Skip to main content
British Journal of Clinical Pharmacology logoLink to British Journal of Clinical Pharmacology
. 2016 Dec 20;83(5):1097–1107. doi: 10.1111/bcp.13181

Population pharmacokinetics of temsirolimus and sirolimus in children with recurrent solid tumours: a report from the Children's Oncology Group

Tomoyuki Mizuno 1,, Tsuyoshi Fukuda 1,4, Uwe Christians 3, John P Perentesis 2,4, Maryam Fouladi 2,4, Alexander A Vinks 1,4,
PMCID: PMC5401981  PMID: 28000286

Abstract

Aims

Temsirolimus is an inhibitor of the mammalian target of rapamycin (mTOR). Pharmacokinetic (PK) characterization of temsirolimus in children is limited and there is no paediatric temsirolimus population PK model available. The objective of this study was to simultaneously characterize the PK of temsirolimus and its metabolite sirolimus in paediatric patients with recurrent solid or central nervous system tumours and to develop a population PK model.

Methods

The PK data for temsirolimus and sirolimus were collected as a part of a Children's Oncology Group phase I clinical trial in paediatric patients with recurrent solid tumours. Serial blood concentrations obtained from 19 patients participating in the PK portion of the study were used for the analysis. Population PK analysis was performed by nonlinear mixed effect modelling using NONMEM.

Results

A three‐compartment model with zero‐order infusion was found to best describe temsirolimus PK. Allometrically scaled body weight was included in the model to account for body size differences. Temsirolimus dose was identified as a significant covariate on clearance. A sirolimus metabolite formation model was developed and integrated with the temsirolimus model. A two‐compartment structure model adequately described the sirolimus data.

Conclusion

This study is the first to describe a population PK model of temsirolimus combined with sirolimus formation and disposition in paediatric patients. The developed model will facilitate PK model‐based dose individualization of temsirolimus and the design of future clinical studies in children.

Keywords: mTOR inhibitor, paediatrics, population pharmacokinetics, sirolimus, temsirolimus

What is Already Known About This Subject

  • Temsirolimus is a prodrug of sirolimus and both are pharmacologically active inhibitors of mammalian target of rapamycin.

  • Clinical studies have demonstrated that temsirolimus is promising for the treatment of solid tumours in paediatric patients; however, large interindividual variability has been observed in the pharmacokinetics of temsirolimus and sirolimus.

  • Characterization of the transformation of temsirolimus to sirolimus in children is limited.

What This Study Adds

  • A combined population pharmacokinetic model of temsirolimus with its metabolite sirolimus in children was developed with pharmacokinetic data collected in a phase I study in paediatric patients with recurrent solid tumours.

  • The developed model will facilitate prediction of both temsirolimus and sirolimus concentrations and exposure in children and can be used to identify age‐appropriate dosing regimens as part of clinical trial design simulations and for Bayesian guided precision dosing.

Tables of Links

These Tables list key protein targets and ligands in this article that are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY 1, and are permanently archived in the Concise Guide to PHARMACOLOGY 2015/16 2.

Introduction

The mammalian target of rapamycin (mTOR) is a serine/threonine kinase that is involved in many critical cellular functions including cell growth, proliferation, angiogenesis and cellular survival. Temsirolimus and its primary metabolite sirolimus form complexes with the FK506‐binding protein, an intracellular immunophilin, and these complexes exert antiangiogenesis and antitumour activities by inhibiting mTOR. To date, clinical trials have revealed that intravenous administered temsirolimus prolongs overall survival in patients with advanced renal cell carcinoma (RCC) compared with interferon‐α therapy 3. Temsirolimus was approved for the treatment of advanced RCC in 2007 in the USA and Europe. Since then, many clinical trials have been conducted to explore the effectiveness of temsirolimus for the treatment of other tumours in adult patients 4, 5, 6, 7, 8 and also in children 9, 10. In addition, recently, many combination therapies with other anticancer drugs and/or antibodies have been investigated 11, 12, 13, 14, 15, 16, 17, 18.

Several studies have documented large interpatient variability in temsirolimus pharmacokinetics (PK) 19, 20, 21. In these clinical trials, temsirolimus PK and exposure rather than dose have been associated with adverse drug reactions (ADRs) 21, 22, 23, suggesting that temsirolimus PK is one of the determinants and markers of temsirolimus‐induced toxicity. Indeed, clinically important associations of temsirolimus and sirolimus area under the curve (AUC) with ADR severity were reported for thrombocytopenia, pruritus and hyperlipaemia 22. A correlation between temsirolimus concentration at end of infusion and severity of ADRs has also been observed 22.

In humans, temsirolimus is rapidly hydrolyzed by carboxyesterases to its major metabolite sirolimus 24. Sirolimus exhibits comparable mTOR inhibitory activity and is predominantly metabolized by CYP3A4 and CYP3A5 to multiple metabolites including hydroxyl‐ and demethyl‐forms 25, 26, 27. To date, two temsirolimus population PK analyses have been reported in adult patients, one in patients with advanced RCC 22 and one in patients with breast cancer 5. PK characterization of temsirolimus in children is limited and there is no paediatric population PK model of temsirolimus available 10, 28, 29.

In this study, we aimed to characterize the PK of temsirolimus in children and develop a population PK model of both temsirolimus and sirolimus. The population PK model could serve as a basis for clinical trial design simulations and for PK‐guided dosing using Bayesian adaptive control.

Methods

Patients and sampling

The population PK analysis was performed using PK data collected in a subset of patients who participated in a Children's Oncology Group phase I dose finding and safety trial of the anti‐ insulin‐like growth factor I receptor antibody cixutumumab (IMC‐A12), combined with temsirolimus in children with recurrent solid tumours (ADVL0813, ClinicalTrials.gov Identifier: NCT00880282) 28. Demographic characteristics of participants are described in Table 1. In brief, 6 mg kg−1 of IMC‐A12 in combination with 8, 10 or 15 mg m−2 of temsirolimus was administered once weekly in 28 day cycles. As described previously 28, blood samples were collected at predose, at 15 and 30 min, and 1, 3, 6 and 24 h after intravenous infusion of temsirolimus on day 1 of cycle 1, and were used for the PK analysis. In addition to those data, the later blood samples collected at 48, 72 and 168 h postdose were also used in this population PK analysis. The infusion duration was documented and ranged from 15 to 63 min. Whole blood concentrations of temsirolimus and sirolimus were determined by validated liquid chromatography tandem‐mass spectrometry assay 30. The range of reliable quantification, the interday imprecision and interday accuracy for the temsirolimus assay are 0.5–100 ng ml−1, 3.8–7.0% and 99.9–108.2%, respectively. For the sirolimus assay, ranges are 0.1–100 ng ml−1, 6.0–12.4% and 98.5–106%, respectively. The Institutional Review Boards of Cincinnati Children's Hospital Medical Center and other participating institutions approved the protocol 28. Informed consent and assent, as appropriate, were obtained according to local institutional guidelines 28.

Table 1.

Demographic characteristics of patients at enrolment in this study

Characteristic Total (n = 19)
Age, years
Mean ± SD 11.9 ± 5.7
Median 10.9
Range 1–19
Body weight, kg
Mean ± SD 45.7 ± 28
Median 35.7
Range 7.3–114.7
Sex, n (%)
Female 8 (42.1)
Male 11 (57.9)
Race, n (%)
Caucasian 17 (89.5)
African–American 1 (5.3)
Asian 1 (5.3)
Temsirolimus dose level, n (%)
8 mg m −2 11 (57.9)
10 mg m −2 3 (15.8)
15 mg m −2 5 (26.3)

SD, standard deviation

Population PK modelling

Population PK analysis was performed by nonlinear mixed effect modelling using NONMEM (version 7.2, ICON, Ellicott City, MD, USA) with Perl speaks NONMEM (PsN) version 3.6.2 31 and Pirana version 2.7.1 (Pirana Software & Consulting BV, http://pirana.sourceforge.net) as the interface. The first‐order conditional estimation with interaction method (FOCE‐I) was applied for all runs. Different compartment models were explored to describe the temsirolimus and sirolimus blood concentration‐time profiles. Model selection was based on goodness‐of‐fit diagnostic plots, comparisons based on the minimum objective function value (OFV) and evaluation of the estimates of population fixed and random effect parameters. Interpatient variability was assessed using an exponential variability model (Equation (1)):

Pi=Ppop×expηi (1)

where Pi is the estimated parameter value for individual i, Ppop is the typical population value (geometric mean) of the PK parameters such as clearance and volume of distribution, ηi is an interindividual random effect for individual i with the mean of zero and variance of ω2. A proportional error model and a combined proportional and additive error model were examined to describe the residual error. All PK models were parameterized in terms of values of clearance (CL), volume of distribution (V) and intercompartmental clearances (Q).

Allometrically scaled body weight was used to account for differences in body size as follows (Equation (2)):

Pi=Ppop×BWiBWstandardpower (2)

where BWi is body weight for individual i, BWstandard is 70 kg, and power is the coefficient set at 0.75 for CL and 1 for V 32. The temsirolimus dose was examined as a covariate to account for the nonlinearity between dose and AUC, which was observed in the prior noncompartmental analysis 28 (Equation (3)).

Pi=Ppop×DoseiDosemedianθdose (3)

where Dosei is the temsirolimus dose normalized by body weight. Age and sex of the patient were examined in the covariate analysis with following equations (Equations (4) and (5)):

Pi=Ppop×AgeiAgemedianθage (4)
Pi=Ppop×θgenderpower (5)

where power is 0 or 1 when the patient is male or female, respectively.

Model evaluation

The following diagnostic plots were used to evaluate the models: observed value (DV) vs. population predicted value (PRED), DV vs. individual predicted value (IPRED), conditional weighted residuals vs. PRED and conditional weighted residuals vs. time after dose to identify a bias corresponding to model mis‐specification. The final model was evaluated using nonparametric bootstrap analysis 33. The original dataset was resampled with 500 replicate data sets, and the estimated medians and 95% confidence intervals (CIs) of parameter estimates from the bootstrap analysis were compared to the final model estimates. The prediction‐corrected visual predictive check (pcVPC) was used for the validation 34. One thousand replicates of simulated datasets were generated using the final model and the distribution of simulated observations was compared with the actual observations.

Results

Population PK modelling for temsirolimus

A total of 135 temsirolimus blood concentration results from 19 participants were available for the population PK analysis. Five to nine blood samples were collected from each patient. Two samples were below the lower limit of quantification 30. A three‐compartment model with zero‐order infusion was found to adequately describe the temsirolimus PK profiles after intravenous infusion (Figure 1). A combined proportional and additive residual error model provided better model fit than a proportional error model. Including allometrically scaled body weight resulted in significant improvement in model fit (ΔOFV = 39.6, P < 0.001). This model was defined as the base model and the population PK parameter estimates are summarized in Table 2. The temsirolimus dose was tested as a covariate in order to account for the observed nonlinearity between the temsirolimus dose and AUC. Dose was retained in the final model for CL, Q2 and Q3 because of significant improvement in model fit (ΔOFV = 6.20, P < 0.05). No further significant improvement in model fit was observed by adding any of the other covariates. PK parameter estimates for the final model are also summarized in Table 2. The IIV for V3 could not be estimated reasonably and then was not included in the model. The η‐shrinkage values are summarized in Table 2. Goodness‐of‐fit plots indicated that the developed final model predicts the temsirolimus blood concentration profiles adequately (Figure 2). The bootstrap analysis indicated that the final model was stable (Table 2). The simulated blood concentrations by pcVPC were in reasonable agreement with the observed concentrations (Figure 3).

Figure 1.

Figure 1

Pharmacokinetic structural model for temsirolimus and sirolimus. IV, intravenous; CL, clearance; V, volume of distribution; Q, intercompartmental clearance; Fm, fraction of metabolism of temsirolimus to sirolimus

Table 2.

Parameter estimates for the temsirolimus population pharmacokinetic model

Parameters Base model Final model
Estimates RSE (%) Shrinkage (%) Estimates RSE (%) Shrinkage (%) Bootstrap analysis
Median 95% CI
Lower Upper
CL (l h −1 70 kg −1 ) 3.40 27 3.61 18 3.71 2.22 5.80
V1 (l 70 kg −1 ) 9.49 30 14.2 8.9 12.8 8.51 18.6
Q2 (l h −1 70 kg −1 ) 40.8 17 30.7 17 29.9 20.9 42.8
V2 (l 70 kg −1 ) 24.7 8 22.9 16 23.0 16.4 29.2
Q3 (l h −1 70 kg −1 ) 7.76 11 7.39 14 7.38 5.45 9.46
V3 (l 70 kg −1 ) 244 18 220 14 217 108 296
θ dose 0.855 19 0.878 0.428 1.65
Interindividual variability (CV%)
IIV for CL 85.6 24 6 70.9 21 6 63.5 36.2 104
IIV for V1 37.4 26 55 56.0 25 33 53.6 22.8 76.9
IIV for Q2 68.0 29 20 50.7 33 27 53.0 12.8 77.8
IIV for V2 64.1 23 7 69.8 24 9 59.1 32.4 99.5
IIV for Q3 54.5 18 12 55.2 17 15 55.1 38.2 80.1
IIV for V3
Residual variability
Proportional error (CV%) 16.1 17 25 16.6 16 25 16.2 11.0 20.3
Additive error (ng ml −1 ) 6.84 31 25 6.60 34 25 6.03 2.74 10.3

CI, confidential interval; CL, clearance; CV, coefficient of variation; IIV, interindividual variability; Q, intercompartmental clearance; RSE, relative standard error; V, volume of distribution.

Figure 2.

Figure 2

Goodness‐of‐fit plots for the final pharmacokinetic model of temsirolimus. Observed vs. (A) population‐predicted and (B) individual‐predicted temsirolimus concentrations (line of identity shown for clarity). The conditional weighted residuals (CWRES) vs. (C) time after dose and (D) population‐predicted temsirolimus concentration

Figure 3.

Figure 3

Prediction‐corrected visual predictive check (pcVPC) for the final model of temsirolimus. (A) All observations and (B) enlarged picture from 0 to 25 h. Open circle, observed blood concentrations; lines represent the median, 5th and 95th percentiles of the simulated data (n = 1000)

Population PK modelling of temsirolimus combined with its metabolite sirolimus

The population PK model for sirolimus was developed by integrating metabolite formation with the temsirolimus PK model. A total of 137 sirolimus concentration measurements were obtained in 19 patients and used for the analysis. To develop the combined model, mass balance and concentrations were adjusted based on the respective molecular weights. A two‐compartment structural sirolimus model linked with the three‐compartmental temsirolimus model best described the PK data (Figure 1). PK parameter estimates for the final combined model are summarized in Table 3. The exponent for dose was fixed to 0.855 in the analysis. A combined proportional and additive error model best described residual error. Inclusion of allometrically scaled body weight on clearance improved the model fit (ΔOFV = 20.1, P < 0.001). The η‐shrinkage values for CL and V1 are summarized in Table 3. IIV for Q2, Q4, V2, V3 and V5 could not be estimated reasonably and then were not included. Goodness‐of‐fit plots showed that the developed final model adequately predicted the concentration profiles of both temsirolimus and sirolimus (Figure 4). Bootstrap analysis confirmed stability of the final model (Table 3). The simulated concentrations by pcVPC were in reasonable agreement with the observed values (Figure 5).

Table 3.

Parameter estimates for the combined population pharmacokinetic model of temsirolimus with its metabolite sirolimus

Parameters Final model
Estimates RSE (%) Shrinkage (%) Bootstrap analysis
Median 95% CI
Lower Upper
CL TEM (l h −1 70 kg −1 ) 4.31 21 4.42 2.40 6.16
F m 0.459 29 0.476 0.389 0.707
V1 (l 70 kg −1 ) 18.9 23 18.1 10.7 26.3
Q2 (l h −1 70 kg −1 ) 10.4 23 10.4 7.00 22.8
V2 (l 70 kg −1 ) 12.9 23 12.9 7.46 19.4
Q3 (l h −1 70 kg −1 ) 9.15 29 9.01 5.87 12.3
V3 (l 70 kg −1 ) 140 24 129 92.5 254
θ dose 0.855 FIXED 0.855
CL SIR (l h −1 70 kg −1 ) 6.08 31 6.44 3.15 10.9
V4 (l 70 kg −1 ) 48 30 51.9 22.9 95.0
Q5 (l h −1 70 kg −1 ) 11.6 36 13.3 9.51 22.0
V5 (l 70 kg −1 ) 72.8 41 86.4 55.8 140
Interindividual variability (CV%)
IIV for CL TEM 49.5 17 8 51.7 31.7 79.4
IIV for V1 67 23 14 68.0 38.6 102
IIV for Q2
IIV for V2
IIV for Q3 79.1 29 5 73.5 43.4 120
IIV for V3
IIV for CL SIR 103 14 7 103 67.7 147
IIV for V4 121 34 3 118 41.9 169
IIV for Q5
IIV for V5
Residual variability
Temsirolimus
Proportional error (CV%) 23.9 20 14 21.7 14.7 29.3
Additive error (ng ml −1 ) 7.25 92 14 8.06 1.01 14.2
Sirolimus
Proportional error (CV%) 25.5 20 14 24.2 8.92 32.9
Additive error (ng ml −1 ) 1.69 47 14 1.52 0.833 3.89

CI, confidential interval; CL, clearance; CV, coefficient of variation; Fm, fraction of metabolism to sirolimus; IIV, interindividual variability; Q, intercompartmental clearance; RSE, relative standard error; V, volume of distribution.

Figure 4.

Figure 4

Goodness‐of‐fit plots for the final pharmacokinetic model of temsirolimus with sirolimus. The observed versus population predicted (A) and individual predicted (B) temsirolimus (open circle) and sirolimus (blue circle) concentrations (line of identity shown for clarity). The conditional weighted residuals (CWRES) vs. time after dose (C) and population predict temsirolimus (open circles) and sirolimus (blue circles) concentrations (D)

Figure 5.

Figure 5

Prediction‐corrected visual predictive check (pcVPC) for the final model of temsirolimus with sirolimus. (A, C) All observations and (B, D) enlarged picture from 0 to 25 h. Open circles, observed temsirolimus concentrations (A, B) and sirolimus concentrations (C, D); lines represent the median, 5th and 95th percentiles of the simulated data (n = 1000)

Individual estimates

The PK parameters for each individual patient were generated using posthoc Bayesian estimation with NONMEM. When CL was standardized to allometrically scaled body weight, no age effects were observed over the age range of patients in this study (Age range 1–19 years, with only one patient younger than 2 years; Figure S1).

Discussion

This study generated a combined population PK model of temsirolimus with its metabolite sirolimus in paediatric patients with recurrent solid tumours. To the best of our knowledge, this is the first population PK modelling analysis of temsirolimus in children. The analysis confirms that temsirolimus PK is nonlinear with dose consistent with that reported in adult patients 5, 22.

Nonlinearity in the relationship between temsirolimus dose and systemic exposure has been well documented 10, 19, 20, 21, 23, 35. In a previous population PK analysis in 50 adult patients, Boni et al. 22 reported a less‐than‐proportional AUC increase with increased dose that they were able to model by including an exponential function of dose 24. Similarly, we incorporated dose as a covariate for CL, Q2 and Q3 in this PK modelling, resulting in a significant improvement in model fit despite the relatively narrow dose range (8–15 mg m−2) and the small number of patients. Although the mechanism underlying this phenomenon has not been well characterized, the saturable specific binding to tissue and binding proteins in blood cells are thought to contribute the nonlinear PK behaviour 22 Indeed, a decreased blood‐to‐plasma ratio with increasing dose has been reported 36, indicating that distribution of temsirolimus to blood cells is saturable. It has been suggested that unbound temsirolimus distributes to some tissues that could serve as ‘sink’ compartments and this phenomenon could be an explanation of the nonlinear relationship between dose and drug exposure. A study including a mechanism‐based approach will be required to further elucidate the precise mechanism of this phenomenon.

We did not observe an age‐effect on temsirolimus CL after adjusting for body weight. This could be explained by the age distribution of the patients in the present study, with all subjects being older than 4 years except for one patient who was age 1 year. The observation of no age‐effect on temsirolimus CL is in line with the expectation that age‐dependent changes in clearance due to maturation of organ function and ontogeny of drug metabolic enzymes typically are observed between ages of 0 and 2–3 years 37. For sirolimus we showed that maturation of the CYP3A4/5 pathways in neonates and infants is responsible for lower drug clearance only in the younger patients 38, 39. We also compared our results with temsirolimus CL estimates reported in one paediatric and three adult studies in patients who were treated at a similar dose level. Temsirolimus CL estimates in the current study are somewhat lower than values reported in the adult studies 19, 20, 35 while clearance was comparable to the previous paediatric study results 10. To document developmental changes in temsirolimus PK and to update the population model with a descriptor of maturation, as described for sirolimus 38, further studies in neonates and infants will be needed.

Sirolimus is the primary metabolite of temsirolimus in humans; however, information on the fraction of parent compound that is converted into sirolimus and contribution by other metabolic pathways is still limited. In this analysis, the fraction of temsirolimus metabolized into sirolimus was estimated to be 45.9% (95% CI: 38.9–70.7%). It should be noted that the observed sirolimus to temsirolimus AUC ratios were highly variable indicating large variability in clearance and the fraction metabolized. To date, lower sirolimus exposures (AUC) have been observed in paediatric studies including this study as compared to what has been reported for adults. For instance, the AUC0‐∞ ratio of sirolimus to temsirolimus was 1.21 ± 1.18 (mean ± SD) in patients receiving a dose of 15 mg m−2. The mean AUC ratio in our study was approximately two‐fold lower that observed in adult patients (2.28 ± 0.99) receiving the same temsirolimus dose of 15 mg m−2 19. In addition to the present study, other paediatric clinical studies have reported lower sirolimus to temsirolimus AUC ratios compared to the ratios in adult patients 9, 10, 19. These observations suggest that the temsirolimus fraction converted to sirolimus in adults may be higher than in children.

Temsirolimus‐induced severe adverse events often lead to discontinuation of treatment. The most common adverse reactions are asthenia, skin/mucosal toxicity and laboratory abnormalities such as hypertriglyceridemia, hypophosphatemia and anaemia. To date, several clinical trials/studies demonstrated the possible relationship between the temsirolimus exposure and toxicity. For instance, a clinical study of temsirolimus mono‐therapy in adult patients with advanced RCC revealed significant associations between temsirolimus AUC and severity of adverse events such as thrombocytopenia, pruritus and hyperlipaemia 22. Furthermore, in a phase I study of temsirolimus monotherapy in paediatric patients, one patient who developed grade 4 thrombocytopenia, showed an unusually high temsirolimus Cmax value 10. These observations indicate that high temsirolimus (and sirolimus) concentrations are likely to increase the risk of adverse events, although no clear target exposure range has been defined. In terms of therapeutic outcomes, no definitive relationship between temsirolimus dose, PK (Cmax or AUC achieved) and PD such as complete tumour response, progression free survival or relative phosphorylation of the mTOR‐targeted proteins such as S70S6 and 4 EB1 has been documented 5, 10. Further well‐controlled studies are warranted to establish these relationships. As part of such studies, a PK‐guided dosing strategy would be attractive as it would allow temsirolimus (and sirolimus) concentration control using sparse sampling and a model based prediction of the drug exposure–response relationship. A similar approach has been successfully used for sirolimus PK guided dosing in infants and children with neurofibromatosis and vascular anomalies 40, 41, 42.

A limitation of this study is the relatively small number of patients (n = 19) available to describe the population PK of temsirolimus in children. In the covariate analysis inclusion of age and sex did not significantly improve the temsirolimus model. However, given the limited sample size, these covariates should still be evaluated in an adequately powered future study. Finally, this analysis was based on the data obtained in patients receiving a combination therapy of temsirolimus and cixutumumab. The CL observed in the previous Phase I study of combination therapy was comparable with that observed at the same dose in the study of temsirolimus monotherapy 10, 28. However, sample size may not have been large enough to conclude no effect of coadministration of cixutumumab on temsirolimus pharmacokinetics. Further study is warranted.

Conclusion

We successfully developed a population PK model of temsirolimus and its metabolite sirolimus in paediatric patients with recurrent solid tumours. The findings of this study and the developed model will facilitate model‐based dose individualization of temsirolimus and further optimize clinical trial design in children.

Competing Interests

There are no competing interests to declare.

This work was supported in part by the Children's Oncology Group Phase I and Pilot Consortium (study ADVL0813) and the research was supported by the National Cancer Institute (NCI) of the National Institutes of Health (NIH) under award number CA097452 (U01 & UM1). The content is solely the responsibility of the authors and does not necessarily represent the official views of the NCI. T.M. was supported by the Japan Research Foundation for Clinical Pharmacology and the Uehara Memorial Foundation.

Supporting information

Figure S1 Individual posthoc Bayesian temsirolimus clearance (CL) estimates. (A) CL (l h–1) vs. age (years) and (B) allometrically scaled CL (l h–1 70 kg–1) vs. age. Solid line represents the line of fit by the Emax model

Supporting info item

Mizuno, T. , Fukuda, T. , Christians, U. , Perentesis, J. P. , Fouladi, M. , and Vinks, A. A. (2017) Population pharmacokinetics of temsirolimus and sirolimus in children with recurrent solid tumours: a report from the Children's Oncology Group. Br J Clin Pharmacol, 83: 1097–1107. doi: 10.1111/bcp.13181.

References

  • 1. Southan C, Sharman JL, Benson HE, Faccenda E, Pawson AJ, Alexander SP, et al. The IUPHAR/BPS Guide to PHARMACOLOGY in 2016: towards curated quantitative interactions between 1300 protein targets and 6000 ligands. Nucl Acids Res 2016; 44: D1054–D1068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Alexander SPH, Fabbro D, Kelly E, Marrion N, Peters JA, Benson HE, et al. The Concise Guide to PHARMACOLOGY 2015/16: Enzymes. Br J Pharmacol 2015; 172: 6024–6109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Kwitkowski VE, Prowell TM, Ibrahim A, Farrell AT, Justice R, Mitchell SS, et al. FDA approval summary: temsirolimus as treatment for advanced renal cell carcinoma. Oncologist 2010; 15: 428–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Galanis E, Buckner JC, Maurer MJ, Kreisberg JI, Ballman K, Boni J, et al. Phase II trial of temsirolimus (CCI‐779) in recurrent glioblastoma multiforme: a North Central Cancer Treatment Group Study. J Clin Oncol 2005; 23: 5294–5304. [DOI] [PubMed] [Google Scholar]
  • 5. Chan S, Scheulen ME, Johnston S, Mross K, Cardoso F, Dittrich C, et al. Phase II study of temsirolimus (CCI‐779), a novel inhibitor of mTOR, in heavily pretreated patients with locally advanced or metastatic breast cancer. J Clin Oncol 2005; 23: 5314–5322. [DOI] [PubMed] [Google Scholar]
  • 6. Smith SM, van Besien K, Karrison T, Dancey J, McLaughlin P, Younes A, et al. Temsirolimus has activity in non‐mantle cell non‐Hodgkin's lymphoma subtypes: The University of Chicago phase II consortium. J Clin Oncol 2010; 28: 4740–4746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Okuno S, Bailey H, Mahoney MR, Adkins D, Maples W, Fitch T, et al. A phase 2 study of temsirolimus (CCI‐779) in patients with soft tissue sarcomas: a study of the Mayo phase 2 consortium (P2C). Cancer 2011; 117: 3468–3475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Gerullis H, Eimer C, Ecke TH, Georgas E, Freitas C, Kastenholz S, et al. A phase II trial of temsirolimus in second‐line metastatic urothelial cancer. Med Oncol 2012; 29: 2870–2876. [DOI] [PubMed] [Google Scholar]
  • 9. Geoerger B, Kieran MW, Grupp S, Perek D, Clancy J, Krygowski M, et al. Phase II trial of temsirolimus in children with high‐grade glioma, neuroblastoma and rhabdomyosarcoma. Eur J Cancer 2012; 48: 253–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Spunt SL, Grupp SA, Vik TA, Santana VM, Greenblatt DJ, Clancy J, et al. Phase I study of temsirolimus in pediatric patients with recurrent/refractory solid tumors. J Clin Oncol 2011; 29: 2933–2940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Bauman JE, Arias‐Pulido H, Lee SJ, Fekrazad MH, Ozawa H, Fertig E, et al. A phase II study of temsirolimus and erlotinib in patients with recurrent and/or metastatic, platinum‐refractory head and neck squamous cell carcinoma. Oral Oncol 2013; 49: 461–467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Alvarez EA, Brady WE, Walker JL, Rotmensch J, Zhou XC, Kendrick JE, et al. Phase II trial of combination bevacizumab and temsirolimus in the treatment of recurrent or persistent endometrial carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol 2013; 129: 22–27. [DOI] [PubMed] [Google Scholar]
  • 13. Margolin KA, Moon J, Flaherty LE, Lao CD, Akerley WL 3rd, Othus M, et al. Randomized phase II trial of sorafenib with temsirolimus or tipifarnib in untreated metastatic melanoma (S0438). Clin. Cancer Res 2012; 18: 1129–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Gandhi L, Bahleda R, Tolaney SM, Kwak EL, Cleary JM, Pandya SS, et al. Phase I study of neratinib in combination with temsirolimus in patients with human epidermal growth factor receptor 2‐dependent and other solid tumors. J Clin Oncol 2014; 32: 68–75. [DOI] [PubMed] [Google Scholar]
  • 15. Diaz‐Padilla I, Hirte H, Oza AM, Clarke BA, Cohen B, Reedjik M, et al. A phase Ib combination study of RO4929097, a gamma‐secretase inhibitor, and temsirolimus in patients with advanced solid tumors. Invest New Drugs 2013; 31: 1182–1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Lee EQ, Kuhn J, Lamborn KR, Abrey L, DeAngelis LM, Lieberman F, et al. Phase I/II study of sorafenib in combination with temsirolimus for recurrent glioblastoma or gliosarcoma: North American Brain Tumor Consortium study 05‐02. Neuro Oncol 2012; 14: 1511–1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Kollmannsberger C, Hirte H, Siu LL, Mazurka J, Chi K, Elit L, et al. Temsirolimus in combination with carboplatin and paclitaxel in patients with advanced solid tumors: a NCIC‐CTG, phase I, open‐label dose‐escalation study (IND 179). Ann Oncol 2012; 23: 238–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Bryce AH, Rao R, Sarkaria J, Reid JM, Qi Y, Qin R, et al. Phase I study of temsirolimus in combination with EKB‐569 in patients with advanced solid tumors. Invest New Drugs 2012; 30: 1934–1941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Punt CJ, Boni J, Bruntsch U, Peters M, Thielert C. Phase I and pharmacokinetic study of CCI‐779, a novel cytostatic cell‐cycle inhibitor, in combination with 5‐fluorouracil and leucovorin in patients with advanced solid tumors. Ann Oncol 2003; 14: 931–937. [DOI] [PubMed] [Google Scholar]
  • 20. Hidalgo M, Buckner JC, Erlichman C, Pollack MS, Boni JP, Dukart G, et al. A phase I and pharmacokinetic study of temsirolimus (CCI‐779) administered intravenously daily for 5 days every 2 weeks to patients with advanced cancer. Clin Cancer Res 2006; 12: 5755–5763. [DOI] [PubMed] [Google Scholar]
  • 21. Atkins MB, Hidalgo M, Stadler WM, Logan TF, Dutcher JP, Hudes GR, et al. Randomized phase II study of multiple dose levels of CCI‐779, a novel mammalian target of rapamycin kinase inhibitor, in patients with advanced refractory renal cell carcinoma. J Clin Oncol 2004; 22: 909–918. [DOI] [PubMed] [Google Scholar]
  • 22. Boni JP, Leister C, Bender G, Fitzpatrick V, Twine N, Stover J, et al. Population pharmacokinetics of CCI‐779: correlations to safety and pharmacogenomic responses in patients with advanced renal cancer. Clin Pharmacol Ther 2005; 77: 76–89. [DOI] [PubMed] [Google Scholar]
  • 23. Raymond E, Alexandre J, Faivre S, Vera K, Materman E, Boni J, et al. Safety and pharmacokinetics of escalated doses of weekly intravenous infusion of CCI‐779, a novel mTOR inhibitor, in patients with cancer. J Clin Oncol 2004; 22: 2336–2347. [DOI] [PubMed] [Google Scholar]
  • 24. Boni JP, Hug B, Leister C, Sonnichsen D. Intravenous temsirolimus in cancer patients: clinical pharmacology and dosing considerations. Semin Oncol 2009; 36 (Suppl. 3): S18–S25. [DOI] [PubMed] [Google Scholar]
  • 25. Cai P, Tsao R, Ruppen ME. In vitro metabolic study of temsirolimus: preparation, isolation, and identification of the metabolites. Drug Metab Dispos 2007; 35: 1554–1563. [DOI] [PubMed] [Google Scholar]
  • 26. Jacobsen W, Serkova N, Hausen B, Morris RE, Benet LZ, Christians U. Comparison of the in vitro metabolism of the macrolide immunosuppressants sirolimus and RAD. Transplant Proc 2001; 33: 514–515. [DOI] [PubMed] [Google Scholar]
  • 27. Emoto C, Fukuda T, Cox S, Christians U, Vinks AA. Development of a physiologically‐based pharmacokinetic model for sirolimus: predicting bioavailability based on intestinal CYP3A content. CPT Pharmacometrics Syst Pharmacol 2013; 2: e59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Fouladi M, Perentesis JP, Wagner LM, Vinks AA, Reid JM, Ahern C, et al. A Phase I study of cixutumumab (IMC‐A12) in combination with temsirolimus (CCI‐779) in children with recurrent solid tumors: a Children's Oncology Group Phase I consortium report. Clin Cancer Res 2015; 21: 1558–1565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Bagatell R, Norris R, Ingle AM, Ahern C, Voss S, Fox E, et al. Phase 1 trial of temsirolimus in combination with irinotecan and temozolomide in children, adolescents and young adults with relapsed or refractory solid tumors: a Children's Oncology Group Study. Pediatr Blood Cancer 2014; 61: 833–839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Christians U, Jacobsen W, Serkova N, Benet LZ, Vidal C, Sewing KF, et al. Automated, fast and sensitive quantification of drugs in blood by liquid chromatography‐mass spectrometry with on‐line extraction: immunosuppressants. J Chromatogr B Biomed Sci Appl 2000; 748: 41–53. [DOI] [PubMed] [Google Scholar]
  • 31. Lindbom L, Pihlgren P, Jonsson EN. PsN‐Toolkit‐‐a collection of computer intensive statistical methods for non‐linear mixed effect modeling using NONMEM. Comput Methods Programs Biomed 2005; 79: 241–257. [DOI] [PubMed] [Google Scholar]
  • 32. Anderson BJ, Holford NH. Mechanism‐based concepts of size and maturity in pharmacokinetics. Annu Rev Pharmacol Toxicol 2008; 48: 303–332. [DOI] [PubMed] [Google Scholar]
  • 33. Ette EI. Stability and performance of a population pharmacokinetic model. J Clin Pharmacol 1997; 37: 486–495. [DOI] [PubMed] [Google Scholar]
  • 34. Holford N. The visual predictive check ‐ superiority to standard diagnostic (Rorschach) plots. Abstract of the Annual Meeting of the Population Approach Group in Europe 2005; abstr 738.
  • 35. Fujisaka Y, Yamada Y, Yamamoto N, Horiike A, Tamura T. A Phase 1 clinical study of temsirolimus (CCI‐779) in Japanese patients with advanced solid tumors. Jpn J Clin Oncol 2010; 40: 732–738. [DOI] [PubMed] [Google Scholar]
  • 36. U.S. Food and Drug Administration, Drug Approval Package, Clinical Phamracology and Biopharmaceutics Review. Available at http://www.accessdata.fda.gov/drugsatfda_docs/nda/2007/022088s000TOC.cfm (last accessed 24 June 2016).
  • 37. Kearns GL, Abdel‐Rahman SM, Alander SW, Blowey DL, Leeder JS, Kauffman RE. Developmental pharmacology‐‐drug disposition, action, and therapy in infants and children. N Engl J Med 2003; 349: 1157–1167. [DOI] [PubMed] [Google Scholar]
  • 38. Emoto C, Fukuda T, Johnson TN, Adams DM, Vinks AA. Development of a pediatric physiologically based pharmacokinetic model for sirolimus: applying principles of growth and maturation in neonates and infants. CPT Pharmacometrics Syst Pharmacol 2015; 4: 127–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Emoto C, Fukuda T, Mizuno T, Cox S, Schniedewind B, Christians U, et al. Age‐dependent changes in sirolimus metabolite formation in patients with neurofibromatosis type 1. Ther Drug Monit 2015; 37: 395–399. [DOI] [PubMed] [Google Scholar]
  • 40. Weiss B, Widemann BC, Wolters P, Dombi E, Vinks AA, Cantor A, et al. Sirolimus for non‐progressive NF1‐associated plexiform neurofibromas: an NF clinical trials consortium phase II study. Pediatr Blood Cancer 2014; 61: 982–986. [DOI] [PubMed] [Google Scholar]
  • 41. Scott JR, Courter JD, Saldana SN, Widemann BC, Fisher M, Weiss B, et al. Population pharmacokinetics of sirolimus in pediatric patients with neurofibromatosis type 1. Ther Drug Monit 2013; 35: 332–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Adams DM, Trenor CC 3rd, Hammill AM, Vinks AA, Patel MN, Chaudry G, et al. Efficacy and safety of sirolimus in the treatment of complicated vascular anomalies. Pediatrics 2016; 137: e20153257. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Figure S1 Individual posthoc Bayesian temsirolimus clearance (CL) estimates. (A) CL (l h–1) vs. age (years) and (B) allometrically scaled CL (l h–1 70 kg–1) vs. age. Solid line represents the line of fit by the Emax model

Supporting info item


Articles from British Journal of Clinical Pharmacology are provided here courtesy of British Pharmacological Society

RESOURCES