Abstract
Objective
Linezolid (LZD), a commonly used antimicrobial agent in clinical practice, has not undergone adequate pharmacokinetic (PK) assessment in pediatric populations with renal impairment (RI). Physiologically based pharmacokinetic (PBPK) modeling provides unique benefits for investigating drug pharmacokinetics in specific patient groups. This study aimed to employ the PBPK model to refine and optimize the therapeutic dosing protocol of LZD for RI pediatric patients.
Methods
The model was developed and validated for both healthy adults and RI adults, which was subsequently adapted for pediatric applications. Upon verification of the pediatric Based on clinical PK data and real-world study findings, the PBPK model demonstrated precise prediction of LZD exposure in pediatric populations with varying degrees of RI, encompassing weight- and age-associated PK variations.
Results
The PBPK modeling simulations exhibited robust agreement with observational data for LZD across both oral and intravenous delivery routes under diverse dosing protocols, as evidenced by the fold error (FE) always between 0.5 and 2 times, geometric mean fold error (GMFE) was less than 2.0 and mean absolute prediction error (MAPE) was within 100%. Pediatric populations with severe or end-stage RI exhibited 1.21-fold and 1.28-fold elevations in plasma concentration-time curve (AUC) values, respectively, relative to healthy pediatric counterparts when administered equivalent 10 mg/kg LZD doses. Pharmacodynamic analysis confirmed that the proposed dosing regimens—8 mg/kg every 8 hours for children with severe or end-stage RI —were effective in achieving the target AUC0-24/MIC ratio of ≥80 at a susceptible inhibitory concentration of ≤ 2 mg/L.
Conclusion
Our model provides a predictive instrument to enhance precision in determining therapeutic LZD dosage regimens for pediatric populations through systematic integration of developmental PK parameters.
Keywords: pediatric, physiologically based pharmacokinetic model, renal impairment, linezolid, pharmacodynamics
Introduction
Linezolid (LZD), a first-in-class synthetic oxazolidinone antimicrobial agent, is clinically indicated for the management of multidrug-resistant Gram-positive bacterial pathogens, including methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus (VRE), and Mycobacterium tuberculosis infections.1,2 LZD can be administered either intravenously or orally owing to its absolute bioavailability of close to 100%.3 Sadahiro et al’s research shows that age and body weight (BW) were influential covariates on clearance.4 The pharmacokinetic (PK) parameters of LZD in adolescents aged 12–18 were shown to be similar to those of adult patients, for children younger than 12 years old, the clearance of LZD increased in an age-dependent manner.5 The established dosage protocol specifies distinct administration schedules based on patient age demographics. For patients aged ≥12 years, a twice-daily regimen of 600 mg per dose is recommended. In contrast, pediatric populations encompassing neonates and children under 12 years require a weight-adjusted regimen of 10 mg/kg administered three times daily.6
LZD is predominantly metabolized through the oxidation of its morpholine ring to an inactive form by nonenzymatic oxidative reactions, namely aminoethoxy acetic metabolite (PNU-142300) and hydroxyethyl glycine metabolite (PNU-142586), which is unrelated to the cytochrome P450 system. This non-enzymatic metabolic pathway circumvents the influence of liver development, so individual differences related to liver metabolic regulation have a weaker impact on the in vivo processes of LZD. Nonrenal clearance accounts for approximately 65% of the total clearance of LZD. Under steady-state conditions, approximately 30% of the dose appears in the urine as LZD, 40% as metabolite PNU-142586, and 10% as metabolite PNU-142300.7 Previous studies have shown that as renal function declines in patients, the clearance rate of LZD and its two main metabolites decreases, resulting in an increased rate of metabolite accumulation and elevated plasma concentrations in patients with renal impairment. Therefore, although the parent drug accounts for only 35% of renal clearance, the renal-dependent excretion of metabolites results in total body exposure that is highly correlated with renal function.8 Consequently, caution is warranted regarding the risk of accumulation in cases of severe renal impairment.
While current LZD prescribing information states that dosage adjustments are not indicated for renally impaired patients, this recommendation is primarily derived from an early PK study involving only 18 renal impairment (RI) patients,9 and subsequent research have demonstrated the clinical necessity of therapeutic drug monitoring (TDM) in LZD regimens10 Clinical investigations have identified a significant correlation between renal function parameters and the incidence of LZD-induced thrombocytopenia (LIT) in treated patients,11 and it could be arise from the altered exposure of the drug. Prior PK investigations revealed significantly elevated risks of systemic overexposure in renally impaired patients (creatinine clearance ≤40 mL/min), demonstrating a 4.27-fold increase in plasma exposure in comparison with cohorts with preserved renal function (eGFR >80 mL/min),12 while the median LZD concentration in RI patients was 1.46-fold that of patients with normal function.13
The PK of the majority of drugs vary between pediatric and adult populations, necessitating the use of distinct, age-appropriate dosing regimens. Additionally, in cases of renal impairment or renal failure when renal function has a significantly influences in-vivo exposure of the drug.14 According to the expert consensus statement on therapeutic drug monitoring and individualization of LZD, for RI patients who are not undergoing hemodialysis, the LZD dosage can be adjusted to a regimen of 300 mg administered q12h,6 however, there is no research and recommendation for the dose adjustment for RI pediatric patients.
Considering the physiological distinctions between adults and pediatric populations, research has illustrated the benefits of PBPK modeling in predicting and simulating drug exposure across various age groups,15–18 recognized by the FDA and the European Medicines Agency (EMA), to explore and quantitatively predict the PK of drugs, to evaluate drug–drug interactions (DDIs), and to support clinical study design, dose selection.19,20 Consequently, our study constructed PBPK models tailored for children and RI pediatric patients, aiming to using validated PBPK models to predict changes in drug exposure in children with renal impairment, thereby guiding dose information when clinical trials are not feasible.
Methods
PK Data
A systematic review of published literature was conducted to aggregate intravenous LZD PK parameters across distinct populations, encompassing healthy adult and pediatric cohorts as well as adult subjects with renal impairment.7,9,21 Using the digitizing software GetData Graph Digitizer version 2.26, the observational concentration–time curves were extracted directly from the graphs.
Establishment of the PBPK Model
The models were developed with the processes shown in Figure S1. The PBPK model for LZD was constructed using GastroPlus version 9.9 (Simulations Plus Inc., Lancaster, CA, USA) and utilized for whole simulations involving both healthy individuals and RI patients. Grounded in the physiological, biochemical, and anatomical principles of the human body, the PBPK model incorporates 13 non-eliminative compartments, including the brain, lungs, heart, muscle, adipose tissue, pancreas, liver, skin, skin, spleen, stomach, intestines, bone, and thymus, along with one eliminative compartment, the kidneys. These organs and tissues are interconnected by arterial and venous blood, each exhibiting its own hemodynamic flow rates, compartmental volumes, tissue-plasma partition coefficients, and transvacuolar permeability characteristics in accordance with the principle of mass conservation. In the present investigation, LZD, exhibiting properties such as a low molecular weight, high lipid solubility, and excellent tissue penetrability,22–24 was employed within a perfusion-limited to constructing a PBPK model. The basic physicochemical and biopharmaceutical parameters for LZD are listed in Table S1.
Estimation of Clearance
LZD is not considered as a substrate for any known major renal transport system, despite the fact that renal reabsorption plays a role in its overall renal clearance (CLR).25 According to the drug label of LZD, the mean total clearance following intravenous administration is roughly 8.76 L/h. It has been reported that the CLR subsequent to tubular reabsorption falls within the range of 1.8–3.0 L/h.7 When the CLR is 1.8 L/h, the predicted alteration in the plasma concentration–time curve (AUC), which is obtained by comparing the RI adults (creatinine clearance <30 mL/min) to the healthy adults, is 1.3-fold. This predicted value is in close proximity to the clinical observation of a 1.2-fold change.7,9 Therefore, a CLR value of 1.8 L/h was established as the foundational PK parameter in this investigation. Given that the intricate and detailed mechanism underlying the liver metabolism of the substance in question remains unknown,25 in the current research, the hepatic intrinsic clearance (Hep CLint) was estimated through the application of the retrogressive approach within the GastroPlus software. The estimated value of Hep CLint was determined to be 0.996 μL/min/mg protein.
Healthy Adults’ LZD PBPK Model
We developed PBPK models for both oral and intravenous administration in healthy adult populations. Utilizing available PK data, we conducted simulations for single-dose and multiple-dose regimens in healthy adults. The experimental design incorporated both single-dose intravenous regimens (250 and 500 mg) and a single 600 mg oral administration,7 alongside multi-dose intravenous protocols delivering 500 mg and 625 mg doses at 12-hour intervals (q12h).21 The PBPK model for LZD was verified using clinical PK data from previous PK studies.
Pediatric Patients’ LZD PBPK Model
We developed PBPK models based on injection administration in pediatric patients. The simulation in pediatric patients with 13–17, 7–12, 3–6, 1–2, 0.25–1 years included a LZD single-dose injection administration of 10 mg/kg according to the previous PK studies.7 The PBPK model for LZD was verified using clinical PK data from previous PK studies.
Hospitalized Pediatric Populations Individualization LZD PBPK Model
We retrospectively collected 16 pediatric inpatients receiving intravenous LZD therapy, with systematic quantification of post-administration plasma drug concentrations were performed using HPLC-MS. Patient-individualized PBPK models were developed by integrating demographic covariates (age, gender, body mass index), clinical parameters (kidney and hepatic function), and pharmacotherapeutic variables (administration frequency, dose, and route), and we used the calculated concentrations obtained from the models to compare with the measured concentrations to complete validation of the models. Moreover, we further used a t-test to analyze the predicted plasma concentrations.
PBPK Model of LZD for Adult and Pediatric Populations Across a Spectrum of RI Severity
Specific differences in physiological parameters, including glomerular filtration rate (GFR), hematocrit, plasma protein levels, and blood pressure, between healthy and RI populations were incorporated into the GastroPlus model. The software follows the renal function classification of the FDA guidance (FDA/CDER 2010, accessible via www.accessdata.fda.gov/scripts/cder/daf/) using the GFR ranges 60–90, 30–59, 15–29, and <15mL/min/1.73m2, which represent mild, moderate, and end-stage RI, respectively.
Model Validation
The simulations were executed under conditions mirroring those of actual clinical trials, encompassing demographic data and dosing regimens. According to the EMA guidelines for PBPK modelling and simulation assessment and reporting,20 quantitative metrics evaluate model predictive performance as follows: the fold error (FE) and geometric mean fold error (GMFE) of PK parameters (AUC and Cmax) representing exposure levels were utilized to evaluate the predictive capability of the model. Prediction error (PE) and mean absolute prediction error (MAPE) are derived by comparing predicted and literature-reported meaning plasma drug concentrations. If FE is between 0.5 and 2 times, GMFE was less than 2.0 and MAPE is within 100%, indicating that the model’s predictive performance is good.26–28
![]() |
(1) |
![]() |
(2) |
![]() |
(3) |
![]() |
(4) |
Population Simulation
To evaluate covariate effects arising from interindividual variability in physiological parameters and compound-specific determinants on the physiological characteristics of the population and compound variables, population simulation for LZD was performed with a sample size of 100. The simulation incorporated the following population traits: virtual adult individuals ranging in the age range of 18 to 65 years, and pediatric individuals classified into age brackets of 13–17, 7–12, 3–6, 1–2, and 0.25–1 years. The virtual cohort consisted of 100 subjects, with an equal gender distribution (50% male). The dosing regimen aligned with prior PK studies.15
Prediction of LZD PK and Dose Adjustment Strategies for Pediatric Populations Across a Spectrum of RI Severity
The PBPK model for LZD disposition in pediatric cohorts with preserved renal function underwent development and validation prior to its extrapolative application in simulating PK at a 10 mg/kg dosing regimen for RI pediatric populations. The software’s embedded parameters were utilized to simulate pediatric populations across a spectrum of RI severity. Observational and predicted PK parameters, involving AUC and the maximum plasma concentration (Cmax), were paralleled, with the normal population serving as the reference group. The dosage optimization for RI pediatrics was determined by multiplying the geometric mean ratio of the normal population to the RI population by the standard dose used in healthy individuals, calculated as:
![]() |
(5) |
Pharmacodynamic Evaluation
LZD has a time-dependent killing effect on susceptible bacteria,29 and the PK/PD target of AUC0-24/MIC ≥80 has been established.30 The minimum inhibitory concentration (MIC) of LZD is 0.5–2.0 mg/L for most common clinical isolates of gram-positive bacteria.31 Consequently, we employed the population simulation feature of GastroPlus, in conjunction with human physiological parameters, to assess the pharmacodynamics (PD) when the MIC values were set at 0.5, 1, 1.5, and 2 mg/L, respectively. For each of the recommended doses, the simulation was carried out 10,000 times. We obtained blood concentration data, and a statistical analysis was performed on the PD metrics associated with the recommended LZD dosages for various degrees of renal function. Leveraging these PK/PD data, the LZD’s PBPK model was utilized to make predictions regarding the efficacious dosing regimens for RI pediatric patients.
Results
Development, Verification and Population Simulation of Healthy Adults’ LZD PBPK Model
The average Plasma Concentration–Time curves, which were both simulated and observational, for single and multiple intravenous doses of LZD in adult individuals with normal renal activity are illustrated in Figures 1 and 2. As presented in Table 1, for the dosing regimens with different doses in healthy adults, the FE values are between 0.8 and 1.4, the GMFE values were found to be less than 2.0. The outcomes of the population simulations are depicted in Figure 3. It was noted that the concentration values following the administration of LZD all fell within the 95% simulated probability range.
Figure 1.
Average plasma concentration–time curve of healthy adult subjects after single dose of LZD. (a and b) Intravenous Injection, (c) Oral Administration, squares are observed values and blue lines are predicted values for LZD.
Figure 2.
Average plasma concentration–time curves following twice-daily oral administration of 500 mg and 625 mg LZD in healthy subjects. Squares are observed values and blue lines are predicted values for LZD.
Table 1.
Observational and Modeled PK Parameters of Intravenous Linezolid in Adult Populations
| Population | Dose | AUC0-t(μg∙h/mL) | Cmax(μg/mL) | ||||||
|---|---|---|---|---|---|---|---|---|---|
| Observed | Predicted | FE | GMFE | Observed | Predicted | FE | GMFE | ||
| Healthy adult | I.v. 250mg (n=16) | 24.792 | 26.342 | 1.062 | 1.130 | 6.331 | 5.958 | 0.941 | 1.165 |
| I.v. 500mg (n=16) | 79.334 | 74.799 | 0.943 | 10.816 | 12.405 | 1.147 | |||
| P.O.600mg (n=16) | 104.130 | 85.955 | 0.825 | 10.615 | 10.590 | 0.998 | |||
| I.v.500mg BID (n=6) | 94.847 | 78.962 | 0.833 | 14.393 | 18.166 | 1.262 | |||
| I.v.625mg BID (n=6) | 109.846 | 97.639 | 0.889 | 15.622 | 21.768 | 1.393 | |||
Figure 3.
Population simulation of LZD after a single intravenous infusion in adults and children. (a) 18–65yrs, (b) 13–17yrs, (c) 7–12yrs, (d) 3–6yrs, (e) 1–2yrs, (f) 0.25–1yrs, population simulation results are shown as a black line, plasma concentration-time profiles with a 95% probability are shown as blue line, and observational values are shown as red squares.
Development, Verification and Population Simulation of Pediatric Patients’ LZD PBPK Model
The average Plasma Concentration–Time curves, encompassing both the simulated and observational data, for a single intravenous dose of LZD in pediatric patients are presented in Figure 4. Additionally, as evidenced in Table 2, for the single-dose regimens administered to children, the FE values are between 0.8 and 1.4, the GMFE values were found to be less than 2.0. The results of the population simulation are depicted in Figure 3. Notably, the concentration values of LZD following its administration all fell within the 95% simulation probability range.
Figure 4.
Average plasma concentration–time curves following a single intravenous dose of 10 mg/kg LZD in pediatric age subgroups. (a) 13–17yrs, (b) 7–12yrs, (c) 3–6yrs, (d) 1–2yrs, (e) 0.25–1yrs, squares are observed values and blue lines are predicted values for LZD.
Table 2.
Observational and Modeled PK Parameters of Intravenous Linezolid Across Pediatric Age Subgroups
| Population | Dose | Age | AUC0-t (μg∙h/mL) | Cmax (μg/mL) | ||||||
|---|---|---|---|---|---|---|---|---|---|---|
| Observed | Predicted | FE | GMFE | Observed | Predicted | FE | GMFE | |||
| Paediatric patients | I.v. 10mg/kg | 13-17 yrs (n=12) |
75.365 | 77.691 | 1.031 | 1.101 | 14.093 | 18.146 | 1.288 | 1.293 |
| 7-12 yrs (n=15) |
73.830 | 74.855 | 1.012 | 17.272 | 19.171 | 1.110 | ||||
| 3-6 yrs (n=14) |
47.114 | 52.791 | 1.120 | 14.310 | 19.952 | 1.394 | ||||
| 1-2 yrs (n=12) |
63.766 | 55.980 | 0.878 | 14.717 | 19.104 | 1.298 | ||||
| 0.25–1yrs (n=10) |
40.707 | 49.495 | 1.216 | 13.122 | 18.308 | 1.395 | ||||
Development and Verification of Hospitalized Pediatric Patients’ Individualization LZD PBPK Model
The Plasma Concentration–Time curves, which include both the simulated and measured data, for intravenous administration of LZD in 16 hospitalized pediatric patients are illustrated in Figure 5. Table 3 demonstrated that the MAPE value of 35.4% for simulated and measured blood concentrations under different dosing regimens and the P-value of 0.292 showed no statistically significant differences.
Figure 5.
Plasma concentration–time curves of LZD in hospitalized pediatric patients. (a–p) 16 patients numbered a~p in Table 3 respectively, squares are observed values and blue lines are predicted values for LZD.
Table 3.
Observational and Modeled Blood Concentrations of Intravenous Linezolid in Hospitalized Pediatric Patients
| Pediatric Patient Number | Age | Weight (kg) |
Dosage | Frequency of Administration | Blood Sampling Point (h) | Plasma Concentration (μg∙h/mL) | |||
|---|---|---|---|---|---|---|---|---|---|
| Observed | Predicted | MAPE | P_value | ||||||
| a | 3.0 | 12.0 | I.v. 120mg | q8h | 131.00 | 4.05 | 2.23 | 35.4% | 0.292 |
| b | 3.0 | 12.0 | I.v. 120mg | q12h | 31.22 | 1.84 | 2.47 | ||
| 3.0 | 12.0 | I.v. 120mg | q12h | 199.23 | 2.13 | 2.62 | |||
| c | 3.0 | 12.0 | I.v. 120mg | q12h | 54.88 | 1.91 | 2.81 | ||
| d | 5.5 | 14.5 | I.v. 150mg | q12h | 54.82 | 2.25 | 1.85 | ||
| e | 5.6 | 22.5 | I.v. 200mg | q12h | 30.95 | 0.97 | 1.26 | ||
| 5.6 | 22.5 | I.v. 200mg | q12h | 126.67 | 1.33 | 1.40 | |||
| f | 7.9 | 28.0 | I.v. 300mg | q12h | 54.42 | 6.28 | 3.80 | ||
| g | 11.2 | 15.0 | I.v. 150mg | q12h | 79.37 | 0.55 | 0.29 | ||
| h | 11.8 | 31.5 | I.v. 300mg | bid | 106.00 | 1.07 | 1.48 | ||
| i | 12.0 | 26.0 | I.v. 300mg | q12h | 54.65 | 4.83 | 2.52 | ||
| j | 12.0 | 26.0 | I.v. 300mg | q12h | 79.12 | 2.74 | 1.42 | ||
| k | 12.7 | 28.0 | I.v. 260mg | qd | 78.92 | 1.04 | 0.70 | ||
| l | 12.8 | 28.0 | I.v. 280mg | qd | 150.33 | 0.71 | 0.43 | ||
| m | 13.2 | 26.0 | I.v. 250mg | q8h | 78.65 | 1.10 | 1.53 | ||
| n | 13.3 | 30.0 | I.v. 300mg | bid | 31.00 | 1.11 | 1.20 | ||
| o | 13.4 | 30.0 | I.v. 200mg | q8h | 19.78 | 5.77 | 3.78 | ||
| 13.4 | 30.0 | I.v. 200mg | q8h | 107.00 | 4.52 | 5.09 | |||
| p | 13.6 | 26.0 | I.v. 300mg | q12h | 55.33 | 2.16 | 2.79 | ||
Development and Verification of the PBPK Model of LZD in RI Adults
Table 4 showed the FE values are between 0.6 and 1.0, the GMFE values were found to be less than 2.0 for simulated and observational single doses of intravenous LZD in RI adults. The predicted values indicated a 20–30% decrease in complete LZD elimination in RI adults, resulting in an increase in the AUC linked to RI. Specifically, the AUC is 1.16-fold and 1.20-fold higher in patients with severe and end-stage RI, respectively, in comparison with healthy adults.
Table 4.
PK Parameters of Linezolid: Observational Versus Simulated Data Following Oral Administration in Adults with Renal Impairment
| Group | AUC0-∞(μg∙h/mL) | Cmax(μg/mL) | ||||||
|---|---|---|---|---|---|---|---|---|
| Observed | Predicted | FE | GMFE | Observed | Predicted | FE | GMFE | |
| Moderate RI (n=6) |
128±53 | 98.763 | 0.772 | 1.263 | 15.5±7.1 | 10.412 | 0.672 | 1.365 |
| Severe RI (n=6) |
127±66 | 105.32 | 0.830 | 10.8±3.1 | 9.913 | 0.918 | ||
| End-stage RI (n=6) |
141±45 | 109.22 | 0.775 | 15.4±5.0 | 9.808 | 0.637 | ||
Predicting of the PK of LZD and Developing Dose Adjustment Regimens in Pediatric Patients Across a Spectrum of RI Severity
The simulated PK changes in RI pediatric patients are presented in Table 5. While no significant alterations were observed in the volume of distribution at steady state (Vss) or half-life (Thalf) in children, the AUC increased by 20–30% in those with severe and end-stage RI in comparison with healthy children. Based on these AUC variations, appropriate dose adjustments for LZD are recommended for children with severe and end-stage RI, as detailed in Table 6.
Table 5.
Simulated PK Profiles of Intravenous Linezolid Administration in Pediatric Age Subgroups Across a Spectrum of Kidney Function
| Group | Condition | AUC0-ꝏ (μg∙h/mL) |
Cmax (μg/mL) |
CL (L/h) |
GFR (mL/s) |
CLnon-renal (L/h) |
CLrenal (L/h) |
Vss (L) |
Thalf (h) |
|---|---|---|---|---|---|---|---|---|---|
| 13–17yrs | Normala | 346.47 | 20.367 | 7.347 | 1.897 | 5.101 | 2.246 | 36.883 | 3.479 |
| Mild RI | 361.25 | 27.749 | 7.046 | 1.580 | 5.152 | 1.894 | 37.197 | 3.658 | |
| Moderate RI | 396.18 | 28.220 | 6.424 | 0.948 | 5.261 | 1.163 | 37.847 | 4.083 | |
| Severe RI | 427.73 | 28.403 | 5.950 | 0.474 | 5.358 | 0.592 | 38.367 | 4.468 | |
| End-stage RI | 451.95 | 28.389 | 5.630 | 0.157 | 5.431 | 0.199 | 38.712 | 4.765 | |
| 7–12yrs | Normala | 497.5 | 23.736 | 4.685 | 1.575 | 3.242 | 1.443 | 22.783 | 3.370 |
| Mild RI | 518.89 | 29.826 | 4.491 | 1.312 | 3.274 | 1.217 | 22.976 | 3.545 | |
| Moderate RI | 569.8 | 30.692 | 4.089 | 0.788 | 3.342 | 0.747 | 23.237 | 3.961 | |
| Severe RI | 616.02 | 31.258 | 3.782 | 0.394 | 3.402 | 0.380 | 23.692 | 4.341 | |
| End-stage RI | 651.61 | 31.553 | 3.575 | 0.131 | 3.447 | 0.128 | 23.907 | 4.634 | |
| 3–6yrs | Normala | 341.88 | 25.508 | 3.523 | 1.021 | 2.794 | 0.730 | 11.706 | 2.302 |
| Mild RI | 350.7 | 25.708 | 3.335 | 0.851 | 2.807 | 0.615 | 11.803 | 2.453 | |
| Moderate RI | 370.35 | 25.868 | 3.252 | 0.511 | 2.875 | 0.377 | 11.998 | 2.557 | |
| Severe RI | 386.37 | 25.865 | 3.117 | 0.255 | 2.925 | 0.192 | 12.154 | 2.702 | |
| End-stage RI | 397.74 | 25.731 | 3.028 | 0.085 | 2.964 | 0.064 | 12.260 | 2.806 | |
| 1–2yrs | Normala | 364.65 | 25.855 | 2.120 | 0.677 | 1.601 | 0.519 | 7.389 | 2.416 |
| Mild RI | 376.58 | 26.039 | 2.052 | 0.564 | 1.615 | 0.437 | 7.448 | 2.515 | |
| Moderate RI | 403.58 | 26.409 | 1.915 | 0.338 | 1.647 | 0.268 | 7.569 | 2.739 | |
| Severe RI | 426.79 | 26.893 | 1.811 | 0.169 | 1.675 | 0.136 | 7.665 | 2.934 | |
| End-stage RI | 443.66 | 26.545 | 1.742 | 0.056 | 1.696 | 0.046 | 7.730 | 3.076 | |
| 0.25–1yrs | Normala | 304.09 | 23.03 | 1.853 | 0.439 | 1.334 | 0.519 | 5.489 | 2.053 |
| Mild RI | 316.12 | 23.223 | 1.782 | 0.366 | 1.346 | 0.436 | 5.533 | 2.152 | |
| Moderate RI | 343.54 | 23.627 | 1.640 | 0.219 | 1.372 | 0.268 | 5.626 | 2.378 | |
| Severe RI | 368.13 | 23.871 | 1.530 | 0.110 | 1.394 | 0.136 | 5.698 | 2.581 | |
| End-stage RI | 386.59 | 23.971 | 1.457 | 0.036 | 1.411 | 0.046 | 5.748 | 2.734 |
Note: aNormal renal function.
Table 6.
Recommended Dose Adjustments for Linezolid in RI Pediatrics
| Normala | Mild RI | Moderate RI | Severe RI | End-stage RI | |
|---|---|---|---|---|---|
| fAUC 0-24h (μg∙h/mL) | 128.29 | 136.90 | 146.76 | 155.28 | 164.12 |
| Geometric mean ratio (RI/healthy) | 1.07 | 1.14 | 1.21 | 1.28 | |
| Proposed recommended dose | 10mg/kgq8 h | 10mg/kgq8 h | 10mg/kgq8 h | 8mg/kg q8 h | 8mg/kg q8 h |
Note: a Normal renal function.
Abbreviations: fAUC0-24h, free area under the concentration vs. time curve from 0 to 24h calculated by trapezoidal rule, which was based on children aged 6 years with different levels of renal function; RI, renal impairment.
Pharmacodynamic Evaluation
The dose was adjusted to 8 mg/kg every 8 hours (q8h), and the PD evaluation results for children with severe and end-stage RI are summarized in Table S2. At a MIC of 1.5 mg/L, 98.7% and 98.9% of children with severe and end-stage RI, respectively, achieved the PK/PD target at the recommended dose. At a MIC of 2 mg/L, the target achievement rate (PTA) for AUC0-24/MIC ≥80 at the recommended dose exceeded 85% in both severe and end-stage RI children, as illustrated in Figure 6.
Figure 6.
The probability of achieving PK goals in children with mild and moderate RI receiving 10 mg/kg LZD, q8h, and in children with severe RI receiving the recommended dose of LZD 8mg/kg, q8h, respectively.
Discussion
Pediatric drug development is faced with many difficulties, such as ethical problems, low consent rates from neonate parents, and limited blood volume availability.32 The PBPK model functions as a robust framework for synthesizing diverse data streams, encompassing in vitro, preclinical, and clinical data, to elucidate PK variations in pediatric populations. Consequently, it offers significant advantages in predicting PK of adult and extrapolating these findings to pediatric cohorts,33 which has been extensively utilized as a critical tool in determination-making processes, study design optimization, and data analysis within academic research, pharmaceutical development, and supervising evaluations of pediatric drug therapies,34–36 numerous modeling reports have been either published or formally submitted to health supervising agencies, serving to optimize clinical trial designs or to support requests for clinical research waivers.37–39
A comprehensive PBPK model incorporating physiological variability was initially established and rigorously validated for intravenous LZD administration in both healthy populations and RI adults, based on which a PBPK model for RI patients can be developed and adequately evaluated. Computational analyses of multi-age cohorts (pediatric and adult subjects) with varying degrees of renal impairment demonstrated a statistically 10–30% reduction in systemic drug clearance relative to age-matched controls with preserved renal function. The analysis of PK parameters showed that values for Thalf, Vss, and total clearance almost no change with decrease in creatinine clearance. In pediatric patients with renal impairment, with the decrease in renal clearance, the nonrenal clearance increased, the observed compensatory increase in nonrenal LZD clearance may arise from declining renal function. This reduction diminishes renal excretion, leading to metabolite accumulation and decreased plasma protein binding.40 The resultant elevation of free plasma drug concentrations accelerates non-enzymatic oxidation rates, thereby compensating for the loss of renal clearance.
To advance the practical application of this model in our hospital, we further analyzed data from our hospital patients. Since the number of patients with measured LZD plasma concentrations in our hospital is limited, we currently have data from only 16 patients. The results with MAPE within 100% further validate the model’s estimation performance. For the 16 patients, we collected data from, we further used a T-test to analyze the predicted plasma concentrations, and the results showed no statistically significant differences. In the future, we will further validate and optimize the model through practical application in a larger population of patients with diverse characteristics.
Previous research showed that LZD may be the substrate of P-glycoprotein (P-gp) and the AUC over the dosing interval and Cmax for LZD were reduced by approximately 32% and 21% when LZD was co-administered with rifampin,41 in comparison with the monotherapy group, the combined use of 500 mg clarithromycin increased the AUC of LZD by 44%.42 However, Madin-Darby canine kidney cells transfected with the human multidrug resistance 1 gene cell-based assay indicated that LZD was not a substrate of P-gp.41 The idea that LZD acts as a P-gp substrate is likewise a subject of debate. Consequently, the parameters of P-gp have not yet been incorporated into our model, the verification results demonstrated acceptable predictive accuracy, so further additional studies are warranted to ascertain whether the inclusion of P-gp would enhance the model’s predictive capabilities.
Previous research has indicated that RI patients experienced LIT more frequently during constant-dose therapy.43–45 Specifically, the proportion of males was 76.5% of 221 patients (20–98yrs) receiving LZD, examination based on the patients’ renal function showed that compared with patients with normal renal function (n=44) LIT occurs twice, 8times, and 9times as often in patients with mild (n=54), moderate (n=56), and severe (n=43) renal impairment, respectively, and 8 times more often in patients undergoing hemodialysis (n=24).46 The results of a retrospective cohort study of children showed that 57% of the 37 children with baseline renal impairment developed LIT.47 RI has been identified as a crucial risk factor for increased LZD Cmin in real-world clinical studies.12,48 Nevertheless, current therapeutic guidelines maintain a uniform 10 mg/kg q8h dosing regimen across all pediatric cohorts, despite documented instances of overexposure and LIT in cases of RI.9 TDM and dose-adjustment strategies have been advocated by multiple researchers to enhance the safety and efficacy of LZD therapy in RI adult patients,44,49,50 whereas our research can provide reference for the formulation of drug administration scheme for RI children in clinical.
Based on our research findings, age demonstrates a significant correlation with drug clearance, clearance rates were observed to increase with age, which in turn influences drug exposure, ie, drug concentration. Clinically, since children’s body weight typically increases with age, pediatric dosages have primarily been determined based on weight. However, beyond body weight, renal function emerges as another critical factor affecting exposure to LZD. The appropriate LZD dosage in RI pediatric patients could theoretically achieve the same free AUC (fAUC) as in healthy children when multiplied by the correction factor for total drug in the indicated category of renal impairment. Based on changes in AUC in RI children in comparison with healthy children, we can draw some conclusions. For children aged 12 years and older, the recommended dosage aligns with that for adults, consistent with prescribing guidelines; for pediatric patients aged 3 months to 11 years, modification is not required for those with mild or moderate RI. However, in cases of severe and end-stage RI, the recommended dosage should be adjusted to 8 mg/kg administered every 8 hours (q8h)., supported by PD analyses.
There were several limitations to this study. First, the conduct of pediatric clinical trials is inherently constrained by numerous practical considerations, such as ethical limitations and difficulties in participant recruitment,51 PK data for LZD in RI pediatric patients are not available to verify the model; therefore, it will be necessary to accumulate relevant clinical data to further optimize and verify future studies of the model. Second, our assumptions may not be appropriate for children with overweight or underweight suffering from renal impairment because we only considered children at the normal level of development. Further studies on children with abnormal development states should be conducted in the future.
Conclusion
In summary, the present study was conducted with the objective of developing, optimizing, and validating PBPK models for LZD in pediatric patients with impaired renal function in order to predict LZD exposure to RI children and to complete PD assessments. The study proposed available optimized dosage regimens for pediatric patients with severe and end-stage RI, respectively. Our model has the potential to serve as a tool for predicting LZD PK and facilitating dose modifications or other further applicable therapeutic decisions.
Acknowledgments
A fellowship from the Chongqing Science and Health Joint Medical Research Project (2023GDRC014 and 2024QNXM012) and Chongqing clinical pharmacy key specialty construction project to this study is gratefully acknowledged.
Funding Statement
This research was funded by grants from the Chongqing Science and Health Joint Medical Research Project (Grant Numbers: 2023GDRC014 and 2024QNXM012) and the Chongqing Key Specialty Construction Project in Clinical Pharmacy.
Ethical Approval
The study complies with the Declaration of Helsinki and was approved by the Ethics Committee of the Children’s Hospital of Chongqing Medical University with an informed consent exemption considering the observational and retrospective nature of the study, and the data were collected without identifiers. (Approval No. 2020–282).
Author Contributions
All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Disclosure
The authors affirm that this research was carried out without any commercial or financial affiliations that might be perceived as potential conflicts of interest.
References
- 1.Perry CM, Jarvis B. Linezolid: a review of its use in the management of serious gram-positive infections. Drugs. 2001;61(4):525–551. doi: 10.2165/00003495-200161040-00008 [DOI] [PubMed] [Google Scholar]
- 2.Lee M, Lee J, Carroll MW, et al. Linezolid for treatment of chronic extensively drug-resistant tuberculosis. N Engl J Med. 2012;367(16):1508–1518. doi: 10.1056/NEJMoa1201964 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Beringer P, Nguyen M, Hoem N, et al. Absolute bioavailability and pharmacokinetics of linezolid in hospitalized patients given enteral feedings. Antimicrob Agents Chemother. 2005;49(9):3676–3681. doi: 10.1128/AAC.49.9.3676-3681.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Abe S, Chiba K, Cirincione B, et al. Population pharmacokinetic analysis of linezolid in patients with infectious disease: application to lower body weight and elderly patients. J Clin Pharmacol. 2009;49(9):1071–1078. doi: 10.1177/0091270009337947 [DOI] [PubMed] [Google Scholar]
- 5.Jungbluth GL, Welshman IR, Hopkins NK. Linezolid pharmacokinetics in pediatric patients: an overview. Pediatr Infect Dis J. 2003;22(9 Suppl):S153–7. doi: 10.1097/01.inf.0000086954.43010.63 [DOI] [PubMed] [Google Scholar]
- 6.Lin B, Hu Y, Xu P, et al. Expert consensus statement on therapeutic drug monitoring and individualization of linezolid. Front Public Health. 2022;10:967311. doi: 10.3389/fpubh.2022.967311 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Drugs@fda. Clinical pharmacology and biopharmaceutics review for NDA 021130. Available from: http://www.accessdata.fda.gov/. Accessed 18, April 2000.
- 8.Sakurai N, Kawaguchi H, Abe J, et al. Population pharmacokinetics of linezolid and its major metabolites PNU-142300 and PNU-142586 in adult patients. Pharmacotherapy. 2022;42(9):707–715. doi: 10.1002/phar.2719 [DOI] [PubMed] [Google Scholar]
- 9.Brier ME, Stalker DJ, Aronoff GR, et al. Pharmacokinetics of linezolid in subjects with renal dysfunction. Antimicrob Agents Chemother. 2003;47(9):2775–2780. doi: 10.1128/AAC.47.9.2775-2780.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kawasuji H, Tsuji Y, Ogami C, et al. Proposal of initial and maintenance dosing regimens with linezolid for renal impairment patients. BMC Pharmacol Toxicol. 2021;22(1):13. doi: 10.1186/s40360-021-00479-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Shi C, Xia J, Ye J, et al. Effect of renal function on the risk of thrombocytopaenia in patients receiving linezolid therapy: a systematic review and meta-analysis. Br J Clin Pharmacol. 2022;88(2):464–475. doi: 10.1111/bcp.14965 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Pea F, Cojutti PG, Baraldo M. A 10-year experience of therapeutic drug monitoring (TDM) of linezolid in a hospital-wide population of patients receiving conventional dosing: is there enough evidence for suggesting TDM in the majority of patients? Basic Clin Pharmacol Toxicol. 2017;121(4):303–308. doi: 10.1111/bcpt.12797 [DOI] [PubMed] [Google Scholar]
- 13.Souza E, Crass RL, Felton J, et al. Accumulation of major linezolid metabolites in patients with renal impairment. Antimicrob Agents Chemother. 2020;64(5). doi: 10.1128/AAC.00027-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.van den Anker J, Reed MD, Allegaert K, et al. Developmental changes in pharmacokinetics and pharmacodynamics. J Clin Pharmacol. 2018;58(Suppl 10):S10–s25. [DOI] [PubMed] [Google Scholar]
- 15.Freriksen JJM, van der Heijden JEM, de Hoop-Sommen MA, et al. Physiologically based pharmacokinetic (PBPK) model-informed dosing guidelines for pediatric clinical care: a pragmatic approach for a special population. Paediatr Drugs. 2023;25(1):5–11. doi: 10.1007/s40272-022-00535-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lin W, Chen Y, Unadkat JD, et al. Applications, challenges, and outlook for PBPK modeling and simulation: a regulatory, industrial and academic perspective. Pharm Res. 2022;39(8):1701–1731. doi: 10.1007/s11095-022-03274-2 [DOI] [PubMed] [Google Scholar]
- 17.Verscheijden LFM, Koenderink JB, Johnson TN, et al. Physiologically-based pharmacokinetic models for children: starting to reach maturation? Pharmacol Ther. 2020;211:107541. doi: 10.1016/j.pharmthera.2020.107541 [DOI] [PubMed] [Google Scholar]
- 18.Khalil F, Läer S. Physiologically based pharmacokinetic models in the prediction of oral drug exposure over the entire pediatric age range-sotalol as a model drug. Aaps J. 2014;16(2):226–239. doi: 10.1208/s12248-013-9555-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.US Food and Drug Administration. Clinical drug interaction studies ‐ study design, data analysis, implications for dosing, and labeling recommendations. Draft Guidance for Industry; Available from: https://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm292362.pdf. Accessed 18, April 2000. [Google Scholar]
- 20.European Medicines Agency. Draft guideline on the qualification and reporting of physiologically based pharmacokinetic (PBPK) modelling and simulation. 2016. Available from: https://www.ema.europa.eu/documents/scientific-guideline/draft-guideline-qualification-reporting-physiologically-based-pharmacokinetic-pbpk-modelling_en.pdf>. Accessed September 5, 2025.
- 21.Stalker DJ, Jungbluth GL, Hopkins NK, et al. Pharmacokinetics and tolerance of single- and multiple-dose oral or intravenous linezolid, an oxazolidinone antibiotic, in healthy volunteers. J Antimicrob Chemother. 2003;51(5):1239–1246. doi: 10.1093/jac/dkg180 [DOI] [PubMed] [Google Scholar]
- 22.Wen S, Zhang T, Yu X, et al. Bone penetration of linezolid in osteoarticular tuberculosis patients of China. Int J Infect Dis. 2021;103:364–369. doi: 10.1016/j.ijid.2020.11.203 [DOI] [PubMed] [Google Scholar]
- 23.Hosmann A, Moser MM, van Os W, et al. Linezolid brain penetration in neurointensive care patients. J Antimicrob Chemother. 2024;79(3):669–677. doi: 10.1093/jac/dkae025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wishart DS, Feunang YD, Guo AC, et al. DrugBank 5.0: a major update to the drugbank database for 2018. Nucleic Acids Res. 2018;46(D1):D1074–d82. doi: 10.1093/nar/gkx1037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Wynalda MA, Hauer MJ, LCJDm W, et al. Oxidation of the novel oxazolidinone antibiotic linezolid in human liver microsomes. Drug Metabol Disp. 2000;28(9):1014–1017. [PubMed] [Google Scholar]
- 26.Jones HM, Parrott N, Jorga K, et al. A novel strategy for physiologically based predictions of human pharmacokinetics. Clin Pharmacokinet. 2006;45(5):511–542. doi: 10.2165/00003088-200645050-00006 [DOI] [PubMed] [Google Scholar]
- 27.Zhang T, Heimbach T, Lin W, et al. Prospective predictions of human pharmacokinetics for eighteen compounds. J Pharm Sci. 2015;104(9):2795–2806. doi: 10.1002/jps.24373 [DOI] [PubMed] [Google Scholar]
- 28.Britz H, Hanke N, Volz AK, et al. Physiologically-based pharmacokinetic models for CYP1A2 drug-drug interaction prediction: a modeling network of fluvoxamine, theophylline, caffeine, rifampicin, and midazolam. CPT Pharmacometrics Syst Pharmacol. 2019;8(5):296–307. doi: 10.1002/psp4.12397 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mazzei T, Novelli A. Pharmacological rationale for antibiotic treatment of intra-abdominal infections. J Chemother. 2009;21 Suppl 1(sup1):19–29. doi: 10.1179/joc.2009.21.Supplement-1.19 [DOI] [PubMed] [Google Scholar]
- 30.Vardakas KZ, Kioumis I, Falagas ME. Association of pharmacokinetic and pharmacodynamic aspects of linezolid with infection outcome. Curr Drug Metab. 2009;10(1):2–12. doi: 10.2174/138920009787048446 [DOI] [PubMed] [Google Scholar]
- 31.Ross JE, Farrell DJ, Mendes RE, et al. Eight-year (2002-2009) summary of the linezolid (Zyvox ® annual appraisal of potency and spectrum; ZAAPS) program in European countries. J Chemother. 2011;23(2):71–76. doi: 10.1179/joc.2011.23.2.71 [DOI] [PubMed] [Google Scholar]
- 32.Laughon MM, Avant D, Tripathi N, et al. Drug labeling and exposure in neonates. JAMA Pediatr. 2014;168(2):130–136. doi: 10.1001/jamapediatrics.2013.4208 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Maharaj AR, Edginton AN. Physiologically based pharmacokinetic modeling and simulation in pediatric drug development. CPT Pharmacometrics Syst Pharmacol. 2014;3(11):e150. doi: 10.1038/psp.2014.45 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Rowland Yeo K, Gil Berglund E, Chen Y. Dose Optimization Informed by PBPK Modeling: state-of-the Art and Future. Clin Pharmacol Ther. 2024;116(3):563–576. doi: 10.1002/cpt.3289 [DOI] [PubMed] [Google Scholar]
- 35.Mackie C, Arora S, Seo P, et al. Physiologically based biopharmaceutics modeling (PBBM): best practices for drug product quality, regulatory and industry perspectives: 2023 workshop summary report. Mol Pharm. 2024;21(5):2065–2080. doi: 10.1021/acs.molpharmaceut.4c00202 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Frechen S, Rostami-Hodjegan A. Quality assurance of pbpk modeling platforms and guidance on building, evaluating, verifying and applying pbpk models prudently under the umbrella of qualification: why, when, what, how and by whom? Pharm Res. 2022;39(8):1733–1748. doi: 10.1007/s11095-022-03250-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Wu F, Shah H, Li M, et al. Biopharmaceutics applications of physiologically based pharmacokinetic absorption modeling and simulation in regulatory submissions to the U.S. food and drug administration for new drugs. Aaps j. 2021;23(2):31. doi: 10.1208/s12248-021-00564-2 [DOI] [PubMed] [Google Scholar]
- 38.Grimstein M, Yang Y, Zhang X, et al. Physiologically based pharmacokinetic modeling in regulatory science: an update from the U.S. food and drug administration’s office of clinical pharmacology. J Pharm Sci. 2019;108(1):21–25. doi: 10.1016/j.xphs.2018.10.033 [DOI] [PubMed] [Google Scholar]
- 39.Zhang X, Yang Y, Grimstein M, et al. Application of PBPK modeling and simulation for regulatory decision making and its impact on US prescribing information: an update on the 2018-2019 submissions to the US FDA’s office of clinical pharmacology. J Clin Pharmacol. 2020;60(Suppl 1):S160–s78. [DOI] [PubMed] [Google Scholar]
- 40.Suzuki Y, Aoki Y, Matsuyama Y, et al. Clinical evaluation of serum albumin reductivity in patients with renal dysfunction: a comparison between conservative renal failure patients and hemodialysis patients. Nihon Jinzo Gakkai shi. 2008;50(4):513–520. [PubMed] [Google Scholar]
- 41.Gandelman K, Zhu T, Fahmi OA, et al. Unexpected effect of rifampin on the pharmacokinetics of linezolid: in silico and in vitro approaches to explain its mechanism. J Clin Pharmacol. 2011;51(2):229–236. doi: 10.1177/0091270010366445 [DOI] [PubMed] [Google Scholar]
- 42.Bolhuis MS, van Altena R, van Soolingen D, et al. Clarithromycin increases linezolid exposure in multidrug-resistant tuberculosis patients. Eur Respir J. 2013;42(6):1614–1621. doi: 10.1183/09031936.00001913 [DOI] [PubMed] [Google Scholar]
- 43.Huo BN, Wu YE, Shu L, et al. Relationship between linezolid exposure and the typical clinical laboratory safety and bacterial clearance in chinese pediatric patients. Front Pharmacol. 2022;13:926711. doi: 10.3389/fphar.2022.926711 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Crass RL, Cojutti PG, Pai MP, et al. Reappraisal of linezolid dosing in renal impairment to improve safety. Antimicrob Agents Chemother. 2019;63(8). doi: 10.1128/AAC.00605-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zhang D, Xu Y, Wang X, et al. Risk factors for thrombocytopenia in patients receiving linezolid therapy: a systematic review and meta-analysis. Eur J Clin Pharmacol. 2023;79(10):1303–1314. doi: 10.1007/s00228-023-03542-z [DOI] [PubMed] [Google Scholar]
- 46.Hanai Y, Matsuo K, Ogawa M, et al. A retrospective study of the risk factors for linezolid-induced thrombocytopenia and anemia. J Infect Chemotherap. 2016;22(8):536–542. doi: 10.1016/j.jiac.2016.05.003 [DOI] [PubMed] [Google Scholar]
- 47.Jones SJ, Nichols KR, DeYoung HL, et al. Linezolid-associated thrombocytopenia in children with renal impairment. J Pedia Infect Dis Soc. 2015;4(3):272–275. doi: 10.1093/jpids/piu035 [DOI] [PubMed] [Google Scholar]
- 48.Morata L, De la Calle C, Gómez-Cerquera JM, et al. Risk factors associated with high linezolid trough plasma concentrations. Expert Opin Pharmacother. 2016;17(9):1183–1187. doi: 10.1080/14656566.2016.1182154 [DOI] [PubMed] [Google Scholar]
- 49.Tsuji Y, Holford NHG, Kasai H, et al. Population pharmacokinetics and pharmacodynamics of linezolid-induced thrombocytopenia in hospitalized patients. Br J Clin Pharmacol. 2017;83(8):1758–1772. doi: 10.1111/bcp.13262 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Matsumoto K, Shigemi A, Takeshita A, et al. Analysis of thrombocytopenic effects and population pharmacokinetics of linezolid: a dosage strategy according to the trough concentration target and renal function in adult patients. Int J Antimicrob Agents. 2014;44(3):242–247. doi: 10.1016/j.ijantimicag.2014.05.010 [DOI] [PubMed] [Google Scholar]
- 51.Zhou X, Dun J, Chen X, et al. Predicting the correct dose in children: role of computational pediatric physiological-based pharmacokinetics modeling tools. CPT Pharmacometrics Syst Pharmacol. 2023;12(1):13–26. doi: 10.1002/psp4.12883 [DOI] [PMC free article] [PubMed] [Google Scholar]











