Validation of GLP Time-Dependent
Cytochrome P450 Inhibition Assays
with Assays with LC/MS Detection
Application Note 478
1
Elke S. Perloff,
1
Andrew Mason,
1
Shangara S. Dehal,
1
Andrew P. Blanchard,
1
Ling Morgan,
1
Andre Dandeneau,
2
Thuy Ho,
1
Ronell M. Crocker,
1
Catherine M. Chandler,
2
Nathalie Boily,
2
Charles L. Crespi, and
2
David M. Stresser
1
BD Biosciences – Woburn, MA
2
Corning Life Sciences – Tewksbury, MA
Abstract
Time-dependent inhibition of cytochrome P450 (CYP)
metabolism can lead to nonlinear pharmacokinetics and drug-
drug interactions. Recent guidance from the USFDA and drug
failures attributable to toxicity that may have a basis in time-
dependent inhibition have elevated interest in this long-known
undesirable property of drug candidates. We have validated
cytochrome P450 inhibition assays in pooled human liver
microsomes that incorporate IC
50
, IC
50
shifts, K
I
, K
i
and k
inact
endpoints. The assays incorporate CYP-isoform selective probe
substrates, LC/MS/MS analysis using stable-labeled isotope
internal standards, positive and negative control inhibitors, low
total protein to minimize non-specic binding and a step-wise
analysis designed to improve K
inact
experimental design. Our
data was found to compare well with literature values. Among
the enzymes, probes and inhibitors validated, with enhanced
consideration for CYP3A4 because of its major role in drug
metabolism and its propensity to exhibit substrate-dependent
inhibition response, were as follows: CYP1A2/phenacetin/alpha
naphthoavone/furafylline; CYP2C9/diclofenac/sulfaphenazole/
tienilic acid; CYP2C19/S mephenytoin/S-benzylnirvanol; CYP2D6/
dextromethorphan/quinidine/paroxetine; CYP3A4/testosterone/
midazolam/ketoconazole/azamulin/verapamil/diltiazem.
Introduction
Drug candidate failures resulting from metabolism-based drug
interactions are now becoming infrequent, attributable to
robust and routine screening procedures aimed at eliminating
potent inhibitors of cytochrome P450 (CYP)a. Addressing drug
toxicity as a cause of drug failures is now a top-priority. In turn,
this has driven more focus on screening for mechanism-based
CYP inhibitors. This is because mechanism-based inhibitors can
be associated with toxicity, ostensibly as a result of reactive
metabolite formation and subsequent covalent binding to
cellular macromolecules. Moreover, recent guidance from the
USFDA has advocated testing for this endpoint
1
. Although this
testing is not required to be performed according to GLP as
outlined in Title 21 CFR pt. 58, many pharmaceutical companies
prefer a conservative approach to in vitro drug-drug interaction
testing and therefore, this was incorporated as a success criteria
for this project.
Analytical Methods
Metabolites were quantitated by LC/MS using authentic
reference standards obtained from Corning Life Sciences or
Sigma-Aldrich. To control for ion suppression and provide optimal
quantitation, stable-labeled isotopes, obtained from Corning
Life Sciences, were used as internal standards. Metabolites
were quantied using an ABI/MDS Sciex 4000 Q TRAP
system,
equipped with TurboIonSpray
®
source. Samples were injected
onto a C18 column [Waters
®
Symmetry
®
(C18, 2.1x50 mm,
5 µm)] with a mobile phase consisting of 0.1% FA in H
2
O,
0.1% FA in ACN at a ow rate of 0.4 mL/min. Run times
were <3.5 min. Parameters for analytical method validation,
including mass transitions, range, interday precision are shown
in Table 1. All standard curves were prepared in a matrix of
0.1 mg/mL pooled HLM (Cat. No. 452161) in potassium
phosphate buffer pH 7.4 containing an NADPH regeneration
system. All methods were validated in accordance with the FDA
Bioanalytical Method Validation Guidance, 2001.
Assay Methods
Assays were conducted in 0.1 M potassium phosphate buffer
pH 7.4 in a volume of 400 µL. For IC
50
shift experiments, test
articles were incubated with HLM with and without NADPH for
10 or 30 min prior to transfer of 40 µL to 360 µL into a secondary
incubation containing probe substrate at a concentration at or
near the K
m
and supplemental NADPH. Secondary incubations
proceeded according to the parameters in Table 2. Reactions
were terminated by addition of 0.1% FA in ACN containing IS
to give nal concentrations between 0.02 and 2 µM, which
in general is in the lower range of each metabolite standard
curve. The ratio of the IC
50
value determined with incubations
lacking NADPH in the preincubation compared to same with
NADPH in preincubation are reported as a “shift”. For K
I
and
K
inact
experiments, at multiple time-points, 40 µL aliquots were
transferred into secondary incubations as above, except the
substrate concentration was approximately 5X the K
m
. The K
I
and k
inact
values were determined using non-linear regression
(SigmaPlot™ v. 8.0 equipped with Enzyme Kinetics module v 1.1).
2
Enzyme Substrate Metabolite
Mass
Transition Internal Standard
Mass
Transition
Ioniz-
ation
LLOQ
(µM) RE
Standard Curve Range and Interday Precision
CV% Range R2 RE CV
CYP1A2 Phenacetin Acetaminophen 151111 Acetaminophen-[
13
C
2
15
N] 155110 ESI+ 0.0760 106 11.9 0.076-5.0 0.9988 98-101 5.3
CYP2C9 Diclofenac 4'-OH Diclofenac 312268 4'-OH Diclofenac-[
13
C
6
] 316272 ESI - 0.0087 107 4.20 0.0087-2.0 0.9998 99-102 3.4
CYP2C19 S-mephenytoin 4'-OH S-Mephenytoin 235150 4'-OH S-Mephenytoin-[D3] 238150 ESI+ 0.0040 101 10.4 0.004-10.0 0.9979 97-103 9.8
CYP2D6 Dextromethorphan Dextrorphan 258157 Dextrorphan-[D3] 261157 ESI+ 0.0025 94 7.90 0.0025-1.0 0.9975 94-103 7.9
CYP3A4 Midazolam 1'-OH Midazolam 342203 1'-OH Midazolam-[
13
C
3
] 347208 ESI+ 0.0025 93 10.0 0.0025-1.0 0.9967 93-112 13.2
CYP3A4 Testosterone 6β-OH Testosterone 305269 6β-OH Testosterone-[D7] 312276 ESI+ 0.0160 107 5.3 0.016-10.0 0.9994 98-102 7.2
Table 1. Analytical method parameters
Enzyme Substrate K
m
Model [S] Inc time (min) HLM (mg/mL) Competitive inhibitor IC
50
a
(nM) K
i
a
(nM) Model for K
i
CYP1A2 Phenacetin 37 MM 40 15 0.1 α-Naphthoavone 13, 12 18, 20 Mixed
CYP2C9 Diclofenac 3.7 MM 5 5 0.05 Sulfaphenazole 410, 630 200, 190 Competitive
CYP2C19 S-mephenytoin 43 MM 40 20 0.3 S-Benzylnirvanol 440, 310 130 Competitive
CYP2D6 Dextromethorphan 4.9 MM 5 5 0.1 Quinidine 58, 65 58, 41 Competitive
CYP3A4 Midazolam 2.2 MM 3 5 0.02 Ketoconazole 13, 19 8.6, 9.2 Mixed
CYP3A4 Testosterone 65
b
Hill 50 10 0.05 Ketoconazole 18, 19 24, 18 Competitive
Table 2. Reversible Inhibition Assay Parameters, IC
50
and K
i
values
a – values determined in duplicate on independent days unless a single value is shown
b – K
s
, Hill coefcient n = 1.3