throbber
‘6ZKB]/\[I10S['8U11'10[1123.I0‘S[ELI.II’l0I18dSB'pIJ.Ip{I101}PSPEOIIIAAOG
`
`
`
`
`
`
`
`0090-9556/99/271l—l350—l359$02.00/0
`DRUG METABOLISM AND DISPOSITION
`Copyright © 1999 by The American Society for Pharmacology and Experimental Therapeutics
`
`Vol. 27, No. 11
`Printed in U.S.A.
`
`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
`
`R. SCOTT OBACH
`
`Drug Metabolism Department, Candidate Synthesis, Enhancement, and Evaluation, Central Research Division, Pfizer, Inc., Groton, Connecticut
`
`(Received April 9, 1999; accepted July 30, 1999)
`
`This paper is available online at http://www.dmd.org
`
`ABSTRACT:
`
`Twenty-nine drugs of disparate structures and physicochemical
`properties were used in an examination of the capability of human
`liver microsomal Iability data (“in vitro T,,2" approach) to be useful
`in the prediction of human clearance. Additionally, the potential
`importance of nonspecific binding to microsomes in the in vitro
`incubation milieu for the accurate prediction of human clearance
`was investigated. The compounds examined demonstrated a wide
`range of microsomal metabolic labilities with scaled intrinsic clear-
`ance values ranging from less than 0.5 ml/min/kg to 189 ml/min/kg.
`Microsomal binding was determined at microsomal protein con-
`centrations used in the Iability incubations. For the 29 compounds
`studied, unbound fractions in microsomes ranged from 0.11 to 1.0.
`Generally, basic compounds demonstrated the greatest extent of
`binding and neutral and acidic compounds the least extent of
`
`binding. In the projection of human clearance values, basic and
`neutral compounds were well predicted when all binding consid-
`erations (blood and microsome) were disregarded, however, in-
`cluding both binding considerations also yielded reasonable pre-
`dictions. Including only blood binding yielded very poor projections
`of human clearance for these two types of compounds. However,
`for acidic compounds, disregarding all binding considerations
`yielded poor predictions of human clearance. It was generally most
`difficult to accurately predict clearance for this class of com-
`pounds; however the accuracy was best when all binding consid-
`erations were included. Overall, inclusion of both blood and micro-
`some binding values gave the best agreement between in vivo
`clearance values and clearance values projected from in vitro
`intrinsic clearance data.
`
`The use of in vitro drug metabolism data in the understanding of in
`vivo pharmacokinetic data has recently become an area of scientific
`interest (Houston, 1994; Houston and Carlile, 1997; Iwatsubo et al.,
`1997). This has partially stemmed from a trend in the pharmaceutical
`industry to use in vitro drug metabolism data, using human—derived
`reagents, as a criterion to select compounds for further development
`(Rodrigues, 1997). Thus, in vitro metabolism data is used in a pro-
`spective manner to choose those compounds for further development
`that are expected to possess commercially acceptable pharmacokinetic
`properties (e.g., half-life permitting once-per—day administration reg-
`imens, low oral clearance to reduce dose, etc.). Several investigators
`have recently described methods whereby preclinical drug metabo-
`lism and pharmacokinetic data can be used to predict human pharma-
`cokinetic parameters (Obach et al., 1997; Lave et al., 1997a,b; Mah-
`mood, l998a,b).
`The first demonstration of the correlation between in vivo clearance
`values and clearance values calculated from liver microsomal metab-
`
`olism intrinsic clearance data was made by Rane et al. (1977) for the
`rat. Intrinsic clearance data were obtained by determination of the
`enzyme kinetic parameters (Vmax and KM). In our work, we described
`two related methods whereby human clearance could be predicted
`from in vitro metabolism data (Obach et al., 1997). In one method, the
`
`
`
`enzyme kinetic parameters Vmax and KM were determined and con-
`verted to intrinsic clearance (CL',,,,)1, which is similar to that de-
`scribed by Rane et al. (1977). In the other method, referred to as the
`“in vitro T1,; method”, CL’,m was determined by measuring the
`first-order rate constant for consumption of the substrate at a low
`concentration. Interestingly, for both of these methods, a better cor-
`relation was observed between the actual and predicted clearance
`values if the free fraction in blood was disregarded in the well-stirred
`or parallel-tube equations describing hepatic extraction.
`One possible reason for the observation that a better prediction of
`human clearance was made when disregarding plasma protein binding
`was that the substrates were bound in the microsomal incubations, and
`that the extent of this binding could be great enough so as to almost
`cancel out the plasma protein binding term in the well-stirred and
`parallel-tube equations (Obach, 1996). This possibility was further
`substantiated in an examination of probe substrates propranolol, imip-
`ramine, and warfarin (Obach, 1997). In this report, it was demon-
`strated that the lipophilic amines propranolol and imipramine were
`bound to microsomes, and that incorporation of this binding term
`aided in the accurate prediction of human clearance from in vitro
`intrinsic clearance data. The acidic drug, warfarin, exhibited this
`phenomenon to a much lesser extent. However, for all three drugs
`overall, incorporation of both plasma protein and microsome binding
`
`Send reprint requests to: R. Scott Obach, Ph.D., Drug Metabolism Depart-
`ment, Candidate Synthesis, Enhancement, and Evaluation, Central Research
`Division, Pfizer, lnc., Groton, CT 06340. E-mail: obachr@pfizer.com
`
`1 Abbreviations used are: CL',,,,, intrinsic clearance; f,,(,,,,C,, unbound fraction in
`microsomal incubation mixtures; f,,(b.,,,,,,,, unbound traction in blood; Q, hepatic
`blood flow; ISTD, internal standard.
`1350
`
`WOCK- EXHIBIT 1024
`
`Z 9 [
`
`-1
`27:‘
`
`O a
`
`t E ,
`
`0
`L23
`
`2V
`
`J
`
`1'3
`
`0 i
`
`t
`
`+-
`H-1
`
`2 Ba
`
`t D
`
`

`
`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on May 29, 2015
`
`MICROSOME BINDING IMPACT ON CLEARANCE PREDICTIONS
`
`1351
`
`FIG. 1.Chemical structures of the 29 drugs examined in this study.
`
`terms generally yielded more accurate predictions of human clear-
`ance.
`The objective of the experiments described herein is to more
`exhaustively test the hypothesis that microsomal binding is an impor-
`tant phenomenon in the prediction of in vivo pharmacokinetics from
`in vitro drug metabolism data. To this end, human hepatic microsomal
`metabolism data were gathered for 29 drugs, using the in vitro T1/2
`
`approach. Additionally, the extent of nonspecific binding to micro-
`somes in the in vitro matrix was measured for each drug. The drugs
`used in these experiments span a broad range of structural types (Fig.
`1) and include basic compounds (positively charged at pH 7.5), acidic
`compounds (negatively charged at pH 7.5), and neutral compounds
`(no charge at pH 7.5). The data set was used to project human
`clearance from the in vitro intrinsic clearance data to determine
`
`

`
`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on May 29, 2015
`
`1352
`
`OBACH
`
`TABLE 1
`
`Sample processing and HPLC-MS conditions for 29 drugs used in this analysis
`
`Drug
`
`Internal Standard
`
`Incubation
`Termination
`
`Mobile Phase
`System
`
`CH3CN
`
`MS
`Polarity
`
`Basic compounds
`Chlorpromazine
`Propafenone
`Verapamil
`Diphenhydramine
`Lorcainide
`Diltiazem
`Amitriptyline
`Desipramine
`Imipramine
`Ketamine
`Quinidine
`Clozapine
`Neutral compounds
`Dexamethasone
`Prednisone
`Diazepam
`Midazolam
`Methoxsalen
`Alprazolam
`Triazolam
`Zolpidem
`Acidic compounds
`Diclofenac
`Ibuprofen
`Tolbutamide
`Warfarin
`Tenidap
`Tenoxicam
`Amobarbital
`Hexobarbital
`Methohexital
`
`Amitriptyline
`Verapamil
`Propafenone
`Propafenone
`Propafenone
`Propafenone
`Imipramine
`Amitriptyline
`Amitriptyline
`Metoprolol
`Ondansetron
`Diltiazem
`
`Prednisone
`Dexamethasone
`Midazolam
`Diazepam
`Diazepam
`Triazolam
`Alprazolam
`Quinine
`
`Ibuprofen
`Diclofenac
`Warfarin
`Tolbutamide
`Warfarin
`Piroxicam
`Methohexital
`Methohexital
`Amobarbital
`
`NaOH
`NaOH
`NaOH
`NaOH
`NaOH
`NaOH
`NaOH
`NaOH
`NaOH
`NaOH
`NaOH
`NaOH
`
`NaOH
`NaOH
`NaOH
`NaOH
`CH3CN
`NaOH
`NaOH
`NaOH
`
`HCl
`HCl
`HCl
`HCl
`HCl
`HCl
`HCl
`HCl
`HCl
`
`1
`1
`1
`1
`1
`1
`1
`1
`1
`1
`1
`1
`
`1
`1
`1
`1
`1
`1
`1
`1
`
`2
`2
`2
`2
`2
`2
`2
`2
`2
`
`%
`
`36.5
`32.0
`32.0
`32.0
`32.0
`32.0
`36.5
`36.5
`36.5
`18.5
`18.5
`27.5
`
`32.0
`32.0
`50.0
`50.0
`50.0
`41.0
`41.0
`23.0
`
`32.0
`32.0
`27.5
`27.5
`32.0
`27.5
`45.5
`45.5
`45.5
`
`1
`1
`1
`1
`1
`1
`1
`1
`1
`1
`1
`1
`
`1
`1
`1
`1
`1
`1
`1
`1
`
`2
`2
`2
`2
`2
`2
`2
`2
`2
`
`m/z
`
`318.8
`341.9
`455.1
`256.0
`371.0
`415.0
`278.0
`266.5
`281.0
`237.8
`325.0
`326.9
`
`393.1
`359.1
`284.9
`325.8
`217.0
`309.0
`342.9
`308.0
`
`294.0
`205.1
`269.0
`307.3
`319.1
`336.1
`225.2
`235.1
`261.1
`
`Rt
`
`min
`
`1.2
`1.4
`1.6
`0.8
`1.4
`1.0
`1.0
`0.8
`1.0
`0.8
`1.5
`1.2
`
`1.8
`1.1
`1.4
`0.8
`1.0
`0.9
`1.0
`1.5
`
`1.1
`1.3
`1.2
`1.2
`1.3
`0.8
`0.8
`0.8
`1.5
`
`whether the most accurate projections are made by disregarding all
`binding data, including only blood binding values, or including both
`blood and microsomal binding values.
`
`Experimental Procedures
`
`Materials. The 29 drugs examined in these experiments were obtained from
`Sigma Chemical Co. (St. Louis, MO) with the exception of lorcainide (ob-
`tained from ICN, Aurora, OH), methoxsalen (obtained from Aldrich Chemical,
`Milwaukee, WI), zolpidem (obtained from Research Biochemicals Interna-
`tional, Natick, MA), and methohexital (obtained from Radian Inc., Dallas,
`TX). NADPH was obtained from Sigma. Solvents and other reagents were
`from common sources and were of HPLC grade or better. Human liver
`microsomes were from an in-house bank of liver microsomes maintained at
`Pfizer Central Research (Groton, CT). A pool was prepared from six liver
`microsomal preparations from six individual donors that were selected on the
`basis of having average activities for five of the major drug metabolizing
`cytochrome P-450 (CYP) enzymes (CYP1A2, CYP2C9, CYP2C19, CYP2D6,
`and CYP3A) normalized per microsomal protein content. Microsomes from
`putative CYP2D6 and CYP2C19 poor metabolizers were excluded. The P-450
`content of this pool, as determined by spectral means (Omura and Sato, 1964)
`was 0.26 nmol/mg microsomal protein. CYP isoform specific marker substrate
`activities were as follows: CYP1A2, phenacetin O-deethylase of 0.147 nmol/
`min/mg protein (at 50 mM phenacetin); CYP2C9, tolbutamide 4-hydroxylase
`of 0.23 nmol/min/mg protein (at 1.0 mM tolbutamide); CYP2C19, S-mephe-
`nytoin 49-hydroxylase of 0.093 nmol/min/mg protein (at 1.0 mM S-mepheny-
`toin); CYP2D6, bufuralol 19-hydroxylase of 0.075 nmol/min/mg protein (at 10
`mM bufuralol); and CYP3A4,
`testosterone 6b-hydroxylase of 2.7 nmol/
`min/mg protein (at 250 mM testosterone). All glassware was subjected to gas
`phase silylation before use.
`Metabolic Incubations. Human liver microsomal incubations were con-
`ducted in triplicate. General conditions are described as follows with details
`specific to each drug listed in Table 1. Incubation mixtures consisted of liver
`microsomes (0.3–10 mg microsomal protein/ml), substrates (1.0 mM), MgCl2
`
`(3.3 mM), and NADPH (1.3 mM) in a total volume of 0.5 ml potassium
`phosphate buffer (25 mM, pH 7.5). Reactions were commenced with the
`addition of NADPH and shaken in a water bath open to the air at 37°C. At T 5
`0 and at five time points ranging to 40 min, aliquots (50 ml) were removed and
`added to termination mixtures containing internal standards as listed in Table
`1. The samples were processed by extraction into methy t-butyl ether (3 ml),
`the aqueous layer was frozen in a dry ice-acetone bath, the organic solvent was
`decanted and evaporated under N2 at 30°C. The residue was reconstituted in 50
`ml HPLC mobile phase A (see below). For methoxsalen samples, the work-up
`procedure consisted of precipitation of protein with CH3CN (100 ml), removal
`of precipitated materials by centrifugation, and analysis of the supernatant by
`HPLC-mass spectrometry (MS).
`Equilibrium Dialysis. Drugs (1.0 mM) were mixed with human liver
`microsomes (at protein concentrations used for the respective metabolic incu-
`bations), MgCl2 (3.3 mM) and potassium phosphate buffer (25 mM; pH 7.5).
`The mixtures were subjected to equilibrium dialysis versus buffer/MgCl2 at
`37°C using a Spectrum apparatus (Spectrum Industries, Los Angeles, CA) as
`per instructions of the manufacturer. Spectra-Por no. 4 membranes, with
`molecular mass cutoff of 12 to 14 kDa, were used and the cells were rotated
`at 20 rpm for 5 h. (These dialysis conditions had been previously shown to give
`equilibrium for this dialysis apparatus; Obach, 1997). Dialysis experiments
`were done in triplicate. On completion of the dialysis period, the microsome
`and buffer samples were removed, processed as described above, and analyzed
`by HPLC-MS. Microsome samples (50 ml) were mixed with control buffer
`(100 ml), and buffer samples (100 ml) were mixed with control microsomes (50
`ml) to yield an identical matrix before sample work-up. Drug recovery through
`the dialysis procedure was determined by analyzing samples of the mixtures
`that were not subjected to dialysis, and recovery values were 86% or greater.
`HPLC-MS Analysis. The HPLC-MS system consisted of a Hewlett-
`Packard 1100 quaternary gradient HPLC pump with membrane degasser
`(Hewlett-Packard, Palo Alto, CA), a CTC PAL autoinjector (Leap Technolo-
`gies, Carrboro, NC), and a PE-Sciex API 100 single quadrupole mass spec-
`
`

`
`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on May 29, 2015
`
`MICROSOME BINDING IMPACT ON CLEARANCE PREDICTIONS
`
`1353
`
`Values for systemic clearance, fraction unbound in plasma, and blood-to-plasma ratio for 29 drugs examined in this analysis
`
`TABLE 2
`
`Drug
`
`Fraction Unbound
`in Plasma (fu)
`
`Blood-to-Plasma
`Ratio
`
`Basic compounds
`Chlorpromazine
`Propafenone
`Verapamil
`Diphenhydramine
`Lorcainide
`Diltiazem
`Amitriptyline
`Desipramine
`Imipramine
`Ketamine
`Quinidine
`Clozapine
`Neutral compounds
`Dexamethasone
`Prednisone
`Diazepam
`Midazolam
`Methoxsalen
`Alprazolam
`Triazolam
`Zolpidem
`Acidic compounds
`Diclofenac
`Ibuprofen
`Tolbutamide
`Warfarin
`Tenidap
`Tenoxicam
`Amobarbital
`Hexobarbital
`Methohexital
`
`0.05
`0.04
`0.10
`0.22
`0.15
`0.22
`0.05
`0.18
`0.10
`0.88
`0.13
`0.05
`
`0.32
`0.25
`0.013
`0.05
`0.09
`0.32
`0.10
`0.08
`
`0.005
`0.01
`0.04
`0.01
`0.0007
`0.009
`0.39
`0.53
`0.27
`
`0.78
`0.70
`0.77c
`0.65c
`0.77
`1.0
`0.86
`0.96
`1.1
`0.82c
`0.92
`0.87
`
`0.93
`0.83c
`0.71
`0.53
`0.67
`0.78c
`0.62c
`0.76c
`
`0.55c
`0.55c
`0.55c
`0.55
`0.56
`0.67
`1.5
`1.0
`0.70c
`
`Nonrenal Clearancea
`
`Plasma
`
`Blood
`
`ml/min/kg
`
`8.6b
`13
`15
`6.2
`14
`12
`10
`12
`13
`16
`2.5
`2.5
`
`3.5
`4.1
`0.4
`4.6
`12
`0.59
`2.9
`4.3
`
`4.2
`0.8
`0.2
`0.045
`0.058
`0.02
`0.53
`3.6
`11
`
`11
`19
`19
`9.5
`18
`12
`12
`12
`12
`20
`2.7
`2.9
`
`3.8
`4.9
`0.6
`8.7
`18
`0.76
`4.7
`5.7
`
`7.6
`1.5
`0.36
`0.081
`0.10
`0.03
`0.35
`3.6
`16
`
`References
`
`Dahl and Strandjard, 1974; Maxwell et al., 1972; Lund, 1980
`Bryson et al., 1993
`Eichelbaum et al., 1984
`Blyden et al., 1986
`Somani et al., 1987; Klotz et al., 1978
`Echizen and Eichelbaum, 1986; Smith et al., 1983
`Schulz et al., 1983
`Brosen and Gram, 1988
`Sallee and Pollack, 1990; Abernathy et al., 1985
`White et al., 1985
`Greenblatt et al., 1977; Rakhit et al., 1984; Hughes et al., 1975
`Cheng et al., 1988
`
`Tseui et al., 1979; Peterson et al., 1983
`Schalm et al., 1977
`Greenblatt et al., 1980; Maguire et al., 1980
`Heizmann et al., 1983
`Billard et al., 1995; Pibouin et al., 1987
`Smith et al., 1984
`Smith et al., 1987
`Durand et al., 1992
`
`Willis et al., 1979; Chan et al., 1987
`Martin et al., 1990
`Balant, 1981; Scott and Poffenbarger, 1979
`O’Reilly, 1972
`Gardner et al., 1995
`Heintz et al., 1984
`Bachmann, 1987; Sawada et al., 1985
`Breimer et al., 1975; Sawada et al., 1985
`Breimer, 1976; Gillis et al., 1976
`
`a All clearance values from the literature were from i.v. dosing. In the case of dependence of clearance on genetic polymorphism of drug-metabolizing enzymes, data from poor metabolizers was
`excluded. Nonrenal clearance values were calculated by: Clnon-renal 5 Cltotal z (1 2 fraction of the dose excreted unchanged in urine).
`b Chlorpromazine clearance values from i.m. dose; assumes complete absorption from i.m. route.
`c Denotes blood-to-plasma ratios that were unavailable in the scientific literature. Values were determined in duplicate after incubation of drug at 1.0 mg/mL in whole blood at ambient temperature
`for 45 min.
`
`trometer (Sciex, Thornhill, Ontario, Canada) with a turbo ionspray interface.
`There were various mobile phases used for the different drugs as listed in Table
`1. Mobile phase system 1 consisted of 20 mM acetic acid (adjusted to pH 4
`with NH4OH) and CH3CN used at various percentages of organic solvent (as
`listed in Table 1). System 2 consisted of 5 mM NH4OAc (pH unadjusted) and
`CH3CN at various percentages as listed in Table 1. The column used was a
`Phenomenex Luna C18 narrow bore column (2.5 3 50 mm) with a 3-mm
`particle size (Phenomenex, Torrance, CA). The flow rate was 0.5 ml/min and
`the mobile phase composition was held isocratically for each analyte. The
`injection volume was 25 ml.
`The effluent was split with approximately 0.25 ml/min introduced into the
`turbo ionspray source of the mass spectrometer. Source parameters (e.g.,
`orifice voltage, temperature, gas flow rates, etc.) were individually optimized
`for each drug, and the molecular ion (either M 1 H1 or M 2 H2, depending
`on the orifice polarity) was followed for each compound and internal standard
`in the selected ion monitoring mode.
`Calculations. In the determination of the in vitro t1/2, the analyte/ISTD peak
`height ratios were converted to percentage drug remaining, using the T 5 0
`peak height ratio values as 100%. The slope of the linear regression from log
`percentage remaining versus incubation time relationships (2k) was used in
`5 20.693/k. Conversion
`the conversion to in vitro T1/2, values by in vitro T1/2
`to in vitro CL9
`int (in units of ml/min/kg) was done using the following formula
`(Obach et al., 1997):
`
`CL9int 5
`
`0.693
`in vitro T1/ 2
`
`z
`
`ml incubation
`mg microsomes
`
`z
`
`45 mg microsomes
`gm liver
`
`z
`
`20 gm liver
`kg b.w.
`
`For microsomal binding, the fraction unbound in the incubation mixture was
`calculated by:
`
`fu(mic) 5
`
`drug/ISTD peak height ratio in buffer sample
`2 z drug/ISTD peak height ratio in microsome sample
`
`with the factor of 2 in the denominator because the aliquot volume of buffer
`samples analyzed was twice that analyzed for the microsome samples (see
`above).
`The overall accuracies of clearance prediction methods were determined by
`(Obach et al., 1997):
`
`average fold error 5 10
`
`U(logSpredicted
`
`actual
`N
`
`D
`
`U
`
`Literature values for i.v. clearance, plasma binding, and blood-to-plasma ratio
`for the 29 compounds are listed in Table 2. For those compounds in which
`renal excretion of unchanged drug represents a significant component of total
`clearance, clearance values were corrected to nonrenal clearance values by:
`
`CLnonrenal 5 CLtotal z ~1 2 fraction of dose excreted unchanged in urine)
`
`Results
`The use of HPLC-atmospheric pressure ionization-MS was an
`important tool in the gathering of these metabolic lability and micro-
`somal binding data. The selectivity and sensitivity of this instrumen-
`tation permitted facile quantitation of a wide variety of drug struc-
`tures. Chromatographic methods were developed for each compound
`
`

`
`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on May 29, 2015
`
`1354
`
`OBACH
`
`TABLE 3
`
`In vitro intrinsic clearance values and fraction unbound in the incubation conditions for 29 drugs examined
`Each in vitro T1/2 and microsomal binding value represents mean 6 S.D. for triplicate determinations. Intrinsic clearance values were calculated from in vitro T1/2 data as described in
`Experimental Procedures.
`
`Drug
`
`Basic compounds
`Chlorpromazine
`Propafenone
`Verapamil
`Diphenhydramine
`Lorcainide
`Diltiazem
`Amitriptyline
`Desipramine
`Imipramine
`Ketamine
`Quinidine
`Clozapine
`Neutral compounds
`Dexamethasone
`Prednisone
`Diazepam
`Midazolam
`Methoxsalen
`Alprazolam
`Triazolam
`Zolpidem
`Acidic compounds
`Diclofenac
`Ibuprofen
`Tolbutamide
`Warfarin
`Tenidap
`Tenoxicam
`Amobarbital
`Hexobarbital
`Methohexital
`
`Microsomal
`Concentration
`
`mg/ml
`
`1.0
`0.5
`0.5
`6.0
`1.0
`2.0
`0.5
`0.5
`0.5
`1.0
`5.0
`5.0
`
`5.0
`5.0
`5.0
`1.0
`0.5
`5.0
`1.0
`5.0
`
`0.3
`2.0
`10
`10
`3.0
`10
`10
`5.0
`1.0
`
`In Vitro
`T1/2
`
`min
`
`25 6 6
`8.0 6 0.4
`10 6 0.2
`49 6 24
`13 6 2
`21 6 3
`92 6 13
`74 6 24
`66 6 5
`23 6 3
`37 6 5
`27 6 5
`
`42 6 3
`47 6 1
`54 6 19
`3.9 6 0.1
`31 6 3
`105 6 66
`33 6 2
`44 6 5
`
`11 6 3
`36 6 4
`71 6 12
`.120
`26 6 2
`38 6 11
`66 6 5
`48 6 6
`13 6 2
`
`CL9int
`
`ml/min/kg
`
`25 6 6
`166 6 8
`122 6 2
`2.1 6 0.9
`50 6 6
`15 6 2
`14 6 2
`17 6 7
`19 6 2
`27 6 4
`3.4 6 0.5
`4.6 6 0.9
`
`3.0 6 0.2
`2.7 6 0.0
`2.3 6 0.7
`160 6 3
`40 6 3
`1.6 6 1.0
`19 6 1
`2.8 6 0.3
`
`189 6 39
`8.8 6 0.9
`0.90 6 0.15
`,0.52
`8.3 6 0.7
`1.7 6 0.4
`0.94 6 0.07
`2.3 6 0.3
`49 6 8
`
`fu(mic)
`
`0.11 6 0.02
`0.26 6 0.04
`0.43 6 0.10
`0.29 6 0.02
`0.52 6 0.03
`0.76 6 0.10
`0.15 6 0.04
`0.21 6 0.01
`0.18 6 0.04
`0.49 6 0.02
`0.32 6 0.17
`0.13 6 0.01
`
`1.00 6 0.07
`0.20 6 0.02
`0.28 6 0.05
`0.88 6 0.12
`0.94 6 0.11
`0.66 6 0.04
`0.78 6 0.09
`0.58 6 0.10
`
`1.00 6 0.13
`0.84 6 0.13
`0.95 6 0.03
`0.47 6 0.05
`0.32 6 0.01
`0.78 6 0.03
`0.76 6 0.08
`0.81 6 0.05
`0.86 6 0.13
`
`using the same column and only two types of mobile phases, with
`virtually the only customization required for each compound being
`determination of an optimal percentage of organic modifier (CH3CN)
`to effect elution of drug and internal standard within a reasonable run
`time.
`In vitro T1/2 data in pooled human liver microsomes for the 29
`compounds examined are listed in Table 3. Metabolic lability of this
`set of compounds spanned a wide range, the most stable compound
`being warfarin (in vitro T1/2 was immeasurably long at a microsomal
`protein concentration of 10 mg/ml), and the most labile being diclofe-
`nac, propafenone, and midazolam (scaled CL9
`int values of 160 ml/
`min/kg or greater). Within each general class of compounds (weak
`bases, weak acids, and neutral compounds), intrinsic clearance values
`spanned a broad range. Bases ranged from low intrinsic clearance
`values of 3.4 and 4.6 ml/min/kg for quinidine and clozapine, respec-
`tively, to high intrinsic clearance values of 122 and 166 ml/min/kg for
`verapamil and propafenone, respectively. Intrinsic clearance values
`for acids ranged from less than 0.52 ml/min/kg for warfarin and 0.90
`and 0.94 ml/min/kg for tolbutamide and amobarbital, respectively, up
`to 189 ml/min/kg for diclofenac. Intrinsic clearance values for the
`neutral compounds ranged from 1.6 ml/min/kg for alprazolam to 160
`ml/min/kg for midazolam.
`The extent of microsomal binding was determined for each com-
`pound using a microsomal protein concentration equal to that used in
`the metabolic incubations (Table 3). Because different protein con-
`centrations were used, the compounds cannot be rank ordered with
`regard to extent of binding to microsomes. The values ranged from no
`binding to approximately 90% bound. Furthermore, those compounds
`
`that exhibited the greatest extent of binding were not necessarily those
`in which the microsomal protein concentration was highest. In gen-
`eral, the weak bases demonstrated greater binding to microsomes,
`despite the fact that microsomal concentrations used for the bases
`were, on average, lower than those used for the neutral and acidic
`compounds.
`A summary of human blood clearance predictions from the in vitro
`data is presented in Table 4 and predicted clearance values are plotted
`versus actual clearance values in Fig. 2. Equations for both the
`well-stirred and the parallel-tube models of hepatic extraction were
`applied under three variations: disregarding all binding values (Table
`4, eqs. 1 and 4), including only blood binding (Table 4, eqs. 2 and 5),
`and including both blood and in vitro microsome binding (Table 4,
`eqs. 3 and 6). Overall accuracy values, determined as described in
`Experimental Procedures, are listed in Table 5. For all compounds
`examined (n 5 29), average fold error values were just over 2-fold in
`the cases in which either no binding values were considered or all
`binding values were considered. The most accurate method was the
`use of the parallel-tube model with both blood and microsome binding
`incorporated (average fold error of 2.13). Using only the blood bind-
`ing value in either model of hepatic extraction yielded very poor
`predictions of human clearance. When subsets of compounds were
`considered, some differences as to which were the most accurate
`methods were observed. For weak bases and neutral compounds,
`disregarding all binding in either model of hepatic extraction yielded
`the best agreement between actual human clearance values and those
`projected from in vitro intrinsic clearance data. However, for the
`acidic compounds, the most accurate clearance prediction methods
`
`

`
`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on May 29, 2015
`
`MICROSOME BINDING IMPACT ON CLEARANCE PREDICTIONS
`
`1355
`
`Summary of human clearance values predicted from in vitro intrinsic clearance, protein binding, and microsome binding values using well-stirred and parallel-tube
`models of hepatic extraction
`
`TABLE 4
`
`Actual
`Human
`CLblood
`
`mL/min/kg
`
`No Binding
`(eq. 1)
`
`Well-Stirred
`
`fu(bl) Only
`(eq. 2)
`
`Predicted Human CLblood
`
`a
`
`Parallel-Tube
`
`fu(bl) and fu(mic)
`(eq. 3)
`
`No binding
`(eq. 4)
`
`fu(bl) Only
`(eq. 5)
`
`fu(bl) and fu(mic)
`(eq. 6)
`
`11
`19
`19
`9.5
`18
`12
`12
`12
`12
`20
`2.7
`2.9
`
`3.8
`4.9
`0.6
`8.7
`18
`0.76
`4.7
`5.7
`
`7.6
`1.5
`0.36
`0.08
`0.10
`0.03
`0.35
`3.6
`16
`
`11
`19
`18
`1.9
`15
`8.7
`8.2
`9.4
`10
`12
`2.9
`3.8
`
`2.6
`2.4
`2.1
`19
`14
`1.5
`10
`2.5
`
`19
`6.2
`0.86
`0.46
`5.9
`1.6
`0.90
`2.1
`15
`
`1.5
`6.5
`9.0
`0.7
`6.7
`2.9
`0.8
`2.8
`1.6
`12
`0.5
`0.3
`
`1.0
`0.8
`0.04
`8.8
`4.3
`0.64
`2.7
`0.3
`
`1.6
`0.2
`0.07
`0.01
`0.01
`0.02
`0.24
`1.2
`9.9
`
`8.6
`13
`13
`2.2
`9.9
`3.6
`4.2
`8.8
`6.6
`15
`1.4
`1.9
`
`1.0
`3.4
`0.2
`9.4
`4.5
`0.95
`3.3
`0.5
`
`1.6
`0.2
`0.07
`0.02
`0.03
`0.03
`0.32
`1.40
`11
`
`15
`21
`21
`2.0
`19
`11
`10
`12
`12
`15
`3.1
`4.1
`
`2.8
`2.5
`2.2
`21
`18
`1.5
`12
`2.6
`
`21
`7.2
`0.88
`0.46
`6.8
`1.6
`0.92
`2.2
`19
`
`1.5
`7.6
`11
`0.7
`7.8
`3.0
`0.8
`2.9
`1.6
`15
`0.5
`0.3
`
`1.0
`0.8
`0.04
`11
`4.7
`0.64
`2.8
`0.3
`
`1.6
`0.2
`0.07
`0.01
`0.01
`0.02
`0.24
`1.2
`12
`
`11
`17
`17
`2.3
`12
`3.9
`4.6
`11
`7.7
`20
`1.4
`1.9
`
`1.0
`3.6
`0.2
`12
`5.0
`0.97
`3.6
`0.5
`
`1.6
`0.2
`0.07
`0.02
`0.03
`0.03
`0.32
`1.45
`14
`
`Drug
`
`Basic compounds
`Chlorpromazine
`Propafenone
`Verapamil
`Diphenhydramine
`Lorcainide
`Diltiazem
`Amitriptyline
`Desipramine
`Imipramine
`Ketamine
`Quinidine
`Clozapine
`Neutral compounds
`Dexamethasone
`Prednisone
`Diazepam
`Midazolam
`Methoxsalen
`Alprazolam
`Triazolam
`Zolpidem
`Acidic compounds
`Diclofenac
`Ibuprofen
`Tolbutamide
`Warfarin
`Tenidap
`Tenoxicam
`Amobarbital
`Hexobarbital
`Methohexital
`
`Equations used are as follows:
`
`CLblood 5
`
`Q z CL9int
`Q 1 CL9int
`
`(1)
`
`CLblood 5
`
`Q z fu(blood) z CL9int
`Q 1 fu(blood) z CL9int
`
`(2)
`
`CLblood 5
`
`(3)
`
`Q z fu(blood) z
`
`Q 1 fu(blood) z
`
`CL9int
`fu(mic)
`CL9int
`fu(mic)
`S 2CLint
`D!
`S 2fu(blood)zCLint
`CLblood 5 Q z ~1 2 eS 2fu(blood)zCLint
`
`~5!
`
`~6!
`
`CLblood 5 Q z ~1 2 e
`
`Q
`
`CLblood 5 Q z ~1 2 e
`
`Q
`
`Qzfu(mic)
`
`~4!
`D!
`D!
`a A value of 21 ml/min/kg was used for human hepatic blood flow (Q).
`
`incorporated both blood and microsome binding. Figure 3 contains
`plots of accuracy of predicted clearance values using the six equations
`versus the respective human clearance values.
`
`Discussion
`The prediction of human pharmacokinetic parameters for new
`chemical entities has become an important approach in the drug
`discovery process. For any given drug discovery approach, hun-
`dreds of compounds will satisfy potency objectives, however few
`
`can be examined in humans. New chemical entities require exten-
`sive investigation and investment of resources prior to administra-
`tion to humans, and therefore it is desirable to be able to exclude
`compounds from this process that would be expected to exhibit
`unsatisfactory human pharmacokinetic properties. Recently, sev-
`eral investigators have described various methods and approaches
`whereby human pharmacokinetic parameters can be predicted from
`in vitro and/or preclinical pharmacokinetic data (Hoener, 1994;
`Gobburu and Shelver, 1995; Lave et al., 1997a,b; Obach et al.,
`
`

`
`1356
`
`OBACH
`
`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on May 29, 2015
`
`FIG. 2.Plots of human clearance values predicted from in vitro intrinsic clearance data versus actual human clearance values.
`A, well-stirred model disregarding all binding parameters. B, well-stirred model incorporating only plasma protein binding. C, well-stirred model incorporating both
`plasma protein and microsome binding. D, parallel-tube model disregarding all binding parameters. E, parallel-tube model incorporating only plasma protein binding. F,
`parallel-tube model incorporating both plasma protein and microsome binding.
`
`1997; Kuhnz and Gieschen, 1998; Sarver et al., 1997; Mahmood,
`1998a,b). These methods vary in complexity and the amount of
`data required for accurate predictions.
`One of the simplest methods described to predict human clearance
`is the use of human hepatic microsomal lability data, termed the in
`vitro T1/2 approach (Obach et al., 1997). In this method, one incubates
`the test compound with human liver microsomes in the presence of
`appropriate cofactors (NADPH for CYP catalyzed reactions) and
`measures the first-order rate of consumption of the test compound.
`This rate is converted to an in vitro CL9
`int value, scaled-up to reflect
`CL9
`int in vivo, and inserted into a model of hepatic extraction. A high
`degree of success was previously reported for this particular approach,
`however the number of test compounds was low, and a majority of the
`
`compounds were of a similar physicochemical class (lipophilic
`amines). One of the objectives of the present experimentation was to
`further test the in vitro T1/2 approach with more compounds and
`greater structural diversity.
`The use of hepatic microsomes in the prediction of clearance
`requires acceptance of several assumptions and caveats: 1) metabolic
`..
`clearance is the major mechanism of clearance (i.e., CLmetabolism
`1 CLbiliary
`1 CLother); 2) the liver is the major organ of
`CLrenal
`.. SCLall other organs); 3) oxidative metabo-
`clearance (i.e., CLhepatic
`lism predominates over other metabolic routes such as direct conju-
`gative metabolism, reduction, hydrolysis, etc.; 4) rates of metabolism
`and enzyme activities in vitro are truly reflective of those that exist in
`vivo. Additionally, a tenet of the well-stirred and parallel-tube models
`
`

`
`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on May 29, 2015
`
`MICROSOME BINDING IMPACT ON CLEARANCE PREDICTIONS
`
`1357
`
`TABLE 5
`Accuracy of clearance predictions from liver microsomal in vitro T1/2 values including and excluding values for blood binding and microsome binding
`
`Average Fold Error (predicted CLblood/actual CLblood)
`
`Basic compounds
`only (n 5 12)
`
`Neutral compounds
`only (n 5 8)
`
`Acidic compounds
`only (n 5 9)
`
`All compounds
`(n 5 29)
`
`Well-stirred model
`No binding considered (eq. 1)
`Including fu(blood) (eq. 2)
`Including fu(blood) and fu(mic) (eq. 3)
`Parallel-tube model
`No binding considered (eq. 4)
`Including fu(blood) (eq. 5)
`Including fu(blood) and fu(mic) (eq. 6)
`
`1.37
`5.17
`1.86
`
`1.31
`4.75
`1.60
`
`1.99
`3.90
`2.55
`
`1.99
`3.90
`2.53
`
`5.05
`3.91
`2.83
`
`5.35
`3.79
`2.74
`
`2.28
`4.39
`2.31
`
`2.28
`4.20
`2.14
`
`of hepatic extraction is that the unbound concentration of drug in the
`plasma is equal to the unbound concentration in the hepatocyte.
`Therefore, facilitated transport processes that could possibly be re-
`sponsible for drug uptake or drug extrusion from hepatocytes are not
`accounted for in these models. The in vitro T1/2 approach has two
`additional inherent assumptions that are not required if intrinsic clear-
`ance were determined using the more rigorous approach of calculating
`intrinsic clearance from enzyme kinetic data (i.e., Vmax/KM). These
`are: 1) the substrate concentration employed (1.0 mM in the case of
`this report) is well below the apparent KM for substrate turnover, and
`2) there is no significant product inhibition, nor is there any mecha-
`nism based inactivation of enzyme. Overall, clearance for these 29
`compounds was generally underestimated using any of the six ap-
`proaches, giving credence to the notion that the aforementioned as-
`sumptions are not completely valid in many cases. It should be noted
`that the 29 compounds chosen for this examination represent a set for
`which other clearance mechanisms (e.g., renal clearance, nonoxida-
`tive clearance, etc.) are known to be less important than hepatic
`oxidative metabolic clearance, but some are known to fall outside the
`scope of the aforementioned assumptions, e.g., nonhepatic metabo-
`lism of dexamethasone (Diederich et al., 1996; Tomlinson et al.,
`1997), nonoxidative components of metabolic clearance such as the
`reductive metabolism of warfarin (Moreland and Hewick, 1975) or
`glucuronidation of ibuprofen (Rudy et al., 1991), and product inhibi-
`tion of diltiazem (Sutton et al., 1997).
`A second objective of this work was to further explore the potential
`importance of nonspecific reversible binding

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket