throbber
BIOPHARMACEUTICS & DRUG DISPOSITION
`Biopharm. Drug Dispos. 27: 421–426 (2006)
`Published online in Wiley InterScience
`(www.interscience.wiley.com) DOI: 10.1002/bdd.524
`
`Everolimus Drug Interactions: Application of a Classification
`System for Clinical Decision Making
`
`John M. Kovarik*, Doris Beyer and Robert L. Schmouder
`Novartis Pharmaceuticals, Basel, Switzerland and East Hanover, NJ, USA
`
`ABSTRACT: Introduction. More than half of all drugs used in medical practice are metabolized by
`cytochrome CYP3A. Coadministration of drugs that share this elimination pathway may lead to
`pharmacokinetic drug interactions. Efforts are underway by clinical, drug development and
`regulatory scientists to classify CYP3A-related drug interactions with the ultimate goal of
`improving guidance for clinical intervention. The CYP3A inhibitory classification system ranks
`inhibitors according to the fold-increase in area-under-the-curve (AUC) of a probe substrate as:
`strong (55-fold), moderate (>2.0- to 4.9-fold), or weak (42.0-fold). This classification system was
`applied to characterize everolimus as a CYP3A substrate.
`Methods. Five open-label crossover drug interaction studies were performed in 12–16 healthy
`subjects each. Subjects received a single 2 mg dose of everolimus alone and again during single- or
`multiple-dose treatment with the probe inhibitors ketoconazole, erythromycin, verapamil,
`cyclosporine and atorvastatin.
`Results. The fold-increase in everolimus AUC was: 15.0 with the strong inhibitor ketoconazole;
`4.4, 3.5 and 2.7 with the moderate inhibitors erythromycin, verapamil and cyclosporine; and no
`change with the weak inhibitor atorvastatin. Subjects with low baseline AUCs when everolimus was
`given alone tended to have AUC increases of a higher magnitude (more potent interaction) in the
`presence of an inhibitor.
`Conclusions. Strong CYP3A inhibitors should be avoided when possible during everolimus
`treatment as compensatory everolimus dose reductions could be difficult to manage. Everolimus
`therapeutic drug monitoring should be used to guide individualized dose adjustments when
`moderate CYP3A inhibitors are added to or withdrawn from the regimen. Routine everolimus
`therapeutic drug monitoring should be sufficient to determine whether dose adjustments are
`needed when weak CYP3A inhibitors are coadministered. This rational and systematic approach to
`drug interactions on everolimus yielded clinically useful, structured guidelines for dose
`adjustment. Copyright # 2006 John Wiley & Sons, Ltd.
`
`Key words: drug interactions; enzyme inhibition; everolimus
`
`Introduction
`
`A large proportion of drugs used in clinical
`medicine}up to 60% by some estimates}
`utilize the cytochrome P450 (CYP) 3A pathway
`for biotransformation [1]. Consequently,
`the
`
`*Correspondence to: Novartis Pharma AG, Building WSJ 103.426,
`4002 Basel, Switzerland. E-mail: john.kovarik@Novartis.com
`
`potential for pharmacokinetic drug interactions
`when coadministering agents that share this
`pathway is clearly of clinical relevance. It is
`understandable that drug development scientists
`and regulatory authorities share the desire to
`harmonize approaches to assess drug interac-
`tions and to develop a CYP3A classification
`system to communicate risk to health care
`providers and patients. While several regulatory
`
`Copyright # 2006 John Wiley & Sons, Ltd.
`
`Received 14 October 2005
`Revised 10 March 2006
`Accepted 28 June 2006
`
`Ex. 1049-0001
`
`

`
`422
`
`J.M. KOVARIK ET AL.
`
`agencies worldwide have issued general drug
`interaction guidances for industry [2–4],
`the
`specifics of study design and methodology have
`not been addressed in a comprehensive manner.
`The drug metabolism and clinical pharmacol-
`ogy technical working groups of the Pharmaceu-
`tical Research and Manufacturers of America
`have issued a consensus statement to begin to
`address some of these issues and to define a drug
`interaction data package that can be expected by
`regulatory authorities in the submission dossier
`of a new drug [5].
`In addition to defining
`currently preferred and alternate probe CYP
`substrates to use in clinical drug interaction
`studies,
`they also cite a CYP3A inhibitory
`classification system to allow ranking the magni-
`tude of
`interactions via this pathway. Using
`midazolam as a prototype CYP3A substrate, this
`system classifies the inhibitory potential of a new
`drug as strong if the plasma area-under-the-
`curve (AUC) of midazolam increases 55-fold,
`moderate if the increase is >2.0- to 4.9-fold, and
`weak if the increase is 42.0-fold [5]. This system
`could also be used to characterize a new drug as
`a substrate of CYP3A, placing it in reference to
`preferred probe inhibitors in the three classes.
`With the exception of midazolam, which was
`used to define the system,
`there are to our
`knowledge no examples in the scientific literature
`applying this classification system in a consistent
`manner during the development of a new drug.
`We present our attempt to do so with everolimus,
`a macrolide immunosuppressant, that acts as a
`proliferation signal
`inhibitor on T-lymphocytes.
`
`Everolimus is used to prevent acute rejection
`episodes after kidney and heart transplantation.
`It is primarily biotransformed via CYP3A with
`metabolites eliminated in the bile and is also a
`substrate of P-glycoprotein [6]. A series of clinical
`drug interaction studies were performed during
`the development of everolimus demonstrating that
`it fits into this classification system as a CYP3A
`substrate. The classification system provided a
`useful structure to rank the potential for interac-
`tions from coadministered drugs on everolimus
`and to recommend everolimus dose adjustments.
`
`Methods
`
`Five open-label crossover drug interaction stu-
`dies were performed in healthy subjects using
`the strong CYP3A inhibitor ketoconazole [7]; the
`moderate inhibitors erythromycin [8] verapamil
`[9] and cyclosporine microemulsion [10]; and the
`weak inhibitor atorvastatin [11]. All study proto-
`cols were approved by medical ethics committees
`and all subjects gave written informed consent to
`participate.
`In these studies the reference treatment was a
`2 mg single dose of everolimus (Certican, Novar-
`tis Pharmaceuticals) administered after an over-
`night fast of at least 10 h. The test treatment
`consisted of single-dose everolimus administered
`simultaneously with the inhibitor either under
`single-dose conditions or after steady state was
`reached during multiple-dose administration of
`the inhibitor as itemized in Table 1. Blood
`
`Table 1. Clinical drug interaction studies of CYP3A inhibitors on everolimus
`
`Inhibitor
`class
`
`Number of
`subjects
`
`Test regimen
`
`Strong
`
`Moderate
`
`Moderate
`
`Moderate
`
`Weak
`
`12
`
`16
`
`16
`
`12
`
`12
`
`Ketoconazole 200 mg b.i.d.  8
`days, Everolimus on day 4
`Erythromycin 500 mg t.i.d.  9
`days, Everolimus on day 5
`Verapamil 80 mg t.i.d.  6 days,
`Everolimus on day 2
`Cyclosporine 175 mg single dose,
`Everolimus on day 1
`Atorvastatin 20 mg single dose,
`Everolimus on day 1
`
`Everolimus
`AUC alone
`(mg.h/l)
`
`Everolimus AUC
`with inhibitor
`(mg.h/l)
`
`Fold-increase in
`everolimus
`AUCa
`
`90  23
`
`1324  232
`
`15.0 (13.6–16.6)
`
`116  37
`
`115  45
`
`74  26
`
`120  37
`
`524  225
`
`392  142
`
`193  47
`
`118  46
`
`4.4 (3.5–5.4)
`
`3.5 (3.1–3.9)
`
`2.7 (2.2–3.2)
`
`1.0 (0.8–1.2)
`
`Ref
`
`[7]
`
`[8]
`
`[9]
`
`[10]
`
`[11]
`
`aFold-increase is point estimate and 90% confidence interval.
`AUC, area under the concentration-time curve; b.i.d., twice daily; t.i.d., thrice daily.
`
`Copyright # 2006 John Wiley & Sons, Ltd.
`
`Biopharm. Drug Dispos. 27: 421–426 (2006)
`DOI: 10.1002/bdd
`
`Ex. 1049-0002
`
`

`
`EVEROLIMUS DRUG INTERACTIONS
`
`423
`
`samples were obtained before each everolimus
`dose and then at 0.5, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 12, 24,
`36, 48, 72, 96 and 120 h postdose. Everolimus
`whole blood concentrations were determined by
`a validated liquid chromatography method with
`mass spectrometric detection as previously de-
`scribed [12]. The assay limit of quantification was
`0.3 ng/ml. The area under the concentration-time
`curve extrapolated to infinity (AUC) was derived
`by noncompartmental methods. The AUCs were
`log-transformed and compared between treat-
`ments by conventional bioequivalence testing to
`yield the test/reference ratio of the geometric
`means and 90% confidence interval. The ratio
`served as the estimate of the magnitude of the
`drug interaction on everolimus expressed as the
`fold-increase in everolimus exposure. The inter-
`subject variability in the magnitude of
`the
`interaction was derived as the standard deviation
`divided by the mean of these AUC-ratios (AUC
`with inhibitor/AUC alone). Relationships be-
`tween the reference everolimus AUC and the
`fold-increase in the test AUC were explored by
`graphical evaluation and fitting a descriptive log
`function through the data.
`
`Results
`
`Interaction magnitude
`
`As summarized in Table 1 the population
`average fold-increase in everolimus AUC in the
`presence of strong, moderate and weak CYP3A
`inhibitors fell within the bounds of the classifica-
`tion system [5] in all cases. A related rapamycin
`macrolide, sirolimus, reported similar fold-in-
`creases in sirolimus AUC of 10.9 in the presence
`of ketoconazole, 4.2 in the presence of erythro-
`mycin, 3.3 in the presence of cyclosporine
`microemulsion, 2.2 in the presence of verapamil,
`and no relevant change in the presence of
`atorvastatin [13]. The fold-increases in everoli-
`mus and sirolimus AUC were similar to that of
`the prototype CYP3A substrate, midazolam, in
`the presence of
`these inhibitors. Published
`studies reported mean fold-increases in midazo-
`lam AUC in the presence of multiple-dose
`ketoconazole of 8.7, 11 and 16 [14–16], in the
`presence of multiple-dose erythromycin of 4.4
`
`Copyright # 2006 John Wiley & Sons, Ltd.
`
`and 3.8 [17–18] and in the presence of multiple-
`dose verapamil of 2.9 [19]. The lack of change in
`the AUC of atorvastatin}a recommended and
`sensitive probe substrate of CYP3A}in the
`presence of everolimus [11] indicates that ever-
`olimus does not inhibit CYP3A to a clinically
`relevant extent.
`
`Interaction variability
`
`Figure 1 depicts the individual fold-increases in
`everolimus AUC. The associated intersubject
`variability was generally moderate to high at
`20% in the presence of ketoconazole, 57% for
`erythromycin, 28% for verapamil, 34% for
`cyclosporine microemulsion and 36% for ator-
`vastatin. While all subjects exhibited a strong
`interaction magnitude (55-fold increase) with
`ketoconazole and a weak interaction magnitude
`(52.0-fold) with atorvastatin,
`some subjects
`receiving moderate inhibitors had fold-increases
`which transgressed the categorical boundaries.
`Specifically, for cyclosporine microemulsion 3 of
`12 subjects (25%) had weak interactions between
`1.5- to 1.9-fold. For erythromycin 6 of 16 subjects
`(38%) had a strong interaction ranging from
`5.1- to 12.6-fold and for verapamil 1 of 16 subjects
`(6%) had a strong interaction of 6.3-fold.
`
`Exposure-response associations
`
`The pharmacokinetic data were explored to look
`for predictive or explanatory factors for the
`
`Magnitude: Wk Mod
`
`Strong
`
`Ketoconazole
`
`Erythromycin
`
`Verapanil
`
`Cyclosporine
`
`Atorvastatin
`
`0
`
`2
`
`4
`
`6
`8 10 12 14 16 18 20 22 24
`Fold-increase in everolimus AUC
`
`Figure 1. Fold-increase in everolimus AUC in the presence of
`various CYP3A inhibitors against a categorical background
`for weak inhibition (Wk, 42.0-fold increase), moderate
`inhibition (Mod, 2.0- to 4.9-fold increase) and strong inhibition
`(Strong, 55-fold increase). Shown are the individual fold-
`increases (open circles), the population average (vertical bar),
`and the 90% confidence interval (horizontal bar)
`
`Biopharm. Drug Dispos. 27: 421–426 (2006)
`DOI: 10.1002/bdd
`
`Ex. 1049-0003
`
`

`
`J.M. KOVARIK ET AL.
`
`by monitoring predose blood levels along with
`the clinical condition of the patient [6]. Given the
`small influence of weak CYP3A inhibitors on
`everolimus pharmacokinetics,
`it is anticipated
`that routine therapeutic drug monitoring would
`indicate whether an individual patient needs an
`everolimus dose adjustment when a weak
`CYP3A inhibitor is added to or removed from
`the regimen. At the other extreme, the large
`influence of strong CYP3A inhibitors on ever-
`olimus exposure would make compensatory
`dose adjustments difficult to achieve given the
`tablet strengths available. Hence, coadministra-
`tion of strong CYP3A inhibitors with everolimus
`is not recommended.
`Addition or removal of moderate CYP3A
`inhibitors to an everolimus regimen should be
`accompanied by therapeutic drug monitoring to
`guide compensatory everolimus dose adjust-
`ments. The study data shown in Figure 1 indicate
`that most everolimus AUC increases were in the
`range of 2- to 4-fold and this can serve as a
`general
`indicator of what
`to expect when
`coadministering a moderate CYP3A inhibitor.
`However, some increases in everolimus exposure
`may extend into the strong interaction range.
`Figure 2 suggests that patients who may be more
`susceptible to a drug interaction from a CYP3A
`inhibitor appear to be those with low baseline
`everolimus exposure. Such patients would likely
`require a higher everolimus dose than the
`population average dose in order to maintain
`everolimus blood levels in the therapeutic range
`in the absence of CYP3A inhibitors. Clinicians
`treating such patients should be more alert for
`drug interactions when adding a moderate
`CYP3A inhibitor to the regimen until everolimus
`therapeutic drug monitoring results are available
`from the analytical
`laboratory to confirm the
`actual magnitude of the interaction.
`
`Discussion
`
`This overview of the everolimus drug interaction
`program during drug development demonstrates
`that
`the influence of
`five different CYP3A
`inhibitors on everolimus fits into the proposed
`CYP3A inhibitory classification system proposed
`
`Biopharm. Drug Dispos. 27: 421–426 (2006)
`DOI: 10.1002/bdd
`
`Ketoconazole
`
`Erythromycin
`
`0
`
`25
`
`Verapamil
`75 100 125 150 175 200 225 250
`50
`Baseline everolimus AUC (µg.h/L)
`
`424
`
`24
`
`21
`
`18
`
`15
`
`12
`
`0369
`
`Fold-increase in everolimus AUC
`
`Figure 2. Relationship between everolimus AUC in the absence
`of coadministered drugs (baseline AUC) and the fold-increase
`in everolimus AUC in the presence of CYP3A inhibitors
`ketoconazole (filled triangles), erythromycin (open squares)
`and verapamil (filled circles). Shown is the best fit of a
`for ketoconazole, y ¼
`logarithmic trendline to the data:
`9 lnðxÞ þ 55 ðr2 ¼ 0:617Þ; for erythromycin, y ¼ 1:6 lnðxÞ þ
`11:3 ðr2 ¼ 0:423Þ; for verapamil,y ¼ 4:5 lnðxÞ þ 26 ðr2 ¼ 0:275Þ
`
`magnitude of the interaction on everolimus or its
`intersubject variability. The blood or plasma level
`of the inhibitor during the test treatment was in
`no case predictive of
`the magnitude of
`the
`interaction on everolimus [7–11]. The baseline
`everolimus AUC, however, was inversely related
`to the fold-increase in everolimus AUC in the
`presence of the inhibitor as shown in Figure 2 for
`ketoconazole, erythromycin, and verapamil. In
`each case a linear regression did not adequately
`account for the curvature in these relationships;
`hence,
`they were described with logarithmic
`trendlines as specified in the figure legend. In
`general, the lower the baseline everolimus AUC,
`the greater the interaction magnitude in the
`presence of the inhibitor. There was no apparent
`relationship for everolimus with cyclosporine for
`which the scatterplot appeared flat. Since ator-
`vastatin essentially did not change the ever-
`olimus AUC, no relationship was observed for
`this weak inhibitor.
`
`Clinical implications
`
`Everolimus is marketed at dose strengths of 0.25,
`0.5, 0.75 and 1 mg tablets and 0.1 and 0.25 mg
`dispersible tablets [20].
`In organ transplant
`medicine, everolimus dosing is individualized
`
`Copyright # 2006 John Wiley & Sons, Ltd.
`
`Ex. 1049-0004
`
`

`
`EVEROLIMUS DRUG INTERACTIONS
`
`425
`
`by the Pharmaceutical Research and Manufac-
`turer’s Association [5]. Such a classification
`system begins to address the desire expressed
`by industry sponsors to develop a ranking
`system to allow better assessment and compar-
`ison of different drugs in their drug interaction
`potential and to communicate risk to health care
`providers and patients [5].
`With regard to comparing drug interaction
`potential among drugs, the two macrolide im-
`munosuppressants, everolimus and sirolimus,
`differ in chemical structure and biotransforma-
`tion pathways [21–22] which in theory could
`yield differences in their susceptibility to meta-
`bolic drug interactions. Nonetheless, comparing
`the everolimus
`results presented here with
`published data from sirolimus suggests that they
`share a generally similar risk for CYP3A inhibi-
`tion drug interactions as interpreted against the
`background of this classification system.
`This classification system also serves as a
`framework to communicate drug interaction risk
`to clinicians. The risk of increased drug exposure
`when coadministering strong CYP3A inhibitors
`led to the recommendation to avoid these come-
`dications in the product labels of both everolimus
`[20] and sirolimus [13]. Coadministration of
`moderate CYP3A inhibitors, however, appears to
`be associated with a generally lower magnitude in
`the fold-increase of everolimus AUC; however, the
`healthy subject data indicate that some individuals
`are more susceptible to CYP3A inhibition interac-
`tions resulting in larger increases in exposure. The
`data further suggest that these individuals tend to
`have lower everolimus exposure in the absence of
`comedications as shown in Figure 2. This pattern
`has been observed in other drug interaction
`studies with CYP3A substrates [23–25].
`It
`is
`speculated that individuals with higher expression
`of CYP3A (manifested by lower exposure when
`the drug is given alone) have more enzyme to
`inhibit and thereby elicit a stronger drug interac-
`tion in the presence of a CYP3A inhibitor [24–26].
`In other words, intersubject variability in enzyme
`levels is likely a strong contributor to variability in
`the magnitude of drug interactions. In light of this
`underlying variability, moderate CYP3A inhibitors
`may generally be used with everolimus provided
`that
`it
`is accompanied by therapeutic drug
`monitoring to quantify the increase in everolimus
`
`Copyright # 2006 John Wiley & Sons, Ltd.
`
`exposure after adding the moderate inhibitor and
`to individually adjust
`the everolimus dose to
`compensate for the interaction [20].
`the CYP3A
`An additional application of
`inhibitor classification system in risk manage-
`ment is as a qualitative guide to drug interactions
`not specifically tested by the sponsor. This is
`explicitly recognized in the European Agency for
`the Evaluation of Medicinal Products drug
`interaction guidance which states:
`‘It seems
`in vivo studies with strong
`reasonable that
`inducers/inhibitors may be used to extrapolate
`qualitatively to other inducers/inhibitors of the
`same enzyme’ [4]. In the case of everolimus, the
`summary of product characteristics extends the
`recommendation to avoid strong CYP3A inhibi-
`tors based on ketoconazole data to include
`itraconazole, voriconazole, clarithromycin and
`ritonivir [20]. Similar qualitative extensions are
`made for moderate CYP3A inhibitors [20].
`While the CYP3A inhibitor classification sys-
`tem has many desirable traits, some investigators
`cite points for caution as well [27]. For example,
`when using the system to classify a new drug as
`an inhibitor with midazolam as the probe
`substrate, the data might not be extrapolatable
`to other substrates. The classification results may
`be study design-specific if the magnitude of the
`interaction depends on the drug doses studied,
`administration under single-dose or multiple-
`dose conditions, or relative timing of coadminis-
`tration (simultaneously versus delayed). Further-
`more,
`the potency of an individual drug
`interaction may be understated if multiple ‘mild’
`or ‘moderate’ inhibitors are coadministered in a
`patient’s drug regimen leading to a stronger
`combined interaction or if the victim drug is a
`substrate of multiple CYP enzymes or transpor-
`ters inhibited by the perpetrator drug.
`
`Conclusions
`
`Mindful of the caveats mentioned above, drug
`development scientists, regulatory reviewers and
`clinicians can use this CYP3A inhibitory classifi-
`cation system as a tool to design drug interaction
`programs,
`to manage regulatory risk and to
`guide pharmacotherapy in the patient care
`setting. The example of everolimus demonstrates
`
`Biopharm. Drug Dispos. 27: 421–426 (2006)
`DOI: 10.1002/bdd
`
`Ex. 1049-0005
`
`

`
`426
`
`J.M. KOVARIK ET AL.
`
`that applying a rational and systematic approach
`to drug interactions on everolimus yielded
`clinically useful, structured guidelines for dose
`adjustment.
`
`Acknowledgements
`
`The clinical studies described herein were spon-
`sored by Novartis Pharmaceuticals.
`
`References
`
`1. Venkatakrishnan K, von Moltke LL, Greenblatt DJ.
`Human drug metabolism and the cytochrome P450:
`J Clin
`in vitro models.
`application and relevance of
`Pharmacol 2001; 41: 1149–1179.
`2. United States Food and Drug Administration. Guidance for
`Industry: Drug Metabolism/Drug Interactions in the Drug
`Development Process: Studies In vitro. 1997.
`3. United States Food and Drug Administration. Guidance for
`Industry: In vivo Drug Metabolism/Drug Interaction Studies:
`Study Design, Data Analysis, and Recommendations for
`Dosing and Labeling. 1999.
`the Evaluation of Medicinal
`4. European Agency for
`Products. Note for Guidance on the Investigation of Drug
`Interactions. 1997.
`5. Bjornsson TD, Callaghan JT, Einolf HJ, et al. The conduct
`of in vitro and in vivo drug–drug interaction studies: a
`PhRMA perspective. J Clin Pharmacol 2003; 43: 443–469.
`6. Kovarik JM. Everolimus: a proliferation signal inhibitor
`targeting primary causes of allograft dysfunction. Drugs
`Today 2004; 40: 101–109.
`7. Kovarik JM, Beyer D, Bizot MN, et al. Blood concentra-
`tions of everolimus are markedly increased by ketocona-
`zole. J Clin Pharmacol 2005; 45: 514–518.
`8. Kovarik JM, Beyer D, Bizot MN, et al. Effect of multiple-
`dose erythromycin on everolimus pharmacokinetics. Eur J
`Clin Pharmacol 2005; 61: 35–38.
`9. Kovarik JM, Beyer D, Bizot MN, et al. Pharmacokinetic
`interaction between verapamil and everolimus in healthy
`subjects. Br J Clin Pharmacol 2005; 60: 434–437.
`10. Kovarik JM, Kalbag J, Figueiredo J, et al. Differential
`influence of two cyclosporine formulations on everolimus
`pharmacokinetics: a clinically relevant pharmacokinetic
`drug interaction. J Clin Pharmacol 2002; 42: 95–99.
`11. Kovarik JM, Hartmann S, Hubert M, et al. Pharmacoki-
`netic and pharmacodynamic assessments of HMG-CoA
`reductase inhibitors when coadministered with ever-
`olimus. J Clin Pharmacol 2002; 42: 222–228.
`12. Brignol N, McMahon LM, Luo S, et al. High-throughput
`semi-automated 96-well
`liquid/liquid extraction and
`liquid chromatography/mass spectrometric analysis of
`
`everolimus and cyclosporin A in whole blood. Rapid
`Commun Mass Spectrom 2001; 15: 898–907.
`13. Rapamune (sirolimus) [package insert] 2005.
`14. Lam Y, Ereshefsky L, Alfaro C, et al. In vivo inhibition of
`midazolam disposition by ketoconazole and fluoxetine,
`and comparison to in vitro prediction. Clin Pharmacol Ther
`1999; 65: 143.
`15. Olkkola KT, Backman JT, Neuvonen. Midazolam should
`be avoided in patients receiving the systemic antimycotics
`ketoconazole or itraconazole. Clin Pharmacol Ther 1994; 55:
`481–485.
`16. Tsunoda SM, Velez RL, von Moltke LL, Greenblatt DJ.
`Differentiation of intestinal and hepatic cytochrome P450
`3A4 activity with use of midazolam as an in vivo probe:
`effect of ketoconazole. Clin Pharmacol Ther 1999; 66:
`461–471.
`17. Olkkola KT, Aranko K, Luurila H, et al. A potentially
`hazardous interaction between erythromycin and mid-
`azolam. Clin Pharmacol Ther 1993; 53: 298–305.
`18. Zimmermann T, Yeates RA, Laufen H, et al. Influence of
`the antibiotics erythromycin and azithromycin on the
`pharmacokinetics and pharmacodynamics of midazolam.
`Arzneimittelforschung 1996; 46: 213–217.
`19. Backman JT, Olkkola KT, Aranko K, et al. Dose of
`midazolam should be reduced during diltiazem and
`treatments. Br J Clin Pharmacol 1994; 37:
`verapamil
`221–225.
`20. Certican (everolimus) [package insert] 2004. Available at
`www.mpa.se/eng.
`21. Jacobsen W, Serkova N, Hausen B, et al. Comparison of
`the in vitro metabolism of the macrolide immunosuppres-
`sants sirolimus and RAD. Transplant Proc 2001; 33:
`514–515.
`22. Kuhn B, Jacobsen W, Christians U, et al. Metabolism of
`sirolimus and its derivative everolimus by cytochrome
`P4503A4: insights from docking, molecular dynamics, and
`quantum chemical calculations. J Med Chem 2001; 44:
`2027–2034.
`23. Gross AS, Goh YD, Addison RS, et al.
`Influence of
`juice on cisapride pharmacokinetics. Clin
`grapefruit
`Pharmacol Ther 1999; 65: 395–401.
`24. Gorski JC, Jones DR, Haehner-Daniles BD, et al. The
`contribution of
`intestinal and hepatic CYP3A to the
`interaction between midazolam and clarithromycin. Clin
`Pharmacol Ther 1998; 64: 133–143.
`25. Pinto AG, Wang YH, Chalasani N, et al. Inhibition of
`human intestinal wall metabolism by macrolide antibio-
`tics: effect of clarithromycin on cytochrome P450 3A4/5
`activity and expression. Clin Pharmacol Ther 2005; 77:
`178–188.
`26. Huang SM, Hall SD, Watkins P, et al. Drug interactions
`with herbal products and grapefruit juice: a conference
`report. Clin Pharmacol Ther 2004; 75: 1–12.
`27. Huang SM. Drug–drug interactions. In Applications of
`Pharmacokinetic Principles in Drug Development, Krishna R
`(ed.). Kluwer Academic/Plenum Publishers: New York,
`2004: 307–331.
`
`Copyright # 2006 John Wiley & Sons, Ltd.
`
`Biopharm. Drug Dispos. 27: 421–426 (2006)
`DOI: 10.1002/bdd
`
`Ex. 1049-0006

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