Abstract
Purpose of Review
Prior to human studies, knowledge of drug disposition in the body is useful to inform decisions on drug safety and efficacy, first in human dosing, and dosing regimen design. It is therefore of interest to develop predictive models for primary pharmacokinetic parameters, clearance, and volume of distribution. The volume of distribution of a drug is determined by the physiological properties of the body and physiochemical properties of the drug, and is used to determine secondary parameters, including the half-life. The purpose of this review is to provide an overview of current methods for the prediction of volume of distribution of drugs, discuss a comparison between the methods, and identify deficiencies in current predictive methods for future improvement.
Recent Findings
Several volumes of distribution prediction methods are discussed, including preclinical extrapolation, physiological methods, tissue composition-based models to predict tissue:plasma partition coefficients, and quantitative structure-activity relationships. Key factors that impact the prediction of volume of distribution, such as permeability, transport, and accuracy of experimental inputs, are discussed. A comparison of current methods indicates that in general, all methods predict drug volume of distribution with an absolute average fold error of 2-fold. Currently, the use of composition-based PBPK models is preferred to models requiring in vivo input.
Summary
Composition-based models perfusion-limited PBPK models are commonly used at present for prediction of tissue:plasma partition coefficients and volume of distribution, respectively. A better mechanistic understanding of important drug distribution processes will result in improvements in all modeling approaches.
Keywords: Distribution, Volumeofdistribution, Tissue:plasmapartitioncoefficients, Membranepartitioning, Predictionmodels
Introduction
Prediction of drug pharmacokinetics in humans is critically important early in drug discovery and development. Noncompartmental analysis, compartmental models, and physiologically based pharmacokinetic (PBPK) models are commonly utilized to characterize the absorption, distribution, metabolism, and elimination of drugs. Primary PK parameters—drug clearance (CL), volume of distribution (V, or Vss for steady-state volume of distribution), and bioavailability (F) upon a non-systemic dose—aid in the characterization of drug plasma concentration-time profiles. This manuscript focuses on Vss, and methods for the prediction of Vss in drug discovery.
Volume of distribution (V) of a drug is a proportionality constant to describe the relationship between the concentration of drug in the plasma to the amount of drug in the body [1]. It is often described as an “apparent volume of distribution,” describing a mathematical volume in which the drug distributes in the body [2]. The Vss of a drug is determined by the physiological properties of the body and physiochemical properties of the drug, and is used to determine secondary parameters, including the half-life [2]. These factors include plasma protein binding, tissue partitioning, and drug transporters as discussed below (Fig. 1). The volume of distribution can be experimentally determined with a variety of PK modeling techniques, with clinical plasma concentration-time data. Any estimation of Vss requires IV dosing (bolus or infusion) data. If only oral data are available, an oral volume (V/F) can be calculated, but the extent of distribution is unknown. In a drug discovery setting, while the volume of distribution is often considered in drug design, it does not necessarily need to be optimized for drugs. For example, increasing volume of distribution not only increases the elimination half-life of a compound but also decreases the unbound target-site concentration. Understanding the disposition of a drug early in drug discovery is important in order to determine efficacious human doses, to design dosing regimens, as well as to characterize the therapeutic window of a drug. Only unbound drug is assumed to cross membranes and to distribute into cells out of the plasma, but as discussed below, poor permeability across membranes and/or the presence of transporters can alter the equilibrium assumed by the free drug hypothesis [1]. Unbound drug concentrations at the target site are relevant in order to characterize the therapeutic index of a drug, and in vitro assays as well as predictive models should ideally provide a useful correlation between the unbound target-site drug concentration and the total plasma drug concentration.
Fig. 1.

Determinants of drug distribution. Drug (D) can bind to erythrocytes as well as plasma proteins in the blood. Unbound drug diffuses out of capillaries into extracellular fluid (ECF), where it can bind to plasma proteins in the ECF. Unbound drug reversibly diffuses across the plasma membrane, or can be actively transported into or out of cells. Cell partitioning in intracellular organelles such as lysosomes, as well as partitioning into phospholipids and neutral lipids in the cell, is depicted
To understand the kinetics of drug distribution, both the rate and extent of distribution must be considered. The rate of distribution for permeable drugs in many tissues is often perfusion-limited (blood-flow limited), as the only restriction to the rate of distribution is the blood perfusion rate to the tissue. In contrast, some drugs in some tissues exhibit permeability-limited distribution, when membrane permeability is a barrier that restricts partitioning into tissues. Early in drug discovery/development, predictive models often assume perfusion-limited distribution of drugs. The extent of distribution is determined by tissue:plasma partition coefficients (Kp) and tissue volumes, which together determine the Vss of a drug.
Plasma Protein Binding
Plasma protein binding of drugs is a key determinant of the volume of distribution. Drugs generally bind to plasma proteins (including albumin, alpha acid glycoprotein or AAG, and lipoproteins) in a relatively nonspecific manner. In general, acidic drugs bind primarily to plasma albumin, bases predominantly to AAG, and hydrophobic compounds to lipoproteins. However, there are many exceptions, e.g., some bases and many neutral molecules bind albumin. The fraction of drug unbound in plasma (fup) is calculated as the unbound plasma concentration compared with the total concentration in the plasma at equilibrium. Drugs with high plasma protein binding tend to have smaller volumes of distribution compared to drugs that are not highly plasma protein bound [1]. Plasma protein binding in the extracellular fluid also occurs, and the smallest volume that a highly albumin-bound drug can have is about 7 L in a healthy human [1]. This corresponds to the volume of distribution of plasma proteins. In contrast to plasma, the blood additionally has red blood cells (RBCs), and drug binding to RBC membranes is typically measured with a blood/plasma ratio (BP). The hematocrit of the blood sample, fup, and the affinity of the drug for the RBCs determines the value of BP [1].
Accurate determination of fup is critical, since this value is used for the prediction of drug clearance as well as volume of distribution [3]. Equilibrium dialysis under controlled pH conditions (in the presence of 5–10% CO2) provides more consistent estimates of fup compared with assays conducted in the absence of CO2, or with ultracentrifugation methods. The determination of BP is relatively straightforward.
Tissue Partitioning
Another key determinant of the extent of drug distribution is the partitioning of drugs into tissues. Tissue partitioning is determined by the physicochemical properties of the drug, as well as the composition of each specific tissue. Drugs with high tissue partitioning will generally have a large Vss. Drugs typically partition into tissue membrane components, including phospholipids and neutral lipids, as well as cellular lipid components and tissue proteins. Tissue partitioning is characterized by the equilibrium tissue to plasma partition coefficient of a drug in a tissue (Kp). Currently, the most commonly used collection of experimental data for tissue Kp values is provided in Rodgers et al. [4, 5] and includes the Kp of 67 drugs in various tissues in rats. As discussed below, various methods are employed to predict tissue Kp values of drugs, for prediction of Vss, and for use in PBPK modeling.
Overall, the observed Vss of drugs is a result of the competition between plasma protein binding and tissue partitioning. However, extensive plasma protein binding does not necessarily result in a low Vss, when tissue partitioning is extensive. For example, tamoxifen is > 98% plasma protein bound but has a Vss of ~ 4000 L [6].
Blood Flow and Perfusion-Limited Distribution
In perfusion-limited distribution, the rate-limiting step to distribution is the blood flow to the organ/tissue. The rate of distribuion of highly permeable drugs is typically perfusion-limited, and uptake or efflux transporters generally do not have a significant impact. Distribution occurs rapidly in highly perfused tissues, and in tissues with fenestrated or discontinuous capillaries, e.g., liver and spleen. Tissues like muscle and skin, which have continuous capillaries and low perfusion rates, often exhibit slower rates of drug distribution [7]. Even more restrictive is the blood-brain barrier (BBB), which has tight junctions between endothelial cells, and drugs must cross membranes to reach the extracellular space. For most PBPK modeling efforts in early drug discovery, drugs are assumed to exhibit perfusion-limited distribution in all organs, and the impact of membrane permeability and transporters is often ignored. Resulting models may predict the extent of drug distribution accurately but poorly predict the rate of distribution.
Membrane Permeability, Permeability-Limited Distribution, and Transporters
Poorly permeable drugs are limited in their distribution into tissues by membrane barriers at the capillary or the cell. Organs such as the brain are protected by the BBB, and most drugs exhibit permeability-limited distribution in the brain. Efflux transporters can lower the unbound drug concentration in the tissue compared to the plasma, as seen with the efflux of drugs by P-glycoprotein at the BBB [8]. Drugs with poor permeability are also often substrates for uptake transporters. The impact of uptake transporters in drug distribution is exemplified by atorvastatin uptake in the liver [9]. Active uptake transport results in higher unbound drug concentration in the tissue compared to the plasma at equilibrium.
The various physiologic complexities interplay with physicochemical properties of drug molecules to eventually determine Vss of drugs. Various methods have been developed for the prediction of Vss, including use of in vitro data to predict Vss directly, use of in vitro and in silico methods to predict Kp, and quantitative structure activity relationships (QSAR). Some of these methods are discussed below.
Vss Prediction Methods
Volume of distribution prediction methods are important, as they allow an early understanding of drug behavior, before clinical data is obtained, in order to drive decisions in drug discovery and development. Several methods have been used to predict Vss, including (a) preclinical extrapolation [10], (b) physiological equations, (c) Kp prediction models, and (d) QSAR models [11]. All these methods attempt to incorporate physicochemical (drug) and physiological (body) determinants of distribution.
(a). Preclinical Extrapolation
Prior to the clinic, human volume of distribution values are often extrapolated from a preclinical species, often through direct correlation or allometry. Preclinical parameters can be empirically extrapolated to predict human Vss [3]. These methods are often accurate because tissue composition is generally conserved across mammalian species. Preclinical Vss is first used to predict the fraction of drug unbound in tissue (fut), and human Vss is then predicted based on the predicted fut and human unbound plasma fraction (fup) from in vitro assays. This method is based on the assumptions that partitioning into tissue lipids is relatively independent of species, and plasma protein binding may show significant deviations. While Vss is often scaled from a preclinical species, classical allometric relationships are also used as described below.
Allometric scaling relates body weight (BW) to a given physiological parameter or pharmacokinetic parameter [10]. This allows prediction of these parameters across species, based on their body weight. An allometric equation can be expressed as:
| (1) |
where Y is the parameter, a and b are the coefficients which are parameterized from data from different species, and BW is the body weight. Boxenbaum initially showed that allometry could be used for the prediction of human volume of distribution [10]. The allometric exponents vary for different PK parameters, and the exponents used for clearance and Vss are generally 0.75 and 1, respectively [12]. An exponent of 1 implies that weight normalized Vss is constant across species, which is a trivial allometric relationship. Allometric predictions of PK parameters are empirical and do not account for physiological differences across species, including but not limited to, enzymes, transporters, and plasma protein binding. Plasma protein binding of drugs can differ greatly from one species to the next [13]. Sugita et al. showed that allometry could not be used for tolbutamide, which is a highly protein-bound drug [14]. Obach et al. reported that correction for protein binding led to better allometric predictions [3]. Sawada et al. compared Vss predictions for 10 weakly basic drugs with direct extrapolation across species versus with an equation using fup and BP (a form of the Oie-Tozer equation, discussed below) [15], and reported that Vss predictions were erroneous upon simple extrapolation. Jones et al. evaluated 24 different methods for the prediction of Vss and found that generally, methods using in vivo preclinical data (when available for more than two species) were more predictive than those that relied solely on in vitro data [16].
(b). Physiological Models
Various models that use in vitro data to directly predict Vss have been proposed. The commonly used Gillette and Oie-Tozer equations, modifications thereof, and a method based on membrane partitioning published previously by our laboratory are briefly described.
In 1971, Gillette described a relationship between Vss and physiologic plasma and tissue volumes, drug protein binding, and drug tissue partitioning [17]:
| (2) |
where Vp is the volume of the plasma and VT is the volume of the tissue. This relationship was further explored mechanistically in several seminal PK discussions [18, 19]. Equation 2 is the origin of essentially all methods to predict Vss. For example, if various tissues are considered, the sum of all tissue volumes would equal VT. Also, the term fup/fut equals Kp, since unbound concentration in the plasma is assumed to equal unbound concentration in tissue. The Oie-Tozer equation [20] was based on the Gillette equation and made the distinction between drug bound to plasma proteins in the plasma versus drug bound to plasma proteins in the extracellular fluid:
| (3) |
where RE/I is the ratio of total binding sites in the extracellular fluid outside plasma to the total binding sites in plasma, VE is the extracellular fluid volume, and VR is the aqueous volume outside extracellular fluid into which drug distributes. The Oie-Tozer equation has been used in an attempt to better predict the Vss, as well as be more mechanistic [21, 22]. Lombardo used the Oie-Tozer equation to predict fut (n = 64 drugs) and Vss (n = 14 drugs) of neutral and basic drugs [21]. Of the 14 drugs in the validation set, Vss for 8 drugs was predicted within 2-fold of the experimental value. These results were expanded and corroborated in a subsequent study by the same group [22].
Membrane partitioning is an important mechanism for drug distribution and the subsequent parameterization of Vss. Using two experimental inputs—plasma protein binding (fup) and microsomal partitioning (fum) to represent membrane partitioning—Vss predictions were attempted for 63 drugs (acids, bases, and neutrals) [23••]. The resulting model had an R2 of 0.83 and 1.6-fold error for 60 drugs, with three outliers. This base model was expanded to include other factors including neutral lipid interactions as well as interactions with lysosomes (for bases). These different additions did not significantly improve Vss predictions.
(c). Kp Models
Vss can also be predicted by using Kp values [15]. Vss is determined by taking the sum of the tissue volumes multiplied by the respective drug Kp values, and adding this sum to the plasma volume. This method requires knowledge of drug Kp for different tissues in the body. Kp values can be determined in preclinical species, but this is costly and time-intensive. Early in drug discovery, it is therefore helpful to predict Kp values in order to predict Vss. These Kp values are also useful in PBPK modeling to describe tissue distribution and are used to simulate drug concentration-time profiles. Kp values are generally predicted from physicochemical properties (e.g., LogP, pKa), in vitro studies, in vivo preclinical studies, and physiological parameters including tissue composition and protein ratios. Equation 4, derived from the Gillette equation, is used:
| (4) |
where Vti is the volume of the ith tissue and Kpi is the corresponding Kp. Table 1 provides a brief comparison of various Kp models in the literature. Bjorkman used the relationship in Eq. 4 to determine which tissue Kp values were necessary in order to predict an initial Vss [26]. Distribution to adipose and muscle accounted for an average 84% of the total estimated Vss for the bases (n = 17) evaluated. For acids (n = 18), distribution to adipose and muscle accounted for only 65% of Vss. Many published Kp prediction models require an in vivo component such as a Kp,muscle term or experimental Vss [30,33–35]. Clausen and Bickel compared drug binding in blood to drug binding in tissue homogenate for in vitro determination of Kp. These predicted values generally under-predicted the experimental Kp values in rat tissue homogenates for 10 drugs [34]. Berry et al. [33] used a similar technique by using ultracentrifugation to determine the fraction unbound in tissues as well as in plasma, to determine Vss for different compounds. These values were compared to Vss determined by composition-based Kp prediction methods, as well as to experimental Vss values. Using tissue binding data, Vss was predicted within 2-fold of experimental values for 61% of all drugs studied, compared to 42 or 53% with composition-based methods (Berezhokivsiy and Rodgers and Rowland methods, respectively) within Symcyp Rat version 8.0. These in vitro binding methods had some limitations. Homogenization likely disrupted elements of the tissues which contribute to distribution, including lysosomal partitioning and transporters. Also, choice of drugs used, experimental errors, and literature inputs probably influenced the results. While these methods provide reasonable predictions, they are more labor intensive and less high-throughput than computational methods. Jansson et al. used preclinical Vss values and drug lipophilicity to predict tissue Kp for 22 drugs. The Kp values were generally well predicted for non-eliminating tissues (R2 = 0.81), with 72% of predicted values within 2-fold of experimental values. Various additional algorithms for predicting Kp have been previously reported [30, 35, 36] and have been compared in the literature [37].
Table 1.
Summary of key Kp prediction methods
| Reference | Summary of study | Comments |
|---|---|---|
| Arundel [24] |
|
|
| Jansson et al. [25] |
|
|
| Poulin et al. [27–29] |
|
|
| Poulin and Theil [30] |
|
|
| Berezkovskiy [32] |
|
|
| Rodgers and Rowland [4, 5] |
|
|
| Rodgers and Rowland [5] |
|
|
| Korzekwa and Nagar [23••] |
|
|
Tissue composition-based models attempt to predict drug distribution based on tissue composition, drug physicochemical properties, and plasma protein binding. These methods usually require simple input parameters such as LogP, fup, and BP. These methods do not rely on in vivo components but instead use in vitro surrogates to represent different distribution processes [4, 5, 27, 38]. Poulin et al. calculated Kp as a function of a lipophilicity measure, tissue-specific concentration of lipids and fraction unbound in plasma [27, 28, 39]. However, as discussed previously [40••], the lipophilicity measures used in these composition-based models rely on mechanistically unsound calculations of lipid partitioning. For example, a vegetable oil:water partition coefficient calculated with equations inaccurate for this purpose is used to describe neutral lipid partitioning. Also, neutral phospholipids are modeled as 70% water and 30% octanol. Berezhkovskiy revised the Poulin method by correcting for the ratio of unbound fraction in tissue to that in plasma [32]. Next, the Rodgers and Rowland models assumed that moderate to strong bases partition only in acidic phospholipids [4] and further developed two separate equations to predict the Kp of moderate-to-strong bases [4], and neutrals, weak bases, and acidic drugs [5]. A major assumption of the Rodgers and Rowland method for bases is that ionized bases only bind to acidic phospholipids, and neutral bases bind to neutral phospholipids. The validity of this assumption has been questioned previously [40••]. Also, the method uses BP to parameterize acidic phospholipid binding. It has been shown that erythrocyte partitioning is highly correlated with unbound volumes of distribution for bases [41]. Therefore, use of BP has two important consequences. First, when used to parameterize acidic phospholipid binding, the subsequent Vss prediction becomes insenitive to errors in fup [42]. Second, the acidic phospholipid term dominates Kp predictions, and Vss is ultimately determined by the BP value [40••].
Additionally, several QSAR models have been used to predict Kp and/or Vss in humans. Many groups have used descriptor-based models, to determine important interactions in tissues and physicochemical properties that dictate distribution [43–45]. Lombardo et al. used a mixture discriminant analysis-random forest method to predict Vss and found a 1.8-fold geometric mean fold error for an external validation set [44]. This was compared to a 2.1-fold error for animal-based predictions suggesting that this method was comparable to the much more expensive in vivo method. In another study, a mixture of experimental and computed structural descriptors was used with step-wise regression to predict volume of distribution (both Vss and Varea were used) [43]. It was found that models for unbound volumes had higher errors than total volumes (2.3-fold versus 2.0-fold, respectively). A model using genetic algorithm and stepwise regression with molecular descriptors to predict the Vss of acidic drugs resulted in an AAFE of 2.1 for an external validation set [45]. All three methods result in an approximate fold error of two for an external validation set. It should be noted that if the external validation set contains drugs from the same class as the training set, the predictability of a compound in a new chemical class is unknown. This is a general concern when descriptor-based QSAR models are used to predict non-specific ADME properties.
Factors Influencing Vss Predictions
Key factors that impact Vss prediction accuracy include permeability, transporters, and accuracy of experimental inputs. BCS Class 3 and Class 4 drugs have low permeability across membranes. Polar drugs exhibit permeability-limited distribution due to low permeability across the hydrophobic lipid bilayer. In drug discovery, compound permeability is usually measured with standard Caco-2 cell assays. However, compound permeability is often ignored in Vss predictions. For example, perfusion-limited PBPK models ignore drug permeability. Poorly permeable compounds may not reach equilibrium with unbound plasma concentrations within the exposure time of the drug. While outside the scope of this discussion, permeability is also critical to consider in the absorption of drugs via the gut. When permeability impacts the rate of drug distribution, perfusion-limited PBPK models will inaccurately predict the shape of the concentration-time profile, even though Vss may be predicted accurately [42]. Incorporation of in vitro permeability data into simple compartmental models with explicit membranes has been previously reported [46]. Models with explicit membranes can be used to improve predictions of Vss as well as concentration-time profiles (i.e., extent and rate of drug distribution). Although currently unavailable, full perfusion- and permeability-limited models may ultimately predict the rate and extent of distribution.
A factor closely related to drug permeability is the impact of transporters. Poorly permeable drugs are often substrates for uptake transporters. The family of organic anion transporter proteins (OATPs) has emerged as a key uptake transporter protein family expressed in the liver, with clinical implications for drug pharmacokinetics and drug-drug interactions [9, 47•]. Numerous reports have been published on active uptake of drugs such as statins by OATPs into the liver, and the impact of this uptake on drug clearance as well as drug-drug interactions [48]. Our laboratory evaluated the active hepatic uptake of atorvastatin in a rat model, and it was observed that active uptake significantly increased the total atorvastatin concentration in the rat liver [49•]. OATP-mediated transport was predicted to alter hepatic intracellular concentrations by two orders of magnitude [49•]. Therefore, uptake into the liver alone can result in a 2-fold increase in Vss. Efflux transporters such as P-glycoprotein significantly impact distribution of drugs in the brain [8]. Incorporation of transporter kinetics into Vss predictions will be important in improving predictions. With current Vss prediction models, the Vss of acids is usually under-predicted. One reason for this underprediction could be the lack of inclusion of transporters in Vss models.
As is true for all modeling efforts, accuracy of experimental inputs is critical for accurate predictive outputs. In the context of Vss prediction models, experimental data such as plasma protein binding measurements are one example where inaccuracies have been observed. The pH of plasma samples can greatly impact the plasma protein binding assay and the subsequent calculation of fup [50••]. Kochansky et al. reported that for 40% of the 55 drugs tested, the ratio of fup at pH 7.4 (assay conducted under 10% CO2) vs. pH 8.7 (assay conducted in air) was ≥ 2.0 [51]. Another experimental input, the blood/plasma ratio, is less prone to experimental errors. Similarly, equilibrium dialysis assays are standard procedure for determination of fum. In the authors’ experience, experimental values of LogP are preferred over values calculated with in silico methods. A bigger concern with the use of LogP is that octanol:water partitioning, although a general measure of hydrophobicity, does not correspond to the physiological process of drug distribution [40••].
Finally, errors in in vivo pharmacokinetic data can result from experimental design or data analysis. For animal studies, if data collection or bioanalytical assay sensitivity is insufficient, the resulting calculated Vss (used as an “experimenta” Vss value) may be under-predicted. Also, use of Varea instead of Vss overpredicts Vss and may not translate to the human pharmacokinetic parameters. Use of preclinical pharmacokinetics to predict human drug disposition may be subject to several species differences. With respect to distribution, plasma protein binding and transporter protein homology, substrate specificity, binding kinetics, and genetic polymorphisms are all known to differ across species. These factors may result in significant errors in Vss prediction.
Comparison of Different Prediction Methods
Many groups have reported comparative studies on various Vss prediction methods [3, 16, 30, 31, 52••, 53–55]. Graham et al. compared six different Kp prediction methods for their prediction of unbound Kp (Kpu) as well as 4 Vss prediction models, for 81 compounds in 11 rat tissues [31]. The Rodgers and Rowland method [4, 5] provided the best prediction of Kpu (77% within 3-fold of experimental values). The Poulin and Theil method [27, 28] was able to predict the Vss best (87% within 3-fold error). Jones et al. [16] compared 24 methods for Vss prediction, and the maximum success rate (prediction within 2-fold) was 78% across methods. A recent comparison by Chan et al. compared two Vss prediction methods for a large data set (over 150 marketed drugs) [52••]. The methods included preclinical extrapolation and in silico prediction based on the Rodgers and Rowland equations. Use of composition-based Kp prediction methods for determination of the Vss was more accurate than preclinical extrapolation, with Vss of 65.8% (out of 152 drugs) predicted within 2-fold.
Conclusions
Extrapolation directly from preclinical species, physiological models, composition-based Kp prediction models, and QSAR methods can all be used to predict the Vss of a drug. Overall, the accuracy of all current methods in general is a 2-fold absolute average fold error. As with all modeling exercises, there is a trade-off between the quality of inputs and the accuracy of the predictions. For example, any model with experimental inputs (e.g., fup, fum, LogP) will result in better predictions than a model using calculated values. Currently, the use of composition-based PBPK models is preferred to models requiring in vivo input. Perfusion-limited PBPK models are commonly used, but we anticipate that membrane-based models that include permeability-limited distribution will be used more frequently. QSAR methods are an active area of research, but careful selection of training and validation sets is required to prevent over-fitting. A better mechanistic understanding of important drug distribution processes will result in improvements in all modeling approaches.
Funding Information
The authors acknowledge funding from the National Institutes of Health grants (R01GM104178 and R01GM114369).
Footnotes
Conflict of Interest The authors have no conflicts of interest.
Human and Animal Rights and Informed Consent This article does not contain any studies with human or animal subjects performed by any of the authors.
Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
Papers of particular interest, published recently, have been highlighted as:
• Of importance
•• Of major importance
- 1.Rowland M, Tozer T. Clinical Pharmacokinetics and Pharmacodynamics: Concepts and Applications. Fourth ed. 2011. [Google Scholar]
- 2.Gabrielsson J, Weiner D. Pharmacokinetic and Pharmacodynamic Data Analysis: Concepts and Applications. Fourth ed. 2010. [Google Scholar]
- 3.Obach RS, Baxter JG, Liston TE, Silber BM, Jones BC, MacIntyre F, et al. The prediction of human pharmacokinetic parameters from preclinical and in vitro metabolism data. J Pharmacol Exp Ther. 1997;283(1):46–58. [PubMed] [Google Scholar]
- 4.Rodgers T, Leahy D, Rowland M. Physiologically based pharmacokinetic modeling 1: predicting the tissue distribution of moderate-to-strong bases. J Pharm Sci. 2005;94(6):1259–76. 10.1002/jps.20322. [DOI] [PubMed] [Google Scholar]
- 5.Rodgers T, Rowland M. Physiologically based pharmacokinetic modelling 2: predicting the tissue distribution of acids, very weak bases, neutrals and zwitterions. J Pharm Sci. 2006;95(6):1238–57. 10.1002/jps.20502. [DOI] [PubMed] [Google Scholar]
- 6.Hardman JG, Limbird LE. Goodman and Gilman’s the pharmacological basis of therapeutics 10th edition. New York: McGraw-Hill; 2001. [Google Scholar]
- 7.Peters SA. Physiologically-based pharmacokinetic modeling and simulations. Hoboken, NJ: Wiley; 2012. [Google Scholar]
- 8.Cole S, Bagal S, El-Kattan A, Fenner K, Hay T, Kempshall S, et al. Full efficacy with no CNS side-effects: unachievable panacea or reality? DMPK considerations in design of drugs with limited brain penetration. Xenobiotica. 2012;42(1):11–27. 10.3109/00498254.2011.617847. [DOI] [PubMed] [Google Scholar]
- 9.Shitara Y, Maeda K, Ikejiri K, Yoshida K, Horie T, Sugiyama Y. Clinical significance of organic anion transporting polypeptides (OATPs) in drug disposition: their roles in hepatic clearance and intestinal absorption. Biopharm Drug Dispos. 2013;34(1):45–78. 10.1002/bdd.1823. [DOI] [PubMed] [Google Scholar]
- 10.Boxenbaum H. Interspecies scaling, allometry, physiological time, and the ground plan of pharmacokinetics. J Pharmacokinet Biopharm. 1982;10(2):201–27. 10.1007/BF01062336. [DOI] [PubMed] [Google Scholar]
- 11.Freitas AA, Limbu K, Ghafourian T. Predicting volume of distribution with decision tree-based regression methods using predicted tissue:plasma partition coefficients. J Cheminformatics. 2015;7: 17. 10.1186/s13321-015-0054-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mahmood I Theoretical versus empirical allometry: facts behind theories and application to pharmacokinetics. J Pharm Sci. 2010;99(7):2927–33. 10.1002/jps.22073. [DOI] [PubMed] [Google Scholar]
- 13.Colclough N, Ruston L, Wood JM, MacFaul PA. Species differences in drug plasma protein binding. Med Chem Commun. 2014;5:963–7. [Google Scholar]
- 14.Sugita O, Sawada Y, Sugiyama Y, Hanano M, Iga T. Effect of sulfaphenazole on tolbutamide distribution in rabbits - analysis of interspecies differences in tissue distribution of tolbutamide. J Pharm Sci. 1984;73(5):631–4. 10.1002/jps.2600730513. [DOI] [PubMed] [Google Scholar]
- 15.Sawada Y, Hanano M, Sugiyama Y, Harashima H, Iga T. Prediction of the volumes of distribution of basic drugs in humans based on data from animals. J Pharmacokinet Biopharm. 1984;12(6):587–96. 10.1007/bf01059554. [DOI] [PubMed] [Google Scholar]
- 16.Jones R, Jones HM, Rowland M, Gibson CR, Yates JWT, Chien JY, et al. PhRMA CPCDC initiative on predictive models of human pharmacokinetics, part 2: comparative assessment of prediction methods of human volume of distribution. J Pharm Sci. 2011;100(10):4074–89. 10.1002/jps.22553. [DOI] [PubMed] [Google Scholar]
- 17.Gillette JR. Factors affecting drug metabolism. Ann N YAcad Sci. 1971;179:43–66. [DOI] [PubMed] [Google Scholar]
- 18.Gibaldi M, McNamara PJ. Apparent volumes of distribution and drug binding to plasma proteins and tissues. Eur J Clin Pharmacol. 1978;13(5):373–80. [DOI] [PubMed] [Google Scholar]
- 19.Wilkinson GR, Shand DG. Commentary: a physiological approach to hepatic drug clearance. Clin Pharmacol Ther. 1975;18(4):377–90. [DOI] [PubMed] [Google Scholar]
- 20.Oie S, Tozer TN. Effect of altered plasma-protein binding on apparent volume of distribution. J Pharm Sci. 1979;68(9):1203–5. 10.1002/jps.2600680948. [DOI] [PubMed] [Google Scholar]
- 21.Lombardo F, Obach RS, Shalaeva MY, Gao F. Prediction of volume of distribution values in humans for neutral and basic drugs using physicochemical measurements and plasma protein binding data. J Med Chem. 2002;45(13):2867–76. 10.1021/jm0200409. [DOI] [PubMed] [Google Scholar]
- 22.Lombardo F, Obach RS, Shalaeva MY, Gao F. Prediction of human volume of distribution values for neutral and basic drugs. 2. Extended data set and leave-class-out statistics. J Med Chem. 2004;47(5):1242–50. 10.1021/jm030408h. [DOI] [PubMed] [Google Scholar]
- 23.••.Korzekwa K, Nagar S. Drug Distribution Part 2. Predicting volume of distribution from plasma protein binding and membrane partitioning. Pharm Res. 2017;34(3):544–51. 10.1007/s11095-016-2086-y [DOI] [PMC free article] [PubMed] [Google Scholar]; This article describes a new method for the prediction of the Vss ,which utilizes partitioning into microsomes to represent phospholipid partitioning in a physiological-based Vss equation. This study also looked at other tissue interactions which may be important for describing the distribution of a drug.
- 24.Arundel P. A multi-compartmental model generally applicable to physiologically-based pharmacokinetics. IFAC Proceedings Volumes. 1997;30(2):129–33. 10.1016/S1474-6670(17)44557-5. [DOI] [Google Scholar]
- 25.Jansson R, Bredberg U, Ashton M. Prediction of drug tissue to plasma concentration ratios using a measured volume of distribution in combination with lipophilicity. J Pharm Sci. 2008;97(6): 2324–39. 10.1002/jps.21130. [DOI] [PubMed] [Google Scholar]
- 26.Bjorkman S Prediction of the volume of distribution of a drug: which tissue-plasma partition coefficients are needed? J Pharm Pharmacol. 2002;54(9):1237–45. 10.1211/002235702320402080. [DOI] [PubMed] [Google Scholar]
- 27.Poulin P, Theil F-P. A priori prediction of tissue:plasma partition coefficients of drugs to facilitate the use of physiologically-based pharmacokinetic models in drug discovery. J Pharm Sci. 2000;89(1):16–35. . [DOI] [PubMed] [Google Scholar]
- 28.Poulin P, Schoenlein K, Theil FP. Prediction of adipose tissue: plasma partition coefficients for structurally unrelated drugs. J Pharm Sci. 2001;90(4):436–47. . [DOI] [PubMed] [Google Scholar]
- 29.Poulin P, Krishnan K. A biologically-based algorithm for predicting human tissue-blood partition coefficients of organic chemicals. Hum Exp Toxicol. 1995;14(3):273–80. 10.1177/096032719501400307. [DOI] [PubMed] [Google Scholar]
- 30.Poulin P, Theil F-P. Development of a novel method for predicting human volume of distribution at steady-state of basic drugs and comparative assessment with existing methods. J Pharm Sci. 2009;98(12):4941–61. 10.1002/jps.21759. [DOI] [PubMed] [Google Scholar]
- 31.Graham H, Walker M, Jones O, Yates J, Galetin A, Aarons L. Comparison of in-vivo and in-silico methods used for prediction of tissue: plasma partition coefficients in rat. J Pharm Pharmacol. 2012;64(3):383–96. 10.1111/j.2042-7158.2011.01429.x. [DOI] [PubMed] [Google Scholar]
- 32.Berezhkovskiy LM. Volume of distribution at steady state for a linear pharmacokinetic system with peripheral elimination. J Pharm Sci. 2004;93(6):1628–40. 10.1002/jps.20073. [DOI] [PubMed] [Google Scholar]
- 33.Berry LM, Roberts J, Be X, Zhao Z, Lin MHJ. Prediction of Vss from in vitro tissue-binding studies. Drug Metab Dispos. 2010;38(1):115–21. 10.1124/dmd.109.029629. [DOI] [PubMed] [Google Scholar]
- 34.Clausen J, Bickel MH. Prediction of drug distribution in distribution dialysis and in vivo from binding to tissues and blood. J Pharm Sci. 1993;82(4):345–9. 10.1002/jps.2600820402. [DOI] [PubMed] [Google Scholar]
- 35.Poulin P, Ekins S, Theil F-P. A hybrid approach to advancing quantitative prediction of tissue distribution of basic drugs in human. Toxicol Appl Pharmacol. 2011;250(2):194–212. 10.1016/j.taap.2010.10.014. [DOI] [PubMed] [Google Scholar]
- 36.Yun YE, Edginton AN. Correlation-based prediction of tissue-to-plasma partition coefficients using readily available input parameters. Xenobiotica. 2013;43(10):839–52. 10.3109/00498254.2013.770182. [DOI] [PubMed] [Google Scholar]
- 37.Yun YE, Cotton CA, Edginton AN. Development of a decision tree to classify the most accurate tissue-specific tissue to plasma partition coefficient algorithm for a given compound. J Pharmacokinet Pharmacodyn. 2014;41(1):1–14. 10.1007/s10928-013-9342-0. [DOI] [PubMed] [Google Scholar]
- 38.Schmitt W. General approach for the calculation of tissue to plasma partition coefficients. Toxicol In Vitro. 2008;22(2):457–67. 10.1016/j.tiv.2007.09.010. [DOI] [PubMed] [Google Scholar]
- 39.Poulin P, Theil FP. Prediction of pharmacokinetics prior to in vivo studies. 1. Mechanism-based prediction of volume of distribution. J Pharm Sci. 2002;91(1):129–56. 10.1002/jps.10005. [DOI] [PubMed] [Google Scholar]
- 40.••.Korzekwa K, Nagar S. On the nature of physiologically-based pharmacokinetic models –a priori or a posteriori? Mechanistic or empirical? Pharm Res. 2017;34(3):529–34. 10.1007/s11095-016-2089-8 [DOI] [PMC free article] [PubMed] [Google Scholar]; This article provides a commentary on the current assumptions and methods used in physiologically-based pharmacokinetic models.
- 41.Hinderling PH. Red blood cells: a neglected compartment in pharmacokinetics and pharmacodynamics. Pharmacol Rev. 1997;49(3): 279–95. [PubMed] [Google Scholar]
- 42.Ye M, Nagar S, Korzekwa K. A physiologically based pharmacokinetic model to predict the pharmacokinetics of highly protein-bound drugs and the impact of errors in plasma protein binding. Biopharm Drug Dispos. 2016;37(3):123–41. 10.1002/bdd.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ghafourian T, Barzegar-Jalali M, Hakimiha N, Cronin MTD. Quantitative structure-pharmacokinetic relationship modelling: apparent volume of distribution. J Pharm Pharmacol. 2004;56(3): 339–50. 10.1211/0022357022890. [DOI] [PubMed] [Google Scholar]
- 44.Lombardo F, Obach RS, DiCapua FM, Bakken GA, Lu J, Potter DM, et al. Hybrid mixture discriminant analysis-random forest computational model for the prediction of volume of distribution of drugs in human. J Med Chem. 2006;49(7):2262–7. 10.1021/jm050200r. [DOI] [PubMed] [Google Scholar]
- 45.Zhivkova Z, Doytchinova I. Prediction of steady-state volume of distribution of acidic drugs by quantitative structure-pharmacokinetics relationships. J Pharm Sci. 2012;101(3):1253–66. 10.1002/jps.22819. [DOI] [PubMed] [Google Scholar]
- 46.Korzekwa KR, Nagar S, Tucker J, Weiskircher EA, Bhoopathy S, Hidalgo IJ. Models to predict unbound intracellular drug concentrations in the presence of transporters. Drug Metab Dispos. 2012;40(5):865–76. 10.1124/dmd.111.044289. [DOI] [PubMed] [Google Scholar]
- 47.•.Kovacsics D, Patik I, Özvegy-Laczka C. The role of organic anion transporting polypeptides in drug absorption, distribution, excretion and drug-drug interactions. Expert Opin Drug Metab Toxicol. 2017;13(4):409–24. 10.1080/17425255.2017.1253679 [DOI] [PubMed] [Google Scholar]; This article is a current review discussing the OATP family of transporters and the importance of OATPs in the absorption and distribution of drugs, as well as their role in drug-drug interactions.
- 48.Maeda K. Organic anion transporting polypeptide (OATP)1B1 and OATP1B3 as important regulators of the pharmacokinetics of substrate drugs. Biol Pharm Bull. 2015;38(2):155–68. 10.1248/bpb.b14-00767. [DOI] [PubMed] [Google Scholar]
- 49.•.Kulkarni P, Korzekwa K, Nagar S. Intracellular unbound atorvastatin concentrations in the presence of metabolism and transport. J Pharmacol Exp Ther. 2016;359(1):26–36. 10.1124/jpet.116.235689 [DOI] [PMC free article] [PubMed] [Google Scholar]; This article used a 5-compartmental model for the prediction of intracellular concentrations of atorvastation, to understand the influence of transporters on the intracellular concentration.
- 50.••.Di L, Breen C, Chambers R, Eckley ST, Fricke R, Ghosh A, et al. Industry perspective on contemporary protein-binding methodologies: considerations for regulatory drug-drug interaction and related guidelines on highly bound drugs. J Pharm Sci. 2017;106(12): 3442–52. 10.1016/j.xphs.2017.09.005 [DOI] [PubMed] [Google Scholar]; This article offers an industry perspective on the current methods used to determine the plasma protein binding of a drug, as well as factors which should be considered in current methodology.
- 51.Kochansky CJ, McMasters DR, Lu P, Koeplinger KA, Kerr HH, Shou M, et al. Impact of pH on plasma protein binding in equilibrium dialysis. Mol Pharm. 2008;5(3):438–48. 10.1021/mp800004s. [DOI] [PubMed] [Google Scholar]
- 52.••.Chan R, De Bruyn T, Wright M, Broccatelli F. Comparing mechanistic and preclinical predictions of volume of distribution on a large set of drugs. Pharm Res. 2018;35(4):11. 10.1007/s11095-018-2360-2 [DOI] [PubMed] [Google Scholar]; This article compared the use of composition-based tissue: plasma partition coefficient prediction models, as well as preclinical extrapolation for the prediction of the Vss for a set of 152 drugs.
- 53.Zou P, Zheng N, Yang YS, Yu LX, Sun DX. Prediction of volume of distribution at steady state in humans: comparison of different approaches. Expert Opin Drug Metab Toxicol. 2012;8(7):855–72. 10.1517/17425255.2012.682569. [DOI] [PubMed] [Google Scholar]
- 54.Sui XF, Sun J, Li HY, Wang YJ, Liu JF, Liu XH, et al. Prediction of volume of distribution values in human using immobilized artificial membrane partitioning coefficients, the fraction of compound ionized and plasma protein binding data. Eur J Med Chem. 2009;44(11):4455–60. 10.1016/j.ejmech.2009.06.004. [DOI] [PubMed] [Google Scholar]
- 55.De Buck SS, Sinha VK, Fenu LA, Gilissen RA, Mackie CE, Nijsen MJ. The prediction of drug metabolism, tissue distribution, and bioavailability of 50 structurally diverse compounds in rat using mechanism-based absorption, distribution, and metabolism prediction tools. Drug Metab Dispos. 2007;35(4):649–59. 10.1124/dmd.106.014027. [DOI] [PubMed] [Google Scholar]
