Open Access

Initial dosage optimization of ciclosporin in pediatric Chinese patients who underwent bone marrow transplants based on population pharmacokinetics

  • Authors:
    • Xiao Chen
    • Xin Yu
    • Dong‑Dong Wang
    • Hong Xu
    • Zhiping Li
  • View Affiliations

  • Published online on: May 8, 2020     https://doi.org/10.3892/etm.2020.8732
  • Pages: 401-408
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Bone marrow transplants (BMT) are an established therapeutic strategy for patients with severe aplastic anemia, acute lymphoblastic leukemia, acute myeloid leukemia or chronic myeloid leukemia. However, the successful application of BMT is limited by graft‑vs.‑host disease (GVHD). Ciclosporin has been widely used for treating GVHD in pediatric patients who underwent BMT. The present study aimed to optimize the dosage of ciclosporin for safety and effectiveness based on population pharmacokinetics. A non‑linear mixed‑effects model was used to analyze the clinical data of pediatric patients who underwent BMT between September 2016 and September 2019 at the Children's Hospital of Fudan University. Monte Carlo simulations were used to identify the optimal dose of ciclosporin. The final population pharmacokinetic model indicated that body weight and days post‑transplant influenced the clearance of ciclosporin in pediatric patients who underwent BMT. The present study indicated that the optimal initial dose of ciclosporin for pediatric patients weighing 5‑30 kg who underwent BMT was 6 mg/kg/day split into 2 doses.

Introduction

Bone marrow transplants (BMT) are used to treat patients with severe aplastic anemia (1), acute lymphoblastic leukemia (2), acute myeloid leukemia (3) and chronic myeloid leukemia (4,5). However, the clinical application of BMT is limited by graft-vs.-host disease (GVHD) (6). Acute GVHD is a common complication which results in severe morbidity and mortality following BMT and has an occurrence rate of 30-50% (7-9). Acute GVHD primarily occurs in the skin, intestines and liver (10). Cytokine dysregulation resulting from an allogeneic interaction causes tissue injury that is characteristic of acute GVHD (11,12). Therefore, identifying a novel therapeutic strategy for GVHD is required.

Ciclosporin, an immunosuppressant drug, has been widely used as a treatment strategy for GVHD in pediatric patients who have undergone BMT (12-14); however, the therapeutic range is relatively narrow (15) and the drug exhibits wide inter-individual pharmacokinetic variability (16-18). Identifying the optimal dose regimen of ciclosporin to achieve and maintain the target concentration of the drug is crucial (19,20).

Population pharmacokinetics is a tool that can be used to collect sparse clinical data in order to model and simulate approaches to assess dosing regimens in specific patients (16,18). Numerous studies have established population pharmacokinetics of ciclosporin. For example, Ni et al (21) established population pharmacokinetics of ciclosporin in Chinese children with aplastic anemia, Fanta et al (22) and Irtan et al (23) in patients receiving pediatric renal transplants and Wilhelm et al (24) in patients receiving hematopoietic allogeneic stem cell transplants. Therefore, the present study aimed to optimize the initial dosage of ciclosporin for safety and effectiveness in pediatric Chinese patients who underwent BMT based on population pharmacokinetics.

Materials and methods

Patients and data collection

Pediatric patients who underwent BMT between September 2016 and September 2019 at the Children's Hospital of Fudan University (Shanghai, China) were retrospectively recruited to the present study. The criteria for inclusion were as follows: i) Age, <16 years; ii) treated with cyclosporin; and iii) full set of therapeutic drug monitoring (TDM) data for cyclosporin. Patients with other transplant statuses, including liver or kidney transplants were excluded. Ciclosporin concentrations and clinical data were gathered via TDM records and from medical records, respectively. The present study was approved by the Research Ethics Committee of the Children's Hospital of Fudan University [approval no. (2019)021]. A total of 18 patients were included in the current study (male/female ratio, 13/5; mean age, 1.60±1.15 years; age range, 0.29-6.49 years).

Drug administration and concentration detection

The initial ciclosporin dosage range was 14-100 mg/day. The dose was later adjusted based on clinical efficacy, adverse events and the trough concentration based on TDM. TDM was measured twice per week or more frequently if required, especially in suspected cases of intolerance or adverse events, using the Emit® 2000 Cyclosporin Specific assay (cat. no. 6R079UL; Siemens Healthcare Diagnostics, Inc.) according to the manufacturer's protocol. Blood samples (≥100 µl) were collected from the elbow vein immediately before the next drug administration.

Population pharmacokinetic modeling

A Nonlinear Mixed-Effects Modeling tool (NONMEM®; version VII; ICON Development Solutions Ltd.) was used to analyze the clinical data of pediatric patients who underwent BMT. The absorption phase was described by a one-compartment model with first-order elimination, where pharmacokinetic parameters included apparent oral clearance/bioavailability (CL/F) and apparent volume of distribution (V/F). Based on published literature, the absorption rate constant (Ka) of the model was 0.68 h-1 (16,21,25).

Random effect model

Equation A was used to calculate inter-individual variability: Pi=T(P) x exp (ηi), where Pi represented the individual parameter value, T(P) was the typical individual parameter value and ηi was the symmetrical distribution, which was a zero-mean chance variable with variance term. Equation B was used to calculate random residual variability: Y=F x (1+ε1) + ε2, where Y was the observation, F was the individual predicted concentration, and ε1 and ε2 were symmetrically distributed, zero-mean random variables with variance terms.

Covariate model

The relationship between weight and pharmacokinetic parameters was calculated using Equation C: Pi=Pnorm x (WTi/WTnorm)POW, where Pi represented the ith individual pharmacokinetic parameter, WTi represented the ith individual weight, WTnorm represented the standard weight of 70 kg, Pnorm represented the typical individual parameter whose weight was WTnorm and POW represented the allometric coefficient (0.75 for CL/F; 1 for V/F) (26).

The relationship between continuous covariates or categorical covariates and pharmacokinetic parameters was calculated by Equations D and E, respectively. Equation D: Pi=T(P) x (Covi/Covmedian)θ. Equation E: Pi=T(P) x (1+θ x Covi). Pi represented the individual parameter value, T(P) was the typical individual parameter value, θ was the parameter to be estimated, Covi was the covariate of the ith individual and Covmedian was the population median for the covariate.

To explain the variability of pharmacokinetic parameters, the correlations between covariates were investigated and the pharmacokinetic parameters were estimated. The potential covariates, which were obtained from the medical records, included sex, age, weight, days post-transplant (POD), albumin, alanine transaminase, aspartate transaminase, creatinine, urea, total protein, total bile acid, direct bilirubin, total bilirubin, hematocrit, hemoglobin, mean corpuscular hemoglobin, mean corpuscular hemoglobin concentration and co-medications (glucocorticoids, mycophenolate mofetil, omeprazole, phenobarbital and tacrolimus).

Statistical analysis

Alterations to the objective function values (OFV) were generated using covariate inclusions and a decrease in OFV of >3.84 [χ2; α=0.05; degrees of freedom (df)=1] was considered sufficient for inclusion of the base model. After establishing a full regression model, the model was further assessed by eliminating covariates from each pharmacokinetic parameter to obtain the final model. An increase in OFV of >6.64 (χ2; α=0.01; df=1) was considered to indicate a statistically significant difference.

Model validation

The reliability and stability of the final parameters were assessed by bootstrap, an internal validation method, which was performed using Wings for NONMEM (version VII; ICON plc) and repeated 1,000 times using different random draws. The medians and 2.5-97.5% percentiles of the bootstrap results were compared with the final pharmacokinetic parameter estimates and the absolute threshold of bias was set at <15% which was calculated using the following formula: Bias = (median-estimate)/estimate x100%. The final model was evaluated using goodness of fit plots and prediction-corrected visual predictive check plots, which were used to analyze model precision and predictability, respectively.

Simulation of dosing regimens

The dosage regimen simulations were performed using the parameter estimates obtained from the final model. The probability to achieve the target concentration was investigated using Monte Carlo simulations (NONMEM®; version VII; ICON plc), based on the established model. According to previous studies, the target concentrations were determined as 50-350 ng/ml (27-29). A total of 1,000 virtual patients were simulated in each of the four weight groups (5, 10, 20 and 30 kg) and for seven initial dosage regimens (2, 3, 4, 5, 6, 7 and 8 mg/kg/day ciclosporin split into 2 doses). Due to the large differences between individuals, the simulation results were presented with 70% confidence interval.

Results

Data collection

Clinical data of 18 pediatric patients who underwent BMT (13 male and 5 female) were collected for the present study. Patient characteristics and co-medications are presented in Table I.

Table I

Demographic data of patients and co-medications (n=18).

Table I

Demographic data of patients and co-medications (n=18).

CharacteristicMean ± SDMedian (range)
Sex, male/female13/5n/a
Age, years1.60±1.151.22 (0.29-6.49)
Weight, kg8.40±3.287.60 (5.20-25.60)
POD61.16±40.1651.50 (1.00-188.00)
Albumin, g/l34.07±5.5134.40 (1.20-45.20)
Alanine transaminase, IU/l51.26±65.9730.00 (1.00-439.00)
Aspartate transaminase, IU/l56.00±57.5836.30 (5.80-392.00)
Creatinine, µmol/l17.59±5.2316.00 (8.00-38.00)
Urea, mmol/l3.62±1.863.45 (0.60-11.10)
Total protein, g/l57.74±8.4958.80 (38.40-77.80)
Total bile acid, µmol/l21.62±32.0910.85 (0.40-201.20)
Direct bilirubin, µmol/l20.73±45.604.60 (0.10-301.90)
Total bilirubin, µmol/l33.30±64.4611.60 (1.30-384.20)
Hematocrit, %30.64±6.7630.40 (10.50-44.30)
Hemoglobin, g/l101.16±21.86101.00 (35.00-149.00)
Mean corpuscular hemoglobin, pg29.78±3.3029.70 (20.40-42.00)
Mean corpuscular hemoglobin concentration, g/l331.48±19.62333.00 (273.00-396.00)
Comedication, n  
     Glucocorticoids15n/a
     Mycophenolate mofetil7n/a
     Omeprazole16n/a
     Phenobarbital2n/a
     Tacrolimus2n/a

[i] SD, standard deviation; POD, days post-transplant.

Modeling

The final covariate models were displayed by equations F and G: i) Equation F, CL/F=θCL/F x (WT/70)0.75 x (POD/51.5)θPOD; and ii) Equation G: V/F=θV/F x (WT/70). θCL/F and θV/F represented the typical population values of CL/F and V/F, respectively, whilst θPOD represented the coefficient of the POD. From Table II, the value of θCL/F was found to be 29.200, θV/F was 6550.000 and θPOD was 0.749. Using these two models, WT and POD were included as the covariates of CL/F, whilst WT was included as the covariate of V/F. If the potential influencing factors could be included in the model as covariates, it indicated that there was an influence on ciclosporin. Conversely, if not, it indicated that there was no influence on ciclosporin. In the final models co-medications (glucocorticoids, mycophenolate mofetil, omeprazole, phenobarbital and tacrolimus) were could not be included into the final covariate models, suggesting that there was no significant drug interaction with ciclosporin.

Table II

Parameter estimates of the final model and bootstrap validation.

Table II

Parameter estimates of the final model and bootstrap validation.

ParameterEstimateSE (%)Bootstrap median95% confidence intervalBias (%)
CL/F (l/h)29.20015.328.700(20.000, 38.300)-1.71
V/F (l)6,550.00029.96,620.000(3,500.000, 11,800.000)1.07
Ka (h-1)0.680 (fixed)----
θPOD0.74925.40.730(0.330, 1.120)-2.54
ωCL/F0.62716.50.611(0.338, 0.826)-2.55
ωV/F0.99820.10.955(0.522, 1.367)-4.31
σ10.44715.60.442(0.296, 0.597)-1.12
σ270.07130.267.749(14.765, 104.403)-3.31

[i] 95% confidential interval is presented as (2.5th, 97.5th percentile) of bootstrap estimates. Bias=(median-estimate)/estimate x100%. SE, standard error; CL/F, apparent oral clearance; V/F, apparent volume of distribution; Ka, absorption rate constant; θPOD was the coefficient of days post-transplant; ωCL/F, inter-individual variability of CL/F; ωV/F, inter-individual variability of V/F; σ1, residual variability, proportional error; σ2, residual variability, additive error; bias, prediction error; -, not applicable.

Validation

Goodness of fit plots, representing the observed and predicted drug concentrations in the blood, are presented in Fig. 1, including observations vs. population predictions, observations vs. individual predictions, conditional weighted residuals (WRES) vs. population predictions and WRES vs. time after the start of therapy. In Fig. 1A and B, the black dashed lines represent the line of unity, where the predictions matched the observed values and the smooth yellow line represents the trend of the data. Hence, the closer the yellow smooth line is to the black dashed line, the more accurate the predictive the model. In Fig. 1C and D, the yellow smooth line represents the trend of the data such that the closer the yellow smooth line is to the line of unity, the more accurate the predictive model. Therefore, the final model exhibit higher precision and predictability. The parameter estimates of the final model and bootstrap validation are presented in Table II, where the median values of the parameter estimates of bootstraps were close to the respective values of the final population model. The absolute value of bias was found to be <5, <15% of the standard, which indicated that the final population model was accurate and reliable. The prediction-corrected visual predictive check plots of the final model are presented in Fig. 2. The majority of the observations were within the 95% prediction intervals of the simulation data, which suggested that the prediction-corrected concentrations were well predicted by the final model.

Simulation

Weight and POD influenced the clearance of ciclosporin in pediatric patients who underwent BMT (Fig. 3). Specifically, ciclosporin clearance was found to be increased as POD increased, whilst ciclosporin clearance was reduced with increasing weight. Additionally, lighter weight resulted in higher clearance rates in pediatric patients with the same POD. The initial dosage of 2-5 and 8 mg/kg/day split into 2 doses for pediatric patients weighing 5-30 kg exhibited lower probabilities of achieving the target concentrations, whilst the initial dosage of 6 or 7 mg/kg/day split into 2 doses for pediatric patients weighing 5-30 kg displayed higher probabilities to achieve the target concentrations (Fig. 4). However, the 7 mg/kg/day dose split into 2 doses exceeded the upper limit of the treatment window (350 ng/ml) in all weight groups (Table III). Therefore, an initial dose of 6 mg/kg/day ciclosporin split into 2 doses for pediatric patients weighing 5-30 kg who underwent BMT was identified as the optimal dose.

Table III

Predicted median (15th percentile-85th percentile) concentrations (ng/ml) of ciclosporine in each group.

Table III

Predicted median (15th percentile-85th percentile) concentrations (ng/ml) of ciclosporine in each group.

Dosea5 kg10 kg20 kg30 kg
2 mg/kg/day41.5 (15.05-104.81)41.41 (15.07-105.82)41.49 (15.09-107.50)41.54 (15.11-108.09)
3 mg/kg/day61.72 (22.57-157.22)62.11 (22.61-158.74)62.23 (22.63-161.25)62.31 (22.67-162.14)
4 mg/kg/day82.30 (30.10-209.65)82.82 (30.14-211.65)82.98 (30.18-215.00)83.08 (30.22-216.20)
5 mg/kg/day102.87 (37.63-262.02)103.52 (37.68-264.56)103.72 (37.72-268.75)103.85 (37.77-270.24)
6 mg/kg/day123.45 (45.15-314.43)124.22 (45.21-317.47)124.46 (45.26-322.50)124.62 (45.32-324.29)
7 mg/kg/day144.02 (52.68-366.84)144.93 (52.75-370.39)145.20 (52.81-376.25)145.39 (52.88-378.34)
8 mg/kg/day164.60 (60.20-419.24)165.63 (60.28-423.29)165.94 (60.35-430.00)166.16 (60.43-432.38)

[i] aSplit into 2 doses.

Discussion

The immunosuppressive drug ciclosporin was initially approved for use to prevent rejection in organ transplants, such as in liver (30,31), kidney (32,33), lung (34) and heart (35,36). Additionally, ciclosporin has been approved by the US Federal Drug Association for severe psoriasis and rheumatoid arthritis treatment (37). It has also been reported that ciclosporin can be used to treat alopecia (38), chronic autoimmune urticaria (39), pyoderma gangrenosum (40), severe atopic dermatitis (41), systemic lupus erythematosus (42), aplastic anemia (43), Crohn's disease (44) and ulcerative colitis (45). Furthermore, ciclosporin has also been used for the treatment of GVHD in pediatric patients who underwent BMT (1,2,13,14).

While ciclosporin has a wide range of clinical applications, the potential risks in numerous conditions have not been fully elucidated (46). Clinically, ciclosporin displays pharmacokinetic challenges which vary considerably between patients receiving the same dose (46). Additionally, ciclosporin has a narrow therapeutic range (47,48). Low doses are closely associated with the risk of graft rejection or loss, and overexposure is associated with acute or chronic toxicity, and irreversible renal damage (48). Therefore, a key challenge for the clinical use of ciclosporin is maintaining constant drug exposure in the narrow therapeutic window for each patient (15,49). While clinical TDM is often used to determine the optimal ciclosporin concentration and to provide reference for subsequent dose adjustments, a concentration reference for the initial dose has not been identified.

Population pharmacokinetics has the potential to aid individualized therapy by integrating different effects of variables on drug exposure (50); therefore, it can be used to determine the initial dose in different diseases. Population pharmacokinetics has been used for dosage optimization of tacrolimus in patients with nephritic syndrome (51,52), oxcarbazepine in pediatric Chinese patients with epilepsy (53), azithromycin in children with community-acquired pneumonia (54), vancomycin in neonates and young infants (55) and cyclosporin in pediatric patients with hemophagocytic lymphohistiocytosis (16). Therefore, the present study aimed to optimize the initial dosage of ciclosporin in pediatric patients who underwent BMT based on population pharmacokinetics and Monte Carlo simulations.

In the present study, the typical value of CL/F from the final population pharmacokinetic model was 29.200 l/h, which was similar to the previously reported value for pediatric patients receiving stem cell or kidney transplants (23.1-29.3 l/h) (23,27). Weight and POD influenced the clearance of ciclosporin in pediatric patients who underwent BMT. Ciclosporin clearance associated positively and negatively with POD and weight, respectively. A similar previous study demonstrated a non-linear relationship between drug clearance and body weight in pediatric patients (26). The association between body weight and clearance may scale with 0.75 power and a coefficient of 1 for volume (26,56,57). Therefore, in the present study, the following allometric coefficient was selected: 0.75 for CL/F and 1 for V/F. For ease of comparison between studies, body weight is typically standardized to 70 kg and the standardization of weight is particularly important in studies investigating children and neonates (21,56,58,59). Furthermore, in the present study, POD was found to associate positively with ciclosporin clearance, which may be explained by the association between patient recovery and an increased ability to metabolize exogenous drugs (60). Additionally, lighter weight was associated with higher clearance rates in pediatric patients with the same POD.

Subsequently, whether there was a significant difference in initial dose between children with different body weights was investigated. Monte Carlo simulations were used to simulate the optimal initial dose, including four weight groups (5, 10, 20 and 30 kg) and seven initial dosing regimens (2, 3, 4, 5, 6, 7 and 8 mg/kg/day split into 2 doses). According to previous studies, the target concentration range was determined as 50-350 ng/ml (27-29). The results of the present study suggested that the doses of 6 and 7 mg/kg/day split into 2 doses displayed a similar probability to achieve the target concentrations. However, the dose of 7 mg/kg/day resulted in an increased number of cases where the dose exceeded the upper limit of the therapeutic window. Therefore, an initial dose of 6 mg/kg/day ciclosporin split into 2 doses for pediatric patients weighing 5-30 kg who underwent BMT was identified as the optimal dose. Additionally, the present study also considered combination drugs, including glucocorticoid, mycophenolate mofetil, omeprazole, phenobarbital and tacrolimus; however, no significant interaction with ciclosporin was identified.

The present study had an important limitation. Ciclosporin is primarily eliminated via biotransformation by cytochrome P450. Therefore, whether the inclusion of genotyping in the model generated in the present study explains the variability of ciclosporin in pediatric Chinese patients who underwent BMT requires further investigation.

In conclusion, the present study indicated that weight and POD influenced the clearance of ciclosporin in pediatric patients who underwent BMT. Furthermore, the results indicated that the optimal initial dose of ciclosporin was 6 mg/kg/day split into 2 doses for pediatric patients weighing 5-30 kg who underwent BMT.

Acknowledgements

Not applicable.

Funding

The present study was supported by Important Weak Subject Construction Project of Shanghai (grant no. 2016ZB0305), Clinical Pharmacy Key Specialty Construction Project of Shanghai (grant no. 2017/26), Scientific Research Project of Science and Technology Commission of Shanghai Municipality (grant no. 18DZ1910604/19DZ1910703) and Shanghai Science and Technology Commission (grant no. 19XD1400900).

Availability of data and materials

The dataset used and/or analyzed during the present study are available from the corresponding authors on reasonable request.

Authors' contributions

ZL and HX conceived and designed the study. XC, XY and DW collected and analyzed the data. XC drafted the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Research Ethics Committee of Children's Hospital of Fudan University. Patient consent was waived by the Ethics Committee.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

McCann S, Passweg J, Bacigalupo A, Locasciulli A, Locatelli F, Ryan J, Schrezenmeier H and Lawler M: The influence of cyclosporin alone, or cyclosporin and methotrexate, on the incidence of mixed haematopoietic chimaerism following allogeneic sibling bone marrow transplantation for severe aplastic anaemia. Bone Marrow Transplant. 39:109–114. 2007.PubMed/NCBI View Article : Google Scholar

2 

Teuffel O, Schrauder A, Sykora KW, Zimmermann M, Reiter A, Welte K and Schrappe M: The impact of cyclosporin A on acute graft-versus-host disease after allogeneic bone marrow transplantation in children and adolescents with acute lymphoblastic leukemia. Bone Marrow Transplant. 36:145–150. 2005.PubMed/NCBI View Article : Google Scholar

3 

Bacigalupo A, Vitale V, Corvò R, Barra S, Lamparelli T, Gualandi F, Mordini N, Berisso G, Bregante S, Raiola AM, et al: The combined effect of total body irradiation (TBI) and cyclosporin A (CyA) on the risk of relapse in patients with acute myeloid leukaemia undergoing allogeneic bone marrow transplantation. Br J Haematol. 108:99–104. 2000.PubMed/NCBI View Article : Google Scholar

4 

Brandenburg U, Gottlieb D and Bradstock K: Antileukemic effects of rapid cyclosporin withdrawal in patients with relapsed chronic myeloid leukemia after allogeneic bone marrow transplantation. Leuk Lymphoma. 31:545–550. 1998.PubMed/NCBI View Article : Google Scholar

5 

Goldman JM, Apperley JF, Jones L, Marcus R, Goolden AW, Batchelor R, Hale G, Waldmann H, Reid CD, Hows J, et al: Bone marrow transplantation for patients with chronic myeloid leukemia. N Engl J Med. 314:202–207. 1986.PubMed/NCBI View Article : Google Scholar

6 

Kohno A, Morishita Y, Iida H, Sakamaki H, Yokozawa T, Kitaori K, Ozeki K, Matsuo K and Sao H: Low-dose cyclosporin A with short-term methotrexate for graft-versus-host disease prophylaxis in allogeneic bone marrow transplantation from human leukocyte antigen-identical siblings: A prospective phase II study in Japanese patients. Int J Hematol. 84:83–89. 2006.PubMed/NCBI View Article : Google Scholar

7 

Rowlings PA, Przepiorka D, Klein JP, Gale RP, Passweg JR, Henslee-Downey PJ, Cahn JY, Calderwood S, Gratwohl A, Socie G, et al: IBMTR Severity Index for grading acute graft-versus-host disease: Retrospective comparison with Glucksberg grade. Br J Haematol. 97:855–864. 1997.PubMed/NCBI View Article : Google Scholar

8 

Martin PJ, Schoch G, Fisher L, Byers V, Anasetti C, Appelbaum FR, Beatty PG, Doney K, McDonald GB, Sanders JE, et al: A retrospective analysis of therapy for acute graft-versus-host disease: Initial treatment. Blood. 76:1464–1472. 1990.PubMed/NCBI

9 

Martin PJ, Schoch G, Fisher L, Byers V, Appelbaum FR, McDonald GB, Storb R and Hansen JA: A retrospective analysis of therapy for acute graft-versus-host disease: Secondary treatment. Blood. 77:1821–1828. 1991.PubMed/NCBI

10 

Ferrara JL, Levine JE, Reddy P and Holler E: Graft-versus-host disease. Lancet. 373:1550–1561. 2009.PubMed/NCBI View Article : Google Scholar

11 

Hill GR and Ferrara JL: The primacy of the gastrointestinal tract as a target organ of acute graft-versus-host disease: Rationale for the use of cytokine shields in allogeneic bone marrow transplantation. Blood. 95:2754–2759. 2000.PubMed/NCBI

12 

Fowler DH, Foley J, Whit-Shan Hou J, Odom J, Castro K, Steinberg SM, Gea-Banacloche J, Kasten-Sportes C, Gress RE and Bishop MR: Clinical ‘cytokine storm’ as revealed by monocyte intracellular flow cytometry: Correlation of tumor necrosis factor alpha with severe gut graft-versus-host disease. Clin Gastroenterol Hepatol. 2:237–245. 2004.PubMed/NCBI View Article : Google Scholar

13 

Gérard C, Bleyzac N, Girard P, Freyer G, Bertrand Y and Tod M: Links between cyclosporin exposure in tissues and graft-versus-host disease in pediatric bone marrow transplantation: Analysis by a PBPK model. Pharm Res. 28:531–539. 2011.PubMed/NCBI View Article : Google Scholar

14 

Kobayashi R, Yabe H, Hara J, Morimoto A, Tsuchida M, Mugishima H, Ohara A, Tsukimoto I, Kato K, Kigasawa H, et al: Preceding immunosuppressive therapy with antithymocyte globulin and ciclosporin increases the incidence of graft rejection in children with aplastic anaemia who underwent allogeneic bone marrow transplantation from HLA-identical siblings. Br J Haematol. 135:693–696. 2006.PubMed/NCBI View Article : Google Scholar

15 

Bowers LD: Therapeutic monitoring for cyclosporine: Difficulties in establishing a therapeutic window. Clin Biochem. 24:81–87. 1991.PubMed/NCBI View Article : Google Scholar

16 

Wang DD, Ye QF, Chen X, Xu H and Li ZP: Population pharmacokinetics and initial dosing regimen optimization of cyclosporin in pediatric hemophagocytic lymphohistiocytosis patients. Xenobiotica. 50:435–441. 2020.PubMed/NCBI View Article : Google Scholar

17 

Li TF, Hu L, Ma XL, Huang L, Liu XM, Luo XX, Feng WY and Wu CF: Population pharmacokinetics of cyclosporine in Chinese children receiving hematopoietic stem cell transplantation. Acta Pharmacol Sin. 40:1603–1610. 2019.PubMed/NCBI View Article : Google Scholar

18 

Liu YO, Jia B, Chen CY, Zhou Y, Cui YM and Zhou FD: Population pharmacokinetics of cyclosporine A in Chinese patients with nephrotic syndrome in individualized drug administration. Int J Clin Pharmacol Ther. 58:1–9. 2020.PubMed/NCBI View Article : Google Scholar

19 

Martin P, Bleyzac N, Souillet G, Galambrun C, Bertrand Y, Maire PH, Jelliffe RW and Aulagner G: Relationship between CsA trough blood concentration and severity of acute graft-versus-host disease after paediatric stem cell transplantation from matched-sibling or unrelated donors. Bone Marrow Transplant. 32:777–784. 2003.PubMed/NCBI View Article : Google Scholar

20 

Carlens S, Aschan J, Remberger M, Dilber M and Ringden O: Low-dose cyclosporine of short duration increases the risk of mild and moderate GVHD and reduces the risk of relapse in HLA-identical sibling marrow transplant recipients with leukaemia. Bone Marrow Transplant. 24:629–635. 1999.PubMed/NCBI View Article : Google Scholar

21 

Ni SQ, Zhao W, Wang J, Zeng S, Chen SQ, Jacqz-Aigrain E and Zhao ZY: Population pharmacokinetics of ciclosporin in Chinese children with aplastic anemia: Effects of weight, renal function and stanozolol administration. Acta Pharmacol Sin. 34:969–975. 2013.PubMed/NCBI View Article : Google Scholar

22 

Fanta S, Jonsson S, Backman JT, Karlsson MO and Hoppu K: Developmental pharmacokinetics of ciclosporin-a population pharmacokinetic study in paediatric renal transplant candidates. Br J Clin Pharmacol. 64:772–784. 2007.PubMed/NCBI View Article : Google Scholar

23 

Irtan S, Saint-Marcoux F, Rousseau A, Zhang D, Leroy V, Marquet P and Jacqz-Aigrain E: Population pharmacokinetics and bayesian estimator of cyclosporine in pediatric renal transplant patients. Ther Drug Monit. 29:96–102. 2007.PubMed/NCBI View Article : Google Scholar

24 

Wilhelm AJ, de Graaf P, Veldkamp AI, Janssen JJ, Huijgens PC and Swart EL: Population pharmacokinetics of ciclosporin in haematopoietic allogeneic stem cell transplantation with emphasis on limited sampling strategy. Br J Clin Pharmacol. 73:553–563. 2012.PubMed/NCBI View Article : Google Scholar

25 

Wang D, Chen X and Li Z: Cyclosporin population pharmacokinetics in pediatric refractory nephrotic syndrome based on real-world studies: Effects of body weight and spirolactone administration. Exp Ther Med. 17:3015–3020. 2019.PubMed/NCBI View Article : Google Scholar

26 

Anderson BJ and Holford NH: Mechanism-based concepts of size and maturity in pharmacokinetics. Annu Rev Pharmacol Toxicol. 48:303–332. 2008.PubMed/NCBI View Article : Google Scholar

27 

Willemze AJ, Cremers SC, Schoemaker RC, Lankester AC, den Hartigh J, Burggraaf J and Vossen JM: Ciclosporin kinetics in children after stem cell transplantation. Br J Clin Pharmacol. 66:539–545. 2008.PubMed/NCBI View Article : Google Scholar

28 

Weiss M, Steinbach D, Zintl F, Beck J and Gruhn B: Superior outcome using cyclosporin A alone versus cyclosporin A plus methotrexate for post-transplant immunosuppression in children with acute leukemia undergoing sibling hematopoietic stem cell transplantation. J Cancer Res Clin Oncol. 141:1089–1094. 2015.PubMed/NCBI View Article : Google Scholar

29 

Choi JS, Lee SH, Chung SJ, Yoo KH, Sung KW and Koo HH: Assessment of converting from intravenous to oral administration of cyclosporin A in pediatric allogeneic hematopoietic stem cell transplant recipients. Bone Marrow Transplant. 38:29–35. 2006.PubMed/NCBI View Article : Google Scholar

30 

Muduma G, Saunders R, Odeyemi I and Pollock RF: Systematic review and meta-analysis of tacrolimus versus ciclosporin as primary immunosuppression after liver transplant. PLoS One. 11(e0160421)2016.PubMed/NCBI View Article : Google Scholar

31 

Muduma G, Odeyemi I and Pollock RF: A cost-utility analysis of prolonged-release tacrolimus relative to immediate-release tacrolimus and ciclosporin in liver transplant recipients in the UK. J Med Econ. 19:995–1002. 2016.PubMed/NCBI View Article : Google Scholar

32 

Riegersperger M, Plischke M, Jallitsch-Halper A, Steinhauser C, Födinger M, Winkelmayer WC, Dunkler D and Sunder-Plassmann G: A non-randomized trial of conversion from ciclosporin and tacrolimus to tacrolimus MR4 in stable long-term kidney transplant recipients: Graft function and influences of ABCB1 genotypes. PLoS One. 14(e0218709)2019.PubMed/NCBI View Article : Google Scholar

33 

Gathogo E, Harber M, Bhagani S, Levy J, Jones R, Hilton R, Davies G and Post FA: UK HIV Kidney Transplantation Study Group. Impact of tacrolimus compared with cyclosporin on the incidence of acute allograft rejection in human immunodeficiency virus-positive kidney transplant recipients. Transplantation. 100:871–878. 2016.PubMed/NCBI View Article : Google Scholar

34 

Penninga L, Penninga EI, Møller CH, Iversen M, Steinbrüchel DA and Gluud C: Tacrolimus versus cyclosporin as primary immunosuppression for lung transplant recipients. Cochrane Database Syst Rev: CD008817, 2013.

35 

Jia Y, Meng X, Li Y, Xu C, Zeng W, Jiao Y and Han W: Optimal sampling time-point for cyclosporin A concentration monitoring in heart transplant recipients. Exp Ther Med. 16:4265–4270. 2018.PubMed/NCBI View Article : Google Scholar

36 

Robertsen I, Falck P, Andreassen AK, Naess NK, Lunder N, Christensen H, Gullestad L and Asberg A: Endomyocardial, intralymphocyte, and whole blood concentrations of ciclosporin A in heart transplant recipients. Transplant Res. 2(5)2013.PubMed/NCBI View Article : Google Scholar

37 

Rosmarin DM, Lebwohl M, Elewski BE and Gottlieb AB: National Psoriasis Foundation. Cyclosporine and psoriasis: 2008 National Psoriasis Foundation Consensus Conference. J Am Acad Dermatol. 62:838–853. 2010.PubMed/NCBI View Article : Google Scholar

38 

Gupta AK, Ellis CN, Cooper KD, Nickoloff BJ, Ho VC, Chan LS, Hamilton TA, Tellner DC, Griffiths CE and Voorhees JJ: Oral cyclosporine for the treatment of alopecia areata. A clinical and immunohistochemical analysis. J Am Acad Dermatol. 22:242–250. 1990.PubMed/NCBI View Article : Google Scholar

39 

Boubouka CD, Charissi C, Kouimintzis D, Kalogeromitros D, Stavropoulos PG and Katsarou A: Treatment of autoimmune urticaria with low-dose cyclosporin A: A one-year follow-up. Acta Derm Venereol. 91:50–54. 2011.PubMed/NCBI View Article : Google Scholar

40 

Ahronowitz I, Harp J and Shinkai K: Etiology and management of pyoderma gangrenosum: A comprehensive review. Am J Clin Dermatol. 13:191–211. 2012.PubMed/NCBI View Article : Google Scholar

41 

Berth-Jones J, Finlay AY, Zaki I, Tan B, Goodyear H, Lewis-Jones S, Cork MJ, Bleehen SS, Salek MS, Allen BR, et al: Cyclosporine in severe childhood atopic dermatitis: A multicenter study. J Am Acad Dermatol. 34:1016–1021. 1996.PubMed/NCBI View Article : Google Scholar

42 

Zavada J, Pesickova S, Rysava R, Olejarova M, Horák P, Hrncír Z, Rychlík I, Havrda M, Vítova J, Lukác J, et al: Cyclosporine A or intravenous cyclophosphamide for lupus nephritis: The Cyclofa-Lune study. Lupus. 19:1281–1289. 2010.PubMed/NCBI View Article : Google Scholar

43 

Zheng Y, Liu Y and Chu Y: Immunosuppressive therapy for acquired severe aplastic anemia (SAA): A prospective comparison of four different regimens. Exp Hematol. 34:826–831. 2006.PubMed/NCBI View Article : Google Scholar

44 

McDonald JW, Feagan BG, Jewell D, Brynskov J, Stange EF and Macdonald JK: Cyclosporine for induction of remission in Crohn's disease. Cochrane Database Syst Rev: CD000297, 2005.

45 

Meier J and Sturm A: Current treatment of ulcerative colitis. World J Gastroenterol. 17:3204–3212. 2011.PubMed/NCBI View Article : Google Scholar

46 

Paziana K, Del Monaco M, Cardonick E, Moritz M, Keller M, Smith B, Coscia L and Armenti V: Ciclosporin use during pregnancy. Drug Saf. 36:279–294. 2013.PubMed/NCBI View Article : Google Scholar

47 

Barr WH: Cyclosporine: The case for expanding bioequivalence criteria to include measures of individual bioequivalence in relevant population subsets. Transplant Proc. 31:25S–30S. 1999.PubMed/NCBI View Article : Google Scholar

48 

Kahan BD, Welsh M and Rutzky LP: Challenges in cyclosporine therapy: The role of therapeutic monitoring by area under the curve monitoring. Ther Drug Monit. 17:621–624. 1995.PubMed/NCBI View Article : Google Scholar

49 

Colaizzi JL and Lowenthal DT: Critical therapeutic categories: A contraindication to generic substitution? Clin Ther. 8:370–379. 1986.PubMed/NCBI

50 

Zheng QS and Li LJ: Pharmacometrics: A quantitative tool of pharmacological research. Acta Pharmacol Sin. 33:1337–1338. 2012.PubMed/NCBI View Article : Google Scholar

51 

Lu T, Zhu X, Xu S, Zhao M, Huang X, Wang Z and Zhao L: Dosage optimization based on population pharmacokinetic analysis of tacrolimus in Chinese patients with nephrotic syndrome. Pharm Res. 36(45)2019.PubMed/NCBI View Article : Google Scholar

52 

Wang X, Han Y, Chen C, Ma L, Xiao H, Zhou Y, Cui Y, Wang F, Su B, Yao Y, et al: Population pharmacokinetics and dosage optimization of tacrolimus in pediatric patients with nephrotic syndrome. Int J Clin Pharmacol Ther. 57:125–134. 2019.PubMed/NCBI View Article : Google Scholar

53 

Chen CY, Zhou Y, Cui YM, Yang T, Zhao X and Wu Y: Population pharmacokinetics and dose simulation of oxcarbazepine in Chinese paediatric patients with epilepsy. J Clin Pharm Ther. 44:300–311. 2019.PubMed/NCBI View Article : Google Scholar

54 

Zheng Y, Liu SP, Xu BP, Shi ZR, Wang K, Yang JB, Huang X, Tang BH, Chen XK, Shi HY, et al: Population pharmacokinetics and dosing optimization of azithromycin in children with community-acquired pneumonia. Antimicrob Agents Chemother. 62:e00686–18. 2018.PubMed/NCBI View Article : Google Scholar

55 

Chen Y, Wu D, Dong M, Zhu Y, Lu J, Li X, Chen C and Li Z: Population pharmacokinetics of vancomycin and AUC-guided dosing in Chinese neonates and young infants. Eur J Clin Pharmacol. 74:921–930. 2018.PubMed/NCBI View Article : Google Scholar

56 

Anderson BJ and Holford NH: Tips and traps analyzing pediatric PK data. Paediatr Anaesth. 21:222–237. 2011.PubMed/NCBI View Article : Google Scholar

57 

Holford NH: A size standard for pharmacokinetics. Clin Pharmacokinet. 30:329–332. 1996.PubMed/NCBI View Article : Google Scholar

58 

Hao GX, Huang X, Zhang DF, Zheng Y, Shi HY, Li Y, Jacqz-Aigrain E and Zhao W: Population pharmacokinetics of tacrolimus in children with nephrotic syndrome. Br J Clin Pharmacol. 84:1748–1756. 2018.PubMed/NCBI View Article : Google Scholar

59 

Wang DD, Chen X and Li ZP: Wuzhi capsule and haemoglobin influence tacrolimus elimination in paediatric kidney transplantation patients in a population pharmacokinetics analysis: A retrospective study. J Clin Pharm Ther. 44:611–617. 2019.PubMed/NCBI View Article : Google Scholar

60 

Wang DD, Chen X, Fu M, Zheng QS, Xu H and Li ZP: Model extrapolation to a real-world dataset: Evaluation of tacrolimus population pharmacokinetics and drug interaction in pediatric liver transplantation patients. Xenobiotica. 50:371–379. 2020.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

July-2020
Volume 20 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen X, Yu X, Wang DD, Xu H and Li Z: Initial dosage optimization of ciclosporin in pediatric Chinese patients who underwent bone marrow transplants based on population pharmacokinetics. Exp Ther Med 20: 401-408, 2020
APA
Chen, X., Yu, X., Wang, D., Xu, H., & Li, Z. (2020). Initial dosage optimization of ciclosporin in pediatric Chinese patients who underwent bone marrow transplants based on population pharmacokinetics. Experimental and Therapeutic Medicine, 20, 401-408. https://doi.org/10.3892/etm.2020.8732
MLA
Chen, X., Yu, X., Wang, D., Xu, H., Li, Z."Initial dosage optimization of ciclosporin in pediatric Chinese patients who underwent bone marrow transplants based on population pharmacokinetics". Experimental and Therapeutic Medicine 20.1 (2020): 401-408.
Chicago
Chen, X., Yu, X., Wang, D., Xu, H., Li, Z."Initial dosage optimization of ciclosporin in pediatric Chinese patients who underwent bone marrow transplants based on population pharmacokinetics". Experimental and Therapeutic Medicine 20, no. 1 (2020): 401-408. https://doi.org/10.3892/etm.2020.8732