++
Drugs are carried by blood flow from the administration (input) site to various body organs, where the drug rapidly equilibrates with the interstitial water in the organ. Physiologic pharmacokinetic models are mathematical models describing drug movement and disposition in the body based on organ blood flow and the organ spaces penetrated by the drug. In its simplest form, a physiologic pharmacokinetic model considers the drug to be blood flow limited. Drugs are carried to organs by arterial blood and leave organs by venous blood (Fig. 22-2). In such a model, transmembrane movement of drug is rapid, and the capillary membrane does not offer any resistance to drug permeation. Uptake of drug into the tissues is rapid, and a constant ratio of drug concentrations between the organ and the venous blood is quickly established. This ratio is the tissue/blood partition coefficient:
++
where P is the partition coefficient.
++
The magnitude of the partition coefficient can vary depending on the drug and on the type of tissue. Adipose tissue, for example, has a high partition for lipophilic drugs. The rate of drug carried to a tissue organ and tissue drug uptake depend on the rate of blood flow to the organ and the tissue/blood partition coefficient, respectively.
++
The rate of blood flow to the tissue is expressed as Qt (mL/min), and the rate of change in the drug concentration with respect to time within a given tissue organ is expressed as
++
where Cart is the arterial blood drug concentration and Cven is the venous blood drug concentration. Qt is blood flow and represents the volume of blood flowing through a typical tissue organ per unit of time.
++
If drug uptake occurs in the tissue, the incoming concentration, Cart, is higher than the outgoing venous concentration, Cven. The rate of change in the tissue drug concentration is equal to the rate of blood flow multiplied by the difference between the blood drug concentrations entering and leaving the tissue organ. In the blood flow–limited model, drug concentration in the blood leaving the tissue and the drug concentration within the tissue are in equilibrium, and Cven may be estimated from the tissue/blood partition coefficient in Equation 22.1. Substituting in Equation 22.3 with Cven = Ctissue/Ptissue yields
++
Equation 22.4 describes drug distribution in a noneliminating organ or tissue group. For example, drug distribution to muscle, adipose tissue, and skin is represented in a similar manner by Equations 22.5, 22.6, and 22.7, respectively, as shown below. For tissue organs in which drug is eliminated (Fig. 22-3), parameters representing drug elimination from the liver (kLIV) and kidney (kKID) are added to account for drug removal through metabolism or excretion. Equations 22.8 and 22.9 are derived similarly to those for the noneliminating organs above.
++
Removal of drug from any organ is described by drug clearance (Cl) from the organ. The rate of drug elimination is the product of the drug concentration in the organ and the organ clearance.
++
The rate of drug elimination may be described for each organ or tissue (Fig. 22-4).
++
where LIV = liver, SP = spleen, GI = gastrointestinal tract, KID = kidney, LU = lung, FAT = adipose, SKIN = skin, and MUS = muscle.
++
The mass balance for the rate of change in drug concentration in the blood pool is
++
Lung perfusion is unique because the pulmonary artery returns venous blood flow to the lung, where carbon dioxide is exchanged for oxygen and the blood becomes oxygenated. The blood from the lungs flows back to the heart (into the left atrium) through the pulmonary vein, and the quantity of blood that perfuses the pulmonary system ultimately passes through the remainder of the body. In describing drug clearance through the lung, perfusion from the heart (right ventricle) to the lung is considered venous blood (Fig. 22-4). Therefore, the terms in Equation 22.11 describing lung perfusion are reversed compared to those for the perfusion of other tissues. With some drugs, the lung is a clearing organ besides serving as a merging pool for venous blood. In those cases, a lung clearance term could be included in the general model.
++
After intravenous drug administration, drug uptake in the lungs may be very significant if the drug has high affinity for lung tissue. If actual drug clearance is at a much higher rate than the drug clearance accounted for by renal and hepatic clearance, then lung clearance of the drug should be suspected, and a lung clearance term should be included in the equation in addition to lung tissue distribution.
++
The system of differential equations used to describe the blood flow–limited model is usually solved through computer programs. The Runge–Kutta method is often used in computer methods for using a series of differential equations. Because of the large number of parameters involved in the mass balance, more than one set of parameters may fit the experimental data. This is especially true with human data, in which many of the organ tissue data items are not available. The lack of sufficient tissue data sometimes leads to unconstrained models. As additional data become available, new or refined models are adopted. For example, methotrexate was initially described by a flow-limited model, but later work described the model as a diffusion-limited model.
++
Because invasive methods are available for animals, tissue/blood ratios or partition coefficients can be determined accurately by direct measurement. Using experimental pharmacokinetic data from animals, physiologic pharmacokinetic models may yield more reliable predictions.
+++
Physiologic Pharmacokinetic Model with Binding
++
The physiologic pharmacokinetic model assumes flow-limited drug distribution without drug binding to either plasma or tissues. In reality, many drugs are bound to a variable extent in either plasma or tissues. With most physiologic models, drug binding is assumed to be linear (not saturable or concentration dependent). Moreover, bound and free drug in both tissue and plasma are in equilibrium. Further, the free drug in the plasma and in the tissue equilibrates rapidly. Therefore, the free drug concentration in the tissue and the free drug concentration in the emerging blood are equal:
++
++
++
++
where fb is the blood free drug fraction, ft is the tissue free drug fraction, Ct is the total drug concentration in tissue, and Cb is the total drug concentration in blood.
++
Therefore, the partition ratio, Pt, of the tissue drug concentration to that of the plasma drug concentration is
++
++
By assuming linear drug binding and rapid drug equilibration, the free drug fraction in tissue and blood may be incorporated into the partition ratio and the differential equations. These equations are similar to those above except that free drug concentrations are substituted for Cb. Drug clearance in the liver is assumed to occur only with the free drug. The inherent capacity for drug metabolism (and elimination) is described by the term Clint (see Chapter 11). General mass balance of various tissues is described by Equation 22.16:
++
++
++
++
++
++
++
++
The mass balance for the drug in the blood pool is
++
++
++
The influence of binding on drug distribution is an important factor in interspecies differences in pharmacokinetics. In some instances, animal data may predict drug distribution in humans by taking into account the differences in drug binding. For the most part, extrapolations from animals to humans or between species are rough estimates only, and there are many instances in which species differences are not entirely attributable to drug binding and metabolism.
+++
Blood Flow-Limited versus Diffusion-Limited Model
++
Most physiologic pharmacokinetic models assume rapid drug distribution between tissue and venous blood. Rapid drug equilibrium assumes that drug diffusion is extremely fast and that the cell membrane offers no barrier to drug permeation. If no drug binding is involved, the tissue drug concentration is the same as that of the venous blood leaving the tissue. This assumption greatly simplifies the mathematics involved. Table 22-1 lists some of the drugs that have been described by a flow-limited model. This model is also referred to as the perfusion model. A more complex type of physiologic pharmacokinetic model is called the diffusion-limited model or the membrane-limited model. In the diffusion-limited model, the cell membrane acts as a barrier for the drug, which gradually permeates by diffusion. Because blood flow is very rapid and drug permeation is slow, a drug concentration gradient is established between the tissue and the venous blood (Lutz and Dedrick, 1985). The rate-limiting step of drug diffusion into the tissue depends on the permeation across the cell membrane rather than blood flow. Because of the time lag in equilibration between blood and tissue, the pharmacokinetic equation for the diffusion-limited model is very complicated.
++
+++
Application and Limitations of Physiologic Pharmacokinetic Models
++
The physiologic pharmacokinetic model is related to drug concentration and tissue distribution using physiologic and anatomic information. For example, the effect of a change in blood flow on the drug concentration in a given tissue may be estimated once the model is characterized. Similarly, the effect of a change in mass size of different tissue organs on the redistribution of drug may also be evaluated using the system of physiologic model differential equations generated. When several species are involved, the physiologic model may predict the pharmacokinetics of a drug in humans when only animal data are available. Changes in drug–protein binding, tissue organ drug partition ratios, and intrinsic hepatic clearance may be inserted into the physiologic pharmacokinetic model.
++
Most pharmacokinetic studies are modeled based on blood samples drawn from various venous sites after either IV or oral dosing. Physiologists have long recognized the unique difference between arterial and venous blood. For example, arterial tension (pressure) of oxygen drives the distribution of oxygen to vital organs. Chiou (1989) and Mather (2001) have discussed the pharmacokinetic issues when differences in drug concentrations in arterial and venous are considered (see Chapter 10). The implication of venous versus arterial sampling is hard to estimate and may be more drug dependent. Most pharmacokinetic models are based on sampling of venous data. In theory, mixing occurs quickly when venous blood returns to the heart and becomes reoxygenated again in the lung. Chiou (1989) has estimated that for drugs that are highly extracted, the discrepancies may be substantial between actual concentration and concentration estimated from well-stirred pharmacokinetic models.
++
Various approaches have been used to compare the toxicity and pharmacokinetics of a drug among different species. Interspecies scaling is a method used in toxicokinetics and the extrapolation of therapeutic drug doses in humans from nonclinical animal drug studies. Toxicokinetics is the application of pharmacokinetics to toxicology and pharmacokinetics for interpolation and extrapolation based on anatomic, physiologic, and biochemical similarities (Mordenti and Chappell, 1989; Bonate and Howard 2000; Mahmood, 2000, 2007; Hu and Hayton, 2001; Evans et al, 2006).
++
The basic assumption in interspecies scaling is that physiologic variables, such as clearance, heart rate, organ weight, and biochemical processes, are related to the weight or body surface area of the animal species (including humans). It is commonly assumed that all mammals use the same energy source (oxygen) and energy transport systems across animal species (Hu and Hayton, 2001). Interspecies scaling uses a physiologic variable, y, that is graphed against the body weight of the species on log–log axes to transform the data into a linear relationship (Fig. 22-5). The general allometric equation obtained by this method is
++
++
where y is the pharmacokinetic or physiologic property of interest, b is an allometric coefficient, W is the weight or surface area of the animal species, and a is the allometric exponent. Allometry is the study of size.
++
++
Both a and b vary with the drug. Examples of various pharmacokinetic or physiologic properties that demonstrate allometric relationships are listed in Table 22-2. In the example shown in Fig. 22-5, methotrexatevolume of distribution is related to body weight B of five animal species by the equation Vβ = 0.859B0.918.
++
++
The allometric method gives an empirical relationship that allows for approximate interspecies scaling based on the size of the species. Not considered in the method are certain specific interspecies differences such as gender, nutrition, pathophysiology, route of drug administration, and polymorphisms. Some of these more specific cases, such as the pathophysiologic condition of the animal or human, may preclude pharmacokinetic or allometric predictions.
++
Interspecies scaling has been refined by considering the aging rate and life span of the species. In terms of physiologic time, each species has a characteristic life span, its maximum life-span potential (MLP), which is controlled genetically (Boxenbaum, 1982). Because many energy-consuming biochemical processes, including drug metabolism, vary inversely with the aging rate or life span of the animal, the allometric approach has been used for drugs that are eliminated mainly by hepatic intrinsic clearance.
++
Through the study of various species in handling several drugs that are metabolized predominantly by the liver, some empirical relationships regarding drug clearance of several drugs have been related mathematically in a single equation. For example, drug hepatic intrinsic clearance of biperiden in rat, rabbit, and dog was extrapolated to humans (Nakashima et al, 1987). Equation 22.20 describes the relationship between biperiden intrinsic clearance with body weight and MLP:
++
++
where MLP is the maximum life-span potential of the species, B is the body weight of the species, and Clint is the hepatic intrinsic clearance of the free drug.
++
Although further model improvements are needed before accurate prediction of pharmacokinetic parameters can be made from animal data, some interesting results were obtained by Sawada et al (1985) on nine acid and six basic drugs. When interspecies differences in protein–drug binding are properly considered, the volume of distribution of many drugs may be predicted with 50% deviation from experimental values (Table 22-3).
++
++
The application of MLP to pharmacokinetics has been described by Boxenbaum (1982). Initially, hepatic intrinsic clearance was considered to be related to volume or body weight. Indeed, a plot of the log drug clearance versus body weight for various animal species resulted in an approximately linear correlation (ie, a straight line). However, after correcting intrinsic clearance by MLP, an improved log–linear relationship was achieved between free drug Clint and body weight for many drugs. A possible explanation for this relationship is that the biochemical processes, including Clint, in each animal species are related to the animal's normal life expectancy (estimated by MLP) through the evolutionary process. Animals with a shorter MLP have higher basal metabolic rates and tend to have higher intrinsic hepatic clearance and thus metabolize drugs faster. Boxenbaum (1982, 1983) postulated a constant “life stuff” in each species, such that the faster the life stuff is consumed, the more quickly the life stuff is used up. In the fourth-dimension scale (after correcting for MLP), all species share the same intrinsic clearance for the free drug.
++
++
++
Extensive work with caffeine in five species (mouse, rat, rabbit, monkey, and humans) by Bonati and associates (1985) verified this approach. Caffeine is a drug that is metabolized predominantly by the liver. For caffeine,
++
++
++
where B is body weight, L is liver weight, and Q is blood flow.
++
Hepatic clearance for the unbound drug did not show a direct correlation among the five species. After intrinsic clearance was corrected for MLP (calculation based on brain weight), an excellent relationship was obtained among the five species (Fig. 22-6).
++
++
More recently, the subject of interspecies scaling was investigated using Cl values for 91 substances for several species by Hu and Hayton (2001). These investigators used Y = a (BW)b in their analysis, similar to Equation 22.19 above but with different symbols: Y = biological variable dependent on the body weight of the species, a = allometric coefficient, b = allometric exponent, and BW = body weight of the species. One issue discussed by Hu and Hayton is the uncertainty in the allometric exponent (b) of xenobiotic clearance (CL). Published literature has focused on whether the basal metabolic rate scale is a 2/3 or 3/4 power of the body mass (BW). When the uncertainty in the determination of a b value is relatively large, a fixed-exponent approach might be feasible according to Hu and Hayton. In this regard, 0.75 might be used for substances that are eliminated mainly by metabolism or by metabolism and excretion combined, whereas 0.67 might apply for drugs that are eliminated mainly by renal excretion. The researchers pointed out that genetic (intersubject) difference may be a limitation for using a single universal constant.
++
Brightman et al (2006) demonstrated the application of a PK-PD model, based on human parameters to estimate plasma pharmacokinetics of xenobiotics in humans. The model was parameterized through an optimization process, using a training set of in vivo data taken from the literature. On average, the vertical divergence of the predicted plasma concentrations from the observed data was 0.47 log units, on a semilog concentration-time plot. They also evaluated the method against other predictive methods that involve scaling from in vivo animal data. In terms of predicting human clearance for the test set, the model was found to match or exceed the performance of three published interspecies scaling methods, which tend to give overprediction. The article concludes that the generic physiologically based pharmacokinetic model is a means of integrating readily determined in vitro and/or in silico data, and useful for predicting human xenobiotic kinetics in drug discovery.
+++
Physiologic versus Compartment Approach
++
Compartmental models represent a simplified kinetic approach to describe drug absorption, distribution, and elimination (see Chapters 3 and 4). The major advantage of compartment models is that the time course of drug in the body may be monitored quantitatively with a limited amount of data. Generally, only plasma drug concentrations and limited urinary drug excretion data are available. Compartmental models have been applied successfully to prediction of the pharmacokinetics of the drug and the development of dosage regimens. Moreover, compartmental models are very useful in relating plasma drug levels to pharmacodynamic and toxic effects in the body.
++
The simplicity and flexibility of the compartment model is the principal reason for its wide application. For many applications, the compartmental model may be used to extract some information about the underlying physiologic mechanism through model testing of the data. Thus, compartment analysis may lead to a more accurate description of the underlying physiologic processes and the kinetics involved. In this regard, compartmental models are sometimes misunderstood, overstretched, and even abused. For example, the tissue drug levels predicted by a compartment model represent only a composite pool for drug equilibration between all tissue and the circulatory system (plasma compartment). However, extrapolation to a specific tissue drug concentration is inaccurate and analogous to making predictions without experimental data. Although specific tissue drug concentration data are missing, many investigators may make general predictions about average tissue drug levels.
++
Compartment models account accurately for the mass balance of the drug in the body and the amount of drug eliminated. Mass balance includes the drug in the plasma, the drug in the tissue pool, and the amount of drug eliminated after dosage administration. The compartment model is particularly useful for comparing the pharmacokinetics of related therapeutic agents. In the clinical pharmacokinetic literature, drug data comparisons are based on compartment models. Though alternative pharmacokinetic models have been available for approximately 20 years, the simplicity of the compartment model allows easy tabulation of parameters such as VDSS, alpha t1/2, and beta t1/2. The alternative pharmacokinetic models, including the physiologic and statistical moment (mean residence time) approaches, are used much less frequently, even though a substantial body of data has been generated using both of these models.
++
In spite of these advantages, the compartmental model is generally regarded as somewhat empirical and lacking physiologic relevance. Many disease-related changes in pharmacokinetics are the result of physiologic changes, such as impairment of blood flow or a change in organ mass. These pathophysiologic changes are better evaluated using a physiologic-based pharmacokinetic model.
++
Because of its simplicity, the compartment model often serves as a “first model” that requires further refinement in order to describe the physiologic and drug distribution processes in the body accurately. The physiologic pharmacokinetic model—which accounts for processes of drug distribution, drug binding, metabolism, and drug flow to the body organs—is much more realistic. Disease-related changes in physiologic processes are more readily related to changes in the pharmacokinetics of the drug. Furthermore, organ mass, volumes, and blood perfusion rates are often scalable, based on size, among different individuals and even among different species. This allows a perturbation in one parameter and the prediction of changing physiology on drug distribution and elimination.
++
The physiologic pharmacokinetic model may also be modified to include a specific feature of a drug. For example, for an antitumor agent that penetrates into the cell, both the drug level in the interstitial water and the intracellular water may be considered in the model. Blood flow and tumor size may even be included in the model to study any change in the drug uptake at that site.
++
The physiologic pharmacokinetic model can calculate the amount of drug in the blood and in any tissues for any time period if the initial amount of drug in the blood is known and the dose is given by IV bolus. In contrast, the tissue compartment in the compartmental model is not related to any actual anatomic tissue groups. The tissue compartment is needed when the plasma drug concentration data are fitted to a multicompartment model. In theory, when tissue drug concentration data are available, the multiple-compartment models may be used to fit both tissue and plasma drug data together, including the drug concentration in a specific tissue. In such a case, the compartment model would mimic the system of equations used in the physiologic model, except that in place of blood flows, transfer constants would be used to describe the mass transfer in the model. The latter approach would probably, at best, yield less useful information than that obtained from the physiologic model.
+++
Physiologic Pharmacokinetic Model Incorporating Hepatic Transporter-Mediated Clearance
++
It is now well recognized that drug transporters play important roles in the processes of absorption, distribution, and excretion and should be accounted for in models. Predicting human drug disposition, especially when involving hepatic transport, is difficult during drug development. However, drug transport may be a critical process in overall drug disposition in the body such that without a realistic description of transport processes in the body, model accuracy may be deficient. Watanabe et al (2009) describe a model with hepatobiliary excretion mediated by transporters, organic anion-transporting polypeptide (OATP) 1B1 and multidrug resistance–associated protein (MRP) 2, for the HMG-CoA reductase inhibitor drug, pravastatin. While the classical blood flow–based physiologic pharmacokinetic models developed 40 years ago using systems of differential equations that are still useful in describing the mass balance and transfer of drug within major organs, the models are inadequate in light of new discoveries in molecular biology and pharmacogenomics. Drug disposition and drug targeting are better understood based upon using influx/efflux and binding mechanisms in micro structures such as interior cellular structures, membrane transporters, surface receptors, genomes, and enzymes. The liver is a complex organ intimately connected to drug transport and bile movement. Compartment concepts are needed to track the mass of drug transfer in and out of those fine structures as shown by the example in Fig. 22-7. Human liver microsomes are used to help predict the metabolic clearance of drugs in the body.
++
++
The PBPK model with pravastatin (Watanabe et al, 2009) is used to evaluate the concentration-time profiles for drugs in the plasma and peripheral organs in humans using physiological parameters, subcellular fractions (cells lysed and contents fractionated based on density), and drug-related parameters (unbound fraction and metabolic and membrane transport clearances extrapolated from in vitro experiments). The principle of the prediction was as follows. First, subcellular fractions were obtained by comparing in vitro and invivo parameters in rats. Then, the in vitro human parameters were extrapolated in vivo using the subcellular fractions obtained in rats. Pravastatin was selected as the model compound because many studies have investigated the mechanisms involved in the drug disposition in rodents, and clinical data after intravenous and oral administration are available.
++
When multiple drug metabolites are involved, the physiologic model of the cascade events can be quite complicated and an abbreviated approach may be used. St-Pierre et al (1988) developed a simple one-compartment open model, based on the liver as the only organ of drug disappearance and metabolite formation. The model was used to illustrate the metabolism of a drug to its primary, secondary, and tertiary metabolites. The model encompassed the cascading effects of sequential metabolism (Fig. 22-8). The concentration-time profiles of the drug and metabolites were examined for both oral and intravenous drug administration. Formation of the primary metabolite from drug in the gut lumen, with or without further absorption, and metabolite formation arising from first-pass metabolism of the drug and the primary metabolite during oral absorption were considered. Mass balance equations, incorporating modifications of the various absorption and conversion rate constants, were integrated to provide the explicit solutions.
++
++