throbber
Differentiation of intestinal and hepatic
`cytochrome P450 3A activity with use of
`midazolam as an in vivo probe: Effect of
`ketoconazole
`
`Background: The cytochrome P450 3A (CYP3A) isoforms are responsible for the metabolism of a major-
`ity of therapeutic compounds, and they are abundant in the intestine and liver. CYP3A activity is highly
`variable, causing difficulty in the therapeutic use of CYP3A substrates. A practical in vivo probe method
`that characterizes both intestinal and hepatic CYP3A activity would be useful.
`Objectives: To determine the intestinal and hepatic contribution to the bioavailability of midazolam with
`use of the CYP3A inhibitor ketoconazole.
`Methods: The pharmacokinetics of midazolam was assessed in nine (six men and three women) healthy indi-
`viduals after single doses of 2 mg intravenous and 6 mg oral midazolam (phase I). These pharmacokinetic
`values were compared with those obtained after single doses of 2 mg intravenous and 6 mg oral midazo-
`lam and three doses of 200 mg oral ketoconazole (phase II).
`Results: After ketoconazole therapy, area under the concentration versus time curve of midazolam increased
`5-fold after intravenous midazolam administration (P ≤ .001) and 16-fold after oral midazolam adminis-
`tration (P ≤ .001). Intrinsic clearance decreased by 84% (P = .003). Total bioavailability increased from
`25% to 80% (P < .001). The intestinal component of midazolam bioavailability increased to a greater extent
`than the hepatic component (2.3-fold [P = .003] and 1.5-fold [P ≤ .001], respectively). In the control
`phase, female subjects had greater midazolam clearance values than the male subjects.
`Conclusions: Ketoconazole caused marked inhibition of CYP3A activity that was greater in the intestine
`than the liver. Administration of single doses of oral and intravenous midazolam with and without oral
`ketoconazole exemplifies a practical method for differentiating intestinal and hepatic CYP3A activity. (Clin
`Pharmacol Ther 1999;66:461-71.)
`
`Shirley M. Tsunoda, PharmD, Rebecca L. Velez, PharmD, Lisa L. von Moltke, MD,
`and David J. Greenblatt, MD Boston, Mass
`
`The cytochrome P450 3A (CYP3A) subfamily is the
`most important of the cytochrome P450 superfamily
`because of its abundance in the liver and intestine and
`
`From the School of Pharmacy, Bouvé College of Health Sciences,
`Northeastern University, and the Department of Pharmacology and
`Experimental Therapeutics, Tufts University School of Medicine.
`Supported by the Burroughs Wellcome Fund and the American Foun-
`dation for Pharmaceutical Education and by grants M01 RR00054,
`MH-01237, and MH-34223 from the National Institutes of Health
`(Bethesda, Md).
`Received for publication April 23, 1999; accepted Aug 17, 1999.
`Reprint requests: Shirley M. Tsunoda, PharmD, School of
`Pharmacy, Bouvé College of Health Sciences, 206 Mugar Life
`Sciences Building, Northeastern University, Boston, MA 02115.
`E-mail: s.tsunoda@nunet.neu.edu
`Copyright © 1999 by Mosby, Inc.
`0009-9236/99/$8.00 + 0 13/1/102335
`
`its ability to metabolize a wide variety of therapeutic
`compounds. The activity of CYP3A is highly variable
`both between and within individuals, which makes the
`dosing and therapeutic use of many CYP3A substrates
`difficult, especially those with narrow therapeutic
`ranges. The CYP3A subfamily consists of three known
`isoforms
`in humans—CYP3A4, CYP3A5,
`and
`CYP3A7. The CYP3A4 isoform is thought to be the
`dominant form in humans1; however, recent evidence
`reports the presence of CYP3A5 in 10% to 25% of adult
`human livers2 and CYP3A7 in 50% of adult liver sam-
`ples.3 It has not been established whether the presence
`or absence of CYP3A5 and/or CYP3A7 in the intestine
`or liver contributes to a significant difference in meta-
`bolic capacity.4-8 Therefore, to maintain consistency,
`we will use the general term “CYP3A” to refer to the
`
`461
`
`1
`
`TEVA1007
`
`

`

`462 Tsunoda et al
`
`CLINICAL PHARMACOLOGY & THERAPEUTICS
`NOVEMBER 1999
`
`subfamily of enzymes when appropriate. The CYP3A
`activity of an individual is probably determined both
`by genetics and by environmental factors that can mod-
`ulate the activity. Many factors may potentially influ-
`ence the activity of CYP3A, including age,9 gender,10
`menopausal status,11 hormones,12 and numerous xeno-
`biotics.13 However, in general these factors do not have
`a predictable effect on CYP3A activity. In addition, the
`CYP3A activities of an individual in the intestine and
`the liver do not appear to be coordinately regulated14,15;
`therefore it is likely that these many factors may dif-
`ferentially modulate intestinal and hepatic activity.
`Because of the importance of the CYP3A subfamily
`and because of the many factors that influence CYP3A
`activity, a practical method to determine the CYP3A
`activity of an individual is desirable. There have been
`many attempts to use various CYP3A substrates as
`probe compounds to predict CYP3A activity, including
`dapsone, 6-b -hydroxylation of testosterone, nifedipine,
`6-b -hydroxycortisol, and the erythromycin breath test.
`Although the erythromycin breath test has shown cor-
`relations with cyclosporine (INN, ciclosporin) clear-
`ance,16 OG 37-325,17 and midazolam clearance,18 there
`are limitations to this test, including the administration
`of radioactivity, the variability in correlating radioac-
`tive exhaled carbon dioxide and N-demethylation of
`erythromycin, the role of CYP3A in erythromycin
`metabolism,1 and the lack of prediction of intestinal
`CYP3A activity. Therefore an optimal CYP3A probe
`has yet to be identified.
`Midazolam is a benzodiazepine that is used clini-
`cally for conscious sedation. It has pharmacokinetic
`properties that make it an attractive in vivo probe: it is
`specifically metabolized by CYP3A19 to one predomi-
`nant metabolite (1¢ -hydroxymidazolam); it has a short
`half-life (t1⁄2), so estimation of total area under the con-
`centration versus time curve (AUC) is easily measur-
`able and it can be given intravenously and orally (with
`the injection preparation). In addition, hepatic CYP3A
`content measured in vitro has been shown to be highly
`correlated with midazolam intravenous clearance (r =
`0.93; P < .001),20 and plasma 1¢ -hydroxymidazo-
`lam/midazolam ratio.21
`Ketoconazole is one of the most potent inhibitors of
`CYP3A in clinical use. Its estimated in vitro inhibition
`constant (Ki) with use of human liver microsomes for
`1-hydroxymidazolam formation is less than 0.01
`m mol/L (1 m mol/L = 0.53 m g/mL).22 Administration of
`the CYP3A substrate midazolam intravenously (when
`the contribution of intestinal CYP3A activity is
`assumed to be negligible) and orally both in the absence
`and presence of the CYP3A inhibitor ketoconazole
`
`allows the contributions of intestinal and hepatic
`CYP3A to the bioavailability of midazolam to be esti-
`mated. Our study population consisted of healthy indi-
`viduals; therefore potential confounding variables such
`as abnormal liver or intestinal function and concomi-
`tant medications were minimized.
`
`METHODS
`Nine healthy nonsmoking human subjects (six men
`and three women) were enrolled in the study after each
`gave written informed consent. Their mean age was 26
`years (age range, 19 to 41 years). The mean body
`weight of male subjects was 77.5 kg, and the mean
`body weight of female subjects was 59.7 kg. All sub-
`jects were white. The study was approved by the
`Human Investigation Review Committee at New Eng-
`land Medical Center and Tufts University School of
`Medicine and by the Institutional Review Board at
`Northeastern University. The study was conducted in
`the General Clinical Research Center at New England
`Medical Center. All subjects were healthy adults with
`no evidence of medical disease. No subjects took any
`medications. Female subjects were not taking oral con-
`traceptives.
`The study was conducted in two phases. In phase I,
`baseline oral and intravenous midazolam pharmacoki-
`netic parameters were established. Subjects received a
`single dose of 2 mg intravenous midazolam (Versed, 2
`mg/mL for intravenous injection, Hoffmann-LaRoche
`Inc, Nutley, NJ) or 6 mg oral midazolam as 3 mL of the
`parenteral preparation diluted with 30 mL water.
`Although midazolam bioavailability has been estimated
`to average 41%,9 we chose the conservative estimate of
`33% to ensure adequate measurement of plasma con-
`centrations. In phase II, subjects received single doses
`of 2 mg intravenous or 6 mg oral midazolam with 200
`mg ketoconazole (Nizoral, Janssen Pharmaceutica,
`Titusville, NJ). The first dose was administered 12
`hours before the midazolam dose and subsequent doses
`were administered every 12 hours for a total of three
`doses to maintain plasma ketoconazole levels in excess
`of the Ki. The order of treatments was randomized; how-
`ever, treatments remained constant between phase I and
`II for each subject. Before each study day, subjects were
`instructed to fast from midnight until they were given
`a standardized light breakfast approximately 90 min-
`utes before midazolam administration. All subsequent
`meals during each study day were standardized. Alco-
`hol, caffeine, and grapefruit juice were prohibited dur-
`ing each study day. Blood samples were collected
`before and at 15, 30, and 45 minutes and 1, 11⁄2, 2, 21⁄2,
`3, 4, 5, 6, and 8 hours after midazolam dosing during
`
`2
`
`

`

`CLINICAL PHARMACOLOGY & THERAPEUTICS
`VOLUME 66, NUMBER 5
`
`Tsunoda et al 463
`
`Table I. Intravenous and oral midazolam pharmacokinetic parameters: Noncompartmental analysis
`
`Control
`
`With ketoconazole
`
`Intravenous
`
`Oral
`
`Intravenous
`
`Oral
`
`Pharmacokinetic parameters
`AUC (ng · h/mL)
`CL (mL/min/kg)
`CLint (mL/min/kg)
`Varea (L/kg)
`Elimination t1⁄2 (h)
`Cmax (ng/mL)
`tmax (h)
`F (%)
`FH (%)
`FABS · FG (%)
`Ratios (ketoconazole/control)
`AUC
`CLint
`F
`FH
`FABS · FG
`
`70.2 ± 25.8
`7.6 ± 2.9
`13.3 ± 8.9
`1.9 ± 0.8
`3.4 ± 2.0
`—
`—
`—
`—
`—
`
`54.2 ± 28.0
`34.2 ± 18.7
`
`—
`19.1 ± 8.1
`0.8 ± 0.2
`25.3 ± 9.6
`64.6 ± 13.4
`40.1 ± 14.7
`
`354 ± 185*
`1.6 ± 0.8*
`1.8 ± 1.0†
`1.6 ± 0.5
`14.0 ± 8.2†
`—
`—
`—
`—
`—
`
`5.1 ± 1.9
`0.16 ± 0.06
`
`738 ± 191*
`2.1 ± 0.6*
`
`—
`81.0 ± 28.9*
`0.8 ± 0.6
`80.0 ± 31.6*
`92.4 ± 3.7*
`87.8 ± 38.5†
`
`16.0 ± 6.1
`—
`3.4 ± 1.7
`1.5 ± 0.3
`2.3 ± 0.8
`
`Data are mean values ± SD.
`AUC, Area under the concentration versus time curve; CL, clearance; CLint, intrinsic clearance; Varea, volume of distribution; t1⁄2
`tration; tmax, time to reach Cmax; F, total bioavailability; FH, hepatic bioavailability; FABS · FG, intestinal bioavailability.
`*P ≤ .001.
`†P = .003.
`
`, half-life; Cmax, maximum concen-
`
`phase I, with additional samples taken at 12 and 24
`hours after midazolam dosing for phase II.
`Plasma was analyzed for midazolam concentrations
`by electron capture gas chromatography according to a
`previously published method.23 In addition, plasma keto-
`conazole concentrations were determined by HPLC.24
`Pharmacokinetic analysis was conducted by use of
`two methods. The first method used noncompartmen-
`tal methods in which midazolam plasma AUC values
`were calculated by use of the log-linear and linear
`trapezoidal methods for decreasing and increasing
`concentrations, respectively, with extrapolation from
`the last measured concentration to infinity. Estimation
`of the terminal elimination rate constant was per-
`formed by linear regression of at least the last three
`concentration–time points. Oral and intravenous clear-
`ance rates (CLoral and CLIV, respectively) were calcu-
`lated by dividing the respective dose by the AUC after
`oral and intravenous administration, respectively.
`Intrinsic clearance (CLint), defined as the clearance
`attributed solely to hepatic metabolism, was calculated
`with the equation:
`
`(CLIV · QH)/(QH – CLIV)
`in which QH is the liver blood flow. Liver blood flow
`was assumed to be 1500 mL/min/70 kg, normalized for
`body weight of each individual subject. Volume of dis-
`
`tribution (Varea) was calculated with use of the area
`method. The second method used nonlinear regression
`with a linear sum of two exponential terms to analyze
`plasma midazolam concentrations after intravenous
`administration. Coefficients and exponents from the fit-
`ted function were used to estimate total AUC, Varea,
`elimination t1⁄2, and total clearance. After oral adminis-
`tration, AUC until the last detectable concentration was
`calculated by use of the linear trapezoidal method and
`extrapolated to infinity by addition of the final concen-
`tration divided by b
`. The maximum concentration
`(Cmax) and the time to reach Cmax (tmax) were deter-
`mined by the highest measured concentration and
`respective time.
`Bioavailability (F) was determined from dose-
`corrected AUC values, assuming dose-independent
`pharmacokinetics. The components of bioavailability
`were further predicted on the basis of the equation:
`
`F = FABS · FG · FH
`in which FABS is the fraction of the dose absorbed from
`the gut lumen, FG is the fraction of the dose not metab-
`olized by intestinal metabolic enzymes, and FH is the
`fraction of the dose absorbed into the hepatic portal
`vein that escapes first-pass liver metabolism. FH was
`defined as follows:
`
`1 – ERH
`
`3
`
`

`

`464 Tsunoda et al
`
`CLINICAL PHARMACOLOGY & THERAPEUTICS
`NOVEMBER 1999
`
`Fig 1. Mean ± SEM plasma concentration versus time curves after intravenous and oral midazo-
`lam in the control phase versus with ketoconazole.
`
`Table II. Intravenous and oral midazolam pharmacokinetic parameters: Nonlinear regression with a linear sum of
`two exponential terms
`
`Control
`
`With ketoconazole
`
`Intravenous
`
`Oral
`
`Intravenous
`
`Oral
`
`Pharmacokinetic parameters
`AUC (ng · h/mL)
`CL (mL/min/kg)
`CLint (mL/min/kg)
`Varea (L/kg)
`Elimination t1⁄2 (h)
`Cmax (ng/mL)
`tmax (h)
`F (%)
`FH (%)
`FABS · FG (%)
`Ratios (ketoconazole/control)
`AUC
`CLint
`F
`FH
`FABS · FG
`
`Data are mean values ± SD.
`*P ≤ .001.
`
`63.4 ± 18.7
`8.1 ± 2.8
`14.6 ± 9.4
`1.3 ± 0.4
`1.9 ± 0.5
`—
`—
`—
`—
`—
`
`58.2 ± 31.1
`32.7 ± 19
`
`1.7 ± 0.4
`19.1 ± 8.1
`0.8 ± 0.2
`29.3 ± 11.3
`62.4 ± 13.0
`47.5 ± 16.6
`
`300 ± 104*
`1.7 ± 0.5*
`1.9 ± 0.6*
`1.3 ± 0.4
`9.7 ± 4.1*
`—
`—
`—
`—
`—
`
`4.8 ± 1.4
`0.15 ± 0.06
`
`719 ± 181*
`2.1 ± 0.6*
`
`9.9 ± 3.3*
`81.0 ± 28.9*
`0.8 ± 0.5
`83.8 ± 21.0*
`92.0 ± 2.4*
`91.4 ± 24.4*
`
`15.1 ± 6.7
`—
`3.2 ± 1.3
`1.5 ± 0.4
`2.1 ± 0.7
`
`in which ERH is the hepatic extraction ratio defined
`as clearance of intravenously administered midazo-
`lam divided by liver blood flow: CLIV/QH. Non-
`hepatic contributions to clearance were assumed to
`
`be negligible. Therefore, assuming linear pharmaco-
`kinetics, the product FABS · FG was determined and
`compared with and without the presence of the
`enzyme inhibitor ketoconazole. It was assumed that
`
`4
`
`

`

`CLINICAL PHARMACOLOGY & THERAPEUTICS
`VOLUME 66, NUMBER 5
`
`Tsunoda et al 465
`
`ketoconazole does not significantly affect liver blood
`flow or FABS.
`Statistical analysis was conducted by use of the
`paired Student t test, with a = .05.
`
`RESULTS
`Midazolam pharmacokinetic parameters before (con-
`trol) and with ketoconazole calculated by use of
`noncompartmental analysis are shown in Table I; param-
`eters calculated by use of nonlinear regression with a
`linear sum of two exponential terms are shown in Table
`II. In the presence of ketoconazole, intravenous mid-
`azolam AUC increased 5-fold and oral midazolam AUC
`increased 16-fold (P < .001). The comparatively larger
`increase in oral midazolam AUC versus intravenous
`midazolam AUC is shown graphically in Fig 1. Accord-
`ingly, midazolam clearance after intravenous and oral
`administration decreased from 7.6 ± 2.9 to 1.6 ± 0.8
`(P < .001) and 34.2 ± 18.7 to 2.1 ± 0.6 mL/min/kg
`(P < .001) for control versus administration with keto-
`conazole, respectively. Consistent with the presumed
`effect of ketoconazole on CYP3A metabolism, the
`CLint decreased dramatically after ketoconazole admin-
`istration: 13.3 ± 8.9 versus 1.8 ± 1.0 mL/min/kg (P =
`.003; Tables I and II). The comparative declines in all
`three clearance values are shown in Fig 2.
`Ketoconazole caused total measured bioavailability
`to increase from 25% to 80% (P < .001). Ketoconazole
`also caused both the hepatic and intestinal components
`of total bioavailability to be increased, although the
`intestinal component was increased to a greater extent
`(Tables I and II). There was a lack of correlation
`between FH and FABS · FG (r2 = 0.13; Fig 3, A).
`The Cmax of oral midazolam increased 5-fold with
`ketoconazole, consistent with the large gut effect. The
`tmax was not changed with ketoconazole (0.78 ± 0.21
`versus 0.82 ± 0.57 hour, control versus with ketocona-
`zole, respectively; P = NS). The t1⁄2 increased approxi-
`mately 5-fold in the presence of ketoconazole (P =
`.003; Fig 1).
`Interestingly, in the control phase, the three female
`subjects had a mean midazolam clearance that was
`greater than the six male subjects when corrected for
`weight (10.2 ± 3.2 versus 6.3 ± 1.8 mL/min/kg; P =
`.047). However, this difference disappeared during the
`ketoconazole phase (1.8 ± 1.3 versus 1.5 ± 0.5
`mL/min/kg; P = .64). The FH was significantly lower
`in the women compared with the men in the control
`phase only (52.6% ± 14.8% versus 70.7% ± 8.4%; P =
`.047), which was consistent with the change in clear-
`ance. Women also had a larger weight-adjusted Varea
`than men that reached statistical significance during the
`
`Fig 2. Mean ± SEM plasma clearance rates in the control
`phase versus with ketoconazole. Intravenous and oral clear-
`ance values were measured. Intrinsic clearance was calcu-
`lated as described in the text.
`
`ketoconazole phase (2.17 ± 0.25 versus 1.32 ± 0.34 L/kg;
`P = .007), when calculated with noncompartmental
`methods. It should be noted that there were no changes
`in weight between the control and ketoconazole phases.
`However, using the two-compartment method, females
`had larger weight-adjusted Varea compared to males in
`both the control (1.65 ± 0.49 versus 1.08 ± 0.20 L/kg,
`P = .03) and ketoconazole (1.70 ± 0.68 versus 1.10 ±
`0.23 L/kg, P = .025) phases. In addition, there were no
`gender-related differences in clearance or the elimina-
`tion rate constant during the ketoconazole phase. There
`were no other gender-related differences in any other
`pharmacokinetic parameter.
`Ketoconazole plasma concentrations ranged from
`0.077 to 4.852 m g/mL, thereby exceeding the in vitro
`Ki at most time points (Fig 4).
`
`DISCUSSION
`The CYP3A subfamily is clearly important in deter-
`mining the pharmacokinetics and subsequent pharmaco-
`
`5
`
`

`

`466 Tsunoda et al
`
`CLINICAL PHARMACOLOGY & THERAPEUTICS
`NOVEMBER 1999
`
`A
`
`C
`
`B
`
`D
`
`Fig 3. A, Lack of correlation between midazolam hepatic bioavailability (FH) and intestinal bioavail-
`ability (FABS · FG ). B, Lack of correlation between total bioavailability (F) in the control phase
`(without ketoconazole) and the relative change with ketoconazole. C, Correlation between FH in
`the control phase and the relative change in FH with ketoconazole. D, Lack of correlation between
`FABS · FG in the control phase and the relative change with ketoconazole.
`
`dynamics of many compounds that encompass a variety
`of therapeutic areas. In addition, the intestinal contri-
`bution of CYP3A is also important in mechanisms
`of drug-drug interactions and the determination of
`the bioavailability of a number of CYP3A sub-
`strates.20,21,25-30 It appears that the intestinal and
`hepatic CYP3A activities of an individual are regulated
`
`independently14,15; therefore an ideal in vivo probe
`must quantitate both. Our study used a CYP3A com-
`pound that can be administered intravenously and orally
`in the absence and presence of a potent CYP3A
`inhibitor to partition intestinal and hepatic activity.
`In the control phase, the midazolam pharmacokinetic
`parameters clearance and total bioavailability were
`
`6
`
`

`

`CLINICAL PHARMACOLOGY & THERAPEUTICS
`VOLUME 66, NUMBER 5
`
`Tsunoda et al 467
`
`Fig 4. Mean ± SEM plasma concentration versus time curve after intravenous and oral midazolam.
`
`comparable to those previously reported. Clearance val-
`ues ranged from 4.7 to 8.1 mL/min/kg and total
`bioavailability values ranged from 27 to 40%9,28,30-32
`in healthy volunteers.
`Midazolam pharmacokinetic parameters changed
`dramatically during the administration of ketoconazole,
`which was expected. The magnitude of change was
`consistent with the results of Olkkola et al33 in which
`7.5 mg oral midazolam was administered with 400 mg
`ketoconazole for four doses in nine healthy volunteers.
`Administering a 20% higher dose of midazolam and a
`2-fold higher dose of ketoconazole gave a mean mid-
`azolam AUC that increased 16-fold compared with our
`mean oral AUC, which also increased 16-fold. The inhi-
`bition of CYP3A by ketoconazole does not appear to
`be dose- or concentration-dependent at this dosing
`range; and it is likely that a dose of 200 mg causes
`nearly complete CYP3A inhibition. Total systemic
`clearance decreased by 78% in the presence of keto-
`conazole. This magnitude is slightly greater than that
`in the study by Gorski et al,30 who found a 64%
`decrease in midazolam clearance in the presence of
`clarithromycin. The inhibitory effect of ketoconazole
`on CYP3A metabolism is more accurately illustrated
`by the 84% decrease in CLint. Because midazolam is
`an intermediate extraction ratio compound (mean
`extraction ratio, 35%), CLint more accurately reflects
`the contribution of clearance attributable to drug metab-
`olism, without the influence of hepatic blood flow. In
`
`the presence of ketoconazole, CLoral, CLIV, and CLint
`fall to a similar level (approximately 2 mL/min/kg),
`again illustrating the potent inhibitory effect of keto-
`conazole on CYP3A-mediated intestinal and hepatic
`metabolism (Fig 2). Consistent with the intestinal
`effect, ketoconazole caused a 5-fold increase in Cmax
`without any change in the tmax. We also found an almost
`5-fold lengthening of the elimination t1⁄2 with ketocona-
`zole, similar to the 310% increase in elimination t1⁄2
`found by Olkkola et al,33 and the almost 3-fold increase
`found by Gorski et al.30
`Although several midazolam studies have failed to
`show a statistically significant gender-related effect in
`young subjects,9,28,34,35 we found a gender-related
`effect in the control midazolam phase of 60% greater
`weight-corrected clearance in the young women versus
`the young men (P = .05). This is qualitatively consis-
`tent with that shown with other CYP3A substrates such
`as erythromycin, methylprednisolone, tirilazad, and
`verapamil, as recently reviewed.10 In addition, on the
`basis of blood concentrations, Gorski et al30 reported a
`statistically significant 27% higher midazolam clear-
`ance in young healthy women versus men. They also
`found weight-corrected CLoral to be 50% higher in
`young women versus men, with a corresponding
`decrease in the AUC.30 Our data are also consistent
`with that of Gorski et al30 with the CYP3A inhibitor
`clarithromycin in that the gender-related effect disap-
`peared during the inhibition phase. Some investigations
`
`7
`
`

`

`468 Tsunoda et al
`
`CLINICAL PHARMACOLOGY & THERAPEUTICS
`NOVEMBER 1999
`
`imply that intestinal metabolism may be different in
`men versus women because gender-related differences
`have been found in either CLoral or bioavailability with
`oral triazolam,36 oral cyclosporine,14 and oral verap-
`amil.37 On the other hand, other studies with oral
`triazolam38 and other oral CYP3A substrates, such as bu-
`spirone,39 alprazolam,40 and trazodone,41 have not found
`a gender-related difference. The studies that have found
`a gender-related difference have shown that young
`females have greater CYP3A activity compared with
`young males. Our data with midazolam are consistent
`with this; however, confirmation in a larger population
`is required because we enrolled a small number of
`female subjects. The gender issue is complex, may
`involve other intestinal proteins, and may not prove to
`be consistent with all CYP3A substrates. Our data illus-
`trated a larger weight-adjusted Varea in women com-
`pared with men, which is consistent with other reports
`in the literature.9,28,34
`Ketoconazole decreased the variability in hepatic
`bioavailability. It has been suggested that inhibition of
`CYP3A causes a “normalization” of activity across the
`study population; that is, subjects with the greatest
`CYP3A activity exemplified by high clearance rates or
`low bioavailability show the greatest change. This phe-
`nomenon has been shown with grapefruit juice and
`felodipine42 and with clarithromycin and midazolam.30
`We found a correlation between FH in the control phase
`and the relative increase in FH during ketoconazole
`inhibition (r2 = 0.96; Fig 3, C). A lack of correlation
`was found in these respective relationships with total
`bioavailability (r2 = 0.22) and FABS · FG (r2 = 0.14; Figs
`3, B and D).
`Total measured bioavailability increased by a factor
`of 3.4 in the presence of ketoconazole. The intestinal
`availability increased 2.3-fold, whereas the hepatic
`availability increased 1.5-fold in the presence of keto-
`conazole. These results are qualitatively consistent with
`other CYP3A inhibition studies reported in the litera-
`ture in that the intestinal component is affected to a
`greater extent than the hepatic component. Gorski et
`al30 found a 197% increase in midazolam intestinal
`availability and a 120% increase in hepatic availability
`after 7 days of 500 mg clarithromycin twice daily. Sim-
`ilarly, Gomez et al26 demonstrated a 2.3-fold increase
`in intestinal availability and a 1.15-fold increase in
`hepatic availability of cyclosporine after 6 to 10 days
`of 200 mg ketoconazole therapy. Midazolam is readily
`absorbed,30 so these effects are likely the result of an
`intestinal metabolism effect. These effects are proba-
`bly not modulated by P-glycoprotein because midazo-
`lam is not a P-glycoprotein substrate.43 It is likely that
`
`higher local concentrations of the inhibitor, in this case
`ketoconazole, are achieved in the intestine, leading to
`greater CYP3A inhibition compared with that in the
`liver. In addition, the dynamics and variability of gas-
`tric motility probably contribute to the complexity of
`the interaction. However, we did not test this directly.
`Nonetheless, the potentially greater effect on intestinal
`CYP3A inhibition has important clinical implications
`for drug-drug and drug-food interactions with oral com-
`pounds in that separating the timing of administration
`of potentially interacting oral CYP3A substrates may
`lessen or even ameliorate an interaction.44
`Consistent with other reports that have suggested that
`intestinal and hepatic CYP3A activity are regulated
`independently, we found that there was no correlation
`between FH and FABS · FG (r2 = 0.13; Fig 3, A).
`Our conclusions are based on the assumption that
`midazolam exhibits dose-independent pharmacokinet-
`ics, which is a reasonable assumption because it has
`been shown by several studies in young healthy indi-
`viduals over a varied dosing range.9,34,45 We did not
`measure the protein binding of midazolam; however,
`because the extraction ratio of midazolam (estimated
`from our study to be 35% ± 13% [mean ± SD]) is
`much greater than the estimated percent fraction
`unbound, which has been reported to be 2.4% to
`3.7%,9,28 protein binding cannot completely restrict
`hepatic uptake. We also assumed that enzyme activity
`and blood flow remained constant between oral and
`intravenous midazolam administration. Although
`simultaneous administration of oral and intravenous
`substrate may be theoretically preferable, the practi-
`cal issues, such as the need for isotope-labeled drug
`and the high midazolam dose required, precluded this
`method. In addition, because all subjects were healthy
`and had no longer than a 7-day washout period
`between study days, variability in these processes is
`not likely to have influenced the results. A recent
`investigation showed insignificant interday variability
`in hepatic CYP3A4 metabolism in young men by use
`of midazolam intravenous pharmacokinetics.46 We
`used plasma rather than blood concentrations for cal-
`culation of pharmacokinetic parameters, which may
`lead to overestimation of AUC values. In addition,
`rather than measuring hepatic blood flow directly, we
`assumed it to be 1500 mL/min/70 kg. Based on lido-
`caine clearance,47 this is a reasonable assumption;
`however, measured hepatic blood flow would have
`been more accurate. Finally, fluctuations in ketocona-
`zole concentrations may have complicated the inter-
`action with midazolam (Fig 4); however, in general,
`plasma concentrations exceeded the in vitro Ki.
`
`8
`
`

`

`CLINICAL PHARMACOLOGY & THERAPEUTICS
`VOLUME 66, NUMBER 5
`
`Tsunoda et al 469
`
`The results of several studies6,48,49 show that mid-
`azolam is predominantly and virtually exclusively
`metabolized by enzymes of the CYP3A subfamily and
`that measuring midazolam clearance in vivo correlates
`well with content and activity in vitro. In vitro studies
`with hepatic microsomes that contain CYP3A4 were
`found to hydroxylate midazolam to its two primary
`metabolites, 1¢ -hydroxymidazolam and 4¢ -hydroxy-
`midazolam.6,22 Total midazolam clearance has been
`shown to be highly correlated with hepatic CYP3A con-
`tent(r = 0.93; P < .001) and cyclosporine clearance
`(r = 0.81; P < .001)20 but less strongly correlated with
`the erythromycin breath test result (r = 0.52; P = .03).18
`In addition, the apparent maximum rate (Vmax) for 1¢ -
`hydroxymidazolam formation was weakly correlated
`with total hepatic CYP3A4 content (r2 = 0.31; P < .05),48
`suggesting the importance of the intestinal contribution.
`Finally, 43% of an intraduodenally administered dose of
`midazolam was shown to be metabolized through the
`intestinal mucosa during the anhepatic phase of 10 liver
`transplant patients.49 These data6,18,20,48,49 support the
`use of midazolam as an in vivo probe, although a num-
`ber of the studies20,49 used invasive methods. Therefore
`the method used in our study with use of relatively small
`doses of midazolam intravenously and orally in the pres-
`ence and absence of the inhibitor ketoconazole exempli-
`fies a less invasive model for partitioning intestinal and
`hepatic CYP3A activity.
`
`We gratefully acknowledge the assistance of the following indi-
`viduals: Anna-Liza Durol for plasma analysis of midazolam and keto-
`conazole, Jerold S. Harmatz for pharmacokinetic and statistical analy-
`ses, Richard I. Shader, MD, for advice and collaboration, and the
`staff of the General Clinical Research Center, New England Medical
`Center.
`
`References
`1. Shimada T, Yamazaki H, Mimura M, Inui Y, Guengerich
`FP. Interindividual variations in human liver cytochrome
`P-450 enzymes involved in the oxidation of drugs, car-
`cinogens and toxic chemicals: studies with liver micro-
`somes of 30 Japanese and 30 Caucasians. J Pharmacol
`Exp Ther 1994;270:414-23.
`2. Wrighton SA, Ring BJ, Watkins PB, Vandenbranden M.
`Identification of a polymorphically expressed member of
`the human cytochrome P-450III family. Mol Pharmacol
`1989;36:97-105.
`3. Schuetz JD, Beach DL, Guzelian PS. Selective expres-
`sion of cytochrome P450 CYP3A mRNAs in embryonic
`and adult human liver. Pharmacogenetics 1994;4:11-20.
`4. Gibbs MA, Thummel KE, Shen DD, Kunze KL. Inhibi-
`tion of cytochrome P-450 3A (CYP3A) in human intesti-
`nal and liver microsomes: comparison of Ki values and
`impact of CYP3A5 expression. Drug Metab Dispos 1999;
`27:180-7.
`
`5. Gillam EM, Wunsch RM, Ueng YF, Shimada T, Reilly
`PE, Kamataki T, et al. Expression of cytochrome P450
`3A7 in Escherichia coli: effects of 5¢ modification
`and catalytic characterization of recombinant enzyme
`expressed
`in bicistronic
`format with NADPH–
`cytochrome P450 reductase. Arch Biochem Biophys
`1997;346:81-90.
`6. Gorski JC, Hall SD, Jones DR, Vandenbranden M,
`Wrighton SA. Regioselective biotransformation of mid-
`azolam by members of the human cytochrome P450
`3A (CYP3A) subfamily. Biochem Pharmacol 1994;47:
`1643-53.
`7. Paine MF, Khalighi M, Fisher JM, Shen DD, Kunze KL,
`Marsh CL, et al. Characterization of interintestinal and
`intraintestinal variations in human CYP3A-dependent
`metabolism. J Pharmacol Exp Ther 1997;283:1552-62.
`8. Tateishi T, Watanabe M, Moriya H, Yamaguchi S, Sato T,
`Kobayashi S. No ethnic difference between Caucasian
`and Japanese hepatic samples in the expression frequency
`of CYP3A5 and CYP3A7 proteins. Biochem Pharmacol
`1999;57:935-9.
`9. Greenblatt DJ, Abernethy DR, Locniskar A, Harmatz
`JS, Limjuco RA, Shader RI. Effect of age, gender, and
`obesity on midazolam kinetics. Anesthesiology 1984;
`61:27-35.
`10. Harris RZ, Benet LZ, Schwartz JB. Gender effects in
`pharmacokinetics and pharmacodynamics. Drugs 1995;
`50:222-39.
`11. Harris RZ, Tsunoda SM, Mroczkowski P, Wong H, Benet
`LZ. The effects of menopause and hormone replacement
`therapies on prednisolone and erythromycin pharmacoki-
`netics. Clin Pharmacol Ther 1996;59:429-35.
`12. Tsunoda SM, Harris RZ, Mroczkowski PJ, Benet LZ.
`Preliminary evaluation of progestins as inducers of
`cytochrome P450 3A4 activity in postmenopausal
`women. J Clin Pharmacol 1998;38:1137-43.
`13. Thummel KE, Wilkinson GR. In vitro and in vivo dr

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