QSPR models for the prediction of apparent volume of distribution

https://doi.org/10.1016/j.ijpharm.2006.03.043Get rights and content

Abstract

An estimate of volume of distribution (Vd) is of paramount importance both in drug choice as well as maintenance and loading dose calculations in therapeutics. It can also be used in the prediction of drug biological half life. This study employs quantitative structure–pharmacokinetic relationship (QSPR) techniques for the prediction of volume of distribution. Values of Vd for 129 drugs were collated from the literature. Structural descriptors consisted of partitioning, quantum mechanical, molecular mechanical, and connectivity parameters calculated by specialized software and pKa values obtained from ACD labs/log D database. Genetic algorithm and stepwise regression analyses were used for variable selection and model development. Models were validated using a leave-many-out procedure. QSPR analyses resulted in a number of significant models for acidic and basic drugs separately, and for all the drugs. Validation studies showed that mean fold error of predictions for the selected models were between 1.79 and 2.17. Although separate QSPR models for acids and bases resulted in lower prediction errors than models for all the drugs, the external validation study showed a limited applicability for the equation obtained for acids. Therefore, the universal model that requires only calculated structural descriptors was recommended. The QSPR model is able to predict the volume of distribution of drugs belonging to different chemical classes with a prediction error similar to that of the other more complicated prediction methods including the commonly practiced interspecies scaling. The structural descriptors in the model can be interpreted based on the known mechanisms of distribution and the molecular structures of the drugs.

Introduction

The concentration of drug in the plasma or tissues depends on the amount of drug systemically absorbed and the volume in which the drug is distributed as well as the clearance. The apparent volume of distribution in the body, Vd, is a key pharmacokinetic parameter which determines the extent of drug distribution. It is simply a proportionality constant which relates the amount of drug in the body and/or compartments of the body to its plasma concentration. Therefore, despite the fact that Vd has no physical or anatomical meaning, it represents a measure of the relative partitioning of drug between plasma (the central compartment) and the tissues. An estimate of Vd is of paramount importance both in drug choice as well as maintenance and loading dose calculations in therapeutics. Moreover, Vd, in conjunction with clearance, are the two pharmacokinetic parameters determining the drug biological half life. A number of different Vd terms have been defined in the literature. Eq. (1) represents the Vd of the central compartment as the dose taken divided by the plasma concentration of the drug at time zero (C0) (Shargel and Yu, 1999).Vd=DoseC0

Two different terms have been used to describe the volume of distribution for drugs that follow multiple exponential decay. The first, designated Vdarea, is calculated as the ratio of clearance to the rate of decline of concentration during the elimination phase of the logarithmic concentration versus time curve:Vdarea=DosekAUCThe second volume term is the volume of distribution at steady state (Vdss) which represents the volume in which a drug would appear to be distributed during steady state if the drug existed throughout that volume at the same concentration as that in the measured fluid (plasma or blood). It should be mentioned that when using pharmacokinetics to make drug dosing decisions, the difference between Vdarea and Vdss is not usually clinically significant (Wilkinson, 2001).

The volume of distribution in man is traditionally predicted from in vivo data in preclinical animals with appropriate scaling to man (Smith et al., 2001). This can be based on allometric scaling using the body weight (BW) of the species as is represented by Eq. (3).Vd=aBWbwhere a and b are regression coefficients, and b is ca. 0.9–1.0. In cases where plasma protein binding varies across the species, allometric scaling should be based upon the volume of distribution corrected for the extent of protein binding (Vd of unbound drug). The more successful animal scaling prediction methods include the scaling of fractal volume of distribution (Karalis et al., 2001) and a method based on the incorporation of unbound fraction of drug in tissues of animals as well as human plasma protein binding values for the estimation of Vd in human taking into account physiological parameters such as extracellular fluid and plasma volumes (Obach et al., 1997).

Quantitative structure–pharmacokinetic relationships (QSPRs) offer a convenient alternative to animal scaling (Van de Waterbeemd, 2005). This is particularly of interest in view of the need for high-throughput in vitro screening of absorption, distribution, metabolism, and excretion (ADME) in earlier stages of drug development process. Previous QSPR studies have focused on classification into different ratings of volume of distribution (Hirono et al., 1994) as well as regression models for congeneric series of molecules (Gobburu and Shelver, 1995, Turner et al., 2003), and structurally unrelated drugs (Ritschel et al., 1995, Karalis et al., 2002, Lombardo et al., 2004, Ghafourian et al., 2004). In a previous study we developed QSPR models for the prediction of Vd of structurally unrelated drugs (Ghafourian et al., 2004). In this investigation, a larger numbers of drug Vd values have been collated from the literature and a wider range of structural descriptors have been incorporated. The special emphasis of the present investigation is on the interpretability of the models and rigorous leave-many-out validation process. The large number of compounds used in the study as well as the fact that they cover a wide range of chemical and pharmacological classes can add to the significance and consistency of the models. Predictions have been made based on different QSPR models for acidic drugs and basic drugs separately, as well as for all the drugs together. A comparison with previous QSPR, and other prediction methods, is made.

Section snippets

Dataset

Volume of distribution was collected from the literature for 129 drug entities, belonging to different pharmacological and chemical classes (Ritschel et al., 1995, Moffat et al., 1986, Perry, 2002, Ritschel and Hammer, 1980, Ritschel, 1976, Durnas et al., 1990, Raaflaub and Speiser-Courvoisier, 1974, Lam et al., 1997, Nattell et al., 1987, Schoerlin et al., 1990, Glare and Walsh, 1991, Sonne et al., 1988, Greenblatt, 1981, Fulton and Sorkin, 1995). These included, among others, benzodiazepines,

Results

The apparent volume of distribution (Vd) and the extent of protein binding for the compounds used in this study are listed in Table 1, together with the relevant references. Also included in the table are pKa values from ACD labs database or the calculated values. Drugs used in the study covered a wide range of chemical and pharmacological classes with the Vd values ranging from 0.1 to 112.4 L kg−1. Stepwise regression and genetic algorithm led to a number of significant QSPR models from which

Discussion

Volume of distribution is an important pharmacokinetic property that needs to be determined during drug development process. It is normally estimated using animal scaling which is associated with certain levels of error (Mahmood, 1998). QSPR technique can offer an alternative method for the estimation, especially in early stages of drug development. One particular limitation of QSPR could be the narrow range of applicability which arises from the limited chemical space covered by the training

Conclusion

Apparent volume of distribution for drug entities belonging to different chemical classes was studied using a QSPR approach. Some of the suggested QSPR models resulted in encouragingly low prediction errors. The errors were within the range of more complicated prediction methods such as interspecies scaling and a method requiring experimentally determined parameters, e.g. extent of plasma protein binding. Furthermore, the structural descriptors used in the models can be interpreted based on the

Acknowledgments

We are grateful to the Research Council of Tabriz Medical Sciences University for financial support of parts of this study. T.G. thanks Dr. Mark Cronin from Liverpool John Moores University for providing some of the descriptors used in the study.

References (37)

  • M. Feher et al.

    A simple model for the prediction of blood-brain partitioning

    Int. J. Pharm.

    (2000)
  • J.V.S. Gobburu et al.

    Quantitative structure pharmacokinetic relationships (QSPR) of beta blockers derived using neural networks

    J. Pharm. Sci.

    (1995)
  • J.V. Turner et al.

    Multiple pharmacokinetic parameter prediction for a series of cephalosporins

    J. Pharm. Sci.

    (2003)
  • M.H. Abraham et al.

    Hydrogen bonding—Part 46. A review of the correlation and prediction of transport properties by an LFER method: physicochemical properties, brain penetration and skin permeability

    Pestic. Sci.

    (1999)
  • G. Barbeau et al.

    Pharmacokinetics of nalidixic acid in old and young volunteers

    J. Clin. Pharmacol.

    (1982)
  • G.E. Blakey et al.

    Quantitative structure–pharmacokinetics relationships: I. Development of a whole body physiologically based model to characterize changes in pharmacokinetics across a homologous series of barbiturates in the rat

    J. Pharmacokinet. Biopharm.

    (1997)
  • R.E. Cutler et al.

    Flucytosine kinetics in subjects with normal and impaired renal function

    Clin. Pharmacol. Ther.

    (1978)
  • A.M. Davis et al.

    Robust assessment of statistical significance in the use of unbound/intrinsic pharmacokinetic parameters in quantitative structure–pharmacokinetic relationships with lipophilicity

    Drug Metab. Dispos.

    (2000)
  • J.C. Dearden et al.

    Hydrogen bonding parameters for QSAR: comparison of indicator variables, counts, molecular orbital and other parameters

    J. Chem. Inf. Comp. Sci.

    (1999)
  • C. Durnas et al.

    Hepatic drug metabolism and aging

    Clin. Pharmacokinet.

    (1990)
  • B. Fulton et al.

    Propofol an overview of its pharmacology and a review of its clinical efficacy in intensive care sedation

    Drugs

    (1995)
  • T. Ghafourian et al.

    Quantitative structure-pharmacokinetics relationships study of the apparent volume of distribution of CNS drugs

    J. Pharm. Pharmacol.

    (2004)
  • P.A. Glare et al.

    Clinical pharmacokinetics of morphine

    Ther. Drug Monit.

    (1991)
  • D.J. Greenblatt

    Clinical pharmacokinetics of oxazepam and lorazepam

    Clin. Pharmacokinet.

    (1981)
  • S. Hirono et al.

    Non-congeneric structure–pharmacokinetic property correlation studies using fuzzy adaptive least squares: volume of distribution

    Biol. Pharm. Bull.

    (1994)
  • V. Karalis et al.

    Fractal volume of drug distribution: it scales proportionally to body mass

    Pharm. Res.

    (2001)
  • V. Karalis et al.

    Multivariate statistics of disposition pharmacokinetic parameters for structurally unrelated drugs used in therapeutics

    Pharm. Res.

    (2002)
  • E. Klinge et al.

    Pharmacokinetics of methenamine in healthy volunteers

    J. Antimicrob. Chemother.

    (1982)
  • Cited by (57)

    • Biopharmaceutical characters and bioavailability improving strategies of ginsenosides

      2018, Fitoterapia
      Citation Excerpt :

      The elimination half-life(t1/2β), peak time(Tmax), apparent volume of distribution(Vd)and bioavailability(F) are important parameters to describe pharmacokinetics in vivo. Specifically, t1/2β is the main parameter to express the elimination rate of drugs, Tmax is mainly used to describe the rate of absorption, Vd is a key pharmacokinetic parameter which determines the extent of drug distribution, F is used to evaluate the absorption degree of drugs [61, 62]. Clinically, oral administration is the most common route of administration.

    View all citing articles on Scopus
    View full text