265–274. [Google Scholar] [CrossRef] [Green Version]
  223. Bhatt, D.K.; Gaedigk, A.; Pearce, R.E.; Leeder, J.S.; Prasad, B. Age-dependent Protein Abundance of Cytosolic Alcohol and Aldehyde Dehydrogenases in Human Liver. Drug Metab. Dispos. 2017, 45, 1044–1048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Pacifici, G.M.; Franchi, M.; Giuliani, L.; Rane, A. Development of the glucuronyltransferase and sulphotransferase towards 2-naphthol in human fetus. Dev. Pharmacol. Ther. 1989, 14, 108–114. [Google Scholar] [CrossRef]
  225. Ladumor, M.K.; Bhatt, D.K.; Gaedigk, A.; Sharma, S.; Thakur, A.; Pearce, R.E.; Leeder, J.S.; Bolger, M.B.; Singh, S.; Prasad, B. Ontogeny of Hepatic Sulfotransferases and Prediction of Age-Dependent Fractional Contribution of Sulfation in Acetaminophen Metabolism. Drug Metab. Dispos. 2019, 47, 818–831. [Google Scholar] [CrossRef]
  226. Miyagi, S.J.; Collier, A.C. The development of UDP-glucuronosyltransferases 1A1 and 1A6 in the pediatric liver. Drug Metab. Dispos. 2011, 39, 912–919. [Google Scholar] [CrossRef] [Green Version]
  227. Strange, R.; Howie, A.; Hume, R.; Matharoo, B.; Bell, J.; Hiley, C.; Jones, P.; Beckett, G. The developmental expression of alpha-, mu- and pi-class glutathione S-transferases in human liver. Biochim. Biophys. Acta 1989, 993, 186–190. [Google Scholar] [CrossRef]
  228. Stojanović, V.D.; Barišić, N.A.; Radovanović, T.D.; Kovač, N.B.; Djuran, J.D.; Antić, A.P.E.; Doronjski, A.D. Serum glutathione S-transferase Pi as predictor of the outcome and acute kidney injury in premature newborns. Pediatr. Nephrol. 2018, 33, 1251–1256. [Google Scholar] [CrossRef]
  229. Rogers, Z.; Hiruy, H.; Pasipanodya, J.G.; Mbowane, C.; Adamson, J.; Ngotho, L.; Karim, F.; Jeena, P.; Bishai, W.; Gumbo, T. The Non-Linear Child: Ontogeny, Isoniazid Concentration, and NAT2 Genotype Modulate Enzyme Reaction Kinetics and Metabolism. EBioMedicine 2016, 11, 118–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. 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. [Google Scholar] [CrossRef] [PubMed]
  231. Johnson, T.N.; Jamei, M.; Rowland-Yeo, K. How Does In Vivo Biliary Elimination of Drugs Change with Age? Evidence from In Vitro and Clinical Data Using a Systems Pharmacology Approach. Drug Metab. Dispos. 2016, 44, 1090–1098. [Google Scholar] [CrossRef] [PubMed]
  232. Lumbers, E.R.; Kandasamy, Y.; Delforce, S.J.; Boyce, A.C.; Gibson, K.J.; Pringle, K. Programming of Renal Development and Chronic Disease in Adult Life. Front. Physiol. 2020, 11, 757. [Google Scholar] [CrossRef] [PubMed]
  233. Filler, G.; Bhayana, V.; Schott, C.; de Ferris, M.E.D. How should we assess renal function in neonates and infants? Acta Paediatr. 2021, 110, 773–780. [Google Scholar] [CrossRef]
  234. Kandasamy, Y.; Rudd, D.; Smith, R.; Lumbers, E.R.; Wright, I. Extra uterine development of preterm kidneys. Pediatr. Nephrol. 2018, 33, 1007–1012. [Google Scholar] [CrossRef] [Green Version]
  235. Kandasamy, Y.; Rudd, D.; Lumbers, E.R.; Smith, R. An evaluation of preterm kidney size and function over the first two years of life. Pediatr. Nephrol. 2020, 35, 1477–1482. [Google Scholar] [CrossRef]
  236. Smeets, N.J.; IntHout, J.; van der Burgh, M.J.; Schwartz, G.J.; Schreuder, M.F.; de Wildt, S.N. Maturation of GFR in Term-Born Neonates: An Individual Participant Data Meta-Analysis. J. Am. Soc. Nephrol. 2022, 33, 1277–1292. [Google Scholar] [CrossRef]
  237. Salem, F.; Johnson, T.N.; Hodgkinson, A.B.J.; Ogungbenro, K.; Rostami-Hodjegan, A. Does “Birth” as an Event Impact Maturation Trajectory of Renal Clearance via Glomerular Filtration? Reexamining Data in Preterm and Full-Term Neonates by Avoiding the Creatinine Bias. J. Clin. Pharmacol. 2021, 61, 159–171. [Google Scholar] [CrossRef]
  238. Correa, L.P.; Marzano, A.C.S.; Filha, R.S.; Magalhães, R.C.; Simoes-E-Silva, A.C. Biomarkers of renal function in preterm neonates at 72 h and 3 weeks of life. J. Pediatr. 2021, 97, 508–513. [Google Scholar] [CrossRef]
  239. Iacobelli, S.; Guignard, J.-P. Maturation of glomerular filtration rate in neonates and infants: An overview. Pediatr. Nephrol. 2021, 36, 1439–1446. [Google Scholar] [CrossRef]
  240. Coleman, C.; Perez, A.T.; Selewski, D.T.; Steflik, H.J. Neonatal Acute Kidney Injury. Front. Pediatr. 2022, 10, 842544. [Google Scholar] [CrossRef]
  241. Rhone, E.T.; Carmody, J.B.; Swanson, J.R.; Charlton, J.R. Nephrotoxic medication exposure in very low birth weight infants. J. Matern Fetal Neonatal Med. 2014, 27, 1485–1490. [Google Scholar] [CrossRef]
  242. Jančič, S.G.; Močnik, M.; Varda, N.M. Glomerular Filtration Rate Assessment in Children. Children 2022, 9, 1995. [Google Scholar] [CrossRef] [PubMed]
  243. Wang, J.; Kumar, S.S.; Sherwin, C.; Ward, R.; Baer, G.; Burckart, G.J.; Wang, Y.; Yao, L.P. Renal Clearance in Newborns and Infants: Predictive Performance of Population-Based Modeling for Drug Development. Clin. Pharmacol. Ther. 2019, 105, 1462–1470. [Google Scholar] [CrossRef] [PubMed]
  244. Penicillin, G. DrugBank Online. Available online: https://go.drugbank.com/drugs/DB01053 (accessed on 1 February 2023).
  245. Padari, H.; Metsvaht, T.; Germovsek, E.; Barker, C.I.; Kipper, K.; Herodes, K.; Standing, J.F.; Oselin, K.; Tasa, T.; Soeorg, H.; et al. Pharmacokinetics of Penicillin G in Preterm and Term Neonates. Antimicrob. Agents Chemother. 2018, 62, e02238-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Peechakara, B.V.; Gupta, M. Ampicillin/Sulbactam. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK526117 (accessed on 1 February 2023).
  247. Le, J.; Greenberg, R.G.; Yoo, Y.; Clark, R.H.; Benjamin, D.K.; Zimmerman, K.O.; Cohen-Wolkowiez, M.; Wade, K.C. Best Pharmaceuticals for Children Act–Pediatric Trials Network Steering Committee Ampicillin dosing in premature infants for early-onset sepsis: Exposure-driven efficacy, safety, and stewardship. J. Perinatol. 2022, 42, 959–964. [Google Scholar] [CrossRef]
  248. Stępnik, K.E.; Malinowska, I. Determination of binding properties of ampicillin in drug-human serum albumin standard solution using N-vinylpyrrolidone copolymer combined with the micellar systems. Talanta 2017, 162, 241–248. [Google Scholar] [CrossRef]
  249. Soto, E.; Shoji, S.; Muto, C.; Tomono, Y.; Marshall, S. Population pharmacokinetics of ampicillin and sulbactam in patients with community-acquired pneumonia: Evaluation of the impact of renal impairment. Br. J. Clin. Pharmacol. 2014, 77, 509–521. [Google Scholar] [CrossRef] [Green Version]
  250. DrugBank Online. Amoxicillin. Available online: https://go.drugbank.com/drugs/DB01060 (accessed on 1 February 2023).
  251. Pullen, J.; Stolk, L.M.; Nieman, F.H.; Degraeuwe, P.L.; van Tiel, F.H.; Zimmermann, L.J. Population pharmacokinetics and dosing of amoxicillin in (pre)term neonates. Ther. Drug Monit. 2006, 28, 226–231. [Google Scholar] [CrossRef]
  252. CharlesX, B.G.; Preechagoon, Y.; Lee, T.C.; Steer, P.A.; Flenady, V.J.; Debuse, N. Population pharmacokinetics of intravenous amoxicillin in very low birth weight infants. J. Pharm. Sci. 1997, 86, 1288–1292. [Google Scholar] [CrossRef]
  253. Landersdorfer, C.; Kirkpatrick, C.; Kinzig-Schippers, M.; Bulitta, J.B.; Holzgrabe, U.; Drusano, G.L.; Sörgel, F. Population pharmacokinetics at two dose levels and pharmacodynamic profiling of flucloxacillin. Antimicrob. Agents Chemother. 2007, 51, 3290–3297. [Google Scholar] [CrossRef] [Green Version]
  254. Pullen, J.; de Rozario, L.; Stolk, L.M.L.; Degraeuwe, P.L.J.; van Tiel, F.H.; Zimmermann, L.J.I. Population pharmacokinetics and dosing of flucloxacillin in preterm and term neonates. Ther. Drug Monit. 2006, 28, 351–358. [Google Scholar] [CrossRef] [PubMed]
  255. Anderson, P.; Bluhm, G.; Ehrnebo, M.; Herngren, L.; Jacobson, B. Pharmacokinetics and distribution of flucloxacillin in pacemaker patients. Eur. J. Clin. Pharmacol. 1985, 27, 713–719. [Google Scholar] [CrossRef]
  256. Oh, K.-H.; Kim, C.; Lee, H.; Lee, H.; Jung, J.Y.; Kim, N.J.; Yu, K.-S.; Shin, K.-H.; Jang, I.-J.; Ahn, C. Pharmacokinetics of intravenous piperacillin administration in patients undergoing on-line hemodiafiltration. Antimicrob. Agents Chemother. 2009, 53, 3266–3268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Cohen-Wolkowiez, M.; Watt, K.M.; Zhou, C.; Bloom, B.T.; Poindexter, B.; Castro, L.; Gao, J.; Capparelli, E.V.; Benjamin, D.K.; Smith, P.B. Developmental pharmacokinetics of piperacillin and tazobactam using plasma and dried blood spots from infants. Antimicrob. Agents Chemother. 2014, 58, 2856–2865. [Google Scholar] [CrossRef] [Green Version]
  258. Libke, R.D.; Clarke, J.T.; Ralph, E.D.; Luthy, R.P.; Kirby, W.M.M. Ticarcillin vs carbenicillin: Clinical pharmacokinetics. Clin. Pharmacol. Ther. 1975, 17, 441–446. [Google Scholar] [CrossRef]
  259. Watt, K.M.; Hornik, C.P.; Balevic, S.J.; Mundakel, G.; Cotten, C.M.; Harper, B.; Benjamin, D.K.; Anand, R.; Laughon, M.; Smith, P.B.; et al. Pharmacokinetics of ticarcillin–clavulanate in premature infants. Br. J. Clin. Pharmacol. 2019, 85, 1021–1027. [Google Scholar] [CrossRef]
  260. De Groot, R.; Hack, B.D.; Weber, A.; Chaffin, D.; Ramsey, B.; Smith, A.L. Pharmacokinetics of ticarcillin in patients with cystic fibrosis: A controlled prospective study. Clin. Pharmacol. Ther. 1990, 47, 73–78. [Google Scholar] [CrossRef] [PubMed]
  261. Patel, K.B.; Nicolau, D.P.; Nightingale, C.H.; Quintiliani, R. Pharmacokinetics of cefotaxime in healthy volunteers and patients. Diagn. Microbiol. Infect. Dis. 1995, 22, 49–55. [Google Scholar] [CrossRef]
  262. Shang, Z.-H.; Wu, Y.-E.; Lv, D.-M.; Zhang, W.; Liu, W.-Q.; Anker, J.V.D.; Xu, Y.; Zhao, W. Optimal dose of cefotaxime in neonates with early-onset sepsis: A developmental pharmacokinetic model-based evaluation. Front. Pharmacol. 2022, 13, 916253. [Google Scholar] [CrossRef]
  263. Aardema, H.; Bult, W.; Van Hateren, K.; Dieperink, W.; Touw, D.; Alffenaar, J.-W.C.; Zijlstra, J. Continuous versus intermittent infusion of cefotaxime in critically ill patients: A randomized controlled trial comparing plasma concentrations. J. Antimicrob. Chemother. 2020, 75, 441–448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Pacifici, G.; Marchini, G. Clinical Pharmacology of Cefotaxime in Neonates and Infants: Effects and Pharmacokinetics. Int. J. Pediatr. 2017, 5, 6111–6138. [Google Scholar] [CrossRef]
  265. Pais, G.M.; Chang, J.; Barreto, E.F.; Stitt, G.; Downes, K.J.; Alshaer, M.H.; Lesnicki, E.; Panchal, V.; Bruzzone, M.; Bumanglag, A.V.; et al. Clinical Pharmacokinetics and Pharmacodynamics of Cefepime. Clin. Pharmacokinet. 2022, 61, 929–953. [Google Scholar] [CrossRef]
  266. Zhao, Y.; Yao, B.-F.; Kou, C.; Xu, H.-Y.; Tang, B.-H.; Wu, Y.-E.; Hao, G.-X.; Zhang, X.-P.; Zhao, W. Developmental Population Pharmacokinetics and Dosing Optimization of Cefepime in Neonates and Young Infants. Front. Pharmacol. 2020, 11, 14. [Google Scholar] [CrossRef]
  267. Capparelli, E.; Hochwald, C.; Rasmussen, M.; Parham, A.; Bradley, J.; Moya, F. Population pharmacokinetics of cefepime in the neonate. Antimicrob. Agents Chemother. 2005, 49, 2760–2766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  268. Zhou, J.; Sulaiman, Z.; Llorin, R.M.; Hee, K.-H.; Lee, L.S.-U.; Lye, D.C.; Fisher, D.A.; Tam, V.H. Pharmacokinetics of ertapenem in outpatients with complicated urinary tract infections. J. Antimicrob. Chemother. 2014, 69, 2517–2521. [Google Scholar] [CrossRef] [Green Version]
  269. Pacifici, G.M.; Allegaert, K. Clinical pharmacology of carbapenems in neonates. J. Chemother. 2014, 26, 67–73. [Google Scholar] [CrossRef]
  270. Mouton, J.W.; Mouton, J.W.; Anker, J.N.V.D. Meropenem clinical pharmacokinetics. Clin. Pharmacokinet. 1995, 28, 275–286. [Google Scholar] [CrossRef]
  271. Smith, P.B.; Cohen-Wolkowiez, M.; Castro, L.M.; Poindexter, B.; Bidegain, M.; Weitkamp, J.-H.; Schelonka, R.L.; Ward, R.M.; Wade, K.; Valencia, G.; et al. Population pharmacokinetics of meropenem in plasma and cerebrospinal fluid of infants with suspected or complicated intra-abdominal infections. Pediatr. Infect. Dis. J. 2011, 30, 844–849. [Google Scholar] [CrossRef]
  272. Dao, K.; Fuchs, A.; André, P.; Giannoni, E.; Decosterd, L.A.; Marchetti, O.; Asner, S.A.; Pfister, M.; Widmer, N.; Buclin, T.; et al. Dosing strategies of imipenem in neonates based on pharmacometric modelling and simulation. J. Antimicrob. Chemother. 2022, 77, 457–465. [Google Scholar] [CrossRef]
  273. DrugBank Online. Imipenem. Available online: https://go.drugbank.com/drugs/DB01598 (accessed on 1 February 2023).
  274. Paterson, D.L.; DePestel, D.D. Doripenem. Clin. Infect. Dis. 2009, 49, 291–298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Cirillo, I.; Vaccaro, N.; Castaneda-Ruiz, B.; Redman, R.; Cossey, V.; Bradley, J.S.; Allegaert, K. Open-Label Study to Evaluate the Single-Dose Pharmacokinetics, Safety, and Tolerability of Doripenem in Infants Less than 12 Weeks in Chronological Age. Antimicrob. Agents Chemother. 2015, 59, 4742–4749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  276. DrugBank Online. Azithromycin. Available online: https://go.drugbank.com/drugs/DB00207 (accessed on 1 February 2023).
  277. Merchan, L.M.; Hassan, H.E.; Terrin, M.L.; Waites, K.B.; Kaufman, D.A.; Ambalavanan, N.; Donohue, P.; Dulkerian, S.J.; Schelonka, R.; Magder, L.S.; et al. Pharmacokinetics, Microbial Response, and Pulmonary Outcomes of Multidose Intravenous Azithromycin in Preterm Infants at Risk for Ureaplasma Respiratory Colonization. Antimicrob. Agents Chemother. 2015, 59, 570–578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  278. Bandín-Vilar, E.; García-Quintanilla, L.; Castro-Balado, A.; Zarra-Ferro, I.; González-Barcia, M.; Campos-Toimil, M.; Mangas-Sanjuan, V.; Mondelo-García, C.; Fernández-Ferreiro, A. A Review of Population Pharmacokinetic Analyses of Linezolid. Clin. Pharmacokinet. 2022, 61, 789–817. [Google Scholar] [CrossRef]
  279. Thibault, C.; Kassir, N.; Goyer, I.; Théorêt, Y.; Litalien, C.; Moussa, A.; Ovetchkine, P.; Autmizguine, J. Population Pharmacokinetics of Intravenous Linezolid in Premature Infants. Pediatr. Infect. Dis. J. 2019, 38, 82–88. [Google Scholar] [CrossRef]
  280. Jungbluth, G.L.; Welshman, I.R.; Hopkins, N.K. Linezolid pharmacokinetics in pediatric patients: An overview. Pediatr. Infect. Dis. J. 2003, 22 (Suppl. S9), S153–S157. [Google Scholar] [CrossRef] [PubMed]
  281. Xie, F.; Mantzarlis, K.; Malliotakis, P.; Koulouras, V.; DeGroote, S.; Koulenti, D.; Blot, S.; Boussery, K.; Van Bocxlaer, J.; Colin, P. Pharmacokinetic evaluation of linezolid administered intravenously in obese patients with pneumonia. J. Antimicrob. Chemother. 2019, 74, 667–674. [Google Scholar] [CrossRef] [PubMed]
  282. Gostelow, M.; Gonzalez, D.; Smith, P.B.; Cohen-Wolkowiez, M. Pharmacokinetics and safety of recently approved drugs used to treat methicillin-resistant Staphylococcus aureus infections in infants, children and adults. Expert Rev. Clin. Pharmacol. 2014, 7, 327–340. [Google Scholar] [CrossRef] [Green Version]
  283. Estes, K.S.; Derendorf, H. Comparison of the pharmacokinetic properties of vancomycin, linezolid, tigecyclin, and daptomycin. Eur. J. Med. Res. 2010, 15, 533–543. [Google Scholar] [CrossRef]
  284. Cohen-Wolkowiez, M.; Watt, K.M.; Hornik, C.P.; Benjamin, D.K.; Smith, P.B. Pharmacokinetics and tolerability of single-dose daptomycin in young infants. Pediatr. Infect. Dis. J. 2012, 31, 935–937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Stass, H.; Lettieri, J.; Vanevski, K.M.; Willmann, S.; James, L.P.; Sullivan, J.E.; Arrieta, A.C.; Bradley, J.S. Pharmacokinetics, Safety, and Tolerability of Single-Dose Intravenous Moxifloxacin in Pediatric Patients: Dose Optimization in a Phase 1 Study. J. Clin. Pharmacol. 2019, 59, 654–667. [Google Scholar] [CrossRef]
  286. Yeung, T.; Chung, E.; Chen, J.; Erdman, L.K.; Smiljkovic, M.; Wong, W.; Rolnitsky, A.; Morris, S.K.; El Shahed, A.; Banihani, R.; et al. Therapeutic Drug Monitoring of Moxifloxacin to Guide Treatment of Mycoplasma hominis Meningitis in an Extremely Preterm Infant. J. Pediatr. Pharmacol. Ther. 2021, 26, 857–862. [Google Scholar] [CrossRef] [PubMed]
  287. DrugBank Online. Amikacin. Available online: https://go.drugbank.com/drugs/DB00479 (accessed on 1 February 2023).
  288. An, S.H.; Kim, J.Y.; Gwak, H.S. Outcomes of a new dosage regimen of amikacin based on pharmacokinetic parameters of Korean neonates. Am. J. Health Syst. Pharm. 2014, 71, 122–127. [Google Scholar] [CrossRef]
  289. Gonzalez, D.; Melloni, C.; Yogev, R.; Poindexter, B.B.; Mendley, S.R.; Delmore, P.; Sullivan, J.E.; Autmizguine, J.; Lewandowski, A.; Harper, B.; et al. Use of opportunistic clinical data and a population pharmacokinetic model to support dosing of clindamycin for premature infants to adolescents. Clin. Pharmacol. Ther. 2014, 96, 429–437. [Google Scholar] [CrossRef] [Green Version]
  290. Rowland, M. Clinical Pharmacokinetics of Teicoplanin. Clin. Pharmacokinet. 1990, 18, 184–209. [Google Scholar] [CrossRef] [PubMed]
  291. Kontou, A.; Sarafidis, K.; Begou, O.; Gika, H.G.; Tsiligiannis, A.; Ogungbenro, K.; Dokoumetzidis, A.; Agakidou, E.; Roilides, E. Population Pharmacokinetics of Teicoplanin in Preterm and Term Neonates: Is It Time for a New Dosing Regimen? Antimicrob. Agents Chemother. 2020, 64, e01971-19. [Google Scholar] [CrossRef] [PubMed]
  292. Zaric, R.Z.; Milovanovic, J.; Rosic, N.; Milovanovic, D.; Zecevic, D.R.; Folic, M.; Jankovic, S. Pharmacokinetics of Vancomycin in Patients with Different Renal Function Levels. Open Med. 2018, 13, 512–519. [Google Scholar] [CrossRef]
  293. Mulubwa, M.; Griesel, H.A.; Mugabo, P.; Dippenaar, R.; Van Wyk, L. Assessment of Vancomycin Pharmacokinetics and Dose Regimen Optimisation in Preterm Neonates. Drugs R D 2020, 20, 105–113. [Google Scholar] [CrossRef] [Green Version]
  294. Leroux, S.; Anker, J.N.V.D.; Smits, A.; Pfister, M.; Allegaert, K. Maturational changes in vancomycin protein binding affect vancomycin dosing in neonates. Br. J. Clin. Pharmacol. 2019, 85, 865–867. [Google Scholar] [CrossRef]
  295. Leroux, S.; Jacqz-Aigrain, E.; Biran, V.; Lopez, E.; Madeleneau, D.; Wallon, C.; Zana-Taïeb, E.; Virlouvet, A.-L.; Rioualen, S.; Zhao, W. Clinical Utility and Safety of a Model-Based Patient-Tailored Dose of Vancomycin in Neonates. Antimicrob. Agents Chemother. 2016, 60, 2039–2042. [Google Scholar] [CrossRef] [Green Version]
  296. Pullen, J.; Stolk, L.M.L.; Degraeuwe, P.L.J.; van Tiel, F.H.; Neef, C.; Zimmermann, L.J.I. Pharmacokinetics of intravenous rifampicin (rifampin) in neonates. Ther. Drug Monit. 2006, 28, 654–661. [Google Scholar] [CrossRef] [PubMed]
  297. Cohen-Wolkowiez, M.; Ouellet, D.; Smith, P.B.; James, L.P.; Ross, A.; Sullivan, J.E.; Walsh, M.C.; Zadell, A.; Newman, N.; White, N.R.; et al. Population Pharmacokinetics of Metronidazole Evaluated Using Scavenged Samples from Preterm Infants. Antimicrob. Agents Chemother. 2012, 56, 1828–1837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  298. DrugBank Online. Metronidazole. Available online: https://go.drugbank.com/drugs/DB00916 (accessed on 4 March 2023).
  299. Li, S.; Xie, F. Foetal and neonatal exposure prediction and dosing evaluation for ampicillin using a physiologically-based pharmacokinetic modelling approach. Br. J. Clin. Pharmacol. 2022, 89, 1402–1412. [Google Scholar] [CrossRef] [PubMed]
  300. Padari, H.; Soeorg, H.; Tasa, T.; Metsvaht, T.; Kipper, K.; Herodes, K.; Oselin, K.; Hallik, M.; Ilmoja, M.-L.; Lutsar, I. Ampicillin Pharmacokinetics During First Week of Life in Preterm and Term Neonates. Pediatr. Infect. Dis. J. 2021, 40, 464–472. [Google Scholar] [CrossRef] [PubMed]
  301. Wu, Y.-E.; Wang, T.; Yang, H.-L.; Tang, B.-H.; Kong, L.; Li, X.; Gao, Q.; Li, X.; Yao, B.-F.; Shi, H.-Y.; et al. Population pharmacokinetics and dosing optimization of azlocillin in neonates with early-onset sepsis: A real-world study. J. Antimicrob. Chemother. 2021, 76, 699–709. [Google Scholar] [CrossRef] [PubMed]
  302. Bijleveld, Y.; Mathôt, R.; van der Lee, J.; Groenendaal, F.; Dijk, P.; van Heijst, A.; Simons, S.; Dijkman, K.; van Straaten, H.; Rijken, M.; et al. Population Pharmacokinetics of Amoxicillin in Term Neonates Undergoing Moderate Hypothermia. Clin. Pharmacol. Ther. 2018, 103, 458–467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  303. Quinton, M.-C.; Bodeau, S.; Kontar, L.; Zerbib, Y.; Maizel, J.; Slama, M.; Masmoudi, K.; Lemaire-Hurtel, A.-S.; Bennis, Y. Neurotoxic Concentration of Piperacillin during Continuous Infusion in Critically Ill Patients. Antimicrob. Agents Chemother. 2017, 61, e00654-17. [Google Scholar] [CrossRef] [Green Version]
  304. Bradley, J.S.; Stone, G.G.; Chan, P.L.S.; Raber, S.R.; Riccobene, T.; Casullo, V.M.; Yan, J.L.; Hendrick, V.M.; Hammond, J.; Leister-Tebbe, H.K. Phase 2 Study of the Safety, Pharmacokinetics and Efficacy of Ceftaroline Fosamil in Neonates and Very Young Infants with Late-onset Sepsis. Pediatr. Infect. Dis. J. 2020, 39, 411–418. [Google Scholar] [CrossRef]
  305. Shi, Z.-R.; Chen, X.-K.; Tian, L.-Y.; Wang, Y.-K.; Zhang, G.-Y.; Dong, L.; Jirasomprasert, T.; Jacqz-Aigrain, E.; Zhao, W. Population Pharmacokinetics and Dosing Optimization of Ceftazidime in Infants. Antimicrob. Agents Chemother. 2018, 62, e02486-17. [Google Scholar] [CrossRef] [Green Version]
  306. Leroux, S.; Roué, J.-M.; Gouyon, J.-B.; Biran, V.; Zheng, H.; Zhao, W.; Jacqz-Aigrain, E. A Population and Developmental Pharmacokinetic Analysis to Evaluate and Optimize Cefotaxime Dosing Regimen in Neonates and Young Infants. Antimicrob. Agents Chemother. 2016, 60, 6626–6634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  307. De Cock, R.F.W.; Smits, A.; Allegaert, K.; De Hoon, J.; Saegeman, V.; Danhof, M.; Knibbe, C.A.J. Population pharmacokinetic modelling of total and unbound cefazolin plasma concentrations as a guide for dosing in preterm and term neonates. J. Antimicrob. Chemother. 2014, 69, 1330–1338. [Google Scholar] [CrossRef] [Green Version]
  308. Ganguly, S.; Edginton, A.N.; Gerhart, J.G.; Cohen-Wolkowiez, M.; Greenberg, R.G.; Gonzalez, D. Best Pharmaceuticals for Children Act-Pediatric Trials Network Steering Committee. Physiologically Based Pharmacokinetic Modeling of Meropenem in Preterm and Term Infants. Clin. Pharmacokinet. 2021, 60, 1591–1604. [Google Scholar] [CrossRef] [PubMed]
  309. Steffens, N.A.; Zimmermann, E.S.; Nichelle, S.M.; Brucker, N. Meropenem use and therapeutic drug monitoring in clinical practice: A literature review. J. Clin. Pharm. Ther. 2021, 46, 610–621. [Google Scholar] [CrossRef] [PubMed]
  310. Mohzari, Y.; Aljobair, F.; Alrashed, A.; Asdaq, S.; Alshuraim, R.; Asfour, S.; Al-Mouqdad, M.; Bamogaddam, R.; Al-Anazi, D.; Zeilinger, C.; et al. Safety and Efficacy of Daptomycin in Neonates with Coagulase-Negative Staphylococci: Case Series Analysis. Antibiotics 2021, 10, 168. [Google Scholar] [CrossRef]
  311. Newby, B.D.; Timberlake, K.E.; Lepp, L.M.; Mihic, T.; Dersch-Mills, D.A. Levofloxacin Use in the Neonate: A Case Series. J. Pediatr. Pharmacol. Ther. 2017, 22, 304–313. [Google Scholar] [CrossRef] [Green Version]
  312. Smits, A.; De Cock, R.F.W.; Allegaert, K.; Vanhaesebrouck, S.; Danhof, M.; Knibbe, C.A.J. Prospective Evaluation of a Model-Based Dosing Regimen for Amikacin in Preterm and Term Neonates in Clinical Practice. Antimicrob. Agents Chemother. 2015, 59, 6344–6351. [Google Scholar] [CrossRef] [Green Version]
  313. Low, B.Y.S.; Tan, M.S.L.; Wan, M.A.S. Extended-Interval gentamicin dosing in achieving therapeutic concentrations in malaysian neonates. J. Pediatr. Pharmacol. Ther. 2015, 20, 119–127. [Google Scholar] [CrossRef] [PubMed]
  314. Neeli, H.; Hanna, N.; Abduljalil, K.; Cusumano, J.; Taft, D.R. Application of Physiologically Based Pharmacokinetic-Pharmacodynamic Modeling in Preterm Neonates to Guide Gentamicin Dosing Decisions and Predict Antibacterial Effect. J. Clin. Pharmacol. 2021, 61, 1356–1365. [Google Scholar] [CrossRef] [PubMed]
  315. Valitalo, P.A.J.; Anker, J.N.V.D.; Allegaert, K.; de Cock, R.F.W.; de Hoog, M.; Simons, S.H.P.; Mouton, J.W.; Knibbe, C.A.J. Novel model-based dosing guidelines for gentamicin and tobramycin in preterm and term neonates. J. Antimicrob. Chemother. 2015, 70, 2074–2077. [Google Scholar] [CrossRef] [Green Version]
  316. Jacqz-Aigrain, E.; Leroux, S.; Thomson, A.H.; Allegaert, K.; Capparelli, E.V.; Biran, V.; Simon, N.; Meibohm, B.; Lo, L.; Marques, R.; et al. Population pharmacokinetic meta-analysis of individual data to design the first randomized efficacy trial of vancomycin in neonates and young infants. J. Antimicrob. Chemother. 2019, 74, 2128–2138. [Google Scholar] [CrossRef] [PubMed]
  317. Janssen, E.J.H.; Välitalo, P.A.J.; Allegaert, K.; de Cock, R.F.W.; Simons, S.H.P.; Sherwin, C.M.T.; Mouton, J.W.; Anker, J.N.V.D.; Knibbe, C.A.J. Towards Rational Dosing Algorithms for Vancomycin in Neonates and Infants Based on Population Pharmacokinetic Modeling. Antimicrob. Agents Chemother. 2015, 60, 1013–1021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  318. Traunmüller, F.; Popovic, M.; Konz, K.-H.; Joukhadar, C.; Vavken, P.; Leithner, A.; Joukhadar, C. A Reappraisal of Current Dosing Strategies for Intravenous Fosfomycin in Children and Neonates. Clin. Pharmacokinet. 2011, 50, 493–503. [Google Scholar] [CrossRef] [PubMed]
  319. Cohen-Wolkowiez, M.; Sampson, M.; Bloom, B.T.; Arrieta, A.; Wynn, J.; Martz, K.; Harper, B.; Kearns, G.L.; Capparelli, E.V.; Siegel, D.; et al. Determining population and developmental pharmacokinetics of metronidazole using plasma and dried blood spot samples from premature infants. Pediatr. Infect. Dis. J. 2013, 32, 956–961. [Google Scholar] [CrossRef] [Green Version]
  320. Procianoy, R.S.; Silveira, R.D.C. The challenges of neonatal sepsis management. J. Pediatr. Rio J. 2019, 96 (Suppl. S1), 80–86. [Google Scholar] [CrossRef]
  321. Zyryanov, S.K.; Ushkalova, E.A.; Kondratyeva, E.I.; Butranova, O.I.; Kondakova, Y.A. Gene Polymorphism of Biotransformation Enzymes and Ciprofloxacin Pharmacokinetics in Pediatric Patients with Cystic Fibrosis. Biomedicines 2022, 10, 1050. [Google Scholar] [CrossRef]
  322. Yalçın, N.; Flint, R.B.; van Schaik, R.H.; Simons, S.H.; Allegaert, K. The Impact of Pharmacogenetics on Pharmacokinetics and Pharmacodynamics in Neonates and Infants: A Systematic Review. Pharm. Pers. Med. 2022, 15, 675–696. [Google Scholar] [CrossRef]
Figure 1. PK/PD targets for optimizing efficacy and minimizing antibacterial resistance for time-dependent, concentration-dependent, and concentration-dependent with time dependence antibiotics.
Figure 1. PK/PD targets for optimizing efficacy and minimizing antibacterial resistance for time-dependent, concentration-dependent, and concentration-dependent with time dependence antibiotics.
Biomedicines 11 00940 g001
Figure 2. Factors affecting GFR in neonates.
Figure 2. Factors affecting GFR in neonates.
Biomedicines 11 00940 g002
Table 1. Time of expression, age-associated changes, and antibiotic-substrates for the main intestinal transport proteins.
Table 1. Time of expression, age-associated changes, and antibiotic-substrates for the main intestinal transport proteins.
Transport ProteinTime of Expression in
Different Cells
Age-Dependent Change of ExpressionTransported Antibiotics
BCRPIntestinal epithelium—from 5.5 to 28 weeks of gestation.
Hepatocytes—from 10 to 11 weeks [45]
Similar levels in preterm newborns, full-terms, and adults [43,46]Delafloxacin, ciprofloxacin, enrofloxacin, nitrofurantoin, norfloxacin, ofloxacin [47,48]
P-gpEnterocytes—from the 12 weeks of gestation [45]Expression in fetus is lower than in adult samples [43]Erythromycin, tetracycline [49]
Azithromycin [50]
Levofloxacin, sparfloxacin [51]
Dicloxacillin [52,53]
MRP2Appears in the liver of 14-week-old fetuses, strong expression at 19 weeks [54]Lower protein expression in fetuses and term newborns than in adults [43]
MRP2 mRNA was 30-fold lower in fetal, 200-fold lower in neonatal, and 100-fold lower in infant liver compared to adults [55]
Ampicillin, azithromycin, and ceftriaxone, cefodizime, ceftriaxone [44,56]
OATP1A2--Levofloxacin [49]
Ciprofloxacin, enoxacin, gatifloxacin, levofloxacin, lomefloxacin, norfloxacin [57]
Erythromycin [58]
Tebipenem [59]
OATP1B1-High expression in the fetus and low expression in the term newborns, with stable protein levels further on [43]
OATP1B1 expression was 20-fold lower in fetal, 500-fold lower in neonatal, and 90-fold lower in infant liver compared to adults [55]
Benzylpenicillin, rifampicin, rifampin, rifampicin [49]
Cefazolin, cefditoren, cefoperazone, nafcillin [60]
OATP1B3-OATP1B3 mRNA was 30-fold lower in fetal, 600-fold lower in neonatal, and 100-fold lower in infant liver compared to adults [55]
OATP1B3 exhibited high expression at birth, decline over the first months of life, and then increase in the preadolescent period [61].
Rifampicin, rifampin [49]
Cefadroxil, cefazolin, cefditoren, cefmetazole, cefoperazone, cephalexin, nafcillin [60]
Erythromycin [58]
OATP2B1-Similar levels in samples from all age groups [43]
Intestinal OATP2B1 expression in neonates was significantly higher than in adults [55]
Benzylpenicillin [62]
Tebipenem pivoxil [63]
PEPT1Enterocytes—from 24.7 to 40th week of gestation (comparable level with full-terms) [43]The PEPT1 mRNA expression of the neonates/infants was only lightly lower (0.8-fold) than the older children (p < 0.05) [55]Cefadroxil, ceftibuten, cefixime, cephradine, cephalexin [60]
Table 2. The summary of GIT features in neonates and their effects on absorption and bioavailability of orally administered drugs.
Table 2. The summary of GIT features in neonates and their effects on absorption and bioavailability of orally administered drugs.
ParameterAge-Associated ChangeEffect on Absorption and
Bioavailability
pHValue close to neutral at birth was described in some studies with rapid change to acidic valuesHigh gastric pH may provide positive effect on the oral bioavailability of acid-labile drugs (e.g., ampicillin, amoxicillin, penicillin, nafcillin, erythromycin), it favors the ionization, and reduces the absorption of weak acids and may negatively affect absorption of weak bases by decreasing their solubility [64]
Gastric volumeDecreasedSolubility of some drugs can be decreased
Gastric emptying and intestinal transitSome slowing is specific, intestinal transit in preterm newborns is 4 times longer than in adults [37]Delay and decrease in absorption
Biliary functionDecreased compared with older children and adults [31]Possible decrease in the intestinal absorption of lipid-soluble drugs
Pancreatic enzymesDecrease at birth with further rise through the first year of lifeDecreased intra-duodenal hydrolysis may result in incomplete absorption
Intestinal surface areaReduced surface-to-volume ratio in children when compared with adults [64]Reduced absorptive surface may influence absorption of some drugs
Intestinal permeabilityIn preterm infants (26–36 weeks gestation), intestinal permeability is higher than in healthy term infants only if measured within two days of birth [65]Increase in absorption may by for drugs with paracellular route of absorption [66]
Transport proteinsDecreased p-gp, MRP1, MRP2—decreased efflux functionPossible increase in absorption of corresponding substrates
Light decrease in PEPT1Possible decrease in absorption of corresponding substrates
Intestinal wall drug-metabolizing enzymesPossible decrease in cytochrome P450 isoenzime 3A4 (CYP3A4) in neonates at birth with further rise through childhood period [67]Possible increase in absorption and bioavailability of CYP3A4 substrates
Table 3. Food effects on absorption of oral antibiotics in pediatric and adult populations.
Table 3. Food effects on absorption of oral antibiotics in pediatric and adult populations.
Drug (Oral
Administration)
Food Effect on PK Parameters in Pediatric PopulationFood Effect on PK Parameters in Adults
Penicillin V (phenoxymethylpenicillin)Cmax decreasedPenicillin V (phenoxymethylpenicillin)
AmoxicillinCmax decreased at lower doses, unchanged at higher doses
AUC unchanged at all doses
No effect
AmpicillinCmax unchanged
AUC unchanged
Some studies suggest reduction in Cmax and AUC with food
Cefpodoxime proxetil Tmax prolonged
Cmax unchanged
Cmax and AUC increase with any meal intake
CephalexinCmax lightly decreased
AUC lightly increased
Delay in absorption
AUC unchanged
CefaclorCmax decreasedTmax prolonged
Cmax decreased
CefadroxilCmax decreasedNo effect
CephradineCmax decreasedNo effect
CefiximeNo clinically significant changes in Cmax and AUCTmax prolonged
Cmax and AUC unchanged
ClarithromycinCmax unchanged
AUC unchanged
Cmax unchanged
AUC unchanged
Table 4. Age-associated change of absorption for non-oral routes of drugs administration.
Table 4. Age-associated change of absorption for non-oral routes of drugs administration.
Routes of
Administration
Main Physiological Factors Affecting PK Parameters in NeonatesChange of Absorption
IntramuscularBlood flow to muscle:
variable decrease over the first 2–3 weeks of life.
The ratio of muscle mass to body mass:
less in neonates than in adults
Water content:
higher proportion of water in neonates [118]
Possible variability in absorption, though for many antibiotics it nearly reaches adult values.
Hydrophilic drugs may have greater intramuscular
absorption in neonates than children or adults due to high water content in muscles
RectalpH:
decreased in neonates compared with older infants (mean pH 6.47 vs. 6.90) [119] and with adults (pH 7.2–7.4) [120]
Nearly no change in absorption of drugs (absorbable drugs should have pKa values near or above the physiological range)
PercutaneousSkin thickness and keratinization:
reduced
Hydration of the stratum corneum:
increased
Surface area to bodyweight ratio:
higher than in adults [38,121]
Increased absorption of drugs including some topical antibiotics with risk of overexposure, and potential toxic effects
Inhalation
Intranasal
Stage of lung development:
late preterm neonates (28–34 weeks of gestation) may have saccular stage of lung development, more preterm infants—just the stage of development of bronchioles and alveolar epithelium, both with surfactant deficiency and high risks for respiratory distress syndrome [122]
Permeability of the mucosa of the nasal cavity:
Increased in neonates [121]
Reduced absorption from the lower respiratory department and increased absorption from the upper respiratory department including nasal cavity (e.g., with facemask for inhalation)
Table 5. Relative rates of antibiotics distribution to the different organs and tissues.
Table 5. Relative rates of antibiotics distribution to the different organs and tissues.
AntibioticCNSLungsGallbladder, BileUrinary TractBones and Joints
Fosfomycin++++++++++++
Lincosamides+
High doses may result in therapeutic concentrations achievement
+++++/++++/+++
Rifamycins+++++++ (?)+/++++/+++
Fluoroquinolones++/+++
Minimum values for ciprofloxacin, higher value for respiratory fluoroquinolones
+++
High in alveolar cells, inflammation may decrease penetration
+/++
Higher for levofloxacin, ofloxacin, ciprofloxacin
++/++++++
Macrolides+
Rapid removement by P-gp of BBB
+++
Higher values in the lung parenchyma, less in epithelial lining fluid
−/+−/+++
Aminoglycosides+
Penetration may be increased in preterm infants with increased risks of aminoglycoside-dependent ototoxicity
++
Only in epithelial lining fluid and bronchial mucus, no cellular entry
−/++++++
Glycopeptides?
Variable data, not predicting clinical outcome, penetration may increase in neonates with meningitis
+++++/++++++++/+++
Lipoglycopeptides+
Poor penetration
++++ (?)++/++++++
Cephalosporines++/+++
Especially high for ceftriaxone, cefotaxime, ceftazidime, cefixime and cefepime
Low—for cefuroxime.
Meningeal inflammation increases penetration
+++
High concentrations in the lung parenchyma, epithelial lining fluid and bronchial mucus
++/+++
The highest value is for cefoperazone, cefotetan, minimum—for cefotaxime
+++++/+++
Penicillins+
Negligible with non-inflamed BBB and increased in meningitis. Dose increase is needed in CNS infections
++/+++
High in the epithelial lining fluid and bronchial mucus
++/+++
The highest value is for piperacillin/tazobactam
+++
High concentrations in urine
−/+
Undetectable, exception for flucloxacillin, piperacillin tazobactam, amoxicillin clavulanat
Carbapenems+++
Imipenem reaches higher CSF concentrations, but causes seizures in neonates, especially preterms, thus meropenem is preferred
++/++++
Despite low bile concentrations, some studies demonstrated clinical efficacy of imipenem and meropenem)
+++
Imipenem: adding cilastatin prevents degradation with dehydropeptidase-I in kidneys
++/+++
Oxazolidinones++
Penetration in CNS tissues and CSF, brain concentration of linezolid is higher than for tedizolid
+++
Concentrations in epithelial lining fluid > serum > alveolar macrophages)
++ (?)+++++
?—more studies are needed; + detectible concentrations (low levels); ++ moderate concentrations; +++ high concentrations.
Table 6. Antibacterials with different bilirubin displacing activity (Reprinted with permission from [175] Copyright year, 2023; Copyright owner, Elsevier).
Table 6. Antibacterials with different bilirubin displacing activity (Reprinted with permission from [175] Copyright year, 2023; Copyright owner, Elsevier).
Moderate to Strong Displacing EffectMinimal Displacing EffectNo Displacing Effect
Sulfamethoxazole
Sulfisoxazole
Sulfadiazine
Ceftriaxone
Cefotetan
Moxalactam
Dicloxacillin
Carbenicillin
Cefazolin
Nafcillin
Ampicillin
Oxacillin
Methicillin
Cefotaxime
Aminoglycosides
Table 7. CYP450 isoenzymes ontogeny and antibiotics-substrates.
Table 7. CYP450 isoenzymes ontogeny and antibiotics-substrates.
CYP450 IsoenzymeAntibiotic-SubstrateComment on Ontogeny
CYP1A1Linezolid [186]Absent-to-low expression in the neonate; activity reaches 50% of adult values by 1 year of age [193]
CYP1A2-Becomes active at 1 to 3 months [194]
The median expression was significantly greater after 15 months postnatal age (n = 55) than in fetal and younger postnatal samples [195]
CYP2A6-Low activity during fetal age (<1% of adult values) with increase up to 50% adult activity values at neonatal age. Activity and protein expression reach adult values between 1 and 15 years of age [196]
CYP1B1Linezolid [186]Appears in the early stages in ontogenesis [197]
CYP2B6-2-fold increase in expression and activity in the first month of life, it is suggested that there are no additional changes between 1 month and 18 years or a 7-fold increase between 1 year and adulthood [198]
CYP2C8Linezolid [186]The median expression was significantly greater among samples from subjects older than 35 postnatal days (n = 122) compared with fetal samples and very young infants (fetal to 35 days postnatal, n = 100) [195]
CYP2C9-Expression at 1 to 2% of mature values during the first trimester, with progressive increases during the second and third trimesters to levels approximately 30% of adults. From birth to 5 months values varied 35-fold and were significantly higher than those observed during the late fetal period, with 51% of samples exhibiting values commensurate with mature levels. Less variability was observed between 5 months and 18 years [199]
CYP2C19-Expression did not change at birth, increased linearly over the first 5 postnatal months, and varied 21-fold from 5 months to 10 years [199]
CYP2D6Linezolid [186]Hepatic expression in neonates <7 days was higher than in first and second trimesters, but close to the values of third trimester. Expression in neonates >7 days was higher than in earlier ages [200]
Adult activity is achieved by 3–5 years of age [193]
CYP2E1-Lower value in neonatal samples compared with infants 31 to 90 days of age, increased with age. Median (range) values of microsomal protein for older infants, children, and young adults were 8.8 (0–70); 23.8 (10–43); 41.4 (18–95) pmol/mg, respectively [201]
CYP3A4Erythromycin [156]
Linezolid [186]
Clindamycin [202]
Activity is extremely low in the fetus; it begins to increase after birth reaching 30–40% of the adult value after one month, 50% at 6–12 months, and 100% at age 1–5 years [203]
Protein expression was 200-fold lower in the fetal tissues compared to either the pediatric or adult tissues [191]
CYP3A5Linezolid [186]
Clindamycin [111]
Protein expression is relatively stable (0.001–0.005 nmol/mg) in fetal, pediatric, and adult samples [191]
CYP3A7Clarithromycin [204]Highly active in fetuses with a subsequent decrease in activity after first week of life, reaching minimal levels in adults [204]
Protein expression was 10-fold higher in the fetal tissues compared to either the pediatric or adult tissues [191]
Table 8. Effect of antibiotics on human CYP450 enzymes (direct and indirect).
Table 8. Effect of antibiotics on human CYP450 enzymes (direct and indirect).
CYP450 IsoenzymeAntibiotics-InhibitorsAntibiotics-Inducers
CYP1A2Ciprofloxacin [207]
Caderofloxacin, antofloxacin, moxifloxacin, gatifloxacin * [208].
Oritavancin * [214]
-
CYP2B6Oritavancin * [214]Rifampicin [215]
CYP2C8Piperacillin [211]Rifampicin [215]
Trimethoprim [216,217]
CYP2C9Oritavancin * [214].Rifampicin [215]
Sulfamethoxazole [216]
Dicloxacillin [53]
Levofloxacin [208]
CYP2C19Oritavancin * [214]
Meropenem [213]
Rifampicin [215]
Dicloxacillin [53]
CYP2D6Oritavancin * [214]Oritavancin [214]
CYP3A4Erythromycin, roxithromycin, clarithromycin, troleandomycin, telithromycin, josamycin [205]
Oritavancin * [214]
Meropenem [213]
Flucloxacillin [212]
Dicloxacillin [53]
Rifampin (inducer of CYP 3A4) [206,215]
Oritavancin * [214]
CYP3A5Erythromycin, clarithromycin [205]-
CYP3A7Erythromycin [218]Rifampicin, troleandomycin, erythromycin (upregulation of expression) [218,219]
* Negligible effect.
Table 9. PPB, Vd and CL of antibiotics in neonates and adults.
Table 9. PPB, Vd and CL of antibiotics in neonates and adults.
DrugVdPPB RateCLT1/2
AdultsNeonatesAdultsNeonatesAdultsNeonatesAdultsNeonates
Penicillin Group
Penicillin G0.53–0.67 L/kg
[244]
Extremely preterm neonates comparable with full-terms, mean value 0.50 L/kg, [245]45–68%
[244]
60%
[245]
560mL/min
[244]
Median—0.21 L/h (interquartile range, 0.16 to 0.29 L/kg) [245]0.4–0.9 h
[244]
Median—3.6 h (interquartile range, 3.2 to 4.3 h)
[245]
Ampicillin and
Ampicillin sulbactam
14.2 to 15.1 L
[246]
GA 22–24 weeks:
0.25 L/kg;
GA 25–27 weeks:
0.34 L/kg
[247]
20% [248]Some decrease is proposed
[247]
10.7 L/h
[249]
GA 22–24 weeks:
0.023 L/h/kg;
GA 22–24 weeks:
0.036 L/h/kg
[247]
1 h
[246]
GA 22–24 weeks:
7.6
[6.4–9.0] h;
GA 25–27 weeks:
6.6
[5.6–7.9] h
[247]
Amoxicillin27.7 L
[250]
GA 25 to 42 weeks:
0.65 L/kg
[251];
<20%
[252]
Less than in adults
[252]
21.3 L/h
[250]
0.096 ± 0.036 L/kg−1 h−1
[251]
~1 h
[250]
5.2 ± 1.9 h
[251]
FlucloxacillinAbout 10 L
[253]
GA 26 to 42 weeks:
0.54 ± 0.17 L/kg
[254]
95–96%
[124]
GA 25 to 41 weeks:
86.3%
[139]
93.1 mL/min
[255]
GA 26 to 42 weeks:
0.18 L/kg/h
[254]
0.88 h
[255]
2.6 ± 1.6
hours
[254]
Piperacillin0.27 ± 0.13 L/kg
[256]
PNA < 14 weeks:
0.42 L/kg
[257]
20–30%
[256]
Median GA of 30 weeks:
30%
[257]
0.183 L/h/kg
[258]
Median GA of 30 weeks:
median value of 0.085 L/h/kg
[257]
1.1 ± 0.6 h
[256]
3.5
(1.7–8.9) h
[257]
Ticarcillin15.7 L
[258]
Preterms > 30 weeks of gestation:
0.26–0.34 L/kg
Fullterms: 0.22–0.23 L/kg
Total population
0.48 L/kg
[259]
45–65%
[258,260]
Less than 45%
[259]
46.2 ± 10.9 mL/min/m2
[260]
PNA < 14 days:
0.078 L/kg/h;
PNA ≥ 4–45 days: 0.078 L/kg/h
[259]
1.23 h
[258]
PNA < 14 days:
24.3
(11.2–35.7) h;
PNA = 14 days:
15.1
(5.0–44.8) h
[259]
Cephalosporins
Cefotaxime33.3 L/1.73 m2
[261]
GA 30 to 40.1 weeks:
median value of 0.36 L/kg
[262]
Median value of 29.45–29.86% in critically ill adults against 27–38% in healthy volunteers
[261,263]
GA 30 to 40.1 weeks:
median value of 40%
[262]
138.4 mL/min
[261]
0.04 (0.02–0.09) L/h/kg
[262]
1.1 hours
[261]
GA < 32 weeks,
PNA < 7 days:
3.49 ± 0.45 h,
PNA ≥ 7 days:
3.72 ± 0.85 h;
GA 32–63 weeks,
PNA < 7 days:
3.28 ± 0.35 h,
PNA ≥ 7 days:
2.0 ± 0.24 h;
GA ≥ 37 weeks,
PNA < 7 days:
2.77 ± 0.49 h,
PNA ≥ 7 days:
2.01 ± 0.57 h [264]
Cefepime0.2 L/kg
[265]
Mean GA 38.1 weeks: median value of 0.62 L/kg
[266]
20%
[265]
Mean GA 38.1 weeks:
≤20%
[266]
7.2 ± 0.48 L/h
[265]
0.18 (0.13–0.24) L/h/kg
[266]
2–2.3 h
[265]
4.9 ± 2.1 h
[267]
Carbapenems
Ertapenem0.7 L
[268]
0.4–0.6 L/kg
[269]
92–95%
[146]
Some decrease is proposed0.7 L/h
[268]
0.05–0.25 L/h/kg
[269]
6.1 h
[268]
NA
Meropenem21 L
[270]
GA 23–41 weeks:
0.4 L/kg
[271]
2%
[271]
NA27.6–36.3 mL/min
[146]
GA 23–41 weeks:
0.104 L/h/kg
[271]
0.093–0.238 L/h/kg
[269]
1 h
[270]
PNA 5–44 days:
1.3–5.4 h
[269]
Imipenem0.22 L/kg
[272]
GA 24–41 weeks:
0.73 L
[272]
20%
[272]
<20%
[272]
0.2 L/h/kg
[273]
0.21 L/h
[272]
1–3 h
[272]
1.9 h in term neonates,
2.4 h in preterm neonates
[272]
Doripenem16.8 L
(8.09–55.5)
[274]
GA <32 weeks:
~0.56 L/kg
Infants:
~0.55 L/kg
[275]
8%
[274]
NA16.0 L/h
[274]
PNA < 4 weeks:
2.03 mL/min/kg;
PNA > 4 weeks:
3.03 mL/min/kg [275]
1 h
[274]
2.98 h
[275]
Macrolides
Azithromycin31.1 L/kg
[276]
GA 24–28 weeks:
1.88 L/kg
[277]
30%
[158]
NA
[158]
37.8 L/h [276]0.15 L/h/kg0.75 [277]68 h
[276]
58 h
[159]
Oxazolidinones
LinezolidIn most studies
50–60 L
[278]
GA < 34 weeks:
0.92 L/kg;
GA ≥ 34 weeks:
0.72 L/kg
[279]
Preterms < 1 week:
0.81 L/kg;
Fullterms < 1 week:
0.78 L/kg;
Fullterms ≥ 1 week ≤ 28 days: 0.66 L/kg
[280]
10.5–31%
[278,280]
NA5.54 L/h for 30-year-old patients;
1.35 L/h for 85-year-old patients
[281]
GA < 34 weeks:
0.17 (0.07–0.31) L/h/kg;
GA ≥ 34 weeks:
0.16 (0.14–0.19) L/h/kg
[279]

Preterms < 1 week PNA:
2.0 mL/min/kg;
Fullterms < 1 week PNA:
3.8 mL/min/kg;
Fullterms ≥ 1 week ≤28 days: 5.1 mL/min/kg
[280]
4–6 h
[280]
Preterms
<1 week PNA:
5.6
[2.4–9.8] h;
Fullterms
<1 week:
3.0
[1.3–6.1] h;
Fullterm
≥1 week ≤28 days:
1.5
[1.2–1.9]
[280]
Lipoglycopeptides
Telavancin0.1–0.15 L/kg
[282]
NA 90–93%
[282]
NA 0.012–0.014 L/h/kg
[282]
NA 6–8 h
[282]
NA
Lipopeptides
Daptomycin0.1 L/kg
[283]
GA 23–40 weeks:
262.4 mg × h/L
[284]
>90%
[283]
NA0.01 L/h/kg
[283]
GA 23–40 weeks:
0.21 L/kg
[284]
8–9 h
[282]
NA
Fluoroquinolones
Levofloxacin1.09–1.26 L/kg
[169]
0.5–2 years old:
1.56 L/kg (IV),
2.32 L/kg (PO)
[103]
24–38%
[115]
NA0.5–2 years old:
0.35 L/h/kg (IV),
0.31 L/h/kg (PO)
[103]
5.76–8.52 L/h
[169]
6–8 h
[169]
0.5 to <2 years:
4.1 ± 1.3 h
[103]
Moxifloxacin2.1 L/kg
[285]
2.25 L/kg
[285]
39%
[285]
NA12 ± 2 L/h
[171]
0.35 L/h/kg
[285]
11.5–15.6 h
[171]
8.6 h [286]
Aminoglycosides
Amikacin24 L
[287]
0.47–0.61 L/kg
[288]
≤10%
[287]
NA100 mL/min—serum clearance
94mL/min—renal clearance
[287]
0.025–0.097 L/kg/hr
[288]
2–3 h [287]PMA ≤ 29 weeks,
PNA 0–7 days:
17.6 ± 4.8 h,
PNA ≥ 8 days: 9.8 ± 2.1 h;
PMA 30–33 weeks,
PNA 0–7 days:
12.3 ± 0.5 h,
PNA ≥ 8 days:
7.3 ± 3.2 h;
PMA 34–36 weeks,
PNA 0–7 days:
6.3 ± 1.4 h,
PNA ≥ 8 days:
5.2 ± 1.6 h.
[288]
Lincosamides
Clindamycin0.79 L/kg
[289]
PMA < 28 weeks:
1.20 L/kg;
PMA > 28–32 weeks:
1.3 L/kg;
PMA > 32–40 weeks:
1.03 L/kg;
PMA > 40–60 weeks:
0.99 L/kg
[165]
78–94%
[289]
81%
[164]
0.3–0.4 L/h/kg
[289]
PMA < 28 weeks:
0.14 L/kg/h;
PMA > 28–32 weeks:
0.19 L/kg/h;
PMA > 32–40 weeks:
0.24 L/kg/h;
PMA > 40–60 weeks:
0.28 L/kg/h
[164]
2.4 h
[202]
PMA ≤ 38 weeks:
5.89
(2.42–12.90) h;
PMA > 28–32 weeks:
5.25
(2.34–8.87) h;
PMA > 32–40 weeks:
3.96
(1.30–8.83) h;
PMA > 40–60 weeks:
2.35
(0.94–6.44) h;
PMA > 5 months < 1 year:
2.05
(1.26–3.47) h
[164]
Glycopeptides
Teicoplanin0.86 L/kg
[290]
PMA 26 to 43 weeks:
0.283 L
[291];
90–95%
[149]
80.5–71.9%
[150]
0.0114 L/h/kg
[290]
0.0227 L/h
[291]
70–100 h
[149]
29.75 ± 8.86 h
[291]
Vancomycin0.4–1 L/kg
[292]
Preterms, median age 14 (3–58) days: 0.884 L [293];
GA 23–34 weeks:
0.54 ± 0.10 L/kg
[294]
10–50%
[292]
NA2.64 L/h
[292]
Preterms, median age 14 (3–58) days: 0.1 L/h
[293];
GA 23–34 weeks:
0.06 L/h/kg
[294]
4–12 h [283]3.5–10 h [295]
Other groups of antibiotics
Rifampin1.6 L/kg
[117]
1.84 ± 0.59
[296]
72–92%
[173]
NA10–19.3 L/h
[117]
3.6 L/h, 70 kg
[117]
2.5 h
[117]
PNA < 14 days:
7.1
(3–23.9) h;
PNA ≥ 14 days:
3.5
(1.9–6.5) h
[117]
Fosfomycin0.42 L/kg
[113]
GA 38–40 weeks:
0.38 L/kg
[172]
Negligible
[172]
NA8.7 ± 1.7 L/h
[113]
0.14 L/h
[172]
2.4–2.8 h
[113]
2.4 ± 0.4 h
[113]
Metronidazole 0.25–0.85 L/kg
[297]
0.71 L/kg [297]20% [297]NAGA < 26 weeks:
0.024
(0.010–0.086) L/h;
GA 26–29 weeks:
0.026 (0.012–0.076) L/h;
GA 30–32 weeks:
0.029 (0.015–0.074) L/h [297]
6–10 h
[298]
8 h [297]GA < 26 weeks:
20.5
(5.7–49.9) h;
GA 26–29 weeks:
18.6
(6.5–38.7) h;
GA 30–32 weeks:
16.7
(6.5–38.7) h [297]
GA—gestational age; PMA—postmenstrual age; PNA—postnatal age.
Table 10. Antibiotic dosing regimens in neonates.
Table 10. Antibiotic dosing regimens in neonates.
AntibioticRegimen and Indications for Critically Ill
Neonates
CommentsPK/PD TargetSafety
Penicillin Group
Penicillin G
[245]
EOS:
25,000 IU/kg every 12 h for all MIC values tested (up to 2 mg/L).
GA of ≥32 weeks
PNA of <72 h
40% fT > MIC
MIC < 2 mg/L
100% fT > MIC
MIC ≤ 0.5 mg/L
100 μg/mL
At high doses—risk of neurotoxicity and encephalopathy
[245]
Ampicillin
[299]
50 mg/kg every 8 hPNA 1—10 days75% T > MIC
MIC ≤ 8 mg/L
140 μg/mL
At high doses—risk of neurotoxicity
[247]
Ampicillin
[300]
50 mg/kg every 8 or 12 hGA ≥ 32 weeks40% fT > MIC;
100% fT > MIC
Ampicillin
[247]
EOS:
A short-course ampicillin regimen:
2 doses, 50 mg/kg every 12 h
GA 22–27 weeks
PNA < 7 days,
BW < 1500 g
100% fT > MIC
MIC ≤ 8 mg/L
Azlocillin [301]100 mg/kg every 8 hGA 30.1–41.1 weeks
PNA 1.0–2.0 days
70% fT > MIC
MIC = 2–16 mg/L
Risk of hepatotoxicity
Amoxicillin
[302]
50 mg/kg every 24 h
75 mg/kg every 24 h
GA 36–37 weeks
GA 38–42
weeks
100% T > MIC
MIC = 0.25–1 mg/L
Doses > 140–150 mg/L:
risk of nephrotoxicity
Flucloxacillin
[254]
25 mg/kg every 4 h
10 mg/kg every 6 h
For all neonates, irrespective of their GA and PNA.40% T > MIC
MIC = 0.25–2 mg/L
200 mg/L
Piperacillin/Tazobactam
[257]
100 mg/kg every 8 hPMA ≤ 30 weeks50–75% fT > MIC
MIC = 0.5–32 mg/L
157.2 mg/L
Risk of neurotoxicity
[303]
80 mg/kg every 6 hPMA 30–35 weeks
80 mg/kg every 4 hPMA 35–49 weeks
Ticarcillin/Clavulanic acid
[259]
75 mg/kg every 12 hPNA < 14 weeks75% fT > MIC
MIC = 0.5–16 mg/L
75 mg/kg every 8 hPNA ≥ 14–45 weeks
Cephalosporins
Ceftoraline Fosamil
[304]
6 mg/kg every 8 h<2 months35–44%
fT > MIC
MIC = 0.5–2 mg/L
High
risk of diarrhea
Ceftazidime
[305]
25 mg/kg every 8 h
for MIC 4 mg/L
0.1–2 years70% fT > MIC
MIC = 4–8 mg/L
50 mg/kg every 8 h
for MIC 8 mg/L
Cefotaxime
[262]
50 mg/kg every 12 hGA 30.0–41.1 weeks
PNA 1.0–3.0 days
70% fT > MIC
MIC = 2 mg/L
Feeding intolerance
Diarrhea
[262]
Cefotaxime
[306]
50 mg/kg every 12 hPNA < 7 days75% fT > MIC
MIC = 2–4 mg/L
50 mg/kg every 8 hGA < 32 weeks
PNA ≥ 7 days
50 mg/kg every 6 hGA ≥ 32 weeks
PNA ≥ 7 days
Cefepime
[266]
50 mg/kg every 12 h
for MIC 4 mg/L
PMA < 38 weeks70% fT > MIC
MIC = 4–8 mg/L
40 mg/kg every 8 h
for MIC 8 mg/L
PMA ≥ 38 weeks
Cefazoline
[307]
25 mg/kg every 12 hPNA ≤ 7 days
BW ≤ 2000 g
60% fT > MIC
MIC > 8 mg/L
50 mg/kg every 12 hPNA ≤ 7 days
BW > 2000 g
25 mg/kg every 8 hPNA 8–28 days
BW ≤ 2000 g
50 mg/kg every 8 hPNA 8–28 days
BW > 2000 g
Carbapenems
Meropenem
[308]
20 mg/kg every 12 hGA < 32 weeks
PNA < 14 weeks
50% T > MIC MIC = 4 mg/L
75% T > MIC MIC = 2 mg/L
64.2 mg/L—risk of neurotoxicity
44.45 mg/L for Cmin—risk of nephrotoxicity
[309]
20 mg/kg every 8 hGA < 32 weeks
PNA ≥ 14 weeks
GA ≥ 32 weeks
PNA < 14 weeks
30 mg/kg every 8 hGA ≥ 32 weeks
PNA ≥ 14 weeks
Doripenem
[275]
5 mg/kg
every 24 h
<8 weeks
PNA < 12 weeks
70–100% T > MIC
MIC = 1 mg/L
Well tolerated
8 mg/kg
every 24 h
>8 weeks
PNA < 12 weeks
Imipenem
[272]
20–25 mg/kg
every 6–12 h
GA 24–41 weeks
PNA 2–153 days
100% T > MIC
MIC = 2 mg/L
Lipopeptides
Daptomycin
[310]
6 mg/kg every 12 hMedian GA 27 weeks; PNA 5 daysNO PK/PD STUDYRisk of elevation of liver enzymes
Fluoroquinolones
Levofloxacin
[311]
10 mg/kg every 12 hPMA 27–42 weeksNO PK/PD STUDYRisk of diarrhea, hepatotoxicity,
Dysglycemia
Moxifloxacin
[285]
6 mg/kg every 12 h>3 months to <2 yearsAUC(0–24) at steady state 20–60 mg·h/L
Cmax at steady state 2–6 mg/L
Risk of individual intolerance
risk of QT prolongation
Moxifloxacin
[286]
5 mg/kg every 24 hGA 25 weeksCmax/MIC: >10
AUC0–24/MIC: ≥100
Macrolides
Azithromycin
[277]
20 mg/kg every 24 h
× 3 days
GA 24–28 weeks
PMA < 72 h
AUC24/MIC90
of >4 h
Risk of necrotizing enterocolitis, intraventricular hemorrhage
Lincosamides
Clindamycin
[164]
5 mg/kg every 8 hPMA ≤ 32 weeksAUCss,0–8 = 33.8 μg·h/mL
Css,max = 7.9 μg/mL
fCss,50 = 0.12 μg/mL
No adverse events related to clindamycin use in this study
7 mg/kg every 8 hPMA 32–40 weeks
9 mg/kg every 8 hPMA > 40–60 weeks
Aminoglycosides
Amikacin
[312]
15 mg/kg
30–36 h
PNA ≤ 14 days
BW
2000 g; ≥2800 g
Ctrough < 3 μg/mL
Cmax 24 g/mL
Toxicity risk at Cmin > 5 μg/mL
[312]
Amikacin
[288]
13 mg/kg every 48 hPNA 0–7 days
PCA ≤ 29 weeks
Serum peak concentration 20–30 mg/L
13 mg/kg every 36 hPNA ≥ 8 days
PCA ≤ 29 weeks
PNA 0–7 days
PCA 30–33 weeks
13 mg/kg every 24 hPNA ≥ 8 days
PCA 30–33 weeks
PNA 0–7 days
PCA 34–36 weeks
13 mg/kg every 18 hPNA ≥ 8 days
PCA 34–36 weeks
13 mg/kg every 24 hPNA 0–7 days
PCA ≥ 37 weeks
13 mg/kg every 24 hPNA ≥ 8 days
PCA ≥37 weeks
Gentamicin
[313]
5 mg/kg every 48 hPremature BW
<1200 g
Cmax/MIC ratio at least 8–10Toxicity risk at Cmin > 2 μg/mL and Cmax > 35 μg/mL
Risk of nephrotoxicity, neurotoxicity, ototoxicity
5 mg/kg every 36 hPremature
BW 1200–2500 g
5 mg/kg every 24 hBW > 2500 g;
Term neonates 1–28 days
Gentamicin
[314]
5 mg/kg every 36 hPMA 30–34 weeks
PNA 8–28 days
PMA ≥ 35 weeks
PNA 0–7 days
Ctrough < 1 μg/mL
Gentamicin/Tobramicin
[315]
4.5 mg/kg of gentamicin
and
5.5 mg/kg of tobramycin
every 72 h
PNA ≤ 5 daysCmax 5–12 μg/mL
Ctrough < 0.5 μg/mL
Glycopeptides
Vancomycin
[293]
12 mg/kg every 8 hGA 23–34 weeksAUC0–24 h of ≥400 mg·h/L>20 μg/mL—risk of nephrotoxicity
[293]
Vancomycin
[316]
25 mg/kg (LD)
15 mg/kg (MD)
every 12 h
PMA < 35 weeksAUC0–24 of 400 mg·h/L at steady-state
25 mg/kg (LD)
15 mg/kg (MD)
every 8 h
PMA ≥ 35 weeks
Vancomycin
[317]
32–60 mg/kg/day
(4 doses)
<1 yearAUC0–24/MIC > 400
60 mg/kg/day
(4 doses)
>1 year
Vancomycin
[294]
10 mg/kg
every 12 h
PMA < 30 weeksAUC0–24/MIC ≥ 267
at steady state
10 mg/kg
every 8 h
PMA ≥ 35 weeks
Teicoplanin
[291]
16 mg/kg
10–11 mg/kg
every 24 h
BW: <1000 g
BW: 1000
<2000 g
AUC/MIC ratio of ≥400
Other groups of antibiotics
Rifampin
[117]
8 mg/kg every 24 hPNA < 14 daysAUC = 55.2 μg·h/mL
Cmax,ss = 8.3 ± 1.8 μg/mL
Well tolerated
15 mg/kg every 24 hPNA 14–61 daysAUC = 55.2 μg·h/mL
Cmax,ss = 13.2 ± 3.3 μg/mL
Rifampin
[296]
10 mg/kg every 24 hGA 29.9 (4.1) weeks
PNA 24.8 (13.4) days
Plasma concentrations
>MIC
Fosfomycin
[318]
50–100 mg/kg every 8 h
50–70 mg/kg every 6 h
Healthy Pre-Term and Full-Term NeonatesMIC = 64 mg/LWell tolerated
100 mg/kg every 24 h
2 partial doses
Pre-Term Neonates aged 1–3 daysMIC = 32 mg/L
150–200 mg/kg every 24 h
3–4 partial doses
Pre-Term neonates aged 3–5 weeksMIC = 32 mg/L
Fosfomycin
[172]
100 mg/kg every 12 hNeonates aged < 7 days or weighing < 1500 gMIC = 32 mg/L
Metronidazole
[319]
7.5 mg/kg every 12 h
15 mg/kg (LD)
PMA < 34 weeksCminss > 2 mg/L
7.5 mg/kg every 8 h
15 mg/kg (LD)
PMA 34–40 weeks
% fT > MIC—Percent of time for free drug concentration remains above the minimum inhibitory concentration; % T > MIC—Percent of time for total drug concentration remains above the minimum inhibitory concentration; AUC—Area under curve; BW—Body weight; Cmax—Maximum (peak) plasma concentration; Cmin—Minimum plasma concentration; Css—Steady-state concentration; Ctrough—Pre-dose plasma concentration; EOS—Early onset sepsis; fCss—free plasma steady—state concentration; GA—Gestational age, weeks; LD—Loading dose; MD—Maintenance dose; MIC—Minimal inhibitory concentration; PCA—Post-conceptual age, weeks; PK/PD—Pharmacokinetics/Pharmacodynamics; PMA—Postmenstrual age, weeks; PNA—Postnatal age, weeks, days, hours.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

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. https://doi.org/10.3390/biomedicines11030940

AMA Style

Butranova OI, Ushkalova EA, Zyryanov SK, Chenkurov MS. Developmental Pharmacokinetics of Antibiotics Used in Neonatal ICU: Focus on Preterm Infants. Biomedicines. 2023; 11(3):940. https://doi.org/10.3390/biomedicines11030940

Chicago/Turabian Style

Butranova, Olga I., Elena A. Ushkalova, Sergey K. Zyryanov, and Mikhail S. Chenkurov. 2023. "Developmental Pharmacokinetics of Antibiotics Used in Neonatal ICU: Focus on Preterm Infants" Biomedicines 11, no. 3: 940. https://doi.org/10.3390/biomedicines11030940

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop