throbber
Research Paper
`
`Mechanism of cytochrome P450-3A inhibition
`by ketoconazolejphp_1202
`
`214..221
`
`David J. Greenblatta, Yanli Zhaoa, Karthik Venkatakrishnanb,
`Su X. Duana, Jerold S. Harmatza, Sarah J. Parenta, Michael H. Courta
`and Lisa L. von Moltkec
`
`aDepartment of Pharmacology and Experimental Therapeutics, Tufts University School of Medicine and
`Tufts Medical Center, Boston, bMillennium Pharmaceuticals Inc. and cGenzyme Corp., Cambridge, MA,
`USA
`
`Abstract
`Objectives Ketoconazole is extensively used as an index inhibitor of cytochrome P450-3A
`(CYP3A) activity in vitro and in vivo, but the mechanism of ketoconazole inhibition of
`CYP3A still is not clearly established.
`Methods Inhibition of metabolite formation by ketoconazole (seven concentrations from
`0.01 to 1.0 mm) was studied in human liver microsomes (n = 4) at six to seven substrate
`concentrations for triazolam, midazolam, and testosterone, and at two substrate concentra-
`tions for nifedipine.
`Key findings Analysis of multiple data points per liver sample based on a mixed
`competitive–noncompetitive model yielded mean inhibition constant Ki values in the range
`of 0.011 to 0.045 mm. Ketoconazole IC50 increased at higher substrate concentrations,
`thereby excluding pure noncompetitive inhibition. For triazolam, testosterone, and mida-
`zolam a-hydroxylation, mean values of a (indicating the ‘mix’ of competitive and noncom-
`petitive inhibition)
`ranged from 2.1 to 6.3. However,
`inhibition of midazolam
`4-hydroxylation was consistent with a competitive process. Determination of Ki and a based
`on the relation between 50% inhibitory concentration values and substrate concentration
`yielded similar values. Pre-incubation of ketoconazole with microsomes before addition of
`substrate did not enhance inhibition, whereas inhibition by troleandomycin was significantly
`enhanced by pre-incubation.
`Conclusions Ketoconazole inhibition of triazolam a- and 4-hydroxylation, midazolam
`a-hydroxylation, testosterone 6b-hydroxylation, and nifedipine oxidation appeared to be a
`mixed competitive–noncompetitive process, with the noncompetitive component being
`dominant but not exclusive. Quantitative estimates of Ki were in the low nanomolar range for
`all four substrates.
`Keywords competitive inhibition; cytochrome P450-3A; ketoconazole; mechanism-based
`inhibition; noncompetitive inhibition
`
`Introduction
`Ketoconazole is extensively used as an ‘index’ inhibitor of human cytochrome P450-3A
`(CYP3A) isoforms in the academic, regulatory, and drug development communities.[1–3]
`Although ketoconazole has been used in this context for some twenty years, the kinetic
`mechanism of CYP3A inhibition by ketoconazole in human liver microsomal preparations
`still is not clearly established. Published in-vitro studies of ketoconazole inhibition of
`CYP3A have yielded differing and conflicting conclusions as to whether inhibition can be
`explained by competitive, noncompetitive, or mixed competitive–noncompetitive mecha-
`nisms.[4] Accurate assignment of the mechanism of inhibition affects understanding of the
`fundamental nature of the substrate–enzyme and inhibitor–enzyme interactions, as well as
`the interpretation of in-vitro inhibition constants (Ki), the validity of which is dependent on
`the correctness of the underlying model of inhibition.[5]
`We have evaluated the mechanism of CYP3A inhibition by ketoconazole in human liver
`microsomal preparations. To our knowledge this is among the few studies to assess this
`question using CYP3A substrates from different categories, with a sufficient number of
`
`JPP 2011, 63: 214–221
`© 2011 The Authors
`JPP © 2011 Royal
`Pharmaceutical Society
`Received July 27, 2010
`Accepted September 9, 2010
`DOI
`10.1111/j.2042-7158.2010.01202.x
`ISSN 0022-3573
`
`Correspondence: David J.
`Greenblatt, Department of
`Pharmacology and,
`Experimental Therapeutics, Tufts
`University School of Medicine,
`136 Harrison Ave, Boston, MA
`02111, USA.
`E-mail: dj.greenblatt@tufts.edu
`
`214
`
`1
`
`TEVA1024
`
`

`

`Ketoconazole inhibition of CYP3A
`
`David J. Greenblatt et al.
`
`215
`
`substrate and inhibitor concentrations such that enzyme
`kinetic analysis could proceed without a predetermined deci-
`sion as to whether the inhibition mechanism was competitive,
`noncompetitive, or mixed.[6]
`
`Materials and Methods
`Preparation of human liver microsomes
`Liver samples from four individual human donors with no
`known liver disease were provided by the International Insti-
`tute for the Advancement of Medicine (Exton, PA, USA); the
`Liver Tissue Procurement and Distribution System, Univer-
`sity of Minnesota (Minneapolis, MN, USA); or the National
`Disease Research Interchange (Philadelphia, PA, USA). The
`ages of the four donors were: 21, 34, 37, and 62 years. All
`were male and the cause of death was either physical trauma
`or sudden illness.
`ultracentrifugation;
`by
`prepared
`Microsomes were
`microsomal pellets were suspended in 0.1 m potassium phos-
`phate buffer containing 20% glycerol and were stored at
`-80°C until use.[7]
`
`Preparation of reagents
`incubations and
`Chemical reagents used for microsomal
`HPLC analysis were purchased from commercial sources.
`Triazolam, midazolam,
`testosterone, nifedipine, ketocona-
`zole, and troleandomycin (triacetyloleandomycin) were pur-
`chased from commercial sources or kindly provided by their
`pharmaceutical manufacturers.
`Stock solutions of the drug entities were prepared in
`methanol, and subsequently diluted with methanol as needed.
`Solutions were stored at -18°C.
`
`In-vitro study design
`Four CYP3A substrates were selected for study: triazolam,
`midazolam, testosterone and nifedipine.[8,9] The choice of sub-
`strates was based on categories proposed by Kenworthy
`triazolam a-hydroxylation and
`studies of
`et al.[6] For
`4-hydroxylation, midazolam a-hydroxylation
`and
`4-
`hydroxylation, and testosterone 6b-hydroxylation,
`incuba-
`tions were performed at multiple substrate concentrations. At
`each concentration, additional incubations were done with
`co-addition of ketoconazole at concentrations of 0, 0.01,
`0.025, 0.05, 0.1, 0.25, 0.5 or 1.0 mm. Within this range keto-
`conazole is established as a relatively specific CYP3A inhibi-
`tor. Studies were done with microsomal preparations from
`
`four different human liver samples and all individual incuba-
`tions were performed in duplicate.
`Nifedipine dehydrogenation studies were done at two fixed
`concentrations of 25 and 250 mm. Incubations were performed
`with co-addition of varying concentrations of ketoconazole as
`described above.
`For testosterone as substrate, an additional study was
`conducted to evaluate the effect of 20 min pre-incubation of
`microsomes with ketoconazole, prior to the addition of test-
`osterone (100 mm) on the inhibitory effect of ketoconazole at
`concentrations of 0.05 and 0.25 mm. Troleandomycin was
`used as the positive control in this study. Troleandomycin
`concentrations were 0, 1, 5, 10, 25, and 50 mm.
`
`Incubation procedures
`Incubation mixtures contained 50 mm phosphate buffer, 5 mm
`MgCl2, 0.5 mm nicotinamide adenine dinucleotide phosphate,
`and an isocitrate and isocitric dehydrogenase regenerating
`system.[7,8] Substrates (Table 1) and ketoconazole or trolean-
`domycin, as appropriate, were added to a series of incubation
`tubes. The solvent was evaporated to dryness at 40°C under
`mild vacuum conditions.
`After addition of the incubation mixture components to
`yield a final volume of 250 ml, reactions were initiated by the
`addition of microsomal protein (0.1–0.25 mg/ml). The incu-
`bation duration ranged from 5 to 40 min, depending on the
`intrinsic clearance of the substrate.[8,10–12] All reactions were
`shown to be in the linear range with respect to protein con-
`centration and incubation duration.
`At the appropriate time reactions were stopped by cooling
`on ice and the addition of 100 ml acetonitrile. Internal stan-
`dard was added, the incubation mixture was centrifuged, and
`the supernatant was transferred to an autosampling vial for
`HPLC analysis of metabolites (Table 1). The analytic column
`was stainless steel, 15 cm ¥ 3.9 mm, containing reverse-phase
`C18 Novapak (Waters Associates, Milford, MA, USA)
`(Table 1).
`
`Analysis of data
`For triazolam, midazolam, and testosterone, analyses were
`performed for each individual liver sample using combina-
`tions of reaction velocity (V) at each substrate concentration
`([S]) and inhibitor concentration ([I]). The following equa-
`tion, consistent with mixed competitive–noncompetitive inhi-
`bition, was fitted to the data points by nonlinear regression
`using SAS PROC NLIN:[13–15]
`
`Table 1 Summary of reaction characteristics and chromatographic conditions
`
`Substrate
`
`Product(s)
`
`Internal standard
`
`Mobile phase composition
`
`Ultraviolet absorbance
`wavelength
`
`Triazolam
`
`Midazolam
`
`Testosterone
`Nifedipine
`
`a-OH-triazolam,
`4-OH-triazolam
`a-OH-midazolam,
`4-OH-midazolam
`6b-OH-testosterone
`Oxidized nifedipine
`
`Phenacetin
`
`Phenacetin
`
`Androstenedione
`Diazepam
`
`aPotassium dihydrogen phosphate (pH unadjusted).
`
`70% buffera (10 mm), 20% acetonitrile,
`10% methanol
`45% buffera (10 mm), 20% acetonitrile,
`35% methanol
`55% water, 45% acetonitrile
`45% water, 55% methanol
`
`220 nm
`
`220 nm
`
`240 nm
`270 nm
`
`2
`
`

`

`216
`
`Journal of Pharmacy and Pharmacology 2011; 63: 214–221
`
`a∞ 5
`
`0
`
`20
`
`10
`
`5
`
`12
`
`100
`
`50
`
`20
`
`10
`
`C50/Ki
`
`5I
`
`2 1
`
`0.1 0.2
`
`0.5
`
`1
`
`2
`
`5
`[S]/Km
`
`10
`
`20
`
`50 100
`
`Figure 1 For a mixed competitive–noncompetitive inhibitor, the rela-
`tionship between the [S]/Km ratio (x-axis) and the IC50/Ki ratio, at varying
`values of a ranging from 1.0 to ‘infinity’. The functions were predicted
`based on equation 4.
`
`⋅[ ]
`max S
`+
`
`(1)
`
`⎞⎠⎟
`
`+ [ ]
`I
`K
`
`i
`
`1
`
`⎛⎝⎜
`
`m
`
`K
`
`⎞⎠⎟
`
`i
`
`V K
`
`[ ] + [ ]
`I
`
`⋅α
`
`1
`
`⎛⎝⎜
`
`S
`
`=
`
`V
`
`Iterated variables were: Vmax, the maximum reaction veloc-
`ity; Km, the Michaels–Menten constant; Ki, the inhibition con-
`stant; and a, a number ⱖ 1.0 indicating the ‘mix’ of
`competitive and noncompetitive mechanisms. In the case of
`testosterone, a sigmoidal (Hill) kinetic pattern was observed
`without and with the addition of ketoconazole. For purposes
`of nonlinear regression analysis of testosterone data, equa-
`tion 1 was empirically modified by addition of an exponent to
`[S] and to S50 (replacing Km).
`For determination of 50% inhibitory concentration (IC50)
`values, reaction velocities with co-addition of inhibitor were
`expressed as a percentage ratio (Rv) of the control velocity
`with no inhibitor present. The relationship of Rv to inhibitor
`concentration ([I]) was analysed by nonlinear regression
`to determine the IC50 value, based on the following
`equation:
`
`=
`
`R
`v
`
`100 1
`
`−
`
`max
`b
`
`b
`
`b
`
`(2)
`
`For triazolam, midazolam and testosterone, ketoconazole
`IC50 values were available at multiple substrate concentra-
`tions. Using mean Km (or S50) values determined as described
`above (eqn 1) as fixed entries in equation 4, this equation was
`fitted to data points ([S] and IC50) by nonlinear regression.
`Iterated variables were a and Ki. For nifedipine, the same
`procedure was applied using the available data points, with
`the Km value from a previously reported study.[8]
`
`Results
`Nonlinear regression based on equation 1 (modified by an
`exponent in the case of testosterone) yielded convergent solu-
`tions (Figures 2 and 3).
`For triazolam, Km values for the 4-OH-triazolam pathway
`the a-OH-triazolam pathway
`for
`exceeded Km values
`(Table 2). This was similar to previous studies from our labo-
`ratory and elsewhere. [8,10,11,17] Ketoconazole inhibition of tria-
`zolam metabolite formation was consistent with the mixed
`competitive–noncompetitive scheme (Figure 2). The mean
`values of a were 2.11 and 2.14 for the a-hydroxylation and
`4-hydroxylation pathways, respectively, indicating a predomi-
`nance of noncompetitive inhibition in the balance.
`Consistent with previous reports, Km values for the mida-
`zolam a-hydroxylation pathway were on average more than
`tenfold lower than for the 4-hydroxylation pathway (Table 2,
`of a-OH-
`inhibition
`Figure 3).[8,11,12,18,19] Ketoconazole
`midazolam formation was explained by the mixed model,
`with a mean a of 6.30. However, inhibition of midazolam
`4-hydroxylation was consistent with a competitive process,
`inasmuch as estimates of a from nonlinear regression were
`very large.
`testosterone 6b-hydroxylation
`As described previously,
`a
`sigmoidal
`(Hill)
`kinetic
`was
`best
`described
`by
`pattern.[5,8,20–25] Inhibition by ketoconazole was consistent with
`the mixed competitive–noncompetitive model
`(Figure 2).
`
`⎞ ⎠⎟
`
`[ ]
`I
`E
`[ ] +
`I
`IC
`
`⎛ ⎝⎜
`
`Iterated variables were: Emax, the maximum degree of inhi-
`bition; IC, the inhibitor concentration producing an Rv value
`of 50% of (100 – Emax); and b, an exponent. The actual IC50
`was calculated as:
`
`IC50
`
`=
`
`IC
`
`(
`
`2
`
`E
`
`max
`
`−
`
`)
`1 1
`
`b
`
`(3)
`
`IC50 values were determined based on mean data
`points across four liver samples at corresponding inhibitor
`concentrations.
`The relationship between IC50 and substrate concentration
`can be used as an alternative method for determining a and Ki
`for mixed competitive–noncompetitive inhibition.[16] That
`relationship is:
`
`(4)
`
`⎞⎠⎟
`
`⎞⎠⎟
`
`+ [ ]
`1
`+ [ ]
`
`⋅α
`
`⎛⎝⎜
`
`K
`
`i
`
`1
`
`⎛⎝⎜
`
`IC
`
`50
`
`=
`
`S K S K
`
`m
`
`m
`
`This is illustrated in Figure 1. Note that for pure competi-
`tive inhibition (a = ‘infinity’) this reduces to:
`+ [ ]
`
`⎞⎠⎟
`
`S K
`
`m
`
`1
`
`⎛⎝⎜
`
`IC
`
`50
`
`=
`
`K
`
`i
`
`(5)
`
`whereas for pure noncompetitive inhibition (a = 1.0), this
`reduces to:
`
`50 = K
`
`IC
`
`i
`
`(6)
`
`3
`
`

`

`Ketoconazole inhibition of CYP3A
`
`David J. Greenblatt et al.
`
`217
`
`Keto (mM)
`
`0 0
`
`.01
`
`0.025
`
`0.05
`
`0.1
`
`0.25
`0.5
`1.0
`
`Vmax = 3.36
`S50 = 70.5 mM
`Ki = 0.025 mM
`a = 3.15
`
`(b)
`
`3.5
`
`3.0
`
`2.5
`
`2.0
`
`1.5
`
`1.0
`
`0.5
`
`(relative velocity units)
`
`6b-OH-testosterone fromation rate
`
`Keto (mM)
`
`0 0
`
`.01
`
`0.025
`
`0.05
`
`0.1
`
`0.25
`0.5
`1.0
`
`Vmax = 1.02
`Km = 72 mM
`Ki = 0.032 mM
`a = 2.06
`
`(a)
`
`1.0
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`(relative velocity units)
`
`a-OH-triazolam formation rate
`
`0
`
`100
`
`300
`200
`Triazolam (mM)
`
`400
`
`500
`
`0
`
`100
`
`300
`200
`Testosterone (mM)
`
`400
`
`500
`
`Figure 2 Rates of formation of a-OH-triazolam from triazolam (a) and formation of 6b-OH-testosterone from testosterone (b) in a human liver
`sample. Lines represent the functions determined by nonlinear regression based on equation 1. For testosterone, equation 1 was empirically modified
`by addition of an exponent to account for sigmoidal kinetics.
`
`(a)
`
`0.35
`
`0.30
`
`Vmax = 0.296
`Km = 3.38 mM
`Ki = 0.007 mM
`a = 7.16
`
`(b)
`
`0.21
`
`Vmax = 0.287
`Km = 48.8 mM
`Ki = 0.0175 mM
`
`Keto (mM)
`
`Keto (mM)
`
`0 0
`
`.01
`
`0.025
`
`0.05
`
`0.1
`
`0.25
`0.5
`1.0
`
`0
`
`20
`
`60
`40
`Midazolam (mM)
`
`80
`
`100
`
`0.18
`
`0.15
`
`0.12
`
`0.09
`
`0.06
`
`0.03
`
`(relative velocity units)
`
`4-OH-midazolam formation rate
`
`0 0
`
`.01
`
`0.025
`
`0.05
`
`0.1
`
`0.25
`0.5
`1.0
`
`0
`
`20
`
`60
`40
`Midazolam (mM)
`
`80
`
`100
`
`0.25
`
`0.20
`
`0.15
`
`0.10
`
`0.05
`
`(relative velocity units)
`
`a-OH-midazolam formation rate
`
`Figure 3 Rates of formation of a-OH-midazolam (a) and 4-OH-midazolam (b) from midazolam in a human liver sample. Lines represent the
`functions determined by nonlinear regression based on equation 1.
`
`Table 2 Enzyme kinetic parameters for in-vitro biotransformation of cytochrome P450-3A substrates with co-addition of ketoconazole as inhibitor
`
`Substrate
`
`Product
`
`Triazolam
`
`Midazolam
`
`Testosterone
`Nifedipine
`
`a-OH-triazolam
`4-OH-triazolam
`a-OH-midazolam
`4-OH-midazolam
`6b-OH-testosterone
`Oxidized nifedipine
`
`Km (mm)
`
`63.1 (⫾ 3.8)
`319 (⫾ 45)
`3.34 (⫾ 0.15)
`51.3 (⫾ 8.2)
`69 (⫾ 7)a
`–
`
`Based on equation 1 (mean ⫾ SE, n = 4)
`a
`Ki (mm)
`
`Based on equation 4
`a
`
`Ki
`
`0.031 (⫾ 0.003)
`0.045 (⫾ 0.005)
`0.011 (⫾ 0.004)
`0.019 (⫾ 0.003)
`0.019 (⫾ 0.002)
`–
`
`2.14 (⫾ 0.13)
`2.11 (⫾ 0.43)
`6.30 (⫾ 1.77)
`b
`3.07 (⫾ 0.23)
`–
`
`0.041
`0.052
`0.0083
`0.019
`0.028
`0.072
`
`1.51
`1.41
`8.54
`b
`
`2.11
`4.25
`
`aS50. bValues of a for 4-OH-midazolam formation were very large, consistent with competitive inhibition.
`
`4
`
`

`

`218
`
`Journal of Pharmacy and Pharmacology 2011; 63: 214–221
`
`TST = 50 mM (IC50 = 0.030 mM)
`TST = 500 mM (IC50 = 0.056 mM)
`
`100
`
`80
`
`60
`
`40
`
`20
`
`(% control without inhibitor)
`
`6b-OH-testosterone fromation rate
`
`Triazolam = 50 mM (IC50 = 0.040 mM)
`Triazolam = 500 mM (IC50 = 0.065 mM)
`
`0.01 0.02
`
`0.2
`0.1
`0.05
`Ketoconazole (mM)
`
`0.5
`
`1
`
`0.01 0.02
`
`0.2
`0.1
`0.05
`Ketoconazole (mM)
`
`0.5
`
`1
`
`Nifedipine = 25 mM (IC50 = 0.11 mM)
`Nifedipine = 250 mM (IC50 = 0.30 mM)
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0.01
`
`0.02
`
`0.2
`0.1
`0.05
`Ketoconazole (mM)
`
`0.5
`
`1
`
`(% control without inhibitor)
`
`Oxidized nifedipine formation rate
`
`Midazolam = 10 mM (IC50 = 0.092 mM)
`Midazolam = 100 mM (IC50 = 0.185 mM)
`
`0.01 0.02
`
`0.2
`0.1
`0.05
`Ketoconazole (mM)
`
`0.5
`
`1
`
`100
`
`80
`
`60
`
`40
`
`20
`
`(% control without inhibitor)
`a-OH-triazolam formation rate
`
`100
`
`80
`
`60
`
`40
`
`20
`
`(% control without inhibitor)
`
`a-OH-midazolam formation rate
`
`Figure 4 Inhibition curves for ketoconazole vs four different substrates at low and high concentrations. Points are mean (⫾ SE, n = 4) percentage
`ratios of reaction velocity with coaddition of ketoconazole divided by the control velocity without ketoconazole. Lines are the functions consistent with
`equation 2 determined by nonlinear regression. TST, testosterone.
`
`Values of a fell between 2.7 and 3.7 (mean 3.07), indicating
`the predominance of noncompetitive inhibition in the mixed
`model (Table 2).
`For all four CYP3A substrates, ketoconazole IC50 values
`increased at higher concentrations of substrate (Figure 4).
`Analysis of the relation between IC50 and [S] based on equa-
`tion 4 yielded estimates of Ki and a that were similar to those
`determined from the analysis using equation 1 (Table 2). For
`4-OH-midazolam formation, equation 4 again yielded very
`large estimates of a, indicating competitive inhibition. Keto-
`conazole inhibition of nifedipine oxidation was explained via
`equation 4 as a mixed competitive–noncompetitive process,
`with Ki = 0.072 mm and a = 4.25.
`Inhibition of testosterone 6b-hydroxylation by troleando-
`mycin was clearly enhanced by pre-incubation (Figure 5),
`consistent with mechanism-based inhibition.[26–30] However,
`inhibition
`by
`ketoconazole was
`not
`enhanced
`by
`pre-incubation (Figure 5),
`confirming reversible
`rather
`than mechanism-based
`inactivation
`of CYP3A by
`ketoconazole.[30–33]
`
`Discussion
`
`While ketoconazole is generally recognized as a strong inhibi-
`tor of human CYP3A isoforms, numerous studies published
`over the last two decades have indicated extensive variability
`in quantitative Ki values for ketoconazole vs different CYP3A
`substrates, as well as differences among studies of the same
`substrate.[1,2,4] Ketoconazole Ki may also differ even for two
`parallel metabolic pathways for the same substrate such as
`alprazolam, triazolam, midazolam and terfenadine.[10–12,34,35]
`Quantitative determination of Ki values from in-vitro
`studies is dependent on the assumed or established underlying
`mechanism of inhibition. Any given set of data points will
`yield a different estimate of Ki if the mechanism of inhibition
`is assumed to be competitive or noncompetitive. Therefore it
`is probable that some component of the variability among
`studies in Ki values is explained by differing assignments of
`inhibitory mechanism.
`reporting in-vitro Ki
`Among 51 published studies
`values for ketoconazole inhibition of CYP3A substrate
`
`5
`
`

`

`Ketoconazole inhibition of CYP3A
`
`David J. Greenblatt et al.
`
`219
`
`Without pre-incubation
`With pre-incubation
`
`(b)
`
`40
`
`35
`
`30
`
`25
`
`20
`
`15
`
`10
`
`5
`
`(% control without inhibitor)
`
`6b-OH-testosterone fromation rate
`
`Without pre-incubation
`
`With pre-incubation
` (IC50 = 4.6 mM)
`
`1
`
`2
`
`10
`5
`Troleandomycin (mM)
`
`20
`
`50
`
`Keto = 0.05 mM
`
`Keto = 0.25 mM
`
`(a)
`120
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`(% control without inhibitor)
`
`6b-OH-testosterone fromation rate
`
`Figure 5 (a) Effect of troleandomycin on the rate of 6b-OH-testosterone formation from testosterone. (b) Mean percentage ratios of reaction
`velocities with coaddition of ketoconazole divided by the control velocity without ketoconazole. For (a): points are mean (⫾ SE, n = 4) percentage
`ratios of reaction velocities with coaddition of troleandomycin divided by the control velocity without troleandomycin. Studies were done without and
`with pre-incubation of troleandomycin with microsomes before addition of the substrate. Dashed line is the function consistent with equation 2
`determined by nonlinear regression. For (b): mean ⫾ SE, n = 4. Ketoconazole was used at 0.05 or 0.25 mm. Studies were done without and with
`pre-incubation of microsomes with ketoconazole.
`
`biotransformation, the majority assigned a fully competitive
`mechanism.[4] In some of these cases, the competitive process
`was assumed or ‘forced’ a priori, without an evaluation of
`whether that mechanism was most appropriate for the data. In
`other studies, the choice of competitive, noncompetitive or
`mixed inhibition was based on the appearance of data when
`plotted after some form of linearizing transformation (typi-
`cally a Dixon plot of the reciprocal of reaction velocity vs
`inhibitor concentration at different substrate concentrations).
`These transformations may provide useful
`information
`(including estimates of Ki), but may be misleading or distor-
`tive since they involve calculations of reciprocals. Data points
`having the smallest numerical values – and therefore subject
`to greater measurement inaccuracy – become inappropriately
`magnified in importance. Small experimental errors in such
`points are thereby expanded, and may bias slopes and inter-
`cepts of reciprocal plots.
`This study evaluated the kinetics of ketoconazole inhibi-
`tion of human CYP3A in vitro, using four different substrates
`to assess the inhibitory mechanism. The pre-incubation study
`with testosterone as substrate verified that inhibition by keto-
`conazole was reversible as opposed to mechanism-based,
`as
`reported previously from our own laboratory and
`elsewhere.[30–33] The finding of increased IC50 values at higher
`concentrations of all four substrates (Figure 4) effectively
`excluded purely noncompetitive inhibition as the underlying
`mechanism, since the IC50 value would be unchanged (and
`equal to Ki) regardless of substrate concentration (eqn 6). For
`triazolam, midazolam, and testosterone, nonlinear regression
`of all data points allowed simultaneous determination of Ki
`and of the ‘mix’ of competitive and noncompetitive processes,
`with no a priori assumptions about the mechanism, and
`without the potential biases of reciprocal transformations. The
`outcome of the analysis demonstrated mixed-mechanism
`inhibition for
`triazolam,
`testosterone
`and midazolam
`a-hydroxylation. The mean values of a were in the range of
`
`2–7, and the Ki values for ketoconazole were in the low
`nanomolar range. It is of interest that the Ki values vs the
`4-OH-triazolam pathway were higher than for the a-OH-
`triazolam pathway. This confirmed our previous observations
`and emphasized that Ki values for a given inhibitor of CYP3A
`may differ even between two parallel metabolic pathways for
`the same substrate.[10,11] Finally, we observed that ketocona-
`zole inhibition of midazolam 4-hydroxylation, as opposed to
`a-hydroxylation, was consistent with a competitive process.
`These phenomena might be explained by cooperativity and
`multisite kinetic behaviour of human CYP3A.[5,20,23,25,36]
`The relation between the IC50 value and substrate concen-
`tration provided an alternative approach to determination of
`the Ki and a-values. Since the IC50 value increased at
`higher substrate concentrations, pure noncompetitive inhibi-
`triazolam a-hydroxylation and
`tion was excluded. For
`testosterone 6b-hydroxylation and mida-
`4-hydroxylation,
`zolam a-hydroxylation, application of equation 4 indicated
`mixed-mechanism inhibition, with a predominance of the
`noncompetitive component. The numerical values of Ki and a
`were similar to those determined from equation 1. Application
`of equation 4 to midazolam 4-hydroxylation indicated com-
`petitive inhibition, as was found using equation 1. Therefore,
`two independent approaches to data analysis yielded similar
`or identical conclusions regarding the mechanism of inhibi-
`tion by ketoconazole, as well as the numerical values of Ki.
`The four CYP3A substrates used in this study were based
`on the hypothetical categories originally proposed by Ken-
`worthy et al.[6] The conclusion from that study was that probe
`substrates could be divided into two major groups, one of
`which included triazolam and midazolam, and another which
`included testosterone. Nifedipine was described as not falling
`into either of the discrete groups. Those authors made the
`point that there was a high degree of consistency among
`CYP3A substrates in terms of extensive inhibition caused by
`established strong CYP3A inhibitors, and minimal inhibition
`
`6
`
`

`

`220
`
`Journal of Pharmacy and Pharmacology 2011; 63: 214–221
`
`caused by weak inhibitors. We have reported a very high
`degree of correlation of IC50 values for 22 different CYP3A
`inhibitors using midazolam and testosterone as CYP3A sub-
`strates (see Figure 2.2 of Volak et al.[15]) based on data pub-
`lished by Obach et al.[37] A similar point has been made in
`other publications.[20,38,39] In this study, ketoconazole was dem-
`onstrated to be a strong inhibitor of CYP3A – with Ki or IC50
`values in the low nanomolar range – regardless of the hypo-
`thetical category of the substrate. Although current Food and
`Drug Administration guidelines recommend the use of more
`than one CYP3A substrate for in-vitro studies of CYP3A
`inhibition, our findings suggested that a single CYP3A index
`substrate may be sufficient for in-vitro characterization of a
`potential CYP3A inhibitor.[40] There appeared to be low prob-
`ability of acquiring unique information from study of one or
`more additional CYP3A substrates.
`
`Conclusions
`Although ketoconazole is a strong CYP3A inhibitor regard-
`less of the specific substrate, there are nonetheless variations
`among substrates in the quantitative potency of ketoconazole
`inhibition even under standardized in-vitro study conditions.
`The mechanism of inhibition, though reversible, in general
`cannot be characterized as fully competitive or fully noncom-
`petitive. This study has suggested that a mixed competitive–
`noncompetitive mechanism was likely to be applicable. The
`noncompetitive component appeared dominant
`in most
`instances, but the quantitative ‘mix’ of competitive and non-
`competitive processes will vary among substrates and even
`between parallel biotransformation pathways for the same
`substrate.
`
`Declarations
`Conflict of interest
`The Author(s) declare(s) that they have no conflicts of interest
`to disclose.
`
`Funding
`This research received no specific grant from any funding
`agency in the public, commercial, or not-for-profit sectors.
`
`References
`
`1. Venkatakrishnan K et al. Effects of the antifungal agents on
`oxidative drug metabolism in humans: clinical relevance. Clin
`Pharmacokinet 2000; 38: 111–180.
`2. Greenblatt DJ et al. The CYP3 family. In: Ioannides C, ed.
`Cytochrome P450: Role in the Metabolism and Toxicology of
`Drugs and Other Xenobiotics. Cambridge, UK: Royal Society of
`Chemistry, 2008; 354–383.
`3. Greenblatt DJ, von Moltke LL. Clinical studies of drug-drug
`interactions: design and interpretation. In: Pang KS et al., eds.
`Enzyme and Transporter-Based Drug-Drug Interactions:
`Progress and Future Challenges. New York: Springer, 2010;
`625–649.
`4. Greenblatt DJ et al. Sources of variability in ketoconazole inhi-
`bition of human cytochrome P450 3A in vitro. Xenobiotica 2010;
`40: 713–720.
`
`5. Houston JB, Galetin A. Modelling atypical CYP3A4 kinetics:
`principles and pragmatism. Arch Biochem Biophys 2005; 433:
`351–360.
`6. Kenworthy KE et al. CYP3A4 drug interactions: correlation of
`10 in vitro probe substrates. Br J Clin Pharmacol 1999; 48:
`716–727.
`7. von Moltke LL et al. Alprazolam metabolism in vitro: studies of
`human, monkey, mouse, and rat liver microsomes. Pharmacol-
`ogy 1993; 47: 268–276.
`8. Patki KC et al. In vitro metabolism of midazolam, triazolam,
`nifedipine, and testosterone by human liver microsomes and
`recombinant cytochromes P450: role of CYP3A4 and CYP3A5.
`Drug Metab Dispos 2003; 31: 938–944.
`9. Galetin A et al. Multisite kinetic analysis of interactions between
`prototypical CYP3A4 subgroup substrates: midazolam,
`test-
`osterone, and nifedipine. Drug Metab Dispos 2003; 31: 1108–
`1116.
`10. von Moltke LL et al. Triazolam biotransformation by human
`liver microsomes in vitro: effects of metabolic inhibitors, and
`clinical confirmation of a predicted interaction with ketocona-
`zole. J Pharmacol Exp Ther 1996; 276: 370–379.
`11. Perloff MD et al. Midazolam and triazolam biotransformation in
`mouse and human liver microsomes: relative contribution of
`CYP3A and CYP2C isoforms. J Pharmacol Exp Ther 2000; 292:
`618–628.
`12. von Moltke LL et al. Midazolam hydroxylation by human liver
`microsomes in vitro: inhibition by fluoxetine, norfluoxetine, and
`by azole antifungal agents. J Clin Pharmacol 1996; 36: 783–791.
`13. Segel IH. Enzyme Kinetics. New York: John Wiley and Sons,
`1975.
`14. Venkatakrishnan K et al. Human drug metabolism and the cyto-
`chromes P450: application and relevance of in vitro models.
`J Clin Pharmacol 2001; 41: 1149–1179.
`15. Volak LP et al. In vitro approaches to anticipating clinical drug
`interactions. In: Li AP, ed. Drug-Drug Interactions in Pharma-
`ceutical Development. Hoboken, NJ: John Wiley & Sons, 2008;
`75–93.
`16. Brown HS et al. Use of isolated hepatocyte preparations for
`cytochrome
`P450
`inhibition
`studies:
`comparison with
`microsomes for Ki determination. Drug Metab Dispos 2007; 35:
`2119–2126.
`17. Schrag ML, Wienkers LC. Triazolam substrate inhibition: evi-
`dence of competition for heme-bound reactive oxygen within the
`CYP3A4 active site. Drug Metab Dispos 2001; 29: 70–75.
`18. Ghosal A et al. Inhibition and kinetics of cytochrome P4503A
`activity in microsomes from rat, human, and cDNA-expressed
`human cytochrome P450. Drug Metab Dispos 1996; 24: 940–
`947.
`19. Gorski JC et al. Regioselective biotransformation of midazolam
`by members of the human Cytochrome P450 3A (CYP3A) sub-
`family. Biochem Pharmacol 1994; 47: 1643–1653.
`20. Galetin A et al. CYP3A4 substrate selection and substitution in
`the prediction of potential drug-drug interactions. J Pharmacol
`Exp Ther 2005; 314: 180–190.
`21. Hosea NA et al. Elucidation of distinct ligand binding sites for
`cytochrome P450 3A4. Biochemistry 2000; 39: 5929–5939.
`22. Lin Y et al. Substrate inhibition kinetics for cytochrome P450-
`catalyzed reactions. Drug Metab Dispos 2001; 29: 368–374.
`23. Kenworthy KE et al. Multisite kinetic models for CYP3A4:
`simultaneous activation and inhibition of diazepam and testoster-
`one metabolism. Drug Metab Dispos 2001; 29: 1644–1651.
`24. Harlow GR, Halpert JR. Analysis of human cytochrome P450
`3A4 cooperativity: construction and characterization of a site-
`directed mutant that displays hyperbolic steroid hydroxylation
`kinetics. Proc Natl Acad Sci USA 1998; 95: 6636–6641.
`
`7
`
`

`

`Ketoconazole inhibition of CYP3A
`
`David J. Greenblatt et al.
`
`221
`
`25. Ueng Y-F et al. Cooperativity in oxidations catalyzed by cyto-
`chrome P450 3A4. Biochemistry 1997; 36: 370–381.
`26. Obach RS et al. Mechanism-based inactivation of human cyto-
`chrome P450 enzymes and the prediction of drug-drug interac-
`tions. Drug Metab Dispos 2007; 35: 246–255.
`27. Obach RS et al. Inactivation of human Cytochrome P450
`enzymes and drug-drug interactions. In: Pang KS et al., eds.
`Enzyme- and Transporter-Based Drug-Drug Interactions:
`Progress and Future Challenges. New York: Springer, 2010;
`473–492.
`28. Bertelsen KM et al. Apparent mechanism-based inhibition
`of human CYP2D6 in vitro by paroxetine: comparison with
`fluoxetine and quinidine. Drug Metab Dispos 2003; 31: 289–
`293.
`29. Fontana E et al. Cytochrome P450 enzymes mechanism based
`inhibitors: common sub-structures and reactivity. Curr Drug
`Metab 2005; 6: 413–454.
`30. von Moltke LL et al. Potent mechanism-based inhibition of
`human CYP3A in vitro by amprenavir and ritonavir: comparison
`with ketoconazole. Eur J Clin Pharmacol 2000; 56: 259–261.
`31. Weemhoff JL et al. Apparent mechanism-based inhibition of
`human CYP3A in-vitro by lopinavir. J Pharm Pharmacol 2003;
`55: 381–386.
`32. Perloff ES et al. Atazanavir: effects on P-glycoprotein transport
`and CYP3A metabolism in vitro. Drug Metab Dispos 2005; 33:
`764–770.
`
`33. Sekiguchi N et al. Prediction of drug-drug interactions based on
`time-dependent inhibition from high throughput screening of
`cytochrome P450 3A4 inhibition. Drug Metab Pharmacokinet
`2009; 24: 500–510.
`34. von Moltke LL et al. Inhibitors of alprazolam metabolism in vitro:
`effect of serotonin-reuptake-inhibitor antidepressants, ketocona-
`zole and quinidine. Br J Clin Pharmacol 1994; 38: 23–31.
`35. von Moltke LL et al. In vitro prediction of the terfenadine-
`ketoconazole pharmacokinetic interaction. J Clin Pharmacol
`1994; 34: 1222–1227.
`36. Niwa T et al. Heterotropic cooperativity in oxidation mediated
`by Cytochrome P450. Curr Drug Metab 2008; 9: 453–462.
`37.

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


Or .

Accessing this document will incur an additional charge of $.

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

Accept $ Charge
throbber

Still Working On It

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

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

throbber

A few More Minutes ... Still Working

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

Thank you for your continued patience.

This document could not be displayed.

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

Your account does not support viewing this document.

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

Your account does not support viewing this document.

Set your membership status to view this document.

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

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

Become a Member

One Moment Please

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

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

Your document is on its way!

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

Sealed Document

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

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


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket