throbber
ORIINAL RESARCH ARTILE
`
`
`Clin Phormocokinei 2007: 46 (8): 681—696
`03126963/07/0008—0681 /3411 95/0
`© 2007 Acts Data Information BV. All rights resen/ed
`
`General Framework for the
`
`Quantitative Prediction of
`CYP3A4-Mediated Oral Drug
`Interactions Based on the AUC
`Increase by Coadministration
`of Standard Drugs
`
`Yoshiyuki Chi/10,1 Akihiro Hisozka1 and Hiroshi Suzukz‘lr2
`
`1 Department of Pharmacy, University of Tokyo Hospital Faculty of Medicine, University of
`Tokyo, Tokyo, Japan
`2 Center for Advanced Medical Engineering and Informatics, Osaka University, Osaka, Japan
`
`
`Abstract
`
`Background: Cytochrome P450 (CYP) 3A4 is the most prevalent metabolising
`enzyme in the human liver and is also a target for various drug interactions of
`significant clinical concern. Even though there are numerous reports regarding
`drug interactions involving CYP3A4, it is far from easy to estimate all potential
`interactions, since too many drugs are metabolised by CYP3A4. For this reason, a
`comprehensive framework for the prediction of CYP3A4-mediated drug interac-
`tions would be of considerable clinical importance.
`Objective: The objective of this study was to provide a robust and practical
`method for the prediction of drug interactions mediated by CYP3A4 using
`minimal in vivo information from drug-interaction studies, which are often carried
`out early in the course of drug development.
`Data sources: The analysis was based on 113 drug-interaction studies reported in
`78 published articles over the period 1983—2006. The articles were used if they
`contained sufficient information about drug interactions. Information on drug
`names, doses and the magnitude of the increase in the area under the concentra—
`tion-time curve (AUC) were collected.
`Methods: The ratio of the contribution of CYP3A4 to oral clearance (CRCYP3A4)
`was calculated for 14 substrates (midazolam, alprazolam, buspirone, cerivas-
`tatin, atorvastatin, ciclosporin, felodipine,
`lovastatin, nifedipine, nisoldipine,
`simvastatin, triazolam, zolpidem and telithromycin) based on AUC increases
`observed in interaction studies with itraconazole or ketoconazole. Similarly, the
`time-averaged apparent inhibition ratio of CYP3A4 (IRCYP3A4) was calculated
`for 18 inhibitors
`(ketoconazole, von'conazole,
`itraconazole,
`telithromycin,
`clarithromycin, saquinavir, nefazodone, erythromycin, diltiazem, fluconazole,
`verapamil, cimetidinc, ranitidine, roxithromycin, fluvoxamine, azithromycin, gat—
`ifloxacin and fluoxetine) primarily based on AUC increases observed in drug-
`interaction studies with midazolam. The increases in the AUC of a substrate
`
`TEVA1 027
`
`me
`
`my
`
`oak.“«a-4
`
`
`
`ewvewww.«av
`
`
`myfiffifi,MWJ®_W.WWJ{§MMW¢A¢§Mn-4,-,M43».
`
`
`.1w..raft?“
`3%”A“Meta.
`
`tvflm'éfi‘~-w¢2a%rim
`
`1
`
`TEVA1027
`
`

`

`682QWM
`
`associated with coadministration of an inhibitor were estimated using the equation
`1/(] — CRCYP3A4 . IRCYP3A4), based on pharmacokinetic considerations.
`Results: The proposed method enabled predictions of the AUC increase by
`interactions with any combination of these substrates and inhibitors (total 251
`matches). In order to validate the reliability of the method, the AUC increases in
`60 additional studies were analysed. The method successfully predicted AUC
`increases within 67~150% of the observed increase for 50 studies (83%) and
`within 50—200% for 57 studies (95%). Midazolam is the most reliable standard
`substrate for evaluation of the in viva inhibition of CYP3A4. The present analysis
`suggests that simvastatin, lovastatin and buspirone can be used as alternatives. To
`evaluate the in viva contribution of CYP3A4, ketoconazole or itraconazole is the
`selective inhibitor of choice.
`
`Conclusion: This method is applicable to (i) prioritise clinical trials for investi-
`gating drug interactions during the course of drug development and (ii) predict the
`clinical significance of unknown drug interactions. If a drug—interaction study is
`carefully designed using appropriate standard drugs, significant
`interactions
`involving CYP3A4 will not be missed. In addition,
`the extent of CYP3A4-
`mediated interactions between many other drugs can be predicted using the
`current method.
`
`Background
`
`Cytochrome P450 (CYP) 3A4 is the most preva—
`lent CYP enzyme in the human liver. It accounts for
`=30% of the total CYP enzymes in hepatic micro—
`somes and is involved in the metabolism of >50% of
`
`the drugs currently on the market?!” CYP3A4 is
`also the target enzyme for a number of drug interac—
`tions of significant clinical concern. Drug interac—
`tions are one of the major sources of adverse events,
`and some have actually led to drug withdrawals in
`the past.[3'5] Even though there are numerous reports
`of CYP3A4 drug interactions, it is far from easy to
`estimate all potential interactions, since too many
`drugs are metabolised by CYP3A4. For this reason,
`a comprehensive framework for the prediction of
`drug interactions would be of significant clinical
`importance. In addition, pharmaceutical companies
`are encouraged to carry out many in viva drug-
`interaction studies during the drug development pro—
`cess, and the cost of these studies is increasing.
`Consequently, it is important to prioritise significant
`drug interactions to be confirmed as early as possi—
`ble during the course of development. A reliable
`method for the prediction of CYP3A4 drug interac-
`tions would be advantageous in such circumstances.
`A great deal of effort has already been devoted to
`establish a method for the accurate prediction of in
`
`viva drug interactions using in vitra experimental
`data.[6““] These predictions in principle rely on the
`[I]/Ki ratio, i.e. a ratio of the unbound concentration
`of the inhibitor at the interaction site to the in vitro
`inhibition constant. The results of these studies have
`
`increased our understanding of the mechanisms of
`drug interactions. Nowadays, both human liver
`specimens and expressed human CYP enzymes are
`commercially available, and it
`is not difficult to
`determine a profile of metabolic drug interactions in
`vitra. However, the proper interpretation and quanti-
`tative extrapolation of in vitra data to in viva situa-
`tions require a detailed understanding of the overall
`pharmacokinetics of the drugs involved. Considera-
`tion should be given to the site of interaction, the
`time—courses of the unbound drug concentration at
`the site,
`the effects of drug transporters on the
`pharmacokinetics, and the possible contribution of
`metabolites to the interactionm]
`
`the quantitative prediction of drug
`Moreover,
`interactions is difficult for the following reasons:
`
`0
`
`Intestinal CYP3A4 plays a significant role in the
`first—pass metabolism of orally administered
`drugs. For example, several human in viva stud-
`ies have shown that midazolam,
`felodipine,
`ciclosporin
`and
`buspirone
`are
`extensively
`metabolised in the intestinelm Although Caco—Z
`
`© 2007 Adis Data Information BV, All rights reserved.
`
`Clin Pharmacokinei 2007: 46 (8)
`
`2
`
`

`

`P
`
`683
`
`
`cells are used in predicting the extent of intestinal
`absorption, it is difficult to predict the intestinal
`metabolism because of the very low expression
`of CYP3A4 in this cell linen“ It is also known
`
`that CYP3A4 does not distribute uniformly along
`the length of the intestine — it is expressed more
`in the jejunum than in the ileumils] In addition,
`quantitative prediction of oral bioavailability is
`difficult because of the synergistic role of
`CYP3A4 and efflux transporters, such as mul—
`tidrug resistance-l (MDRI), in reducing the in—
`testinal
`absorption of
`substrate
`drugs.[16'20]
`MDRl is expressed more in the ileum than in the
`jejunum?” Although the intestine is also consid-
`ered an important site of drug interactions, the
`extent of intestinal metabolism is unpredictable
`in many cases.
`0 CYP3A4 recognises a wide range of substrates,
`and some structural flexibility has been suggest-
`ed at the substrate recognition siteml Conse—
`quently,
`the enzyme kinetics of CYP3A4 are
`sometimes complicated. Indeed, simple competi—
`tive inhibition theory has often failed to explain
`interactions via CYP3A4.[23]
`
`0 A series of CYP3A4 substrates apparently act as
`mechanism-based inhibitors which covalently
`bind to the enzyme. For these drugs, the recovery
`of CYP3A4 activity depends on regeneration of
`the enzyme at the target site. For this reason,
`predictions of the mechanism—based interactions
`from in vitra data require more complicated ki—
`netic models compared with reversible inhibi-
`tors.[7,8,l3,24-26]
`
`Considering these complex factors, it is reasona—
`ble to conclude that, by using only in vitra experi-
`mental data, precise prediction of in viva drug inter—
`actions is not easy for the variety of drugs that are
`metabolised by CYP3A4. One of the practical meth—
`ods to overcome this problem is to use in viva
`information on interaction data of probe drugs of
`CYP3A4. This approach would enable the predic—
`tion of various drug interactions from results of a
`small number of drug-interaction studies carried out
`early in the course of drug development.
`The objective of the present study was to con—
`struct a framework for the prediction of various drug
`interactions mediated by CYP3A4 using minimum
`in viva information on drug interactions. We select—
`
`ed midazolam as a standard substrate and ketocona—
`zole or itraconazole as a standard inhibitor. We
`
`aimed to keep the method as simple as possible from
`a practical viewpoint while, at the same time,
`re—
`maining theoretically accurate.
`
`Methods
`
`The analysis is based on 113 in viva studies
`reported in 78 articles published over the period
`1983—2006 (table I). Based on information from a
`comprehensive review,[9] we added some new data
`from the literature. Some substrates and inhibitors
`
`information be-
`were removed from the original
`cause of the small contribution or low selectivity for
`CYP3A4. Studies were used if the report included
`information on the dosage regimen and the increase
`in the area under the concentration-time curve
`
`(AUC). A survey of a series of articles revealed that,
`in general, inhibitor drugs were administered con-
`secutively for more than several days to attain
`steady—state conditions, and substrate drugs were
`given as a single—dose administration.
`The oral clearance (CLorai) of drugs is given by
`equation 1, where CLtot, CLH, CLR, Fa, PG and FH
`are the total body clearance, hepatic clearance, renal
`clearance, absorption ratio, and gastrointestinal and
`hepatic availabilities, respectively. The reason why
`Fa is located at the left side in equation 1 is that the
`current analysis (right—side terms) should focus on
`events after absorption.
`
`CLtot _ CLH+ CLR
`CLoral ' Fa: m -
`FG . FH
`
`(Eq- 1)
`Assuming the well—stirred organ modellmsl equa-
`tion 1 can be converted to equation 2 where fu and
`CLim(H) are the unbound fraction of the drug in the
`blood and the intrinsic clearance of the liver, respec—
`tively. This equation represents a general relation-
`ship between the oral clearance and the intrinsic
`clearance of the liver.
`
`CLoral ° Fa =
`
`fu’ €14an)
`F6
`
`
`+ CLR
`F6 ° FH
`
`(1301.2)
`In the present study, two simplifications were
`made with respect to equation 2. First, we assumed
`that CLR can be ignored, which is frequently the
`case for lipophilic CYP3A4 substrates. Second, we
`
`© 2007 Adis Data Information BV. All rights reserved.
`
`Clin Photmocokinet 2007: 46 (8)
`
`3
`
`

`

`684
`
`Ohno et al.
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`0:0.v60::00:0_.:.02:00£0:0500.000:v<mm>om:_>_o>:_00630:0=00:0E_.m:.6_00_:__o00:000.fir:000:0..000:0:000:6.0:o_fim=00>:_c0050:00_:0:ozm_0m._0.00...
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`._oN_E4m:5:002“:20:2>0:d“.06:000:0020.2
`
`
`
`a.0000R_N<
`
`
`
`
`
`05020552:.
`
`
`
`NV3,300.0000005“00.00«0.00002.0
`
`00.005a.ona?0v2};0::5
`
`50:3.0000%<40
`
`N0.5F0.00600000.00:000mm«0>00.
`
`8000000N0“.
`
`KE.0000%xi
`
`.000:mu3:
`
`.320
`
`
`
`
`
`.0000.00000.00.00.00000000.00,5.?.00Ex
`
`00.00S:02
`
`
`
`N:8.500.00.00z<m
`
`5,?.9“0:ES
`
`00.00.m:
`
`mi:09:9
`
`.2:02x0:
`
`“we90
`
`
`
`
`
`00.0000$00.00“00.030.00.550.03:.0500E0:0:«E
`
`
`
`.0050::E00050:“0030:00::050002059:00:0:02,0:028:030089000:
`
`
`
`
`
`
`
`
`
`000062002:0:“0:0Av<0m>omov080.806.059v<mn_>oD552:05:000%.:00:0:0:0000.:2002.000:0:02,:0082025000::0:000:0005:00::00:05.m
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`.000:0:0_0__m>05020:20058::0000:02.0.05005..?<mn_>om_v0:0::oEnEEE90000
`
`
`
`
`
`
`
`
`
`
`
`
`
`MEmNSEUn.=o”:_0>E2£:0_0n<40”0:_U:0E_0u5:05500206n0.052000250ummo03:00::Nmam”:_0>E0:£_N0n_N<6:300:20n0.5.”E53990un3<
`
`
`
`
`
`006305302060000.320:00020.:mE.0._”00000.00:02:00020:.0
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`”$305023n._.m_v_”0650:0008:um...—EUmonEmn._.<0”0:300:02:nx._n_”0:_me0>:=n:4...”05:60.2M4m".”98080::nNon.”:_0>E2£>._0n>mm”:_0>Eo._£__9n.._m._.”:_§00>E_0u5.5u_>0:_:000uO<m”:_0>Eo:£_x2u08$”0:_.:0:nz<m050.20g:nmi”05060::n”=2”0:003:20:u"52”E3803:u0:2M_§00>o_u>o._
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`.E00_0_0Nu._ON”0.0Nm:00_:o>nmo>25.0003;nmm>”EEONmEHE...
`
`© 2007 Adis Data Informofion BV. All rights reserved.
`
`Clin Phormocokine‘r 2007,- 46 (8)
`
`4
`
`
`
`

`

`General Prediction of CYP3A4 Drug Interactions
`
`
`
`
`
`
`assumed that FG can be regarded as 1.0 hypotheti-
`cally. This simplification does not necessarily mean
`that the gastric metabolism is insignificant. Rather,
`it means that the gastric metabolism is assumed to
`occur in proportion to the hepatic metabolism as if it
`is a part of the liver. This may allow some overesti-
`mation of CLim(H) as a consequence. Significant
`gastric first-pass effects are well established facts
`for some CYP3A4 substrates; FG values of midazo—
`lam, felodipine, ciclosporin and buspirone are re-
`ported to be 0.57, 0.45, 0.39 and 0.21, respective—
`ly.[13’16] In the future, it may be advantageous to
`distinguish between gastric and hepatic metabolism
`by CYP3A4. At present, however, we have no alter-
`native but to accept this simplification, since we do
`not know the extent of the in viva gastric metabolism
`for all of the drugs analysed in the present study. In
`this connection, it is worth noting that incorporation
`of gut wall CYP3A4 inhibition did not result in a
`general improvement in drug—interaction predictions
`in a recent reportiml Overall, equation 2 becomes
`equation 3 with these simplifications.
`
`CLoral ' Fa 5 1cu ' CLint(H)
`
`(Eq- 3)
`
`We then considered a relationship between the
`intrinsic clearance and metabolic drug interactions
`under an assumption of the rapid equilibrium of the
`drug concentration between the blood and the
`hepatocyte.
`It
`is often the case that a drug is
`metabolised by more than two pathways. In equa-
`tion 4, we assume two intrinsic metabolic clear—
`
`ances, CLint(Cyp3A4) and CLint(others), which re-
`present the metabolism mediated by CYP3A4 and
`the sum of other metabolic pathways, respective—
`1y.[11,1o7]
`
`CLint(H) = CLint(CYP3A4) + CLint(others)
`
`(Eq. 4)
`
`In the following equations, asterisks denote pa-
`rameters altered by a drug interaction. When the
`metabolism of CYP3A4 is inhibited by a drug inter—
`action, the altered clearance is given by equation 5.
`
`Equation 5 is applicable to both competitive and
`non—competitive inhibitions, since the drug concen—
`tration in vivo is usually much lower than the
`Michaelis-Menten constant (Km) value.
`Here, we define the ratio of the contribution of
`
`CYP3A4 to oral clearance (CRCYP3A4) by equation
`6:
`
`CRCYP3A4 =
`
`CLorai ‘ CLoral(—CYP3A4)
`CL 1Ora
`
`(Eq. 6)
`
`is an altered in viva oral
`where CLoraK—CYP3A4)
`clearance when CLim(cyp3A4) is blocked complete-
`1y. CLoraK—CYP3A4) is given by equation 7, based on
`equations 3 and 4.
`
`CLora1(—CYP3A4) ' Fa = fu ° (€14er
`
`CLint(CYP3A4))
`
`(Eq. 7)
`
`From equations 6 and 7, CRCYP3A4 is given by
`equation 8, which indicates that the ratio of the in
`viva contribution of CYP3A4 to oral clearance has
`
`the same value as the fraction metabolised by
`CYP3A4 to inhibition (fm(CYP3A4), Which is deter—
`mined by examining the effect of CYP3A4 selective
`inhibitors/antibodies on drug metabolism by human
`liver microsomesmg’mgl when the urinary excretion
`is very low and rapid equilibrium is achieved in the
`liver. These relationships have been widely used for
`prediction of in viva clearances in the presence of
`drug interactions or CYP enzyme polymorphisms,
`as reported by other groups.[“’1071
`
`CRCYP3A4=
`
`CLint(CYP3A4)
`in
`CL- [(H)
`
`(Eq. 8)
`
`Equation 5 can be converted to equation 9 using
`equations 6 and 8.
`C
`I CLin
`CLiiHKH) :W + (1 _ CRCYP3A4) . CmeH)
`1 + E
`
`= (1 — CRCYP3A4 '
`
`
`I
`[Il[+]Ki
`
`) CLint(H)
`
`(EQ- 9)
`
`CL*int(CYP3A4) =
`
`CLint(CYP3A4)
`1+%]i
`
`To estimate AUC from equation 9, the equation
`needs to be integrated with time. For this pur-
`pose,
`the time—averaged apparent inhibition ratio
`(IRCYP3A4) is defined by equation 10:
`
`(Eq- 5)
`
`© 2007 Adis Data Information BV. All rights resen/ed.
`
`Clin Phormocokine‘r 2007; 46 (8)
`
`5
`
`

`

`Ohm) et al.
`686
`
`’L “mew
`
`
`
`
`Happl
`IRCYP3A4 =W
`
`(Eq. 10)
`
`where [Iapp] is the time-averaged apparent unbound
`concentration of the inhibitor in the liver. The in—
`
`crease in the ratio of the AUC caused by a drug
`interaction (equation 10) is derived from equations 9
`and 10. We assumed here that the value of IRCYP3A4
`
`for an inhibitor is the same for any substrate.
`
`Equation ll indicates that an AUC increase by a
`drug interaction between any oral drug via CYP3A4
`can be predicted if the CRCYP3A4 of the substrate
`and the IRCYP3A4 of the inhibitor are available.
`
`AUC*ora1 : CLoral - Fa _ CLint(H)
`AUCma]
`CL*OmloFa
`CL*int(H)
`
`_
`—
`
`l
`
`l — CR
`
`[lapp]
`' —'—‘-"‘
`[lappl +Ki
`
`CYP3A4
`
`= ____l___
`1 — CRCYP3A4 ° IRCYP3A4
`
`(Eq. ll)
`It needs to be mentioned that the above theory
`may not be directly applicable to mechanism—based
`inhibitors. However, the final form of equation 11
`can be accepted even for mechanism—based inhibi—
`tors by regarding the IRCYP3A4 values as overall
`inhibition ratios of CYP3A4 at
`the equilibrium
`state between inactivation and generation of the
`metabolising enzyme. From this viewpoint, the defi—
`nition of IR by equation 10 is invalid for mecha—
`nism—based inhibitors.
`
`The values of CRCYP3A4 and IRCYP3A4 for vari—
`ous substrates and inhibitors were calculated se-
`
`quentially according to the following steps based on
`AUC increases in the 53 interaction studies, which
`are indicated in table I.
`
`0 We assumed that the CRCYP3A4 value of simvas—
`tatin is 1.0 since it has been reported that simvas-
`tatin is a selective substrate of CYP3A4” and a
`search of the literature showed that the plasma
`AUC of simvastatin tends to be increased most
`
`markedly following inhibition of CYP3A4. It
`was confirmed that a reduction of the CRCYP3A4
`value of simvastatin to 0.95 did not affect the
`
`overall outcomes of the present analysis.
`
`0 Once we assumed the CRCYP3A4 value for
`simvastatin, the IRCYP3A4 value of itraconazole,
`a typical inhibitor, was obtained based on equa-
`tion 11, using the result of a drug—interaction
`study involving simvastatin and itraconazole.
`o The CRCYP3A4 value of midazolam, a typical
`substrate, was calculated from studies with
`midazolam and itraconazole using the calculated
`IRCYP3A4 value of itraconazole with equation ll.
`An algebraic mean of the AUC increase was used
`for the calculation, whenever the results of multi—
`ple studies are reported for an interaction set of
`interest.
`
`0 The IRCYP3A4 values of the other inhibitors in-
`cluding ketoconazole, another typical inhibitor,
`were calculated from interaction studies between
`
`the inhibitor and midazolam, using the calculated
`CRCYP3A4 value of midazolam with equation ll.
`0 The CRCYP3A4 values of other substrates were
`calculated from interaction studies between the
`substrate and itraconazole or ketoconazole when—
`
`ever possible, using the calculated chyp3A4
`value of itraconazole or ketoconazole, respec-
`tively, with equation 11.
`e For nifedipine, no interaction study with itra—
`conazole or ketoconazole has been reported. Ac-
`cordingly, the CRCYP3A4 value of nifedipine was
`calculated from the study with nifedipine and
`diltiazem, using the calculated IRCYP3A4 value of
`diltiazem, with equation 11. Diltiazem was se—
`lected, since the AUC of nifedipine was most
`significantly increased by the administration of
`diltiazem.
`
`Results
`
`We surveyed 113 in viva drug—drug interaction
`studies published in 78 articles
`(table I). The
`CRCYP3A4 values of 14 substrates and the IRCYP3A4
`values of 18 inhibitors were calculated using equa—
`tion ll based on the results of 53 clinical studies
`
`(the estimation set), which are indicated in table I.
`As shown in table 11, high CRCYP3A4 values of
`>095 were calculated for simvastatin,
`lovastatin,
`
`buspirone and nisoldipine, 0.85—0.94 for triazo—
`lam, midazolam and felodipine, and 0.70—0.84
`for ciclosporin, nifedipine and alprazolam. High
`IRCYP3A4 values of >095 were estimated for
`
`© 2007 Adis Data information BV. All rights reserved.
`
`Clin Phormocokiner 2007; 46 (8)
`
`6
`
`

`

`
`W Wfide— e. aam am: 25;; 242, was”
`General Prediction of CYP3A4 Drug Interactions
`
`
`
`Table ll. Calculated ratios of the contribution of CYP3A4 to the oral
`clearance (CRCYP8A4) of substrates
`Substrate
`CRCYP3A4
`Simvastatin
`1.00
`Lovastatin
`1 .00
`Buspirone
`0.99
`Nisoldipine
`0.96
`Triazolam
`0.93
`Midazolam
`0.92
`Felodipine
`0.89
`Ciclosporin
`0.80
`Nifedipine
`0.78
`Alprazolam
`0.75
`Atorvastatin
`0.68
`Telithromycin
`0.49
`Zolpidem
`0.40
`
`Cerivastatin 0.18
`
`ketoconazole (daily dose 200—400mg), voriconazole
`(400mg) and itraconazole (100—200mg), 0.85—0.94
`for
`telithromycin
`(800mg),
`clarithromycin
`(SOO—lOOOmg),
`saquinavir
`(3600mg) and nefa-
`zodone (400mg), and 0.70—0.84 for erythromycin
`(lOOO—ZOOOmg), diltiazem (90—270mg),
`flucona-
`zole (200mg) and veraparnil (240—480mg).
`
`The current method enabled predictions of the
`AUC increase caused by drug-drug interactions of
`any combination of the substrates and inhibitors. In
`order
`to validate the suitability of the present
`method, the extent of the increase in the AUC by
`drug interaction was predicted for the remaining 60
`clinical studies (the validation set), which are indi—
`cated in table I, and the results were compared with
`the observed values (figure 1). This prediction was
`performed by substituting the values of CRCYP3A4
`and IRCYP3A4 shown in table II and table III, respec—
`tively, in equation 11. As shown in figure 1, with the
`current method we could predict the increase in the
`AUC within 67—150% of the observed AUC in—
`
`crease for 50 clinical studies (83%) and within
`50-200% for 57 clinical studies (95%).
`
`In the calculation of CRCYP3A4 and IRCYP3A4
`values, the algebraic mean of the increase in the
`AUC was used when more than one article was
`
`available for the same interaction set (table I). In
`. these cases, however, significant deviation was ob—
`served in the AUC increase between or among re-
`ports. For the analysis, we often combined the data
`of clinical studies with different doses of the inhibi~
`
`tor. Since the lower doses frequently gave more
`AUC increase of a substrate, it is possible that the
`deviation of the inhibitor dose may not largely affect
`the results, as far as the dose of the inhibitor is set
`within the therapeutic range. The extent of this
`deviation is shown in figure 2, which was prepared
`in the same style as figure 1. Each circle and vertical
`bar in figure 2 represents the mean + SD reported in
`each article of the estimation set in table I. If the SD
`
`values were not reported in articles listed in table I,
`the reported mean values are shown by squares. As
`shown by the dotted lines in figure 2, for most of the
`articles the increase in the AUC of substrate drugs
`caused by drug inhibition deviated by 67—150% of
`the algebraic mean values. The predictions within
`50—200% of the observed AUC increase in figure 1
`were regarded as successful, since the correspond-
`ing variation of the AUC in figure 2 (the estimation
`set) was within this range.
`The data shown in figure 1 and figure 2 were then
`reorganised to indicate the relationships between the
`IRCYP3A4 values and the increase in AUC of each
`substrate (figure 3). It was found that
`the AUC
`increased steeply as the IRCYP3A4 value increased
`for highly CYP3A4—dependent substrates, such as
`simvastatin, lovastatin and buspirone, whereas only
`minimal increases were observed for poor CYP3A4
`substrates, such as zolpidem and cerivastatin (figure
`
`Table III. Calculated ratios of the time-averaged apparent inhibition
`ratio of CYP3A4 (IRCYP3A4) for inhibitors
`Inhibitor
`Daily dose (mg)
`Ketoconazole
`200—400
`Voriconazole
`400
`ltraconazole
`100—200
`Telithromycin
`800
`Clarithromycin
`500—1000
`Saquinavir
`3600
`Neiazodone
`400
`Erythromycin
`1000—2000
`Diltiazem
`90—270
`Fluconazoie
`200
`Verapamil
`240—480
`Cimetidine
`800—1200
`Ranitidine
`300—600
`Roxithromycin
`300
`Fluvoxamine
`100—200
`Azithromycin
`250—500
`Gatifloxacin
`400
`Fluoxetine
`20—60
`
`chvp3A4
`1 .00
`0.98
`0.95
`0.91
`0.88
`0.88
`0.85
`0.82
`0.80
`0.79
`0.71
`0.44
`0.37
`0.35
`0.30
`0.1 1
`0.08
`0.00
`
`© 2007 Adis Data lnformotion BV. All rights reserved.
`
`Ciin Phormocokine’r 2007; 46 (8)
`
`7
`
`

`

`Ohm) et al.
`688
`
`
`100
`
`
`
`
`
`ObservedAUCincrease(fold)
`
`0.5
`
`Calculated AUC increase (fold)
`
`Fig. 1. Relationship between the observed and calculated increase in the area under the concentration-time curve (AUC) by drug
`interactions. Using the ratio of the contribution of cytochrome P450 (CYP) 3A4 to oral clearance (CRCYP3A4) and the time-averaged
`apparent inhibition ratio of the CYP3A4 (IRCYP3A4) values shown in table it and table III, respectively, the increase in the AUC of substrate
`drugs by drug interactions reported in 60 clinical studies (the validation set; table I) was predicted with equation 11. Each circle and vertical
`bar represents the mean + SD values of subjects reported in each article. A dashed bar represents the range. Where the SD values or the
`ranges were not reported in the article, the reported mean value is shown by a square. The solid and dotted lines represent 50—200% and
`67—150% ranges, respectively, of the calculated increase.
`
`3). In the same manner, potent inhibitors, such as
`azole antifungals, increased the blood levels of a
`number of CYP3A4 substrates markedly, whereas
`no or only very minor increases were observed for
`weak inhibitors, such as azithromycin, gatifloxacin
`and fluoxetine (figure 4).
`
`Finally, the data were reorganised to show that
`the increase in the AUC in 251 kinds of drug interac-
`tions between 14 substrate diugs and 18 inhibitors
`could be predicted (figure 5; note that telithromycin
`is included with both the substrates and the in—
`
`hibitors). The nomogram in figure 5 indicates that a
`
`very marked increase in the AUC is anticipated
`when substrate drugs with high CRCYP3A4 values
`are administered with potent inhibitors with high
`IRCYP3A4 values.
`
`Discussion
`
`CYP3A4 is the most important drug-metabolis—
`ing enzyme, which preferentially oxidises relatively
`large, lipophilic, neutral to basic molecules. There-
`fore, CYP3A4 is recognised as a key enzyme that
`determines the clearance of various drugs and, in
`some cases, has a major effect on their safety and
`
`© 2007 Adis Data Information BV. All rights reserved.
`
`Cltn Phormocokinet 2007; 46 (8)
`
`8
`
`

`

`General Prediction of CYP3A4 Drug Interactions
`
`
`689
`
`
`efficacy. Although no major polymorphism in the
`CYP3A4 gene has been identified, marked inter—
`individual differences have been reported in the
`activity of CYP3A4.[1101 One possible reason for
`such differences in the activity is that CYP3A4 is
`inducible by various diets and drugs,
`such as
`rifampicin (rifampin) and carbamazepine, via the
`mechanism mediated by pregnane X receptor}1 ”'113]
`Furthermore,
`CYP3A4
`is
`the
`predominant
`metabolising enzyme not only in the liver but also in
`the intestine. It has been reported that intestinal
`metabolism is the major factor determining the
`bioavailability of some drugsimdlim] However, as
`100
`
`far as we know, nobody has succeeded in predicting
`the extent of the first—pass effect on metabolism by
`intestinal CYP3A4 from in vitro data. Although
`there are some established methods to determine the
`
`activity of CYP3A4 in viva, including evaluation of
`the metabolic ratio of selective substrates (midazo—
`
`lam, testosterone and cortisol) and the erythromycin
`breathe test, it has been reported that these methods
`
`do not offer consistent results,m7] possibly due to
`differences in the organ of metabolism (liver or
`
`intestine) and/or the presence of multiple recogni-
`tion sites in the CYP3A4 moleculemsi
`
`50*
`
`_. 0
`
`U'l
`
`
`
`0.5
`
`
`
`
`ObservedAUCincrease(fold)
`
`
`
`,.
`
`Calculated AUC increase (fold)
`
`p
`
`Fig. 2. Relationship between the observed and calculated increase in the area under the concentration-time curve (AUC) by drug
`interactions. This figure was prepared in the same style as figure 1 for the purpose of demonstrating the deviation of AUC values among 53
`clinical studies (the estimation set; table I), the mean values of which were used to determine the CRCYP3A4 and IRcyp3A4 values. Each
`~circle and vertical bar represents the mean + SD values of subjects reported in each article. A dashed bar represents the range. Where the
`SD values or the ranges were not reported in the article, the reported mean value is shown by a square. The solid and dotted lines represent
`50—200% and 67—150% ranges, respectively, of the calculated increase.
`
`© 2007 Adis Data Information BV. All rights reserved.
`
`Clin Pharmacokinet 2007; 46 (8)
`
`9
`
`

`

`
`
`Ohno et al.
`“WWWEWWWMWM”€7MWWWMWSam
`
`100 I Simvastatin
`
`100 ' Lovastatin
`
`100.
`
`Buspirone
`
`100-
`
`Nisordipine
`
`10 '
`
`1
`
`1 r—fifi—T—rfi
`0 0.2 0.4 0.6 0.8 1.0
`
`1o —
`
`10 *
`
`l
`1- 1-
`
`0 0.2 0.4 0.6 0.8 1.0
`0 0.2 0.4 0.6 0.8 1.0
`O 0.2 0.4 0.6 0.8 1.0
`
`
`
`100 -
`
`1o -
`
`Triazolam
`
`I
`
`4
`E,
`r——~—.——n'—i—~—r—u
`I;
`0 0.2 0.4 0.6 0.8 1.0
`g
`'
`‘5
`5 100 Nifedipine
`10 -
`
`1
`
`3<
`
`1
`
`/r‘I/§/
`-
`O 0.2 0.4 0.6 0.8 1.0
`
`100 ' Zolpidem
`
`10 '
`
`10° ' Atorvastatin
`
`10 -
`
`100 _ Telithromycin
`
`10 '
`
`~
`
`1
`
`///‘§“
`0 0.2 0.4 0.6 0.8 1.0
`
`‘
`—
`.3 .
`0 0.2 0.4 0.6 0.8 1.0
`
`
`1 l
`.
`I M 1 Fifi—’4
`O 0.2 0.4 0.6 0.8 1.0
`0 0.2 0.4 0.6 0.8 1.0
`
`Fig. 3. Increase in the area under the concentration—time curve (AUC) reorganised for each substrate drug as a function of the inhibition
`ratio (IR) of the inhibitors. The data shown in figure 1 and figure 2 were reorganised to show the increase in the AUC of each substrate drug
`as a function of the time-averaged apparent inhibition ratio of CYP3A4 (IRCYP3A4) values of the inhibitors. The closed and open symbols
`represent the dataset shown in figure 1 and figure 2, respectively. See figure 1 and figure 2 legends for details.
`
`I RCYP3A4
`
`Despite these issues regarding the in viva evalua—
`tion of CYP3A4 activity, the current rather simple
`method gave satisfactory predictions in most cases.
`The following‘issues may contribute to this success.
`First, uncertain factors were avoided since the cur—
`rent method relies primarily on an overall in viva
`evaluation. For example, the present method satis-
`factorily predicted drug interactions with mecha-
`nism-based
`inhibitors
`such
`as
`azithromycin,
`clarithromycin, diltiazem, erythromycin and rox—
`ithromycin (figure 4), which frequently exhibit com-
`plicated kinetics. Accurate predictions have been
`achieved recently from in vitro data for mechanism-
`based inhibitors by sophisticated analysis. For the
`successful prediction, it has been reported that eval-
`uation of the unbound fraction of the drug in the
`
`incubation medium is importantm’m] Moreover,
`the analysis
`requires
`a
`turnover
`rate of
`the
`metabolising enzyme and a rate constant for the
`irreversible reaction, both of which are not easy to
`estimate from in vitro experiments.
`
`Second, we used simvastatin as a selective sub-
`strate and ketoconazole and itraconazole as selective
`
`inhibitors of CYP3A4, although these drugs are not
`absolutely specific for CYP3A4. For example,
`al—
`though we assumed that the CRCYP3A4 value of
`simvastatin is 1.0, this drug is also metabolised by
`CYP2C8 to a minor extent.[1°5] Ketoconazole is a
`well known selective inhibitor of CYP3A4, but this
`drug also inhibits the activities of CYP2C8,[120]
`CYP2C9ml] and MDR1,[m] which may also affect
`the disposition of substrate drugs analysed in the
`
`© 2007 Adis Data Information BV. All rights reserved.
`
`1 0
`
`Clin Phormocokinet 2007; 4(2 (8)
`
`
`
`
`
`
`
`1001
`
`Midazolam
`
`.13i
`
`1o —
`
`100 -
`
`Felodipine
`
`10'
`
`100 -
`
`Ciclosporin
`
`10
`
`1
`
`0 0.2 0.4 0.6 0.8 1.0
`
`O 0.2 0.4 0.6 0.8 1.0
`
`:/rf
`/1
`
`.
`.
`.
`.
`.
`1 s
`0 0.2 0.4 0.6 0.8 1.0
`-i
`100 Alprazolam
`1o .
`
`1
`
`i
`‘
`.
`0 0.2 0.4 0.6 0.8 1.0
`
`1
`
`100 ' Cerivastatin
`
`10 ‘
`
`
`
`10
`
`

`

`
`i... $9M . 427A» ”m laws/W a“
`. 4A
`General Prediction of CYP3A4 Drug Interactions
`
`691
`
`
`present study. Despite these defects, the success in
`the prediction of drug interactions with the present
`method (figure
`1
`and figure 2)
`suggests
`that
`CYP3A4 plays a crucial role in most of the drug
`interactions analysed in the present study.
`
`A number of probe drugs have been used to
`study the activity of CYP3A4, including midazo—
`lam, nifedipine, simvastatin and erythromycinim]
`Among them,
`it
`is generally recognised that
`the most reliable probe drug is midazolam for
`CYP3A4.[

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