REVIEW ARTICLE

Predictors of sirolimus pharmacokinetic variability identified using a nonlinear mixed effects approach: a systematic review

Janthima Methaneethorn1, 2*, Premsuda Art-arsa1, Ramanya Kosiyaporn1, Nattawut Leelakanok3

1Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, Thailand

2Center of Excellence for Environmental Health and Toxicology, Naresuan University, Phitsanulok, Thailand

3Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Burapha University, Chonburi, Thailand

Abstract

Several sirolimus (SRL) population pharmacokinetics (PopPK) were conducted to explain its pharmacokinetic variability, and the results varied across studies. Thus, we conducted a systematic review to summarize significant predictors influencing SRL pharmacokinetic variability. Moreover, discrepancies in model methodologies across studies were also reviewed and discussed. Four databases (PubMed, CINAHL Complete, Science Direct, and Scopus) were systematically searched. The PICO framework was used to identify eligible studies conducted in humans and employ a nonlinear-mixed effects strategy. Based on the inclusion and exclusion criteria, 20 studies were included. SRL pharmacokinetics were explained using 1- or 2-compartment models. Only one study assessed the model using an external approach, while the rest employed basic or advanced internal approaches. Significant covariates influencing SRL pharmacokinetics were bodyweight, age, CYP3A5 polymorphism, gender, BSA, height, cyclosporine dose or trough concentration, triglyceride, total cholesterol, hematocrit, albumin, aspartate aminotransferase, alanine aminotransferase, and total bilirubin. Of these, bodyweight, age, and CYP3A5 polymorphism were the three most identified significant predictors for SRL clearance. This review summarizes significant predictors to predict SRL clearance, which can subsequently be used to individualize SRL maintenance dose. However, the PopPK model selected for such prediction should be based on the resemblance of population characteristics between the target population and those used to conduct the model. Moreover, the predictability of the models in the target population should be assessed before implementation in clinical practice.

Key words: anticancer, immunosuppressant, nonlinear mixed-effects, population pharmacokinetics, sirolimus

*Corresponding author: Janthima Methaneethorn, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand. Email: janthima.methaneethorn@gmail.com

Submitted: 3 August 2022; Accepted: 2 September 2022; Published: 24 October 2022

DOI: 10.47750/jptcp.2022.940

©2022 Methaneethorn J, et al.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 International (CC BY-NC 4.0). License (http://creativecommons.org/licenses/by-nc/4.0/)

INTRODUCTION

Sirolimus (SRL) or rapamycin is an immunosuppressive agent approved for the prophylaxis of graft rejection in kidney transplant patients aged 13 years or more and for the treatment of patients with lymphangioleiomyomatosis.1 Moreover, SRL exerts anti-tumor action, and recently its derivative, temsirolimus (TEM) or CCI-779, has been developed to treat advanced renal cell carcinoma.2 Though SRL has a similar structure to tacrolimus and binds to FK-binding protein, SRL/FK-binding protein complex does not affect calcineurin phosphatase. Instead, it binds to mammalian targets of rapamycin (mTOR), leading to the inhibition of the progression of the cell cycle from the G1 to the S phase.1,3,4

Following oral administration in stable renal transplant patients, the time to peak concentration (Tmax) is variable, ranging from 0.5 to 3 hours,5 however, the bioavailability is low with a value of approximately 15.0%.5,6 In blood, SRL is preferentially distributed to red blood cells (94.5%) in a concentration-independent manner,7 with the mean blood to plasma ratio of 34.5:1 in stable kidney transplant patients receiving a single oral dose, nonetheless, this ratio is substantially variable ranging from 10 to 70.8 In contrast, in the plasma, approximately 40.0% of the drug is bound to lipoproteins,5 therefore, based on the nature of SRL, therapeutic drug monitoring (TDM) should be performed using whole blood as the appropriate biological matrix. Moreover, due to the lipophilic property, SRL is extensively distributed in lipid membranes of various organs, resulting in a high volume of distribution (Vd) of 5.6–16.7 L/kg in stable renal transplant patients.8

SRL is extensively metabolized by CYP3A4 and CYP3A5. Also, it is a substrate of P-glycoprotein (P-gp), which contributes to its low oral bioavailability.5 Moreover, CYP3A5 also plays a role in SRL metabolism.9 The drug exhibits large interindividual variability (IIV) in metabolism, with the apparent clearance ranging from 0.090-0.416 L/h/kg in stable renal transplant patients receiving a 14-day course of SRL with cyclosporine and prednisolone.8 The elimination half-life (t1/2) was approximately 62 hours;8 thus, the steady-state condition is achieved within 1-2 weeks.

Based on preclinical and clinical studies, the efficacy and adverse effects of SRL are related to blood concentrations, with trough concentrations at steady-state (Css,tr) of greater than 5 µg/L associated with an 89.5% negative predictive value for the occurrence of acute rejection episodes, while Css,tr levels greater than 15 µg/L are related to toxicity such as leucopenia, thrombocytopenia, and hypertriglyceridemia.10,14 This suggests that Css,tr of SRL should be maintained within the range of 5-15 µg/L to achieve an optimal therapeutic outcome,5 and thus TDM is an essential process during SRL therapy.

Since it takes approximately one week for SRL to reach steady-state condition, and a dosage adjustment based on the target Css,tr cannot be performed sooner, a population pharmacokinetic (PopPK) approach can be conducted to aid dosage individualization. This approach and the Bayesian estimation can provide individual pharmacokinetic parameter estimates for the optimization of SRL therapy. To date, several PopPK studies of SRL have been developed to characterize factors influencing SRL pharmacokinetic variability,9,1529 however, significant predictors obtained from these studies are not consistent; for example, some studies identified a significant effect of gender on SRL clearance (CLSRL)27 and Vd25, but other studies could not find such the effects.15,17,19,21,24 Based on the conflicting results, we aimed to systematically summarize factors that significantly influence SRL pharmacokinetic variability and their relationships with pharmacokinetic parameters. In addition, the disparity of model methodologies across studies was also reviewed and discussed.

METHODS

Database Searching and Study Selection

CINAHL Complete, PubMed, Science Direct, and SCOPUS databases were systematically searched to identify apposite studies. The search spanned the period from the database’s inception to May 2021. Search terms were developed using the PICO framework as follows: P: human studies, I: sirolimus OR Rapamune or rapamycin, C: none, O: “population pharmacokinetics” OR “pharmacokinetic model” OR “nonlinear mixed effect” OR NONMEM OR “interindividual variability” OR “intersubject variability” OR “residual variability” OR “intrasubject variability.” Reference lists were also screened for additional studies.

Title and abstracts were screened to exclude non-relevant articles. Screening of full-text articles was subsequently performed to identify studies to be included in this systematic review based on the following inclusion criteria: 1) PopPK studies conducted in humans, 2) SRL was used as a treatment drug, and 3) studies conducted using a nonlinear mixed-effects approach, while the exclusion criteria included: 1) non-English or non-Thai articles, 2) information on model development methodology was not sufficient, and 3) studies that were not original research articles. The Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guideline was adopted and followed during the review process.

Data Extraction

JM, PA, and RK independently extracted the data using the data abstraction form developed by JM. The extracted data were discussed, and the consensus was made by all authors. Three data categories were extracted, which included: 1) population characteristics such as study design, sample size, underlying diseases, age, body size, sex, functions of elimination organs, measurements of relevant laboratory values; 2) pharmacokinetic related information, that is, SRL dosage regimens, concurrent medications, analytical methods of SRL concentrations, and sampling strategy which is categorized into sparse sampling if the number of samples per patients was less than 6, otherwise it was considered an extensive sampling approach, and 3) model development methodologies and model evaluations. For model development, structural and statistical models for IIV and residual variability (RV) were summarized. In addition, tested and significant covariates on SRL pharmacokinetic parameters were compared across studies. Model evaluations were classified into three categories including basic internal, advanced internal, and external evaluation, as previously described by Brendel et al.30

Transparent Report and Clarity of the Included Studies

All reviewers independently assessed the study report quality using the Clinical PK checklist developed by Kanji et al.31 and the PopPK model-building strategies introduced by Dartois et al.32

RESULTS

Study Identification

Based on the systematic search, 992 articles were identified from all databases. Following the removal of duplicates, titles and abstracts of 904 non-redundant studies were screened, and 873 articles were excluded as irrelevant, leaving 31 studies for full-text assessment. Of these, 20 studies published between 1997 and 2021 met the inclusion criteria and were included in this review. Details on study exclusion are summarized and presented in a PRISMA diagram (Figure 1).

Figure 1. A PRISMA diagram of the study identification.

Population Characteristics, Study Design, and Pharmacokinetic Data

Most SRL PopPK studies were performed on various types of transplant patients, including kidney transplants,9,15,17,20,26 heart transplants,19 pancreatic islet transplants,18 and bone marrow transplants,22 while cancer was the second most disease in which SRL PopPK studies were conducted,21,27,28,3335 which included renal cell carcinoma, Kaposiform hemangioendothelioma, and several types of advanced or recurrent solid tumor, and of these two studies developed SRL PopPK models as an active metabolite of TEM.33,34 Other underlying diseases included immune cytopenia,29 tuberous sclerosis complex,36 and vascular anomalies.23,24 Moreover, one study was conducted solely on healthy Chinese subjects,25 and the other one was performed in both healthy Chinese and kidney transplant patients.37 In terms of age category, respective nine and 11 studies were conducted on children and adults. Most SRL PopPK studies were conducted to characterize SRL pharmacokinetics and its variability, but a few studies specifically aimed to determine the initial SRL dose using the developed PopPK models28,36 or create a Bayesian estimator for estimating an individual’s pharmacokinetic parameters.17 The sample sizes of the included studies ranged from 6 to 127 subjects, with half of the included studies retrospectively conducted using data from a clinical trial1921,23,34,37 and TDM data,2628,36 while the rest of the studies were prospective.9,15,17,18,22,24,25,29,33 Most studies used sparse sampling strategy,9,18,20,23,24,2629,36 whereas six and four studies employed an intensive sampling approach15,17,22,25,34,35 and a mixture of both intensive and sparse sampling.19,21,33,37 Concerning bioassay, most studies used liquid chromatography with tandem mass spectrometry (LCMS/MS),17,1921,2325,33,34,37 mass spectrometry (MS),15,22,33,35 or high performance liquid chromatography with ultraviolet detector (HPLC/UV),18,20 whereas the rest utilized various types of immunoassays.9,2629,36 Table 1 summarizes population characteristics, study design, and pharmacokinetic data.

Table 1. Population characteristics, study design, and pharmacokinetic data.

No Author Study design Study site Patient characteristics N (male) Mean age (range) years Mean weight
(range) kg
Mean BSA (range) m2 Route Sampling strategy Bioassay
1 Ferron et al., 199715 Randomized, double-blind, placebo-controlled ascending single-dose study Multicenter KT: high risk of chronic rejection 12 (11) 52.3 ± 24 73.9 ± 17.5 1.87 ± 10.1 Oral Intensive HPLC/MS
KT: compromise kidney function 12 (11) 51 ± 25.5 78.2 ± 28.7 1.90 ± 15.7
KT: stable transplant 12 (9) 43.3 ± 29.0 75.6 ± 18.9 1.88 ± 10.0
2 Boni et al., 200533 Randomized, double-blind, multicenter trial, once-weekly IV infusion Multicenter Advanced renal cell carcinoma 50 (33) 57.9 ± 9.6
(40-81)
82.9 ± 17.1
(53.7-124.7)
1.97 ± 0.2
(1.62-2.45)
IV inf. (30 min) Intensive and Sparse LCMS/MS
3 Djebli et al., 200617 Prospective Adult KT 22 (13) 48.6 (20-69) Wk 1: 60
(48-90)
Wk 2: 63
(48-90)
Mth 1: 58
(41-90)
Mth 2: 58
(41-90)
  Oral Intensive HPLC-MS/MS
4 Sato et al., 200618 Prospective Single-center Pancreatic islet transplant 6 (2) Med: 39
(35-58)
Med: 57
(37-30)
  Oral Sparse HPLC/UV
5 Zahir et al., 200619 Retrospective of heart transplant trial Multicenter Adult heart transplant with or without ischemic heart disease 31 (24) 49.0 ± 12.0
(18-66)
78.3 ± 12.7
(51-12.5)
  Oral Intensive and Sparse LCMS/MS
6 Jiao et al., 200920 Retrospective of clinical trial Multicenter Chinese adult KT 112 (78) 42 ± 9.9 60.4 ± 9.43 1.7 ± 0.144 Oral Intensive LCMS/MS and HPLC/UV
7 Wu et al., 201221 Retrospective of clinical trial Single-center Patients with advanced solid tumors 76 (39) 57.7
(22-83)
79.76
(32.8-154.6)
  Oral Sparse LCMS/MS
8 Goyal et al., 201322 Prospective Multicenter Pediatric blood and bone marrow transplant 37 (27) Med: 10.1 ± 5 (4-22) Med: 34.8 ± 19
(13.2-84.3)
  Oral Sparse HPLC/MS
9 Shi et al., 20169 Prospective Single-center Chinese adult KT 108 (79) 47±11
(21-72)
58.8 ± 5
(45-72)
  Oral Intensive MEIA
10 Emoto et al., 201623 Retrospective of clinical trial   Neonates and infants with complicated vascular anomalies 52 (20) Med: 4.8
(0.058-19)
Med: 18
(4-101)
Med: 0.77
(0.23-2.2)
  Sparse LCMS/MS
11 Mizuno et al., 201634 Retrospective of clinical trial   Children with recurrent solid tumor 19 (11) 11.9 ± 5.7
(21 days-19 years)
45.7 ± 28
(7.3-114.7)
  IV inf. (15-60 min) Intensive and Sparse LCMS/MS
12 Mizuno et al., 201724 Prospective Multicenter Pediatric patients with vascular anomalies 52 (20) Med:4.9
(0.1-18.6)
Med: 18.4
(4-100.6)
  Oral Intensive LCMS/MS
13 Wang et al., 201637 Retrospective of bioavailability and post-market study Multicenter Healthy Chinese and KT patients 127 (89) 37.44±1.02
(19-64)
62±0.91
(37-89)
  Oral Sparse LCMS/MS
14 Peng et al., 201825 Prospective Single-center Healthy Chinese subjects 27 (12) Med: 25.48
(20-36)
Med: 61.93
(45-75)
Med: 1.71
(1.45-1.93)
Oral Sparse LCMS/MS/MS
15 Golubovic et al., 201926 Retrospective of TDM data Single-center Adult KT 25 (18) 43.22 ± 12.62 (16-64) 77.07 ± 18.76
(44-128)
    Sparse CMIA
16 Wang et al., 201927 Retrospective of TDM data Single-center Pediatric Chinese patients with KHE 17 (11) 1.21 ± 1.2
(0.2-6)
7.99 ± 3.04
(3.6-18)
  Oral Sparse EMIT
17 Chen et al., 202028 Retrospective Single-center Pediatric Chinese patients with KHE 14 (9) 1.53 ± 1.40 8.87 ± 4.12   Oral Sparse EMIT
18 Chen et al., 202029 Prospective Single-center Children with immune cytopenia 27 (18) 8.16 ± 3.60 27.03 ± 10.87   Oral Sparse FPIA
19 Wang et al., 202036 Retrospective Single-center Children with tuberous sclerosis complex 15 (7) 6.16 ± 2.80 23.83 ± 8 .88   Oral Sparse EMIT
20 Sabo et al., 202135 Prospective Multicenter Pediatric oncology Day 1: 27 (16) 11.7 ± 5.9 38.5 ± 18.6   Oral Intensive Reverse phase LCMS
Day 8: 34 (21) 12.6 ± 5.6 40.7 ± 17.4  

BSA: body surface area, CMIA: Chemiluminescent microparticle immunoassay, EMIT: enzyme multiplied immunoassay technique, HPLC/MS: high performance liquid chromatography/mass spectrometry, HPLC-MS/MS: high performance liquid chromatography with tandem mass spectrometry, HPLC/UV: high performance liquid chromatography/ultraviolet, Inf: infusion, IV: intravenous, KHE: kaposiform hemangioendothelioma, KT: kidney transplant, LCMS/MS: liquid chromatography with tandem mass spectrometry, Med: median, MEIA: microparticle enzyme immunoassay, Mth: month, TDM: therapeutic drug monitoring, Wk: week.

Population Pharmacokinetic Model Development and Model Evaluation

Most studies conducted a PopPK model using NONMEM® software, except for four studies in which Phoenix NLME,25,29 Monolix,35 and P-Pharm15 were used. Ten studies developed a model with a 1-compartment structure,9, 1820,24,2729,35,36 while nine studies used a 2-CMT disposition,15,17,21,22,25,26,33,34,37 and the other one that explored the developmental trajectory of CLSRL employed a sigmoidal Emax model.23 The first-order absorption process was used in all models, except studies by Djebli et al.17 and Wu et al.21 in which the Erlang distribution and the Michaelis-Menten kinetics were used to explain the absorption process. In addition, one study reported an absorption lag-time of 0.24 h.16 Eight studies9,19, 20,24,2729,36 had to fix the absorption rate constant (ka) at 0.485, 0.752, 2.2, or 2.77 h1 since the information during the absorption phase was insufficient to estimate ka, whereas the estimated ka ranged from 0.0535 to 2.65 h1, with the IIV ranging from 17.5% to 80.0%. The estimated CLSRL without covariate effects ranged from 3.2 to 14.4 L/h, with a wider IIV range than the ka (11.4% to 103.0%). As for the Vd, the estimated values ranged from 88.9 to 3670 L for those with the 1-compartment structure, with the magnitude of IIV of 5.5% to 115.5%. While for the 2-compartment models, the central (Vc) and peripheral (Vp) volumes of distribution were in the range of 26.9 to 676.0 L and 72.8 to 1380 L, with the respective IIV of 7.8% to 164.0% and 10.3% to 38.7%. Concerning the statistical model, the IIV was modeled using an exponential relationship in all studies, except for one study in which an additive model was employed,15 while the proportional model was the most commonly used relationship for the RV, followed by a combined additive and proportional relationship.

Effects of numerous covariates on SRL pharmacokinetics were tested (Table 2), including age, body size (i.e., weight, height, body mass index; BMI, and body surface area; BSA), sex, SRL dose, TEM dose, duration of SRL therapy (DTT), enzyme polymorphisms, concurrent medication, postoperative days, and various laboratory measures such as hemoglobin (Hb), hematocrit (Hct), red blood cell (RBC), white blood cell (WBC), platelet, mean corpuscular hemoglobin (MCH), mean corpuscular hemoglobin concentration (MCHC), aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP), albumin (ALB), total protein (TP), bilirubin (BIL), serum creatinine (SCr), creatinine clearance (CLCR), total cholesterol (TC), triglyceride (TG), high-density lipoprotein (HDL), low-density lipoprotein (LDL), and glucose. Other investigated covariates included study site, ethnicity, graft origin, tumor age, dialysis before transplant, and the presence of ischemic heart disease (IHD). Of the tested covariates, weight was the significant covariate most commonly identified, whereas CYP3A5 polymorphisms and age were the second most commonly identified significant covariates. Other factors significantly affecting SRL pharmacokinetics were gender, BSA, height, cyclosporine concentration at time 0 h (CsA C0), TEM dose, TG, TC, Hct, ALB, AST, ALT, and BIL.

Table 2. Tested covariates for sirolimus pharmacokinetics.

No Author Tested Covariates
Age Body size Sex Blood profile LFT Lipid profile Protein Urea BIL RFT SRL dose CsA dose/conc. Polymorphism Concurrent med Other
1 Ferron et al., 199715 WT, HT, BSA Study site
2 Boni et al., 200533 NR NR NR NR NR NR NR NR NR NR NR NR NR NR NR
3 Djebli et al., 200617 WT, HT, BMI, BSA HCT, HB, RBC, WBC, platelet AST, ALT TC, TG TP, ALB SCr CYP3A5, CYP3A4, MDR1    
4 Sato et al., 200618 ND ND ND ND ND ND ND ND ND ND ND ND ND ND ND
5 Zahir et al., 200619 WT HCT, RBC   TC, TG, HDL, LDL TP, ALB   Ethnicity, IHD, POD
6 Jiao et al., 200920 WT, HT, BMI, BSA HCT, HB, RBC, WBC AST, ALT TC, TG, HDL, LDL   SCr, CLCR Organ source, POD
7 Wu et al., 201221 WT, HT HCT, HB, RBC, WBC, platelet AST, ALT, ALP TC, TG TP, ALB SCr, CLCR Glucose
8 Goyal et al., 201322 HB AST, ALT   ALB SCr
9 Shi et al., 20169 NR NR NR NR NR NR NR NR NR NR NR NR NR NR NR
10 Emoto et al., 201623 ND ND ND ND ND ND ND ND ND ND ND ND ND ND ND
11 Mizuno et al., 201634 ✓ (for TEM)   ✓ (for TEM)   TEM dose
12 Mizuno et al., 201724 NR NR NR NR NR NR NR NR NR NR NR NR NR NR NR
13 Wang et al., 201637 WT, HT, BMI AST SCr population
14 Peng et al., 201825 WT, HT, BSA HCT, HB, RBC, platelet AST, ALT TC, TG TP, ALB SCr     CYP3A5, MDR1    
15 Golubovic et al., 201926 WT HCT AST, ALT, ALP TC, TG TP SCr Graft origin, dialysis before transplant
16 Wang et al., 201927 WT HCT, HB AST, ALT     SCr Duration of treatment with SRL
17 Chen et al., 202028 HCT, HB, MCH, MCHC AST, ALT TP, ALB   SCr CYP3A5
18 Chen et al., 202029 WT HCT, HB, RBC, platelet AST, ALT, ALP TC, TG, LDL ALB SCr
19 Wang et al., 202036 WT HCT, HB, MCH, MCHC AST, ALT TP SCr ABCB1, ABCC4, ABCC8, ABCG2, CYP2C9, CYP2C19, CYP3A4, CYP3A5, CYP4F2, UGT1A1, UGT1A8, UGT2B15
20 Sabo et al., 202135 WT, HT, BMI, BSA   Tumor age, CNS tumor diagnostic, irinotecan dose on day 1, glucose

ABC: ATP binding cassette, ALB: albumin, ALP: alkaline phosphatase, ALT: alanine aminotransferase, AST: aspartate aminotransferase, BIL: bilirubin, BMI: body mass index, BSA: body surface area, CLCR: creatinine clearance, CsA: cyclosporine, CYP: cytochrome P450, HB: hemoglobin, HCT: hematocrit, HDL: high density lipoprotein, HT: height, IHD: ischemic heart disease, LDL: low density lipoprotein, LFT: liver function test, MCH: mean corpuscular hemoglobin, MCHC: mean cell hemoglobin concentration, MDR1: multidrug resistant protein 1, ND: not determined, NR: not reported, POD: postoperative day, RBC: red blood cell, RFT: renal function test, SCr: serum creatinine, SRL: sirolimus, TC: total cholesterol, TEM: temsirolimus, TG: triglyceride, TP: total protein, UGT: Uridine 5’-Diphospho-Glucuronosyl Transferase, WBC: white blood cell, WT: weight.

As for the model evaluation, all studies used basic and advanced internal approaches; however, one study assessed the model based solely on the basic internal method,22 and the other one employed all types of model evaluation techniques.26 Two studies did not have information on the model evaluation.15,18 Software, structural models, and model evaluation are presented in Table 3, while Table 4 summarizes the final models of the included studies and their magnitudes of IIV and RV.

Table 3. Software, structural models, and model evaluation.

No Author Software Structural model Model evaluation
1 Ferron et al., 199715 P-Pharm 2-CMT with first-order absorption and Lag-time NR
2 Boni et al., 200533 NONMEM 2-CMT with first-order formation into the central CMT (SRL was model as a metabolite of TEM) Basic and advanced internal evaluation
3 Djebli et al., 200617 NONMEM 2-CMT with Erlang distribution and first-order elimination Basic and advanced internal evaluation
4 Sato et al., 200618 NONMEM 1-CMT with first-order elimination NR
5 Zahir et al., 200619 NONMEM 1-CMT with first-order absorption and elimination Basic and advanced internal evaluation
6 Jiao et al., 200920 NONMEM 1-CMT with first-order absorption and elimination Basic and advanced internal evaluation
7 Wu et al., 201221 NONMEM 2-CMT with Michaelis-Menten absorption and first-order elimination Basic and advanced internal evaluation
8 Goyal et al., 201322 NONMEM 2-CMT with first-order absorption and elimination Basic internal evaluation
9 Shi et al., 20169 NONMEM 1-CMT with first-order absorption and elimination Basic and advanced internal evaluation
10 Emoto et al., 201623 NONMEM Sigmoidal Emax Basic and advanced internal evaluation
11 Mizuno et al., 201634 NONMEM Temsirolimus: 3-CMT with zero-order infusion
Sirolimus (as a metabolite): 2-CMT with first-order elimination
Basic and advanced internal evaluation
12 Mizuno et al., 201724 NONMEM 1-CMT with first-order input Basic and advanced internal evaluation
13 Wang et al., 201637 NONMEM 2-CMT with first-order absorption and elimination Basic and advanced internal evaluation
14 Peng et al., 201825 Phoenix NLME 2-CMT with first-order absorption and elimination Basic and advanced internal evaluation
15 Golubovic et al., 201926 NONMEM 2-CMT with first-order absorption and elimination Basic and advanced internal and external evaluation
16 Wang et al., 201927 NONMEM 1-CMT with first-order absorption and elimination Basic and advanced internal evaluation
17 Chen et al., 202028 NONMEM 1-CMT with first-order absorption and elimination Basic and advanced internal evaluation
18 Chen et al., 202029 Phoenix NLME 1-CMT with first-order absorption and elimination Basic and advanced internal evaluation
19 Wang et al., 202036 NONMEM 1-CMT with first-order absorption and elimination Basic and advanced internal evaluation
20 Sabo et al., 202135 Monolix 1-CMT with first-order absorption and elimination Basic and advanced internal evaluation

CMT: compartment, NR: not reported, SRL: sirolimus, TEM: temsirolimus.

Table 4. Final models, interindividual variability, and residual variability.

No Authors Final model RV (%CV)
Absorption IIV
(%CV)
Distribution IIV
(%CV)
Elimination IIV (%CV)
1 Ferron et al., 199715 ka (h-1) = 2.18 41.3% Vc (L) = 112.9 31.8% CL/F (L/h) = 8.91 38.2% Prop: NR
tlag (h) = 0.24 40.1% Vp (L) = 452 26.4%
2 Boni et al., 200533 ka (h-1) = 0.087 34.6% Vc (L) = 10.4 164% CL (L/h) = 2.05*Dose0.422 63.7% Prop: 23.24%
Vp (L) = 12.9*Dose0.302*HCT0.719 22.8% Q (L/h) = 44.1 NR
3 Djebli et al., 200617 ktr (h-1) = 5.25 42.7% Vc/F (L) = 218 52.7% CL/F(L/h) = 14.1 + 14.2*CYP3A5 49.3% Prop: 5.86%
Add: 3.08 ng/mL
Vp/F (L) = 292 20.2% Q/F (L/h) = 38.7 78.1%  
4 Sato et al., 200618,ε NA NA V/F (L) = 790 ± 659 NA CL/F (L/h) = (0.0776 + 0.167*POD/12)*WT for POD ≤ 12 NA NA
CL/F (L/h) = (0.0776 + 0.167)*WT for POD > 12
5 Zahir et al., 200619 ka (h-1) = 0.752 (fixed) NA (fixed) V/F (L) = 1350 NR CL/F (L/h) = 7.09 - 0.0147*CsA dose-1.37*(1-TG) + 2.2*(1-IHD) 27.5% Add: 24.1%
6 Jiao et al., 200920 ka (h-1) = 0.752 (fixed) NA (fixed) V/F (L) = 3670-7.27*(CsA C0-104) 56.7% CL/F (L/h) = [10.1-0.662*(TC-5.66)-0.00417*(CsA C0-104)] *0.65SLM *0.661GLZ *(DDS/2)0.479 23.8% Expo: 29.9%
7 Wu et al., 201221 Vmax(µg/L*h) = 4.56 NE Vc/F (L) = 53.4 52.4% CL/F (L/h) = 12.9*(35.1/HCT)0.14 52.4% Prop: 2.17%
Add: 0.5 ng/mL
Km (mg) = 13.8 NE Vp/F (L) = 611 19.3% Q/F (L/h) = 29.0 52.4%  
8 Goyal et al., 201322 ka (h-1) = 0.0535±0.0104 NR Vc/F (L) = 26.9±7.7 91% CL/F (L/h) = 6.66±1.10 78% Prop: 21%
Add: 0.84 ng/mL
    Vp/F (L) = 630±171 NR Q/F (L/h) = 4.62±2 NR  
9 Shi et al., 20169 ka (h-1) = 2.20 (fixed) NA (fixed) Vd/F (L) = 322 22.6 CL/F (L/h) = 14.4*(1+WT/58.6*0.19)*exp((-ALB/38.9)*0.26)*exp(CYP3A5*-0.30) 19.6 Expo: 25.6%
10 Emoto et al., 201623         CL = CLmatured * PMAHill/+ PMAHill
CLmatured (L/h/70 kg) = 18.7
11.3% Prop: 25.7%
TM50 (weeks) = 62.9 17.4%
Hill = 2.94 102%
11 Mizuno et al., 201634 CLTEM (L/h/70 kg) = 3.4*Dose0.855 70.9% Vc (L/h/70kg) = 48 121% CLSRL (L/h/70kg) = 6.08 103% Prop: 25.5%
Add: 1.69 ng/mL
Vp (L/h/70kg) = 72.8 NE Q (L/h/70kg) = 11.6 NE
12 Mizuno et al., 201724 ka (h-1) = 2.77 fixed NA (fixed) Vpediatric = Vadult*(BW/70)
Vadult = 1030
62.3% CLpediatric = CLadult*(BW/70)0.73*MF
MF = PMAHill/(0 + PMAHILL)
CLadult = 18.5
Hill = 2.94
31.2% Prop: 38.1%
13 Wang et al., 201637 ka (h-1) = 0.24 0 Vc/F (L) = 676*(SCR/592.3)1.4 10.3% CL/F (L/h) = 8.81*[1-0.219*(CsA/300)] * [1-0.0171*(age-40)] 50.9% Prop: 62.2%
Vp/F (L) = 1380 103.4% Q/F (L/h) = 32.9 12.7%
14 Peng et al., 201825 ka (h-1) = 2.651   Vc = 184.461 for female
Vc = 184.461*exp(0.266) for male
7.8 CL = 10.813*(CYP3A5*1/*1)
CL = 10.813*exp(-0.034)*(CYP3A5*1/*3)
CL = 10.813*exp(-0.250)*(CYP3A5*3/*3)
11.4% Prop: 17.5%
Vp = 170.029*BSA2.165 38.7 Q (L/h) = 23.596 7.6%
15 Golubovic et al., 201926 ka (h-1) = 2.19 38.1% Vc/F (L) = 118 55.3 CL/F (L/h) = 12.2*0.63AST*(1-(age/44)*0.388)
AST = 0 if =< 37 IU/L
AST=1 if >37 IU/L
23.4% Prop: 24.9%
Add: 1.93 ng/mL
Vp/F (L) = 609 25.6 Q/F (L/h) = 5.07 32.1%
16 Wang et al., 201927 ka (h-1) = 0.485 (fixed) NA (fixed) V/F (L) = 165*exp(0.0783*DTT/10) 115.5 CL/F (L/h) = 3.19*exp(0.215*age)*exp(0.0108*ALT)*exp(-0.818*sex)
Sex = 1 for female, 0 otherwise
46.6% Expo: 60.4%
17 Chen et al., 202028 ka (h-1) = 0.485 (fixed) NA (fixed) V/F (L) = 1840*(WT/70) NR CL/F (L/h) = 7.55*(WT/70)0.75*(1-(-0.999)*CYP3A5) 59.0% Prop: 62.4%
18 Chen et al., 202029 ka (h-1) = 0.752 (fixed) NA (fixed) V/F (L) = 144.16 42.9% CL/F (L/h) = 5.63*(TBIL/11.29)-0.32*(WT/28.5)0.5 21.9% NR: 11.8%
19 Wang et al., 202036 ka (h-1) = 0.485 (fixed) NA (fixed) V/F (L) = 124*(WT/70) 5.5% CL/F (L/h) = 6.48*(WT/70)0.75 25.7% Prop: 55.9%
Add: 1.25 ng/mL
20 Sabo et al., 202135 For day 1: ka (h-1) = 0.46 80% For day 1: V/F (L/h) = 88.9*(BSAi/Med BSA)1.35 62.5% For day 1: CL/F (L/h) = 23.9*(BSAi/Med BSA) 92.9% Prop:
Day 1: 0.54%
For day 8: ka (h-1) = 0.97 168.5% For day 8: V/F (L/h) = 238*(BSAi/Med BSA)1.41 29.4% For day 8: CL/F (L/h) = 11.9*(BSAi/Med BSA)1.09 50.1% Prop: Day 8: 0.31%

εThis study compared two models and did not determine the magnitude and sources of variability ALB: albumin, AST: aspartate aminotransferase, BSA: body surface area, BSAi: individual body surface area, CL: clearance, CLmatured: clearance at fully matured level, CsA C0: cyclosporine trough concentration, CsA: Cyclosporine, CYP: cytochrome P450, DDS: daily dose sirolimus, F: bioavailability, GLZ: glycyrrhizin, HCT: hematocrit, IHD: ischemic heart disease, ka: absorption rate constant, km: sirolimus amount at 50% of Vmax, ktr: transfer rate constant, Med: median, MF: maturation function, NA: not applicable, NE: Not estimated, PMA: postmenstrual age, POD: postoperative day, Q: intercompartmental clearance, SLM: silymarin, SRL: sirolimus, TBIL: total bilirubin, TC: total cholesterol, TEM: temsirolimus, TG: triglyceride, tlag: absorption lag time, TM: postmenstrual age at which clearance is half of CLmatured, Vc: central volume of distribution, Vmax: maximum absorption rate, Vp: peripheral volume of distribution, WT: weight.

Transparent Report of the Included Studies

Most studies complied with the guideline of the transparent report for clinical pharmacokinetic studies,31,32 with a compliance rate greater than 80%. Only one study had a compliance rate of approximately 60%,18 while four studies reported their results with compliance rates between 70% to 80%.16,29,36,38 The most common non-reported items of each section, identified in more than 10 studies were “route of administration” in the introduction section, “formulation details” and “sample storage” in the method section, and “study withdrawals or lost to follow-up” in the result section.

DISCUSSION

Several PopPK models of SRL have been conducted to determine factors influencing its pharmacokinetic variability. The impacts of these factors on SRL pharmacokinetics were summarized and discussed below.

Absorption

The rate of SRL absorption was variable, with the estimated ka ranging from 0.0535 to 2.65 h1, which was consistent with a traditional pharmacokinetic study in stable transplant patients that reported a wide range of Tmax of 0.5–3 h.39 The difference in ka among studies could not be clearly explained, however, it has been reported that administration of SRL with food results in a 3.5-fold increase in Tmax, while the maximum concentration (Cmax) is decreased by 34%.1 The administration process of some of the included studies, e.g., fasted or fed, was not described and might contribute to such difference. Though the magnitude of IIV on ka ranged from 17.5% to 80.0%, no studies identified significant predictors for ka, and this was consistent with a study by Kahan et al. that reported no association between sex, age, weight, or ethnicity and Cmax, the minimum concentration at steady-state (Cmin,ss), or AUC.14 Notably, the slowest ka of 0.0535 h1 was reported by a study conducted in pediatric bone marrow transplant patients whose ages ranged from 4 years to 22 years, and some patients were co-administered fluconazole.22 It has been shown that neonates and infants have longer gastric emptying, which can delay the rate of drug absorption, however, the age at which gastric emptying time approaches that in adults was not specifically determined.40

Distribution

The estimated Vd for the 1-compartment studies ranged from 88.9 L to 1840 L, excluding the one with a substantially high value of 3670 L, while those of the 2-compartment studies had the Vd of 120.8 L to 2056 L. The large Vd of SRL can be explained by its lipophilic property, which contributes to the distribution of the lipid membrane of various tissues.5 The ranges of Vd from SRL PopPK studies were more comprehensive than those of the traditional pharmacokinetics conducted in stable renal transplant patients (392 L to 1169 L for a 70 kg patient),8 which could be due to different patients’ characteristics. PopPK studies that reported the low Vd values (88.9 L to 165 L) were conducted in children,27,29,35,36 and evidence has indicated that children contain lower fat mass than adults.41

Significant predictors for SRL Vd included CsA C0,20 weight,24,28,36 BSA,35 Scr,37 sex,25 and DTT.27 Jiao et al.20 indicated that 1 ng/mL increase in CsA C0 from the median value of 104 ng/mL resulted in a decrease in the apparent volume of distribution (Vd/F) of 7.27 L. CsA is a substrate and an inhibitor of intestinal CYP3A4 and P-gp,42 thus co-administration of CsA and SRL increased SRL bioavailability, and in turn, a decrease in Vd/F.

The effect of weight on Vd was explained using an allometric scaling relationship with the exponent of one, suggesting that the Vd is linearly related to weight. This relationship is well accepted since it is based on physiologic principles describing size in relation to blood volume and vital capacity.43 Another study; however, used BSA instead of weight as an index of body size with the exponent of 1.35 and 1.09 for Vd on day 1 and Vd on day 8.35 This was deemed feasible since this study was conducted in pediatric patients with solid tumors in which SRL dose was titrated based on BSA.

Wang et al. reported a nonlinear increase in Vc/F with an increase in SCr.37 This could be rationalized by a decrease in plasma protein binding in patients with impaired renal function,44 increasing free SRL concentrations that can distribute to red blood cells. Moreover, Peng et al. reported higher Vc of SRL in males than in females, which was expected given that males generally have larger body configurations than females.25 As for the DTT, Vd of SRL increased as the DTT increased, which could be due to an increase in the number of erythrocytes with the improvement of clinical outcomes as the duration of treatment was lengthened.27

Elimination

Significant predictors for CLSRL were different among studies. Jiao et al.20 reported a nonlinear increase in CLSRL/F with an increase in SRL dose. However, this could be due to the TDM effects since subjects with higher CLSRL/F tend to receive higher SRL doses. This effect was previously described by Ahn et al.45 Boni et al.33 also reported a similar effect using a model conducted in patients with advanced renal cancer receiving TEM, in which CLSRL increased with an increase in TEM dose.

Several studies found a significant effect of CsA on CLSRL. Zahir et al.19 identified that a 100 mg increase in CsA dose led to approximately 20.7% decrease in CLSRL/F, while Wang et al.37 reported a smaller effect of a 7.3% decrease in CLSRL/F. Moreover, Jiao et al.20 found a 4.5% decrease in CLSRL/F for a 100 ng/mL increase in CsA C0. CsA is an inhibitor of CYP3A4 and P-gp. Thus concomitant administration of CsA and SRL leads to an increase in SRL bioavailability and in turn, a decrease in CLSRL/F.42 Therefore, in patients undergoing CsA dose minimization, the SRL dose should be increased by 7% to 20% for a 100 mg decrease in the CsA dose.

Significant effects of several laboratory values were identified. First, an increase in Hct contributed to a modest decrease in CLSRL/F.21 This effect was previously reported for tacrolimus.46 Since SRL is concentrated in erythrocytes,7 as Hct increases, free SRL levels available for elimination decrease. Second, CLSRL/F is decreased as TC20 or TG increases.19 This could be due to increased SRL bioavailability following high-fat meals since patients with hyperlipidemia tend to consume high-fat meals.19,20 Zahir et al.19 also proposed that SRL may be a low intrinsic clearance drug in which hepatic clearance depends on fraction unbound. Patients with hyperlipidemia may have a lower unbound SRL fraction since the drug extensively distributes across the plasma membrane and binds to erythrocytes, decreasing hepatic clearance. The authors also reported that heart transplant patients with non-IHD had lower CLSRL/F than IHD patients, which might also be associated with dyslipidemia since IHD patients tend to consume a high-fat diet. Nonetheless, this covariate (IHD) can be confounded by the effect of TG and should be interpreted with a caveat. Third, a one-fold of ALB lower than average level contributed to a modest increase in CLSRL/F of 17%.9 This would be expected since it increased the free SRL fraction available for elimination. Fourth, and impaired liver function, expressed as AST above upper limit normal contributed to lower CLSRL/F,26 which is not surprising given that SRL is extensively metabolized by the liver. Fifth, Cheng et al. reported that the higher BIL was associated with, the lower CLSRL/F; however, underlying mechanism of this effect could not be clarified.29

Several studies identified a significant association between weight and CLSRL/F, with an increase in CLSRL/F with bodyweight.9,24,28,29,36 This association is commonly described using a power relationship43 and is widely applied since higher body weight may relate to larger elimination organs. Whereas Sabo et al.35 reported a significant association between BSA and CLSRL/F in pediatric patients with solid tumors using a power relationship, which is deemed appropriate given that SRL dose is given based on BSA in patients with cancer. Moreover, one study reported that females had approximately 40% lower CLSRL/F than males,27 which is incongruent with a physiological basis that females have lower body weight, corresponding to smaller elimination organs.

Glolubovic et al. identified that CLSRL/F of adults decreased with advancing age,26 which is in agreement with physiological basis, while Wang et al.27 reported an increase in CLSRL/F with age in a pediatric population aged 0.2-6 years. This was expected based on the development of elimination organs that approaches adults with increasing age. Moreover, Emoto et al.23 described the developmental trajectory of CLSRL in neonates and infants using postmenstrual age (PMA) and a sigmoidal Emax model, which could aid dosing recommendations in this population. Based on their model, CLSRL approached the mature level at the PMA of approximately 144-196 weeks.

CYP3A5 polymorphisms significantly influence CLSRL/F, as Djebli et al.17 and Chen et al.28 found that non-expressers (CYP3A5*3/*3) had approximately 50% lower CLSRL/F than expressers (CYP3A5*1/*1 and CYP3A5*1/*3). With a similar trend, Shi et al.9 and Peng et al.25 reported that patients carrying CYP3A5*1/*1, CYP3A5*1/*3, CYP3A5*3/*3 had a ratio of CLSRL/F of 1: 0.74: 0.55 and 1: 0.96: 0.78, respectively. This effect could assist SRL dosing when CYP3A5 genotyping is available.

In conclusion, PopPK models of SRL conducted using a nonlinear mixed-effects approach were summarized, and significant predictors for CLSRL were identified. These models, with Bayesian forecasting, can be used to guide SRL dosage individualization. However, the choice of model selection should be based on the characteristics of the target population in which the model is to be used. Moreover, most models were not externally evaluated. Therefore, the predictive performance of such models should be assessed before applying them in clinical practice.

Conflict of Interest

The authors declare no conflict of interest.

Authorship contributions

Conceptualizations: Janthima Methaneethorn. Data screening and extraction: Janthima Methaneethorn, Premsuda Art-arsa, and Ramanya Kosiyaporn. Quality assessment: Janthima Methaneethorn, Premsuda Art-arsa, Ramanya Kosiyaporn, and Nattawut Leelakanok. Drafting the manuscript: Janthima Methaneethorn. Reviewing and approving the manuscript: Janthima Methaneethorn, Premsuda Art-arsa, Ramanya Kosiyaporn, and Nattawut Leelakanok.

REFERENCES

1. Rapamune [package insert]. Philadelphia, PA: Wyeth-Ayerst Pharmaceuticals; 2017.

2. Rini BI. Temsirolimus, an inhibitor of mammalian target of rapamycin. Clin Cancer Res. 2008;14(5):1286–90. 10.1158/1078-0432.CCR-07-4719

3. Stenton SB, Partovi N, Ensom MH. Sirolimus. Clin Pharmacokinet. 2005;44(8):769–86. 10.2165/00003088-200544080-00001

4. MacDonald A, Scarola J, Burke JT, Zimmerman JJ. Clinical pharmacokinetics and therapeutic drug monitoring of sirolimus. Clin Ther. 2000;22:B101–B21. 10.1016/s0149-2918(00)89027-x

5. Mahalati K, Kahan BD. Clinical pharmacokinetics of sirolimus. Clin Pharmacokinet. 2001;40(8):573–85. 10.2165/00003088-200140080-00002

6. Yatscoff R. Pharmacokinetics of rapamycin. Transplant Proc. 1996;28(2):970–3.

7. Yatscoff R, LeGatt D, Keenan R, Chackowsky P. Blood distribution of rapamycin. Transplantation. 1993;56(5):1202–6. 10.1097/00007890-199311000-00029

8. Zimmerman JJ, Kahan BD. Pharmacokinetics of sirolimus in stable renal transplant patients after multiple oral dose administration. J Clin Pharmacol. 1997;37(5):405–15. 10.1002/j.1552-4604.1997.tb04318.x

9. Shi HQ, Yang J, Zhang LQ, et al. The population pharmacokinetics of sirolimus and CYP3A5*3 polymorphism in Chinese renal transplant patients. Int J Clin Exp Med. 2016;9(3):5854–66. 10.1111/j.1365-2125.2009.03392.x

10. Fryer J, Yatscoff RW, Pascoe EA, et al. The relationship of blood concentrations of rapamycin and cyclosporine to suppression of allograft rejection in a rabbit heterotopic heart transplant model. Transplantation. 1993;55(2):340–5. 10.1097/00007890-199302000-00021

11. Granger DK, Cromwell JW, Chen SC, et al. Prolongation of renal allograft survival in a large animal model by oral rapamycin monotherapy. Transplantation. 1995;59(2):183–6.

12. Kahan BD, Julian BA, Pescovitz MD, et al. Sirolimus reduces the incidence of acute rejection episodes despite lower cyclosporine doses in Caucasian recipients of mismatched primary renal allografts: a phase II trial. Transplantation. 1999;68(10):1526–32. 10.1097/00007890-199911270-00016

13. Kahan BD, Podbielski J, Napoli KL, et al. Immunosuppressive effects and safety of a sirolimus/cyclosporine combination regimen for renal transplantation. Transplantation. 1998;66(8):1040–6. 10.1097/00007890-199810270-00013

14. Kahan B, Napoli K, Kelly P, et al. Therapeutic drug monitoring of sirolimus: correlations with efficacy and toxicity. Clin Transplant. 2000;14(2):97–109. 10.1034/j.1399-0012.2000.140201.x

15. Ferron GM, Mishina EV, Zimmerman JJ, et al. Population pharmacokinetics of sirolimus in kidney transplant patients. Clin Pharmacol Ther. 1997;61(4):416–28. 10.1016/S0009-9236(97)90192-2

16. Ferron GM, Mishina EV, Jusko WJ, et al. Population pharmacokinetics of sirolimus. Clin Pharmacol Ther. 1998;63(4):494. 10.1111/j.1365-2125.2009.03392.x

17. Djebli N, Rousseau A, Hoizey G, et al. Sirolimus population pharmacokinetic/pharmacogenetic analysis and bayesian modelling in kidney transplant recipients. Clin Pharmacokinet. 2006;45(11):1135–48. 10.2165/00003088-200645110-00007

18. Sato E, Shimomura M, Masuda S, et al. Temporal decline in sirolimus elimination immediately after pancreatic islet transplantation. Drug Metab Pharmacokinet. 2006;21(6):492–500.

19. Zahir H, Keogh AM, Akhlaghi F. Apparent clearance of sirolimus in heart transplant recipients: impact of primary diagnosis and serum lipids. Ther Drug Monit. 2006;28(5):614–22. 10.1097/01.ftd.0000246765.05248.fa

20. Jiao Z, Shi XJ, Li ZD, et al. Population pharmacokinetics of sirolimus in de novo Chinese adult renal transplant patients. Br J Clin Pharmacol. 2009;68(1):47–60. 10.1111/j.1365-2125.2009.03392.x

21. Wu K, Cohen EE, House LK, et al. Nonlinear population pharmacokinetics of sirolimus in patients with advanced cancer. CPT: Pharmacomet Syst Pharmacol. 2012;1(12):e17. 10.1038/psp.2012.18

22. Goyal RK, Han K, Wall DA, et al. Sirolimus pharmacokinetics in early postmyeloablative pediatric blood and marrow transplantation. Biol Blood Marrow Transplant. 2013;19(4):569–75. 10.1016/j.bbmt.2012.12.015

23. Emoto C, Fukuda T, Mizuno T, et al. Characterizing the developmental trajectory of sirolimus clearance in neonates and infants. CPT: Pharmacomet Syst Pharmacol. 2016;5(8):411–7. 10.1002/psp4.12096

24. Mizuno T, Emoto C, Fukuda T, et al. Model-based precision dosing of sirolimus in pediatric patients with vascular anomalies. Eur J Pharm Sci. 2017;109s:s124–s31. 10.1016/j.ejps.2017.05.037

25. Peng M, Zhao G, Li X, et al. Population pharmacokinetics of sirolimus in healthy Chinese subjects. Indian J Pharm Sci. 2018;80(2):291–7. 10.4172/pharmaceutical-sciences.1000357

26. Golubović B, Vučićević K, Radivojević D, et al. Exploring sirolimus pharmacokinetic variability using data available from the routine clinical care of renal transplant patients-population pharmacokinetic approach. J Med Biochem. 2019;38(3):323–31. 10.2478/jomb-2018-0030

27. Wang D, Chen X, Li Z. Population pharmacokinetics of sirolimus in pediatric patients with kaposiform hemangioendothelioma: a retrospective study. Oncol Lett. 2019;18(3):2412–9. 10.3892/ol.2019.10562

28. Chen X, Wang DD, Xu H, et al. Initial dose recommendation for sirolimus in paediatric kaposiform haemangioendothelioma patients based on population pharmacokinetics and pharmacogenomics. J Int Med Res. 2020;48(8):300060520947627. 10.1177/0300060520947627

29. Cheng X, Zhao Y, Gu H, et al. The first study in pediatric: population pharmacokinetics of sirolimus and its application in Chinese children with immune cytopenia. Int J Immunopathol Pharmacol. 2020;34:2058738420934936. 10.1177/2058738420934936

30. Brendel K, Dartois C, Comets E, et al. Are population pharmacokinetic and/or pharmacodynamic models adequately evaluated? Clin Pharmacokinet. 2007;46(3):221–34. 10.2165/00003088-200746030-00003

31. Kanji S, Hayes M, Ling A, et al. Reporting guidelines for clinical pharmacokinetic studies: the ClinPK statement. Clin Pharmacokinet. 2015;54(7):783–95. 10.1007/s40262-015-0236-8

32. Dartois C, Brendel K, Comets E, et al. Overview of model-building strategies in population PK/PD analyses: 2002–2004 literature survey. Br J Clin Pharmacol. 2007;64(5):603–12. 10.1111/j.1365-2125.2007.02975.x

33. Boni JP, Leister C, Bender G, et al. Population pharmacokinetics of CCI-779: correlations to safety and pharmacogenomic responses in patients with advanced renal cancer. Clin Pharmacol Ther. 2005;77(1):76–89.

34. Mizuno T, Fukuda T, Christians U, et al. Population pharmacokinetics of temsirolimus and sirolimus in children with recurrent solid tumours: a report from the Children’s oncology group. Br J Clin Pharmacol. 2017;83(5):1097–107. 10.1111/bcp.13181

35. Sabo AN, Jannier S, Becker G, et al. Sirolimus pharmacokinetics variability points to the relevance of therapeutic drug monitoring in pediatric oncology. Pharmaceutics. 2021;13(4):470. 10.3390/pharmaceutics13040470

36. Wang DD, Chen X, Xu H, et al. Initial dosage recommendation for sirolimus in children with tuberous sclerosis complex. Front Pharmacol. 2020;11:890. 10.3389/fphar.2020.00890

37. Wang M, Bao-ling D, Yin-jin Y, et al. Population pharmacokinetic characteristics of sirolimus in healthy Chinese subjects and renal transplant patients. Int J Clin Pharmacol Ther. 2016;54(6):433. 10.5414/cp202499

38. Boni JP, Zhou S, Burns J, et al. 552 POSTER integrated population pharmacokinetic analysis of temsirolimus in cancer patients following weekly IV treatments. Eur J Cancer Suppl. 2006;4(12):167–8. 10.1053/j.seminoncol.2009.10.009

39. Johnson EM, Zimmerman J, Duderstadt K, et al. A randomized, double-blind, placebo-controlled study of the safety, tolerance, and preliminary pharmacokinetics of ascending single doses of orally administered sirolimus (rapamycin) in stable renal transplant recipients. Transplant Proc. 1996;28(2):987.

40. Benedetti MS, Whomsley R, Baltes EL. Differences in absorption, distribution, metabolism and excretion of xenobiotics between the paediatric and adult populations. Expert Opin Drug Metab Toxicol. 2005;1(3):447–71. 10.1517/17425255.1.3.447

41. Weber DR, Leonard MB, Zemel BS. Body composition analysis in the pediatric population. Pediatr Endocrinol Rev. 2012;10(1):130–9.

42. Zimmerman JJ, Harper D, Getsy J, et al. Pharmacokinetic interactions between sirolimus and microemulsion cyclosporine when orally administered jointly and 4 hours apart in healthy volunteers. J Clin Pharmacol. 2003;43(10):1168–76. 10.1177/0091270003257227

43. Anderson BJ, Holford NH. Mechanism-based concepts of size and maturity in pharmacokinetics. Annu Rev Pharmacol Toxicol. 2008;48:303–32. 10.1146/annurev.pharmtox.48.113006.094708

44. Gibaldi M. Drug distribution in renal failure. Am J Med. 1977;62(4):471–4. 10.1016/0002-9343(77)90399-0

45. Ahn JE, Birnbaum AK, Brundage RC. Inherent correlation between dose and clearance in therapeutic drug monitoring settings: possible misinterpretation in population pharmacokinetic analyses. J Pharmacokinet Pharmacodyn. 2005;32(5-6):703-18. 10.1007/s10928-005-0083-6

46. Staatz CE, Willis C, Taylor PJ, et al. Population pharmacokinetics of tacrolimus in adult kidney transplant recipients. Clin Pharmacol Ther. 2002;72(6):660-9. 10.1067/mcp.2002.129304