throbber

`

`114
`
`L. L. von Moltke et al.
`
`tations and drawbacks. The issue of risk to human subjects
`is always of concern, even if the actual risk is small and
`acceptable. The increasingly stringent regulatory require(cid:173)
`ments for control and monitoring of premarketing studies,
`as well as the scientific needs for appropriate design and
`adequate sample size, generally cause these studies to be
`costly and time-consuming. As a consequence, there is a
`realistic limit to the number and scope of clinical drug
`interaction studies that can be performed, whether in the
`premarketing phases of drug development or after approval
`for clinical use. Inevitably some important drug interac(cid:173)
`tions will be overlooked simply because they were not
`among those that were studied; the often-cited case of the
`terfenadine-ketoconazole interaction is an important ex(cid:173)
`ample [12]. On the other hand, many clinical interaction
`studies are negative.
`These dilemmas have stimulated the search for alterna(cid:173)
`tive approaches to studying drug interactions. The possibil(cid:173)
`ity of predicting in vivo metabolic events from in vitro
`models has been discussed for decades and some of the
`fundamental principles and assumptions involved in the in
`vitro-in vivo predictions have been described [13-15]. Iden(cid:173)
`tifying the cytochromes involved in biotransformation of a
`specific drug can identify sources of variation in clearance
`secondary to population demographics, genetic polymor(cid:173)
`phisms, extrahepatic enzyme distribution, and known
`chemical inducers or inhibitors. The ability to accurately
`predict drug interactions secondary to inhibition of cyto(cid:173)
`chrome metabolism has been greatly enhanced by the
`knowledge of which cytochrome mediates a specific reac(cid:173)
`tion.
`
`MODEL COMPONENTS AND ASSUMPTIONS
`
`Predictive models for in vitro-in vivo scaling of pharmaco(cid:173)
`kinetic drug interaction can be constructed from a combi(cid:173)
`nation of laboratory and theoretical components. Many of
`the pieces are based on well-established principles, while
`others represent hypotheses and assumptions. The follow(cid:173)
`ing describes the steps in development of a representative
`model.
`
`Biotransformation In Vitro
`
`The objective of the laboratory component is to replicate in
`vitro the principal metabolic pathways of the index sub(cid:173)
`strate as they occur in vivo, and to characterize the mech(cid:173)
`anism and quantitative inhibiting potency of the model
`inhibitor.
`Human liver microsomes appear to be the most applica(cid:173)
`ble in vitro system for purposes of scaling, since the various
`cytochromes are present in proportion to their in vivo
`representation. This is particularly important when more
`than one specific cytochrome contributes to the biotrans(cid:173)
`formation of the index substrate. Microsomal preparations
`from various animal species have been used for predictive
`
`studies of drug interactions, but no species has been
`generally established as a substitute for human tissue.
`Mathematical analysis of in vitro reaction kinetic data in
`the absence of inhibitors usually includes application of the
`Michaelis-Menten (MM) equation, with or without linear(cid:173)
`izing transformation of the data [16]. However, with in(cid:173)
`creasing numbers of data points and/or range of substrate
`concentrations, it may become apparent that the simplest
`form of the MM equation is not applicable. Examples of
`complicating features requiring modification of modeling
`approaches include: simultaneous contribution of two or
`more enzymes with distinct Km values [17, 18]; cooperative
`binding resulting in apparent substrate activation [19-23];
`concentration-dependent inhibition by substrate and/or
`product [21, 22, 24-26]; sequential metabolism of the
`primary metabolite to yield secondary downstream meta(cid:173)
`bolic products. Goodness-oHit criteria from mathematical
`modeling techniques may tentatively identify which one
`(or more) of these features is applicable to a given data set,
`but definitive confirmation of a biochemical mechanism
`requires supplemental information [21]. In any case, com(cid:173)
`plex reaction kinetic mechanisms may compromise the
`conceptual validity of absolute or relative intrinsic clear(cid:173)
`ance values as calculated from the V maxlKm ratio [27].
`It is nonetheless important to recognize that the accuracy
`of potency calculations for specific chemical inhibitors is
`not necessarily compromised importantly when simplifying
`approximations are used to handle complex reaction kinet(cid:173)
`ics. When two distinct enzyme components contribute to a
`specific biotransformation, the component with the lowest
`Km (the "high-affinity" site) may account for a large
`percentage of net intrinsic clearance. In such cases, the
`high-Km enzyme may be approximated by a simple linear
`function, and inhibition of the low-Km enzyme accounts for
`most of the clinically important activity of chemical inhib(cid:173)
`itors [17].
`
`Chemical Inhibitors In Vitro
`
`Coincubation of a candidate chemical inhibitor with the
`target substrate using the same in vitro system can yield an
`estimate of the intrinsic inhibitory potency of the candidate
`compound. The most straightforward approach utilizes a
`fixed concentration of substrate [5] coincubated with var(cid:173)
`ious concentrations of inhibitor (Fig. 1). Analysis of the
`relation between inhibitor concentration [I] and reaction
`velocity decrement (i.e. reaction velocity at that concen(cid:173)
`tration of inhibitor divided by reaction velocity with no
`inhibitor present) may yield an estimated concentration of
`inhibitor corresponding to a 50% decrease in reaction
`velocity (IC50). This approach has the advantage of being
`model independent. That is, 1C50 can be calculated without
`knowledge of the biochemical mechanism of inhibition.
`The 1C50 values are quite useful when comparing the
`relative inhibitory potency of different candidate inhibitors
`of the same chemical class, such as the SSRI antidepres(cid:173)
`sants [17] or azole antifungal agents [28]. On the other
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2053, Page 2
`
`

`

`

`

`

`

`

`

`118
`
`L. L. von Moltke et al.
`
`TABLE 2. Partitioning of Ketoconazole Between Plasma and
`Liver Tissue
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`>
`:>
`Z
`0
`W
`>
`0::
`W
`CI)
`a:J
`0
`
`/
`
`/
`
`/
`
`/
`
`/,
`
`/ab
`
`/
`
`/
`
`/
`
`c
`
`d
`
`/
`
`/
`
`/
`
`/
`
`/
`
`/
`
`/
`
`/
`
`/
`
`/
`
`/
`
`/
`
`/
`
`/e
`
`/
`
`/
`
`/
`
`20
`
`40
`
`60
`
`80
`
`100
`
`PREDICTED IN VITRO
`FIG. 5. Outcome of six studies evaluating the actual inhibition
`of oral desipramine clearance in humans as compared with that
`predicted for the in vitro model. In vitro competitive K; values
`for six SSRls versus desipramine hydroxylation in human liver
`microsomes were determined as described previously [94-96].
`Predicted inhibition (x-axis) was determined using the model
`described in the text, based on K; values and plasma SSRI
`concentrations which were then corrected for anticipated he(cid:173)
`patic uptake. Actual inhibition of desipramine clearance in vivo
`(y-axis) was determined in clinical pharmacokinetic studies.
`Dashed line is the line of identity (y = x). Symbols a and bare
`studies of fluoxetine/norfluoxetine [97,98]; c and d are studies
`of paroxetine [100, 101]; and e and f are studies of sertraline/
`desmethylsertraline [98, 99].
`
`these SSRls [97-100]. Application of the predictive model
`yields reasonably accurate forecasting of in vivo desipramine
`clearance decrements based on in vitro data, provided that
`values of [I] determined from plasma SSRI concentrations
`are adjusted for anticipated liver partitioning, consistent
`with experimental and human autopsy data (Fig. 5). Use of
`unbound or even total SSRI plasma concentrations in the
`predictive model leads to substantial underestimation of
`observed clearance decrements.
`
`SSRI Antidepressants and P450-3A Substrates
`
`In vivo decrements in clearance of alprazolam, a substrate
`for P450-3A isoforms, due to coadministration of fluoxetine
`[101, 102] or fluvoxamine [103] were reasonably well
`predicted from in vitro data [86, 95]. Minimal clinical
`interaction of terfenadine with paroxetine was predicted in
`vitro [104] and confirmed in vivo [105]. However, the
`predictive model forecasted a significant decrement in
`triazolam clearance due to coadministration of fluoxetine
`[82], whereas a clinical study demonstrated only a minimal
`in vivo pharmacokinetic interaction [106]. The reasons for
`these discrepancies are not established. Gastrointestinal
`P450-3A isoforms are likely to account for an important
`
`Experimental system
`Rat, in vivo
`Rat, in vivo
`Rat, in vivo
`Mouse, in vivo
`In vitro: human plasma
`vs liver homogenate
`* At plasma concentrations <3.8 I-Lg/mL, liver:plasma partition ratios exceeded 3.2.
`
`Liver:plasma
`partition ratio
`
`5
`0.5-1.0
`~3.2*
`2.03
`1.12
`
`Ref.
`79
`80
`81
`82
`82
`
`azole (total plasma concentration 2::1 f.Lg/mL) reduced oral
`midazolam clearance by an average of 94%. By applying
`these Ki values along with the total plasma ketoconazole
`concentrations for [I] in the predictive model, the pre(cid:173)
`dicted degrees of oral midazolam clearance inhibition are
`95% [36] and >95% (assuming [S] « Km) [24], respec(cid:173)
`tively. However, use of the projected unbound plasma
`ketoconazole concentration (0.01 X total plasma concen(cid:173)
`tration) yields underestimations of the degree of clearance
`inhibition (16 and 82%, respectively). Reasonable predic(cid:173)
`tions have also been derived, using total plasma ketocon(cid:173)
`azole concentrations and in vitro Ki values, for the clinical
`interactions of ketoconazole with triazolam [82, 84], terfe(cid:173)
`nadine [12, 85], and alprazolam [86, 87]. It is of interest that
`coadministration of ketoconazole reduces clearance of oral
`alprazolam by an average of 69% [87], although absolute
`bioavailability of oral alprazolam is greater than 80%
`[88,89].
`Itraconazole, like ketoconazole, is 99% bound to blood/
`plasma components [78], whereas liver versus total plasma
`concentration ratios in experimental models range from
`11:1 to 20:1 [79]. The competitive Ki for itraconazole versus
`a-OH-midazolam formation was 0.28 f.LM [24], and the
`impairment of midazolam clearance by coadministration of
`itraconazole (total plasma concentrations generally ~0.1
`f.LM) in three clinical studies was 90, 83, and 85% [83, 90,
`91]. These are well predicted by the scaling model if a
`liver:plasma ratio in the range of 10:1 to 20:1 is assumed for
`itraconazole, but poorly predicted based on total or un(cid:173)
`bound plasma itraconazole levels. It is of interest that
`itraconazole coadministration also impairs clearance of
`intravenous midazolam [91].
`
`SSRI Antidepressants and P450-2D6 Substrates
`
`In vitro inhibition of desipramine hydroxylation, a reaction
`mediated largely if not entirely by human cytochrome
`P450-2D6 [92, 93], has been evaluated for SSRI antidepres(cid:173)
`sants and their pertinent metabolites [94, 95]. Based on in
`vitro competitive Ki values, fluoxetine, norfluoxetine, and
`paroxetine are potent inhibitors, while sertraline, desmeth(cid:173)
`ylsertraline, and fluvoxamine are substantially weaker [96].
`Clinical studies have evaluated the impairment of desipra(cid:173)
`mine clearance due to coadministration of a number of
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2053, Page 6
`
`

`

`Predicting Drug Interactions
`
`119
`
`component of presystemic extraction after oral administra(cid:173)
`tion of drugs such as triazolam. Inhibition of P450-3A
`isoforms by fluoxetine is largely attributable to the metab(cid:173)
`olite norfluoxetine [53, 107], which may have incomplete
`access to gastrointestinal mucosal cells following adminis(cid:173)
`tration of the parent compound. It is also possible that
`fluoxetine and other SSRIs may have inducing effects that
`offset competitive inhibition under some circumstances.
`
`COMMENT
`
`Major advances in the use of in vitro systems to understand
`human drug metabolism have logically led to exploration of
`the application of such systems to focus, supplement, or
`even replace, human pharmacokinetic study programs. In
`vitro models to predict drug interactions based on in vitro
`data have been proposed. The models are preliminary, and
`the drawbacks, limitations, weaknesses, and assumptions
`are numerous. The models can, in principle, forecast the
`magnitude of an interaction, but not its potential clinical
`consequences [96]. Nonetheless, the concept is promising,
`and the potential long-range benefit is a drug-development
`process that is more rapid and cost-effective, with reduced
`risk to human subjects.
`
`This work was supported by Grants MH-34223, DA-OS2S8, MH-
`19924, and RR-000S4 from the Department of Health and Human
`Services, and by a grant-in-aid from Pharmacia & Upjohn, Kalama(cid:173)
`zoo, MI. Dr. von Moltke is the recipient of a Scientist Development
`Award (K21-MH-01237) from the National Institutes of Mental
`Health. Dr. Schmider was the recipient of a Merck Sharp & Dohme
`International Fellowship in Clinical Pharmacology. The authors are
`grateful for the assistance of Su Xiang Duan, Monette M. Cotreau(cid:173)
`Bibbo, and Jeffrey Grassi.
`
`References
`
`1. Beaune PH and Guengerich FP, Human drug metabolism in
`vitro. Pharmacol Ther 37: 193-211, 1988.
`2. Birkett DJ, Mackenzie PI, Veronese ME and Miners JO, In
`vitro approaches can predict human drug metabolism. Trends
`Pharmacol Sci 14: 292-294, 1993.
`3. Br¢sen K, Recent developments in hepatic drug oxidation:
`Implications for clinical pharmacokinetics. Clin Pharmacoki(cid:173)
`net 18: 220-239, 1990.
`4. Gonzalez FJ, Human cytochromes P450: Problems and pros(cid:173)
`pects. Trends Pharmacol Sci 13: 346-352, 1992.
`5. Guengerich FP, Human cytochrome PA50 enzymes. Life Sci
`50: 1471-1478, 1992.
`6. Ketter T A, Flockhart DA, Post RM, Denicoff KD, Pazzaglia
`PJ, Marangell LB, George MS and Callahan AM, The
`emerging role of cytochrome P450 3A in psychopharmacol(cid:173)
`ogy. J Clin Psychopharmacol 15: 387-398, 1995.
`7. Murray M, P450 enzymes: Inhibition mechanisms, genetic
`regulation and effects of liver disease. Clin Pharmacokinet 23:
`132-146, 1992.
`8. Parkinson A, An overview of current cytochrome P450
`technology for assessing the safety and efficacy of new
`materials. Toxicol Pathol 24: 45-57, 1996.
`9. Rodrigues AD, Use of in vitro human metabolism studies in
`drug development. Biochem Phalmacol48: 2147-2156, 1994.
`
`10. Watkins PB, Role of cytochrome P450 in drug metabolism
`and hepatotoxicity. Semin Liver Dis 10: 235-250, 1990.
`11. Wrighton SA and Stevens JC, The human hepatic cyto(cid:173)
`chromes P450 involved in drug metabolism. Crit Rev Toxicol
`22: 1-21, 1992.
`12. Honig PK, Wortham DC, Zamani K, Conner DP, Mullin JC
`and Cantilena LR, T erfenadine- ketoconazole interaction:
`Pharmacokinetic and electrocardiographic consequences. J
`Am Med Assoc 269: 1513-1518, 1993.
`13. Leemann TD and Dayer P, Quantitative prediction of in vivo
`drug metabolism and interactions from in vitro data. In:
`Advances in Drug Metabolism in Man (Eds. Pacifici GM and
`Fracchia GN), pp. 783-830. The European Commission,
`Luxembourg, 1995.
`14. Boobis AR, Prediction of inhibitory drug-drug interactions
`by studies in vitro. In: Advances in Drug Metabolism in Man
`(Eds. Pacifici GM and Fracchia GN), pp. 513-539. The
`European Commission, Luxembourg, 1995.
`15. Tucker GT, The rational selection of drug interaction
`studies: Implications of recent advances in drug metabolism.
`Int J Clin Pharmacol Ther Toxicol 30: 550-553, 1992.
`16. Segel IH, Enzyme Kinetics. Wiley, New York, 1975.
`17. Br¢sen K, Skjelbo E, Rasmussen BB, Poulsen HE and Loft S,
`Fluvoxamine is a potent inhibitor of cytochrome P4501A2.
`Biochem. Pharmacol45: 1211-1214, 1993.
`18. von Moltke LL, Greenblatt DJ, Duan SX, Schmider J,
`Kudchadker L, Fogelman SM, Harmatz JS and Shader RI,
`Phenacetin O-deethylation by human liver microsomes in
`vitro: Inhibition by chemical probes, SSRI antidepressants,
`nefazodone and venlafaxine. Psychopharmacology 128: 398-
`407, 1996.
`19. Ueng Y-F, Kuwabara T, Chun Y-J and Guengerich FP,
`Cooperativity in oxidations catalyzed by cytochrome P450-
`3A4. Biochemistry 36: 370-381, 1997.
`20. Shou M, Grogan J, Mancewicz JA, Krausz KW, Gonzalez FJ,
`Gelboin HV and Korzekwa KR, Activation of CYP3A4:
`Evidence for the simultaneous binding of two substrates in a
`cytochrome P450 active site. Biochemistry 33: 6450-6455,
`1994.
`21. Schmider J, Greenblatt DJ, Harmatz JS and Shader RI,
`Enzyme kinetic modelling as a tool to analyse the behaviour
`of cytochrome P450 catalysed reactions: Application to
`amitriptyline N-demethylation. Br J Clin Pharmacol 41:
`593-604, 1996.
`22. Schmider J, Greenblatt DJ, von Moltke LL, Harmatz JS and
`Shader RI, N-Demethylation of amitriptyline in vitro: Role
`of cytochrome PA50 3A (CYP3A) isoforms and effect of
`metabolic inhibitors. J Pharmacol Exp Ther 275: 592-597,
`1995.
`23. Andersson T, Miners JO, Veronese ME and Birkett DJ,
`Diazepam metabolism by human liver microsomes is medi(cid:173)
`ated by both S-mephenytoin hydroxylase and CYP3A iso(cid:173)
`forms. Br J Clin Pharmacol38: 131-137, 1994.
`24. von Moltke LL, Greenblatt DJ, Schmider J, Duan SX,
`Wright CE, Harmatz JS and Shader RI, Midazolam hydroxy(cid:173)
`lation by human liver micro somes in vitro: Inhibition by
`fluoxetine, norfluoxetine, and by azole antifungal agents.
`J Clin Pharmacol 36: 783-791, 1996.
`25. Ghosal A, Satoh H, Thomas PE, Bush E and Moore D,
`Inhibition and kinetics of cytochrome P4503A activity in
`microsomes from rat, human, and eDNA-expressed human
`cytochrome P450. Drug Metab Dispos 24: 940-947, 1996.
`26. Kronbach T, Mathys D, Umeno M, Gonzalez FJ and Meyer
`UA, Oxidation of midazolam and triazolam by human liver
`cytochrome P450IIlA4. Mol Pharmacol36: 89-96, 1989.
`27. Houston JB, Utility of in vitro drug metabolism data in
`predicting in vivo metabolic clearance. Biochem Pharmacol
`47: 1469-1479, 1994.
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2053, Page 7
`
`

`

`120
`
`L. L. von Moltke et al.
`
`28. Back DJ and Tjia JF, Comparative effects of the antimycotic
`drugs ketoconazole, fluconazole, itraconazole and terbinafine
`on the metabolism of cyclosporin by human liver micro(cid:173)
`somes. Br I Clin Pharmacal 32: 624-626, 1991.
`29. Harmatz JS and Greenblatt DJ, Falling off the straight line:
`Some hazards of correlation and regression. I CUn Psycho(cid:173)
`pharmacal 12: 75-78, 1992.
`30. Barlow RB, Effects of 'rogue' points on non-linear fitting.
`Trends Pharmacol Sci 14: 399-403, 1993.
`31. Halpert JR, Structural basis of selective cytochrome P450
`inhibition. Annu Rev Pharmacal Toxical35: 29-53, 1995.
`32. Silverman RB, Mechanism-based enzyme
`inactivators.
`Methods Enzymol 249: 241-282, 1992.
`33. von Moltke LL, Greenblatt DJ, Schmider J, Harmatz JS and
`Shader RI, Metabolism of drugs by cytochrome P450 3A
`isoforms. Clin Pharmacokinet 29 (Suppl 1): 33-44, 1995.
`34. Wilkinson GR, Clearance approaches in pharmacology.
`Pharmacol Rev 39: 1-47, 1987.
`35. Wilkinson GR and Shand DG, A physiological approach to
`hepatic drug clearance. Clin Pharmacol Ther IS: 377-390,
`1975.
`36. Wrighton SA and Ring BJ, Inhibition of human CYP3A
`catalyzed l' -hydroxy midazolam formation by ketoconazole,
`nifedipine, erythromycin, cimetidine, and nizatidine. Pharm
`Res 11: 921-924, 1994.
`37. Olkkola KT, Aranko K, Luurila H, Hiller A, Saarnivaara L,
`Himberg J-J and Neuvonen PJ, A potentially hazardous
`interaction between erythromycin and midazolam. CUn
`Pharmacol Ther 53: 298-305, 1993.
`38. Echizen H, Kawasaki H, Chiba K, Tani M and Ishizaki T, A
`potent inhibitory effect of erythromycin and other macrolide
`antibiotics on the mono-N-dealkylation metabolism of diso(cid:173)
`pyramide with human liver microsomes. I Pharmacal Exp
`Ther 264: 1425-1431, 1993.
`39. Pang KS, Rowland M and Tozer TN, In vivo evaluation of
`Michaelis-Menten constants of hepatic drug-eliminating
`systems. Drug Metab Dispos 6: 197-200, 1978.
`40. Mitra AK, Thummel KE, Kalhorn TF, Kharasch ED, Unad(cid:173)
`kat JD and Slattery JT, Inhibition of sulfamethoxazole
`hydroxylamine formation by fluconazole in human liver
`microsomes and healthy volunteers. Clin Pharmacol Ther 59:
`332-340, 1996.
`41. Kunze KL and Trager WF, Warfarin-fluconazole III. A
`rational approach to management of a metabolically based
`drug interaction. Drug Metab Dispos 24: 429-435, 1996.
`42. Ervine CM, Matthew DE, Brennan B and Houston JB,
`Comparison of ketoconazole and fluconazole as cytochrome
`P450 inhibitors. Drug Metab Dispos 24: 211-215, 1996.
`43. Ring BJ, Binkley SN, Roskos Land Wrighton SA, Effect of
`fluoxetine, norfluoxetine, sertraline and desmethyl sertraline
`on human CYP3A catalyzed l' -hydroxy midazolam forma(cid:173)
`tion in vitro. I Pharmacol Exp Ther 275: 1131-1135, 1995.
`44. Pacifici GM and Viani A, Methods of determining plasma
`and tissue binding of drugs: Pharmacokinetic consequences.
`CUn Pharmacokinet 23: 449-468, 1992.
`45. Kurz Hand Fichtl B, Binding of drugs to tissues. Drug Metab
`Rev 14: 467-510, 1983.
`46. Tsuru M, Erickson RR and HoltzmanJL, The metabolism of
`phenytoin by isolated hepatocytes and hepatic microsomes
`from male rats. I Pharmacal Exp Ther 222: 658-661, 1982.
`47. Bogeyevitch MA, Gillam EMJ, Reilly PEB and Winzor DJ,
`Physical partitioning as the major source of metoprolol
`uptake by hepatic microsomes. Biochem Pharmacal 36: 4167-
`4168, 1987.
`48. Miyauchi S, Sawada Y, Iga T, Hanano M and Sugiyama Y,
`Comparison of the hepatic uptake clearances of fifteen drugs
`with a wide range of membrane permeabilities in isolated rat
`
`hepatocytes and perfused rat livers. Pharm Res 10: 434-440,
`1993.
`49. Chou C-H, Evans AM, Fornasini G and Rowland M,
`Relationship between lipophilicity and hepatic dispersion
`and distribution for a homologous series of barbiturates in
`the isolated perfused in situ rat liver. Drug Metab Dispos 21:
`933-938, 1993.
`50. Schuhmann G, Fichtl Band Kurz H, Prediction of drug
`distribution in vivo on the basis of in vitro binding data.
`Biopharm Drug Dispos S: 73-86, 1987.
`51. Harashima H, Sugiyama Y, Sawada Y, Iga T and Hanano M,
`Comparison between in vivo and in vitro tissue-to-plasma
`unbound concentration ratios (Kp,f) of quinidine in rats.
`I Pharm Pharmacal 36: 340-342, 1984.
`52. Lin JH, Sugiyama Y, Awazu Sand Hanano M, In vitro and in
`vivo evaluation of the tissue-to-blood partition coefficient for
`physiological pharmacokinetic models. I Pharmacakinet Bio(cid:173)
`pharm 10: 637-647, 1982.
`53. Greenblatt DJ, von Moltke LL, Schmider J, Harmatz JS and
`Shader RI, Inhibition of human cytochrome P450-3A iso(cid:173)
`forms by fluoxetine and norfluoxetine: In vitro and in vivo
`studies. I CUn Pharmacol36: 792-798, 1996.
`54. Aravagiri M, Marder SR, Yuwiler A, Midha KK, Kula NS
`and Baldessarini RJ, Distribution of fluphenazine and its
`metabolites in brain regions and other tissues of the rat.
`Neuropsychopharmacalogy 13: 235-247, 1995.
`55. Scavone JM, Friedman H, Greenblatt DJ and Shader RI,
`Effect of age, body composition, and lipid solubility on
`benzodiazepine tissue distribution in rats. Arzeneimittelfors(cid:173)
`chung 37: 2-6, 1987.
`56. Xie X, Steiner SH and Bickel MH, Kinetics of distribution
`and adipose tissue storage as a function of lipophilicity and
`chemical structure: II. Benzodiazepines. Drug Metab Dispos
`19: 15-19, 1991.
`57. Ferslew KE, Hagardorn AN and McCormick WF, Postmor(cid:173)
`tem determination of the biological distribution of sufentanil
`and midazolam after an acute intoxication. I Forensic Sci 34:
`249-257, 1989.
`58. Jortani SA, Valentour JC and Poklis A, Thioridazine enan(cid:173)
`tiomers in human tissues. Forensic Sci Int 64: 165-170, 1994.
`59. Levine BS, Wu S-C, Goldberger BA and Caplan YH, Two
`fatalities involving haloperidol. I Anal Toxicol15: 282-284,
`1991.
`60. Friedman H, Ochs HR, Greenblatt DJ and Shader RI, Tissue
`distribution of diazepam and its metabolite desmethyldiaz(cid:173)
`epam: A human autopsy study. I CUn Pharmacal 25: 613-
`615, 1985.
`61. Wong RJ, The determination of the trizolobenzodiazepine
`triazolam in post mortem samples. I Anal ToxicalS: 10-13,
`1984.
`62. Levine B, Jenkins AJ and Smialek JE, Distribution of
`sertraline in postmortem cases. I Anal ToxicollS: 272-274,
`1994.
`63. Bailey DN and Shaw RF, Interpretation of blood and tissue
`concentrations in fatal self-ingested overdose involving am(cid:173)
`itriptyline: An update (1978-1979). I Anal Toxicol 4:
`232-236, 1980.
`64. Worm K, Kringsholm Band Steentoft A, Clozapine cases
`with fatal, toxic or therapeutic concentrations. Int I Legal
`Med 106: 115-118, 1993.
`65. Fraser AD, Isner AF and Perry RA, Distribution of trimipra(cid:173)
`mine and its major metabolites in a fatal overdose case. I
`Anal Toxicalll: 168-170, 1987.
`66. Hilberg T, Morland J and Bjorneboe A, Postmortem release
`of amitriptyline from the lungs; a mechanism of postmortem
`drug redistribution. Forensic Sci Int 64: 47-55, 1994.
`67. Jones GR and Pounder DJ, Site dependence of drug concen-
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2053, Page 8
`
`

`

`Predicting Drug Interactions
`
`121
`
`trations in postmortem blood-A case study. ] Anal Toxicol
`11: 186-190, 1987.
`68. Hilberg T, Bugge A, Beylich K-M, Ingum ], Bj\Zlrneboe A
`and M\Zlrland], An animal model of postmortem amitripty(cid:173)
`line redistribution. ] Forensic Sci 38: 81-90, 1993.
`69. Hilberg T, Bugge A, Beylich K-M, M\Zlrland] and Bj\Zlrneboe
`A, Diffusion as a mechanism of postmortem drug redistribu(cid:173)
`tion: An experimental study in rats. IntJ Legal Med 105:
`87-91, 1992.
`70. Morgan D] and Smallwood RA, Clinical significance of
`pharmacokinetic models of hepatic elimination. Clin Phar(cid:173)
`macokinet 18: 61-76, 1990.
`71. Pond SM and Tozer TN, First-pass elimination: Basic
`concepts and clinical consequences. Clin Pharmacokinet 9:
`1-25, 1984.
`72. Kolars ]C, Schmiedlin-Ren P, Schuetz ]D, Fang C and
`Watkins PB, Identification of rifampin-inducible P450IIIA4
`(CYP3A4) in human small bowel enterocytes. ] Clin Invest
`90: 1871-1878, 1992.
`73. Lown KS, Kolars ]C, Thummel KE, Barnett ]L, Kunze KL,
`Wrighton SA and Watkins PB, Interpatient heterogeneity
`in expression of CYP3A4 and CYP3A5 in small bowel: Lack
`of prediction by the erythromycin breath test. Drug Metab
`Dispos 22: 947-955, 1994.
`74. Paine MF, Shen DD, Kunze KL, Perkins ]D, Marsh CL,
`McVicar ]P, Barr DM, Gillies BS and Thummel KE, First(cid:173)
`pass metabolism of midazolam by the human intestine. Clin
`Pharmacal Ther 60: 14-24, 1996.
`75. Thummel KE, O'Shea D, Paine MF, Shen DD, Kunze KL,
`Perkins ]D and Wilkinson GR, Oral first-pass elimination of
`midazolam
`involves both gastrointestinal and hepatic
`CYP3A-mediated metabolism. Clin Pharmacal Ther 59: 491-
`502, 1996.
`76. Hebert MF, Roberts ]P, Pruesksaritanont T and Benet LZ,
`Bioavailability of cyclosporine with concomitant rifampin
`administration is markedly less than predicted by hepatic
`enzyme induction. Clin Pharmacal Ther 52: 453-457, 1992.
`77. Gomez DY, Wacher V], Tomlanovich S], Hebert MF and
`Benet LZ, The effects of ketoconazole on the intestinal
`metabolism and bioavailability of cyclosporine. Clin Pharma(cid:173)
`cal Ther 58: 15-19, 1995.
`78. Como ]A and Dismukes WE, Oral azole drugs as systemic
`antifungal therapy. N EnglJ Med 330: 263-272, 1994.
`79. Heykants ], Michiels M, Meuldermans W, Monbaliu ],
`Lavrijsen K, Van Peer A, Levron ]C, Woestenborghs Rand
`Cauwenbergh G, The pharmacokinetics of itraconazole in
`animals and man: An overview. In: Recent Trends in the
`Discovery, Development and Evaluation of Antifungal Agents
`(Ed. Fromtling RA), pp. 223-249. ]. R. Prous Science
`Publishers, S. A., Barcelona, 1987.
`80. Riley CM and James MO, Determination ofketoconazole in
`the plasma, liver, lung, and adrenal of the rat by high(cid:173)
`performance liquid chromatography. ] Chromatogr 377:
`287-294, 1986.
`81. Matthew D, Brennan B, Zomorodi K and Houston ]B,
`Disposition of azole antifungal agents. 1. Nonlinearities in
`ketoconazole clearance and binding in rat liver. Pharm Res
`10: 418-422, 1993.
`82. von Moltke LL, Greenblatt D], Harmatz ]S, Duan SX,
`Harrel LM, Cotreau-Bibbo MM, Pritchard GA, Wright CE
`and Shader RI, Triazolam biotransformation by human liver
`microsomes in vitro: Effects of metabolic inhibitors and
`clinical confirmation of a predicted interaction with keto(cid:173)
`conazole. ] Pharmacal Exp Ther 276: 370-379, 1996.
`83. Olkkola KT, Backman ]T and Neuvonen P], Midazolam
`should be avoided in patients receiving the systemic anti(cid:173)
`mycotics ketoconazole or itraconazole. Clin Pharmacol Ther
`55: 481-485, 1994.
`
`84. Varhe A, Olkkola KT and Neuvonen P], Oral triazolam is
`potentially hazardous to patients receiving systemic antimy(cid:173)
`catics ketoconazole or itraconazole. Clin Pharmacol Ther 56:
`601-607, 1994.
`85. von Moltke LL, Greenblatt D], Duan SX, Harmatz ]S and
`Shader RI, In vitro prediction of the terfenadine-ketocon(cid:173)
`azole pharmacokinetic interaction. ] Clin Pharmacal 34:
`1222-1227, 1994.
`86. von Moltke LL, Greenblatt D], Cotreau-Bibbo MM, Har(cid:173)
`matz ]S and Shader RI, Inhibitors of alprazolam metabolism
`in vitro: Effect of serotonin-reuptake-inhibitor antidepres(cid:173)
`sants, ketoconazole and quinidine. BrJ Clin Pharmacol 38:
`23-31, 1994.
`87. Wright CE, Greenblatt D], von Moltke LL, Ehrenberg BL,
`Harmatz ]S, Corbett K and Shader RI, Ketoconazole inhi(cid:173)
`bition of triazolam and alprazolam clearance: Differential
`kinetic and dynamic consequences. Clin Pharmacal Ther 61:
`183, 1997.
`88. Lin K-M, Lau ]K, Smith R, Phillips P, Antal E and Poland
`RE, Comparison of alprazolam plasma levels in normal
`Asian and Caucasian male volunteers. Psychopharmacalogy
`96: 365-369, 1988.
`89. Smith RB, Kroboth PD, Vanderlugt]T, Phillips]P and ]uhl
`RP, Pharmacokinetics and pharmacodynamics of alprazolam
`after oral and IV administration. Psychopharmacology 84:
`452-456, 1984.
`90. Ahonen], Olkkola KT and Neuvonen P], Effect of itracon(cid:173)
`azole and terbinafine on the pharmacokinetics and pharma(cid:173)
`codynamics of midazolam in healthy volunteers. Br ] Clin
`Pharmacol40: 270-272, 1995.
`91. Olkkola KT, Ahonen] and Neuvonen P], The effect of the
`systemic antimycotics, itraconazole and fluconazole, on the
`pharmacokinetics and pharmacodynamics of intravenous
`and oral midazolam. Anesth Analg 82: 511-516, 1996.
`92. von Bahr C, Spina E, Birgersson C, Ericsson 6, Goransson
`M, Henthorn T and Sjoqvist F, Inhibition of desmethylimi(cid:173)
`pramine 2-hydroxylation by drugs in human liver micro(cid:173)
`somes. Biochem Pharmacal 34: 2501-2505, 1985.
`93. Ohmori S, Takeda S, Rikihisa T, Kiuchi M, Kanakubo Y and
`Kitada M, Studies on cytochrome P450 responsible for
`oxidative metabolism of imipramine in human liver micro(cid:173)
`somes. Bioi Pharm Bull 16: 571-575, 1993.
`94. von Moltke LL, Greenblatt D], Cotreau-Bibbo MM, Duan
`SX, Harmatz ]S and Shader RI, Inhibition of desipramine
`hydroxylation in vitro by serotonin-reuptake-inhibitor anti(cid:173)
`depressants, and by quinidine and ketoconazole: A model
`system to predict drug interactions in vivo. ] Pharmacol Exp
`Ther 268: 1278-1283, 1994.
`95. von Moltke LL, Greenblatt D], Court MH, Duan SX,
`Harmatz ]S and Shader RI, Inhibition of alprazolam and
`desipramine hydroxylation in vitro by paroxetine and fluvox(cid:173)
`amine: Comparison with other selective serotonin reuptake
`inhibitor antidepressants. ] Clin Psychopharmacol 15: 125-
`131, 1995.
`96. Shader RI, von Moltke LL, Schmider ], Harmatz ]S and
`Greenblatt D], The clinician and drug interactions-An
`update. ] Clin Psychopharmacal16: 197-201, 1996.
`97. Bergstrom RF, Peyton AL and Lemberger L, Quantification
`and mechanism of the fluoxetine and tricyclic antidepres(cid:173)
`sant interaction. Clin Pharmacal Ther 51: 239-248, 1992.
`98. Preskorn SH, Alderman], Chung M, Harrison W, Messig M
`and Harris S, Pharmacokinetics of desipramine coadminis(cid:173)
`tered with sertraline or fluoxetine. ] Clin Psychopharmacol
`14: 90-97, 1994.
`99. Alderman ], Preskorn SH, Greenblatt D], Harrison W,
`Penenberg D, Allison] and Chung M, Desipramine phar(cid:173)
`macokinetics when co administered with paroxetine or ser-
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2053, Page 9
`
`

`

`1

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