Commentary
Target-mediated drug disposition and dynamics

https://doi.org/10.1016/j.bcp.2005.12.041Get rights and content

Abstract

Nonlinear pharmacokinetics and pharmacodynamics may result from several capacity-limited processes and often represent complicating factors in characterizing the pharmacological properties of drugs. Target-mediated drug disposition (TMDD) corresponds to a special case wherein a significant proportion of a drug (relative to dose) is bound with high affinity to a pharmacological target, such that this interaction is reflected in the pharmacokinetic properties of the drug. Dose-dependent effects on apparent pharmacokinetic parameters may manifest, including the steady-state volume of distribution and total systemic clearance. Although a few small molecular weight compounds have been identified to exhibit TMDD, the incidence of TMDD is likely to increase particularly among emerging biotechnology pharmaceuticals. The goal of this commentary is to describe the basic tenets of TMDD and discuss several mathematical modeling approaches for characterizing this phenomenon. Whereas traditional pharmacokinetic/pharmacodynamic models assume that the amount of the drug–target complex is negligible relative to the total amount of drug in the body, integrated mechanism-based models of TMDD incorporate the binding and stoichiometry of drug–target binding. These models may be utilized to infer the time-course of inaccessible system variables, such as the in vivo density of the drug–target complex, and provide a suitable platform for ascertaining the apparent pharmacodynamic implications of TMDD.

Introduction

Nonlinear conditions in drug disposition and pharmacological effects are encountered frequently, and dose-dependent pharmacokinetic (PK) and pharmacodynamic (PD) processes complicate the characterization of drug concentration-effect and -toxicity relationships [1], often with significant clinical implications [2]. The fundamental principle that governs this behavior is that of capacity-limitation, where limited densities of enzymes or other proteins result in saturable processes and disproportionate changes in net drug exposure or responses with increasing dose. Levy introduced the term target-mediated drug disposition (TMDD) in reference to the observation that for some drugs, the capacity-limited substance responsible for their complex nonlinear pharmacokinetics was in fact the pharmacological target of the compound [3]. Whereas plasma concentrations of most drugs greatly exceed receptor or target concentrations, agents exhibiting TMDD are bound with high affinity and to a significant degree (relative to dose), such that this interaction influences the temporal profile of plasma drug concentrations. Although originally posed to describe the effects of extensive drug–target binding in tissues, TMDD has received considerable interest owing in part to its role in saturable clearance mechanisms for specific peptide and protein pharmaceuticals (e.g., receptor-mediated endocytosis) [4], [5]. The utilization of lower doses of these potent compounds, coupled with the means for detecting relatively low drug concentrations in biological fluids offered by advanced analytical methods, further increases the probability of observing this phenomenon.

The pharmacokinetic characteristics imparted by TMDD are now well recognized; however, systematic approaches for understanding the pharmacodynamic implications of TMDD are still in early development. For some compounds, there is an apparent disconnect between the time-course of target-occupancy and the pharmacological response [3]. Pertinent aspects of the in vivo pharmacological properties of drugs derive from the integration of PK/PD systems [6], and progress in comprehending the significance of TMDD will most likely be made through an iterative consideration of experimental data and mechanism-based modeling. This paradigm should yield opportunities for the rational design of new analogues and/or delivery systems for drugs with TMDD properties, as well as clinical dosing regimens that optimize pharmacotherapy across populations and for individual patients.

Section snippets

General pharmacological expectations

The pharmacokinetic consequences of TMDD may be subtle or pronounced, but in either case, the effects are important and it is convenient to categorize compounds based on whether or not binding to the pharmacological target significantly contributes to the elimination of the drug. Classic examples of small molecular weight compounds that demonstrate TMDD characteristics, but for which saturable elimination mechanisms are not implicated, include various angiotensin-converting enzyme (ACE)

PK/PD systems analysis

Noncompartmental analysis of plasma drug concentration–time data represents a useful starting point in characterizing the pharmacokinetic properties of drugs, and curve-fitting single-dose data is a common method for resolving the slopes, heights, area, and moment (SHAM) properties of such curves that are used to calculate primary pharmacokinetic parameters [21], [22]. This approach may be applied to initially identify the dose-dependent properties of TMDD. Natalizumab is a humanized mAb

Future considerations

Appreciation for TMDD properties and the pharmacodynamic implications of such systems will indubitably increase following the acquisition of much needed experimental data coupled with the continued development and refinement of mechanism-based PK/PD models of this phenomenon. Major goals continue to include the identification of drug and system specific parameters that control exposure-response relationships, as well as patient specific characteristics or covariates that account for

Acknowledgments

The author thanks Dr. William J. Jusko and Dr. Gerhard Levy for introducing him to the concepts of TMDD and pharmacodynamics, many insightful discussions, and various collaborative projects in these fields of study. The author also acknowledges Dr. Wojciech Krzyzanski for his derivation of the equilibrium model of TMDD and for his thoughtful comments and suggestions. Part of this work was supported by Grant GM57980 (to W.J.J.) from the National Institutes of Health and presented at The

References (72)

  • C.J.H. Porter et al.

    Lymphatic transport of proteins after subcutaneous administration

    J Pharm Sci

    (2000)
  • A. Sharma et al.

    Precursor-dependent indirect pharmacodynamic response model for tolerance and rebound phenomena

    J Pharm Sci

    (1998)
  • P. Millet et al.

    Wavelet analysis of dynamic PET data: application to the parametric imaging of benzodiazepine receptor concentration

    Neuroimage

    (2000)
  • A.R. Hilgenbrink et al.

    Folate receptor-mediated drug targeting: from therapeutics to diagnostics

    J Pharm Sci

    (2005)
  • J.H. Lin

    Dose-dependent pharmacokinetics: experimental observations and theoretical considerations

    Biopharm Drug Dispos

    (1994)
  • T.M. Ludden

    Nonlinear pharmacokinetics: clinical implications

    Clin Pharmacokinet

    (1991)
  • G. Levy

    Pharmacologic target-mediated drug disposition

    Clin Pharmacol Ther

    (1994)
  • S.M. Abdel-Rahman et al.

    The integration of pharmacokinetics and pharmacodynamics: understanding dose-response

    Annu Rev Pharmacol Toxicol

    (2004)
  • A.E. Till et al.

    Pharmacokinetics of repeated single oral doses of enalapril maleate (MK-421) in normal volunteers

    Biopharm Drug Dispos

    (1984)
  • R. Brazzell et al.

    Dose-dependent pharmacokinetics of the aldose reductase inhibitor imirestat in man

    Pharm Res

    (1991)
  • J.Y. Chien et al.

    Saturable tissue binding and imirestat pharmacokinetics in rats

    Pharm Res

    (1992)
  • K.R. Lees et al.

    Pharmacokinetics of an ACE inhibitor, S-9780, in man: evidence of tissue binding

    J Pharmacokinet Biopharm

    (1989)
  • C. Weber et al.

    Pharmacokinetics and pharmacodynamics of the endothelin-receptor antagonist bosentan in healthy human subjects

    Clin Pharmacol Ther

    (1996)
  • K. Laine et al.

    Multiple-dose pharmacokinetics of selegiline and desmethylselegiline suggest saturable tissue binding

    Clin Neuropharmacol

    (2000)
  • I. Mahmood et al.

    Pharmacokinetic and pharmacodynamic considerations in the development of therapeutic proteins

    Clin Pharmacokinet

    (2005)
  • K. Terashi et al.

    Close association between clearance of recombinant human granulocyte colony-stimulating factor (G-CSF) and G-CSF receptor on neutrophils in cancer patients

    Antimicrob Agents Chemother

    (1999)
  • R.A. Mufson et al.

    Binding and internalization of recombinant human erythropoietin in murine erythroid precursor cells

    Blood

    (1987)
  • S. Chapel et al.

    Changes in erythropoietin pharmacokinetics following busulfan-induced bone marrow ablation in sheep: evidence for bone marrow as a major erythropoietin elimination pathway

    J Pharmacol Exp Ther

    (2001)
  • R.T. Meijer et al.

    Pharmacokinetics of murine anti-human CD3 antibodies in man are determined by the disappearance of target antigen

    J Pharmacol Exp Ther

    (2002)
  • W.J. Jusko

    Guidelines for collection and analysis of pharmacokinetic data

  • L. Steinman

    Blocking adhesion molecules as therapy for multiple sclerosis: natalizumab

    Nat Rev Drug Discov

    (2005)
  • W.A. Sheremata et al.

    A safety and pharmacokinetic study of intravenous natalizumab in patients with MS

    Neurology

    (1999)
  • D.M. Foster

    Noncompartmental vs. compartmental approaches to pharmacokinetic analysis

  • E. Bellissant et al.

    Methodological issues in pharmacokinetic-pharmacodynamic modelling

    Clin Pharmacokinet

    (1998)
  • L. Michaelis et al.

    Die Kinetik der Invertinwirkung

    Biochem Z

    (1913)
  • W.J. Jusko

    Pharmacokinetics of capacity-limited systems

    J Clin Pharmacol

    (1989)
  • Cited by (192)

    • Analyze impact of tumor-associated kinetics on antibody delivery in solid tumors with a physiologically based pharmacokinetics/pharmacodynamics model

      2021, European Journal of Pharmaceutics and Biopharmaceutics
      Citation Excerpt :

      These compartments represent three tumor layers defined based on their relative access to extravasated mAb (Fig. 1B). The target-mediated binding kinetics were described separately for each tumor compartment [14,15]. The apparent extravasation flux was assumed to be the driving force for antibody delivery from tumor microvessels through the three sequentially accessible tumor compartments.

    View all citing articles on Scopus
    View full text