Abstract
Introduction:
Extensive studies have been conducted to predict in vivo metabolic clearance from in vitro human liver metabolism parameters (i.e., in vitro-in vivo-extrapolation; IVIVE) with little success.
Methods:
Here, deriving IVIVE from first principles, we show that the product of fraction unbound in blood and the predicted in vivo intrinsic clearance determined from hepatocyte or microsomal incubations is the lower boundary condition for in vivo hepatic clearance and the prerequisite for IVIVE predictions to be valid, regardless of extraction ratio.
Results and Discussion:
For 60-80% of drugs evaluated here, this product is markedly less than the in vivo measured clearance, a result that violates the lower boundary of the predictive relationship. This can only be explained by (a) suboptimal in vitro metabolic stability assay conditions, (b) significant error in the assumption that in vitro intrinsic clearance determinations will predict in vivo intrinsic clearance simply by scaling-up the amount of enzyme (in vitro incubation to in vivo liver) and/or (c) the methods of determining fraction unbound are incorrect. We further suggest that widely-employed organ blood flow values underpredict the effective blood flow within the organ by approximately 2.5-fold, thus impacting IVIVE of high clearance compounds.
Conclusion:
We propose future pathways that should be investigated in terms of the relationship to experimentally measured clearance values, rather than model dependent intrinsic clearance. IVIVE outcome can be improved by estimating the ratio of unbound drug concentration in the liver tissue to the liver plasma, examining the assumption of the free drug theory (i.e., there are no transporter effects at the blood cell membrane) and the finding that the upper limit of organ clearance may be greater than blood flow entering the organ.
Keywords: IVIVE, Hepatic Clearance, Organ Blood Flow, Fraction Unbound
Introduction
Drug development is an extremely expensive, time-consuming process with an unacceptable, very poor success rate. It is well recognized that if we could predict an NME’s (new molecular entity’s) human ADME (absorption, distribution, metabolism, excretion) pharmacokinetic characteristics prior to dosing the drug to humans, or even animals, this could markedly speed the drug development process (1, 2). It seemed logical that measurements of in vitro elimination in human hepatocytes or subcellular fractions such as microsomes should be a useful pathway to predict in vivo drug clearance for metabolized drugs (3-6). Obach et al. (7) detailed two methods to estimate in vivo human metabolic clearance from in vitro metabolism measurements, designated IVIVE, in vitro to in vivo extrapolation. The first followed that of Rané et al. (3) where in vitro measures of maximum rate of drug elimination and the concentration giving half of the maximum rate were determined and translated into values of intrinsic clearance (CLint). In the second method, designated as the “in vitro T½ method”, CLint was determined by measuring the first order rate constant for drug elimination in a hepatocyte or microsome in vitro incubation and scaling this value up using relevant amounts of human liver metabolic enzymes. It is this second method that is primarily used today, with Obach (8) initially predicting human clearance of 29 drugs from hepatic microsomal intrinsic clearance data. Many further studies have attempted to use this methodology. However, in vitro measures of drug metabolic clearance in human liver tissue cannot adequately predict the in vivo human drug metabolic liver clearance across the panoply of metabolized substrates (9, 10), nor can in vitro measures of drug metabolic clearance in rat liver tissue adequately predict in vivo rat drug metabolic clearance (10). We reviewed multiple human hepatic IVIVE (in vitro-in vivo extrapolation) prediction studies where it is assumed that only unbound drug in blood/plasma is available for metabolism; the weighted average of studies indicates that 66.5% of predictions fell more than 2-fold outside of measured in vivo values with both microsomes and hepatocytes yielding approximately equivalent poor predictions (9). In the great majority of cases IVIVE under predicts human metabolic clearance. Wood et al. (10) also showed comparable poor predictions for hepatocytes and microsomes with a somewhat higher percentage of predictions (~75%) falling more than 2-fold outside of measured in vivo human metabolic clearance values.
Compounding the difficulty is that the field does not understand why the IVIVE predictions are inaccurate, even for purely metabolized drugs. In 2016, Takano et al. (11) wrote, “Theoretically, in vivo functions of enzymes/transporters should be predicted from in vitro metabolic/transporter activities simply by scaling up in vitro parameters with the use of the ratio of expression levels of molecules in whole tissue to that in the expression system.” That is:
| (1) |
where CLint terms represent the clearance capacity of the in vitro metabolic incubation or the in vivo whole liver to clear unbound drug (with units of volume per time per mg of metabolic enzymes) independent of flow and protein binding considerations. Chiba et al. (12) enumerate potential reasons for these CLint inaccuracies “Extrinsic factors such as preparation process and/or storage conditions of liver samples from human are likely responsible for the potential loss of metabolic activity, resulting in the systematic under predictions by biological scaling factors for human samples.” Wood et al. (10, 13) have comprehensively reviewed these reasons and many others in the literature (e.g., protein binding issues, cofactor depletion, unstirred water layers) suggesting that “ultimately, the in vitro causes of under prediction are likely to be multifactorial” and “more investigation is required to resolve the quantitative capabilities of in vitro systems”. We concur with the statement of Hallifax and Houston (14) concerning the need for further “mechanistic elucidation to improve prediction methodology rather than empirical correction of bias”, believing that the marked poor predictability may be a result of unappreciated conceptual understanding rather than poor experimental results. A recent investigation by Riccardi et al. (15) reported that correcting Eq. 1 for the partition of unbound drug between whole liver tissue and liver plasma for 16 out of 19 Extended Clearance Classification System (ECCS) class 1 and 2 drugs yielded predictions within 3-fold of observed values. For ECCS class 3 and 4 drugs, predictions were only improved to within 3-fold for 5 of 13 drugs.
Here we analyze the theoretical basis for IVIVE with focus on each of the individual contributors to total clearance, intrinsic clearance (CLint), protein binding (fu,B ), and hepatic blood flow (QH), to illustrate the reasons for the present inadequacies of the methods used and the potential methodologies that can improve IVIVE predictions. We further suggest a major flaw in IVIVE theory which results from utilizing a “Chemistry” approach to predict a “Pharmacokinetic” parameter. In this context, the term “Chemistry” refers to the in vitro measurement of drug concentration loss (i.e., rate of reaction) in a fixed measurable incubation volume that is drug independent. All pharmacokinetic derivations are based on mass balance (amount) considerations. These amounts are converted to concentrations incorporating a volume of distribution that does not have physiological relevance, that is not independently measurable, and is drug dependent. The term “Pharmacokinetics” refers to the in vivo scenario, where observed drug elimination (based on total drug amounts rather than concentrations) can also be influenced by drug distribution within the liver, can occur to a different degree for each drug, and is an aspect not currently captured by traditional “chemistry”-based incubations.
Materials and Methods
Pharmacokinetics vs. Chemistry
The most critical pharmacokinetic parameter is clearance, CL, a measure of the body’s ability to eliminate drug, since at steady-state during multiple dosing of a drug, the rate into the body will equal the rate out.
| (2) |
where a proper Dosing Rate (dose, D, divided by the dosing interval) can be determined knowing the desired therapeutic concentration, the fractional availability of dosed drug that will reach the systemic circulation intact (F) and CL. Equation 2 is a definition of clearance, a term exclusive to pharmacokinetics, as the amount of drug lost from systemic circulation over a given time period divided by the exposure driving that elimination. More frequently clearance is defined in vivo as
| (3) |
where the amount of drug lost from the systemic circulation following a single dose (available dose, F · D) is divided by the exposure driving that elimination, measured as the total area under the systemic concentration time curve over all time (AUC). Thus, giving an intravenous bolus dose of drug (F=1) and measuring the total drug exposure will allow CL to be determined. Clearance will change in predictable ways as a function of physiology, pharmacogenomics and pathology, as well as a result of drug-drug interactions.
To help understand why in vivo clearance is commonly underpredicted by IVIVE approaches, it would be helpful to review the development of IVIVE practices by the field of Drug Metabolism. Drug metabolism can be considered analogous to a chemical reaction; however, clearance is not a measurement used in chemistry. A chemical reaction can be considered analogous to drug elimination in humans or animals where metabolism is the major route of elimination, and thus for the current discussion we focus only on the simple case of metabolism with no involvement of xenobiotic transporters. In chemistry under conditions of linear processes, the rate of elimination in an incubation mixture is most frequently characterized by the Michaelis-Menten ratio of Vmax, the maximum rate of the decrease in the concentration of a drug in units of concentration/time, to Km, the concentration of drug at half Vmax. This ratio Vmax/Km thus has units of time−1 since in chemistry reaction rates are the measure of relevance, where Vmax is a function of the concentration of enzyme present to carry out metabolism. For linear processes (when drug concentrations are much less than Km), this ratio can be simplified into a single half-life, and such an approach has been adopted for substrate-depletion drug metabolism incubations where drug concentrations are measured i.e., the “in vitro T½ method” of Obach et al. (7). Note that although Vmax was originally and continues to be defined in chemistry in terms of a concentration change, in pharmacokinetics Vmax has been defined as an amount change that results in the ratio Vmax/Km as a clearance parameter (with the units of volume per unit time) rather than as a rate constant. Pharmacokinetic scientists have approached this discrepancy by converting the units of Vmax, as discussed subsequently, rather than derive the classic Michaelis-Menten relationship based on amounts (so that the units of Vmax will be amount per unit time).
So how is the rate constant from the in vitro incubation mixture (with the units of time−1) converted into a clearance (with the units of volume per unit time)? By multiplying this rate constant by the volume of fluid in the incubation mixture. This detail has not been widely-recognized by the field, since the volume term is typically introduced by dividing the measured rate of elimination (units of time−1) by the concentration of enzymes in the incubation (units of amount per volume).
| (1a) |
Comparison of Eq. 1a to Eq. 1 clearly depicts how the fixed volume of incubation is incorporated in the conversion of in vitro rate of drug loss to what the field considers a prediction of in vivo intrinsic clearance. However, this approach does not account for the pharmacokinetic volume of distribution of drug within the liver that cannot be recapitulated nor scaled-up from traditional IVIVE incubations.
Since this parameter is determined for unbound drug (by accounting for fu,inc, the unbound fraction of drug within the incubation) and independent of blood flow to the liver, this clearance is defined as an intrinsic clearance, a concept introduced in the early 1970s (16, 17), which had a revolutionary effect on our ability to select and modify drug dosing regimens as opposed to prior dependence on measures of drug half-lives. This is because drugs can exhibit multiple half-lives in the systemic circulation as concentrations decrease and upon multiple dosing, and require a model of drug elimination incorporating multiple exponential terms, even for drugs exhibiting linear kinetics. In contrast CL is compartment model independent and is defined (and can be determined) as the ratio of the amount of drug lost divided by the drug exposure in the systemic circulation driving that elimination (Eq. 3). There is a second difference between chemistry and pharmacokinetics of relevance to this analysis. In chemistry, the volume of the incubation mixture is fixed and independent of the drug for which the elimination is being measured. While in pharmacokinetics, volume terms are drug dependent and are non-physiologic apparent volumes that relate the concentration of drug measured in the systemic circulation to the amount of drug in the body. In pharmacokinetics, it is well recognized that drug clearance and drug volume of distribution are independent parameters whereas half-life is the dependent variable that can change as a result of change in clearance or in volume of distribution (18). A physiologic condition such as aging can change the volume of distribution of a drug such as diazepam, changing the half-life but not changing the drug clearance (19, 20). It is also well recognized that the volume of distribution is drug dependent and does not represent actual physiologic volumes in the body (18). Pharmacokinetic volume terms do not represent a definable space, such as the volume of fluid in a beaker in a chemistry reaction. Thus, the two major differences between chemistry and pharmacokinetics are in the definition of Vmax and the possibility that volume of distribution may vary depending on the drug evaluated in pharmacokinetics but remains a fixed, drug independent value in chemistry.
The Relationship between Clearance and Intrinsic Clearance for In Vivo Hepatic Elimination
Rowland et al. (16) and Wilkinson and Shand (17) defined the relationship between the in vivo measured clearance of a drug, presented in terms of Eqs. 2 and 3, and a hypothetical in vivo intrinsic clearance based on the well-stirred model of liver elimination as given in Eq. 4:
| (4) |
where fu,B is the ratio of the unbound concentration of drug in the plasma to the whole blood concentration and QH is the hepatic blood flow rate. Since QH and fu,B can be experimentally measured or estimated, it is this CLint,in vivo value that one tries to determine using Eq. 1.
Let us now look at the relationship between fu,B · CLint and CLH,in,vivo to understand how such in vitro measures of drug metabolism (in tandem with predictions of unbound drug in the blood) are directly related to total clearance in vivo. When the product fu,B · CLintis a small value, for instance for low extraction ratio drugs for which QH >> fu,B · CLint, the predictive clearance relationship (Eq. 4) simplifies to fu,B · CLint,in vivo that is:
| (5) |
When the product fu,B · CLint is a large value, for instance for high extraction ratio drugs for which QH << fu,B · CLint, the Eq. 4 clearance relationship simplifies to CLH = QH, that is:
| (6) |
consistent with the belief that an organ cannot eliminate a drug faster than its delivery to that organ, clearance can never exceed the effective blood flow to that organ. Here it is important to note that although the values of CLH range from fu,B · CLint to QH, for high clearance compounds (where CLH approaches QH) the value of fu,B · CLint is significantly larger than QH. Therefore, when comparing the values of fu,B · CLint to CLH, the value of CLH can never be greater than the product fu,B · CLint, that is:
| (7) |
Conceptually, this can be rationalized by considering that for very high clearance drugs (those with high CLint values), the large product fu,B · CLint will result in total clearance values that are limited by the rate at which drug is presented to the organ (QH). Thus, the value of fu,B · CLint will always be equal to or larger than total CLH, and can never be smaller. This conclusion is based on theoretical considerations, and in practice experimental determinations of each parameter may result in under- or over-predictions, as we will evaluate in detail subsequently. Thus, comparison of the product fu,B · CLint to total CLH will provide enhanced insight into the overall predictability of in vitro determinations, as opposed to comparison of CLint values alone.
In the evaluation of IVIVE predictability, we emphasize that the contribution of both experimental and theoretical error in each of the terms CLint, fu,B and QH can have impact on the resulting CLH predictions. Many IVIVE investigations in the literature are based on evaluation of error between CLH,in vivo,predicted and CLint,in vivo where the CLint,in vivo is back-calculated from total CLH measurements under the assumption that 1) measured in vivo CLH is accurate, 2) determinations of fu,B are accurate, and that 3) the value of QH is accurate. These assumptions may be considered reasonable; however, any resulting errors in IVIVE are thus primarily attributed to issues with in vitro determination of CLint. Further, CLH is a quantifiable in vivo parameter, whereas CLint can only be estimated indirectly by assuming a model of hepatic disposition. Since IVIVE unpredictability continues to challenge the field, here we evaluate IVIVE success by comparing CLH values in order to better understand the potential contribution of errors in each term. This was achieved by examining notable IVIVE studies to determine CLH,in vivo,predicted by combining Eqs. 1 and 4.
We specifically analyze the 29 drugs first compiled by Obach (8) for human microsome measurements and the very large data set of Wood et al. (10), which included data for 101 drugs where in vitro measures of CLint,on vitro were determined in human hepatocytes, 83 drugs with human microsome incubations, 127 compounds with rat hepatocyte incubations and 71 compounds with rat microsome incubations. The Obach (8) dataset reported CLH,in vivo,predicted, however the CLint,in vivo values reported by the Wood et al. (10) analysis were utilized to predict CLH,in vivo,predicted and were compared with measured CLH,in vivo values listed in those papers, resulting in the poor predictability detailed above. We also analyzed the results for the 11 ECCS class 1 and 2 drugs of Riccardi et al. (15) where in vitro hepatocyte data were also available in the Wood et al. (10) data base. The fu,B and QH values used by the authors of these three investigations were also utilized in the current analysis.
Results
We first determined how many of the tabulated measurements met the Eq. 7 lower boundary requirement in each of the Wood et al. (10) and Obach (8) data sets, i.e. drugs for which CLH,in vivo ≤ fu,B · CLint,in vitro, as presented in Table I. As can be seen for the Wood et al. (10) data set, 74.3% of the human hepatocyte predictions were used to predict CLH,in vivo from fu,B · CLint, in vivo measures that violate the boundary conditions of the clearance prediction equation. In other words, for 74.3% of the human hepatocyte predictions, it would not be possible to obtain a correct prediction of human metabolic clearance. For human microsomes 52 of 83 predictions (62.7%) used fu,B · CLint,in vitro measures that violate the Eq. 7 boundary condition. For the analyses in rats the values are 100 of 127 (78.7%) for hepatocytes and 44 of 71 (62.0%) for microsomes. For the 29 drugs assessed by Obach (8) using human microsomes, the percentage violating the Eq. 7 boundary condition is 69.0%, somewhat higher than the 62.7% for the Wood et al. (10) data using human microsomes. Table II presents the predictability within 2-fold for each of the Wood et al. (10) data sets when the Eq. 7 boundary condition is met (i.e. CLH,in vivo ≤ fu,B · CLint,in vitro). When the boundary condition is not violated, 73.1-77.8% of the predictions are within 2-fold for the Wood et al. (10) data sets, while all 9 (100%) of the Obach (8) predictions are correct within 2-fold. The predictability for data violating the boundary conditions (CLH,in vivo > fu,B · CLint,in vitro) is much poorer, 11.0-22.7% in Table II and 35% in the Obach (8) data set. Note in Fig. I that all of the human predictions that meet the boundary condition but that are not within 2-fold are overpredictions. There are no overpredictions greater than 2-fold for the data violating the Eq. 7 boundary condition. To determine if the observed difference could be improved if drugs where transporters affect clearance are excluded, we also evaluated the subset of Class 1 BDDCS drugs (drugs that exhibit high permeability rate, are extensively metabolized and highly soluble) where transporters are believed to not have a clinically significant effect on systemic clearance (21). As seen in Table I, no significant differences were observed between the BDDCS Class 1 drugs and the entire data set with regard to the distribution of drugs between the two groups for humans. However, from Table II it can be seen that successful IVIVE predictability increases by 10 to 20% when only BDDCS Class 1 marketed drugs not violating the Eq. 7 boundary condition are evaluated.
Table I.
Analysis of reported data from Wood et al. (10) for comparison of the product of fu,B and CLint, in vitro with CLH, in vivo
| Human Hepatocytes | Human Microsomes | ||||
|---|---|---|---|---|---|
| CLH > fu,B · CLint,invitro | CLH ≤ fu,B · CLint,invitro | CLH > fu,B · CLint,invitro | CLH ≤ fu,B · CLint,invitro | ||
|
All Drugs (n = 101) |
74.3% (n=75 of 101) |
25.7% (n=26 of 101) |
All Drugs (n = 83) |
62.7% (n=52 of 83) |
37.3% (n=31 of 83) |
|
BDDCS Class 1 (n = 60) |
71.7% (n=43 of 60) |
28.3% (n=17 of 60) |
BDDCS Class 1 (n = 47) |
66.0% (n=31 of 47) |
34.0% (n=16 of 47) |
| Rat Hepatocytes | Rat Microsomes | ||||
| CLH > fu,B · CLint,invitro | CLH ≤ fu,B · CLint,invitro | CLH > fu,B · CLint,invitro | CLH ≤ fu,B · CLint,invitro | ||
|
All Drugs (n = 127) |
78.7% (n=100 of 127) |
21.3% (n=27 of 127) |
All Drugs (n = 71) |
62.0% (n=44 of 71) |
38.0% (n=27 of 71) |
|
Marketed Drugs (n = 38) |
47.4% (n=18 of 38) |
52.6% (n=20 of 38) |
Marketed Drugs (n = 34) |
35.3% (n=12 of 34) |
64.7% (n=22 of 34) |
|
BDDCS Class 1 (n = 21) |
47.6% (n=10 of 21) |
52.4% (n=11 of 21) |
BDDCS Class 1 (n = 17) |
29.4% (n=5 of 17) |
70.6% (n=12 of 17) |
Table II.
Analysis of reported data from Wood et al. (10) for comparison of the product of fu,B and CLint, in vitro with CLH, in vivo in terms of predictability within 2-fold
| Human Hepatocytes | Human Microsomes | ||||
|---|---|---|---|---|---|
| CLH > fu,B · CLint,invitro | CLH ≤ fu,B · CLint,invitro | CLH > fu,B · CLint,invitro | CLH ≤ fu,B · CLint,invitro | ||
|
All Drugs 2-fold: 30.7% (n=31 of 101) |
2-fold: 16.0% (n=12 of 75) |
2-fold: 73.1% (n=19 of 26) |
All Drugs 2-fold: 42.2% (n=35 of 83) |
2-fold: 23.1% (n=12 of 52) |
2-fold: 74.2% (n=23 of 31) |
|
BDDCS Class 1 2-fold: 35.0% (n=21 of 60) |
2-fold: 16.3% (n=7 of 43) |
2-fold: 82.4% (n=14 of 17) |
BDDCS Class 1 2-fold: 48.9% (n=23 of 47) |
2-fold: 25.8% (n=8 of 31) |
2-fold: 93.8% (n=15 of 16) |
| Rat Hepatocytes | Rat Microsomes | ||||
| CLH > fu,B · CLint,invitro | CLH ≤ fu,B · CLint,invitro | CLH > fu,B · CLint,invitro | CLH ≤ fu,B · CLint,invitro | ||
|
All Drugs 2-fold: 25.2% (n=32 of 127) |
2-fold: 11.0% (n=11 of 100) |
2-fold: 77.8% (n=21 of 27) |
All Drugs 2-fold: 43.7% (n=31 of 71) |
2-fold: 22.7% (n=10 of 44) |
2-fold: 77.8% (n=21 of 27) |
|
Marketed Drugs 2-fold: 57.9% (n=22 of 38) |
2-fold: 38.9% (n=7 of 18) |
2-fold: 75.0% (n=15 of 20) |
Marketed Drugs 2-fold: 61.8% (n=21 of 34) |
2-fold: 33.3% (n=4 of 12) |
2-fold: 77.3% (n=17 of 22) |
|
BDDCS Class 1 2-fold: 71.4% (n=15 of 21) |
2-fold: 50.0% (n=5 of 10) |
2-fold: 90.9% (n=10 of 11) |
BDDCS Class 1 2-fold: 82.4% (n=14 of 17) |
2-fold: 60.0% (n=3 of 5) |
2-fold: 91.7% (n=11 of 12) |
Figure 1.

Comparison of IVIVE error (predicted in vivo clearance/observed in vivo clearance) versus observed in vivo clearance using A. human hepatocytes and B. human microsomes from the Wood et al. (10) data set for analyses where the product of the in vitro measured CLint and fu,B violates hepatic model boundary conditions (red dots) and where it does not (green dots).
Figure I presents the comparison of the accuracy of the human IVIVE prediction for the Wood et al. (10) data set versus the measured in vivo hepatic clearance for drugs where the product fu,B · CLint,in vitro violates the Eq. 7 boundary condition (red dots) and where it does not (green dots). The results for the rat data are presented in Fig. II. It is obvious that the predictions for drugs that do not violate the Eq. 7 boundary condition give a better IVIVE outcome. We also investigated the charge distribution of drugs relative to violation of the Eq. 7 boundary condition as presented in Fig. III and Table III. Acids are over represented in drugs violating the Eq. 7 boundary condition both for hepatocytes and microsomes. The distribution in the 29 drug Obach (8) data set is similar. Only 1 of 9 acids did not violate the Eq. 7 boundary condition, 6 of 12 bases and 2 of 8 neutral compounds.
Figure 2.

Comparison of IVIVE error (predicted in vivo clearance/observed in vivo clearance) versus observed in vivo clearance using A. rat hepatocytes and B. rat microsomes from the Wood et al. (10) data set for analyses where the product of the in vitro measured CLint and fu,B violates hepatic model boundary conditions (red dots) and where it does not (green dots).
Figure 3.

Distribution by charge for compounds tabulated by Wood et al. (10) for acids (red), bases (blue), neutrals (green) and zwitterions (purple) in human hepatocytes and human microsomes.
Table III.
Distribution by charge for drugs violating and not violating boundary conditions as tabulated by Wood et al. (10) for A. hepatocyte measurements and B. microsome measurements
| A. | |||
|---|---|---|---|
| Human Hepatocytes | |||
| CLH,in vivo > fu,B · CLint,invitro | CLH,in vivo ≤ fu,B · CLint,invitro | ||
|
All Drugs (n = 101) |
n=75 (74.3% of 101) |
n=26 (25.7% of 101) |
|
|
Acids (n = 19) |
n=17 (22.7% of 75) (89.5% of 19) |
n=2 (7.7% of 26) (10.5% of 19) |
|
|
Bases (n = 47) |
n=32 (42.7% of 75) (68.1% of 47) |
n=15 (57.7% of 26) (31.9% of 47) |
|
|
Neutrals (n = 31) |
n=22 (29.3% of 75) (71.0% of 31) |
n=9 (34.6% of 26) (29.0% of 31) |
|
|
Zwitterions (n = 4) |
n=4 (5.3% of 75) (100% of 4) |
n=0 (0% of 26) (0% of 4) |
|
| B. | |||
| Human Microsomes | |||
| CLH,in vivo > fu,B · CLint,invitro | CLH,in vivo ≤ fu,B · CLint,invitro | ||
|
All Drugs (n = 83) |
n=52 (62.7% of 83) |
n=31 (37.3% of 83) |
|
|
Acids (n = 15) |
n=14 (26.9% of 52) (93.3% of 15) |
n=1 (3.2% of 31) (6.7% of 15) |
|
|
Bases (n = 35) |
n=20 (38.5% of 52) (57.1% of 35) |
n=15 (48.4% of 31) (42.9% of 35) |
|
|
Neutrals (n = 30) |
n=17 (32.7% of 52) (56.7% of 30) |
n=13 (41.9% of 31) (43.3% of 30) |
|
|
Zwitterions (n = 3) |
n=1 (1.9% of 52) (33.3% of 3) |
n=2 (6.5% of 31) (66.6% of 3) |
|
For the 19 ECCS class 1 and 2 drugs analyzed by Riccardi et al. (15), 11 were in the data base of Wood et al. (10). Ten of the 11 violated the Eq. 7 boundary condition. Only theophylline did not, but theophylline was one of three drugs analyzed by Riccardi et al. (15) that was not improved by correcting Eq. 1 for the partition of unbound drug between whole liver tissue and liver plasma. Since Riccardi et al. (15) do not actually provide the in vitro experimental CLint, in vitro measures in their paper, we do not know if their values would lead to the same Eq. 7 boundary condition violations for the 11 drugs. But at this point the Riccardi method improves the prediction for the drugs violating the Eq. 7 boundary condition, but does not for the one drug not in violation, theophylline.
More recently, Hallifax and Houston (22) presented analyses to attempt to explain the increasing degree of underprediction of CLint observed with increasing values of in vivo CLint, calculated from measures of in vivo clearance by rearrangement of Eq. 4. These studies evaluate IVIVE prediction success by comparing the calculated in vivo CLint (from Eq. 4) to the prediction of in vivo CLint derived from hepatocyte and microsome in vitro CLint measurements (Eq. 1). We prefer rather to evaluate the total CLin vivo, predicted determined using Eqs. 1 and 4 with CLin vivo, measured, since ultimately total clearance is the most relevant parameter, and because IVIVE errors based on CLint for high clearance compounds may not translate to significant error in total clearance predictions. In Figs. I and II, IVIVE underprediction also becomes greater with increasing observed measured clearance, as observed by Hallifax and Houston (22). This trend is observed for all data, and separately for the predictions where Eq. 7 is violated (red dots) and where it is not (green dots). The underprediction error becomes greater and greater as the measured in vivo clearance approaches the upper boundary Eq. 6 condition, where CLin vivo, predicted approaches hepatic blood flow. The potential reasons for such an outcome have recently been discussed (10) and have primarily been attributed to inadequacies of in vitro methodologies. However, this outcome could also be explained by proposing that the measured hepatic blood flow in both humans and rats is not the relevant value to use in Eq. 4. In Table IV we have taken the human hepatocyte and microsome data of Wood et al. (10) that do not violate the Eq. 7 boundary condition and examined the effect of changing hepatic blood flow. As can be seen, increasing QH from 15 ml/min to 20 [the value used by Wood et al. (10) was 20.7] to 25, 30, 40 and 50 ml/min brings the slope very close to 1.0 for both hepatocytes and microsomes, eliminating the trend of high clearance drugs giving a greater underprediction. As the change in QH will have little effect on low clearance drugs, this is reflected in only minimal change in the Y-intercept values. Thus, we suggest that widely-employed organ blood flow values could underpredict the effective blood flow within the organ by approximately 2.5-fold, thus impacting IVIVE of high clearance compounds.
Table IV.
Regression of CLin vivo predictive vs CLin vivo, measured with changing hepatic blood flow and percent predictions falling within 2-fold for: A. the 26 drugs not violating Eq. 5 for hepatocyte measurements and B. the 31 drugs not violating Eq. 5 for microsome measurements from the Wood et al. (10) data set
| A. | |||||||
|---|---|---|---|---|---|---|---|
| Matrix | n | QH (ml/min/kg) |
r2 | Slope | Y-Intercept | Percent Within 2-Fold |
|
| Human Hepatocytes | 26 | 15 | 0.660 | 0.485 | 2.40 | 73.1% | |
| Human Hepatocytes | 26 | 20 | 0.649 | 0.600 | 2.49 | 73.1% | |
| Human Hepatocytes | 26 | 25 | 0.638 | 0.697 | 2.54 | 73.1% | |
| Human Hepatocytes | 26 | 30 | 0.626 | 0.780 | 2.55 | 73.1% | |
| Human Hepatocytes | 26 | 40 | 0.605 | 0.916 | 2.53 | 73.1% | |
| Human Hepatocytes | 26 | 50 | 0.588 | 1.02 | 2.49 | 73.1% | |
| B. | |||||||
| Matrix | n | QH (ml/min/kg) |
r2 | Slope | Y-Intercept | Percent Within 2-Fold |
|
| Human Microsomes | 31 | 15 | 0.601 | 0.397 | 3.84 | 77.4% | |
| Human Microsomes | 31 | 20 | 0.591 | 0.521 | 4.05 | 74.2% | |
| Human Microsomes | 31 | 25 | 0.576 | 0.633 | 4.14 | 67.7% | |
| Human Microsomes | 31 | 30 | 0.558 | 0.736 | 4.16 | 67.7% | |
| Human Microsomes | 31 | 40 | 0.523 | 0.920 | 4.07 | 61.3% | |
| Human Microsomes | 31 | 50 | 0.492 | 1.08 | 3.91 | 58.1% | |
This increasing error with measured CLin vivo values raises the question as to whether the errors involved with the violation of the boundary condition of Eq. 7 are greater for high clearance compounds versus low clearance compounds. To address this, we evaluated the relative error observed in Figs. 1 and 2 with increasing measured CLin vivo values and found no relationship (analysis not shown).
Discussion
In the latter half of the 2010s our laboratory began investigating the reasons that IVIVE predictions were so poor. We first confirmed this finding and showed that it was probably not a function of confounding elimination of metabolized drugs with drugs that were also substrates for transporters (9) and these results were further confirmed by Wood et al. (10). We followed by examining interlaboratory variability in human hepatocyte intrinsic clearance values and trends with physicochemical properties (23), confirming the hepatic clearance-dependent underprediction (24) and identifying an apparent microsomal IVIVE anomaly for CYP3A4 substrates for which microsomes give markedly higher IVIVE success in humans than for other enzymes, but the same trend was not observed in hepatocytes (25). We also addressed the measurement of fu,B and protein-facilitated uptake relating to IVIVE (26) and proposed the presence of a transporter-induced protein binding shift as a new explanation for protein-facilitated uptake and to improve IVIVE (27). During the course of these studies we found that all theoretical aspects related to IVIVE may not be fully recognized and specifically that although CLint may be relevant for all models of hepatic elimination, we have recently suggested that it can only be determined for the well-stirred model via Eq. 4 (28). However, it is important to recognize for the present analysis that the total clearance boundary conditions expressed in Eqs. 5 and 6 hold for all models of hepatic elimination. Thus, the violation of the Eq. 7 boundary condition by underprediction (Tables I and II, Figs. I and II) cannot be explained by examining different models of hepatic elimination.
What then are the possible explanations for the poor IVIVE predictability? It could be that all or some of the published measurements of CLH,in vivo, CLint, in vitro,, fu,B and QH are invalid. This could be, but since many of these parameters have been investigated by many scientists, with a reasonable degree of concordance, this is probably not the answer. Let us look now at the individual parameters.
Correctly measuring in vivo blood clearance.
We believe that most investigators would agree that measurements of CLH,in vivo determined from clinical studies in humans using versions of Eqs. 2 and 3 would be reasonably accurate. However, it is important to recognize that this assumption depends on a number of factors, including that total clearance values must be determined following IV dosing or with an accurate estimate of F. In addition, the influence of inter-individual variability and potential of saturable processes such as absorption or metabolism, adequate early sampling for high clearance compounds (to accurately capture initial concentrations) as well as adequate terminal-phase sampling to minimize errors associated with AUC extrapolation. In general, many of these aspects are given consideration in clearance determinations conducted in the drug approval process. Of concern, however, is the recognition that the total clearance value must be in terms of blood concentrations, not plasma. For example, the largest compilation of 1352 drug compound IV clearance values are all presented as plasma clearances (29). To convert these plasma values to blood clearances one must have an accurate measurement of the blood to plasma concentration ratio (). When such measurements are unavailable investigators often assume the value to be equal to 1 for a basic or neutral compound and 0.55 for an acidic compound, as was done by Wood et al. (10). Riccardi et al. (15) measured for each of the compounds studied and Obach (8) measured this value for the 11 drugs for which literature values were not available at the time.
Protein binding determinations.
Most investigators may also believe that fu,B measurements are relatively error free and reproducible. However, there is a theoretical issue that needs to be recognized. The experimental fu,B values are not the fraction unbound in blood, but rather the ratio of the unbound concentration of drug in the plasma to the whole blood concentration as given in Eq. 8.
| (8) |
where fu,P is the fraction unbound in plasma. Typically, measurements of fu,P and are conducted in separate in vitro incubations and fu,B is calculated via Eq. 8. For fu,B to be the fraction unbound in blood, which is what should be required in Eq. 4, the free drug hypothesis must hold. That is, free drug concentrations must be equal in the plasma and the blood cell, or simply that there must be no transporter effects in blood cell to plasma distribution. Transporters have certainly been identified in blood cell membranes and one might suggest that the poor predictability of acids, as demonstrated in Fig. III and Table III, could be explained by transporter effects in blood cells, since acids are very frequently substrates for transporters. The great majority of the compounds studied by Riccardi et al. (15) were acids. Further investigations into the implications of the free drug theory assumption in calculating fu,B are currently underway in our laboratory, allowing further insight into when in vitro measurement of fu,P and can provide reliable estimates of fu,B, and the impact this will have on IVIVE predictability. Further consideration of the potential for red blood cells to support the energetics of active transport processes is warranted, as these cells lack mitochondria.
Another protein binding issue relates to determining the fraction unbound in the in vitro incubation mixture (fu,inc) to obtain the CLint, in vitro parameter in Eq. 1. Here fraction unbound in the microsomal or hepatocyte incubation is measured or when unavailable calculated from predictive regression equations as reported by Wood et al. (10).
Correctly determining in vivo intrinsic clearance.
The most probable source of error is in the determination of CLint, in vivo in Eq. 1. From the in vitro incubation, utilizing the “in vitro T½ method”, we calculate a “chemistry” clearance as the product of the rate constant for drug elimination multiplied by the volume of fluid in the incubation mixture, a volume term that is drug independent. But in vivo, clearances are the product of the rate constant for elimination multiplied by a pharmacokinetic drug dependent volume of distribution. That is, the volume of distribution for each drug is different and not reflective of an actual fluid space (18). We are suggesting that the volume of distribution within the liver itself, for instance into lipophilic regions of the liver, is currently unaccounted for in IVIVE practices. Thus, it is possible that in vivo the extent of distribution into lipophilic regions of the liver (away from the intracellular hepatocyte water where metabolic enzymes exist) will be different for each drug, depending on the unique physicochemical properties of each drug. This is an area of research that needs to be further investigated, and significant efforts towards understanding the impact of liver volume of distribution on IVIVE are currently being pursued by our laboratory. We point out that the suggested liver volume of distribution should not be confused with the total-body volume of distribution at steady state (Vss). It is possible that the in vivo the volume of distribution of drug in the whole liver may be different than the volume of distribution of drug in contact with metabolic enzymes, versus the in vitro condition where the volume of distribution of drug and enzymes will be the same. A similar difference between the in vitro and in vivo conditions was proposed by Riccardi et al. (15) who utilize a measurement of the partition of unbound drug between whole liver tissue and liver plasma to improve IVIVE predictions primarily for acid drugs where transporter effects may be anticipated. We propose that the in vitro experimental measure of drug elimination in a hepatocyte incubation may not reflect the difference found in the in vivo liver where the unbound concentration of drug directly in contact with the liver enzymes and throughout the whole liver may not be the same, possibly due to drug distribution into lipophilic regions of the liver. It appears that for the drugs that are consistent with Eq. 7 in Figs. I and II (green dots) this correction is not needed, but for drugs not consistent with Eq. 7 (red dots), the correction is required. There are some red dots that fall within the 2-fold estimation bands. But the estimation, of course, also depends on correctly determining fu,B, as we have discussed above. At present, we believe that a correction for an NME using the Riccardi et al. (15) methodology, or other to-be-developed methodologies, will be necessary when the NME violate the Eq. 7 boundary condition. But at this point, there is no way to determine if the correction is needed or not a priori.
Organ flow characterization.
Probably the most surprising finding in the analysis presented here is that actual organ blood flow may be underestimating the parameter to be utilized in Eq. 4. It is universally agreed in pharmacokinetics that organ blood clearance cannot exceed organ blood flow, but here we are suggesting that relevant organ blood flow for metabolism may be greater than the blood flow into the organ. Perhaps this could be the result of blood flow circling and becoming available to the hepatocyte two or three times for each ml/min entering the liver, however, we currently do not have a theory based on liver architecture to explain this potential phenomenon. This only becomes evident when CLin vivo, predicted continues to decrease from CLin vivo measured for higher and higher extraction ratio drugs, and of course is consistent with Eq. 4. The data in Table IV shows the decrease in slope for the predicted vs measured CLin vivo values, with little change in intercept. However, we also note that r2 values for the regression also become poorer with increasing flow, and perhaps this may reflect the influence of fu,B errors that become more obvious as slope decreases. These simulations are by no means direct evidence of this phenomenon, but this is a starting point and a potentially fruitful area requiring further investigation.
Conclusions
Future experimental pathways.
The present manuscript was written to examine the theoretical basis for IVIVE, to identify the assumptions that have been made, in some cases unknowingly, and provide direction for future experimental pathways for the field to pursue to improve IVIVE practices. We critically re-evaluate the experimental data for the recently published large data set of Wood et al. (10), the initial 29 drug data set of Obach (8) and the drugs investigated by Riccardi et al. (15), which includes a number of transporter substrates. We point out that from two-thirds to three-fourths of the data examined to predict IVIVE (supposedly for drugs where transporter effects are minimal) violate the fundamental assumption of all models of hepatic elimination, i.e., that fu,B · CLint must be greater than CLH,in vivo, again confirming the significant IVIVE-underprediction trend that the field has struggled with for decades. This outcome depends on the assumption that measured in vivo clearance values are reasonably accurate and noting that accurate blood clearance values depend on blood concentration to plasma concentration ratios when plasma clearance is reported, which may be inaccurate for transporter substrates due to the presence of transporters in the red blood cell membranes. The assumption of the free drug theory in calculating fu,B may result in significant errors that not only applies to total clearance values, but also in scaling up in vitro CLint for prediction of CLH,in vivo, and should be further investigated to elucidate its impact on IVIVE success for transporter substrates. We derived the relationship between the in vitro and in vivo intrinsic clearances, showing that calculating the in vitro intrinsic clearance employs a chemistry approach where the volume multiplied by the rate constant of elimination is fixed. In contrast, for the in vivo pharmacokinetic approach, clearance may be considered the product of the rate constant of elimination multiplied by a drug dependent (and physiologically unidentifiable) volume of distribution. We propose that the relationship between the in vitro and in vivo intrinsic clearances should consider the physiologic difference of drug distribution in the whole liver versus the uniform distribution in vitro. The methodology of Riccardi et al. (15) provides a good starting point in examining when the correction is necessary and when it is not. Finally, the analysis here suggests that for high clearance compounds, the value for blood flow entering the liver may be underpredicting the blood flow that sets the upper limit (Eq. 6) for in vivo clearance. Future experimental studies should investigate this possibility and the other issues raised in this manuscript
Acknowledgements
The authors thank members of our laboratory Dr. Jin Dong (present address: Simulations Plus), Alan R. Wolfe, Shufang Liu (present address: SUNY Buffalo) and Dr. Giovani Bocci (present address: University of New Mexico) for very helpful discussions.
This work was supported in part by a Mary Anne Koda-Kimble Seed Award for Innovation. Dr. Benet is a member of the UCSF Liver Center supported by NIH Grant P30 DK026743. Ms. Sodhi was supported in part by an American Foundation for Pharmaceutical Education Pre-Doctoral Fellowship, NIGMS grant R25 GM56847 and a Louis Zeh Fellowship.
Abbreviations
- ADME
absorption, distribution, metabolism, excretion
- AUC
area under the concentration time curve
- BDDCS
biopharmaceutics drug disposition classification system
blood to plasma partitioning ratio
- CL
clearance
- CLint
intrinsic clearance
- CYP3A
cytochrome P450 3A
- D
dose
- ECCS
Extended Clearance Classification System
- F
bioavailability
- fu,B
fraction of unbound drug in blood
- fu,inc
fraction of unbound drug in an in vitro incubation
- fu,P
fraction of unbound drug in plasma
- IVIVE
in vitro-in vivo extrapolation
- NME
new molecular entity
- QH
hepatic blood flow
References:
- 1.Page KM. Validation of early human dose prediction: a key metric for compound progression in drug discovery. Mol Pharmaceut. 2015;13:609–620. [DOI] [PubMed] [Google Scholar]
- 2.Krauss M, Hofmann U, Schafmayer C, Igel S, Schlender J, Mueller C, Brosch M, von Schoenfels W, Erhart W, Schuppert A, Block M, Schaeffeler E, Boehmer G, Goerlitz L, Hoecker J, Lippert J, Kerb R, Hampe J, Kuepfer L, Schwab M. Translational learning from clinical studies predicts drug pharmacokinetics across patient populations. NPJ Syst Biol Appl. 2017;3:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Rané A, Wilkinson GR, Shand DG. Prediction of hepatic clearance ratio from in vitro measurement of intrinsic clearance. J Pharmacol Exp Ther. 1977;200:420–424. [PubMed] [Google Scholar]
- 4.Houston JB. Utility of in vitro drug metabolism data in predicting in vivo metabolic clearance. Biochem Pharmacol. 1994;47:1469–1479. [DOI] [PubMed] [Google Scholar]
- 5.Houston JB. Prediction of hepatic clearance from microsomes, hepatocyte and liver slices. Drug Metab Rev. 1997;29:891–922. [DOI] [PubMed] [Google Scholar]
- 6.Iwatsubo T, Hirota N, Ooie T, Suzuki H, Shimada N, Chiba K, Ishizaki T, Green CE, Tyson CA, Sugiyama Y. Prediction of in vivo drug metabolism in the human liver from in vitro metabolism data. Pharmacol Ther. 1997;73:147–171. [DOI] [PubMed] [Google Scholar]
- 7.Obach RS, Baxter JG, Liston TE, Silber BM, Jones BC, MacIntyre F, Rance DJ, Wastall P. The prediction of human pharmacokinetic parameters from preclinical and in vitro metabolism data. J Pharmacol Exp Ther. 1997;283:46–58. [PubMed] [Google Scholar]
- 8.Obach RS. Prediction of human clearance of twenty-nine drugs from hepatic microsomal intrinsic clearance data: an examination of in vitro half-life approach and nonspecific binding to microsomes. Drug Metab Dispos. 1999;27:1350–1359. [PubMed] [Google Scholar]
- 9.Bowman CM, Benet LZ. Hepatic clearance predictions from in vitro-in vivo extrapolation and the Biopharmaceutics Drug Disposition Classification System. Drug Metab Dispos. 2016;44:1731–1735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Wood FL, Houston JB, Hallifax D. Clearance prediction methodology needs fundamental improvement: Trends common to rat and human hepatocytes/microsomes and implications for experimental methodology. Drug Metab Dispos. 2017;45:1178–1188. [DOI] [PubMed] [Google Scholar]
- 11.Takano J, Maeda K, Bolger MB, Sugiyama Y. The prediction of the relative importance of CYP3A/P-gp to the nonlinear intestinal absorption of drugs by advanced compartmental absorption and transit model. Drug Metab Dispos. 2016;44:1808–1818. [DOI] [PubMed] [Google Scholar]
- 12.Chiba M, Ishii Y, Sugiyama Y. Prediction of hepatic clearance in human from in vitro data for successful drug development. AAPS J. 2009;2:262–276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wood FL, Houston JB, Hallifax D. Importance of the unstirred water layer and hepatocyte membrane integrity in vitro for quantification of intrinsic metabolic clearance. Drug Metab Dispos. 2018;46:268–278. [DOI] [PubMed] [Google Scholar]
- 14.Hallifax D, Houston JB. Evaluation of hepatic clearance prediction using in vitro data: emphasis on fraction unbound in plasma and drug ionization using a database of 107 drugs. J Pharm Sci. 2012;101:2645–2652. [DOI] [PubMed] [Google Scholar]
- 15.Riccardi KA, Tess DA, Lin J, Patel R, Ryu R, Atkinson K, Di L, Li R. A novel unified approach to predict human hepatic clearance from both enzyme- and transporter-mediated mechanisms using suspended human hepatocytes. Drug Metab Dispos. 2019;47:484–492. [DOI] [PubMed] [Google Scholar]
- 16.Rowland M, Benet LZ, Graham GG. Clearance concepts in pharmacokinetics. J Pharmacokinet Biopharm. 1973;1:123–135. [DOI] [PubMed] [Google Scholar]
- 17.Wilkinson GR, Shand DG. Commentary: a physiologic approach to hepatic drug clearance. Clin Pharmacol Ther. 1975;18:377–390. [DOI] [PubMed] [Google Scholar]
- 18.Rowland M, Tozer TN. Clinical Pharmacokinetics and Pharmacodynamics: Concepts and Applications. 4th Ed, 2010, Lippincott Williams & Wilkens. [Google Scholar]
- 19.Klotz U, Avant GR, Hoyumpa A, Schenker S, Wilkinson GR. The effects of age and liver disease on the disposition and elimination of diazepam in adult man. J Clin Invest. 1975;55:347–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Mangoni AA, Jackson SHD. Age-related changes in pharmacokinetics and pharmacodynamics: Basic principles and practical applications. Br J Clin Pharmacol. 2004;57:6–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wu C-Y, Benet LZ. Predicting drug disposition via application of BCS: Transport/absorption/elimination interplay and development of a Biopharmaceutics Drug Disposition Classification System. Pharm Res. 2005;22:11–23. [DOI] [PubMed] [Google Scholar]
- 22.Hallifax D, Houston JP. Use of segregated hepatocyte scaling factors and cross-species relationships to resolve clearance dependence in the prediction of human hepatic clearance. Drug Metab Dispos. 2019;47:320–327. [DOI] [PubMed] [Google Scholar]
- 23.Bowman CM, Benet LZ. Interlaboratory variability in human hepatocyte intrinsic clearance values and trends with physicochemical properties. Pharm Res. 2019;36:113. doi: 10.1007/s11095-019-2645-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bowman CM, Benet LZ. In vitro-in vivo extrapolation and hepatic clearance-dependent underprediction. J Pharm Sci. 2019;108:2500–2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bowman CM, Benet LZ. In vitro-in vivo inaccuracy: The CYP3A4 anomaly. Drug Metab Dispos. 2019;47:1368–1371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bowman CM, Benet LZ. An examination of protein binding and protein-facilitated uptake relating to in vitro-in vivo extrapolation. Eur J Pharm Sci. 2018;123:502–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bowman CM, Okochi H, Benet LZ. The presence of a transporter-induced protein binding shift: A new explanation for protein-facilitated uptake and improvement for in vitro-in vivo extrapolation. Drug Metab Dispos. 2019;47:358–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Benet LZ, Liu S, Wolfe AR. The universally unrecognized assumption in predicting drug clearance and organ extraction ratio. Clin Pharmacol Ther. 2018;103:521–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lombardo F, Berellini G, Obach RS. Trend analysis of a database of intravenous pharmacokinetic parameters in humans for 1352 drug compounds. Drug Metab Dispos. 2018;46:1466–1477. [DOI] [PubMed] [Google Scholar]
