+ All Categories
Home > Documents > Determination of Incubational Binding in In Vitro...

Determination of Incubational Binding in In Vitro...

Date post: 21-Mar-2018
Category:
Upload: lekhanh
View: 215 times
Download: 1 times
Share this document with a friend
15
DMD # 74138 1 Determination of Incubational Binding in In Vitro Microsomal and Hepatocyte Metabolic Stability Incubations: A Comparison of Methods Sofia Chen, Luna Prieto Garcia, Fredrik Bergström, Pär Nordell and Ken Grime Respiratory, Inflammation and Autoimmunity Department of DMPK, Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden (KG, SC, LPG) Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden (PN) Cardiovascular and Metabolic Diseases Department of DMPK, Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden (FB) This article has not been copyedited and formatted. The final version may differ from this version. DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138 at ASPET Journals on May 21, 2018 dmd.aspetjournals.org Downloaded from
Transcript
Page 1: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

1

Determination of Incubational Binding in In Vitro Microsomal and Hepatocyte Metabolic

Stability Incubations: A Comparison of Methods

Sofia Chen, Luna Prieto Garcia, Fredrik Bergström, Pär Nordell and Ken Grime

Respiratory, Inflammation and Autoimmunity Department of DMPK, Innovative Medicines and Early

Development, AstraZeneca, Gothenburg, Sweden (KG, SC, LPG)

Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca,

Gothenburg, Sweden (PN)

Cardiovascular and Metabolic Diseases Department of DMPK, Innovative Medicines and Early

Development, AstraZeneca, Gothenburg, Sweden (FB)

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 2: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

2

Running title

Fraction of Drug Unbound in in vitro Metabolic Stability Assays

Corresponding author: Ken Grime

AstraZeneca R&D, Gothenburg

SE 43183 Mölndal

Sweden

Tel: +46 (0)317761815

FAX: +46 (0)317762800

Email: [email protected]

Number of text pages: 15

Number of tables: 0

Number of figures: 3

Number of references: 17

Words in Abstract: 246

Words in Introduction: 397

Words in Results & Discussion: 962

Non-standard abbreviations used:

CLint,u Unbound metabolic intrinsic clearance

fuinc Fraction of unbound drug in incubation, assumed to be at 1 mg/mL microsomal

protein or 1 million hepatocytes/mL unless otherwise stated.

HLM Human liver microsomes

LogD7.4 A distribution-coefficient describing the ratio of compound concentration in octanol

and pH7.4 buffer when the test system is at equilibrium (lipophilicity measure)

RH Rat hepatocytes

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 3: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

3

Abstract

Fraction of drug unbound in an in vitro intrinsic clearance (CLint) incubation, fuinc, is an important

parameter in the pursuit of accurate clearance predictions and is often predicted using algorithms based

on drug lipophilicity measures. However, analysis of an AstraZeneca database suggests that simple

lipophilicity alone is a relatively poor predictor of fuinc measured using equilibrium dialysis. Fuinc can

also be measured directly in CLint assays using multiple concentrations of hepatocytes or microsomal

protein. Since this approach informs of the unbound drug concentration in the assay used to predict in

vivo clearance, it should be considered the gold standard method. As a starting point for building better

predictive algorithms we aimed to determine if equilibrium dialysis really is an appropriate assay for

assessing fuinc. Employing a large number of compounds with a wide range of lipophilicities,

experiments were performed to measure fuinc using rat hepatocytes (RH) and human liver microsomes

(HLM) in both assay formats. A high percentage (94% and 93% for HLM and RH respectively) of the

fuinc values were within 2-fold when compound logD7.4 values were less than 3.5. However, with logD7.4

values greater than these, the agreement was considerably worse. Additional experimental data

generated indicated that this discrepancy was likely due to failings in the direct method when drug

binding is high. Thus we conclude that unbound CLint can be indeed calculated indirectly by

incorporating equilibrium dialysis data with measured CLint but that simple lipophilicity descriptors

alone may be inadequate for predicting fuinc.

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 4: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

4

Introduction

Prediction of in vivo hepatic metabolic clearance from in vitro data uses measured hepatocyte or

microsomal CLint, fuinc and fraction of drug unbound in blood (fub) (Obach, 1996, Ito and Houston, 2005).

To add confidence to human clearance predictions for any given candidate drug, observed unbound in

vivo CLint should first be predicted to within 2-fold in two preclinical species (Grime et al. 2013). To

meet such stringent criteria, attention to fine detail on experimental conditions and physiological

relevance of the in vitro measurements is paramount. In a Drug Discovery setting, fuinc is typically

measured using equilibrium dialysis in which partitioning of drug between two chambers either side of

the dialysis membrane, one holding inactivated microsomes or hepatocytes and one holding buffer, is

determined. This assay allows a high-throughput of compounds but does not necessarily represent the

conditions experienced by a drug at the site of drug metabolism in a CLint assay and thus assessment of

unbound CLint directly in the metabolic stability assay may be viewed as the ideal. With this in mind, an

approach for determining fuinc using metabolic stability data generated at multiple concentrations of

hepatocytes or microsomal protein can be used, since the rate at which a compound is metabolized is

proportional to the unbound concentration available to enzymes (Giuliano et al., 2005, Grime and Riley,

2006, Nordell et al., 2013). The pharmaceutical industry is under great pressure to reduce costs and as

such it is an appropriate aim to minimise wet screening where possible. As such fuinc in silico prediction

algorithms, based on ion class and lipophilicity (logP or logD7.4 octanol/water partition coefficients,

Austin et al., 2002, Austin et al., 2005, Kilford et al., 2008) as well as other molecular descriptors (Gao

et al., 2008, Gao et al., 2010, Abraham and Austin, 2012; Nair et al., 2016), have become widely used.

On review of an in-house database, we noted that simple lipophilicity based predictions were within

two-fold of equilibrium dialysis measured values for only 64% (HLM) and 62% (RH) of compounds

(Figure 1). As a starting point for building better fuinc predictive algorithms we considered the validity

of equilibrium dialysis measures and compared HLM and RH fuinc values generated by this approach

and directly from the CLint assays. The data sets cover 58 and 74 AstraZeneca proprietary compounds

for HLM and RH, respectively, spanning over five orders of magnitude in measured lipophilicity.

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 5: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

5

Material and Methods

Materials

UltraPool human liver microsomes (mixed gender, lot no 38289) were purchased from BD Gentest

(USA) and cryopreserved Han Wistar rat hepatocytes (mixed gender, lots RJQ and RAL) were

purchased Bioreclamation IVT (Belgium). L-15 Leibovitz medium and phosphate buffers were obtained

from Life technologies (NY, USA). NADPH was obtained from Sigma (USA). Compound stock

solutions DMSO (10 mM) were obtained from the AstraZeneca Compound Management.

Measurement of LogD7.4 and Determination of fuinc by equilibrium dialysis

LogD7.4 was measured as previously described (Austin et al., 2002). Equilibrium dialysis was performed

to determine extent of compound binding to HLM (1mg/mL in 0.1M, pH 7.4 phosphate buffer) and

freshly isolated RH (1 million cells/mL in Leibovitz media). Compound stocks (100 μM in DMSO)

were added at a ratio of 10 L / mL HLM or RH suspensions. Inactivation of drug metabolising enzymes

was performed in accordance with previous studies (Austin et al., 2005) using 1-aminobenzotriazole

(400 mM in DMSO) and salicylamide (300 mM in DMSO), added to the preparations at a ratio of 2.5

L/mL and 5 μL/mL respectively, followed by 1 h of pre-incubation at 37C. Samples were checked for

drug metabolism by analysing parent drug pre- and post- dialysis experiments. Experiments were

performed in triplicate. Dialysis membranes were soaked in ultrapure water for 60 min, in 20% ethanol

for 20 min and finally in phosphate buffer (0.1 M, pH 7.4) for 20 min prior to loading into 96-well

dialysis devices (HTDialysis LLC, Gales Ferry, CT). An aliquot (150 μL) of HLM spiked with test

compound (1 μM) and drug metabolism inhibitors was added to donor wells of the dialysis device with

phosphate buffer (150 μL, 0.1M, pH 7.4) added to acceptor wells. The dialysis device was loaded onto

a rotor housed within an air bath. After 4 hours of incubation (37C, 300 rpm), aliquots (50 μL) from

each donor and acceptor well were transferred to a 96-deep well plate. Samples were quenched with

acetoniltrile (100 µL), diluted, analyzed by LC-MS/MS and quantified from a standard curve. Free

fraction of drug was determined as previously described (Austin et al., 2002). RH were isolated from

adult male Sprague-Dawley rats (250 - 300 g) as described previously (Kenny and Grime, 2006) and

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 6: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

6

resuspended in Leibovitz media at 1 million cells/mL. Leibovitz media (150 μL) was added to the

acceptor chamber and hepatocyte suspension (150 μL) containing compound (1 μM) and drug

metabolising enzyme inhibitors was added to the donor chamber. Dialysis procedure, as above. All LC-

MS/MS analyses were performed on a Waters triple quadrupole mass spectrometer, coupled to a liquid

chromatography sample and solvent manager system, as described previously (Nordell et al., 2013). In

order to investigate if non-linear drug binding, with respect to concentration of enzyme, was likely to

influence the estimation of fuinc, equilibrium dialysis experiments were performed as described

above, at HLM concentrations of 0.125, 0.5, 1 and 4 mg/mL for 25 compounds.

Metabolic stability at multiple microsomal protein or hepatocyte concentrations

CLint was determined in duplicate for each of the HLM or RH concentrations used. HLM stock solutions

were thawed and diluted in phosphate buffer (0.1M, pH 7.4) containing NADPH (1 mM) to the

following protein concentrations: 0.14, 0.21, 0.28, 0.41, 0.56, 0.83, 1.11, 2.22, 4.44 mg protein/mL.

Suspensions were mixed with compound stock solutions (50 M in 50% acetonitrile) to a final

concentration of 0.5 M and incubated in a 96-well plate at 37°C with plate shaking. Reactions were

terminated at 0.5, 5, 10, 15, 20 and 30 minutes by quenching with cold acetronitrile containing 0.1%

formic acid and subsequently analyzed by LC-MS/MS. RH were suspended in Leibovitz media to the

following cell concentrations: 0.13, 0.19, 0.25, 0.38, 0.50, 0.75, 1.0, 2.0 and 4.0 million cells/mL. Cell

suspensions were mixed with compound solutions (50 M in 50% acetonitrile) to a final concentration

of 0.5 M and incubated in 96-well plate at 37°C with plate shaking. Reactions were terminated (0.5, 5,

15, 20, 30, 45, 60, 80, 100 and 120 minutes) and analyzed by as described above.

Model of incubational binding

Incubational binding, treated as an equilibrium between drug in aqueous and the membrane phases, is

described by a partitioning constant Kp which is related to the fraction unbound (fuinc): Kp = drug bound

to the membrane phase/unbound in the aqueous phase = (1 – fuinc / fuinc) (Austin et al., 2002). Since only

unbound drug is available to metabolizing enzymes, measured intrinsic metabolic clearance (CLint)

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 7: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

7

becomes a function of the microsomal or hepatocyte concentration, related to the unbound intrinsic

CLint,u: CLint = fuinc x CLint,u. The fraction unbound at standard conditions (microsomal protein or

hepatocyte concentration of 1 mg/mL or 1 million cells/mL, respectively), fuinc,0, is related to the fraction

unbound and CLint at a second enzyme concentration, allowing definition of the true unbound intrinsic

clearance, CLint,u (where CLint represents observed intrinsic clearance at any enzyme concentration C).

𝐶𝐿𝑖𝑛𝑡 =𝐶𝐿𝑖𝑛𝑡,𝑈

1 +𝐶𝐶0

×1 − 𝑓𝑢𝑖𝑛𝑐,0𝑓𝑢𝑖𝑛𝑐,0

Eq. 1

Direct determination of fuinc from in vitro depletion curves

Analytical peak areas of samples, withdrawn from drug incubations at the various HLM or RH

concentrations, were loge-transformed (as exemplified in Figure 2a) and elimination rate constants k

(min-1) were derived from the slopes of the loge[substrate]-time plots. Since CLint,= k x V, where V

represents the incubation volume (mL/mg protein or mL/million cells), the entire dataset from multiple

microsomal protein or hepatocyte concentrations was used to define fuinc and CLint,U (from Eq. 1) as

follows: Using nonlinear least squares solver lsqnonlin in Matlab 2014b (The MathWorks Inc., Natick,

MA), the sum-of-squares of the vertical distances between loge-transformed percentage substrate

remaining and the linear best fit, at all HLM or RH concentrations, was minimized and the intercept

(from the family of CLint values analyzed with their associated microsomal protein or hepatocyte

concentrations, Figure 2b) was allowed to vary freely for optimal fit. Prior to summation, squared

residuals were weighted to account for number of data points included at each enzyme concentration.

Predicted in vivo rat clearance

Clearance predictions were made using a ‘regression line approach’, whereby an existing in vitro-in vivo

unbound CLint dataset (for which in vivo CLint values represent metabolic clearance only) is used as a

framework for predicting the in vivo clearance for novel compounds (Grime and Riley, 2006; Sohlenius-

Sternbeck et al., 2012).

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 8: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

8

Results and Discussion

With ionic class and lipophilicity being identified as main drivers of incubational binding (Austin et al.,

2002, Austin et al., 2005, Kilford et al., 2008) a set of compounds was selected from the AstraZeneca

compound library comprising acids (A), bases (B) and neutrals (N) that each covered a broad range of

logD7.4 values. The fraction unbound was determined in triplicate by equilibrium dialysis. For HLM, 58

(A: 14, B: 20, N: 22) compounds spanning logD7.4 values of 0.5 - 5.3 (A: 1.9 - 4.2, B: 0.5 - 4.8, N: 0.2 -

5.3) had measured fuinc (at 1 mg/mL HLM protein) values of 0.001 - 0.95 (A: 0.02 - 0.75, B: 0.001 -

0.87, N: 0.001 - 0.95) defined. For RH, 74 (A: 21, B: 20, N: 20) compounds spanning logD7.4 values of

-0.3 - 5.3 (A: -0.4 - 4.5, B: -0.3 - 4.1, N: 0.9 - 5.3) had fuinc (at 1 million cells/mL), values of 0.004 -

0.996 (A: 0.009 - 0.71, B: 0.03 - 0.91, N: 0.004 - 0.995) defined. Fuinc was also determined directly from

the CLint assays as described. In the RH equilibrium dialysis experiments, 1-aminobenzotriazole was

used as a CYP inhibitor. Although a very effective CYP3A4 inhibitor, other enzymes may be less

effectively inhibited including CYP2C9, for which over 60% of activity may remain using 1 mM and a

30 minute pre-incubation (Linder et al., 2009). This was a compromise to avoid the use of several

individual CYP inhibitors designed to remove all CYP activity, but at a cost of greater organic solvent

addition. The pre-incubation was 60 minutes and since the HLM equilibrium dialysis incubations

contained no NADPH and the results were in line with the RH experimental data, it is apparent that the

use of 1-aminobenzotriazole as a CYP inhibitor did not compromise the results. In confirmation of this,

biotransformation assessment of all equilibrium dialysis incubations revealed no drug metabolism in the

RH incubates. The DMSO concentration of 1% v/v used in all metabolic stability incubations may give

rise to sub-optimal rate of metabolism for some drug metabolizing enzymes. However, since the relative

contribution of individual CYPs to the overall CLint is not relevant in determining fuinc by equation 1,

this compromise was made to ensure the solubility of all compounds since many were relatively

lipophilic.

Figure 3 shows a log-scale representation of the agreement between incubational binding estimates

given by the two assays for acids, bases and neutrals in RH and HLM incubations. Only 65% (HLM)

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 9: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

9

and 77% (RH) of the fuinc values from the direct assay fell within 2-fold of the values provided by the

equilibrium dialysis assay. However, 94 and 93% (HLM and RH respectively) of the fuinc values were

within 2-fold when compound logD7.4 values were less than 3.5. Although a somewhat arbitrary cut-off,

it allows some quantification of the lipophilicity effect. When considering compounds with logD7.4 >

3.5 only 25% of HLM and 27% of RH fuinc values were within 2-fold. For these compounds (logD7.4 >

3.5), there was significant bias (p = 110-4 and 710-4 for HLM and RH, respectively), with fuinc values

determined by equilibrium dialysis consistently being lower than estimates from direct assessment using

substrate depletion data. Compounds with logD7.4 < 3.5 showed no indication of bias. Using HLM

concentrations of 0.125, 0.5, 1 and 4 mg protein/mL in equilibrium dialysis experiments, we confirmed

that non-linear drug binding, with respect to HLM concentration, was not an influencing factor in the

difference in fuinc values from the two assay formats: For 25 compounds, equilibrium dialysis

incubational binding increased proportionally with increasing HLM concentration (data not shown).

Overall the analysis indicates that more lipophilic compounds may have a strong bias towards

equilibrium dialysis yielding lower fuinc estimates than the direct approach, independent of whether

whole cells or microsomes are studied. In line with this, Guiliano et al. (2005) identified two outliers,

both extensively bound, deviating from the otherwise good correlation between equilibrium dialysis and

metabolic stability based fuinc when investigating HLM-drug binding behaviour. A possible explanation

for these observations is that when incubational binding is high, the rapidly changing CLint-enzyme

concentration curve (Figure 2b) is not sufficiently well-described by the input data at low hepatocyte or

microsomal protein concentrations to allow appropriate back extrapolation to the true CLint,u value (Y-

axis intercept), making fuinc direct from CLint approach less reliable for more lipophilic compounds.

In order to substantiate the idea that, for molecules with higher hepatocyte binding, equilibrium dialysis

gives a more accurate description of binding, we investigated the in vivo rat clearance of two of the

compounds with a big discrepancy in fuinc from the two assays. Using the previously described method

for predicting clearance at AstraZeneca with input rat hepatocyte CLint, fuinc and plasma protein binding

data (Sohlenius-Sternbeck et al., 2012), the clearance predictions were 37 & 1 mL/min/kg when using

fuinc determined from the equilibrium dialysis method and the direct from CLint approach respectively

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 10: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

10

for the first compound. The observed rat plasma clearance was 65 mL/min/kg. Similarly, for the second

compound, the predicted clearance values were 48 and 10 mL/min/kg using equilibrium dialysis and

direct method fuinc respectively, whilst the observed clearance was 51 mL/min/kg. Although such an

assessment is not completely robust since there can be other explanations for poor clearance prediction,

this evidence does point to the greater general utility of equilibrium binding data for making accurate

clearance predictions. This study supports the long-held assumption that unbound CLint, required for

predicting in vivo hepatic metabolic clearance, can indeed be calculated indirectly using equilibrium

dialysis data and CLint taken under standard conditions. Additionally, our analysis of an in-house data

base indicates that for prediction of incubational binding requires more sophisticated prediction

approaches than simple lipophilicty algorithms, as discussed elsewhere (Gao et al., 2010; Nair et al.,

2016).

Authorship contributions

Participated in research design: Grime, Nordell, Bergström, Chen, Prieto Garcia.

Conducted experiments: Chen and Prieto Garcia

Performed data analysis: Chen, Prieto Garcia, Bergström and Nordell

Wrote the manuscript: Grime and Nordell

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 11: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

11

References

Abraham MH and Austin RP. (2012) The effect of ionized species on microsomal binding. Eur J Med

Chem 47:202-205.

Austin RP, Barton P, Mohmed S, and Riley RJ. (2005) The binding of drugs to hepatocytes and its

relationship to physicochemical properties. Drug Metab Disposition 33:419-425.

Austin RP, Barton P, Cockroft SL, Wenlock MC, and Riley RJ. (2002) The influence of nonspecific

microsomal binding on apparent intrinsic clearance, and its prediction from physicochemical

properties. Drug Metab Disposition 30:1497-1503.

Gao H, Steyn SJ, Chang G, and Lin J. (2010) Assessment of in silico models for fraction of unbound

drug in human liver microsomes. Expert Opin Drug Metab Toxicol 6:533-542.

Gao H, Yao L, Mathieu HW, Zhang Y, Maurer TS, Troutman MD, Scott DO, Ruggeri RB, and Lin J.

(2008) In silico modeling of nonspecific binding to human liver microsomes. Drug Metab Dispos

36:2130-2135.

Giuliano C, Jairaj M, Zafiu CM, and Laufer R. (2005) Direct determination of unbound intrinsic drug

clearance in the microsomal stability assay. Drug Metab Disposition 33:1319-1324.

Grime K and Riley RJ. (2006) The impact of in vitro binding on in vitro - In vivo extrapolations,

projections of metabolic clearance and clinical drug-drug interactions. Curr Drug Metab 7:251-264.

Hallifax D and Houston JB. (2007) Saturable uptake of lipophilic amine drugs into isolated

hepatocytes: Mechanisms and consequences for quantitative clearance prediction. Drug Metab

Disposition 35:1325-1332.

Ito K and Houston JB. (2005) Prediction of human drug clearance from in vitro and preclinical data

using physiologically based and empirical approaches. Pharm Res 22:103-112.

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 12: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

12

Kenny JR and Grime K. (2006) Pharmacokinetic consequences of time-dependent inhibition using the

isolated perfused rat liver model. Xenobiotica 36:351-365.

Kilford PJ, Gertz M, Houston JB, and Galetin A. (2008) Hepatocellular binding of drugs: Correction

for unbound fraction in hepatocyte incubations using microsomal binding or drug lipophilicity data.

Drug Metab Disposition 36:1194-1197.

Linder CD, Renaud NA and Hutzler JM (2009) Is 1-aminobenzotriazole an appropriate in vitro tool as

a nonspecific cytochrome P450 inactivator? Drug Metab Disposition 37:10-13.

Nair PC, McKinnon RA and Miners JO (2016) A fragment based approach for the computational

prediction of the nonspecific binding of drugs to hepatic microsomes. Drug Metab Disposition Fast

Forward, DOI: 10.1124/dmd.116.071852

Nordell P, Svanberg P, Bird J, and Grime K. (2013) Predicting Metabolic Clearance for Drugs That

Are Actively Transported into Hepatocytes: Incubational Binding as a Consequence of in Vitro

Hepatocyte Concentration Is a Key Factor. Drug Metabolism and Disposition 41:836-843.

Obach RS. (1996) The importance of nonspecific binding in in vitro matrices, its impact on enzyme

kinetic studies of drug metabolism reactions, and implications for in vitro-in vivo correlations. Drug

Metab Disposition 24:1047-1049.

Smith DA, Di L, and Kerns EH. (2010) The effect of plasma protein binding on in vivo efficacy:

Misconceptions in drug discovery. Nature Reviews Drug Discovery 9:929-939.

Sohlenius-Sternbeck A-K, Jones C, Ferguson D, Middleton BJ, Projean D, FlobyE, Bylund, J,

Afzelius L, (2012) Use of the regression offset approach for the prediction of in vivo intrinsic

clearance from hepatocytes. Xenobiotica 42: 841−853.

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 13: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

13

Figure legends

Figure 1. Human liver microsomal (A) and rat hepatocyte (B) incubational binding: Comparison of

lipophilicity based predicted (Austin et al 2002; Austin et al., 2005) and measured (equilibrium

dialysis) for acidic compounds (upward red triangle), bases (downward blue triangle), neutrals (green

circles) and zwitterions (orange squares). Lines depict unity (solid) and two-fold of unity (dashed).

The number of compounds represented: 512 (RH) and 544 (HLM) of which 319 and 347 respectively

are within 2-fold (observed/predicted).

Figure 2. Principle of data analysis for fuinc determined directly from the CLint assay (RH data shown

for compound 1 in Figure 3). A: loge-transformed percentage of drug remaining for incubations run

between 4 (top-left) and 0.125 (bottom-right) million cells/mL. B: effect of hepatocyte concentration on

CLint. While fuinc reflects the curvature, CLint,U is given by the intercept with the Y-axis according to

equation 1 (Methods). Circles indicate hepatocyte concentration at which incubations were performed.

Figure 3. Figure 3. Comparison between fuinc determined by equilibrium dialysis and directly from

substrate depletion CLint data for HLM (A) and RH (B) incubations. Acidic compounds (upward red

triagles), bases (downward blue triangles), neutrals (green circles). In 3B the solid green circle is

compound 1 (see Figure 2). Comparison between observed and predicted plasma clearance were made

for two compounds indicated by solid red upward triangles. C: Comparison of the fuinc methods

(substrate depletion fuinc / equilibrium dialysis fuinc) with increasing logD7.4 for HLM (upward red

triangles) and RH (downward blue triangles) data. The horizontal lines indicate unity and two-fold of

unity (dashed), the vertical line indicates logD7.4 > 3.5 threshold.

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 14: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

14

Figure 1:

Figure 2a and 2b

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from

Page 15: Determination of Incubational Binding in In Vitro ...dmd.aspetjournals.org/content/dmd/early/2017/01/25/dmd.116.074138... · Determination of Incubational Binding in In Vitro Microsomal

DMD # 74138

15

Figure 3

This article has not been copyedited and formatted. The final version may differ from this version.DMD Fast Forward. Published on January 25, 2017 as DOI: 10.1124/dmd.116.074138

at ASPE

T Journals on M

ay 21, 2018dm

d.aspetjournals.orgD

ownloaded from


Recommended