Next Article in Journal
Electrochemical and Mechanistic Study of Superoxide Elimination by Mesalazine through Proton-Coupled Electron Transfer
Next Article in Special Issue
Design and Mechanism of Action of a New Prototype of Combi-Molecule “Programed” to Release Bioactive Species at a pH Range Akin to That of the Tumor Microenvironment
Previous Article in Journal
Integrative Genomic–Epigenomic Analysis of Clozapine-Treated Patients with Refractory Psychosis
Previous Article in Special Issue
Simulation-Based Assessment of the Impact of Non-Adherence on Endoxifen Target Attainment in Different Tamoxifen Dosing Strategies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Feasibility of Extrapolating Randomly Taken Plasma Samples to Trough Levels for Therapeutic Drug Monitoring Purposes of Small Molecule Kinase Inhibitors

by
Ruben A. G. van Eerden
1,*,
Esther Oomen-de Hoop
1,
Aad Noordam
1,
Ron H. J. Mathijssen
1 and
Stijn L. W. Koolen
1,2
1
Department of Medical Oncology, Erasmus MC Cancer Institute, 3015GD Rotterdam, The Netherlands
2
Department of Hospital Pharmacy, Erasmus MC, University Medical Center, 3015GD Rotterdam, The Netherlands
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2021, 14(2), 119; https://0-doi-org.brum.beds.ac.uk/10.3390/ph14020119
Submission received: 11 January 2021 / Revised: 28 January 2021 / Accepted: 29 January 2021 / Published: 4 February 2021

Abstract

:
Small molecule kinase inhibitors (SMKIs) are widely used in oncology. Therapeutic drug monitoring (TDM) for SMKIs could reduce underexposure or overexposure. However, logistical issues such as timing of blood withdrawals hamper its implementation into clinical practice. Extrapolating a random concentration to a trough concentration using the elimination half-life could be a simple and easy way to overcome this problem. In our study plasma concentrations observed during 24 h blood sampling were used for extrapolation to trough levels. The objective was to demonstrate that extrapolation of randomly taken blood samples will lead to equivalent estimated trough samples compared to measured Cmin values. In total 2241 blood samples were analyzed. The estimated Ctrough levels of afatinib and sunitinib fulfilled the equivalence criteria if the samples were drawn after Tmax. The calculated Ctrough levels of erlotinib, imatinib and sorafenib met the equivalence criteria if they were taken, respectively, 12 h, 3 h and 10 h after drug intake. For regorafenib extrapolation was not feasible. In conclusion, extrapolation of randomly taken drug concentrations to a trough concentration using the mean elimination half-life is feasible for multiple SMKIs. Therefore, this simple method could positively contribute to the implementation of TDM in oncology.

1. Introduction

During the last decades many small molecule kinase inhibitors (SMKIs) have been developed and become widely available for the treatment of multiple types of malignancies. SMKIs changed the treatment paradigm towards a targeted therapy approach with oral anticancer drugs instead of the traditional intravenous chemotherapy as cancer treatment. For many SMKIs an exposure–response relation has been published in recent literature [1,2,3]. Despite the narrow therapeutic window and large inter-patient variability, they are still prescribed at a fixed dose [4,5,6,7,8,9,10]. Consequently, the risk for severe toxicity due to high exposure or ineffectiveness due to low exposure, is high.
Personalized dosing by therapeutic drug monitoring (TDM) is a way to address and possibly solve the problem of under- or overdosing patients [5]. TDM based dosing is already standard clinical practice for many antibiotic and immunosuppressive drugs and is increasingly being explored for oncological drugs [1,2,11,12,13]. It has been proven to be feasible in oncology and could improve the treatment outcome [4,6,14]. However, logistical issues of TDM often hamper the introduction of personalized dosing.
Targets used for TDM are based on plasma exposure that is measured as AUC (area under the concentration-time curve) or as trough concentration (Ctrough) [2,5,15]. The trough concentration is usually the concentration measured immediately before the administration of a new dose. This specific time point as the target for TDM causes some logistical difficulties. Outpatient visits are planned randomly during the day and a part of the patients take their drugs during the evening when it is very difficult to withdraw blood. A real Ctrough sample is therefore almost impossible to measure. This difficulty could potentially be overcome by the estimation of the Ctrough from a randomly taken sample over time.
A simple method for this estimation might be extrapolation of randomly measured concentration to a trough concentration using the mean population terminal half-life of a drug. This does not require specific patient characteristics. This method was thoroughly investigated by Wang et al. for imatinib and was found feasible [16]. This method is easy to implement in the daily routine. The objective of our study was to investigate whether this method is also feasible for more SMKIs, as this might be helpful for the implementation of TDM in daily oncological practice.

2. Results

In total 2105 blood samples with corresponding trough samples were analyzed. Samples were taken from patients participating in nine different prospective pharmacokinetic studies (Table 1) [17,18,19,20,21,22,23,24,25]. Table 2 presents per drug pharmacokinetic parameters used, analyzed cohorts, relative differences and 90% confidence interval (90%CI) of the relative difference.

2.1. Afatinib

In total, 271 samples were included from 13 patients drawn between 2 h and 12.5 h after intake of afatinib [25]. Based on time after intake and available pharmacokinetic (PK) samples obtained during the prospective PK study, samples were divided into six cohorts based on the available measurements: 2 h to 3 h, 3 h to 4 h, 4 h to 4.5 h, 5.5 h to 6.5 h, 7.5 h to 8.5 h and 11.5 h to 12.5 h, respectively. Relative differences and corresponding 90% confidence interval remained within the range of −20% to +25% for all cohorts (Figure 1A). The relative difference and 90%CI per cohort are presented in Table 2.

2.2. Erlotinib

For erlotinib 455 samples from 59 patients were available for analysis [23,24]. The samples were sorted according to time after once daily intake of the erlotinib and available PK samples obtained during the prospective PK study. The 12 h to 13 h cohort was the only cohort in which the relative differences remained within the −20% to +25% range (RD: 10.8%; 90%CI: 6.2% to 15.5%). The other cohorts consisted of samples taken between 4 h to 4.5 h, 6 h to 7 h and 8 h to 8.5 h (Figure 1B). Detailed results are displayed in Table 2.

2.3. Imatinib

In total, 617 imatinib samples were used in the analysis [18,19]. The relative difference for the first cohort of samples taken at 2.5 h to 3 h after intake of once daily imatinib was 20.0% (90%CI: 5.2% to 37.0%). The relative differences and corresponding 90% confidence interval of the other cohorts remained within the range of −20% to +25% (cohort 3 h to 4 h RD: 19.3% (90%CI: 13.9% to 24.9%); cohort 4 h to 5 h RD: 19.2% (90%CI: 14.0% to 24.7%); cohort 5 h to 6 h RD: 4.1% (90%CI: −0.9% to 9.2%); cohort 6 h to 7 h RD: 15.8% (90%CI: 7.9% to 24.3%)) (Figure 1C, Table 2).

2.4. Regorafenib

For regorafenib, 309 samples from 22 patients were available for analysis [17]. The samples were divided into the following five cohorts based on available PK samples obtained during the prospective PK study: 3 h to 4 h, 4 h to 4.5 h, 6 h to 6.5 h, 8 h to 8.5 h and 12 h to 12.5 h. The relative difference and corresponding 90% confidence interval of the 4 h to 4.5 h cohort and 8 h to 8.5 h cohort remained within the range of −20% to +25% RD (RD: −1.3%; 90%CI: −11.6% to 10.1% and RD: 1.5%; 90%CI: −9.2% to 13.5%, respectively). None of the other cohorts met the predefined equivalence requirements (Figure 1D, Table 2).

2.5. Sorafenib

In total, 128 samples were included from 16 patients drawn between 4 h and 10.5 h after intake of twice daily sorafenib [20]. Only the cohort of samples drawn between 10 h and 10.5 h met the predefined definition of equivalence (RD 0.9% (90%CI: −12.0% to 15.8%)). None of the other cohorts met the equivalence requirements with an RD varying between 8.5% and 13.9% with an upper limit of the 90%CI above the 25% (Figure 1E, Table 2).

2.6. Sunitinib and SU12662

For sunitinib and the active metabolite SU12662, 231 samples were taken between 6 h and 13 h after intake of sunitinib [21,22]. Samples were divided into three cohorts based on available PK samples obtained during the prospective PK study (Table 2). The relative differences of sunitinib in the analyzed cohorts varied between −3.8% and 3.1% while the relative difference of SU12662 varied between −3.6% and 3.8%. The corresponding 90% confidence interval of all cohorts remained within the range of −20% to +25% (Figure 1F).

3. Discussion

In this study we have shown that for most analyzed drugs extrapolation of a randomly taken plasma sample to a trough concentration is feasible. Nevertheless, each drug has its own time point after which the extrapolated Ctrough was equivalent to the truly measured Ctrough. Based on our results we formulated for each investigated drug a recommended interval between previous drug intake and blood withdrawal, which are summarized in Table 3 and Figure 1. For afatinib this interval is recommended to be at least 2 h and our recommended intervals are 3 h, 6 h and 12 h for imatinib, sunitinib and erlotinib, respectively. Extrapolation of sorafenib samples is feasible 10 h after intake of the last dose. However, this is thus only of limited benefit since sorafenib has a dose interval of 12 h. Extrapolation of regorafenib samples seems feasible for samples taken between 3–4.5 h and 8–8.5 h after the last dose. However, given the inconsistency in fulfillment of the predefined equivalence requirements we do not recommend extrapolation of randomly taken regorafenib samples to trough samples. If regorafenib samples are extrapolated, results should be interpreted with caution.
After which time point extrapolation of a blood sample is feasible differs depending on the pharmacokinetic characteristics of each individual drug. Most of the analyzed SMKIs (e.g., sunitinib, imatinib, erlotinib, afatinib) are characterized by a first-order elimination [26,27,28,29]. The equation used in our analyses is based on the pharmacokinetic equation that describes first-order elimination, and is therefore likely to be able to estimate a Ctrough from a randomly taken sample. Although the equation should theoretically approximate the Ctrough, we still observed differences between the estimated Ctrough and the measured Ctrough. This might be explained by the deviation of the standardized Ke used in our analyses compared with the Ke of each individual in clinical practice. We calculated the Ke based on the mean elimination half-life described in the summary of product characteristics (SMPC) that does not take into account patient factors that could influence this PK parameter. Therefore, clinicians should be cautious with the use of this equation and population mean values for the terminal half-life when a drug–drug interaction (e.g., a strong induction of inhibition of CYP3A4) or genetic polymorphism (e.g., CYP3A4*22) affects the drug metabolism [10,30]. Another way of estimating the Ke is used by Wang et al. [16], who calculated Ke based on a population PK model of imatinib. However, the deviation between the estimated non corrected Ctrough and the measured Ctrough for the highest sampling interval (i.e., 17 h) described by Wang et al. [16] was 33.2% for the typical Ke while the deviation in all our cohorts ranged between 4% and 20% which is remarkably smaller. Therefore, it could be suggested that our way of Ke calculation leads to a more accurate Ctrough estimation. However, caution is warranted since the elimination half-life in the SMPC is an average half-life that might be accurate in a large average population but might strongly differ from the elimination half-life of a certain other specific population (e.g., other ethnicities than the majority of Caucasian patients in our studies) [31]. Using an ethnicity specific Ke might therefore also be an option and might be more accurate.
Nonetheless, it could be speculated that Ke estimation based on elimination half-life derived from the SMPC leads to a more standardized way of extrapolation since the SMPC is freely accessible for all clinicians while Ke calculations based on the previously described data (e.g., of a specific population) might vary depending on the used reference.
Extrapolation of randomly taken samples from patients treated with regorafenib and sorafenib is less accurate in estimating the Ctrough. This is probably caused by the enterohepatic recycling of regorafenib and sorafenib [17,32,33]. Enterohepatic recycling could cause a second peak in the plasma concentration-time curve [34]. A second peak in this curve was not taken into account in our analyses which causes a less accurate estimation of the Ctrough.
To improve the estimation of the Ctrough concentration, it could also be suggested to add a correction factor to the algorithm for each drug. Wang et al. [16] applied a correction factor ranging between 0.752 and 0.885 for samples drawn before the following intake of imatinib that reduced the deviation of the estimated Ctrough from the measured Ctrough. A correction factor might also be applicable for our imatinib data given the, in most cohorts observed, relative differences of around the 20% between the estimated Ctrough and the true Ctrough.
As this method of extrapolation is feasible for most of the investigated drugs, this method could also be used for TDM purposes in oncological care. TDM is used to personalize the treatment by adjusting the dose based on the plasma drug concentrations of the patient [5]. By personalizing the dose, TDM aims to reduce underexposure and overexposure to a drug which improves the treatment outcome [5]. Given the random timing of outpatient visits and blood withdrawals, targets used for TDM, as AUC or trough concentrations, are unfortunately difficult to determine in clinical practice. Since feasibility of this extrapolation method has been proven, blood withdrawals can from now on be performed during a bigger window of time. This could resolve the aforementioned logical issues related to TDM in the daily care of cancer patients. However, when using this extrapolation method for TDM purposes a pitfall might be misinterpretation of the drug exposure based on the estimated Ctrough. Because of the relative differences observed between the measured and estimated Ctrough, a patients Ctrough might be overestimated or underestimated. Misinterpretation could lead to an inaccurate TDM advice. Overestimation could lead to falsely maintaining the currently used dose that could lead to less effectiveness while underestimation might lead to a redundant dose increment that could lead to increased toxicity and costs. However, this pitfall might be solved by repeated TDM sampling as proposed in the therapeutic drug monitoring protocol of the Dutch Pharmacology Oncology Group [11].
Bayesian estimation based on a known PK-model is another method which could be used for the estimation of a Ctrough level. Bayesian estimates permit flexible blood withdrawals and provide the opportunity to take patient characteristics into account [15,35]. These kind of adjustments are not possible with the formula used for our analysis [36]. Nevertheless, Bayesian estimates also have their drawbacks. To use this method prior awareness of patients’ characteristics, which are sometimes not available or hard to determine, is desirable, because otherwise the estimation will be guided towards the typical population mean [3,15]. Eventually this will lead to loss of individual information of the patient. Moreover, a previously described PK model, specialized software and knowledge is needed to execute and interpret these calculations [3,36]. In contrast to Bayesian estimates, the method we used is simple to execute in clinical practice and does not depend on which model has been chosen.
In conclusion, our analysis proves that a simple equation using the mean elimination half-life can accurately estimate the Ctrough from a randomly taken plasma sample for SMKIs which are not pharmacokinetically characterized by enterohepatic recycling. This method could reduce the variability and standardize the method used for Ctrough estimation as well as simplify the implementation of TDM in the daily practice of oncology. Further studies are warranted to evaluate the feasibility of the analyzed method when used for other SMKIs.

4. Materials and Methods

Prospective pharmacokinetic trials performed between 2000 and 2021 at the Erasmus MC Cancer Institute, Rotterdam, The Netherlands, investigating SMKIs were used in our analysis.
Studies were included for analysis if they investigated the pharmacokinetics (PK) of SMKIs, at least two blood samples within a 24 h time frame were taken including the trough concentration and patients were treated with the approved dose. Moreover, the exact time of drug intake and blood withdrawal had to be known. Studies or samples were excluded if an intervention that influenced the metabolism of the drug was performed during the blood sampling (e.g., CYP3A4 induction by concomitant study drugs). Blood samples taken before Tmax as stated in the SMPC were also excluded from analysis. If a time range for Tmax was stated in the SMPC the lower boundary was used as cut-off point.
The cohort of patients used for this analysis was obtained from nine prospective studies (Table 1). All studies were approved by the medical ethics committee of the Erasmus MC (MEC2004-018, MEC2008-256, MEC2009-302, MEC2012-138, MEC2014-046, MEC2016-165, MEC2016-590, MEC2017-251, MEC2017-490) and all patients gave written informed consent.
The feasibility of extrapolating randomly taken blood samples will be assessed using the criteria of equivalence (i.e., 90%CI of the geometric mean ratio between 0.8 and 1.25) compared to the measured Cmin value.

4.1. Pharmacokinetic Equation

The PK profile of most of the SMKIs is characterized by first-order elimination [26,27,28,29]. Therefore, the concentration at a random time point during the elimination phase (T2) can be estimated based on the concentration measured at another time point during the elimination phase (T1) using the following pharmacokinetic equation:
C t 2 = C t 1   e x p ( k e × Δ t )
And this equation can be rewritten to:
C t 2 = C t 1   e x p ( 0.693 t 1 2 × Δ t )
In this equation Ct1 represents the plasma drug concentration measured at the moment of blood withdrawal (i.e., a random time-point). T1/2 represents the elimination half-life and is taken from the SMPC for each drug. The factor ∆t represents the time between the moment of blood withdrawal (i.e., known and measured drug concentration) and the moment of the next drug intake (time point of Ctrough). By filling in the aforementioned factors the product Ct2 is calculated which represents the estimated trough concentration.

4.2. Statistical Analysis

IBM SPSS statistics (IBM Corporation, version 25.0. Armonk, NY, USA) was used to build the database and to perform the statistical analyses. The estimated Ctrough (Ctrough estimated) was calculated based on the aforementioned equation per included drug measurement. Log transformation was performed on each Ctrough and Ctrough estimated, because a lognormal distribution was assumed for the plasma concentrations. Samples were grouped according to drug and relative time of sample collection. Per drug and per time-point a paired T-test was performed between the Ctrough and Ctrough estimated on the log transformed data. Mean differences and 90% confidence intervals of these differences were calculated. Exponentiation of the mean differences and 90%CI provided the geometric mean ratio and corresponding 90%CI which can be interpreted as the relative differences in percentages. Equivalence between the Ctrough and Ctrough estimated was shown if the 90%CI was within the lower boundary of –20% and upper boundary of +25% (i.e., geometric mean ratio CI within 0.80 and 1.25). These boundaries are based on the EMA guideline of bio-equivalence studies [37]. Based on this guideline a deviation of the true measured Ctrough which is outside this window could lead to a clinically relevant deviation in exposure. Boundaries smaller than −20% and +25% are difficult given the allowed deviation in measurement of bioanalytical methods used [38]. Extrapolating TDM samples to estimate the trough concentration was considered feasible for clinical implementation if the equivalence requirements were met.

Author Contributions

Conceptualization, R.A.G.v.E., R.H.J.M. and S.L.W.K.; Methodology, R.A.G.v.E., E.O.-d.H. and S.L.W.K.; Software, not applicable; Validation, E.O.-d.H.; Formal Analysis, R.A.G.v.E. and A.N.; Investigation, R.A.G.v.E., E.O.-d.H., A.N. and S.L.W.K.; Resources, not applicable; Data Curation, R.A.G.v.E.; Writing—Original Draft Preparation, R.A.G.v.E., E.O.-d.H., R.H.J.M. and S.L.W.K.; Writing—Review and Editing, R.A.G.v.E., E.O.-d.H., A.N.,R.H.J.M. and S.L.W.K.; Visualization, R.A.G.v.E., E.O.-d.H., R.H.J.M. and S.L.W.K.; Supervision, S.L.W.K.; Project Administration, R.A.G.v.E.; Funding Acquisition, NA. All authors have read and agreed to the published version of the manuscript.

Funding

This study was performed without external funding.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki. The studies included in our analysis were all approved by the Ethics Committee of Erasmus MC (MEC2004-018, MEC2008-256, MEC2009-302, MEC2012-138, MEC2014-046, MEC2016-165, MEC2016-590, MEC2017-251, MEC2017-490).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author (RvE) upon reasonable request.

Conflicts of Interest

The authors declare no potential conflict of interest.

References

  1. Groenland, S.L.; van Nuland, M.; Verheijen, R.B.; Schellens, J.H.M.; Beijnen, J.H.; Huitema, A.D.R.; Steeghs, N. Therapeutic Drug Monitoring of Oral Anti-Hormonal Drugs in Oncology. Clin. Pharm. 2019, 58, 299–308. [Google Scholar] [CrossRef]
  2. Verheijen, R.B.; Yu, H.; Schellens, J.H.M.; Beijnen, J.H.; Steeghs, N.; Huitema, A.D.R. Practical Recommendations for Therapeutic Drug Monitoring of Kinase Inhibitors in Oncology. Clin. Pharmacol. Ther. 2017, 102, 765–776. [Google Scholar] [CrossRef] [PubMed]
  3. Mueller-Schoell, A.; Groenland, S.L.; Scherf-Clavel, O.; van Dyk, M.; Huisinga, W.; Michelet, R.; Jaehde, U.; Steeghs, N.; Huitema, A.D.R.; Kloft, C. Therapeutic drug monitoring of oral targeted antineoplastic drugs. Eur. J. Clin. Pharmacol. 2020. [Google Scholar] [CrossRef]
  4. Verheijen, R.B.; Bins, S.; Mathijssen, R.H.; Lolkema, M.P.; van Doorn, L.; Schellens, J.H.; Beijnen, J.H.; Langenberg, M.H.; Huitema, A.D.; Steeghs, N.; et al. Individualized Pazopanib Dosing: A Prospective Feasibility Study in Cancer Patients. Clin. Cancer Res. 2016, 22, 5738–5746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Groenland, S.L.; Mathijssen, R.H.J.; Beijnen, J.H.; Huitema, A.D.R.; Steeghs, N. Individualized dosing of oral targeted therapies in oncology is crucial in the era of precision medicine. Eur. J. Clin. Pharmacol. 2019, 75, 1309–1318. [Google Scholar] [CrossRef] [PubMed]
  6. Lankheet, N.A.; Kloth, J.S.; Gadellaa-van Hooijdonk, C.G.; Cirkel, G.A.; Mathijssen, R.H.; Lolkema, M.P.; Schellens, J.H.; Voest, E.E.; Sleijfer, S.; de Jonge, M.J.; et al. Pharmacokinetically guided sunitinib dosing: A feasibility study in patients with advanced solid tumours. Br. J. Cancer 2014, 110, 2441–2449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Lankheet, N.A.; Knapen, L.M.; Schellens, J.H.; Beijnen, J.H.; Steeghs, N.; Huitema, A.D. Plasma concentrations of tyrosine kinase inhibitors imatinib, erlotinib, and sunitinib in routine clinical outpatient cancer care. Ther. Drug Monit. 2014, 36, 326–334. [Google Scholar] [CrossRef] [PubMed]
  8. Verheijen, R.B.; Beijnen, J.H.; Schellens, J.H.M.; Huitema, A.D.R.; Steeghs, N. Clinical Pharmacokinetics and Pharmacodynamics of Pazopanib: Towards Optimized Dosing. Clin. Pharm. 2017, 56, 987–997. [Google Scholar] [CrossRef] [Green Version]
  9. de Wit, D.; Guchelaar, H.J.; den Hartigh, J.; Gelderblom, H.; van Erp, N.P. Individualized dosing of tyrosine kinase inhibitors: Are we there yet? Drug Discov. Today 2015, 20, 18–36. [Google Scholar] [CrossRef]
  10. Hussaarts, K.; Veerman, G.D.M.; Jansman, F.G.A.; van Gelder, T.; Mathijssen, R.H.J.; van Leeuwen, R.W.F. Clinically relevant drug interactions with multikinase inhibitors: A review. Ther. Adv. Med. Oncol. 2019, 11. [Google Scholar] [CrossRef]
  11. Groenland, S.L.; van Eerden, R.A.G.; Verheijen, R.B.; Koolen, S.L.W.; Moes, D.; Desar, I.M.E.; Reyners, A.K.L.; Gelderblom, H.J.; van Erp, N.P.; Mathijssen, R.H.J.; et al. Therapeutic Drug Monitoring of Oral Anticancer Drugs: The Dutch Pharmacology Oncology Group-Therapeutic Drug Monitoring Protocol for a Prospective Study. Ther. Drug Monit. 2019, 41, 561–567. [Google Scholar] [CrossRef]
  12. Roberts, J.A.; Norris, R.; Paterson, D.L.; Martin, J.H. Therapeutic drug monitoring of antimicrobials. Br. J. Clin. Pharmacol. 2012, 73, 27–36. [Google Scholar] [CrossRef] [Green Version]
  13. Brunet, M.; van Gelder, T.; Asberg, A.; Haufroid, V.; Hesselink, D.A.; Langman, L.; Lemaitre, F.; Marquet, P.; Seger, C.; Shipkova, M.; et al. Therapeutic Drug Monitoring of Tacrolimus-Personalized Therapy: Second Consensus Report. Ther. Drug Monit. 2019, 41, 261–307. [Google Scholar] [CrossRef]
  14. IJzerman, N.S.; Groenland, S.L.; Koenen, A.M.; Kerst, M.; van der Graaf, W.T.A.; Rosing, H.; Beijnen, J.H.; Huitema, A.D.R.; Steeghs, N. Therapeutic drug monitoring of imatinib in patients with gastrointestinal stromal tumours—Results from daily clinical practice. Eur. J. Cancer 2020, 136, 140–148. [Google Scholar] [CrossRef]
  15. de Jonge, M.E.; Huitema, A.D.; Schellens, J.H.; Rodenhuis, S.; Beijnen, J.H. Individualised cancer chemotherapy: Strategies and performance of prospective studies on therapeutic drug monitoring with dose adaptation: A review. Clin. Pharm. 2005, 44, 147–173. [Google Scholar] [CrossRef]
  16. Wang, Y.; Chia, Y.L.; Nedelman, J.; Schran, H.; Mahon, F.X.; Molimard, M. A therapeutic drug monitoring algorithm for refining the imatinib trough level obtained at different sampling times. Ther. Drug Monit. 2009, 31, 579–584. [Google Scholar] [CrossRef]
  17. de Man, F.M.; Hussaarts, K.; de With, M.; Oomen-de Hoop, E.; de Bruijn, P.; van Halteren, H.K.; van der Burg-de Graauw, N.; Eskens, F.; van Gelder, T.; van Leeuwen, R.W.F.; et al. Influence of the Proton Pump Inhibitor Esomeprazole on the Bioavailability of Regorafenib: A Randomized Crossover Pharmacokinetic Study. Clin. Pharmacol. Ther. 2019, 105, 1456–1461. [Google Scholar] [CrossRef] [PubMed]
  18. Eechoute, K.; Franke, R.M.; Loos, W.J.; Scherkenbach, L.A.; Boere, I.; Verweij, J.; Gurney, H.; Kim, R.B.; Tirona, R.G.; Mathijssen, R.H.; et al. Environmental and genetic factors affecting transport of imatinib by OATP1A2. Clin. Pharmacol. Ther. 2011, 89, 816–820. [Google Scholar] [CrossRef]
  19. Eechoute, K.; Fransson, M.N.; Reyners, A.K.; de Jong, F.A.; Sparreboom, A.; van der Graaf, W.T.; Friberg, L.E.; Schiavon, G.; Wiemer, E.A.; Verweij, J.; et al. A long-term prospective population pharmacokinetic study on imatinib plasma concentrations in GIST patients. Clin. Cancer Res. 2012, 18, 5780–5787. [Google Scholar] [CrossRef] [Green Version]
  20. Hussaarts, K.; van Doorn, L.; Eechoute, K.; Damman, J.; Fu, Q.; van Doorn, N.; Eisenmann, E.D.; Gibson, A.A.; Oomen-de Hoop, E.; de Bruijn, P.; et al. Influence of Probenecid on the Pharmacokinetics and Pharmacodynamics of Sorafenib. Pharmaceutics 2020, 12, 788. [Google Scholar] [CrossRef]
  21. Kloth, J.S.; Binkhorst, L.; de Wit, A.S.; de Bruijn, P.; Hamberg, P.; Lam, M.H.; Burger, H.; Chaves, I.; Wiemer, E.A.; van der Horst, G.T.; et al. Relationship Between Sunitinib Pharmacokinetics and Administration Time: Preclinical and Clinical Evidence. Clin. Pharm. 2015, 54, 851–858. [Google Scholar] [CrossRef] [Green Version]
  22. Kloth, J.S.; Klumpen, H.J.; Yu, H.; Eechoute, K.; Samer, C.F.; Kam, B.L.; Huitema, A.D.; Daali, Y.; Zwinderman, A.H.; Balakrishnar, B.; et al. Predictive value of CYP3A and ABCB1 phenotyping probes for the pharmacokinetics of sunitinib: The ClearSun study. Clin. Pharm. 2014, 53, 261–269. [Google Scholar] [CrossRef]
  23. van Leeuwen, R.W.; Peric, R.; Hussaarts, K.G.; Kienhuis, E.; NS, I.J.; de Bruijn, P.; van der Leest, C.; Codrington, H.; Kloover, J.S.; van der Holt, B.; et al. Influence of the Acidic Beverage Cola on the Absorption of Erlotinib in Patients with Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2016, 34, 1309–1314. [Google Scholar] [CrossRef]
  24. Veerman, G.D.M.; Hussaarts, K.; Peric, R.; Oomen-de Hoop, E.; Landa, K.D.; van der Leest, C.H.; Broerse, S.D.; Rutten, H.B.; Belderbos, H.N.A.; Steendam, C.M.J.; et al. Influence of Cow’s Milk and Esomeprazole on the Absorption of Erlotinib: A Randomized, Crossover Pharmacokinetic Study in Lung Cancer Patients. Clin. Pharm. 2020. [Google Scholar] [CrossRef]
  25. BIO-GIO Study—Netherlands Trial Register NL6336 (NTR6652). Available online: https://www.trialregister.nl/trial/6336 (accessed on 16 December 2020).
  26. Freiwald, M.; Schmid, U.; Fleury, A.; Wind, S.; Stopfer, P.; Staab, A. Population pharmacokinetics of afatinib, an irreversible ErbB family blocker, in patients with various solid tumors. Cancer Chemother. Pharmacol. 2014, 73, 759–770. [Google Scholar] [CrossRef]
  27. Delbaldo, C.; Chatelut, E.; Re, M.; Deroussent, A.; Seronie-Vivien, S.; Jambu, A.; Berthaud, P.; Le Cesne, A.; Blay, J.Y.; Vassal, G. Pharmacokinetic-pharmacodynamic relationships of imatinib and its main metabolite in patients with advanced gastrointestinal stromal tumors. Clin. Cancer Res. 2006, 12, 6073–6078. [Google Scholar] [CrossRef] [Green Version]
  28. Houk, B.E.; Bello, C.L.; Kang, D.; Amantea, M. A population pharmacokinetic meta-analysis of sunitinib malate (SU11248) and its primary metabolite (SU12662) in healthy volunteers and oncology patients. Clin. Cancer Res. 2009, 15, 2497–2506. [Google Scholar] [CrossRef] [Green Version]
  29. Evelina, C.; Guidi, M.; Khoudour, N.; Pascaline, B.-R.; Fabre, E.; Tlemsani, C.; Arrondeau, J.; Francois, G.; Vidal, M.; Schneider, M.P.; et al. Population Pharmacokinetics of Erlotinib in Patients with Non-small Cell Lung Cancer: Its Application for Individualized Dosing Regimens in Older Patients. Clin. Ther. 2020, 42, 1302–1316. [Google Scholar] [CrossRef]
  30. Diekstra, M.H.; Klumpen, H.J.; Lolkema, M.P.; Yu, H.; Kloth, J.S.; Gelderblom, H.; van Schaik, R.H.; Gurney, H.; Swen, J.J.; Huitema, A.D.; et al. Association analysis of genetic polymorphisms in genes related to sunitinib pharmacokinetics, specifically clearance of sunitinib and SU12662. Clin. Pharmacol. Ther. 2014, 96, 81–89. [Google Scholar] [CrossRef] [Green Version]
  31. Touma, J.A.; McLachlan, A.J.; Gross, A.S. The role of ethnicity in personalized dosing of small molecule tyrosine kinase inhibitors used in oncology. Transl. Cancer Res. 2017, 6, S1558–S1591. [Google Scholar] [CrossRef]
  32. Jain, L.; Woo, S.; Gardner, E.R.; Dahut, W.L.; Kohn, E.C.; Kummar, S.; Mould, D.R.; Giaccone, G.; Yarchoan, R.; Venitz, J.; et al. Population pharmacokinetic analysis of sorafenib in patients with solid tumours. Br. J. Clin. Pharmacol. 2011, 72, 294–305. [Google Scholar] [CrossRef] [Green Version]
  33. Keunecke, A.; Hoefman, S.; Drenth, H.J.; Zisowsky, J.; Cleton, A.; Ploeger, B.A. Population pharmacokinetics of regorafenib in solid tumours: Exposure in clinical practice considering enterohepatic circulation and food intake. Br. J. Clin. Pharmacol. 2020, 86, 2362–2376. [Google Scholar] [CrossRef] [PubMed]
  34. Bins, S.; van Doorn, L.; Phelps, M.A.; Gibson, A.A.; Hu, S.; Li, L.; Vasilyeva, A.; Du, G.; Hamberg, P.; Eskens, F.; et al. Influence of OATP1B1 Function on the Disposition of Sorafenib-beta-D-Glucuronide. Clin. Transl. Sci. 2017, 10, 271–279. [Google Scholar] [CrossRef] [Green Version]
  35. Rousseau, A.; Marquet, P. Application of pharmacokinetic modelling to the routine therapeutic drug monitoring of anticancer drugs. Fundam. Clin. Pharmacol. 2002, 16, 253–262. [Google Scholar] [CrossRef]
  36. Donagher, J.; Martin, J.H.; Barras, M.A. Individualised medicine: Why we need Bayesian dosing. Intern. Med. J. 2017, 47, 593–600. [Google Scholar] [CrossRef]
  37. Agency, E.M. Guideline on the Investigation of Bioequivalence. Available online: https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-investigation-bioequivalence-rev1_en.pdf (accessed on 25 January 2021).
  38. Agency, E.M. Validation of Analytical Procedures. Available online: https://www.ema.europa.eu/en/documents/scientific-guideline/ich-q-2-r1-validation-analytical-procedures-text-methodology-step-5_en.pdf (accessed on 25 January 2021).
Figure 1. Observed relative differences with corresponding 90% confidence interval of the estimated trough concentration compared to measured trough concentration per drug over time: (A) Relative differences observed in the afatinib cohort; (B) Relative differences observed in the erlotinib cohort; (C) Relative differences observed in the imatinib cohort; (D) Relative differences observed in the regorafenib cohort; (E) Relative differences observed in the sorafenib cohort; (F) Relative differences observed in the sunitinib and SU12662 cohorts). The green fields represent the values of bio-equivalence. The line below the x-axis represents our recommended extrapolation intervals between previous drug intake and blood withdrawal for extrapolation (i.e., red: not recommended, green: extrapolation useful).
Figure 1. Observed relative differences with corresponding 90% confidence interval of the estimated trough concentration compared to measured trough concentration per drug over time: (A) Relative differences observed in the afatinib cohort; (B) Relative differences observed in the erlotinib cohort; (C) Relative differences observed in the imatinib cohort; (D) Relative differences observed in the regorafenib cohort; (E) Relative differences observed in the sorafenib cohort; (F) Relative differences observed in the sunitinib and SU12662 cohorts). The green fields represent the values of bio-equivalence. The line below the x-axis represents our recommended extrapolation intervals between previous drug intake and blood withdrawal for extrapolation (i.e., red: not recommended, green: extrapolation useful).
Pharmaceuticals 14 00119 g001
Table 1. Used prospective pharmacokinetic trials.
Table 1. Used prospective pharmacokinetic trials.
Title of Prospective Pharmacokinetic StudyReference
A long-term prospective population pharmacokinetic study on imatinib plasma concentrations in GIST patients[19]
Environmental and genetic factors affecting transport of imatinib by OATP1A2[18]
Predictive Value of CYP3A and ABCB1 Phenotyping Probes for the Pharmacokinetics of Sunitinib: the ClearSun Study[22]
Relationship Between Sunitinib Pharmacokinetics and Administration Time: Preclinical and Clinical Evidence[21]
Influence of the Acidic Beverage Cola on the Absorption of Erlotinib in Patients With Non-Small-Cell Lung Cancer[23]
Influence of Cow’s Milk and Esomeprazole on the Absorption of Erlotinib: A Randomized, Crossover Pharmacokinetic Study in Lung Cancer Patients[24]
Influence of Probenecid on the Pharmacokinetics and Pharmacodynamics of Sorafenib[20]
Influence of the Proton Pump Inhibitor Esomeprazole on the Bioavailability of Regorafenib: A Randomized Crossover Pharmacokinetic Study[17]
The effects of esomeprazole on the bioavailability of afatinib in patients with non-small-cell lung cancer[25]
Table 2. Investigated cohorts and relative differences of the estimated trough concentration compared to measured trough concentration.
Table 2. Investigated cohorts and relative differences of the estimated trough concentration compared to measured trough concentration.
SMKITmax (h) T1/2 (h)DI (h)Patients (n)Time after Intake (h)Blood Withdrawal (n)RD (%)90%CI RD (%)
Afatinib2 to 53724132.00 to 2.9866−6.4−12.0 to −0.3
133.00 to 3.98701.1−4.2 to 6.7
134.00 to 4.13309.52.6 to 17.0
135.50 to 6.483610.14.5 to 16.0
137.50 to 8.30359.84.2 to 15.8
1311.50 to 12.7334−3.6−8.8 to 1.9
Erlotinib43624554.00 to 4.279833.024.5 to 42.1
595.90 to 6.6312024.718.4 to 31.2
597.87 to 8.3311927.221.2 to 33.5
5911.92 to 13.0011810.86.2 to 15.5
Imatinib2.51824252.58 to 2.983720.05.2 to 37.0
613.00 to 3.9820019.313.9 to 24.9
614.00 to 4.9217619.214.0 to 24.7
585.00 to 6.001504.1−0.9 to 9.2
346.02 to 7.085415.87.9 to 24.3
Regorafenib3 to 420 to 3024223.00 to 3.9810615.97.3 to 25.1
224.00 to 4.5847−1.3−11.6 to 10.1
225.90 to 6.5052−12.0−22.3 to 0.4
227.88 to 8.50521.5−9.2 to 13.5
2211.82 to 12.1052−12.6−22.7 to −1.3
Sorafenib325 to 4812163.98 to 4.223216.90.4 to 36.0
165.97 to 6.15328.5−6.2 to 25.6
167.93 to 8.433213.9−1.6 to 31.7
169.93 to 10.25320.9−12.0 to 15.8
Sunitinib6 to 1240 to 60 24176.00 to 6.2730−3.8−9.1 to 1.8
757.63 to 8.671493.10.6 to 5.8
2411.95 to 12.4252−2.5−5.6 to −0.7
SU126626 to 1280 to 11024176.00 to 6.2730−3.6−9.8 to 3.1
757.63 to 8.671481.3−1.5 to 4.1
2311.95 to 12.42523.8−7.8 to 0.3
Abbrevations: SMKI = Small Molecule Kinase Inhibitor; Tmax = time point when maximum concentration is reached in plasma; T1/2 = elimination half-life of drug; DI = dosing interval; RD = Relative Difference; CI = confidence interval; SU12662 = active metabolite of sunitinib.
Table 3. Recommended intervals between previous drug intake and blood withdrawal.
Table 3. Recommended intervals between previous drug intake and blood withdrawal.
DrugRecommended Interval
Afatinib≥2 h
Erlotinib≥12 h
Imatinib≥3 h
RegorafenibNot recommended
Sorafenib≥10 h
Sunitinib (and SU12662)≥6 h
Abbrevations: SU12662 = active metabolite of sunitinib.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

van Eerden, R.A.G.; Oomen-de Hoop, E.; Noordam, A.; Mathijssen, R.H.J.; Koolen, S.L.W. Feasibility of Extrapolating Randomly Taken Plasma Samples to Trough Levels for Therapeutic Drug Monitoring Purposes of Small Molecule Kinase Inhibitors. Pharmaceuticals 2021, 14, 119. https://0-doi-org.brum.beds.ac.uk/10.3390/ph14020119

AMA Style

van Eerden RAG, Oomen-de Hoop E, Noordam A, Mathijssen RHJ, Koolen SLW. Feasibility of Extrapolating Randomly Taken Plasma Samples to Trough Levels for Therapeutic Drug Monitoring Purposes of Small Molecule Kinase Inhibitors. Pharmaceuticals. 2021; 14(2):119. https://0-doi-org.brum.beds.ac.uk/10.3390/ph14020119

Chicago/Turabian Style

van Eerden, Ruben A. G., Esther Oomen-de Hoop, Aad Noordam, Ron H. J. Mathijssen, and Stijn L. W. Koolen. 2021. "Feasibility of Extrapolating Randomly Taken Plasma Samples to Trough Levels for Therapeutic Drug Monitoring Purposes of Small Molecule Kinase Inhibitors" Pharmaceuticals 14, no. 2: 119. https://0-doi-org.brum.beds.ac.uk/10.3390/ph14020119

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop