Next Article in Journal
Redo DAIR: The Game Is Seldom Worth the Candle
Next Article in Special Issue
Beta-Lactam Probability of Target Attainment Success: Cefepime as a Case Study
Previous Article in Journal
Development of 3D-Printed Collagen Scaffolds with In-Situ Synthesis of Silver Nanoparticles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pharmacokinetics of Antimicrobials in Children with Emphasis on Challenges Faced by Low and Middle Income Countries, a Clinical Review

by
Kevin Meesters
1,*,
Tinsae Alemayehu
2,3,
Sofia Benou
4,
Danilo Buonsenso
5,6,
Eric H. Decloedt
7,
Veshni Pillay-Fuentes Lorente
7,
Kevin J. Downes
8,9 and
Karel Allegaert
10,11,12
1
Department of Pediatrics, BC Children’s Hospital and The University of British Columbia, 4500 Oak Street, Vancouver, BC V6H 3N1, Canada
2
Division of Pediatric Infectious Diseases, Department of Pediatrics and Child Health, St. Paul’s Hospital Millennium Medical College, Addis Ababa P.O. Box 1271, Ethiopia
3
Division of Infectious Diseases and Travel Medicine, American Medical Center, Addis Ababa P.O. Box 62706, Ethiopia
4
Department of Pediatrics, General University Hospital of Patras, Medical School, University of Patras, 26504 Rion, Greece
5
Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy
6
Centro di Salute Globale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
7
Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7500, South Africa
8
Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
9
Division of Infectious Diseases, The Children’s Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
10
Department of Development and Regeneration, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
11
Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
12
Department of Clinical Pharmacy, Erasmus Medical Center, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
*
Author to whom correspondence should be addressed.
Antibiotics 2023, 12(1), 17; https://doi.org/10.3390/antibiotics12010017
Submission received: 8 November 2022 / Revised: 16 December 2022 / Accepted: 18 December 2022 / Published: 22 December 2022
(This article belongs to the Special Issue Therapeutic Drug Monitoring of Antimicrobials - 2nd Volume)

Abstract

:
Effective antimicrobial exposure is essential to treat infections and prevent antimicrobial resistance, both being major public health problems in low and middle income countries (LMIC). Delivery of drug concentrations to the target site is governed by dose and pharmacokinetic processes (absorption, distribution, metabolism and excretion). However, specific data on the pharmacokinetics of antimicrobials in children living in LMIC settings are scarce. Additionally, there are significant logistical constraints to therapeutic drug monitoring that further emphasize the importance of understanding pharmacokinetics and dosing in LMIC. Both malnutrition and diarrheal disease reduce the extent of enteral absorption. Multiple antiretrovirals and antimycobacterial agents, commonly used by children in low resource settings, have potential interactions with other antimicrobials. Hypoalbuminemia, which may be the result of malnutrition, nephrotic syndrome or liver failure, increases the unbound concentrations of protein bound drugs that may therefore be eliminated faster. Kidney function develops rapidly during the first years of life and different inflammatory processes commonly augment renal clearance in febrile children, potentially resulting in subtherapeutic drug concentrations if doses are not adapted. Using a narrative review approach, we outline the effects of growth, maturation and comorbidities on maturational and disease specific effects on pharmacokinetics in children in LMIC.

1. Introduction

Despite widespread implementation of childhood immunization programs, improvements in hygiene and better access to healthcare, infections have remained the most frequent cause of childhood mortality in low and middle income countries (LMIC) [1,2]. Antimicrobials are potentially lifesaving and are among the most frequently administered drugs in children. Unfortunately, the development of antimicrobial resistance (AMR) is a direct consequence of antimicrobial use. LMIC are disproportionately affected by AMR [3,4], driven by numerous circumstances such as excessive use of broad spectrum antimicrobials in healthcare settings, supply issues, limited availability of diagnostic resources, and over-the-counter dispensation [5]. Antimicrobial stewardship (AMS) is a coordinated set of actions to preserve antimicrobials through enhancing the quality of antimicrobial prescribing and has been proposed as a tool in to combat AMR [6]. While unnecessary antimicrobial prescription should be avoided [7], adequate dosing is also of utmost importance since subtherapeutic concentrations result in therapy failure and selection of drug-resistant organisms [8,9]. Achievement of the effective concentration of a drug is governed by the pharmacokinetic processes absorption, distribution, metabolism and elimination, which is depicted in Figure 1.
The need for adapted drug therapies for children has been increasingly acknowledged, and pediatric pharmacokinetic studies have expanded during the most recent years to optimize dosing in children. A good understanding of pharmacokinetics of antimicrobials is critical for successful treatment of infections and to combat AMR. In this clinical review, we outline the key processes that dictate the pharmacokinetics of antimicrobials to treat bacterial infections in children, detailing important considerations for antimicrobial dosing, as well as highlighting specific challenges in LMIC.

2. Pharmacokinetic–Pharmacodynamic Interaction of Antimicrobials

For each bacterium, the lowest concentration at which an antimicrobial inhibits growth is defined as the minimum inhibitory concentration (MIC) [10]. Since antimicrobials target bacteria, MIC is a key component determining whether the concentration achieved at the site of infection will lead to treatment success or failure. The relationship between the free (unbound) concentration of an antimicrobial over time and the bacterial killing effect in vitro, is described by pharmacokinetic-pharmacodynamic (PK/PD) indices (Figure 2). Broadly speaking, antimicrobials can be categorized as concentration-dependent or time-dependent agents [11]. The bacterial killing effect of concentration-dependent antimicrobials is characterized by the ratio between the peak concentration (Cmax) and MIC. Hence, this is directly related to the maximum concentrations that can be achieved at the site of infection. Although bacterial killing of concentration-dependent drugs is dose-dependent, dosing is curtailed by the minimum concentration at which toxicity usually occurs (minimum toxic concentration; MTC); the area between MIC and MTC is called the therapeutic window [12]. Concentration-dependent antimicrobials also tend to exert a post-antimicrobial effect, thereby maintaining a significant antimicrobial effect below the MIC. On the contrary, time-dependent antimicrobials kill bacteria for as long as concentrations are above the MIC. Therefore, the time above the MIC (T > MIC) is the PK-PD index best associated with efficacy for these antimicrobials [13]. Last, the bacterial killing effect of some antimicrobials can be described as being both concentration- and time-dependent; the ratio between area under the concentration-time curve and MIC (AUC/MIC) is the index of choice for concentration-dependent with time-dependent antimicrobials [13].

3. Pharmacokinetics: Drug Transport through Cell Membranes

Antimicrobials are mostly administered distant to their site of action and therefore require transportation to the target site. Drugs that are administered via an extravascular route (e.g., enteral, transdermal, intramuscular) must be absorbed into the bloodstream for distribution to target sites. Once in the circulation, drugs distribute rapidly to the organs to which blood flows instantaneously such as the heart, liver and kidneys; this is called the central compartment [14]. Subsequently, some drugs may distribute to the peripheral compartment, which refers to organs, tissues and cells that are perfused at a slower rate. In order to egress the intravascular space, a drug molecule must cross through cell membranes, either via passive diffusion or using active cell processes that involve receptors, transport proteins or transcytosis [15]. Cell membranes are highly hydrophobic and negatively charged. The drug’s ability to transport across cell membranes is governed by intrinsic physicochemical properties of the drug, such as the amount of protein binding, molecular size, charge, lipophilicity and partition coefficient [16]. Drugs that are bound to a protein, either intravascularly or in the extravascular space, cannot move across membranes due to its molecular size [17]. At any body site, drugs reach an equilibrium between the amount that is either protein bound or free. Albumin, the main drug binding protein, is alkalic and therefore tends to bind to acidic drugs. Generally, lipophilic and uncharged molecules diffuse easily across membranes, whereas hydrophobic and charged molecules are dependent on active processes to disseminate into the peripheral compartment [12]. LogP, the logarithm of the partition coefficient, expresses the affinity of a molecule to dissolve in either water or octanol, and hence the lipophilicity of a drug. In general, a LogP less than 0 indicates a hydrophilic molecule, whereas LogP greater than 0 reflects a lipophilic molecule [18]. The degree of ionization of a drug is predicted by the acid-base dissociation constant (pKa) that relates the pH at which the ionized and unionized forms exist in equal amounts [19]. Therefore, the amount of unionized drug molecules is not constant, since pH varies in different cells and tissues. Table 1 outlines the physicochemical characteristics of antimicrobials.

4. Pharmacokinetic Processes

4.1. Absorption

Absorption, the transfer of a drug from the site of administration into the bloodstream, is relevant for all drugs that are not injected intravascularly. Bioavailability (F), the extent to which a drug enters the circulation following administration, is by definition 100% when the drug is infused directly into the bloodstream, but lower for all other routes of administration [59].
The oral route is the most common route of administration for antimicrobials. Following oral ingestion, the drug needs to dissolve into smaller particles that can subsequently be absorbed [60]. Since liquid formulations are better dissolved than tablets within the GI tract, liquids have typically greater bioavailability than tablets [61]. In addition to molecular characteristics of a drug, different physiologic factors determine the amount of GI absorption, including the pH at the different parts of the GI tract, gastric emptying, intestinal motility and perfusion [62]. Furthermore, directly after intestinal absorption, some drugs undergo first-pass metabolism in the portal circulation, before reaching the systematic circulation. This reduces systemic drug concentrations relative to the amount absorbed of a drug [63].
Within 48 h after birth, gastric pH decreases to around 3, and then gradually returns to neutrality by day 8–10 of life. Thereafter, the gastric pH slowly declines again to reach adult values at about two years of age [62,63]. These pH changes are less apparent in infants born before 32 weeks’ gestation [64]. A smaller fraction of acid-labile antimicrobials (e.g., benzylpenicillin, ampicillin, amoxicillin, nafcillin, flucloxacillin and erythromycin) will become inactivated in a higher intragastric pH, and will therefore attain a higher bioavailability in neonates and infants [65].
Gastric emptying is delayed immediately after birth, but approach adult values within the first six to eight months of life [62,64]. Delayed gastric emptying typically leads to degradation of drug molecules through prolonged exposure to intragastric acid, which impairs drug absorption [66]. Similarly, intestinal motility is also reduced in neonates and young infants. However, this results in increased transit time and improved absorption; intestinal motility gradually increases by 6–8 weeks of life [64]. Last, immaturity of secretion and activity of bile and pancreatic fluid leads to impaired fat digestion in neonates and infants, which particularly reduces the dissolution of lipophilic drugs, and hence impede this absorption [67]. Concluding, gastrointestinal (GI) absorption is complex and highly variable across drugs, particularly in neonates. Contrasting processes lead to large inter-individual variability in absorption rates across infants. Nevertheless, commonly used antimicrobials (amoxicillin, cephalexin, cefpodoxime) often attain adequate serum concentrations to treat most relevant neonatal pathogens (E. coli, S. pneumoniae, S agalactiae) [68].
There is a complex interaction between food and drug absorption. Different food categories affect absorption in varying ways [66]. High fat meals delay gastric emptying and impair absorption of hydrophilic drugs, but improve absorption of lipophilic drugs (e.g., itraconazole) by enhancing solubility. High protein nutrients increase intestinal blood flow and may thereby increase absorption. However, bioavailability of drugs with similar structures to peptides (e.g., cephalexin, cefadroxil) can be lowered if ingested with proteins. High fiber foods delay gastric emptying, reduce solubility of drugs, and decrease bile salt concentrations. Moreover, fasting decreases gastric pH and leads to delayed gastric emptying, while enhancing splanchnic blood flow and stimulating release of bile salts [66]. These complex interactions make it important for clinicians to be aware about when drugs should be taken with food. Table 2 summarizes food effects on commonly used antimicrobials.
Some antimicrobials are administered intramuscularly. Similar physiologic principles determine the bioavailability after intramuscular absorption, such as the local pH and the tissue perfusion rate. As neonates and infants have low muscle mass and low regional blood flow to muscles, bioavailability via intramuscular administration is lower in infants [79].

4.2. Distribution

Volume of distribution (Vd) is a pharmacologic parameter that relates the amount of a drug in the body to its measured concentration in blood or plasma [14]. It is an apparent volume, since it may well exceed any physiologic volume required to contain all the drug in the body at the measured concentration. The magnitude and the sites of drug distribution are dependent on the physicochemical properties of the drug and different biologic factors, such as body composition and various physiological processes, which are all altered by both ontogeny and disease states [80].
Body composition changes markedly throughout childhood. Premature neonates have a much higher total body water content than term born infants. This increases Vd of hydrophilic drugs, such as aminoglycosides and glycopeptides. As a result, higher doses per kilogram are necessary at initiation for premature neonates to attain the same target serum concentration, compared to term born infants [81]. For some drugs, loading doses are given to rapidly attain optimal concentrations. Since Cmax is directly related to Vd, neonates need higher loading doses of hydrophilic antimicrobials [82]. Furthermore, protein composition evolves in childhood. In infants, lower protein binding has been reported [83]. This increases Vd of antimicrobials that are highly protein bound, such as ceftriaxone. Hypoalbuminemia is common in many diseases, such as nephrotic syndrome, liver failure and cachexia. Under these circumstances, the free fraction of protein bound drug rises [84]. However, this does not necessarily translate to higher drug exposure, as unbound molecules are available for excretion, more unbound drug is cleared [85]. Furthermore, free drug molecules may egress the circulation and bind to extravascular proteins.
Tight junctions and multiple cellular mechanisms prevent substances and micro-organisms from entering the brain and cerebrospinal fluid (CSF), which is known as the blood-brain barrier [86]. Therefore, some antimicrobials may not cross the blood-brain barrier at all. But, the integrity of the blood-brain barrier decreases during central nervous system infection (CNS) [87]. In general, unbound lipophilic and uncharged drugs enter the CSF at a higher rate than hydrophilic drugs with a large size or charge. The abilities of antimicrobials to penetrate CSF are displayed in Table 3.
Efflux transporters, such as p-glycoprotein (P-gp), avert intracellular transport of xenobiotics and toxic substrates [88,89]. As these transporters excrete certain drug molecules, this restricts the distribution of some antimicrobials. On top of that, drugs may both induce and inhibit P-gp, such as rifampicin (inducer) and protease inhibitors (inhibitor), and hence further affect exposure to substrates.
Table 3. Cerebrospinal fluid penetration of antimicrobials.
Table 3. Cerebrospinal fluid penetration of antimicrobials.
AgentCerebrospinal Fluid (CSF) Penetration
Aminoglycosides
Amikacin
Gentamicin
Tobramycin
Systemic amikacin, gentamicin and tobramycin penetrate the CSF of inflamed meninges to a limited extent. Their clinical use for CNS infections is restricted by toxicities if administered intravenously. Intrathecal doses of amikacin, gentamicin, and tobramycin have been reported to be effective and well tolerated [90,91]. No information available for other aminoglycosides.
Antimycobacterials
EthambutolLimited data suggest poor to moderate CSF penetration of inflamed meninges [92].
IsoniazidCSF concentration comparable with plasma concentration in inflamed meninges [92].
PyrazinamideCSF concentration comparable with plasma concentration in inflamed meninges [92].
RifabutinHigher CSF penetration than rifampicin, but toxicities may restrict its use in CNS infections [93].]
RifampicinModerate CSF penetration at standard doses, therefore higher doses may be necessary for adequate CSF penetration [91,92].
BedaquilineBedaquiline penetrated freely into the CSF of adults under treatment with pulmonary tuberculosis [94].
ClofazaminePoor CSF penetration, which may be improved by chemical modification [95].
CycloserineGood CSF penetration of inflamed meninges [92,96].
EthionamideGood CSF penetration [90].
DelamanidVery limited clinical data available, low total CSF levels [97].
Beta-lactamase inhibitors
AvibactamNo data available.
Clavulanic acidVery limited data suggest that amoxicillin-clavulanate may be effective for the treatment of bacterial meningitis [98,99].
SulbactamVery high CSF:plasma concentrations in combination with ampicillin [91]. However, clinical experience with this agent for meningitis is limited.
TazobactamNo clinical data available.
VaborbactamNo clinical data available.
Carbapenems
DoripenemNo clinical data available.
ErtapenemNo clinical data available.
ImipenemMeasurable CSF penetrations, but high proconvulsive activity may restrict its use [100].
MeropenemCSF concentrations adequate for treating meningitis [91].
Cephalosporins
CephalexinUsually ineffective due to lower CSF:serum concentrations [91].
CefazolinCSF concentrations of uninflamed meninges close to the MIC of moderately susceptible bacteria [90].
CefadroxilUsually ineffective due to lower CSF:serum concentrations [91].
CefaclorNo clinical data available.
CefotetanNo clinical data available.
CefoxitinNo clinical data available.
CefprozilNo clinical data available.
CefuroximeReaches CSF concentrations in excess of MIC [91].
CephamycinNo clinical data available.
CefdinirNo clinical data available.
CefepimeAdequate CSF penetration for treatment of meningitis [90].
CefiximeCefixime crosses the blood brain barrier of inflamed meninges, but at limited concentrations and should therefore not be used to treat meningitis [91].
CefotaximeAdequate CSF penetration [90,91]
CeftriaxoneCeftriaxone has an adequate CSF penetration of inflamed meninges. CSF concentrations are lower compared with cefotaxime, most likely given the higher degree of protein binding of ceftriaxone. Nevertheless, ceftriaxone is an adequate agent for treatment of meningitis [90,91].
CeftarolineDifferent case studies reported that ceftaroline attained CSF concentration above MIC [101,102,103].
CeftazidimeCSF attains therapeutic levels in CSF [90,91].
CeftizoximeLimited clinical data available suggest that ceftizoxime penetrates CSF [91].
CeftobiproleNo clinical data available, clinical study ongoing (NCT04178629).
CefiderocolVery limited clinical data available in humans suggests that cefiderocol CSF concentrations in meningitis exceed MIC of gram negative organisms [104].
Fluoroquinolones
Ciprofloxacin
Delafloxacin
Gatifloxacin
Gemifloxacin
Levofloxacin
Moxifloxacin
Norfloxacin
Ofloxacin
As a group, fluoroquinolones demonstrate excellent CSF penetration. Clinical data are only available for ciprofloxacin, ofloxacin, levofloxacin and moxifloxacin [90].
Glycopeptides
TeicoplaninThe high protein binding of teicoplanin restricts CSF penetration after IV administration [100].
VancomycinVancomycin is highly hydrophilic and may reach sub therapeutic CSF concentration at conventional doses, but adequate concentrations at increased doses [91].
DalbavancinNo clinical data available.
TelavancinNo clinical data available.
Glycylcycline
TigecyclineLimited clinical data available suggest that tigecycline reaches adequate concentrations of inflamed meninges [90].
Lincosamides
Clindamycin
Lincomycin
Lincomycin and its derivative Clindamycin is considered to have poor CSF penetration [91].
Monobactams
AztreonamScant clinical data available suggest that aztreonam reaches sufficient CSF concentrations after systemic administration in inflamed meninges [45].
Macrolides
Azithromycin
Clarithromycin
Erythromycin
Fidaxomicin
Macrolides have been unable to reach therapeutic CSF concentrations in adults [91].
Nitroimidazoles
MetronidazoleGood CSF penetration in both inflamed and no inflamed meninges [90,91].
TinidazoleNo clinical data available.
Oxazolidinones
LinezolidCSF concentrations above the MIC of susceptible pathogens both with inflamed and uninflamed meninges [90].
TedizolidNo clinical data available.
Penicillins
Penicillin G
Penicillin V
Good CSF concentrations after intravenous administration [91].
TemocillinVery limited clinical data available suggest that temocillin may reach therapeutic concentrations in the CSF of patients with gram negative meningitis, but more data are necessary to assess this [105].
Amoxicillin
Ampicillin
Good CSF penetrations after IV administration [91].
CloxacillinPenetrates in CSF of inflamed meninges to a limited extent, therefore higher doses may be necessary to attain therapeutic targets [106]. Furthermore, therapy failure has been described in patients under treatment for Staphylococcus meningitis [107].
FlucloxacillinPenetrates in CSF of inflamed meninges to a limited extent, therefore higher doses may be necessary to attain therapeutic targets [106].
NafcillinInsufficient CSF penetration for treatment of meningitis [91].
OxacillinLimited CSF diffusion at conventional doses [108].
PiperacillinCrosses the inflamed and non-inflamed blood-brain barrier but in unpredictable amounts [109].
TicarcillinsVery limited data available, rather low and variable CSF concentrations after administration of ticarcillin-clavulanate [110].
Polymyxins
Polymyxin B
Polymyxin E
(Colistin)
Limited clinical data available suggest very low CSF penetration after systemic administration [111].
Sulfonamides
SulfamethoxazoleHigh doses achieve good CSF concentrations both with inflamed and uninflamed meninges [90].
Tetracyclines
DoxycyclineLimited clinical data, same CSF penetration in both inflamed and uninflamed meninges [90].
MinocyclineNo clinical data available.
Miscellaneous
ChloramphenicolChloramphenicol penetrates well into CSF, but significant toxicities prohibit the clinical use [90,91].
DaptomycinLimited PK data available on CSF penetration. Some case reports described the successful use of daptomycin in meningitis.
FosfomycinEnters the CSF in the presence and absence of meningeal inflammation [90].

4.3. Metabolism

Metabolism is the process of chemical modification of a drug molecule (substrate) into a hydrophilic metabolite [112]. This is typically needed to eliminate lipophilic drugs, as these do not dissolve in water and therefore preclude renal excretion [113]. Some metabolites are toxic, other substrates only become active after metabolism; these substrates are therefore called pro-drugs.
Drug metabolism is divided in two main phase reactions [114]. Phase I reactions introduce a functional group to the substrate through oxidation, dealkylation, reduction or hydrolysis; therefore, phase 1 reactions are referred to as functionalization. Importantly, substrates can remain pharmacologically active after phase 1 reactions. Phase II reactions inactivate a substrate through adding a polar conjugate, most commonly glucuronide, which facilitates subsequent elimination through urine or bile. The extent of metabolism of a molecule is determined by the molecular structure, as a substrate may undergo either or both phase reactions [115].
Although most metabolism happens in the liver, other sites of metabolism include the kidneys, lung, intestines, brain and muscle [116]. Cytochrome P450 (CYP) is the major enzyme family responsible for phase 1 reactions [117]. Cytochromes are a superfamily of proteins containing heme as a cofactor, with the role of enzymatic metabolism of both endogenous substrates such as steroids or lipids, and of exogenous substrates such as nutrients and drugs [118].
CYP isoenzymes activity changes during lifetime, increasing significantly during the early years of life, when activities ultimately become similar to adult levels [119]. Therefore, lower doses of drugs that require hepatic metabolism may be needed during the early years of life, while a pro-drug can have lower efficacy early in life. To make drug metabolism even more complex, some isoenzymes like CYP3A7 may be more expressed during the early weeks of life or even in the fetus [120]. Furthermore, phase II enzymes may have varying expression during different periods of life. For example, uridine 5′-diphosphoglucuronic acid glucuronosyltransferases (UGT), which is responsible for about 15% of drug metabolism, is less expressed during early weeks of life. Exemplarily, the grey baby syndrome was observed in infants who were treated with the antimicrobial chloramphenicol, as their low UGT activity restricted metabolism, resulting in mitochondrial chloramphenicol toxicity [121].
Clinically relevant drug interactions may occur in the metabolism processes. Drug interactions are best understood by examining the three main actors: (i) the substrate drug, which is usually metabolized by CYP enzymes, (ii) the inducer drug that can increase the synthesis of CYP enzymes and potentially increase the metabolism of a substrate (thus decreasing serum concentrations of the substrate); (iii) the inhibitor drug that can inhibit CYP and potentially decrease the metabolism of a substrate (increasing serum concentrations of the substrate) [122]. For example, rifampicin is a potent CYP inducer that leads to reduced concentrations of substrate, causing subtherapeutic concentrations [123]. On the contrary, CYP inhibitors such as macrolides and isoniazid increase concentrations of the substrate, therefore increasing the chance of toxicity [124]. An important concept is that inhibition processes require hours and therefore has a relatively quick effect on the substrate’s drug concentration, while induction events require nuclear transcriptional effects that take days to weeks [122].

4.4. Elimination

Elimination or excretion (also called clearance, CL) represents the process of removal of drugs or by-products from the body, sometimes following metabolism [115]. The renal and hepatic routes are the most common ways of elimination, other routes include the lungs, intestine and secretory glands such as sweat, saliva and tears. Half-life (t1/2) is a pharmacokinetic parameter of elimination, which is defined as the time to reduce Cmax by 50%. t1/2 is dependent on both Vd and clearance, as is mathematically expressed by the formula t1/2 = 0.693 × Vd/CL [125].
The renal route is the commonest form of eliminating antimicrobials [126] with glomerular filtration rate (GFR) as its major determinant, which is dependent on renal blood flow. On top of GFR, the tubular processes secretion and reabsorption also can be of relevance for elimination [127]. Both maturational and non-maturational factors affect the performance of renal clearance throughout childhood. The maturational changes associated with renal clearance are related to increases in glomerular filtration rate and tubular secretion with age. Glomerular filtration starting from 2 mL/min in a newborn markedly increases in the first year of life, reaching adult rates by 1–2 years of age. Age-related tubular secretion changes occur in the form of an increase in number and isoforms of transporters mediated by an increase in serum glucocorticoids and thyroid hormone, occurring in synchrony with weaning, and show notable increments after five years of age [128].
Multiple non-maturational factors affect renal excretion in children. However, factitious changes should be discerned. For example, trimethoprim competitively interferes with the tubular secretion of creatinine. Therefore, prolonged administration of trimethoprim results in an increase in serum creatinine, without a decreased GFR, due to impaired tubular secretion [129]. Hence, while trimethoprim has a potential to cause nephrotoxicity, isolated increases in serum creatinine should not be taken as an indicator of renal injury. Disease states are also relevant non-maturational factors affecting antimicrobial clearance in children in both directions, as both an increase (hyperfiltration) or decrease (renal impairment) can occur. Augmented renal clearance triggers low plasma concentrations of administered antimicrobials [129]. High cardiac output and the subsequent raised glomerular filtration (>10% increase from normal clearance rates) are associated with the development of augmented renal clearance [130]. Infants and children, especially those in the post-traumatic or post-operative period, having sepsis, burns or hematologic malignancies are at a higher risk of ARC [131,132]. In order to maintain adequate drug exposure in such children, either prolonged infusions, frequent dosing and increased dosing or change to an alternative antimicrobial drug are required [133,134,135].

5. Therapeutic Drug Monitoring

Therapeutic drug monitoring, which refers to individual patient dose adjustments based on measured drug concentrations, is an important tool to optimize therapy. It may be indicated in patients where high peak concentrations are desired in infections caused by organisms with high MICs, or in patients who are receiving antimicrobials that exert dose-dependent toxicities such as nephrotoxicity. In the case of aminoglycosides, which are nephrotoxic drugs, monitoring through concentrations could limit toxicity. Additionally in cases of treating organisms with higher MICs there may be a need to achieve higher peak concentrations since Cmax/MIC is the PK/PD parameter of efficacy associated with aminoglycosides. Furthermore, pharmacokinetic variability exists with many drugs, this compounded with physiological and anatomical changes seen in pediatric patients further supports the need for therapeutic drug monitoring. Many studies have recommended therapeutic drug monitoring to address the pharmacokinetic variability seen with antituberculosis agents [136,137,138]. Newer drug monitoring approaches are aimed at achieving targeted AUC as opposed to single trough concentrations. Such practices are apt for drugs that utilize AUC/MIC as a PK/PD parameter, such as vancomycin. Newer vancomycin guidelines have proposed targeted therapy by achieving appropriate AUC [139]. Model-based approaches have allowed for easier computing of the AUC for individualization of therapy but requires a drug concentration for dose prediction [139]. Notably, Ewoldt et al. in a recent study reported no beneficial effect of model informed precision dosing of beta-lactam antibiotics and ciprofloxacin on ICU length of stay in critically ill patients however, the study was conducted in adult patients and did not include the pediatric population [140]. In the case of aminoglycosides, which are nephrotoxic drugs, monitoring trough concentrations during the treatment course could limit toxicity. Additionally in cases of treating organisms with higher MICs there may be a need to achieve higher peak concentrations since Cmax/MIC is the PK/PD parameter of efficacy associated with aminoglycosides.

6. Challenges in Attaining Effective Drug Concentrations in Children Living in LMIC

Just under half (45%) of all deaths in children under 5 years of age globally are attributed to undernutrition. Infections are responsible for a large proportion of mortality in malnourished children, due to reduced serum immunoglobulin levels, atrophied immune tissues like the thymus, and impaired epithelial mucosal barriers, such as the skin, respiratory and intestinal tract [141,142]. Therefore, the World Health Organization recommends that all children admitted with severe acute malnutrition receive broad-spectrum antimicrobials. However, different comorbidities and restrictions in resources make it challenging to attain PK-PD targets in LMIC settings. There are relatively few studies available that investigated the effects of protein-energy malnutrition on the pharmacokinetics of medicines in children [143]. While a complete overview of the effects of malnutrition on antimicrobial pharmacology is beyond the scope of this review, it is important for clinicians to recognize that the severity of malnutrition and characteristics of the drug will impact pharmacokinetics. In general, the volume of distribution and drug disposition will be increased for hydrophilic drugs and decreased for lipophilic drugs in severe malnutrition [144]. In addition, alterations in total body water, muscle mass and serum protein concentrations in malnourished children will further impact drug delivery [145]. Dose modifications of enteral medications may be necessary to account for impaired absorption and the reduced total drug clearance associated with malnourishment [146]. Furthermore, hypoalbuminemia associated with severe acute malnutrition can result in hydrophilic drugs having larger volumes of distribution. Therefore, higher doses may be necessary to attain adequate serum concentrations to treat blood stream infections [147,148].

6.1. Co-Morbidities

Apart from the effects of malnutrition, many diseases in LMIC may alter the pharmacokinetics of antimicrobials. The faster intestinal transport of oral antimicrobials during prolonged diarrhea leads to reduced absorption [149]. Furthermore, HIV and tuberculosis are common diseases in LMIC that both require drugs affecting P-gp. Rifampicin is an inducer, while protease inhibitors are either inhibitors or substrates of P-gp, which potentially results in drug-drug interactions [150]. Augmented renal clearance and chronic kidney disease are commonly observed in children with sickle cell disease that may warrant dose modifications to attain therapeutic targets and avoid toxicity [151,152]. Causes of childhood kidney disease in developing countries are diverse and mainly relate to antecedent post streptococcal glomerulonephritis, dehydration, malaria, use of herbal medicines and lead to various syndromes like hemolytic uremic syndrome, acute tubular necrosis and glomerulonephritis. A full understanding of the spectrum of etiologies is hampered by limited diagnostics [153,154]. A reduced creatinine clearance was also observed in children living with HIV in a population pharmacokinetic study of levofloxacin among South African children receiving treatment for multi-drug resistant tuberculosis [155]. Renal dysfunction associated with HIV infection is driven by direct renal parenchymal infection and immune-complex deposition [156].

6.2. Altered Polymorphisms

Besides disease related alterations, polymorphisms in drug metabolizing enzymes may further affect the pharmacokinetics of antimicrobials in affected patients. As an example, isoniazid is included in both prophylactic and therapeutic regimens against tuberculosis while it is widely appreciated that this has potential severe side effects. Its metabolism is highly dependent on the individual acetylation profile of the N-acetyltransferase (NAT2) gene. There is robust evidence that NAT-related polymorphisms already impact isoniazid clearance from neonatal life onwards, as the metabolic activity increases steadily from 4 months until 17 years of age (r = 0.53, age range 4 months to 17 years, 25/88 cases were 4–23 months) [157]. Slow genotypes (no alleles) had a much lower metabolic ratio compared to rapid (two alleles) genotypes (2-fold difference). Schaaf et al. estimated the first order elimination rate constant in 64 children [158], which related both to age and NAT-2 allele frequency (SS = 0.254; FS = 0.51; FF = 0653 h−1). Finally, Zhu et al. quantified the pharmacogenetic specific NAT2 enzyme maturation in perinatal HIV exposed infants receiving isoniazid [159]. Consecutive plasma concentration-time measurements of isoniazid from 151 infants (starting at 3–4 months of age) receiving isoniazid 10 to 20 mg/kg/day orally during the 24-month study were incorporated in a population analysis along with NAT2 genotype, body weight, age, and sex. For fast (FF) and intermediate (SF) acetylators, clearance increased from 14.25 L/h. 70 kg and 10.88 L/h. 70 kg at 3 months to 22.84 L/h. 70 kg and 15.58 L/h. 70 kg at 24 months, while slow (SS) acetylators displayed no changes over age (7.35 L/h). Comparing slow to fast acetylators, there is a 2-fold difference at 3 months, to further increase to a 3-fold difference at 24 months. How to implement such information to attain a ‘precision approach’ within a LMIC remains an issue, but perhaps awareness and considering concentration guided dosing shortly after initiation could be a way forward.

6.3. Challenges with Healthcare Administration

Administrative challenges in hospitals in LMIC include work over-load on behalf of pediatric nurses, who often work in crowded wards, which potentially leads to errors in dosing. Furthermore, skipped doses are often-unnoticed [160]. Under-dosing or inappropriate frequency of antimicrobial dosing will inevitably promote AMR [161]. Limitations in therapeutic drug monitoring in many low-income nations need to be overcome to combat in particular resistance to glycopeptide and aminoglycoside antibiotics [162,163]. LMIC face many challenges due to limited resources and the cost associated with effectively running such facilities. Due to the rural settings of most clinics and hospitals, samples need to be transported over vast distances to reach therapeutic drug monitoring laboratories if one is available, adequate storage of samples are impeded by lack of equipment and conveying of results may be delayed due to the lack of network signal. Addressing these issues would require larger stakeholder engagement with governmental involvement to set up infrastructure, adequately train staff in conducting and interpretation of therapeutic drug monitoring, and devise ways to minimize high-cost burden such as monitoring in selected patients with limited sampling and the use of model-based precision dosing. In the long term, adequate monitoring practices could off-set the cost associated with lack of drug efficacy and/or drug toxicity in the absence of adequate monitoring.
Overall, the practice of infectious diseases and clinical microbiology is hampered by limited clinical bacteriology laboratories, shortage of pharmacokinetic and pharmacodynamics studies as well as lack of access to therapeutic drug monitoring [162,164].

7. Conclusions

A thorough understanding of developmental pharmacokinetics is pivotal for adequate dosing in children. In this review, we outlined the effects of growth and maturation on pharmacokinetics in children. Subsequently, we elaborated how common comorbidities, such as malnourishment, co-morbidities such as tuberculosis, augmented renal clearance, HIV and tuberculosis, in LMIC may further affect the pharmacokinetics of antimicrobials in children. Limited resources for therapeutic drug monitoring restrict the abilities to individualize doses based on measured concentrations. Further pharmacokinetic studies of antimicrobials in children in LMIC are urgently needed to optimize dosing, and hence to attain PK/PD targets and combat antimicrobial resistance.

Author Contributions

Conceptualization, K.M. and K.A.; literature research, K.A., T.A., S.B., D.B., E.H.D., V.P.-F.L., K.J.D. and K.A.; writing—original draft preparation, K.M., T.A., S.B., D.B. and V.P.-F.L.; writing—review and editing, E.H.D., V.P.-F.L., K.J.D. and K.A.; supervision, E.H.D., V.P.-F.L., K.J.D. and K.A. All authors have read and agreed to the published version of the manuscript.

Funding

K.J.D. is supported by the Eunice Kennedy Shriver National Institute of Child Health & Human Development of the National Institutes of Health under Award Number K23HD091365. The other authors did not receive external funding pertinent to this research.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We acknowledge all who have contributed to the online course ‘Pharmacokinetics of Antibiotics’ that was organized by the World Society for Pediatric Infectious Diseases (WSPID). It was this initiative that resulted in the concept of this manuscript, which addresses an important topic in LMIC. WSPID hosts a global e-learning portal (https://edu.wspid.org) that offers educational resources, free of charge, to the global pediatric infectious diseases community.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AMR: antimicrobial resistance. AUC: area under the curve. CL: clearance. Cmax: maximum concentration. CNS: central nervous system. CSF: cerebrospinal fluid. CYP: cytochrome P-450. GFR: glomerular filtration rate. GI: gastrointestinal. HIV: human immunodeficiency virus. LMIC: low and middle income countries. LogP: logarithm of the partition coefficient. MTC: minimum toxic concentration. NAT2: N-acetyltransferase 2. P-gp: p-glycoprotein. pKa: acid-base dissociation constant. PK/PD: pharmacokinetic-pharmacodynamic. t1/2: half-life. UGT: uridine 5′-diphospho-glucuronosyltransferase. Vd: volume of distribution.

References

  1. Strong, K.L.; Pedersen, J.; Johansson, E.W.; Cao, B.; Diaz, T.; Guthold, R.; You, D.; Requejo, J.; Liu, L. Patterns and Trends in Causes of Child and Adolescent Mortality 2000–2016: Setting the Scene for Child Health Redesign. BMJ Glob. Health 2021, 6, e004760. [Google Scholar] [CrossRef] [PubMed]
  2. Perin, J.; Mulick, A.; Yeung, D.; Villavicencio, F.; Lopez, G.; Strong, K.L.; Prieto-Merino, D.; Cousens, S.; Black, R.E.; Liu, L. Global, Regional, and National Causes of under-5 Mortality in 2000–19: An Updated Systematic Analysis with Implications for the Sustainable Development Goals. Lancet Child Adolesc. Health 2022, 6, 106–115. [Google Scholar] [CrossRef] [PubMed]
  3. Le Doare, K.; Bielicki, J.; Heath, P.T.; Sharland, M. Systematic Review of Antibiotic Resistance Rates among Gram-Negative Bacteria in Children with Sepsis in Resource-Limited Countries. J. Pediatr. Infect. Dis. Soc. 2015, 4, 11–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Murray, C.J.; Ikuta, K.S.; Sharara, F.; Swetschinski, L.; Robles Aguilar, G.; Gray, A.; Han, C.; Bisignano, C.; Rao, P.; Wool, E.; et al. Global Burden of Bacterial Antimicrobial Resistance in 2019: A Systematic Analysis. Lancet 2022, 399, 629–655. [Google Scholar] [CrossRef] [PubMed]
  5. Basu, S.; Copana, R.; Morales, R.; Anugulruengkitt, S.; Puthanakit, T.; Maramba-Lazarte, C.; Williams, P.; Musembi, J.; Boga, M.; Issack, M.; et al. Keeping It Real: Antibiotic Use Problems and Stewardship Solutions in Low- and Middle-Income Countries. Pediatr. Infect. Dis. J. 2022, 41, S18–S25. [Google Scholar] [CrossRef]
  6. Villanueva, P.; Coffin, S.E.; Mekasha, A.; McMullan, B.; Cotton, M.F.; Bryant, P.A. Comparison of Antimicrobial Stewardship and Infection Prevention and Control Activities and Resources between Low-/Middle- and High-Income Countries. Pediatr. Infect. Dis. J. 2022, 41, S3–S9. [Google Scholar] [CrossRef]
  7. Gebretekle, G.B.; Haile Mariam, D.; Abebe Taye, W.; Mulu Fentie, A.; Amogne Degu, W.; Alemayehu, T.; Beyene, T.; Libman, M.; Gedif Fenta, T.; Yansouni, C.P.; et al. Half of Prescribed Antibiotics Are Not Needed: A Pharmacist-Led Antimicrobial Stewardship Intervention and Clinical Outcomes in a Referral Hospital in Ethiopia. Front. Public Health 2020, 8, 109. [Google Scholar] [CrossRef] [Green Version]
  8. Guiastrennec, B.; Ramachandran, G.; Karlsson, M.O.; Kumar, A.K.H.; Bhavani, P.K.; Gangadevi, N.P.; Swaminathan, S.; Gupta, A.; Dooley, K.E.; Savic, R.M. Suboptimal Antituberculosis Drug Concentrations and Outcomes in Small and HIV-Coinfected Children in India: Recommendations for Dose Modifications. Clin. Pharmacol. Ther. 2018, 104, 733–741. [Google Scholar] [CrossRef]
  9. Odenholt, I.; Gustafsson, I.; Löwdin, E.; Cars, O. Suboptimal Antibiotic Dosage as a Risk Factor for Selection of Penicillin-Resistant Streptococcus pneumoniae: In Vitro Kinetic Model. Antimicrob. Agents Chemother. 2003, 47, 518–523. [Google Scholar] [CrossRef] [Green Version]
  10. European Committee on Antimicrobial Susceptibility Testing. Available online: https://www.eucast.org/clinical_breakpoints/ (accessed on 6 June 2022).
  11. Onufrak, N.J.; Forrest, A.; Gonzalez, D. Antibiotics PK/PD. Clin. Ther. 2016, 38, 1930–1947. [Google Scholar] [CrossRef]
  12. Turfus, S.C.; Delgoda, R.; Picking, D.; Gurley, B.J. Chapter 25—Pharmacokinetics. In Pharmacognosy; Badal, S., Delgoda, R., Eds.; Academic Press: Cambridge, MA, USA, 2017; pp. 495–512. [Google Scholar] [CrossRef]
  13. Rodríguez-Gascón, A.; Solinís, M.Á.; Isla, A. The Role of Pk/Pd Analysis in the Development and Evaluation of Antimicrobials. Pharmaceutics 2021, 13, 833. [Google Scholar] [CrossRef]
  14. Mansoor, A.; Mahabadi, N. Volume of Distribution. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK545280/ (accessed on 2 September 2022).
  15. Wanat, K. Biological Barriers, and the Influence of Protein Binding on the Passage of Drugs across Them. Mol. Biol. Rep. 2020, 47, 3221–3231. [Google Scholar] [CrossRef] [Green Version]
  16. Gaohua, L.; Miao, X.; Dou, L. Crosstalk of Physiological PH and Chemical pKa under the Umbrella of Physiologically Based Pharmacokinetic Modeling of Drug Absorption, Distribution, Metabolism, Excretion, and Toxicity. Expert Opin. Drug Metab. Toxicol. 2021, 17, 1103–1124. [Google Scholar] [CrossRef]
  17. Deb, P.K.; Al-Attraqchi, O.; Prasad, M.R.; Tekade, R.K. Protein and Tissue Binding: Implication on Pharmacokinetic Parameters. Implication on Pharmacokinetic Parameters; Elsevier: Amsterdam, The Netherlands, 2018; ISBN 9780128144244. [Google Scholar]
  18. Gao, Y.; Gesenberg, C.; Zheng, W. Oral Formulations for Preclinical Studies: Principle, Design, and Development Considerations; Elsevier: Amsterdam, The Netherlands, 2017; ISBN 9780128024478. [Google Scholar]
  19. Manallack, D.T. The pKa Distribution of Drugs: Application to Drug Discovery. Perspect. Med. Chem. 2007, 1, 1177391X0700100. [Google Scholar] [CrossRef]
  20. PubChem. Available online: https://pubchem.ncbi.nlm.nih.gov/ (accessed on 4 September 2022).
  21. DrugBank Online. Available online: https://go.drugbank.com/ (accessed on 7 September 2022).
  22. Asín-prieto, E.; Rodríguez-gasc, A. Applications of the Pharmacokinetic/Pharmacodynamic (PK/PD) Analysis of Antimicrobial Agents. J. Infect. Chemother. 2015, 21, 319–329. [Google Scholar] [CrossRef]
  23. PubChem. Neomycin (Compound). Available online: https://pubchem.ncbi.nlm.nih.gov/compound/8378 (accessed on 22 September 2022).
  24. Gumbo, T.; Angulo-barturen, I.; Ferrer-bazaga, S. Pharmacokinetic-Pharmacodynamic and Dose-Response Relationships of Antituberculosis Drugs: Recommendations and Standards for Industry and Academia. J. Infect. Dis. 2015, 211, S96–S106. [Google Scholar] [CrossRef] [Green Version]
  25. Sekaggya-wiltshire, C.; Dooley, K.E. Expert Opinion on Drug Metabolism & Toxicology Pharmacokinetic and Pharmacodynamic Considerations of Rifamycin Antibiotics for the Treatment of Tuberculosis. Expert Opin. Drug Metab. Toxicol. 2019, 15, 615–618. [Google Scholar] [CrossRef] [Green Version]
  26. WHO. Guideline Updates on the Management of Severe Acute Malnutrition in Infants and Children; World Health Organization: Geneva, Switzerland, 2013. [Google Scholar]
  27. PubChem. Cycloserine (Compound). Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Cycloserine#section=Absorption-Distribution-and-Excretion (accessed on 3 October 2022).
  28. Deshpande, D.; Pasipanodya, J.G.; Mpagama, S.G.; Srivastava, S.; Bendet, P.; Koeuth, T.; Lee, P.S.; Heysell, S.K.; Gumbo, T. Ethionamide Pharmacokinetics/Pharmacodynamics- Derived Dose, the Role of MICs in Clinical Outcome, and the Resistance Arrow of Time in Multidrug-Resistant Tuberculosis. Clin. Infect. Dis. 2018, 67, S317–S326. [Google Scholar] [CrossRef]
  29. Adembri, C.; Novelli, A.; Nobili, S. Some Suggestions from PK/PD Principles to Contain Resistance in the Clinical Setting—Focus on ICU Patients and Gram-Negative Strains. Antibiotics 2020, 9, 676. [Google Scholar] [CrossRef]
  30. Iyer, R.N. Beta Lactam. In Comprehensive Pharmacology; Elsevier: Amsterdam, The Netherlands, 2022; pp. 3–63. [Google Scholar]
  31. DrugBank. Online Monograph Vaborbactam. Available online: https://go.drugbank.com/drugs/DB12107 (accessed on 27 September 2022).
  32. DrugBank. Online Monograph Doripenem. Available online: https://go.drugbank.com/drugs/DB06211 (accessed on 27 September 2022).
  33. Landersdorfer, C.B.; Nation, R.L. Limitations of Antibiotic MIC-Based PK-PD Metrics: Looking Back to Move Forward. Front. Pharmacol. 2021, 12, 770518. [Google Scholar] [CrossRef]
  34. Cohen, R.; Grimprel, E. Antibiotic Pharmacokinetic and Pharmacodynamic Parameters in Pediatric Clinical Practice. Arch. Pédiatrie 2017, 24, S6–S8. [Google Scholar] [CrossRef] [PubMed]
  35. Dellamonica, P. Cefuroxime Axetil. Int. J. Antimicrob. Agents 1994, 4, 23–36. [Google Scholar] [CrossRef] [PubMed]
  36. Jongmans, C.; Muller, A.E.; Van Den Broek, P.; Cruz De Almeida, B.D.M.; Van Den Berg, C.; 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. [Google Scholar] [CrossRef] [PubMed]
  37. Cefmetazole. Available online: http://www.antimicrobe.org/drugpopup/Cefmetazole.htm (accessed on 29 September 2022).
  38. Padda, I.S.; Nagalli, S. Cefotaxime. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK560653/ (accessed on 29 September 2022).
  39. Ramón, J.; Perea, A. Ceftobiprole Review Ceftobiprole: Pharmacokinetics and PK/PD Profile. Rev. Española Quimioter. 2019, 32, 11–16. [Google Scholar]
  40. Prats, G.; Rossi, V.; Salvatori, E.; Mirelis, B. Prulifloxacin: A New Antibacterial Fluoroquinolone. Expert Rev. Anti-Infect. Ther. 2006, 4, 27–41. [Google Scholar] [CrossRef]
  41. Wenzler, E.; Liao, S.; Rodvold, K.A. Pharmacodynamics of Lipoglycopeptides. In Antibiotic Pharmacodynamics; Humana: Towota, NJ, USA, 2016; pp. 285–315. [Google Scholar]
  42. DrugBank Online. Teicoplanin. Available online: https://go.drugbank.com/drugs/DB06149 (accessed on 1 October 2022).
  43. Dalbavancin for Injection, Product Monograph. Available online: https://pdf.hres.ca/dpd_pm/00060897.PDF (accessed on 1 October 2022).
  44. DrugBank Online. Clindamycin. Available online: https://go.drugbank.com/drugs/DB01190 (accessed on 1 October 2022).
  45. Ramsey, C.; Macgowan, A.P. A Review of the Pharmacokinetics and Pharmacodynamics of Aztreonam. J. Antimicrob. Chemother. 2016, 71, 2704–2712. [Google Scholar] [CrossRef] [Green Version]
  46. Child, J.; Chen, X.; Mistry, R.D.; Somme, S.; Macbrayne, C.; Anderson, P.L.; Jones, R.N.; Parker, S.K. Pharmacokinetic and Pharmacodynamic Properties of Metronidazole in Pediatric Patients with Acute Appendicitis: A Prospective Study. J. Pediatr. Infect. Dis. Soc. 2019, 8, 297–302. [Google Scholar] [CrossRef]
  47. Ampicillin. Available online: https://web.archive.org/web/20150712001731/http:/www.drugs.com/monograph/ampicillin.html#r9 (accessed on 10 October 2022).
  48. Fidaxomicin (DificidTM) Monograph. Available online: https://www.pbm.va.gov/PBM/clinicalguidance/drugmonographs/FidaxomicinMonograph.doc (accessed on 12 October 2022).
  49. Oxacillin—Drug Summary. Available online: https://www.pdr.net/drug-summary/Oxacillin-oxacillin-3066 (accessed on 10 October 2022).
  50. Alexandre, K.; Fantin, B. Pharmacokinetics and Pharmacodynamics of Temocillin. Clin. Pharmacokinet. 2018, 57, 287–296. [Google Scholar] [CrossRef]
  51. Al-Shaer, M.H.; Alghamdi, W.A.; Graham, E.; Peloquin, C.A. Meropenem, Cefepime, and Piperacillin Protein Binding in Patient Samples. Ther. Drug Monit. 2020, 42, 129–132. [Google Scholar] [CrossRef]
  52. Sulfadiazine. Available online: http://www.antimicrobe.org/drugpopup/sulfadiazine.htm (accessed on 6 October 2022).
  53. Sulfadoxine. Available online: http://www.antimicrobe.org/drugpopup/sulfadoxine.htm (accessed on 6 October 2022).
  54. Agwuh, K.N.; Macgowan, A. Pharmacokinetics and Pharmacodynamics of the Tetracyclines Including Glycylcyclines. J. Antimicrob. Chemother. 2006, 58, 256–265. [Google Scholar] [CrossRef]
  55. Lee, B.L.; Sachdeva, M.; Chambers, H.F. Effect of Protein Binding of Daptomycin on MIC and Antibacterial Activity. Antimicrob. Agents Chemother. 1991, 35, 2505–2508. [Google Scholar] [CrossRef] [Green Version]
  56. Patel, S.; Saw, S. Daptomycin. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK470407/ (accessed on 8 October 2022).
  57. Van Heeswijk, R.P.G.; Dannemann, B.; Hoetelmans, R.M.W. Bedaquiline: A Review of Human Pharmacokinetics and Drug–Drug Interactions. J. Antimicrob. Chemother. 2014, 69, 2310–2318. [Google Scholar] [CrossRef]
  58. Hoffmann, M.; DeMaio, W.; Jordan, R.A.; Talaat, R.; Harper, D.; Speth, J.; Scatina, J. Metabolism, Excretion, and Pharmacokinetics of [14C]Tigecycline, a First-in-Class Glycylcycline Antibiotic, after Intravenous Infusion to Healthy Male Subjects. Drug Metab. Dispos. Biol. Fate Chem. 2007, 35, 1543–1553. [Google Scholar] [CrossRef] [Green Version]
  59. Doogue, M.P.; Polasek, T.M. The ABCD of Clinical Pharmacokinetics. Ther. Adv. Drug Saf. 2013, 4, 5–7. [Google Scholar] [CrossRef] [Green Version]
  60. 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]
  61. Jamei, M.; Turner, D.; Yang, J.; Neuhoff, S.; Polak, S.; Rostami-Hodjegan, A.; Tucker, G. Population-Based Mechanistic Prediction of Oral Drug Absorption. AAPS J. 2009, 11, 225–237. [Google Scholar] [CrossRef] [Green Version]
  62. Debotton, N.; Dahan, A. A Mechanistic Approach to Understanding Oral Drug Absorption in Pediatrics: An Overview of Fundamentals. Drug Discov. Today 2014, 19, 1322–1336. [Google Scholar] [CrossRef]
  63. Kearns, G.L.; Abdel-Rahman, S.M.; Alander, S.W.; Blowey, D.L.; Leeder, J.S.; Kauffman, R.E. Developmental Pharmacology—Drug Disposition, Action, and Therapy in Infants and Children. New Engl. J. Med. 2003, 349, 1157–1167. [Google Scholar] [CrossRef]
  64. Reiter, P.D. Neonatal Pharmacology and Pharmacokinetics. NeoReviews 2002, 3, e229–e236. [Google Scholar] [CrossRef]
  65. Strolin, M.; 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]
  66. Deng, J.; Zhu, X.; Chen, Z.; Fan, C.H.; Kwan, H.S.; Wong, C.H.; Shek, K.Y.; Zuo, Z.; Lam, T.N. A Review of Food–Drug Interactions on Oral Drug Absorption. Drugs 2017, 77, 1833–1855. [Google Scholar] [CrossRef] [PubMed]
  67. Mooij, M.G.; De Koning, B.A.; Huijsman, M.L.; De Wildt, S.N. Ontogeny of Oral Drug Absorption Processes in Children. Expert Opin. Drug Metab. Toxicol. 2012, 8, 1293–1303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Keij, F.M.; Kornelisse, R.F.; Hartwig, N.G.; Reiss, I.K.M.; Allegaert, K.; Tramper-Stranders, G.A. Oral Antibiotics for Neonatal Infections: A Systematic Review and Meta-Analysis. J. Antimicrob. Chemother. 2019, 74, 3150–3161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Foulds, G.; Luke, D.R.; Teng, R.; Willavize, S.A.; Friedman, H.; Curatolo, W.J. The Absence of an Effect of Food on the Bioavailability of Azithromycin Administered as Tablets, Sachet or Suspension. J. Antimicrob. Chemother. 1996, 37, 37–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Eshelman, F.N.; Spyker, D.A. Pharmacokinetics of Amoxicillin and Ampicillin: Crossover Study of the Effect of Food. Antimicrob. Agents Chemother. 1978, 14, 539–543. [Google Scholar] [CrossRef] [Green Version]
  71. Augmentin® (Amoxicillin/Clavulanate Potassium) Prescribing Information. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2008/050564s051lbl.pdf (accessed on 5 October 2022).
  72. Fassbender, M.; Lode, H.; Schaberg, T.; Borner, K.; Koeppe, P. Pharmacokinetics of New Oral Cephalosporins, Including a New Carbacephem. Clin. Infect. Dis. 1993, 16, 646–653. [Google Scholar] [CrossRef]
  73. Williams, P.E.O.; Harding, S.M. The Absolute Bioavailability of Oral Cefuroxime Axetil in Male and Female Volunteers after Fasting and after Food. J. Antimicrob. Chemother. 1984, 13, 191–196. [Google Scholar] [CrossRef]
  74. Sommers, D.; Wyk, M.; Moncrieff, J.; Schoeman, H. Influence of Food and Reduced Gastric Acidity on the Bioavailability of Bacampicillin and Cefuroxime Axetil. Br. J. Clin. Pharmacol. 1984, 18, 535–539. [Google Scholar] [CrossRef] [Green Version]
  75. Neuvonen, P.J.; Kivistö, K.T.; Lehto, P. Interference of Dairy Products with the Absorption of Ciprofloxacin. Clin. Pharmacol. Ther. 1991, 50, 498–502. [Google Scholar] [CrossRef]
  76. Spenard, J.; Aumais, C.; Massicotte, J.; Brunet, J.-S.; Tremblay, C.; Grace, M.; Lefebvre, M. Effects of Food and Formulation on the Relative Bioavailability of Bismuth Biskalcitrate, Metronidazole, and Tetracycline given for Helicobacter Pylori Eradication. Br. J. Clin. Pharmacol. 2005, 60, 374–377. [Google Scholar] [CrossRef]
  77. Peloquin, C.A.; Namdar, R.; Singleton, M.D.; Nix, D.E. Pharmacokinetics of Rifampin Under Fasting Conditions, with Food, and with Antacids. Chest 1999, 115, 12–18. [Google Scholar] [CrossRef]
  78. Bushra, R.; Aslam, N.; Khan, A. Food Drug Interactions. Oman Med. J. 2011, 26, 77–83. [Google Scholar] [CrossRef]
  79. Rutter, N. Percutaneous Drug Absorption in the Newborn: Hazards and Uses. Clin. Perinatol. 1987, 14, 911–930. [Google Scholar] [CrossRef]
  80. Brunton, L.L.; Hilal-Dandan, R.; Knollmann, B.C. Goodman & Gilman’s The Pharmacological Basis of Therapeutics, 13th ed.; Shanahan, J.F., Lebowitz, H., Eds.; McGraw-Hill Education: New York, NY, USA, 2018; ISBN 978-1-25-958473-9. [Google Scholar]
  81. Shah, S.; Barton, G.; Fischer, A. Pharmacokinetic Considerations and Dosing Strategies of Antibiotics in the Critically Ill Patient. J. Intensive Care Soc. 2015, 16, 147–153. [Google Scholar] [CrossRef] [Green Version]
  82. Katzung, B.G. Basic and Clinical Pharmacology, 14th ed.; McGraw-Hill: New York, NY, USA, 2018; ISBN 978-1-259-64115-2. [Google Scholar]
  83. Batchelor, H.K.; Marriott, J.F. Paediatric Pharmacokinetics: Key Considerations. Br. J. Clin. Pharmacol. 2015, 79, 395–404. [Google Scholar] [CrossRef] [Green Version]
  84. McNamara, P.J.; Alcorn, J. Protein Binding Predictions in Infants. AAPS PharmSci 2002, 4, 19–26. [Google Scholar] [CrossRef] [Green Version]
  85. Ulldemolins, M.; Roberts, J.A.; Rello, J.; Paterson, D.L.; Lipman, J. The Effects of Hypoalbuminaemia on Optimizing Antibacterial Dosing in Critically Ill Patients. Clin. Pharmacokinet. 2011, 50, 99–110. [Google Scholar] [CrossRef]
  86. Saunders, N.R.; Liddelow, S.A.; Dziegielewska, K.M. Barrier Mechanisms in the Developing Brain. Front. Pharmacol. 2012, 3, 46. [Google Scholar] [CrossRef] [Green Version]
  87. Verscheijden, L.F.M.; van Hattem, A.C.; Pertijs, J.C.L.M.; de Jongh, C.A.; Verdijk, R.M.; Smeets, B.; Koenderink, J.B.; Russel, F.G.M.; de Wildt, S.N. Developmental Patterns in Human Blood–Brain Barrier and Blood–Cerebrospinal Fluid Barrier ABC Drug Transporter Expression. Histochem. Cell Biol. 2020, 154, 265–273. [Google Scholar] [CrossRef]
  88. Lam, J.; Baello, S.; Iqbal, M.; Kelly, L.E.; Shannon, P.T.; Chitayat, D.; Matthews, S.G.; Koren, G. The Ontogeny of P-Glycoprotein in the Developing Human Blood–Brain Barrier: Implication for Opioid Toxicity in Neonates. Pediatr. Res. 2015, 78, 417–421. [Google Scholar] [CrossRef]
  89. Haritova, A. A role of p-glycoprotein in modulation of antibiotic pharmacokinetics. Trakia J. Sci. 2008, 6, 1–6. [Google Scholar]
  90. Nau, R.; Seele, J.; Djukic, M.; Eiffert, H. Pharmacokinetics and Pharmacodynamics of Antibiotics in Central Nervous System Infections. Curr. Opin. Infect. Dis. 2018, 31, 57–68. [Google Scholar] [CrossRef] [PubMed]
  91. Sullins, A.K.; Abdel-Rahman, S.M. Pharmacokinetics of Antibacterial Agents in the CSF of Children and Adolescents. Pediatr. Drugs 2013, 15, 93–117. [Google Scholar] [CrossRef] [PubMed]
  92. Donald, P.R. Cerebrospinal Fluid Concentrations of Antituberculosis Agents in Adults and Children. Tuberculosis 2010, 90, 279–292. [Google Scholar] [CrossRef] [PubMed]
  93. Crabol, Y.; Catherinot, E.; Veziris, N.; Jullien, V.; Lortholary, O. Rifabutin: Where Do We Stand in 2016? J. Antimicrob. Chemother. 2016, 71, 1759–1771. [Google Scholar] [CrossRef] [Green Version]
  94. Upton, C.M.; Steele, C.I.; Maartens, G.; Diacon, A.H.; Wiesner, L.; Dooley, K.E. Pharmacokinetics of Bedaquiline in Cerebrospinal Fluid (CSF) in Patients with Pulmonary Tuberculosis (TB). J. Antimicrob. Chemother. 2022, 77, 1720–1724. [Google Scholar] [CrossRef]
  95. de Castro, R.R.; do Carmo, F.A.; Martins, C.; Simon, A.; de Sousa, V.P.; Rodrigues, C.R.; Cabral, L.M.; Sarmento, B. Clofazimine Functionalized Polymeric Nanoparticles for Brain Delivery in the Tuberculosis Treatment. Int. J. Pharm. 2021, 602, 120655. [Google Scholar] [CrossRef]
  96. Kempker, R.R.; Smith, A.G.C.; Avaliani, T.; Gujabidze, M.; Bakuradze, T.; Sabanadze, S.; Avaliani, Z.; Collins, J.M.; Blumberg, H.M.; Alshaer, M.H.; et al. Cycloserine and Linezolid for Tuberculosis Meningitis: Pharmacokinetic Evidence of Potential Usefulness. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2022, 75, 682–689. [Google Scholar] [CrossRef]
  97. Tucker, E.W.; Pieterse, L.; Zimmerman, M.D.; Udwadia, Z.F.; Peloquin, C.A.; Gler, M.T.; Ganatra, S.; Tornheim, J.A.; Chawla, P.; Caoili, J.C.; et al. Delamanid Central Nervous System Pharmacokinetics in Tuberculous Meningitis in Rabbits and Humans. Antimicrob. Agents Chemother. 2019, 63, e00913-19. [Google Scholar] [CrossRef] [Green Version]
  98. Bakken, J.S.; Bruun, J.N.; Gaustad, P.; Tasker, T.C. Penetration of Amoxicillin and Potassium Clavulanate into the Cerebrospinal Fluid of Patients with Inflamed Meninges. Antimicrob. Agents Chemother. 1986, 30, 481–484. [Google Scholar] [CrossRef] [Green Version]
  99. Münch, R.; Lüthy, R.; Blaser, J.; Siegenthaler, W. Human Pharmacokinetics and CSF Penetration of Clavulanic Acid. J. Antimicrob. Chemother. 1981, 8, 29–37. [Google Scholar] [CrossRef]
  100. Nau, R.; Sörgel, F.; Eiffert, H. Penetration of Drugs through the Blood-Cerebrospinal Fluid/Blood-Brain Barrier for Treatment of Central Nervous System Infections. Clin. Microbiol. Rev. 2010, 23, 858–883. [Google Scholar] [CrossRef] [Green Version]
  101. Cies, J.J.; Moore, W.S.; Enache, A.; Chopra, A. Ceftaroline Cerebrospinal Fluid Penetration in the Treatment of a Ventriculopleural Shunt Infection: A Case Report. J. Pediatr. Pharmacol. Ther. JPPT Off. J. PPAG 2020, 25, 336–339. [Google Scholar] [CrossRef]
  102. Balouch, M.A.; Bajwa, R.J.; Hassoun, A. Successful Use of Ceftaroline for the Treatment of MRSA Meningitis Secondary to an Infectious Complication of Lumbar Spine Surgery. J. Antimicrob. Chemother. 2015, 70, 624–625. [Google Scholar] [CrossRef] [Green Version]
  103. Kuriakose, S.S.; Rabbat, M.; Gallagher, J.C. Ceftaroline CSF Concentrations in a Patient with Ventriculoperitoneal Shunt-Related Meningitis. J. Antimicrob. Chemother. 2015, 70, 953–954. [Google Scholar] [CrossRef] [Green Version]
  104. Kufel, W.D.; Abouelhassan, Y.; Steele, J.M.; Gutierrez, R.L.; Perwez, T.; Bourdages, G.; Nicolau, D.P. Plasma and Cerebrospinal Fluid Concentrations of Cefiderocol during Successful Treatment of Carbapenem-Resistant Acinetobacter baumannii Meningitis. J. Antimicrob. Chemother. 2022, 77, 2737–2741. [Google Scholar] [CrossRef]
  105. Lupia, T.; De Benedetto, I.; Stroffolini, G.; Di Bella, S.; Mornese Pinna, S.; Zerbato, V.; Rizzello, B.; Bosio, R.; Shbaklo, N.; Corcione, S.; et al. Temocillin: Applications in Antimicrobial Stewardship as a Potential Carbapenem-Sparing Antibiotic. Antibiotics 2022, 11, 493. [Google Scholar] [CrossRef]
  106. Schievink, H.I.; Mattie, H.; Thomeer, R.T.; Van Strijen, E. The Passage of Cloxacillin into Cerebrospinal Fluid in the Absence of Meningitis. Br. J. Clin. Pharmacol. 1993, 36, 57–60. [Google Scholar] [CrossRef] [Green Version]
  107. Le Turnier, P.; Gregoire, M.; Deslandes, G.; Lakhal, K.; Deschanvres, C.; Lecomte, R.; Talarmin, J.-P.; Dubée, V.; Bellouard, R.; Boutoille, D.; et al. Should We Reconsider Cefazolin for Treating Staphylococcal Meningitis? A Retrospective Analysis of Cefazolin and Cloxacillin Cerebrospinal Fluid Levels in Patients Treated for Staphylococcal Meningitis. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2020, 26, 1415.e1–1415.e4. [Google Scholar] [CrossRef]
  108. Grégoire, M.; Gaborit, B.; Deschanvres, C.; Lecomte, R.; Deslandes, G.; Dailly, É.; Ambrosi, X.; Bellouard, R.; Asseray, N.; Lakhal, K.; et al. High-Dosage Cefazolin Achieves Sufficient Cerebrospinal Diffusion to Treat an External Ventricular Drainage-Related Staphylococcus aureus Ventriculitis. Antimicrob. Agents Chemother. 2019, 63, e01844-18. [Google Scholar] [CrossRef] [Green Version]
  109. Pacifici, G.M. Clinical Pharmacology of Piperacillin-Tazobactam Combination in Infants and Children. Clin. Med. Investig. 2019. [Google Scholar] [CrossRef]
  110. Franz, P.; von Rosen, F.; Garner, C.; Swozil, U.; Schmiedek, P.; Einhäupl, K.; Adam, D. Cerebrospinal Fluid Penetration after Single or Multiple Dosage with Ticarcillin/Clavulanate. J. Antimicrob. Chemother. 1989, 24 (Suppl. B), 107–110. [Google Scholar] [CrossRef] [PubMed]
  111. Markantonis, S.L.; Markou, N.; Fousteri, M.; Sakellaridis, N.; Karatzas, S.; Alamanos, I.; Dimopoulou, E.; Baltopoulos, G. Penetration of Colistin into Cerebrospinal Fluid. Antimicrob. Agents Chemother. 2009, 53, 4907–4910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Seyberth, H.W.; Kauffman, R.E. Basics and Dynamics of Neonatal and Pediatric Pharmacology. In Pediatric Clinical Pharmacology; Springer: Berlin/Heidelberg, Germany, 2011; Volume 258, pp. 3–58. ISBN 354022565X. [Google Scholar]
  113. Susa, S.T.; Preuss, C.V. Drug Metabolism. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK442023/ (accessed on 10 October 2022).
  114. De Wildt, S.N.; Tibboel, D.; Leeder, J.S. Drug Metabolism for the Paediatrician. Arch. Dis. Child. Educ. Pract. Ed. 2014, 99, 1137–1142. [Google Scholar] [CrossRef] [PubMed]
  115. Barreto, E.F.; Larson, T.R.; Koubek, E.J. Drug Excretion. In Reference Module in Biomedical Sciences; Elsevier: Amsterdam, The Netherlands, 2021; pp. 1–18. [Google Scholar] [CrossRef]
  116. Krishna, D.R.; Klotz, U. Extrahepatic Metabolism of Drugs in Humans. Clin. Pharmacokinet. 1994, 26, 144–160. [Google Scholar] [CrossRef]
  117. Lynch, T.; Price, A. The Effect of Cytochrome P450 Metabolism on Drug Response, Interactions, and Adverse Effects. Am. Fam. Physician 2007, 76, 391–396. [Google Scholar]
  118. Stavropoulou, E.; Pircalabioru, G.G.; Bezirtzoglou, E. The Role of Cytochromes P450 in Infection. Front. Immunol. 2018, 9, 89. [Google Scholar] [CrossRef] [Green Version]
  119. Lu, H.; Rosenbaum, S. Developmental Pharmacokinetics in Pediatric Populations. J. Pediatr. Pharmacol. Ther. 2014, 19, 262–276. [Google Scholar] [CrossRef]
  120. Kiss, M.; Mbasu, R.; Nicolaï, J.; Barnouin, K.; Kotian, A.; Mooij, M.G.; Kist, N.; Wijnen, R.M.H.; Ungell, A.L.; Cutler, P.; et al. Ontogeny of Small Intestinal Drug Transporters and Metabolizing Enzymes Based on Targeted Quantitative ProteomicsS. Drug Metab. Dispos. 2021, 49, 1038–1046. [Google Scholar] [CrossRef]
  121. Krasinski, K.; Perkin, R.; Rutledge, J. Gray Baby Syndrome Revisited. Clin. Pediatr. 1982, 21, 571–572. [Google Scholar] [CrossRef]
  122. Cascorbi, I. Drug Interactions—Principles, Examples and Clinical Consequences. Dtsch. Arztebl. Int. 2012, 109, 546–556. [Google Scholar] [CrossRef]
  123. Zanger, U.M.; Schwab, M. Cytochrome P450 Enzymes in Drug Metabolism: Regulation of Gene Expression, Enzyme Activities, and Impact of Genetic Variation. Pharmacol. Ther. 2013, 138, 103–141. [Google Scholar] [CrossRef]
  124. Hakkola, J.; Hukkanen, J.; Turpeinen, M.; Pelkonen, O. Inhibition and Induction of CYP Enzymes in Humans: An Update; Springer: Berlin/Heidelberg, Germany, 2020; Volume 94, ISBN 0123456789. [Google Scholar]
  125. Smith, D.A.; Beaumont, K.; Maurer, T.S.; Di, L. Relevance of Half-Life in Drug Design. J. Med. Chem. 2018, 61, 4273–4282. [Google Scholar] [CrossRef]
  126. van den Anker, J.; Reed, M.D.; Allegaert, K.; Kearns, G.L. Developmental Changes in Pharmacokinetics and Pharmacodynamics. J. Clin. Pharmacol. 2018, 58, S10–S25. [Google Scholar] [CrossRef] [Green Version]
  127. Rodieux, F.; Wilbaux, M.; van den Anker, J.N.; Pfister, M. Effect of Kidney Function on Drug Kinetics and Dosing in Neonates, Infants, and Children. Clin. Pharmacokinet. 2015, 54, 1183–1204. [Google Scholar] [CrossRef] [Green Version]
  128. Gattineni, J.; Baum, M. Developmental Changes in Renal Tubular Transport—An Overview. Pediatr. Nephrol. 2015, 30, 2085–2098. [Google Scholar] [CrossRef] [Green Version]
  129. Quigley, R. Developmental Changes in Renal Function. Curr. Opin. Pediatr. 2012, 24, 184–190. [Google Scholar] [CrossRef]
  130. Baptista, J.P. Augmented renal clearance. In Antibiotic Pharmacokinetic/Pharmacodynamic Considerations in the Critically III; Adis Singapore: Singapore, 2017; Volume 49, pp. 125–150. [Google Scholar] [CrossRef]
  131. Rhoney, D.H.; Metzger, S.A.; Nelson, N.R. Scoping Review of Augmented Renal Clearance in Critically Ill Pediatric Patients. Pharmacotherapy 2021, 41, 851–863. [Google Scholar] [CrossRef]
  132. Hefny, F.; Stuart, A.; Kung, J.Y.; Mahmoud, S.H. Prevalence and Risk Factors of Augmented Renal Clearance: A Systematic Review and Meta-Analysis. Pharmaceutics 2022, 14, 445. [Google Scholar] [CrossRef]
  133. Wacharachaisurapol, N.; Sukkummee, W.; Anunsittichai, O.; Srisan, P.; Sangkhamal, S.; Chantharit, P.; Vandepitte, W.P.; Wattanavijitkul, T.; Puthanakit, T. Dose Recommendations for Intravenous Colistin in Pediatric Patients from a Prospective, Multicenter, Population Pharmacokinetic Study. Int. J. Infect. Dis. 2021, 109, 230–237. [Google Scholar] [CrossRef]
  134. Avedissian, S.N.; Rohani, R.; Bradley, J.; Le, J.; Rhodes, N.J. Optimizing Aminoglycoside Dosing Regimens for Critically Ill Pediatric Patients with Augmented Renal Clearance: A Convergence of Parametric and Nonparametric Population Approaches. Antimicrob. Agents Chemother. 2021, 65, e02629-20. [Google Scholar] [CrossRef] [PubMed]
  135. Chen, I.H.; Nicolau, D.P. Augmented Renal Clearance and How to Augment Antibiotic Dosing. Antibiotics 2020, 9, 393. [Google Scholar] [CrossRef] [PubMed]
  136. Margineanu, I.; Akkerman, O.; Cattaneo, D.; Goletti, D.; Marriott, D.J.E.; Migliori, G.B.; Mirzayev, F.; Peloquin, C.A.; Stienstra, Y.; Alffenaar, J.-W. Practices of Therapeutic Drug Monitoring in Tuberculosis: An International Survey. Eur. Respir. J. 2022, 59, 2102787. [Google Scholar] [CrossRef] [PubMed]
  137. Ghimire, S.; Bolhuis, M.S.; Sturkenboom, M.G.G.; Akkerman, O.W.; de Lange, W.C.M.; van der Werf, T.S.; Alffenaar, J.-W.C. Incorporating Therapeutic Drug Monitoring into the World Health Organization Hierarchy of Tuberculosis Diagnostics. Eur. Respir. J. 2016, 47, 1867–1869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Bolhuis, M.S.; Akkerman, O.W.; Sturkenboom, M.G.G.; de Lange, W.C.M.; van der Werf, T.S.; Alffenaar, J.-W.C. Individualized Treatment of Multidrug-Resistant Tuberculosis Using Therapeutic Drug Monitoring. Int. J. Mycobacteriology 2016, 5, S44–S45. [Google Scholar] [CrossRef] [Green Version]
  139. Rybak, M.J.; Le, J.; Lodise, T.P.; Levine, D.P.; Bradley, J.S.; Liu, C.; Mueller, B.A.; Pai, M.P.; Wong-Beringer, A.; Rotschafer, J.C.; et al. Therapeutic Monitoring of Vancomycin for Serious Methicillin-Resistant Staphylococcus Aureus Infections: A Revised Consensus Guideline and Review by the American Society of Health-System Pharmacists, the Infectious Diseases Society of America, the Pediatr. Clin. Infect. Dis. 2020, 71, 1361–1364. [Google Scholar] [CrossRef]
  140. Ewoldt, T.M.J.; Abdulla, A.; Rietdijk, W.J.R.; Muller, A.E.; de Winter, B.C.M.; Hunfeld, N.G.M.; Purmer, I.M.; van Vliet, P.; Wils, E.-J.; Haringman, J.; et al. Model-Informed Precision Dosing of Beta-Lactam Antibiotics and Ciprofloxacin in Critically Ill Patients: A Multicentre Randomised Clinical Trial. Intensive Care Med. 2022, 48, 1760–1771. [Google Scholar] [CrossRef]
  141. Katona, P.; Katona-Apte, J. The Interaction between Nutrition and Infection. Clin. Infect. Dis. 2008, 46, 1582–1588. [Google Scholar] [CrossRef]
  142. WHO. Fact Sheet Malnutrition. Available online: https://www.who.int/news-room/fact-sheets/detail/malnutrition (accessed on 6 October 2022).
  143. Oshikoya, K.A.; Sammons, H.M.; Choonara, I. A Systematic Review of Pharmacokinetics Studies in Children with Protein-Energy Malnutrition. Eur. J. Clin. Pharmacol. 2010, 66, 1025–1035. [Google Scholar] [CrossRef] [Green Version]
  144. Verrest, L.; Wilthagen, E.A.; Beijnen, J.H.; Huitema, A.D.R.; Dorlo, T.P.C. Influence of Malnutrition on the Pharmacokinetics of Drugs Used in the Treatment of Poverty-Related Diseases: A Systematic Review. Clin. Pharmacokinet. 2021, 60, 1149–1169. [Google Scholar] [CrossRef]
  145. Lazzerini, M.; Tickell, D. Antibiotics in Severely Malnourished Children: Systematic Review of Efficacy, Safety and Pharmacokinetics. Bull. World Health Organ. 2011, 89, 594–607. [Google Scholar] [CrossRef]
  146. Standing, J.F.; Ongas, M.O.; Ogwang, C.; Kagwanja, N.; Murunga, S.; Mwaringa, S.; Ali, R.; Mturi, N.; Timbwa, M.; Manyasi, C.; et al. Dosing of Ceftriaxone and Metronidazole for Children with Severe Acute Malnutrition. Clin. Pharmacol. Ther. 2018, 104, 1165–1174. [Google Scholar] [CrossRef] [Green Version]
  147. Dipasquale, V.; Cucinotta, U.; Romano, C. Acute Malnutrition in Children: Pathophysiology, Clinical Effects and Treatment. Nutrients 2020, 12, 2413. [Google Scholar] [CrossRef]
  148. Otiti, M.I.; Allen, S.J. Severe Acute Malnutrition in Low- and Middle-Income Countries. Paediatr. Child Health 2021, 31, 301–307. [Google Scholar] [CrossRef]
  149. Bolme, P.; Margareta, E. Influence of Diarrhea on the Oral Absorption of Penicillin V and Ampicillin in Children. Scand. J. Infect. Dis. 1975, 7, 141–145. [Google Scholar] [CrossRef]
  150. Justesen, U.S.; Andersen, A.B.; Klitgaard, N.A.; Brosen, K.; Gerstoft, J.; Pedersen, C. Pharmacokinetic Interaction between Rifampin and the Combination of Indinavir and Low-Dose Ritonavir in HIV-Infected Patients. Clin. Infect. Dis. 2004, 38, 426–429. [Google Scholar] [CrossRef]
  151. de Paula, R.P.; Nascimento, A.F.; Sousa, S.M.B.; Bastos, P.R.V.; Barbosa, A.A.L. Glomerular Filtration Rate Is Altered in Children with Sickle Cell Disease: A Comparison between Hb SS and Hb SC. Rev. Bras. Hematol. Hemoter. 2013, 35, 349–351. [Google Scholar] [CrossRef]
  152. Hirschberg, R. Glomerular Hyperfiltration in Sickle Cell Disease. Clin. J. Am. Soc. Nephrol. 2010, 5, 748–749. [Google Scholar] [CrossRef] [Green Version]
  153. Chami, N.; Kabyemera, R.; Masoza, T.; Ambrose, E.; Kimaro, F.; Kayange, N.; Hokororo, A.; Furia, F.F.; Peck, R. Prevalence and Factors Associated with Renal Dysfunction in Children Admitted to Two Hospitals in Northwestern Tanzania. BMC Nephrol. 2019, 20, 79. [Google Scholar] [CrossRef] [Green Version]
  154. Halle, M.P.; Lapsap, C.T.; Barla, E.; Fouda, H.; Djantio, H.; Moudze, B.K.; Akazong, C.A.; Priso, E.B. Epidemiology and Outcomes of Children with Renal Failure in the Pediatric Ward of a Tertiary Hospital in Cameroon. BMC Pediatr. 2017, 17, 202. [Google Scholar] [CrossRef]
  155. Denti, P.; Garcia-Prats, A.J.; Draper, H.R.; Wiesner, L.; Winckler, J.; Thee, S.; Dooley, K.E.; Savic, R.M.; McIlleron, H.M.; Schaaf, H.S.; et al. Levofloxacin Population Pharmacokinetics in South African Children Treated for Multidrug-Resistant Tuberculosis. Antimicrob. Agents Chemother. 2018, 62, e01521-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Fredrick, F.; Francis, J.M.; Ruggajo, P.J.; Maro, E.E. Renal Abnormalities among HIV Infected Children at Muhimbili National Hospital (MNH)—Dar Es Salaam, Tanzania. BMC Nephrol. 2016, 17, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Keller, G.A.; Fabian, L.; Gomez, M.; Gonzalez, C.D.; Diez, R.A.; Girolamo, G. Di Age-Distribution and Genotype-Phenotype Correlation for N-Acetyltransferase in Argentine Children under Isoniazid Treatment. Int. J. Clin. Pharmacol. Ther. 2014, 52, 292–302. [Google Scholar] [CrossRef] [PubMed]
  158. Schaaf, H.S.; Parkin, D.P.; Seifart, H.I.; Werely, C.J.; Hesseling, P.B.; Van Helden, P.D.; Maritz, J.S.; Donald, P.R. Isoniazid Pharmacokinetics in Children Treated for Respiratory Tuberculosis. Arch. Dis. Child. 2005, 90, 614–618. [Google Scholar] [CrossRef] [Green Version]
  159. Zhu, R.; Kiser, J.J.; Seifart, H.I.; Werely, C.J.; Mitchell, C.D.; D’Argenio, D.Z.; Fletcher, C.V. The Pharmacogenetics of NAT2 Enzyme Maturation in Perinatally HIV Exposed Infants Receiving Isoniazid. J. Clin. Pharmacol. 2012, 52, 511–519. [Google Scholar] [CrossRef]
  160. Holgate, S.L.; Bekker, A.; Pillay-Fuentes Lorente, V.; Dramowski, A. Errors in Antimicrobial Prescription and Administration in Very Low Birth Weight Neonates at a Tertiary South African Hospital. Front. Pediatr. 2022, 10, 838153. [Google Scholar] [CrossRef]
  161. Oshikoya, K.A.; Oreagba, I.A.; Ogunleye, O.O.; Senbanjo, I.O.; MacEbong, G.L.; Olayemi, S.O. Medication Administration Errors among Paediatric Nurses in Lagos Public Hospitals: An Opinion Survey. Int. J. Risk Saf. Med. 2013, 25, 67–78. [Google Scholar] [CrossRef]
  162. Nwobodo, N. Therapeutic Drug Monitoring in a Developing Nation: A Clinical Guide. JRSM Open 2014, 5, 205427041453112. [Google Scholar] [CrossRef] [Green Version]
  163. Greybe, L. Newsletter of the African Society for Paediatric Infectious Diseases. Available online: https://cct.mycpd.co.za/FIDSSA/AfSPID_Bulletin_Dec_2020.pdf (accessed on 9 September 2022).
  164. Jacobs, J.; Hardy, L.; Semret, M.; Lunguya, O.; Phe, T.; Affolabi, D.; Yansouni, C.; Vandenberg, O. Diagnostic Bacteriology in District Hospitals in Sub-Saharan Africa: At the Forefront of the Containment of Antimicrobial Resistance. Front. Med. 2019, 6, 205. [Google Scholar] [CrossRef]
Figure 1. Overview of the pharmacokinetic processes absorption, distribution, metabolism and elimination.
Figure 1. Overview of the pharmacokinetic processes absorption, distribution, metabolism and elimination.
Antibiotics 12 00017 g001
Figure 2. Concentration-time curve following oral administration or prolonged infusion (a) and after intravenous administration (b). Abbreviations: AUC0-t4: area under the curve between dosing interval. Cmax: maximum concentration. T: time. MIC: minimum inhibitory concentration. MTC: minimum toxic concentration.
Figure 2. Concentration-time curve following oral administration or prolonged infusion (a) and after intravenous administration (b). Abbreviations: AUC0-t4: area under the curve between dosing interval. Cmax: maximum concentration. T: time. MIC: minimum inhibitory concentration. MTC: minimum toxic concentration.
Antibiotics 12 00017 g002
Table 1. (A) Physicochemical & pharmacokinetic characteristics of antimicrobials mainly eliminated by urine. (B) Physicochemical and pharmacokinetic characteristics of antimicrobials mainly eliminated by feces. (C) Physicochemical and pharmacokinetic characteristics of antimicrobials mainly eliminated by bile.
Table 1. (A) Physicochemical & pharmacokinetic characteristics of antimicrobials mainly eliminated by urine. (B) Physicochemical and pharmacokinetic characteristics of antimicrobials mainly eliminated by feces. (C) Physicochemical and pharmacokinetic characteristics of antimicrobials mainly eliminated by bile.
(A)
ClassAgentPK/PD
Index
Molecular Weight (g/mol) [20]pKa [21]LogP [20]Fraction Protein Binding (%) [20]Metabolism [21]Alternative Route of Elimination [21]
AminoglycosidesAmikacinCmax/MIC [22]585.68.1–12.1−8.8–−7.4<10%Aminoglycosides are not significantly metabolized.
Gentamicin477.610.1–12.6−4.1–−1.90–30%
Kanamycin484.59.5–12.1−6.9–−6.3N/A
Neomycin614.612.9 [23]−9–−3.7N/A
Streptomycin581.611.1–11.6−8–−2.5N/A
Spectinomycin332.47.0–9.2−3.1–−2.3Not significant
Tobramycin467.59.7–12.5−6.2–−5.8Not significant
First line anti-
mycobacterials
IsoniazidAUC/MIC [24]137.11.8–13.6−0.8–−0.70–10%Hepatic
PyrazinamideAUC/MIC [24]123.1−0.5–13−1–−0.6~10%Mainly hepatic
RifabutinAUC/MIC, Cmax/MIC [25]847.06.9–9.04.1–4.785%HepaticFeces
Second line antimycobacterialsCycloserineT > MIC [26]102.14.2–8.4−1.5–−0.9N/AHepatic [27]
EthionamideAUC/MIC [28]166.35–11.90.4–1.1~30%Extensive hepatic metabolism
Beta-lactamase
inhibitors
Clavulanic acidT > MIC [29]199.2−2.6–3.2−2.3–−1.2~25% for amoxicillin-clavulanic acidHepaticFeces,
exhaled air
Sulbactam233.2−3.8–3.1−1~38%<25% is metabolized by the liver [30]
Tazobactam300.30.8–2.9−2~30%Hepatic
Avibactam265.3−3.9–−2−1.85.7–8.2%Not significant
Vaborbactam297.1−2.6–3.81.0–1.9 [31]~33%Not significant
Relebactam348.4−2–10−3.6~22%Not significant
CarbapenemsDoripenemT > MIC [22]420.53.3–9.5−5.6, −1.3 [32]8.1%Limited hepatic metabolism
Ertapenem475.53.2–9.00.3–1.585–95%Limited hepatic metabolism
Imipenem299.43.2–10.9−0.720%Renal metabolism
Meropenem383.53.3–9.4−2.4–−0.6~2%<30% of a dose undergoes hepatic metabolism
First generation cephalosporinsCephalexinT > MIC [33,34]347.43.3–7.20.6–0.710–15%Not significant
Cefazolin454.50.3–2.8−0.674–86%Not significant
Cefadroxil363.43.3–7.2−2.1–−0.428.1%Not significant
Second
generation
cephalosporins
Cefaclor367.82.8–7.2−2.3–0.923.5%Not significant
Cefuroxime 424.4−1.1–3.0−0.8–−0.250%Not significant
Cefuroxime axetil510.5−1.2–10.90.928–38% [35]Axetil is metabolized by the liver
Cefotetan575.6−1.5–3.00.188%Not significant
Cefoxitin427.5−3.8–3.4031–54% [36]Minimal hepatic metabolism
Cefprozil389.43.3–7.2−1.4–0.636%Not significant
Cefmetazole471.5−1.7–3.2−2.2–−0.685% [37]Not significant
Third generation cephalosporinsCefdinir395.42.7–9.7−3.5–060–70%Not significant
Cefditoren 506.62.3–3.70.788%Not significant
Cefixime453.52.5–4.0−0.7–−0.465%Hepatic
Cefpodoxime427.52.8–3.6−1.421–33%Minimal hepatic
metabolism
Ceftazidime546.62.4–4.0−1.6–0.45–23%Not significant
Ceftizoxime383.42.7–3.6030%Not significant
Ceftibuten410.42.9–4.7−0.365%~10% is metabolized by the liver
Ceftriaxone554.62.7–3.4−1.7–−1.395%NegligibleBile
Cefotaxime455.52.7–3.6−1.4–−0.58–41% [36]Partially (15–20%) by the liver [38]
Ceftolozane666.72.5–9.1−6.2–−3.216–21%Not significant
Fourth generation cephalosporinsCefepime480.62.8–3.6−0.120%<1% is metabolized by the liver
Fifth generation cephalosporinsCeftobiprole534.62.9–10.4−2.4<16% [39]Minimal hepatic metabolism [39]
Ceftaroline684.70.4–1.82.3~20%Minimal hepatic metabolismFeces
Siderophore
cephalosporins
Cefiderocol752.22.6–4.0−2.3–140–60%Minimal hepatic metabolism
FluoroquinolonesCiprofloxacinAUC/MIC [34]331.35.6–8.8−1.1–2.320–40%Up to 15% hepatic metabolismFeces
Delafloxacin440.8−1.3–5.62.784%HepaticFeces
Gatifloxacin375.45.5–8.8−0.7–2.620%Limited hepatic
metabolism
Levofloxacin361.45.4–6.7−0.4–2.124–38%Very limited
metabolism
Feces
Norfloxacin319.35.6–8.8−1.0–2.110–15%Hepatic and renalFeces
Ofloxacin361.45.4–6.7−0.4–2.132%HepaticFeces
Prulifloxacin461.55.2–6.01.041–59% [40]HepaticFeces
GlycopeptidesTeicoplaninAUC/MIC [41]1879.73.0–7.10.590–95% [42]Minimal hepatic metabolism
Vancomycin1449.33.0–9.9−3.1–−2.6~50%Not significant
Lipoglyco-
peptides
Dalbavancin1816.71.7–9.9 [43]3.893%Unlikely to have significant metabolismFeces
Telavancin1755.61.6–10.0−2.1>90%Unknown
Oritavancin1793.12.2–10.01.5–4.185%Not significantFeces
LincosamidesClindamycinAUC/MIC [22,34]425.07.6–12.42.260–94% [44]HepaticFeces
Lincomycin406.58.0–12.40.2–0.628–86%HepaticBile
MonobactamsAztreonamT > MIC [45]435.4−1.5–3.90.343–56%6–16% is metabolized by the liver
NitroimidazolesMetronidazoleAUC/MIC, Cmax/MIC [22,46]171.22.6–15.4−0.1–0<20%HepaticFeces
SecnidazoleUndefined185.23.1–15.20.2<5–15%N/A
TinidazoleUndefined247.23.3−0.4–0.712%HepaticFeces
OxazolidonesLinezolidAUC/MIC [22]337.4−1.2–14.90.7–1.3~31%Hepatic
Natural
penicillins
Penicillin GT > MIC [22]334.4−2.8–3.51.5–1.845–68%HepaticBile
AminopenicillinsAmoxicillin365.43.2–7.2−2–0.917%Hepatic
Ampicillin349.43.2–7.2−1.1–1.48–25% [47]Hepatic
Semi-synthetic penicillinsCloxacillin435.9−0.4–3.82.4–3~94%IntestinalBile
Dicloxacillin470.3−0.7–3.82.9–3.796–97% [48]Hepatic
Flucloxacillin453.9−0.9–3.82.6–3.295–96% [48]Hepatic
Oxacillin401.4−0.1–3.82.492–96%45–50% hepatic [49]
Temocillin [50]414.5−4.3–3.11.1~80% [48]N/A
UreidopenicillinsPiperacillin517.6−4.3–3.50.3–0.539.4–71.3% [51]Not significantBile
Carboxy-
penicillins
Ticarcillin384.4−6.3–3.10.845%N/A
Polymixins *Polymyxin BAUC/MIC [33]1203.58.9–11.6−2.579–92%N/A
SulfonamidesSulfadiazineCmax/MIC, AUC/MIC
[22]
250.32.0–6.4−0.2–−0.120–25% [52]Hepatic
Sulfadoxine310.33.4–6.10.7~94% [53]Hepatic
Sulfamethoxazole253.32.0–6.20.7–0.9~70%Hepatic
TetracyclinesDoxycyclineAUC/MIC [54]444.43.1–8.3−0.7–0.6>90%HepaticFeces
Tetracycline444.43.3–9.3−2–−1.320–67%Not significantFeces
MiscellaneousChloramphenicolCmax/MIC, AUC/MIC [22]323.1−2.8–8.70.7–1.150–60% in adults, 32% in premature neonatesExtensive hepatic metabolism
DaptomycinAUC/MIC [22]1619.73.0–9.6−5.190–94% [55]Minimum extent, metabolism site unknown [56]Feces
FosfomycinAUC/MIC [34]138.1−4.3–1.3−1.6–−1.4No plasma bindingNot significant
TrimethoprimCmax/MIC, AUC/MIC [22]290.37.1–17.3 0.6–0.944%Hepatic
NitrofurantoinUndefined238.2−2.2–8.3−0.5<90%Hepatic
(B)
ClassAgentPK/PD IndexMolecular Weight (g/mol)
[20]
pKa [21]LogP [20]Fraction Protein Binding
(%) [20]
Metabolism [21]Alternative Route of Elimination [21]
First line
anti-
mycobacterials
EthambutolCmax/MIC, AUC/MIC [24]204.39.7–14.8−0.4–0.420–30%HepaticUrine
RifabutinAUC/MIC, Cmax/MIC [25]847.06.9–9.04.1–4.785%HepaticUrine
Third line antimycobacterialsBedaquilineAUC/MIC, Cmax/MIC [57]555.58.9–13.67.7>99.9%Hepatic
ClofazimineNot
identified [26]
473.46.6–16.27–7.7N/AN/A
DelamanidNot
Identified [26]
534.55.55.6>99.5%Hepatic
Beta-lactamase
inhibitors
Clavulanic acidT > MIC [29]199.2−2.6–3.2−2.3–−1.2~25% for amoxicillin-clavulanic acidSignificant hepatic metabolismUrine,
exhaled air
FluoroquinolonesGemifloxacinAUC/MIC [34]389.45.4–9.4−0.7–2.360–70%Limited hepatic
metabolism
Urine
Moxifloxacin401.45.5–9.50.6–2.950%<50% hepatic
metabolism
Urine
MacrolidesClarithromycinAUC/MIC, T > MIC [34]748.09–12.51.7–3.2~70%HepaticUrine
Fidaxomicin1058−1.4–5.96.431% [48]Intestinal
OxazolidonesTedizolidAUC/MIC [22]370.3−1.7–14.61.470–90%HepaticUrine
TetracyclinesEravacyclineAUC/MIC [54]558.63.0–9.0179–90%HepaticUrine
Omadacycline556.62.9–10.53~20%Not significantUrine
Tigecycline585.73.2–9.0−0.2–1.171–89%Hepatic [58]Urine
DaptomycinAUC/MIC [22]1619.73.0–9.6−5.190–94% [55]Metabolism site
unknown [56]
Urine
(C)
ClassAgentPK/PD IndexMolecular Weight (g/mol)
[20]
pKa [21]LogP [20]Fraction Protein Binding
(%) [20]
Metabolism [21]Alternative Route of Elimination [21]
First line
Antimyco-
bacterials
RifampicinAUC/MIC, Cmax/MIC [25]822.91.7–7.42.7–4.990%HepaticUrine
Third generation cephalosporinsCefoperazoneT > MIC [33,34]645.7−1.7–3.2−0.782–93%Not significant
LincosamidesLincomycinAUC/MIC [22,34]406.58.0–12.40.2–0.628–86%HepaticUrine
MacrolidesAzithromycinAUC/MIC, T > MIC [34]749.08.5–12.43.0–4.07–51%HepaticUrine
Erythromycin733.99–12.52.6–3.170–93%HepaticUrine
Natural
penicillins
Penicillin VT > MIC [22]350.4−4.9–3.41.4–2.150–80%HepaticUrine
Penicillin G334.4−2.8–3.51.5–1.845–68%HepaticUrine
Semi-synthetic penicillinsCloxacillin435.9−0.4–3.82.4–3~94%IntestinalUrine
Nafcillin414.5−1.9–3.32.9–3.388.4–91.4%Hepatic
UreidopenicillinsPiperacillin517.6−4.3–3.50.3–0.539.4–71.3%
[51]
Not significantUrine
TetracyclinesMinocyclineAUC/MIC [54]457.53.2–8.8−0.6–0.176%HepaticUrine
* The route of elimination for Polymyxin is unknown. For molecular weight, LogP and fraction protein binding refer to reference [20] unless cited otherwise. For pKa, metabolism and main route of elimination refer to reference [21] unless cited otherwise.
Table 2. Summary of antimicrobial-food interactions.
Table 2. Summary of antimicrobial-food interactions.
AgentFood Effect on Absorption
AmoxicillinNo effect of fasting status for infants, children and adults [69,70].
Amoxicillin/clavulanateConcomitant food ingestion may enhance absorption and reduce gastric upset [71].
AmpicillinImpaired when taken with food. Therefore, if administered PO, ampicillin should be administered 1 h before or 2 h after meals [70].
Azithromycin Tablets and suspension present no food effect [69].
Cephalexin, cefadroxil, cefaclor, cefprozil, cefiximeNot affected by food intake [72].
Cefuroxime axetilAbsorption and dissolution into active form are improved when taken with food [73,74].
CiprofloxacinImpaired by dairy products, Ca2+ and Mg2+ supplements [75].
MetronidazoleFood may decrease the rate but not the extent of absorption. However, food may reduce gastric upset [76].
RifampicinImpaired when taken with food, therefore should be taken on an empty stomach [77].
TetracyclineImpaired when taken with food or with divalent metal cations, such as Fe+2 and Ca+2 [78].
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

Meesters, K.; Alemayehu, T.; Benou, S.; Buonsenso, D.; Decloedt, E.H.; Pillay-Fuentes Lorente, V.; 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 2023, 12, 17. https://doi.org/10.3390/antibiotics12010017

AMA Style

Meesters K, Alemayehu T, Benou S, Buonsenso D, Decloedt EH, Pillay-Fuentes Lorente V, Downes KJ, Allegaert K. Pharmacokinetics of Antimicrobials in Children with Emphasis on Challenges Faced by Low and Middle Income Countries, a Clinical Review. Antibiotics. 2023; 12(1):17. https://doi.org/10.3390/antibiotics12010017

Chicago/Turabian Style

Meesters, Kevin, Tinsae Alemayehu, Sofia Benou, Danilo Buonsenso, Eric H. Decloedt, Veshni Pillay-Fuentes Lorente, Kevin J. Downes, and Karel Allegaert. 2023. "Pharmacokinetics of Antimicrobials in Children with Emphasis on Challenges Faced by Low and Middle Income Countries, a Clinical Review" Antibiotics 12, no. 1: 17. https://doi.org/10.3390/antibiotics12010017

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