Vol.1, No.4, 249-262 (2009)
doi:10.4236/health.2009.14041
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
Health
Characterization of in vitro and in vivo metabolism of
AG-024322, a novel cyclin-dependent kinase (CDK)
inhibitor
Wei-Zhu Zhong*, Bojan Lalovic1, Jenny Zhan2
1Clinical Pharmacology, Pfizer Global Research and Development, New London, Connecticut, USA
2Metabolism and Pharmacokinetics, Novartis Institute of Biomedical Research, Cambridge, Massachusetts, USA
*Pharmacokinetics, Dynamics and Metabolism, Pfizer Global Research and Development, San Diego, California, USA;
wei-zhu.zhong@pfizer.com
Received 16 November 2009; revised 25 November 2009; accepted 28 November 2009.
ABSTRACT
Uncontrolled cell proliferation is the hall mark of
many cancers, and is typically manifested by a
deregulation of the cell-division cycle. CDKs
play critical roles in regulating cell cycle, apop-
tosis and cell differentiation. AG-024322 is a
multitargeted CDK inhibitor that has been
shown to induce cancer cell apoptosis and de-
monstrate significant antitumor activity in hu-
man tumor xenograft models. This compound is
under clinical development as an intravenous
anticancer agent. AG-024322 exhibited moder-
ate to high systemic clearance across preclini-
cal species. In vitro metabolism in human liver
microsomes and hepatocytes demonstrates that
glucuronidation and oxidation represent the
major metabolic pathways of AG-024322. The
experiments of chemical inhibition and micro-
somes containing individual CYP or UGT iso-
forms revealed that CYP3A and UGT1A1 appear
to predominantly mediate AG-024322 oxidation
and glucuronidation, respectively. Formation
kinetics of the two pathways in human liver mi-
crosomes suggested that the glucuronidation
activity of AG-024322 was approximately 3-fold
higher as compared to CYP-mediated oxidation,
contributing approximately 37% and 13% of the
total clearance, respectively, based on the pro-
jected human clearance. UGT1A1 is a poly-
morphic isoform involved in glucuronidation of
bilirubin. It is of concern if glucuronidation via
UGT1A1 plays a major role in the elimination of
AG-024322 in humans as competitive inhibition
of UGT1A1 has been associated with toxicity
(Gilbert and Crigler-Najjar syndromes). There-
fore, this information was used to influence the
clinical study design to only include subjects
having constitutive expression of UGT1A1 in
the first human study, thereby decreasing the
potential risk of toxicity to patients.
Keywords: Microsomes; Hepatocytes; Reaction
Phenotyping; Metabolite Formation Kinetics;
CYP3A4; UGT1A1; Mass Balance in Rat
1. INTRODUCTION
AG-024322, N-({5-[3-(4,6-difluoro-1H-benzimidazol-2-
yl)-1H-indazol-5-yl]-4-methylpyridin-3-yl}methyl)ethan-
amine, is a multitargeted CDK inhibitor under deve-
lopment as an anticancer agent. AG-024322 is a potent
ATP-competitive inhibitor of the cell cycle kinases
CDK1, CDK2, and CDK4, with kinetic inhibition con-
stants (Ki) in the 1-3 nM range [1]. CDKs play a key
role in regulating the cell cycle progression and to a
large degree control cellular transitions from growth
phases into phases associated with DNA replication and
mitosis by phosphorylating several cellular proteins
[2-4]. AG-024322 inhibits phosphorylation of CDK sub-
strates in cells, resulting in cell arrest and broad-spec-
trum anti-proliferative activity, and has been shown to
induce cancer cell apoptosis and significant antitumor
efficacy in human tumor xenograft models with tumor
growth inhibition (TGI) ranging from 32% to 86.4% [5].
Single-dose pharmacokinetics of AG-024322 was
evaluated in the rat, dog and monkey following a 30-
minute IV infusion, the intended administration route in
humans. AG-024322 demonstrated moderate clearance
of 26 and 27 ml/min/kg in monkeys and dogs, respec-
tively, and somewhat higher clearance in rats (67
ml/min/kg), with half-lives ranging from 5 to 7 hours.
Minimal contribution of renal clearance was observed in
all 3 species ( 1%). The volume of distribution greatly
exceeded the volume of total body water across species
(9 to 15 l/kg), and was consistent with rapidly and
W. Z. Zhong et al. / HEALTH 1 (2009) 249-262
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
250
widely tissue distribution observed in Long Evans rats
by quantitative whole body autoradiography (QWBA)
using [14C]AG-024322. The plasma protein binding of
AG-024322 was in the range of 85 to 95% across species.
In view of the predominant metabolic pathway of glu-
curonidation observed in human hepatocytes and the
generally low metabolic turnover in in vitro incubations,
the prediction of pharmacokinetics in human was per-
formed using interspecies allometric scaling of single-
dose animal pharmacokinetics data. This approach lead
to a reasonable prediction of human clearance (12.5 ml/
min/kg), which was similar to the clearance actually
observed in humans with IV doses from 20 to 340 mg.
Detailed nonclinical pharmacokinetics and the prediction
of human pharmacokinetics of AG-024322 will be re-
ported elsewhere.
To support the development of AG-024322, a series of
nonclinical in vitro and in vivo studies were conducted to
identify AG-024322 metabolites, and to characterize the
disposition of AG-024322. These studies involved with
either using animal and human liver microsomes and
hepatocytes incubated with C-14 labeled AG-024322
([14C]AG-024322) or using urine, plasma, bile, and fecal
samples from rats administered [14C]AG-024322. Fur-
ther studies to assess the two principal pathways of AG-
024322 biotransformation, CYP-mediated oxidative and
UGT-mediated glucuronidation, and to identify the ma-
jor enzymes involved in these metabolic pathways were
also carried out.
In the present paper, discussion was emphasized on the
in vitro and in vivo characterization of AG-024322 me-
tabolism and the relative importance of CYP3A4 and
UGT1A1 in the biotransformation of AG-024322 in HLM,
as well as the relative contributions of the two metabolic
pathways in the overall clearance of AG-024322.
2. MATERIALS AND METHODS
2.1. Materials
AG-024322 was synthesized at Pfizer Global Research
& Development (PGRD), La Jolla, CA. [14C]AG-
024322 (Specific activity 53.1 mCi/mmol) was synthe-
sized by the Radiosynthesis Group at PGRD (Groton,
CT), and the label was located in the C-1 position of the
benzimidazol ring (Scheme 1). Rat, dog, monkey and
human liver microsomes were obtained from Gentest
Corporation. (Woburn, MA). Human liver microsome
preparation (HL-Mix 14, 21.7 mg protein/ml, 0.3 nmol
CYP/mg protein) and microsomes from insect cells
stably expressing human cDNA CYPs and UGTs (su-
persomes) were also purchased from Gentest Corpora-
tion. Cryopreserved hepatocytes for dogs, monkeys and
humans, and the thawing media were obtained from In
Vitro Technologies (Baltimore, MD). Sprague-Dawley rat
hepatocytes and Hepatocyte Isolation Kit were pur-
Scheme 1. Chemical structure of AG-024322
(asterisk indicates the site of radiolabel).
chased from XenoTech LLC (Lenexa, KS). Ketocona-
zole, quindine, quercetin, furafyline, dibasic and mono-
basic potassium phosphate, NADPH, UDPGA, and
WEM were purchased from Sigma-Aldrich Co. (St.
Louis, MO). HEPES (4-(2-hydroxyethyl)-1-piperazi-
neethanesulfonic acid) buffer was purchased from Invi-
trogen (Carlsbad, CA). Commercially obtained chemi-
cals and solvents were of HPLC or the highest grade
available.
2.2. Liver Microsomal Incubations
Liver microsomes were preincubated with AG-024322
and [14C]AG-024322 in 100 mM potassium phosphate
buffer (pH 7.4) for 5 minutes in a 37ºC shaking water
bath. The reaction was initiated by the addition of
NADPH with a final incubation volume of 0.5 ml con-
taining 10 µM AG-024322, 0.87 µM [14C]AG-024322, 1
mM NADPH, 0.75 mg/ml microsomal protein and 100
mM potassium phosphate buffer (pH 7.4). The incuba-
tion was terminated at 0, 10, 20, 40 and 60 minutes by
the addition of 0.5 ml of acetonitrile and immediately
vortexed and centrifuged at 1900g for 25 minutes. The
supernatants were transferred to a fresh tube, evaporated
to dryness under nitrogen, and reconstituted with 200 µl
of Solvent A for LC/RAM/MS analysis as described
below.
2.3. Hepatocytes Incubation
Cryopreserved hepatocytes were thawed and used ac-
cording to the manufacturer’s instructions. The thawed
cryopreserved hepatocytes were re-suspended into CO2-
bubbled incubation media to determine the total cell
count using the Trypan Blue Exclusion method. The
incubation media consisted of WEM supplemented with
10 mM HEPES buffer. The final cell concentrations
were 1.5, 1.8, 1, and 1.23106 viable cells/ml for rat, dog,
monkey and human hepatocytes in WEM, respectively.
Hepatocytes were incubated in the media containing 10
µM AG-024322 and 0.87 µM [14C]AG-024322 in a
CO2:O2 (5:95) stationary incubator at 37ºC. The final
incubation volume was 1 ml. Reactions were terminated
at several time points from time zero to 5 hours by the
addition of 2 ml of acetonitrile. Samples were then soni-
NN
N
N
N
F
N
F
*
W. Z. Zhong et al. / HEALTH 1 (2009) 249-262
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
251
cated, vortexed and centrifuged at 1900g for 25 min-
utes at 4ºC. The resulting supernatant was evaporated
under nitrogen and reconstituted in 100 µl of water for
LC/RAM/MS analysis.
2.4. CYP Reaction Phenotyping Screen
AG-024322 (1 and 10 µM), each in triplicate, were in-
cubated with a panel of microsomes from insect cells
expressing individual human CYPs (Supersomes) con-
taining 50 pmol of CYP1A2, 2C8, 2D6, 2E1, 3A4 and
3A5 protein or 100 pmol of 2C9*1 and 2C19. The final
incubation volume was 0.5 ml. Incubations containing
AG-024322 and 1 mM NADPH in 100 mM phosphate
buffer (pH 7.4) were preincubated for 5 minutes at 37ºC.
The reactions were started with the addition of 50 µl of
each CYP and were allowed to proceed up to 15 minutes.
Incubations were stopped with the addition of 0.5 ml of
acetonitrile and 20 µl of 1ng/µl AG-024397 (IS), and
immediately capped, vortexed and centrifuged at 1900g
for 25 minutes at 4ºC. The supernatant of each sample
was dried under N2 and reconstituted with 100 µL of
Solvent C for LC-MS analysis as described below.
2.5. Chemical Inhibition
CYP-specific chemical inhibitors, ketoconazole (CYP
3A4), quinidine (CYP2D6), quercetin (CYP2C8), and
furafyline (CYP1A2) were used to identify the isoforms
mediating the formation of N-desethyl AG-024322. In-
cubation mixtures were prepared with ketoconazole and
quinidine at 20, 100, 500 and 1 µM, quercetin at 0.3,
0.75 and 2 µM and furafyline at 1, 10 and 30 µM, and
were preincubated with AG-024322 (1 and 10 µM) and
15 mg HLM protein (0.3 mg/ml) for 5 minutes at 37ºC.
Incubations were started by the addition of NADPH to a
concentration of 1 mM in a final volume of 0.5 ml. In-
cubations with furafyline, a time-dependent inhibitor of
CYP1A2, required a preincubation step in the presence
of 1 mM NADPH for 20 minutes prior to the introduc-
tion of the substrate. The reactions were terminated after
8 minutes by the addition of 0.5 ml of acetonitrile and 20
µl of 1 ng/µl IS. The remaining part of the procedure
was performed as described above for CYP reaction
phenotyping screen.
2.6. UGT Reaction Phenotyping Screen
AG-024322 (1 and 10 µM), each in triplicate, was incu-
bated with 8 UGT isoforms (UGTlA1, 1A3, 1A4, 1A6,
1A8, 1A10, 2B7 and 2B15). The incubation procedure
was adapted from Fisher et al. [6]. Microsomes (0.5 mg/
ml) suspended in 100 mM phosphate buffer (pH 7.1)
were initially treated with alamethicin at 0.025 mg/mg
microsomal protein. The mixture also included 1 mM
MgCl2 and was kept on ice for 15 minutes to permealize
the microsomal membrane. AG-024322 was then added
and the mixture was preincubated at 37‘C for 5 minutes.
The reaction was started with the addition of 1 mM
UDPGA in a total incubation volume of 0.2 ml, and were
terminated after 15 minutes by the addition of 0.5 ml of
acetonitrile and 20 µl of 1 ng/µl IS. The remaining part
of the procedure was performed as described above for
CYP reaction phenotyping screen.
2.7. Metabolite Formation Kinetics in HLM
The formation kinetics of the N-desethyl metabolite and
the parent glucuronides were studies in the same liver
preparation from 14 different human livers (HL-Mix 14)
to afford a comparison of the oxidation and glucuronida-
tion pathways.
To determine the formation rates of N-desethyl me-
tabolite, AG-024322 with a concentration range of 0.5-
100 µM, each in triplicate, was pre-incubated with HLM
containing 0.75 mg/ml protein in 100 mM potassium
phosphate buffer (pH 7.4) for 5 minutes at 37ºC. The
reaction was initiated by the addition of 1 mM NADPH
in a final incubation volume of 0.5 ml. Reactions were
terminated at 15 minutes by the addition of 0.5 ml of
acetonitrile and 40 µl of 1 ng/µl IS. The remaining part
of the procedure was performed as described above for
CYP reaction phenotyping screen.
The experiment conditions to determine the formation
rates of parent glucuronides were optimized in respect to
time and protein concentration. Linear product formation
was observed for up to 1 hour and 2 mg/ml microsomal
protein. The experiments were following the similar
procedure as described above using UGT supersomes,
except that the incubation mixture contains 1 mg/ml
microsome protein, 1 mM MgCl2, 0.05 mg/mg micro-
some protein alamethicin, and 2.5 mM UDPGA in either
0.1 M phosphate buffer (pH 7.1) or WEM, incubated
with a range of AG-024322 concentrations (0.5 to 200
µM) in triplicate, and terminated at 35 minutes.
2.8. Formation Kinetics Calculation
The relative formation rate of each metabolite (peak area
ratio/mg protein/15 or 35 min) was calculated and plot-
ted against AG-024322 concentrations. The metabolite
formation kinetic parameter, Km, was estimated by fitting
the data to the sigmoid Emax model using SAAMII
(SAAM Institute, Seattle, WA). The maximum formation
rate (Vmax) was estimated from separate experiments of
HLM incubation with 10 µM [14C]AG-024322 at the
similar conditions as described above. The formation
rate (V) of each metabolite in the incubations was calcu-
lated based on the radioactivity. The Vmax value was then
calculated based on the Michaelist-Menten equation,
Vmax is the substrate concentration. The in vitro intrinsic
clearance (CLint) was determined by Vmax/Km ratio. The
predicted CLint was scaled up to in vivo by extrapolating
CLint assuming a human liver weight of 1500 g (70 kg body
weight) and that one gram liver tissue contains 45 mg HLM
protein. The total hepatic clearance (CLh) was calculated
W. Z. Zhong et al. / HEALTH 1 (2009) 249-262
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
252
from the well-stirred model: CLh = Q x CLint / Q + CLint,
where Q is the human hepatic blood flow (20 ml/min/kg).
The estimation of CLh was not incorporated with plasma
and microsome protein binding.
2.9. Mass Balance Study in Rats
In this study, [14C]AG-024322 in 5% dextrose in water
(pH 4) was administered as a single 30-minute IV infu-
sion to all animals at a target dose level of 10 mg/kg
(~50 µCi/250 g body weight). The intact animals in
Group 1 (3/gender) were individually housed in metabo-
lism cages for the separate collection of urine and feces
over intervals through 120 hours after dosing. The bile
duct-cannulated (BDC) rats in Group 3 (2/gender) were
individually housed in metabolism cages for the collec-
tion of bile over intervals through 48 hours. Blood sam-
ples were collected from intact animals in Group 2
(2/gender) before dosing and at 0.25, 0.5, 1, 3, 8, 24, 48,
72, 96, and 120 hours from the initiation of infusion.
Blood samples were collected from intact animals in
Group 4 by sacrificing 1 animal/gender at each time
point at 0.5, 2, 4, and 8 hours from the initiation of infu-
sion. Urine, feces, bile, cage wash, cage wipe, blood, and
plasma samples were processed and analyzed for radio-
activity. Selected plasma, urine, bile, and fecal samples
were used for metabolite identification and profiling.
2.10. Plasma, Urine, Fecal and Bile Sample
Preparation
Plasma samples collected from Group 4 animals were
pooled based on the pooling formula described by Ham-
ilton and coworkers [7] to generate plasma pools (2.0
mL). The pooled plasma samples were precipitated by
the addition of 4 volumes of acetonitrile: methanol (3:1,
v/v) with 0.2% formic acid, and then vortexed and cen-
trifuged. Urine samples were pooled on a percent weight
basis and centrifuged. The supernatants from the plasma
or urine pool extracts were concentrated to approxi-
mately 1 ml under N2 before LC/RAM/MS analysis.
Selected fecal homogenate from each animal were
pooled on a percent weight basis to generate fecal ho-
mogenate pools. Each pooled sample was precipitated by
the addition of 4 volumes of acetonitrile containing 0.2%
formic acid, vortexed and centrifuged. The pellet was re-
extracted using the same method, and supernatants were
combined for evaporation under N2 to dryness. The resi-
dues were reconstituted in 1 ml of 20:80 (v/v) metha-
nol:20 mM ammonium acetate with 0.1% formic acid for
LC/RAM/MS analysis. Selected bile samples were pooled
on a percent weight basis to generate bile pools. After
centrifugation, every 100 µl of bile sample was diluted
with 900 µl of 20:80 (v/v) methano l:20 mM ammonium
acetate containing 0.1% formic acid for LC/RAM/MS
analysis. All centrifugation was performed at 15850g
for 10 minutes using Brinkman eppendorf 5415c (West-
bury, NY). The injection volume for LC/RAM/MS was
300-900 µl.
2.11. Determination of Radioactivity
The radioactivity in urine, bile, fecal, blood, and plasma
samples was determined by liquid scintillation counting
(LSC). Triplicate weighed aliquots (approximately 0.02
to 0.1 g each) of urine, bile, and plasma samples were
mixed with scintillation fluid, and analyzed directly by
LSC. Triplicate weighed aliquots of fecal homogenate
(fecal:water, 20:80, w/w, approximately 0.1-0.5 g each)
or blood (approximately 0.02-0.03 g each) were placed
into cones and pad, dried, combusted, and then analyzed
by LSC.
2.12. Metabolite Characterization
Metabolite characterization was performed using an
Agilent 1100 HPLC system (Wilmington, DE) coupled
in-line with an ARC Model C StopFlow System (AIM
Research Company, Newark, DE) and an IN/US Model
3 -RAM radio-detector (Tampa, FL), and a ThermoF-
innigan LCQ-Deca XP ion trap mass spectrometer (San
Jose, CA). When MS detection was used, the HPLC ef-
fluent was split with approximately 20% of the flow
introduced into the MS via electrospray ionization (ESI)
source and 80% diverted to the -RAM detector. The
ion-trap ESI-MS was operated in positive ion model
with a spray voltage of 4.5 kV, sheath gas flow rate of 70
(arbitrary), auxiliary gas flow rate of 20 (arbitrary) and
capillary temperature at 200ºC. Laura 3 V3.0 (IN/US
system, Tampa, FL), ARC data system V2.4 (AIM Re-
search Company, Newark, DE) and Xcalibur V1.4
(ThermoFinnigan, San Jose, CA) software were em-
ployed to control the -RAM detector, the StopFlow
system and the LC-MS system for data acquisition and
processing. The MS spectra were acquired over a mass
range of m/z 100-900 for all samples. Ion-trap LC-ESI-
MS/MS experiments were performed to generate ion
mass spectra for the selected molecular ions representing
possible metabolites of AG-024322. AG-024322 me-
tabolites were identified by chromatographic resolution
and concurrent radiometric and mass-selective quantifi-
cation.
Separation of the parent drug and its metabolites was
accomplished using a Phenomenex Luna 5 µ C18 col-
umn (2504.6 mm, Torrance, CA) or an Agilent Zorbax
5µ C8 column (1504.6 mm, Palo Alto, CA). The mo-
bile phase consisting of a 20 mM ammonium acetate in
water (pH 4.0) as Solvent A and acetonitrile as Solvent B,
and were programmed for each matrix using a linear
gradient at a constant flow rate of 1.0 mL/min as follows:
1) for hepatocyte and microsome incubations: 0 min, 5%
B; 5 min, 5% B; 30 min, 35% B; 31 min, 95% B; 35 min,
95% B; 36 min, 5% B; and 43 min, 5% B; 2) for urine,
feces and bile samples: 0 min, 10% B; 10 min, 18% B;
W. Z. Zhong et al. / HEALTH 1 (2009) 249-262
SciRes Copyright © 2009 http://www.scirp.org/journal/HEALTH/
253
20 min, 18% B; 42 min, 30% B; 50 min, 30% B; 60 min,
55% B; 61 min, 10% B; and 70 min, 10% B; and 3) for
plasma samples: 0 min, 10% B; 50 min, 30% B; 60 min,
55% B; 61 min, 10% B; and 70 min, 10% B.
Openly accessible at
The analysis of N-desethylation metabolite of AG-
024322 and the glucuronides of the parent drug was
using a Zorbax C8 column (2.15 mm, 5 µ) from
Agilent (Palo Alto, CA) to allow a chromatographic
resolution of the metabolites from the parent drug with
a run time of 12 minute. With an API 4000 MS (AB
Sciex Instruments, Toronto, Ontario), the monitored ion
transitions were 419374, 391374 and 595374 for
the parent compound, N-desethyl metabolite and parent
glucuronide, respectively. The mobile phase was pro-
grammed from the initial setting at 95% Solvent C (2
mM ammonium acetate with 0.1% formic acid in water)
and 5% Solvent D (acetonitrile with 0.1% formic acid)
to 35% C and 65% D at 10 minutes by linear gradient,
and restored to original composition at 10.1 min until
the end of run.. The flow rate was maintained constant
at 0.3 ml/min.
3. RESULTS
3.1. In Vitro Metabolism of AG-024322
Metabolic stability of [14C]AG-024322 in liver micro-
somes and hepatocytes are presented in Table 1 as per-
cent of parent drug remaining after incubation in the
preparations for rat, dog, monkey and human. The cor-
responding disappearance half-life was generated from
the available time course data. [14C]AG-024322 depleted
most extensively in rat liver microsome with 11% of
radiochromatogram of the parent drug remaining at
1-hour incubation time, followed by dog (55%). Com-
parable incubations in human and monkey liver micro-
somes yielded significantly less extensive parent drug
depletions with 95% and 92% remaining after 1-hour
incubation, respectively. [14C]AG-024322 also exhibited
metabolic stability in human hepatocytes relative to
other species. After 5 hours incubation, only 14% of the
compound was metabolized in human hepatocytes,
whereas 26% and 27% in dog and rat hepatocytes, re-
spectively. Monkey hepatocytes most extensively me-
tabolized AG-024322 up to 38%. Based on the chroma-
tographic retention time and molecule weight, several
metabolites were tentatively identified as parent glu-
curonides (M1, M2, and M3), N-desethylation (M6) and
metabolites that may arise from N-acetylation of M6 (M8)
or parent methylation (M10) in hepatocytes and micro-
somes of various species (Table 2). The radiochro- ma-
tograms of the 5-hour incubations of [14C]AG-024322 in
rat, dog, monkey and human hepatocytes are shown in
Figure 1. At 5 hours, glucuronidation of the parent drug
accounted for approximately 8% of the total radioactiv-
ity, while M6 and M8/M10 accounted for 4% and 2% of
Table 1. Metabolic stability of [14C]AG-024322 in liver mi-
crosomes and hepatocytes.
Microsomesa Hepatocytesb
Species Remaining
at 1-hour (%)
Half-life
(h)
Remaining
at 5-hour(%)
Half-life
(h)
Human95 12 86 50
Monkey92 37 62 8.5
Dog 55 1.0 74 14
Rat 11 0.3 73 15
a. Incubations contain 0.75 mg/ml microsomal protein; b. Incubations
contain 1.2, 1.0, 1.8 and 1.5 x106 cells/ml in WEM for human, monkey,
dog and rat, respectively.
Table 2. Metabolite profiles of AG-024322 in rat, dog, monkey and human liver microsomes and cryopreserved hepa-
tocytes incubated with [14C] AG-024322.
% Radiochromatogram
Species M1 M2 M3 M5 (Unk)M6 M7(Unk)AG-024322M7/M10 M9(Unk) M10/M7
Liver Microsomes (at 1 Hour of Incubation)
Rata - - - - 7.5 10.6 10.8 8.8 22.1 -
Dogb - - - - 5.2 12.3 55.3 14.0 - -
Monkey - - - - 8.1 - 91.9 - - -
Human - - - - 4.8 - 95.2 - -
Hepatocytes (at 5 hours of Incubation)
Rat - - 8.3 1.2 11.3- 73.1 6.1 - -
Dog - - - - 13.6- 74.2 2.4 3.9 5.8
Monkey 1.9 7.3 21.7 1.8 4.5 0.8 62 - - -
Human 1.5 1.5 5.2 - 4.1 - 85.7 2.1 - -
a. Novel unidentified metabolites accounted for 40.2% of radiochromatogram in rat liver microsome (not listed); b. Novel unidentified
metabolites accounted for 13.2% of radiochromatogram in dog liver microsome (not listed); -: Trace or no RAM detected metabolites;
M1, M2 and M3: Parent drug glucuronides (M.W. 594); M6: N-desethyl AG-024322 (M.W. 386); M7/M10: acetylated M6 or methy-
lated AG-024322 (M.W. 432); Unk: unknown/unidentified metabolite.
W. Z. Zhong et al. / HEALTH 1 (2009) 249-262
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
254
Time (Min)
Monkey
DPM
M6
M1
M2
M3
0:00 10:00 20:0030:00 40:00
0
100
200
300
400
500
600
700
800
AG-024322
Time (Min)
Monkey
DPM
M6
M1
M2
M3
0:00 10:00 20:0030:00 40:00
0
100
200
300
400
500
600
700
800
AG-024322
Human
Time (Min)
DPM
0:0010:00 20:00 30:00 40:00
0
100
200
300
400
500
600
700
800
900
1000
M1 M2 M3 M6
AG-024322
M8/M10
Human
Time (Min)
DPM
0:0010:00 20:00 30:00 40:00
0
100
200
300
400
500
600
700
800
900
1000
M1 M2 M3 M6
AG-024322
M8/M10
Dog
Time (Min)
DPM
0:00 10:00 20:00 30:00 40:00
0
100
200
300
400
500
600
700
800
DPM
M9( Unk)
M7(Unk)
M6
AG-024322
M8/M10
Dog
Time (Min)
DPM
0:00 10:00 20:00 30:00 40:00
0
100
200
300
400
500
600
700
800
DPM
M9( Unk)
M7(Unk)
M6
AG-024322
M8/M10
Rat
Time (Min)
DPM
M8/M10
M5 (Unk)M6
M3
0:0010:0020:0030:00 40:00
0
100
200
300
400
500
600
700
AG -024322
M7 (Unk)
Rat
Time (Min)
DPM
M8/M10
M5 (Unk)M6
M3
0:0010:0020:0030:00 40:00
0
100
200
300
400
500
600
700
AG -024322
M7 (Unk)
Figure 1. Representative HPLC radiochromatograms of a 5-hour [14C]AG-024322 (11 µM) incubation in rat, dog,
monkey and human cryopreserved hepatocytes (1106 cells)
total radioactivity, respectively, in human hepatocytes.
M6 is the only metabolite detected in human liver mi-
crosomes and accounted for 5% of the total radioactivity.
The preliminary proposed in vitro metabolic scheme is
show in Scheme 2.
3.2. CYP Reaction Phenotyping
The CYP enzymes involved in N-desethylation of AG-
024322 were examined by incubations with microsomes
from insect cells expressing cDNA (recombinant) human
CYPs and by CYP-selective chemical inhibition.
CYP3A appear to predominantly catalyzed the N-de-
sethylation of AG-024322 in vitro at 1 and 10 µM (Fig-
ure 2). Incubations of AG-024322 with recombinant
CYP1A2, 2C8 and 2D6, indicated their minor contribu-
tion after scale-up of the velocity using either CYP con-
tent or relative activity factors [8,9]. Follow-up incuba-
tions with CYP-specific inhibitors suggested that only
Scheme 2. Preliminary In Vitro AG-024322 metabolic scheme.
N
N
H
N
N
NN
FF
N
H
2
N
N
N
NN
F
F
N
H
N
N
NN
F
F
N
N
N
N
N
NN
FF
M8
N-acetylation
M10
N
HN
N
N
NN
FF
O
Gluc.
N-meth ylat ion
N-desethylation
AG-024322
UGT
M1, M2, M3
M6
Oxidation
CYP
W. Z. Zhong et al. / HEALTH 1 (2009) 249-262
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
255
ketoconazole inhibited the N-desethylation of AG-024322
(Table 3). Despite the minor projected involvement of
CYP2D6, CYP2C8 and CYP1A2 (Figure 2), quinidine,
quercetin and furafyline had no inhibitory effect on the
formation rate of N-desethyl AG-024322, confirming a
minimal contribution of these enzymes. The involvement
of CYP2C9, CYP2C19 and CYP2E1 in N-desethylation of
AG-024322 was found to be negligible. Formation kinetics
of N-desethyl AG-024322 in human liver microsome were
further investigated over the concentration range of 0.5 to
100 µM AG-024322 and were best described by a single
enzyme system with an apparent Km of 10 µM using
SAAMII, a general nonlinear kinetic modeling program
(Figure 3). The estimated kinetic parameters, Km, Vmax, in
vitro CLint, and extrapolated in vivo intrinsic and hepatic
clearance are presented in Table 4.
3.3. UGT Reaction Phenotyping
The strategy used here for UGT phenotyping is based on-
well-established methods, including screening of re
Figure 2. Comparison of AG-024322 N-desethylation activity
at 1 and 10 µM in microsomes from insect cells individually
expressing human CYPs (CYP supersomes).
Table 3. Percent (%) inhibition or stimulation of AG-024322 metabolism in the presence of CYP-selective Inhibitors.
Inhibitor Concentration (µM)
% Desethylaton Activity
Remaining at 1 µM AG-0243222
% Desethylaton Activity
Remaining at 10 µM AG-0243222
0.02 77.5 ± 4.4 86.2 ± 3.9
0.1 31.4 ± 2.2 62.2 ± 5.2
0.5 16.9 ± 0.5 30.7 ± 1.7
Ketoconazole
(Inhibitor of CYP3A4)
1 14.2 ± 0.4 23.8 ± 2.1
0.02 131 ± 18 117 ± 7
0.1 157 ± 11 115 ± 4
0.5 158 ± 8 122 ± 11
Quinidine
(Inhibitor of CYP2D6)
1 165 ± 4 120 ± 7
0.3 145 ± 6 120 ± 9
0.75 160 ± 5 120 ± 8
Quercetin
(Inhibitor of CYP2C8) 2 156 ± 3 120 ± 9
1 - 126 ± 16
10 - 125 ± 7
Furafyline
(Inhibitor of CYP1A2) 30 - 131 ± 7
-: not determined
Table 4. Enzyme kinetic (michalis-menten) parameters estimates of the formation of AG-024322 oxidation and glu-
curonide metabolites using HL-14 human liver microsome preparation.
Formation Kinetics Buffer Km (µM)Vmax
a (pmol/mg/min) CLint
b (ml/min/kg) CLh
c (ml/min/kg)
N-desethyl AG-024322 KPI 10 20 1.86 1.70
Glucuronide M1 KPI 450 35 0.07 0.07
WEM 98 77 0.74 0.72
Glucuronide M2 KPI 9.1 5.3 0.55 0.54
WEM 19 27 1.34 1.26
Glucuronide M3 KPI 7.8 5.2 0.63 0.61
WEM 11 37 3.17 2.74
a. Formation velocity V was estimated from [14C]AG-024322 incubation. Vmax was calculated from V = Vmax x S/(Km+ S), where S is the sub-
strate concentration; b. CLint was scaled up to in vivo by extrapolating in vitro CLint assuming a human liver weight of 1500 g (70 kg body
weight) and that one gram liver tissue contains 45 mg HLM protein; c. CLh was calculated from the well-stirred model: CLh = Q x CLint / Q +
CLint , where Q is the human hepatic blood flow (20 ml/min/kg); KPI: phosphate buffer, pH 7.1, WEM: William E Medium.
W. Z. Zhong et al. / HEALTH 1 (2009) 249-262
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
256
0
200
400
600
800
1000
1200
1400
1600
1800
020406080100 120 140 160
AG-024322 (M)
Pear Area x 10
3
/0.375 mg/15 min
Observed
Predicted
Figure 3. Formation kinetics of N-desethyl AG-024322 in
human liver microsomes over the AG-024322 concentration
range of 0.5 to 100 µM.
binant UGTs for activity and comparative enzyme ki-
netic analysis. Incubations with microsomes from cells
individually expressing human UGT1A enzymes (UGT
supersomes) yield 3 distinct glucuronide products of the
parent drug (M1, M2 and M3), corresponding to the
peaks seen after incubation in human hepatocytes. A
majority involvement of UGT1A1 at 1 and 10 µM of
AG-024322 (Figure 4) in the formation of parent drug
glucuronides, M2 and M3, and minor involvement of
UGT1A8 in the formation of M1 and M3 were observed.
The involvement of other UGT1A and UGT2B enzymes
in glucuronidation of AG-024322 was negligible. The
formation of these three glucuronides was further inves-
tigated in HLM incubated at an AG-024322 concentra-
tion range of 0.5 to 200 µM, with the system optimized
for glucuronidation. The formation of AG-024322 glu-
curonides in HLM incubations was also tested in two
buffer systems, the phosphate buffer (pH 7.1) and WEM.
The data fits and the estimated kinetic parameters for the
Figure 4. Comparison of AG-024322 glucuronidation (UGT)
activity at 1 and 10 µM in microsomes from insect cells indivi-
dually expressing human UGT1A enzymes (UGT supersomes).
050100 150200
0.0
0.5
1.0
1.5
2.0
2.5
050100 150200
0.0
0.5
1.0
1.5
2.0
2.5
050100 150200
0.0
0.1
0.2
0.3
relative velocity (peak area/mg/30 min)
M2
KPI buffer
WME
M3
AG-024322 (µM)
M1
050100 150200
0.0
0.5
1.0
1.5
2.0
2.5
050100 150200
0.0
0.5
1.0
1.5
2.0
2.5
050100 150200
0.0
0.1
0.2
0.3
relative velocity (peak area/mg/30 min)
M2
KPI buffer
WME
M3
AG-024322 (µM)
M1
050100 150200
0.0
0.5
1.0
1.5
2.0
2.5
050100 150200
0.0
0.5
1.0
1.5
2.0
2.5
050100 150200
0.0
0.1
0.2
0.3
relative velocity (peak area/mg/30 min)
M2
KPI buffer
WME
M3
AG-024322 (µM)
M1
050100 150200
0.0
0.5
1.0
1.5
2.0
2.5
050100 150200
0.0
0.5
1.0
1.5
2.0
2.5
050100 150200
0.0
0.1
0.2
0.3
relative velocity (peak area/mg/30 min)
M2
KPI buffer
WME
M3
AG-024322 (µM)
M1
Figure 5. Comparison of formation kinetics of AG-
024322 glucuronides (M1, M2 and M3) in human liver
microsomes (HL-14) using 100 mM phosphate buffer
(KPI) or WEM (glucose 16 mM) in the liver microsome
incubation.
glucuronidation pathways are shown in Figure 5 and
Table 4, respectively. The extrapolated total in vivo he-
patic clearance via parent drug glucuronidation was 1.22
and 4.72 ml/min/kg using the phosphate buffer and
WEM, respectively.
3.4. [14C]AG-024322 Excretion in Rats
After a 30-min IV infusion of [14C]AG-024322 to intact
Sprague-Dawley rats at 10 mg/kg (~50 µCi/250 g ani-
mal), the total recovery of radioactivity dose averaged
84% by 120 hours, as shown in Table 5. The primary
route of excretion of radioactivity was via the feces
W. Z. Zhong et al. / HEALTH 1 (2009) 249-262
SciRes Copyright © 2009 http://www.scirp.org/journal/HEALTH/
257
(76.5%), indicating extensive biliary excretion. A mean
total of 56% of dose was recovered in bile within 48
hours of administration to bile duct-cannulated rats, con-
sisting with the high recovery of radioactivity in feces
from the intact rats. Urinary excretion was minimal with
an average radioactivity recovery of 3.2%. These data
confirmed that the biliary excretion is the major route of
elimination in rats, which is consistent with the findings
from in vitro metabolism studies. The rate of elimination
of radioactivity was moderately rapid and appeared to be
essentially complete by 96 hours postdose (76.1% in
feces and 3.2% in urine). The overall excretion of radio-
activity was similar for males and females.
3.5. Metabolism of AG-024322 in Rats
Profiling and identification of metabolites of AG-024322
in rats were conducted for plasma, urine, fecal and bile
samples from the excretion mass balance study in rats as
described above. AG-024322 underwent extensive me-
tabolism in rats. The representative HPLC-radiochroma-
tograms from rat plasma, urine, feces, and bile are pre-
sented in Figure 6. Two major metabolites were identi-
fied including a direct glucuronide of the parent drug
and M13, a glucuronidation product of M15. M15 is a
desethyl aminated metabolite (an alcohol) of AG-024322.
Other metabolites identified included the N-desethylated
product of AG-024322 (M6), a combined mono-xygen-
ated and dehydrogenated product at the ethyl amine
group of AG-024322 (M16), M15, and the glucuronides
of M6 and M16 (M12 and M14, respectively). The pro-
posed metabolic scheme of AG-024322 in rats is shown
in Scheme 3. The profiles of these metabolites in plasma,
urine, feces, and bile from intact rats and bile-duct can-
nulated rats are presented in Table 6. In plasma, the
Table 5. Recovery (MeanSD) of [14C]AG-024322 in Rats Following 30-min IV Infusion of [14C]AG-024322 at 10
mg/kg (~50 Ci/Rat).
Total Radioactivity Recovered (%)
Sample
Time Interval Male (n=3) Female (n=2)a Overallb
Urine 0-120 h 4.06 0.24 2.4 3.4 0.94
Feces 0-120 h 74.2 6.7 78.8 76.1 6.3
Cage Wash 0-120 h 7.28 12.3 0.31 4.47 9.52
Cage Wipe 0-120 h 0.24 0.21 0.36 0.29 0.23
Intact Rats
Total 0-120 h 85.8 9.4 81.8 84.2 7.7
Bile Duct-Cannulated Rats Bile 0-48 h 55.5 11.2 (n=2)
a. one female rat excluded from calculation of the mean, as a result of difficulties during infusion; b. male and female combined.
1400.0
1200.0
1000.0
800.0
600.0
400.0
200.0
0.0
0.020. 0040.0060.00
Parent Glucuronide
AG-02432 2
M12
M13
M14
M6 M16
DPM
Time (min)
Ur
1.9
1.4
0.9
0.4
-0.1
Openly accessible at
Plasma AG -0 2432 2
Parent Glucuronide
0.0020. 0040.0060.00
Time (min)
ARC(CPM)
i n e
Urine
0.00
700.0
600.0
500.0
400.0
300.0
200.0
100.0
0.0
20.0040.00 60.00
Parent Glucuronide
AG -024322
M12
M13
M14
M6
UN K
M15
M16
Feces
DPM
Time (min)
Feces
Plasma
650.
0
600.0
550.0
500.0
450.0
400.0
350.0
300.0
250.0
200.0
150.0
100.0
50.0
0.0
0.0020.0040.00 60.00
Parent Glucuronide
AG -02 4322
M12
M13
M14 M6
M16
Bile
Bile
DPM
Time (min)
Figure 6. Representative radiochromatograms of plasma, urine, fecal, and bile samples from rats following 30-minutes IV
infusion of [14C]AG-024322 at 10 mg/kg (50 µCi/250 g body weight).
W. Z. Zhong et al. / HEALTH 1 (2009) 249-262
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
258
parent drug was the predominant radioactive component,
accounting for 80-88% of the circulating radioactivity,
whereas the parent glucuronide represented 3-5% of
circulating radioactivity. In urine, the parent drug and
parent glucuronide represented the two major radioactive
components, accounting for 1-1.2% and 0.6-1.2% of the
dose, respectively. In feces, the major drug-related com-
ponents included the parent drug, the parent glucuronide
and M13, accounting for 9-19%, 12-29% and 9-21% of
the dose, respectively. In bile-duct cannulated male rats,
the major biliary drug-related components were the par-
ent drug, the parent glucuronide and M13, accounting
for 16%, 9% and 8% of the dose, respectively.
3.6. Structure Elucidation of In Vivo
Metabolites in Rats
The tentative structures of the metabolites observed in
rats were proposed based on the product ion mass spec-
trum generated for each identified metabolite peak by
ionspray LC-MS/MS. The combined Q1, product ion,
and multiple reaction monitoring scanning techniques
were used for the structure elucidation. The proposed
structure of each metabolite, along with the interpreta-
tion of its MS/MS spectrum is presented in Scheme 3.
The parent glucuronide had an radiochromatogram re-
tention time of 25.4 min and an [M+H]+ ion at m/z 595
(=419+176) consistent with a direct glucuronide of the
parent AG-024322. The product ion mass spectrum
showed major fragment ions at 550, 419, and 374. M6
had a retention time of 38.2 min and showed an [M+H]+
ion at m/z 391 (=419–28) resulted from N-desethylation
of AG-024322. The product ion mass spectrum of M6
gave a major fragment ion of 374. M12 had a retention
time of 19.2 min and an [M+H]+ ion at m/z 567 (=419–
28+176) consistent with a glucuronide of M6. The
product ion mass spectrum of M12 gave fragment ions
of 550, 391 and 374. M15 showed a retention time of
49.2 min, and exhibited [M+H]+ at m/z 392 (=419-27),
which is likely formed through desethyl amination of
AG-024322. The product ion mass spectra gave frag-
ment ions of 392 and 374. M13 exhibited [M+H]+ at
m/z568 (=419-27+176) consistent with a glucuronide of
M15. The retention time of M13 was 27.0 min and the
MS/MS mass spectrum yield fragment ions of 568 and
392. M16 exhibited [M+H]+ at m/z 433 (=419+14), re-
NN
N
N
N
F
NH2
F
Gluc
NN
N
N
N
F
N
F
NN
N
N
N
F
N
F
Gluc
NN
N
N
N
F
NH2
F
N
NN
N
N
N
FF
Gluc
NN
N
N
N
F
OH
F
N
NN
N
N
N
FF
O
Gluc
NN
N
N
N
F
OH
F
NH2C2H5
NH3
NH2C2H5
OH2
O
AG-024322
(m/z 419)
M6
(m/z 391)
M12
(m/z 567)
M13
(m/z 568)
M14
(m/z 609)
M15
(m/z 392)
- 2H
-2H
Parent Glucuronide
(m/z 595)
550 419
374
374
374
374
550 391
374 -
392
433
550 374
415 433
-
374
391
M16
(m/z 433)
Scheme 3. Proposed metabolic scheme of AG-024322 in rats.
W. Z. Zhong et al. / HEALTH 1 (2009) 249-262
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
259
Table 6. Metabolite Profiles of AG-024322 in plasma, urine, feces, and bile from rats administered [14C] AG-024322 in-
travenously.
Mean % Radiochromatogram for Plasma and Mean % Dose for Urine, Feces and Bile
Matrix
(Time) Gender Parent Gluc M6 M12M13 M14UnkAG-024322 M15 M16
Pooled male 4.52 87.70
Plasma
Pooled female2.61 80.06
Male average1.19 0.19 0.090.35 0.09 1.21 0.13
Urine
Female average0.55 0.05 0.05
1.07
Male average11.61 2.80 2.0421.343.161.439.44 3.16 4.36
Feces
Female average28.98 1.04 1.669.39 1.202.9718.97 1.86 1.15
Bile Male average 8.99 2.62 1.148.28 1.19 16.13 4.13
: not detectable using either LC-MS or RAM; : Trace metabolites detected using LC-MS only; Parent Gluc: parent glucuronide, M1,
M2 or M3 (M.W. 594); M6: N-desethylated metabolite of AG-024322 (M.W. 386); M12: Glucuronide of M6 (M.W. 566); M13: Glu-
curonide of M15 (M.W. 567); M14: Glucuronide of M16 (M.W. 608); M15: Desethyl amination metabolite of AG-024322 (M.W. 391);
M16: Combined mono-oxygenation and dehydrogenation product of AG-024322 (M.W. 432); Unk: Unknown/unidentified metabolite.
sulted from a combined mono-oxygenation and dehy-
drogenation of AG-024322. The retention time of M16
was 50 min, and its MS/MS mass spectra gave fragment
ions at 433, 415 and 374. M14 demonstrated [M+H]+ at
m/z 609 (= 419 + 14 + 176) consistent with a glucuron-
ide of M16. M14 had a retention time of 28.3 min and its
mass spectra gave major fragment ions of 609, 550, 433
and 374. The chemical structure of unknown (Unk) can-
not be determined with no mass spectrum data available.
4. DISCUSSIONS
The in vitro metabolism of AG-024322 was initially in-
vestigated in liver microsomes incubated with [14C]AG-
024322 to assess the metabolic stability across species.
The experiments were performed with NADPH only to
reflect Phase 1 metabolic activity. Subsequent studies
were also conducted using hepatocytes, which has a
complete ensemble of metabolizing enzymes of all iso-
forms, cofactors, and cellular components, providing an
alternate assessment of in vivo clearance particularly for
compounds that are mainly cleared via glucuronidation
[10,11]. AG-024322 exhibited the highest metabolic
stability in both human hepatocytes and liver micro-
somes as compared to other nonclinical species (Table
1). Extensive microsomal metabolism in the rat and dog
was observed, which is consistent with the high in vivo
clearance observed in these species. The metabolites
identified in human liver microsomes and hepatocytes
were also observed in the nonclinical species evaluated.
Several in vitro metabolites of AG-024322 were tenta-
tively identified from microsome and hepatocyte incuba-
tions using chromatographic resolution, and concurrent
radiometric and mass-selective quantification (Figure 1;
Scheme 2). These metabolites including the unidentified
metabolites as well as the parent drug account for all the
major peaks of radioactivity in microsomes and hepato-
cytes. The N-desethyl metabolite and glucuronides of the
parent drug were major metabolites observed across
species. The monkey exhibited the highest extent of glu-
curonidation in hepatocytes, while dog and rat demon-
strated more extensive N-desethylation and methylation/
N-acetylation in hepatocytes, and produced novel uni-
dentified metabolites in microsomes. Metabolism of AG-
024322 in human hepatocytes and HLM is more compa-
rable to those in monkeys. Both species demonstrated
glucuronodation and oxidative N-desethylation as the
predominant metabolic pathways.
The excretion mass balance study conducted in rats dem-
onstrated that the primary route of excretion of [14C]AG-
024322 was via feces (76.5%) with minimal contribu-
tions (3.2%) of urinary excretion (Table 5). The high
fecal elimination is reflected by biliary elimination that
accounted for approximately 56% of the total radioactiv-
ity within 48 hours of dosing. As proposed in Scheme 3,
the glucuronide of the parent drug and M6 identified in
vitro were also observed in rats following 30-minute IV
infusion of [14C]AG-024322. Only one parent glucuron-
ide, likely M3 based on the retention time, was observed
in rats, which was consistent with the findings that only
M3 was detected in the rat hepatocyte incubations (Fig-
ure 1 and Table 2). The parent glucuronide appeared to
be the most abundant metabolite in plasma and in all
excreta collected, representing up to 29% of dose in fe-
ces. Another major metabolite, M13, was the glucuroni-
dation product of a desethyl aminated metabolite (M15),
representing up to 21% of dose in feces, as UGTs are
primarily involved in conjugation of metabolites from
oxidation reactions. M6, M15 and M16 were oxidative
metabolites, each representing less than 5% of dose, and
were also further conjugated to its glucuronides. The
metabolite profile in bile (Table 6) is supportive to the
findings of extensive biliary excretion, which involves
conjugation of the parent drug and oxidative metabolites
with glucuronic acid prior to excretion into the bile and
direct secretion of the parent drug into bile.
W. Z. Zhong et al. / HEALTH 1 (2009) 249-262
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
260
Reaction phenotyping studies using the standard bat-
tery of microsomes from insect cells expressing individ-
ual human CYPs or UGTs were conducted to assess the
two principal pathways of AG-024322 biotransformation;
oxidation and glucuronidation. The formation of M6 in
human recombinant demonstrated the predominant role
of CYP3A4 enzyme in the oxidative metabolism of AG-
024322. The relative contribution of the identified CYPs
was subsequently examined by inhibition studies with
CYP-selective inhibitors. As shown in Table 2, only
ketoconazole (selective CYP 3A inhibitor) inhibited N-
desethylation of AG-024322 with up to 86% inhibition
in HLM over an 8-minute of incubation. Quinidine,
quercetin or furafyline had no inhibitory effects in the
formation rate of N-desethylation. Quinidine is also a
known CYP3A heterotropic activator [12], and in this
case yields indirect additional evidence towards pre-
dominant participation of CYP3A enzymes (Table 3).
Formation kinetics of this pathway were also examined
in a set of representative human liver microsomes and
resulted in an apparent Km of 10 µM and Vmax of 20
pmol/mg/min. The in vivo CLint and CLh extrapolated
from the in vitro kinetic data [13] were estimated to be
1.86 and 1.7 ml/min/kg, respectively. According to the
projected human clearance from interspecies scaling (12.8
ml/min/kg, data not shown), the formation of M6 was
estimated to contribute 13% of the hepatic clearance.
Glucuronidation catalyzed by UGTs is one of major
pathways for drug metabolism and elimination in hu-
mans. Identification of the UGTs responsible for glu-
curonidation of AG-024322 in vitro would assist in the
prediction of adverse reactions resulting from drug-drug
interactions or genetic polymorphism. The integrated
approaches used here for UGT reaction phenotyping
were using recombinant enzymes and human liver mi-
crosomes with primary focusing on identification of the
well-characterized hepatic UGTs, lA1, 1A3, 1A4, 1A6,
1A8, 1A10, 2B7 and 2B15. The involvement of UGT1
A1 was demonstrated from the UGT recombinant incu-
bation experiments, resulting in three glucuronides,
which were identical with the M1, M2 and M3 peaks
that also observed in human hepatocyte incubations.
UGT1A1 is a polymorphic isoform responsible for the
glucuronidation of structurally diverse drugs, non-drug
xenobiotics and endogenous compounds (e.g. bilirubin
and estrogens) [14,15]. It is the primary UGT responsi-
ble for bilirubin glucuronidation. UGT1A1 is highly
expressed in liver, gastrointestinal tract, and bladder
[16-18]. It has been reported that genetic polymorphisms
of the UGT1A1 gene were significantly related to severe
toxicity of irinotecan [19-21]. AG-024322 showed high
affinity towards UGT1A1 in vitro. Potential interaction
liability of AG-024322 with drugs metabolized by UGT1
A1 is also an area of great interest and warrants addi-
tional studies. It is of concern if glucuronidation via
UGT1A1 plays a major role in the elimination of AG-
024322 in humans as competitive inhibition of UGT1A1
may be associated with toxicity, and in patients exhibit-
ing compromised bilirubin metabolism [22].
Formation kinetics of the three glucuronides of AG-
024322 were further investigated in the same set of hu-
man liver preparation as that for the formation kinetic
study of oxidative metabolism. Both phosphate buffer
(pH 7.1) and WEM for this system were evaluated.
WEM is a carbonate-based hepatocyte media reagent.
Engtrakul [11] examined the effects of microsome incu-
bation condition on the formation of AZT-glucuronide,
including various physiological and nonphysiological
buffer systems, such as Tris, phosphate, acetate, carbon-
ate, citrate buffers and WEM. Their experiments sug-
gested that both carbonate buffer and WEM produced
significantly higher amounts of AZT-glucuronide than
other buffers. Therefore, we also included the WEM in
the formation kinetic system to compare with the com-
monly used phosphate buffer. Based on the in vitro CLint,
the extrapolated total in vivo CLint for the three glu-
curonides, M1, M2 and M3, was 1.25 and 5.25 ml/
min/kg using phosphate buffer and WEM, respectively,
contributing 10% and 37% of the predicted human
plasma clearance. In agree with the findings by Engtra-
kul et al. [11], we also found that in the case of glu-
curonidation of AG-024322, WEM appeared to be a
more physiologically relevant buffer yielded almost 4-
fold higher intrinsic clearance by glucuronidation than
the phosphate buffer. Additional studies to examine the
impact of WEM, and more importantly, its components
to probe the function of UGTs in vitro are warranted. In
combination of both contributions for CYP3A mediated
N-desethylation and UGT1A1-mediated glucuronidation,
total metabolic clearance could account at best for 50%
of the predicted human plasma clearance using WEM.
This value may be an underestimation since our calcula-
tion did not account for the biliary excretion, formation
of oxidative metabolite other than M6, and glucuronida-
tion of Phase I metabolites, which might be significant
consideration based on the findings from the excretion
study in rats. Caution should be made when extrapolat-
ing metabolic lability via glucuronidation of UGT en-
zymes from HLM, since this system appeared to under-
estimate the degree of clearance (UGT 1A1 is expressed
in human and rat and in general exhibited comparable
substrate specificities and efficiencies (Vmax/Km) of the
glucuronide formation, and thus rat and human UGT1A1
are functionally similar [23,24]. It is expected to have
comparable glucuronidation profiles between rats and
humans in vivo.
In our calculation for the well-stirred model, protein
binding value was not used. AG-024322 was highly
bound to human plasma (95%). If the plasma protein
binding of AG-024322 is included in the expression de-
scribing the well-stirred model of hepatic extraction and
if the scaling-up in vitro intrinsic clearance used without
W. Z. Zhong et al. / HEALTH 1 (2009) 249-262
SciRes Copyright © 2009 Openly accessible at http://www.scirp.org/journal/HEALTH/
261
microsome binding data, the projected in vivo clearance
is drastically lower. Obach [25] examined the in vivo
hepatic clearance predicted from the in vitro system with
the measured in vivo clearance and was found that in
some cases, the clearance predicted from in vitro using
HLM were substantially lower than those observed in
vivo. The compounds exhibiting such a discrepancy were
all characterized by high protein binding. When plasma
protein binding values were removed from the well-
stirred model for these compounds, scaled-up values for
clearance were remarkably close to those measured in
vivo [26]. Therefore, as an initial estimation of the con-
tribution of the two major metabolic pathways in AG-
024322 clearance, the protein binding value was not
used in the well-stirred model, and was found to be more
reasonable in the clearance projection.
5. CONCLUSIONS
Investigation of in vitro and in vivo metabolism of
AG-024322 demonstrates that UGT1A1 mediated glu-
curonidation and CYP3A mediated oxidation represent
the major metabolic pathways of AG-024322. The esti-
mated contribution of the glucuronidation to total plasma
clearance in humans was approximately 3-fold higher
than that from desethylation of AG-024322. Using the
WE M buffer system in human liver microsomes to as-
sess the metabolic pathway by glucuronidation improved
the clearance predictability of AG-024322. Because of
the polymorphism of UGT1A1 and the reported toxicity
associated with competitive inhibition of UGT1A1 either
by a co-administered drug or in patients exhibiting com-
promised bilirubin metabolism, this information was
used to influence the clinical study design to only in-
clude subjects having normal expression of UGT1A1 in
the first human study thereby decreasing the toxicity risk
to patients.
6. ACKNOWLEDGMENTS
Authors would like to thanks Drs. Hai-Zhi Bu and Deepak Dalvie for
helpful discussion in structure elucidation of AG-024322 metabolites,
Drs. Weiwei Tan and Rachel Courtney for the discussion of the human
study design and sharing the human pharmacokinetic data with us, and
Drs. Scott Fountain, Bill Smith, Ellen Wu and Bhasker Shetty for their
invaluable comments and support of this project.
7. ABBREVIATIONS
CDK, cyclin-dependent kinase; HLM, human liver mi-
crosome; CYP, cytochrome P450; UGT, UDP-glucurono
syltransferase; UDPGA, UDP-glucuronic acid; WEM,
Williams E medium; HPLC, high-performance liquid
chromatography; MS, mass spectrometry; min, min-
ute(s); IS, internal standard; RAM, radiochemical activ-
ity monitor.
REFERENCES
[1] Zhang, C.C., Kephart, S., McAlpine, I., Nonomiya, J.,
Higgins, J., Arango, M.E., Yan, Z., Knighton, D., Ferre,
R.A., Tikhe, J., et al. (2005) AG-024322 is a potent and
selective multi-targeted CDK inhibitor with broad spec-
trum anti-proliferative activity. Proc Amer Assoc Cancer
Res, 46, Abstract #4415.
[2] Morgan, D.O., Fisher, R.P., Espinoza, F.H., Farrell, A.,
Nourse, J., Chamberlin, H. and Jin, P. (1998) Control of
eukaryotic cell cycle progreesion by phosphorylation of
cyclin-dependent kinases. Cancer J Sci Am, 4, S77-83.
[3] Morgan, D.O. (1997) Cyclin-dependent kinases: engines,
clocks, and micoprosessors. Annu Rev Cell Biol, 13,
261-291.
[4] Moriarty, K.J., Koblish, H., Johnson, D.L., Galemmo, J.
and Robert, A. (2007) Progress in the development of
agents. Top Med Chem, 1, 207-291.
[5] Zhang, C.C., Troche, G., Yen, Z., Arango, M.E., Higgins,
J., Romero, D., Kephart, S., McAlpine, I., Koudriakova,
T. and J. Skaptason, et al. (2005) AG-024322 is a multi-
targeted CDK inhibitor with potent antitumor activity in
vivo. Proc Amer Assoc Cancer Res, 46, Abstract #4413.
[6] Fisher, M.B., Campanale, K., Ackermann, B.L., Vanden-
Branden, M. and Wrighton, S.A. (2000) In vitro glu-
curonidation using human liver microsomes and the
pore-forming peptide alamethicin. Drug Metab Dispos,
28, 560-566.
[7] Hamilton, R.A., Garnett, W.R. and Kline, B.J. (1981)
Determination of mean valproic acid serum level by as-
say of a single pooled sample. Clin Pharmacol Ther, 29,
409-413.
[8] Venkatakrishnan, K., von Moltke, L.L., Court, M.H.,
Harmatz, J.S., Crespi, C.L. and Greenblatt, D.J. (2000)
Comparison between cytochrome P450 (CYP) content
and relative activity approaches to scaling from DNA-
expressed CYPs to human liver microsomes: Ratios of
accessory proteins as sources of discrepancies between
the approaches. Drug Metab Dispos, 28, 1493-1504.
[9] Stormer, E., von Moltke L.L. and Greenblatt, D.J. (2000)
Scaling drug biotransformation data from cDNA-ex-
pressed cytochrome P-450 to human liver: A comparison
of relative activity factors and human liver abundance in
studies of mirtazapin metabolism. J Pharmacol Exp Ther,
295, 793-801.
[10] Gomez-Lechon, M.J., Donato, M.T., Castell J.V. and
Jover, R. (2004) Human hepatocytes in primary culture:
the choice to investigate drug metabolism in man. Curr
Drug Metab, 5, 443-462.
[11] Engtrakul, J.J., Foti, R.S., Strelevitz, T.J. and Fisher, M.B.
(2005) Altered AZT (3'-azido-3'-deoxythymidine) glu-
curonidation kinetics in liver microsomes as an explana-
tion for underprediction of in vivo clearance: Comparison
to hepatocytes and effect of incubation environment.
Drug Metab Dispos, 33, 1621-1627.
[12] Tang, W. and Stearns, R.A. (2001) Heterotropic coopera-
tivity of cytochrome P450 3A4 and potential drug-drug
interactions. Current Drug Metabolism, 2, 185-198.
[13] Houston, J.B. (2001) Utility of in vitro drug metabolism
W. Z. Zhong et al. / HEALTH 1 (2009) 249-262
SciRes Copyright © 2009 http://www.scirp.org/journal/HEALTH/Openly accessible at
262
data in predicting in vivo metabolic clearance. BioChem
Pharmacol, 47, 1469-1479.
[14] Jansen, P.L., Mulder, G.J., Burchell, B. and Bock, K.W.
(1992) New developments in glucuronidation research:
Report of a workshop on “glucuronidation, its role in
health and disease”. Hepatology, 15, 532-544.
[15] Beutler, E. Gelbart, T. and Demina, A. (1998) Racial
variability in the UDP-glucuronosyltransferase 1
(UGT1A1) promoter: A balanced polymorphism for
regulation of bilirubin metabolism? Proc Natl Acad Sci,
U.S.A., 95, 8170-8174.
[16] Strassburg, C.P., Kneip, S. Topp, J., Obermayer-Straub, P.,
Barut, A., Tukey, R.H. and Manns, M.P. (2000) Poly-
morphic gene regulation and interindividual variation of
UDP-glucuronosyltransferase activity in human small
intestine. J Biol Chem, 275, 36164-36171.
[17] Tukey R.H. and Strassburg, C.P. (2000) Human UDP-
glucuronosyltransferases: Metabolism, expression, and
disease. Annu Rev Pharmacol Toxicol, 40, 581-616.
[18] Nakamura, A., Nakajima, M., Yamanaka, H., Fujiwara R.
and Yokoi, T. (2008) Expression of UGT1A and UGT2B
mRNA in human normal tissues and various cell lines.
Drug Metab Dispos, 60, 1461-1464.
[19] Ando, Y., Saka, H., Ando, M., Sawa, T., Muro, K., Ueoka,
H., Yokoyama, A., Saitoh, S., Shimokata, K. and Hase-
gawa, Y. (2000) Polymorphisms of UDP- glucuronosyl-
transferase gene and irinotecan toxicity: A pharmacoge-
netic analysis, Cancer Res, 60, 6921-6926.
[20] Mani, S. (2001) UGT1A1 polymorphism predicts iri-
notecan toxicity: Evolving proof. AAPS PharmSci, 3,
1208-1303.
[21] Lyer, L., Hall, D., Das, S., Mortell, M.A., Ramirez, J.,
Kim, S., Di Rienzo, A. and Ratain, M.J. (1999) Pheno-
type-genotype correlation of in vitro SN-38 (active me-
tabolite of irinotecan) and bilirubin glucuronidation in
human liver tissue with UGT1A1 promoter polymor-
phism. Clin Pharmacol Ther, 65, 576-582.
[22] Kadakol, A., Ghosh, S.S., Sappal, B.S., Sharma, G.,
Chowdhury, J.R. and Chowdhury, N.R. (2000) Genetic
lesions of bilirubin urdine-diphosphoglucuronate glu-
curonosyltrabsferase (UGT1A1) causing Crigler-Najjar
and Gilbert syndromes: correlation of genotype to phe-
notype. Hum Mutat, 16, 297-306.
[23] King, C.D., Green, M.D., Rios, G.R., Coffman, B.L.,
Owens, I.S., Bishop, W.P. and Tephly, T.R. (1996) The
glucuronidation of exogenous and endogenous com-
pounds by stably expressed rat and human UDP-glu-
curonosyltrasferase 1.1. Arch Biochem Biophys, 332,
92-100.
[24] King, C.D., Rios, G.R., Green, M.D. and Tephly, T.R.
(2000) UDP-glucuronosyltrasferase. Curr Drug Metab, 1,
143-161.
[25] Obach, R.S. (1996) The importance of nonspecific bind-
ing in in vitro matrices, its impact on enzyme kinetic
studies of drug metabolism reactions, and implications
for in vitro-in vivo correlations. Drug Metab Dispos, 24,
1047-1049.
[26] Obach, R.S. (1997) Nonspecific binding to microsomes:
Impact on scale-up of in Vitro Intrinsic clearance to he-
patic clearance as assessed through examination of war-
farin, imipramine, and propranolol. Drug Metab Dispos,
25, 1359-1369.