Skip to main content
Erschienen in: Cancer Chemotherapy and Pharmacology 1/2020

Open Access 01.01.2020 | Short Communication

A limited sampling schedule to estimate individual pharmacokinetics of pemetrexed in patients with varying renal functions

verfasst von: Nikki de Rouw, Sabine Visser, Stijn L. W. Koolen, Joachim G. J. V. Aerts, Michel M. van den Heuvel, Hieronymus J. Derijks, David M. Burger, Rob ter Heine

Erschienen in: Cancer Chemotherapy and Pharmacology | Ausgabe 1/2020

Abstract

Purpose

Pemetrexed is a widely used cytostatic agent with an established exposure–response relationship. Although dosing is based on body surface area (BSA), large interindividual variability in pemetrexed plasma concentrations is observed. Therapeutic drug monitoring (TDM) can be a feasible strategy to reduce variability in specific cases leading to potentially optimized pemetrexed treatment. The aim of this study was to develop a limited sampling schedule (LSS) for the assessment of pemetrexed pharmacokinetics.

Methods

Based on two real-life datasets, several limited sampling designs were evaluated on predicting clearance, using NONMEM, based on mean prediction error (MPE %) and normalized root mean squared error (NRMSE %). The predefined criteria for an acceptable LSS were: a maximum of four sampling time points within 8 h with an MPE and NRMSE ≤ 20%.

Results

For an accurate estimation of clearance, only four samples in a convenient window of 8 h were required for accurate and precise prediction (MPE and NRMSE of 3.6% and 5.7% for dataset 1 and of 15.5% and 16.5% for dataset 2). A single sample at t = 24 h performed also within the criteria with MPE and NRMSE of 5.8% and 8.7% for dataset 1 and of 11.5% and 16.4% for dataset 2. Bias increased when patients had lower creatinine clearance.

Conclusions

We presented two limited sampling designs for estimation of pemetrexed pharmacokinetics. Either one can be used based on preference and feasibility.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Pemetrexed is an anti-folate drug which is widely used as first and second-line treatment of non-small cell lung cancer and mesothelioma [1, 2]. There is a relationship between pemetrexed pharmacokinetics and toxicity [35]. Despite the introduction of prophylactic use of folic acid and vitamin B12 to reduce the risk of haematological toxicity, neutropenia remains a main exposure-related and treatment-limiting adverse reaction [3]. Latz et al. [3] showed that higher exposure relates to both decrease in neutrophil count and a longer recovery time after neutropenia.
Currently, pemetrexed is dosed based on body surface area (BSA) and this introduces large intraindividual variability in exposure [6]. There are several other factors which can contribute to variability in exposure, such as change in renal function or drug interactions [6-9]. Since pemetrexed exposure correlates well with toxicity [3, 10], pharmacokinetically (PK) guided dosing may be a feasible strategy to optimize treatment. Previously, the proposed target for safe and effective treatment is an AUC of 164 mg*h/L ± 25% [3, 6]. A prerequisite to validate this target for PK-guided dosing is the availability of an accurate, precise and clinically feasible limited sampling schedule (LSS) to assess the AUC.
From a patient’s perspective, a minimally invasive strategy is desired in a short time window. Therefore, our aim was to develop a LSS for the assessment of pemetrexed pharmacokinetics to use in clinical practice.

Methods

Limited sampling design evaluation

The predictive performance of several limited sampling designs to predict the pemetrexed clearance were evaluated. To assess individual exposure, the AUC can be calculated from clearance and the administered dose (\(\mathrm{A}\mathrm{U}\mathrm{C}= \frac{\mathrm{D}\mathrm{o}\mathrm{s}\mathrm{e}}{\mathrm{C}\mathrm{l}\mathrm{e}\mathrm{a}\mathrm{r}\mathrm{a}\mathrm{n}\mathrm{c}\mathrm{e}}\)). The previously developed and validated pharmacokinetic model by Latz et al. [3] was used to obtain the empirical Bayesian estimates for clearance using the post-hoc option in the software package NONMEM v7.4.3 [Icon, Ireland]. First, the full pharmacokinetic curves were fitted and obtained clearances were assumed to be ‘true values’. Subsequently, individual clearances were estimated from several limited sampling strategies based on the original dataset with certain timepoints removed.
The predictive performance was assessed with the mean relative prediction error (MPE %) for precision and normalized root mean squared error (NRMSE %) for accuracy, respectively. For MPE, confidence intervals were calculated as described by Sheiner et al. [11]. For NRMSE, relative uncertainty was determined according to the distribution-free approach of Faber [12]. Subsequently, corresponding confidence intervals were calculated.
Taking both patient’s perspective and statistical considerations into account, the pragmatical criteria for an acceptable LSS were defined as: a maximum of four sampling time points within 8 h with an MPE and NRMSE ≤ 20%. The value of acceptable precision, and, therefore, bias of clearance, depends on multiple factors such as expected analytical error, therapeutic range of the drug and the purpose of the LSS. For pemetrexed, we found this performance acceptable for the estimation of pemetrexed clearance.

Datasets

Two separate datasets were used to evaluate several sampling designs. The first set contained pharmacokinetic data of 15 pemetrexed patients (from Visser et al. 2019) with adequate renal function (range creatinine clearance according to Cockcroft–Gault (CrCl–CG) 60–166 ml/min) [5]. Patients were treated according to label with a pemetrexed dose of 500 mg/m2 over a 10 min intravenous infusion. For dataset 1, the following sampling times were available 0.17–0.5–1–2–4–8–24 h after the start of administration. The second set included rich pharmacokinetic data of 47 individuals from JMAW phase I trial of Eli Lilly, with varying renal function (range CrCl–CG 17–200 ml/min.). These data were obtained through www.​clinicalstudydat​arequest.​com [13]. The dose varied between patients but was administered over a 10 min intravenous infusion. The sampling times were 0.17–0.25–0.5–1–2–4–6–8–12–24–48–72 h after the start of administration. Since the used model of Latz et al. [3] was designed based on sampling up to 36 h after administration of pemetrexed, datapoints after 36 h were excluded from the analysis.

Results

Table 1 presents the relevant baseline characteristics of the patients that were included in the two datasets and the results of the two best performing limited sampling designs. The second dataset contains patients with a wider range of both creatinine clearance and pemetrexed dose. For both datasets, several designs were tested based on the available sampling times. For an adequate estimation of pemetrexed clearance, within a sampling window of 8 h, four sampling times were required to reach acceptable precision and accuracy (MPE and NRMSE < 20%) in both datasets (not all data shown). As can be seen in Table 1, sampling at 0.5–2–4–8 h after administration resulted in an MPE and NRMSE of 3.6% and 5.7% for dataset 1. Using the second dataset, the performance of this sampling strategy was slightly lower but still within the acceptable range, with and MPE and NRMSE 15.5% and 16.5%, respectively. Table 1 also shows the performance of a single sample at t = 24 h. This strategy performed more or less equal to multiple sampling within 8 h, with imprecision and inaccuracy in the same order of magnitude. For all sampling designs, the MPE confidence interval did not include zero in both datasets, indicating a structural bias.
Table 1
Baseline characteristics and predictive performance of best performing limited sampling designs
https://static-content.springer.com/image/art%3A10.1007%2Fs00280-019-04006-x/MediaObjects/280_2019_4006_Tab1_HTML.png
MPE mean percentage error, NRMSE normalized root mean squared error, hrs hours
Figure 1a–d show true versus predicted pemetrexed clearance for the two proposed limited sampling designs. There is an acceptable correlation between the predicted and true clearances. Single sampling at t = 24 h for dataset 1 (see panel C) apparently introduces a slight overprediction of pemetrexed clearance. This is not observed in the second dataset. In Fig. 1e–h, creatinine clearances versus bias (MPE %) are visualized. Generally, for lower creatinine clearances in dataset 2, a larger prediction error (MPE %) was observed.

Discussion

Our aim was to develop a patient-friendly limited sampling strategy for pemetrexed to assess the exposure in clinical practice and for research purposes. We found that two approaches resulted in the acceptable estimation of clearance (which serves as a proxy for the exposure). We propose two possible sampling schedules: the first consists of four sampling times at 0.5–2–4–8 h after pemetrexed administration. The second approach is a single sample at t = 24 h. These sampling schedules can be used for dose optimization and therapeutic drug monitoring, in specific cases as proposed earlier. Either one can be chosen based on preference and practical feasibility.
In general, the selected LSSs seemed to slightly overpredict clearance in both datasets and both sampling strategies. Overprediction of clearance could possibly result in unwarranted dose adjustments resulting in toxic exposure. However, taking the proposed target AUC of 164 mg*h/L ± 25% in mind, this structural overprediction is not considered relevant, because it is still well within the therapeutic range. Especially for dataset 2, bias increased with decreasing creatinine clearance. An explanation for the observation of increasing bias is that the used model of Latz et al. [3] was developed using patients with adequate renal function. In renal impairment, larger variability may be introduced, which is not observed in patients with adequate renal function. Also, with decreasing clearance, early datapoints in the pharmacokinetic curve become less informative. For dataset 2, removing the 8 h timepoint resulted in unacceptable loss of accuracy and precision. Additionally, the result of the t = 24 strategy in dataset 2 showed that at a later sampling time there may be less bias in patients with extremely low creatinine clearance. Altogether, a single sample at t = 24 may a feasible strategy for clinical practice, but it may require an extra hospital visit for the patient instead of a short prolongation of stay.
Our limited samplings strategy aimed to accurately predict pemetrexed AUC. Although Latz et al. have previously suggested that pharmacokinetically-guided dosing using the AUC may result in improved treatment [3], there is currently no conclusive evidence that the AUC is the best pharmacokinetic parameter to predict efficacy and toxicity. For example, the cytotoxicity of other drugs from the antifolate class, like methotrexate, is concentration threshold driven [14]. Prospective studies should confirm the utility of AUC-guided dosing before implementing this in clinical practice.
Altogether, we presented two patient-friendly and reliable limited sampling designs for estimation of pemetrexed pharmacokinetics. We are now using the 4-point LSS for development of personalized dosing strategies for pemetrexed in ongoing clinical studies [1517].

Compliance with ethical standards

Conflict of interest

The author(s) declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Baldwin CM, Perry CM (2009) Pemetrexed: a review of its use in the management of advanced non-squamous non-small cell lung cancer. Drugs 69(16):2279–2302CrossRef Baldwin CM, Perry CM (2009) Pemetrexed: a review of its use in the management of advanced non-squamous non-small cell lung cancer. Drugs 69(16):2279–2302CrossRef
2.
Zurück zum Zitat European Medicine Agency (EMA) (2017) ALIMTA EPAR—Product Information European Medicine Agency (EMA) (2017) ALIMTA EPAR—Product Information
3.
Zurück zum Zitat Latz JE, Rusthoven JJ, Karlsson MO, Ghosh A, Johnson RD (2006) Clinical application of a semimechanistic-physiologic population PK/PD model for neutropenia following pemetrexed therapy. Cancer Chemother Pharmacol 57(4):427–435CrossRef Latz JE, Rusthoven JJ, Karlsson MO, Ghosh A, Johnson RD (2006) Clinical application of a semimechanistic-physiologic population PK/PD model for neutropenia following pemetrexed therapy. Cancer Chemother Pharmacol 57(4):427–435CrossRef
4.
Zurück zum Zitat Latz JE, Schneck KL, Nakagawa K, Miller MA, Takimoto CH (2009) Population pharmacokinetic/pharmacodynamic analyses of pemetrexed and neutropenia: effect of vitamin supplementation and differences between Japanese and Western patients. Clin Cancer Res 15(1):346–354CrossRef Latz JE, Schneck KL, Nakagawa K, Miller MA, Takimoto CH (2009) Population pharmacokinetic/pharmacodynamic analyses of pemetrexed and neutropenia: effect of vitamin supplementation and differences between Japanese and Western patients. Clin Cancer Res 15(1):346–354CrossRef
5.
Zurück zum Zitat Visser SKSLW, de Bruijn P, Belderbos HNA, Cornelissen R, Mathijssen RHJ, Stricker BH, Aerts JGJV (2019) Pemetrexed exposure predicts for toxicity in advanced non-small-cell lung cancer: a prospective cohort study. Eur J Cancer 121:64–73CrossRef Visser SKSLW, de Bruijn P, Belderbos HNA, Cornelissen R, Mathijssen RHJ, Stricker BH, Aerts JGJV (2019) Pemetrexed exposure predicts for toxicity in advanced non-small-cell lung cancer: a prospective cohort study. Eur J Cancer 121:64–73CrossRef
6.
Zurück zum Zitat Latz JE, Chaudhary A, Ghosh A, Johnson RD (2006) Population pharmacokinetic analysis of ten phase II clinical trials of pemetrexed in cancer patients. Cancer Chemother Pharmacol 57(4):401–411CrossRef Latz JE, Chaudhary A, Ghosh A, Johnson RD (2006) Population pharmacokinetic analysis of ten phase II clinical trials of pemetrexed in cancer patients. Cancer Chemother Pharmacol 57(4):401–411CrossRef
7.
Zurück zum Zitat de Rouw N, Croes S, Posthuma R, Agterhuis DE, Schoenmaekers JJAO, Derijks HJ et al (2019) Pharmacokinetically-guided dosing of pemetrexed in a patient with renal impairment and a patient requiring hemodialysis. Lung Cancer 130:156–158CrossRef de Rouw N, Croes S, Posthuma R, Agterhuis DE, Schoenmaekers JJAO, Derijks HJ et al (2019) Pharmacokinetically-guided dosing of pemetrexed in a patient with renal impairment and a patient requiring hemodialysis. Lung Cancer 130:156–158CrossRef
8.
Zurück zum Zitat Ikemura K, Hamada Y, Kaya C, Enokiya T, Muraki Y, Nakahara H et al (2016) Lansoprazole exacerbates pemetrexed-mediated hematologic toxicity by competitive inhibition of renal basolateral human organic anion transporter 3. Drug Metab Dispos 44(10):1543–1549CrossRef Ikemura K, Hamada Y, Kaya C, Enokiya T, Muraki Y, Nakahara H et al (2016) Lansoprazole exacerbates pemetrexed-mediated hematologic toxicity by competitive inhibition of renal basolateral human organic anion transporter 3. Drug Metab Dispos 44(10):1543–1549CrossRef
9.
Zurück zum Zitat Kawazoe H, Yano A, Ishida Y, Takechi K, Katayama H, Ito R et al (2017) Non-steroidal anti-inflammatory drugs induce severe hematologic toxicities in lung cancer patients receiving pemetrexed plus carboplatin: a retrospective cohort study. PLoS ONE 12(2):e0171066CrossRef Kawazoe H, Yano A, Ishida Y, Takechi K, Katayama H, Ito R et al (2017) Non-steroidal anti-inflammatory drugs induce severe hematologic toxicities in lung cancer patients receiving pemetrexed plus carboplatin: a retrospective cohort study. PLoS ONE 12(2):e0171066CrossRef
10.
Zurück zum Zitat Visser S, Koolen SLW, de Bruijn P, Belderbos HNA, Cornelissen R, Mathijssen RHJ et al (2019) Pemetrexed exposure predicts toxicity in advanced non-small-cell lung cancer: a prospective cohort study. Eur J Cancer 121:64–73CrossRef Visser S, Koolen SLW, de Bruijn P, Belderbos HNA, Cornelissen R, Mathijssen RHJ et al (2019) Pemetrexed exposure predicts toxicity in advanced non-small-cell lung cancer: a prospective cohort study. Eur J Cancer 121:64–73CrossRef
11.
Zurück zum Zitat Sheiner LB, Beal SL (1981) Some suggestions for measuring predictive performance. J Pharmacokinet Biopharm 9(4):503–512CrossRef Sheiner LB, Beal SL (1981) Some suggestions for measuring predictive performance. J Pharmacokinet Biopharm 9(4):503–512CrossRef
12.
Zurück zum Zitat Faber NM (1999) Estimating the uncertainty in estimates of root mean square error of prediction: application to determining the size of an adequate test set in multivariate calibration. Chemom Intell Lab Syst 49(1):79–89CrossRef Faber NM (1999) Estimating the uncertainty in estimates of root mean square error of prediction: application to determining the size of an adequate test set in multivariate calibration. Chemom Intell Lab Syst 49(1):79–89CrossRef
14.
Zurück zum Zitat Chabner BA, Young RC (1973) Threshold methotrexate concentration for in vivo inhibition of DNA synthesis in normal and tumorous target tissues. J Clin Invest 52(8):1804–1811CrossRef Chabner BA, Young RC (1973) Threshold methotrexate concentration for in vivo inhibition of DNA synthesis in normal and tumorous target tissues. J Clin Invest 52(8):1804–1811CrossRef
Metadaten
Titel
A limited sampling schedule to estimate individual pharmacokinetics of pemetrexed in patients with varying renal functions
verfasst von
Nikki de Rouw
Sabine Visser
Stijn L. W. Koolen
Joachim G. J. V. Aerts
Michel M. van den Heuvel
Hieronymus J. Derijks
David M. Burger
Rob ter Heine
Publikationsdatum
01.01.2020
Verlag
Springer Berlin Heidelberg
Erschienen in
Cancer Chemotherapy and Pharmacology / Ausgabe 1/2020
Print ISSN: 0344-5704
Elektronische ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-019-04006-x

Weitere Artikel der Ausgabe 1/2020

Cancer Chemotherapy and Pharmacology 1/2020 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.