You are on page 1of 6

J. vet. Pharmacol. Therap. 33, 50–55, doi: 10.1111/j.1365-2885.2009.01101.x.

Canine CYP2B11 metabolizes and is inhibited by anesthetic agents often


co-administered in dogs

M. T. BARATTA* Baratta, M. T., Zaya, M. J., White, J. A., Locuson, C. W. Canine CYP2B11
M. J. ZAYA metabolizes and is inhibited by anesthetic agents often co-administered in dogs.
J. vet. Pharmacol. Therap. 33, 50–55.
J. A. WHITE &
C. W. LOCUSON Medetomidine is a well-established sedative and analgesic for dogs and cats. As
a premedicant for anesthesia regimens that also include other agents,
*Pfizer Global Research and Development, medetomidine can also provide a dose-sparing effect. While there are likely
Pharmacokinetics, Dynamics, and several reasons for the dose-sparing effect of medetomidine, the role of
Metabolism, Creve Coeur, MO, USA; metabolic drug–drug interactions at the single enzyme level has not yet been
Pfizer Animal Health, Veterinary Medicine examined. Using a panel of individually expressed canine cytochromes P450
Research and Development, Metabolism and
cloned from beagle liver, this report demonstrates that medetomidine is an
Safety, Kalamazoo, MI, USA
extremely potent CYP2B11 inhibitor (IC50 < 10 nM) and that ketamine and
midazolam are CYP2B11 substrates with high intrinsic clearances. These
in vitro findings suggest that under some circumstances, medetomidine (i.e.
‘perpetrator’) may inhibit the metabolic clearance of some high metabolic
clearance drugs (i.e. ‘victims’) with which it is commonly co-administered via
the CYP2B11 pathway. However, as the dose-sparing effect of medetomidine
premedication commonly results in anesthetic dose reduction, any increased
plasma concentrations of victim drugs caused by medetomidine would still be
lower than if no dose reduction had been made. Further studies are needed to
characterize whether medetomidine possesses the potential to cause pharma-
cokinetic interactions. In conclusion, the ability of recombinant P450s to define
canine-specific drug clearance pathways and P450 inhibitors should prove
useful in identifying drug combinations that may require dose adjustments in
dogs.
(Paper received 4 February 2009; accepted for publication 29 April 2009)
Charles W. Locuson, Pfizer Animal Health, Veterinary Medicine Research and
Development, Metabolism and Safety, 333 Portage Street, KZO-300-421NW,
Kalamazoo, MI 49001, USA. E-mail: charles.w.locuson@pfizer.com

INTRODUCTION (Crespi & Miller, 1999; McGinnity et al., 2008; Youdim et al.,
2008). However, the availability of expressed canine P450
The growing companion animal health market will likely (cytochrome P450) isoforms has been limited and the relative
encourage veterinary pharmaceutical companies to expand their importance of the canine P450s to canine drug metabolism as a
development of therapeutics for dogs (Evans & Chapple, 2002). whole is largely unknown, in part, because of the lack of
Assuming that instances of polypharmacy in dogs will increase characterization of canine P450s.
with more drugs being developed for companion animals, the Recently, several of the major hepatic canine P450s were
Drug Metabolism ⁄ Pharmacokinetics Scientist will require more co-expressed with canine P450 oxidoreductase in Escherichia coli
tools to assess the potential risks of concomitant medications by the authors (Locuson et al., 2009). During inhibition profiling
altering drug exposure in dogs. Currently, there are few with a panel of the canine rP450s, the authors identified the
pharmaceuticals that have been adequately characterized with anesthetic medetomidine as a potent inhibitor of CYP2B11.
canine enzymes to support their use as specific inhibitors or While the risk of drug–drug interactions involving human
substrates to use in dog interaction studies. For example, one can CYP2B enzymes appears to be low because of minimal involve-
find pharmacokinetic drug interaction studies, but the actual ment in drug oxidation and low hepatic expression (Walsky
enzyme(s) implicated have generally been inferred by analogy to et al., 2006), canine CYP2B11 has shown surprisingly high
human enzymes. One tool that is increasingly being used to link concentrations of activity in vitro towards drugs used in dogs,
pharmacokinetic drug–drug interactions to specific metabolic such as benzodiazepines (Shou et al., 2003). Hence, inhibition of
pathways is rP450 (recombinant, expressed cytochrome P450) CYP2B11 by chloramphenicol (Ciaccio et al., 1987), loperamide,

50  2009 Blackwell Publishing Ltd


Canine CYP2B11 metabolizes anesthetic agents 51

ketoconazole, diazepam, and fluoxetine (Aidasani et al., 2008) Autosampler (Canboro, NC, USA) connected to a Applied
may raise legitimate drug interaction questions in canine Biosystems MDS ⁄ Sciex API 4000 triple quadrupole mass spec-
medicine. Canine CYP2B11 is also expressed at reasonable trometer (Foster City, CA, USA). Detection was accomplished by
concentrations in dog liver, estimated at 10% of total P450 injection of 10 lL of supernatant onto a Phenomenex Syner-
(Eguchi et al., 1996), which would allow it to contribute to drug gi 2.1 · 50 mm Polar-RP 80 Å analytical column (Torrance,
clearance more readily in the dog vs. human CYP2B isoforms. CA, USA) with mobile phases of 0.1% formic acid in water
It is reported here that midazolam and ketamine, which are (mobile phase A) and acetontrile (mobile phase B) using a step
often co-administered with the CYP2B11 inhibitor medetomi- gradient (10% B for 0.4 min, to 90% B in 0.1 min) at a flow rate
dine, demonstrated clearance by rCYP2B11 according to of 0.6 mL ⁄ min. The effluent from the LC was introduced into the
phenotyping experiments. These findings may provide a partial API 4000 mass spectrometer via a turbo ionspray source. Mass
explanation for the dose-sparing effect of medetomidine preme- spectrophotometric analysis was performed in positive ion mode
dication on ketamine and other maintenance anesthetics using reaction monitoring at unit resolution for the transitions of
[reviewed by Cullen (1996)]. The results also demonstrate how 178.8 fi 101.0, 239.0 fi 126.0, 200.9 fi 115.1,
rP450s can be used in drug–drug interaction assessment in 326.9 fi 291.2, 212.8 fi 183.1 and 343.0 fi 230.0 for
veterinary medicine and reinforce the hypothesis that CYP2B11 propofol, ketamine, medetomidine, midazolam, atipamezole, and
may be an important clearance pathway in dogs. triazolam (internal standard), respectively.

CLint,app determination
MATERIALS AND METHODS
In vitro intrinsic clearance (CLint,app) was determined using a
substrate depletion approach as described (Youdim et al., 2008),
Materials
but using 0.1 M potassium phosphate buffer (pH 7.4),
The isomers of medetomidine were synthesized at Pfizer (Sand- 150 pmol ⁄ mL of rP450s or 0.5 mg ⁄ mL beagle liver microsomes,
wich, UK). Commercial formulations of medetomidine (Domi- and 1 mM sodium b-NADPH rather than a NADPH regenerating
tor), ketamine (Ketaset), and atipamezole (Antisedan) were system. Drugs were dissolved DMSO to a final stock concentra-
purchased from Pfizer research veterinarians. Midazolam, prop- tion of 10 mM and further diluted in buffer to achieve a final
ofol, dimethyl sulfoxide (DMSO), b-NADPH, and monobasic and incubation of substrate at 1.0 lM. Liquid handling was con-
dibasic potassium phosphate were purchased from Sigma- ducted on a Beckman Coulter Biomek 2000 (Fullerton, CA, USA)
Aldrich (St Louis, MO, USA). Formic acid (88%) was purchased with a water-filled aluminum plate holder maintained at 37 C.
from Mallinckrodt Baker, Inc. (Paris, KY, USA). High-perfor- Incubations (0.7 mL) were carried out in a ‘one-pot’ method in a
mance liquid chromatography grade solvents were purchased 1.0 mL capacity 96-deep-well titer plate with aliquots removed
from Honeywell Burdick & Jackson (Morristown, NJ, USA). to a quench plate containing 25 lL of formic acid (25 %) with
Canine rP450 Bactosomes (Cypex Ltd., Dundee, Scotland, UK) 500 ng ⁄ mL of triazolam at 0, 5, 10, 20, 30, and 60 min.
were prepared as described (Locuson et al., 2009). Beagle liver Midazolam and ketamine reactions used sampling times of 0, 1,
microsomes purchased from Xenotech, LLC (Lenexa, KS, USA) 2, 5, 10, and 15 min because of their short half-lives, and
were mixed gender from a pool of 23 subjects. regression was only conducted with the first three time points
because of the drug concentrations dropping below 1% remain-
ing. The elimination rate constant and CYP or microsomal
IC50 determination
protein content was then used to calculate CLint,app as
Inhibitor concentration (IC50) values for medetomidine and lL ⁄ min ⁄ pmol rP450 or lL ⁄ min ⁄ mg dog liver microsomes.
atipamezole were determined against phenacetin de-ethylation, Testosterone was used as a positive control for liver microsome
temazepam N-demethylation, diclofenac 4¢-hydroxylation, buf- activity.
uralol 1¢-hydroxylation, and midazolam 1¢-hydroxylation by the
canine rP450s 1A1, 2B11, 2C21, 2D15, and 3A12, respectively.
Inter-system extrapolation factor activity correction
The incubations were conducted as previously described with 12
inhibitor concentrations and substrate concentrations equal to The specific activity of each rP450 preparation might vary
their Km values (Aidasani et al., 2008). Tolbutamide (50 ng ⁄ mL) because of different concentrations of redox partner proteins (e.g.
was used as the internal standard in the acetonitrile quenching NADPH P450 reductase), the lack of inter-isoform P450-P450
solution. protein interactions, and different concentrations of non-P450
proteins that change the nonspecific protein binding of drugs. To
‘normalize’ an observed rP450 activity to those of liver
LC ⁄ MS ⁄ MS analysis
microsomes using the known hepatic expression concentration
Liquid chromatography (LC) and mass spectrometry (MS) of each CYP isoform, inter-system extrapolation factor (ISEF)
detection of all test compounds analyzed for assay endpoints methods were developed to increase the accuracy of in vitro–
was accomplished using a Perkin Elmer Series 200 micro-pump in vivo extrapolation of metabolic clearance (Proctor et al.,
system (Waltham, MA, USA) and a Leap Technologies HTC PAL 2004). Proctor et al. (2004) originally described three levels of

 2009 Blackwell Publishing Ltd


52 M. T. Baratta et al.

rigor in which an ISEF can be determined. We have used a now routine in human drug metabolism phenotyping (Zhang
simple substrate depletion CLint,app approach rather than V or et al., 2007) and drug interaction screening (Trubetskoy et al.,
V ⁄ Km, and did not factor in nonspecific protein binding. The 2005), but canine recombinant CYP1A, 2B, and 2D isoforms
primary goal was to ensure that the relative clearance values for were not previously available for veterinary use. Furthermore,
a drug across the panel of rP450s did not greatly over- or few canine isoform-specific inhibitors, which could be used to
underestimate the role of a particular rP450 isoform. Equation 1 characterize drug metabolism in liver microsomes, have been
was used to determine the CLint ISEF value for each rP450 defined. This led us to report on the cloning and preparation of
isoform with each substrate. several canine rP450s (Locuson et al., 2009).

CLint DLMðlL/min/mg DLMÞ


CLint ISEF ¼ ð1Þ
CLint rCYPðlL/min/pmol CYPÞ  DLM CYP abundanceðpmol/mg DLMÞ

Recently, in-house rP450 screening identified the alpha-2


Inter-system extrapolation factor values were then multiplied by adrenergic receptor agonist medetomidine as a particularly
the rP450 CLint values to arrive at the corrected activities. Liver potent CYP2B11 inhibitor of temazepam N-demethylation
expression concentrations of the dog CYP were taken from (Table 1). The structurally similar reversal agent atipamezole
published findings where immunoquantitation of CYP1A, often used to antagonize the effects of medetomidine also
CYP2B, CYP2C, and CYP3A estimated liver concentrations of inhibited CYP2B11 much more than the other canine rP450s.
17, 48, 160, and 69 pmol ⁄ mg, respectively (Eguchi et al., This was not unexpected as submicromolar IC50s for medetom-
1996), and 160 pmol ⁄ mg for CYP2D (Sakamoto et al., 1995). idine were reported previously in human liver microsomes
Therefore, it has been assumed that the average CYP expression (Kharasch et al., 1992), but the use of rP450s definitively
concentrations from pooled beagle liver microsomes are similar identified the P450 isoform inhibited in dogs. Both medetomidine
between various sources of laboratory beagles even though the and atipamezole were found to induce type-II difference binding
expression variability has not been thoroughly characterized to spectra with rCYP2B11 (data not shown). This suggested that
our knowledge. ISEF values bracketed unity with a range of these compounds likely derive some of their high affinity from
0.77–8.2. the imidazole group coordinating as the sixth ligand to the heme
prosthetic group (Jefcoate, 1978).
Many sedation ⁄ anesthetic agents like medetomidine are
Spectrophotometric studies
almost always used in combination to provide balanced anes-
Spectral binding titration studies were carried out with CYP2B11 thesia. Therefore, the in vitro clearance of commonly employed
Bactosomes (1 lM P450) in 100 mM potassium phosphate buffer anesthetics was analyzed with rP450s to determine whether
pH 7.4. Incubation mixtures were evenly divided into 1.5 mL medetomidine might inhibit their metabolism. Midazolam,
10 mm black-walled cuvettes, and experiments were performed propofol, and ketamine were chosen because they are used
at room temperature by titrating in 1 lL aliquots of Atipamezole extensively in veterinary medicine (Hellebrekers & Sap, 1997;
(DMSO solution) into a sample cuvette (0.02–20 lM), with an Kojima et al., 2002; Seliskar et al., 2007), and several studies
equal aliquot of DMSO in the reference cuvette (DMSO < 1% have examined the effects of combining these anesthetics on
v ⁄ v). After each titration, cuvettes were mixed and allowed to sit cardiac output, respiration, blood gases, and other physiological
for 1 min to allow the samples to reach equilibrium. Difference parameters [reviewed by (Cullen (1996)]. Furthermore, there is
spectra were recorded between 350 and 500 nm using a Hitachi existing evidence that propofol (Hay Kraus et al., 2000) and
(Danbury, CT, USA) U3300 dual-beam spectrophotometer and ketamine (Hijazi & Boulieu, 2002) are partly metabolized by
data were analyzed using UV SOLUTIONS 1.2 software (Hitachi). canine or human microsomal CYP2B isoforms and this allowed
The difference in absorbance between the peak (430 nm) and the opportunity to compare these findings with canine rP450s.
trough (390 nm) for type II spectra were then plotted vs. Most of the drugs tested were substrates of canine CYP 2B11,
titrant concentration and fit to a hyperbolic curve function for 2C21, 2D15, and 3A12 to varying degrees. This suggestion is
estimation of Ks values using GRAPH PAD PRISM 4.0 software based on the substrate depletion experiments conducted at a low
(San Diego, CA, USA). substrate concentration to mimic conditions of linear pharma-
cokinetics (Table 2). However, medetomidine, atipamezole, and
propofol were turned over much slower than midazolam and
RESULTS AND DISCUSSION ketamine with rP450s and dog liver microsomes. This is
consistent with other pathways like glucuronidation contribut-
As P450s are often involved in the clearance of drugs, ing to the clearance of medetomidine (Kaivosaari et al., 2002)
characterizing the specific P450 enzyme(s) that metabolize or and propofol (Soars et al., 2001; Favetta et al., 2002). Propofol
are inhibited by co-administered drugs is critical in assessing the 4-hydroxylation by dog liver microsomes is likely to be carried
risk of pharmacokinetic drug interaction. The use of rP450s is out by CYP2B11 (Hay Kraus et al., 2000), yet it is not clear why

 2009 Blackwell Publishing Ltd


Canine CYP2B11 metabolizes anesthetic agents 53

Table 1. Inhibition of canine rP450 isoforms


Enzyme IC50 (lM)*
by medetomidine and atipamezole
Inhibitor CYP1A1 CYP2B11 CYP2C21 CYP2D15 CYP3A12

rac-Medetomidine 1.1 ± 0.2 0.0048 ± 0.0006 22 ± 4 1.1 ± 0.3 15 ± 4


S-Medetomidine ND 0.034 ± 0.003 ND ND ND
Atipamezole 0.60 ± 0.09 0.0072 ± 0.0013 18 ± 5 0.089 ± 0.014 5.5 ± 1.9

ND, not determined. *Values determined by fitting a titration of twelve inhibitor concentrations
conducted in triplicate to a sigmoidal function; ± values represent the standard error of the fit.

Table 2. Turnover of anesthetics by canine rP450s and liver microsomes

In vitro CLint,app (lL ⁄ min ⁄ pmol, rP450) or (lL ⁄ min ⁄ mg, microsomes)*

Substrate CYP1A1 CYP2B11 CYP2C21 CYP2D15 CYP3A12 DLM

Midazolam ND 2.72 1.08 ND ND 1066


rac-Ketamine ND 26.7 1.24 ND 16.5 1009
Propofol ND ND ND ND ND ND
Atipamezole ND 0.082 0.123 0.141 ND 61.1
S-medetomidine ND 0.047 0.091 0.200 ND 65.5
R-medetomidine ND 0.628 0.262 0.968 ND 104

Half-lives were not extrapolated more than two-fold greater than the value of the last incubation time point of 15 or 60 min (<0.15 lL ⁄ min ⁄ pmol and
<0.039 lL ⁄ min ⁄ pmol). DLM, dog liver microsomes; ND, not detected. *Calculated from a mean elimination rate constant as described in Materials and
methods.

propofol clearance was not demonstrated in our studies since the attributed to the intended pharmacology of medetomidine or
rP450s and liver microsomes were active with other substrates. changes in pharmacokinetics that are unrelated to metabolism.
Low rates of metabolism can prevent measuring substrate Despite the high affinity of medetomidine and atipamezole for
disappearance because of the difficulty in monitoring small CYP2B11, there are also several factors that are likely to
changes in large mass spectrometry signals (e.g. drug concen- minimize their potential for pharmacokinetic interactions with
trations near 1 lM). It is speculated that had we monitored benzodiazepines and ketamine. One factor that likely reduces
formation of hydroxy-propofol it would have been detected, but pharmacokinetic interactions is simply the careful study of
upon calculating rates, would lead to the same result of low anesthesia regimens. Doses of anesthetics given i.v. or by
metabolic conversion. In case the rP450 clearances differed from inhalation are dosed to achieve the desired pharmacodynamic
microsome clearances because of varying redox partner protein effect. This ensures any unpredicted concentration of respiratory,
concentrations and P450–P450 protein interactions, the cardiovascular, or central nervous system depression achieved
CLint,app ISEF method was applied (Proctor et al., 2004). with varying combinations of premedication and induction
According to both corrected and uncorrected in vitro clearance agents is not exacerbated by overdosing the maintenance agent.
values, medetomidine and atipamezole behaved as CYP2B11 Medetomidine is also extremely potent and requires only low
inhibitors because of their high affinity and low turnover. lg ⁄ kg doses, and all of the sedatives ⁄ anesthetics discussed here
Conversely, midazolam and ketamine were excellent CYP2B11 have short half-lives minimizing the time over which P450-
substrates according to high turnover rates (Tables 2 and 3). mediated interactions can occur in vivo. Finally, CYP3A12 and
The above in vitro findings suggest medetomidine and atipa- 2C21 would be expected to compensate for ketamine and ⁄ or
mezole might alter the pharmacokinetics of co-administered midazolam clearance in the event of CYP2B11 inhibition in vivo
midazolam and ketamine via inhibition of CYP2B11. A significant (Tables 2 and 3).
portion of midazolam and ketamine clearance is determined by It is worth noting that the active isomer dexmedetomidine
P450 metabolism (Kaka & Hayton, 1980; Thummel et al., 1996). (S-isomer) provides the same efficacy as medetomidine at half the
However, the authors are not aware of any in vivo studies that have dose (Gomez-Villamandos et al., 2006; Granholm et al., 2007).
examined plasma concentrations of midazolam and ketamine Since levomedetomidine (R-isomer) is largely devoid of alpha-2
when dogs are co-administered with medetomidine or atipamez- adrenergic receptor activity (MacDonald et al., 1991; Kuusela
ole. Studies have documented dose-sparing effects of medetomi- et al., 2001), the use of dexmedetomidine (Dexdomitor, Orion
dine premedication on propofol and ketamine (as maintenance Pharma, Espoo, Finland) would be expected to further decrease
agents; Cullen, 1996), and with inhalation anesthetics (Bloor the risk for CYP2B11 inhibition in vivo vs. medetomidine.
et al., 1992; Gomez-Villamandos et al., 2008). In these cases, the Dexmedetomidine was even measured to be a weaker inhibitor
reduced general anesthesia requirements have previously been than medetomidine in agreement with findings in human liver
microsomes where levomedetomidine was slightly more potent

 2009 Blackwell Publishing Ltd


54 M. T. Baratta et al.

Table 3. ISEF-corrected rP450 isoform activ-


Corrected in vitro CLint,app (lL ⁄ min ⁄ pmol)*
ities
Substrate CYP1A1 CYP2B11 CYP2C21 CYP2D15 CYP3A12

Midazolam NC 22.2 6.66 NC NC


rac-Ketamine NC 20.6 6.19 NC 14.3
Propofol NC NC NC NC NC
Atipamezole NC 1.27 0.382 0.382 NC
S-medetomidine NC 1.36 0.409 0.409 NC
R-medetomidine NC 2.17 0.650 0.650 NC

*Activities from Table 2 were corrected to provide clearance estimates that more closely mimic those
of liver microsomes using published expression concentrations for each CYP isoform. Not calculated
because of lack of detectable turnover.

than dexmedetomidine (Kharasch et al., 1992). Since our source medetomidine that are expected to minimize metabolic
of medetomidine was the formulated product, we cannot rule out interactions.
an added inhibition effect of paraben formulation stabilizers. We
would point out, though, that the medetomidine formulation is
primarily aqueous and it is estimated that dilutions required for ACKNOWLEDGMENTS
IC50 analysis would result in paraben concentrations below
where inhibition effects would be expected (<1.0 lM) (van Gelder The authors thank Dr Scott Brown for support of this research,
et al., 2002). As for atipamezole, its interaction with other Dr David Martin for providing anesthesia expertise, and Dr
anesthetics could theoretically slow patient recovery time, but Kuresh Youdim, Paul Guimond, and C. Locuson, CRNA for
this has not been studied. helpful discussions. We also thank Dr Dawn Merritt and Drs M
Although CYP2B11 has been implicated in propofol clearance Rinkinen, J Salonen, B McKusick, M Huhtinen, H Kaukinen, R
in dogs (Hay Kraus et al., 2000), liver microsomes and rP450s Virtanen, and J Aspegren at Orion Pharma for reviewing the
suggested it is low. However, in this case, there is some evidence manuscript, Richard Nicholas for help with literature searches,
that a pharmacokinetic interaction does not occur during co- and Dr Steven Nederveld for providing anesthetics.
administration of propofol and medetomidine. One study has
demonstrated that the propofol half-life was actually reduced
when co-administered with medetomidine vs. a propofol-only REFERENCES
treatment because of a change in volume of distribution and no
change in plasma clearance (Hall et al., 1994). As expected, total Aidasani, D., Zaya, M.J., Malpas, P.B. & Locuson, C.W. (2008) In vitro
plasma propofol concentrations were lower in dogs receiving drug-drug interaction screens for canine veterinary medicines: evalu-
ation of cytochrome P450 reversible inhibition. Drug Metabolism and
both drugs as less propofol is required to maintain anesthesia
Disposition, 36, 1512–1518.
with medetomidine premedication. By this account, the potential
Bloor, B.C., Frankland, M., Alper, G., Raybould, D., Weitz, J. & Shurtliff,
for a propofol–medetomidine interaction because of pharmaco- M. (1992) Hemodynamic and sedative effects of dexmedetomidine in
kinetics is low. dog. Journal of Pharmacology and Experimental Therapeutics, 263, 690–
In summary, in vitro studies with a panel of canine rP450s 697.
suggest medetomidine and atipamezole are CYP2B11 inhibitors Buhrer, M., Mappes, A., Lauber, R., Stanski, D.R. & Maitre, P.O. (1994)
and that ketamine and midazolam are CYP2B11 substrates. Dexmedetomidine decreases thiopental dose requirement and alters
distribution pharmacokinetics. Anesthesiology, 80, 1216–1227.
Commonly used doses of medetomidine in dogs do result in
Ciaccio, P.J., Duignan, D.B. & Halpert, J.R. (1987) Selective inactivation
plasma drug concentrations near the CYP2B11 IC50 value, but
by chloramphenicol of the major phenobarbital-inducible isozyme of
the exact-free hepatic medetomidine concentrations ‘seen’ by dog liver cytochrome P-450. Drug Metabolism and Disposition, 15, 852–
P450s in vivo are difficult to estimate. Therefore, further 856.
investigation is required to determine whether drug interactions Crespi, C.L. & Miller, V.P. (1999) The use of heterologously expressed
involving some anesthestics used for companion animal sedation drug metabolizing enzymes – state of the art and prospects for the
occur with CYP2B11, which then translate to altered pharma- future. Pharmacology and Therapeutics, 84, 121–131.
Cullen, L.K. (1996) Medetomidine sedation in dogs and cats: a review of
codynamics. In anesthesia, such a result could be viewed as a
its pharmacology, antagonism and dose. British Veterinary Journal,
double-edged sword – the dose-sparing effect can often improve 152, 519–535.
patient outcome, but may simultaneously increase the risk for Eguchi, K., Nishibe, Y., Baba, T. & Ohno, K. (1996) Quantitation of
drug interactions when anesthetics or other surgical drugs are cytochrome P450 enzymes (CYP1A1 ⁄ 2, 2B11, 2C21 and 3A12) in
combined without any knowledge of their drug interaction dog liver microsomes by enzyme-linked immunosorbent assay. Xeno-
potential. However, the dose-to-effect practice of anesthesia, biotica, 26, 755–763.
overlapping pharmacological activities of anesthetics, and Evans, T. & Chapple, N. (2002) The animal health market. Nature
Reviews. Drug Discovery, 1, 937–938.
altered drug volume of distribution (Buhrer et al., 1994) are
currently the reported explanations for the dose-sparing effects of

 2009 Blackwell Publishing Ltd


Canine CYP2B11 metabolizes anesthetic agents 55

Favetta, P., Degoute, C.S., Perdrix, J.P., Dufresne, C., Boulieu, R. & Locuson, C.W., Ethell, B.T., Voice, M., Lee, D. & Feenstra, K.L. (2009)
Guitton, J. (2002) Propofol metabolites in man following propofol Evaluation of Escherichia coli membrane preparations of canine
induction and maintenance. British Journal of Anaesthesia, 88, 653– CYP1A1, 2B11, 2C21, 2C41, 2D15, 3A12, and 3A26 with
658. co-expressed canine P450 reductase. Drug Metabolism and Disposition,
van Gelder, J., Deferme, S., Naesens, L., De Clercq, E., van den Mooter, G., 37, 457–461.
Kinget, R. & Augustijns, P. (2002) Intestinal absorption enhancement MacDonald, E., Scheinin, M., Scheinin, H. & Virtanen, R. (1991) Com-
of the ester prodrug tenofovir disoproxil fumarate through modulation parison of the behavioral and neurochemical effects of the two optical
of the biochemical barrier by defined ester mixtures. Drug Metabolism enantiomers of medetomidine, a selective alpha-2-adrenoceptor ago-
and Disposition, 30, 924–930. nist. Journal of Pharmacology and Experimental Therapeutics, 259, 848–
Gomez-Villamandos, R.J., Palacios, C., Benitez, A., Granados, M.M., 854.
Dominguez, J.M., Lopez, I., Ruiz, I., Aguilera, E. & Santisteban, J.M. McGinnity, D.F., Waters, N.J., Tucker, J. & Riley, R.J. (2008) Integrated in
(2006) Dexmedetomidine or medetomidine premedication before vitro analysis for the in vivo prediction of cytochrome P450-mediated
propofol-desflurane anaesthesia in dogs. Journal of Veterinary Pharma- drug-drug interactions. Drug Metabolism and Disposition, 36, 1126–
cology and Therapeutics, 29, 157–163. 1134.
Gomez-Villamandos, R.J., Palacios, C., Benitez, A., Granados, M.M., Do- Proctor, N.J., Tucker, G.T. & Rostami-Hodjegan, A. (2004) Predicting
minguez, J.M., Estepa, J.C., Ruiz, I., Aguilera, E. & Santisteban, J.M. drug clearance from recombinantly expressed CYPs: intersystem
(2008) Effect of medetomidine infusion on the anaesthetic require- extrapolation factors. Xenobiotica, 34, 151–178.
ments of desflurane in dogs. Research in Veterinary Science, 84, 68–73. Sakamoto, K., Kirita, S., Baba, T., Nakamura, Y., Yamazoe, Y., Kato, R.,
Granholm, M., McKusick, B.C., Westerholm, F.C. & Aspegren, J.C. (2007) Takanaka, A. & Matsubara, T. (1995) A new cytochrome P450 form
Evaluation of the clinical efficacy and safety of intramuscular and belonging to the CYP2D in dog liver microsomes: purification, cDNA
intravenous doses of dexmedetomidine and medetomidine in dogs and cloning, and enzyme characterization. Archives of Biochemistry and
their reversal with atipamezole. Veterinary Record, 160, 891–897. Biophysics, 319, 372–382.
Hall, L.W., Lagerweij, E., Nolan, A.M. & Sear, J.W. (1994) Effect of Seliskar, A., Nemec, A., Roskar, T. & Butinar, J. (2007) Total intravenous
medetomidine on the pharmacokinetics of propofol in dogs. American anaesthesia with propofol or propofol ⁄ ketamine in spontaneously
Journal of Veterinary Research, 55, 116–120. breathing dogs premedicated with medetomidine. Veterinary Record,
Hay Kraus, B.L., Greenblatt, D.J., Venkatakrishnan, K. & Court, M.H. 160, 85–91.
(2000) Evidence for propofol hydroxylation by cytochrome P4502B11 Shou, M., Norcross, R., Sandig, G., Lu, P., Li, Y., Lin, Y., Mei, Q.,
in canine liver microsomes: breed and gender differences. Xenobiotica, Rodrigues, A.D. & Rushmore, T.H. (2003) Substrate specificity and
30, 575–588. kinetic properties of seven heterologously expressed dog cytochromes
Hellebrekers, L.J. & Sap, R. (1997) Medetomidine as a premedicant for p450. Drug Metabolism and Disposition, 31, 1161–1169.
ketamine, propofol or fentanyl anaesthesia in dogs. Veterinary Record, Soars, M.G., Riley, R.J., Findlay, K.A., Coffey, M.J. & Burchell, B. (2001)
140, 545–548. Evidence for significant differences in microsomal drug glucuronida-
Hijazi, Y. & Boulieu, R. (2002) Contribution of CYP3A4, CYP2B6, and tion by canine and human liver and kidney. Drug Metabolism and
CYP2C9 isoforms to N-demethylation of ketamine in human liver Disposition, 29, 121–126.
microsomes. Drug Metabolism and Disposition, 30, 853–858. Thummel, K.E., O’Shea, D., Paine, M.F., Shen, D.D., Kunze, K.L., Perkins,
Jefcoate, C.R. (1978) Measurement of substrate and inhibitor binding to J.D. & Wilkinson, G.R. (1996) Oral first-pass elimination of midazolam
microsomal cytochrome P-450 by optical-difference spectroscopy. involves both gastrointestinal and hepatic CYP3A-mediated metabo-
Methods in Enzymology, 52, 258–279. lism. Clinical Pharmacology and Therapeutics, 59, 491–502.
Kaivosaari, S., Salonen, J.S. & Taskinen, J. (2002) N-Glucuronidation of Trubetskoy, O.V., Gibson, J.R. & Marks, B.D. (2005) Highly miniaturized
some 4-arylalkyl-1H-imidazoles by rat, dog, and human liver micro- formats for in vitro drug metabolism assays using vivid fluorescent
somes. Drug Metabolism and Disposition, 30, 295–300. substrates and recombinant human cytochrome P450 enzymes.
Kaka, J.S. & Hayton, W.L. (1980) Pharmacokinetics of ketamine and two Journal of Biomolecular Screening, 10, 56–66.
metabolites in the dog. Journal of Pharmacokinetics and Biopharmaceu- Walsky, R.L., Astuccio, A.V. & Obach, R.S. (2006) Evaluation of 227
tics, 8, 193–202. drugs for in vitro inhibition of cytochrome P450 2B6. Journal of Clinical
Kharasch, E.D., Herrmann, S. & Labroo, R. (1992) Ketamine as a probe Pharmacology, 46, 1426–1438.
for medetomidine stereoisomer inhibition of human liver microsomal Youdim, K.A., Zayed, A., Dickins, M., Phipps, A., Griffiths, M., Darekar,
drug metabolism. Anesthesiology, 77, 1208–1214. A., Hyland, R., Fahmi, O., Hurst, S., Plowchalk, D.R., Cook, J., Guo, F.
Kojima, K., Nishimura, R., Mutoh, T., Hong, S.H., Mochizuki, M. & & Obach, R.S. (2008) Application of CYP3A4 in vitro data to
Sasaki, N. (2002) Effects of medetomidine-midazolam, acepromazine- predict clinical drug-drug interactions; predictions of compounds as
butorphanol, and midazolam-butorphanol on induction dose of objects of interaction. British Journal of Clinical Pharmacology, 65, 680–
thiopental and propofol and on cardiopulmonary changes in dogs. 692.
American Journal of Veterinary Research, 63, 1671–1679. Zhang, H., Davis, C.D., Sinz, M.W. & Rodrigues, A.D. (2007) Cytochrome
Kuusela, E., Vainio, O., Kaistinen, A., Kobylin, S. & Raekallio, M. (2001) P450 reaction-phenotyping: an industrial perspective. Expert Opinion
Sedative, analgesic, and cardiovascular effects of levomedetomidine on Drug Metabolism & Toxicology, 3, 667–687.
alone and in combination with dexmedetomidine in dogs. American
Journal of Veterinary Research, 62, 616–621.

 2009 Blackwell Publishing Ltd

You might also like