Population pharmacokinetic modelling of unbound and total plasma concentrations of paclitaxel in cancer patients

https://doi.org/10.1016/S0959-8049(03)00126-6Get rights and content

Abstract

The aim of this study was to validate and further develop a mechanism-based population pharmacokinetic model for paclitaxel (Taxol®; Bristol-Myers Squibb Co, Princeton, NJ, USA) based on the knowledge of Cremophor EL (CrEL) micelle entrapment and to evaluate the exposure/toxicity relationships. Paclitaxel (total and unbound) and CrEL concentrations were obtained according to a sparse sampling scheme with on average only 3.5 samples per course from 45 patients with solid tumours who received 3-hour infusions of paclitaxel (final dose range 112–233 mg/m2). The present data were predicted well by the mechanism-based model. In addition, bilirubin and body size were found to be significant as covariates. A change in body surface area (BSA) of 0.1 m2 typically caused a change in clearance (CL) of 22.3 l/h and an increase in bilirubin of 10 μM typically caused a decrease in CL of 41 l/h. Toxicity was best described by a threshold model. In conclusion, even with a sparse sampling scheme, the same mechanism-based binding components as in the previously developed model could be identified. Once the CrEL and total paclitaxel plasma concentrations are known, the unbound concentrations, which are more closely related to the haematological toxicity, can be predicted.

Introduction

The non-linear pharmacokinetics of paclitaxel in plasma has been described in several studies. The non-linearity has mostly been described as both saturable elimination and distribution (saturable transport [1] or saturable binding [2]), but also as caused by the micelle-forming vehicle Cremophor EL (CrEL) 3, 4, 5. CrEL has been suggested to inhibit P-glycoprotein-mediated biliary secretion [6], cause lipoprotein dissociation that would alter protein binding [7] and, recently, it has been suggested to cause an alteration in the distribution in human blood by entrapment in micelles [5]. The free fraction of paclitaxel has been shown to decrease with increasing CrEL concentrations [5]. Paclitaxel has previously been shown to bind to both albumin and α1-acid glycoprotein [8]. In our previous model [9], the non-linearity could be explained to a large extent by binding that was directly proportional to the CrEL concentration.

The pharmacokinetic/pharmacodynamic (PK/PD) relationship for paclitaxel toxicity has previously been described by a threshold model 6, 10 and also by more general models using a non-linear continuous function 11, 12. Since the free fraction of paclitaxel has been shown to vary over time and the drug effect is generally considered to be more closely related to the unbound drug, the PK/PD relationship based on the total concentration needs to be re-evaluated.

The aim of this study was to validate a previously developed mechanism-based population pharmacokinetic model that describes unbound and total plasma concentrations of paclitaxel, to extend the model with covariates and, based on new toxicity data, to characterise the PK/PD relationship based on unbound and total concentrations of paclitaxel.

Section snippets

Patients and methods

This study included 14 male and 31 female patients with different types of solid tumours (colorectal, gastric, gall bladder, breast, uterine, ovarian, pancreas) at advanced stages, not amenable to standard therapy. They received paclitaxel as a 3-hour intravenous (i.v.) infusion with an initial dose of 175 mg/m2 every third week within a trial on the feasibility of chemotherapy selection based on drug sensitivity testing ex vivo. Dose adjustments, escalations and reductions, were based on any

Pharmacokinetics

Observed concentrations of CrEL, unbound (Cu) and total concentrations (Cp) of paclitaxel are shown in Fig. 1. The unbound concentrations were well described by the earlier model. As can be seen in Fig. 2, the predictions from model A and dose (i) (left panel) deviated to some extent from the line of identity. The bias in etas (interindividual variability+interoccasion variability), in the predictions from EB estimates based on Model A, dose and observed unbound concentrations (ii) (middle

Discussion

These data were well described by the previously developed model and also the unbound concentrations were well predicted from total plasma concentrations of paclitaxel, CrEL and the model. Both two and three compartment non-linear pharmacokinetic models have been used when describing paclitaxel pharmacokinetics 1, 2, 6. In this study, there was not enough data to support more than two compartments. No dose dependence could be seen in the free concentrations. The typical value of CL in the

Acknowledgements

We are grateful to the patients who kindly participated in this study. We would also like to thank Britt Jansson, Eric Brouwer and Karin Sjöberg for their technical assistance, Siv Jönsson for many helpful discussions regarding model building and, finally, we thank the Swedish Cancer Society and Bristol Myers Squibb Clinical Oncology Research Department for their financial support.

References (25)

  • A. Sparreboom et al.

    Quantitation of Cremophor EL in human plasma samples using a colorimetric dye-binding microassay

    Anal. Biochem.

    (1998)
  • E. Brouwer et al.

    Linearized colorimetric assay for cremophor ELapplication to pharmacokinetics after 1-hour paclitaxel infusions

    Anal. Biochem.

    (1998)
  • D.S. Sonnichsen et al.

    Saturable pharmacokinetics and paclitaxel pharmacodynamics in children with solid tumors

    J. Clin. Oncol.

    (1994)
  • M.O. Karlsson et al.

    Pharmacokinetic models for the saturable distribution of paclitaxel

    Drug Metab. Dispos.

    (1999)
  • L. van Zuylen et al.

    Pharmacokinetic modeling of paclitaxel encapsulation in Cremophor EL micelles

    Cancer Chemother. Pharmacol.

    (2001)
  • O. van Tellingen et al.

    Cremophor EL causes (pseudo-) non-linear pharmacokinetics of paclitaxel in patients

    Br. J. Cancer

    (1999)
  • A. Sparreboom et al.

    Cremophor EL-mediated alteration of paclitaxel distribution in human bloodclinical pharmacokinetic implications

    Cancer Res.

    (1999)
  • L. Gianni et al.

    Nonlinear pharmacokinetics and metabolism of paclitaxel and its pharmacokinetic/pharmacodynamic relationships in humans

    J. Clin. Oncol.

    (1995)
  • E. Sykes et al.

    Effects of Cremophor EL on distribution of Taxol to serum lipoproteins

    Br. J. Cancer

    (1994)
  • G.N. Kumar et al.

    Binding of taxol to human plasma, albumin and alpha 1-acid glycoprotein

    Res. Commun. Chem. Pathol. Pharmacol.

    (1993)
  • A. Henningsson et al.

    Mechanism-based pharmacokinetic model for paclitaxel

    J. Clin. Oncol.

    (2001)
  • M.T. Huizing et al.

    Pharmacokinetics of paclitaxel and metabolites in a randomized comparative study in platinum-pretreated ovarian cancer patients

    J. Clin. Oncol.

    (1993)
  • Cited by (69)

    • Management of hyperbilirubinaemia in pancreatic cancer patients

      2018, European Journal of Cancer
      Citation Excerpt :

      Seventy per cent of the administered dose was eliminated by bile duct. Therefore, in the presence of liver dysfunction, its metabolism was altered [32]. Some small studies, with heterogeneous patient populations, have examined the influence of hepatic dysfunction and hyperbilirubinemia in pharmacokinetics and toxicity of paclitaxel [32–35].

    • Analytical and clinical validation of a dried blood spot assay for the determination of paclitaxel using high-performance liquid chromatography-tandem mass spectrometry

      2018, Clinical Biochemistry
      Citation Excerpt :

      The interindividual tolerability to these effects is partially related to pharmacokinetics and pharmacogenetics differences among patients, particularly in PCT clearance [8]. PCT pharmacokinetics is nonlinear due to saturable elimination and distribution of the drug [9]. PCT metabolic clearance can be affected by genetic polymorphisms [10], demographic, physiologic and pathologic factors and by drug interactions [11].

    • Differentially expressed proteins in human MCF-7 breast cancer cells sensitive and resistant to paclitaxel

      2015, Experimental Cell Research
      Citation Excerpt :

      Increased levels of these enzymes result in enhanced degradation of paclitaxel [15] and overexpression of these isoforms has been found in some taxane resistant cancer cells [16,17]. Moreover, individual genetic variability between patients exists and can also play a role [18,19]. A possible correlation between polymorphism of these cyps involved in taxane metabolism (and also polymorphism of ABCB1 transporter) and effects of taxane treatment was searched for but results of these studies were controversial [20–22].

    • Pharmacokinetic behaviors of soft nanoparticulate formulations of chemotherapeutics

      2023, Wiley Interdisciplinary Reviews: Nanomedicine and Nanobiotechnology
    View all citing articles on Scopus
    1

    Current address: Clinical Pharmacology Research Core, Medical Oncology Clinical Research Unit, National Cancer Institute, Bethesda, MD 20892, USA.

    View full text