throbber

`

`A. David Rodrigues
`Bristol Myers Squibb Co.
`P.O.Box 4000
`Plinceton
`NJ 08543
`USA
`david.rodrigues @bms.com
`
`Editors
`K. Sandy Pang
`Leslie Dan Faculty of Pharmacy
`University of Toronto
`144 College Street
`Toronto ON M5S 3M2
`Canada
`ks.pang@utoronto.ca
`
`Rairnund M. Peter
`AstraZeneca UK
`Alderley Park
`Macclesfield, Cheshire
`United Kingdom SKlO 4TF
`raimund. peter@astrazeneca.com
`
`ISBN 978-1-4419-0839-1
`DOl 10.1 007/978-1-4419-0840-7
`Springer New York Dordrecht Heidelberg London
`
`e-ISBN 978-1-4419-0840-7
`
`Library of Congress Control Number: 2009938736
`
`© American Association of Pharmaceutical Scientists 20 I 0
`All rights reserved. This work may not be translated or copied in whole or in part without the written
`permission of the publisher (Springer Science+Business Media, LLC, 233 Spring Street, New York,
`NY 10013, USA), except for blief excerpts in cormection with reviews or scholarly analysis. Use in
`connection with any foml of information storage and retrieval, electronic adaptation, computer software,
`or by similar or dissimilar methodology now known or hereafter developed is forbidden.
`The use in this publication of trade names, trademarks, service marks, and similar terms, even if they are
`not identified as such, is not to be taken as an expression of opinion as to whether or not they are subject
`to proprietary rights.
`While the advice and information in this book are believed to be true and accurate at the date of going
`to press, neither the authors nor the editors nor the publisher can accept any legal responsibility for any
`elTors or omissions that may be made. The publisher makes no wananty, express or implied, with respect
`to the material contained herein.
`
`Printed on acid-free paper
`
`Springer is part of Springer Science+Business Media (www.springer.com)
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2049, Page 2
`
`

`

`

`

`626
`
`D.1. Greenblatt and L.L. von Moltke
`
`1999; Greenblatt et aI., 2008). At the same time, in vitro techniques for studying
`human drug metabolism became increasingly developed and refined, including pre(cid:173)
`dictive models for in vitro-in vivo scaling, and the availability of heterologously
`expressed individual human CYPs. At a clinical level, polypharmacy was becoming
`increasingly prevalent, as the population aged, the number of patients with multi(cid:173)
`ple illnesses increased, and our capacity to provide pharmacologic treatments for
`serious disorders became more and more effective. Some newly introduced classes
`of medications - such as the azole antifungal agents and the selective serotonin
`reuptake inhibitor (SSRI) antidepressants - offered unique therapeutic options, but
`also had the secondary property of inhibiting certain human CYPs, thereby ele(cid:173)
`vating the risk of DDls (Greenblatt et aI., 1999; Hemeryck and Belpaire, 2002;
`Venkatakrishnan et aI., 2000). A dramatic and widely publicized event was the inter(cid:173)
`action of the nonsedating antihistamine terfenadine with potent CYP3A inhibitors
`such as ketoconazole and erythromycin (Honig et aI., 1993b; Honig et aI., 1992;
`Honig et aI., 1994; Honig et aI., 1993a). Under usual circumstances, terfenadine
`itself served only as a prodrug, being essentially completely transformed via hepatic
`and enteric CYP3A into fexofenadine, which was the entity having antihistaminic
`properties. Although terfenadine had effects on the cardiac QTc interval (Rampe et
`aI., 1993; Crumb et aI., 1995), this was of minimal concern since intact terfenadine
`does not ordinarily reach the systemic circulation. However, during co-treatment
`with CYP3A inhibitors, conversion of terfenadine to fexofenadine is blocked, and
`potentially hazardous levels of the parent drug reach the circulation (Honig et aI.,
`
`60
`
`..
`
`......
`
`50
`
`en z
`o
`~ o 40
`::i
`III
`::::>
`ll.
`is 30
`C
`LL o
`a: 20
`w
`III
`:2
`::::>
`z
`
`10
`
`1990 1992 1994 1996 1998 2000 2002 2004 2006 2008 2010
`YEAR
`
`Fig.24.1 Number of articles indexed as DDI studies published per year in the Journal of Clinical
`Pharmacology, 1990-2008
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2049, Page 4
`
`

`

`24 Clinical Studies of Drug-Drug Interactions: Design and Interpretation
`
`627
`
`1994; von Moltke et ai., 1994b). A few cases of serious and even fatal cardiac
`arrhythmias were reported as a consequence (Monahan et aI., 1990; Woosley et al.,
`1993). The "terfenadine affair" led to an acutely increased awareness of the poten(cid:173)
`tial importance of DDIs. Terfenadine was withdrawn from clinical practice, and a
`number of regulatory reforms increased the requirements for DDI assessments as
`a component of drug development. The overall shift in focus of the scientific and
`drug development community is clearly evident from the prevalence of DDI studies
`among scientific publications (Fig. 24.1).
`
`24.2 Epidemiology of Drug-Drug Interactions
`
`Given the prevalence of polypharmacy in contemporary clinical practice, the num(cid:173)
`ber of possible DDIs can become very large. If an individual patient is taking 11
`drugs concurrently, the number of pairwise combinations of these two drugs can be
`calculated as follows:
`
`n1
`(n - 2)!2!
`
`(24.1)
`
`The larger the value of 17, the greater the number of different drug combi(cid:173)
`nation pairs, and potential pairwise DDIs (Table 24.1). A patient with diabetes,
`hypertension, ischemic heart disease, and depression might well be taking 10
`drugs concurrently, in which case the number of possible drug interactions is 45.
`Considering this large "denominator" of possibilities, the number of clinically
`important DDIs encountered in contemporary therapeutics actually is relatively
`small.
`
`Table 24.1 Relation of
`number of drugs concunently
`administered to the number of
`possible pairwise drug-drug
`interactions
`
`Number of drugs
`
`Possible pairwise drug interactions
`
`2
`3
`4
`5
`6
`7
`8
`9
`10
`11
`12
`
`1
`3
`6
`10
`15
`21
`28
`36
`45
`5S
`66
`
`The outcome options following concurrent administration of two drugs can be
`constructed based on a probability hierarchy (Fig. 24.2). The most probable outcome
`is that the two drugs act independently, with no evidence of any interaction. Less
`probable is a DDI which can be demonstrated in a controlled laboratory setting,
`but is not detectable in clinical practice either because the magnitude of the change
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2049, Page 5
`
`

`

`

`

`24 Clinical Studies of Drug-Drug Interactions: Design and Interpretation
`
`629
`
`with CYP2D6 substrate drugs such as desipramine (Hemeryck and Belpaire, 2002;
`von Moltke et al., 1994a; von Moltke et al., 1995; Preskorn et al., 1994; Alderman
`et a1., 1997). Yet clinically important drug interactions are rarely reported in actual
`practice (Davies et al., 2004; deVane, 2006; Molden et al., 2005). One possible
`explanation is that the therapeutic index of the victim drug or drugs is large enough
`that even a substantial change in plasma levels is not clinically relevant. Another
`explanation is that clinicians recognize the potential DDI, and make a pre-emptive
`downward adjustment in the dose of the victim to prevent the DDI.
`
`24.3 Drug Interaction Mechanisms and Terminology
`
`We have used the term "perpetrator" to indicate the drug that is causing the DDI,
`while "victim" or "substrate" is the drug that is being interacted with (Greenblatt
`and von Moltke, 2008). In a pure phmmacodynamic DDI, the perpetrator does
`not alter the plasma concentrations or systemic pharmacokinetics of the victim.
`Instead, the two drugs produce either additive or antagonistic pharmacodynamic
`effects. The interaction may occur via additive or opposite actions on the same
`receptor systems that yield additive or opposite clinical actions. Ethyl alcohol and
`benzodiazepines produce additive sedation through their actions on the gamma(cid:173)
`aminobutyric acid (GAB A) receptor system (Chan, 1984; Greenblatt and von
`Moltke, 2008); the pharmacokinetic interaction between alcohol and benzodi(cid:173)
`azepines, if any, is small, and does not account for the additive sedative effects
`(Greenblatt et al., 1978; Greenblatt and von Moltke, 2008; Ochs et a1., 1984;).
`Benzodiazepine agonists and caffeine have antagonistic pharmacodynamic actions.
`Benzodiazepines produce sedation via the GABA-benzodiazepine receptor sys(cid:173)
`tem, whereas caffeine produces alertness due to its action as an adenosine receptor
`antagonist (Biaggioni et al., 1991; Kaplan et al., 1992a, b; Kaplan et al., 1993).
`When caffeine is given together with a benzodiazepine agonist such as zolpidem,
`the sedative effects of zolpidem are partially reversed (Cysneiros et aI., 2007).
`However, there is minimal, if any, pharmacokinetic interaction between these two
`agents.
`A pure phmmacokinetic interaction involves only the effect of the perpetrator
`on the systemic clearance of the victim drug, causing plasma levels of the victim
`to increase or decrease. The clinical actions of the victim may be correspondingly
`increased or decreased, but only because of the indirect effects of the perpetrator
`on systemic clearance, rather than a direct effect of the perpetrator on the target
`receptor mediating clinical action.
`Pharmacokinetic DDIs involving drug-metabolizing enzyme systems (such as
`the CYPs) are generally classified as inhibition or induction. With metabolic inhibi(cid:173)
`tion, the perpetrator impairs the clearance of the victim drug, systemic exposure
`increases, and the clinical concern is toxicity. With induction, clearance of the
`victim increases, systemic exposure decreases, and the clinical concern is lack of
`efficacy (Table 24.2). However, inhibition and induction are not simply the same
`process in opposite directions - they involve fundamentally different mechanisms.
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2049, Page 7
`
`

`

`630
`
`D.J. Greenblatt and L.L. von Moltke
`
`Inhibition of victim drug clearance happens rapidly upon exposure to the perpe(cid:173)
`trator, and represents a direct effect of the perpetrator on the drug-metabolizing
`enzyme. Metabolic inhibition can be studied in vitro using cell homogenates from
`human liver, or cells expressing human metabolic enzymes (Venkatakrishnan et aI.,
`2001; Venkatakrishnan et aI., 2003). From these in vitro systems itis straightforward
`to derive metrics of inhibitory potency such as the inhibition constant (Kj) or the
`50% inhibitory concentration (ICso). In contrast, induction is an indirect process -
`the perpetrator (inducer) initiates a signal for the cell to produce more metabolic
`protein. This is slower than inhibition, and requires cultures of intact cells to study
`in vitro. The metric of induction potency is not so straightforward. Generally, the
`inductive effect of a candidate inducer is expressed as the fractional degree of induc(cid:173)
`tion relative to the hypothetical "maximum" induction by an index inducer such as
`rifampin.
`
`Table 24.2 Comparison of metabolic inhibition and induction
`
`Inhibition
`
`Induction
`
`Effect on victim drug
`Clearance
`Plasma levels
`Principal clinical concern
`Onset (after exposure to
`perpetrator)
`Offset (after perpetrator is
`discontinued)
`Mechanism
`
`In vitro system
`Metric of potency in vitro
`
`Decreased
`Increased
`Toxicity
`Rapid
`
`Rapid
`
`Increased
`Decreased
`Loss of efficacy
`Slow
`
`Slow
`
`Direct chemical
`effect
`Cell homogenates
`Ki or ICso
`
`Indirect signal to increase
`protein synthesis
`Cell culture
`Induction relative to maximum
`
`24.4 The Design of Clinical Drug Interaction Studies
`
`The general objective of DDI studies is to answer the following scientific questions:
`
`1. Given candidate "victim" and "perpetrator" drugs, is there a pharmacokinetic
`interaction between these two drugs that is not a chance event?
`2. What is the magnitude of the pharmacokinetic interaction?
`3. Is the interaction likely to be of clinical importance?
`
`Answers to the first two questions are largely objective and numerical, with little
`need for subjective interpretation or supplemental information. The third question is
`different - unless the DDI study incorporates measures of pharmacodynamic effect
`that are applicable to the target patient population, some supplemental information
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2049, Page 8
`
`

`

`24 Clinical Studies of Drug-Drug Interactions: Design and Interpretation
`
`631
`
`on the exposure-response relationship for the victim substrate drug is needed before
`a judgment can be made.
`
`24.4.1 Study Rationale
`
`The majority of clinical DDI studies involve healthy volunteers who do not have
`a medical need for the drugs under study. As such, study participation for these
`individuals is of no clinical benefit, but does entail some risk (though presumably
`low, and acceptable to an Institutional Review Board). There is also a dollar cost
`involved in the conduct of DDI studies. The cost is borne by the pharmaceutical
`sponsor in the case of an investigational dmg, by the general public in the case of an
`NIH-supported study, or by some other entity. The core assumption is that the risk
`and cost of the DDI study are justified based on the potential public health benefit
`of the information to be acquired.
`Clinical observations raising the possibility of a DDI may form the basis for
`initiating a formal study to either confirm or rule out a DDI. In the course of drug
`development, in vitro data is commonly used to identify dmg pairs for which a DDI
`needs to be evaluated in a clinical study. "Drug X" may be identified as an inhibitor
`of a certain human CYP isofoml in vitro, with a quantitative potency metric of Ki or
`ICso. If [1] is a typical plasma concentration of Drug X encountered during treatment
`with the highest approved dosage, then the ratio [1]/Ki or [1]/ICso is used to judge
`whether a clinical DDI is unlikely, possible, or probable, based on FDA guidelines.
`A DDI is termed "possible" if
`
`[I]/Ki > 0.1
`
`(24.2)
`
`This boundary is arguably too conservative on scientific grounds and triggers a
`large number of clinical DDI studies which tum out to be negative. Nonetheless that
`boundary reflects the current regulatory outlook, and sponsors often will initiate a
`DDI study on that basis.
`A second category of rationale for DDI studies is not directly scientific, but rather
`epidemiologic, based on a high probability of concurrent dmg therapy. Dmg X may
`be under development for a medical condition (such as diabetes, hypertension, or
`hyperlipidemia) that has high co-morbidity with ischemic heart disease. The sponsor
`may choose to initiate DDI studies of Drug X with digoxin or with warfarin because
`the probability of concurrent therapy is high, and because digoxin and warfarin (as
`potential victim dmgs) have a narrow therapeutic index. Even if there is no direct
`scientific rationale raising the possibility of a DDI, it could be argued that clinical
`data excluding DDIs with digoxin or walfarin is needed to assure safe co-treatment
`of Drug X with these potentially hazardous medications.
`Finally, an inevitable consequence of the industry-based system of drug devel(cid:173)
`opment is that research may be initiated solely for business reasons. Within a given
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2049, Page 9
`
`

`

`632
`
`D.1. Greenblatt and L.L. von Moltke
`
`drug class, a number of therapeutic options may be available, for which differences
`in therapeutic efficacy or toxicity may be subtle at most (Table 24.3). Competitive
`advantage then may turn on pharmacokinetic properties, such as mechanism of
`clearance, elimination half-life, and the risk of DDls. A clinical study may be initi(cid:173)
`ated to show that the sponsor's Drug X is not an inhibitor of a specific CYP isoforn1,
`whereas a competitor drug within the same class is in fact a significant inhibitor of
`the same CYP. These properties can be included in a product label, and used by
`pharmaceutical representatives or advertising matelials for competitive advantage.
`An example is the interaction of macrolide antimicrobials with human CYP3A.
`Erythromycin, clarithromycin, and telithromycin are significant CYP3A inhibitors,
`whereas azithromycin is not (Greenblatt et al., 1998a).
`
`Table 24.3 Examples of
`drug classes for which
`individual drugs can be
`distinguished based on
`phatmacokinetic properties or
`drug interaction potential
`
`Newer antidepressants
`Fluoxetine
`Sertraline
`Paroxetine
`Fluvoxamine
`Citalopram
`Venlafaxine
`Drugs to treat erectile dysfunction
`Sildenafil
`Tadalafil
`Vardenafil
`Macrolide antimicrobials
`Erythromycin
`Clarithromycin
`Azithromycin
`Telithromycin
`Hypnotics
`Triazolam
`Zolpidem
`Eszopiclone
`Temazepam
`
`24.4.2 Protocol Construction
`
`The customary design is a typical DDI protocol that involves a randomized, two(cid:173)
`way crossover study in a series of healthy volunteers. On one occasion, the victim
`substrate is administered in the control or baseline condition, without coadministra(cid:173)
`tion of the perpetrator. Total area under the plasma concentration curve from zero to
`infinity is calculated CAUCo). On a separate occasion, area under the curve is deter(cid:173)
`mined in the same subjects during coadministration of inhibitor (AUCr). The AUC
`ratio (RAUc) is calculated as
`
`AUCr
`RAUC = AUCo
`
`(24.3)
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2049, Page 10
`
`

`

`24 Clinical Studies of Drug-Drug Interactions: Design and Interpretation
`
`633
`
`This represents the fractional increase in substrate AUe attributable to coad(cid:173)
`ministration of the perpetrator. The reciprocal of RAUC is the fractional change in
`clearance of the substrate.
`A key requirement is that the exposure of the volunteer subject to the perpetrator
`has to span the duration of blood sampling for plasma concentrations of the sub(cid:173)
`strate. The shorter the half-life of the substrate, the shorter the duration of sampling,
`and the lower the cost and risk of the DDI study. If the perpetrator drug is a metabolic
`inhibitor, this will prolong the necessary exposure duration and sampling time, but
`short half-life victim drugs, nonetheless, are easier to deal with in DDI studies. If
`the perpetrator is an inducer, this if anything decreases the necessary sampling dura(cid:173)
`tion, but this advantage may be offset by the need for a period of pretreatment with
`the inducer due to the time required to attain maximum induction.
`An alternative design is to study the kinetics of the victim drug at steady state.
`With this design, Equation (24.3) is modified to represent the ratio of substrate AUe
`values over a dosage interval segment at steady state. If the intrinsic kinetics of the
`victim drug are nonlinear, this may constitute support for the steady-state DDI study
`design. Beyond that, the steady-state design only has a "showcase" advantage in
`that it more closely mimics the usual therapeutic situation in which the substrate
`is given on an extended basis. However, if the kinetics of the substrate victim are
`linear (dose independent), single-dose kinetics are predictive of what will happen at
`steady state, and the single-dose design provides DDI data of equivalent quality. An
`obvious drawback of the steady-state design is that duration, cost, and risk of the
`study are substantially increased, since the substrate drug must be dosed to steady
`state both in the control condition and during coadministration of the perpetrator.
`
`24.4.3 Studies of Specific Drug Pairs
`
`The initiator of a DDI study may have a clinical or research question that applies
`only to a specific drug pair, without the objective of information that is more general(cid:173)
`izable. With the limited research objective, the study design involves administration
`of the substrate victim on two occasions, with and without perpetrator, as described
`above. The forthcoming research outcome applies to that drug pair, but not neces(cid:173)
`sarily applicable to any other pair. An example is the pharmacokinetic interaction of
`diazepam and fluvoxamine (Perucca et ai., 1994), applicable to that particular com(cid:173)
`bination of substrate and perpetrator, but with no obvious connection to other drug
`combinations.
`
`as Victim
`
`"Drug X" may be identified as a potential DDI victim either through in vitro data,
`clinical observations, or both. The in vitro model may have identified the one or
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2049, Page 11
`
`

`

`634
`
`DJ. Greenblatt and L.L. von Moltke
`
`more CYP isoforms responsible for clearance of Drug X. The commercial entity
`developing Drug X, or a group of academic investigators, then pose the question:
`what happens to the in vivo clearance of Drug X if one or more of the CYP iso(cid:173)
`forms responsible for clearance is either induced or inhibited by a perpetrator. This
`question may represent a critical point in drug development. The outcome could
`influence the drug's clinical safety profile, and the degree of restrictiveness of the
`product label if the drug is eventually approved. The DDI study outcome could even
`lead to discontinuation of the drug as a development candidate.
`The choice of perpetrator in the DDI study usually will be whatever produces
`the "worst case" - that is, the interaction of largest possible magnitude. The scien(cid:173)
`tific community and the FDA largely agree on what those specific perpetrators are,
`sometimes termed "index inhibitors" or "index inducers" (Table 24.4). Whatever the
`degree of inhibition or induction produced by the index compound, no other perpe(cid:173)
`trator will be any worse. Ketoconazole and ritonavir are typical choices of index
`inhibitor for studies of substrate victims metabolized by CYP3A (Lee et aI., 2002;
`Tsunoda et aI., 1999; Knox et aI., 2008; Greenblatt et aI., 2000). The sponsor or
`investigator may also wish to concurrently study a less potent perpetrator, in which
`case the DDI trial design would be modified to become a three-way crossover. For
`example, a candidate drug that is a CYP3A substrate may be studied with ketocona(cid:173)
`zole and erythromycin as perpetrators, representing strong and moderate CYP3A
`inhibitors, respectively.
`The impact of a DDI on the clearance of a victim drug is greatest when that
`drug is extensively metabolized, and a single CYP isoforrn mediates clearance.
`Candidate victim drugs metabolized mainly by CYP3A isoforrns are a target of
`concern, since inhibition of CYP3A by a strong inhibitor such as ketoconazole or
`ritonavir may produce large values of RAve (Equation (24.3» (Lee et aI., 2002;
`Tsunoda et aI., 1999; Knox et aI., 2008; Greenblatt et aI., 2000). Concern is aug(cid:173)
`mented when the substrate victim has high clearance, and undergoes significant
`presystemic extraction after oral dosage (Fig. 24.3).
`An important feature of study design is the optimal duration of pre-exposure to
`the perpetrator drug prior to administration of the substrate victim. To minimize
`study cost and risk, exposure duration should be the minimum necessary to pro(cid:173)
`duce maximum inhibition or induction. In the case of CYP3A inhibition studies,
`there is strong data to indicate that 24 h of pre-exposure to ketoconazole or ritonavir
`is sufficient to produce maximal inhibition (Fig. 24.4) (Stoch et aI., 2009). For a
`time-dependent (mechanism-based) CYP3A inhibitor such as erythromycin or clar(cid:173)
`ithromycin, 48 h of pre-exposure is sufficient (Okudaira et aI., 2007). On the other
`hand, if the perpetrator is an inducer (rifampin), a pre-treatment period of 5-7 days
`is needed for induction to become maximal (Ohnhaus et aI., 1989; Lin, 2006).
`
`24.4.5 Candidate Drug as Pelpetrator
`
`If the candidate drug is being evaluated as a possible perpetrator of DDIs, the
`study design requires selection of an index substrate - that is, a substrate victim
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2049, Page 12
`
`

`

`

`

`

`

`24 Clinical Studies of Drug-Drug Interactions: Design and Interpretation
`
`637
`
`Table 24.4 Representative index substrates, inhibitors, and inducers applicable to the design of
`drug interaction studies*
`
`CYP isoform
`
`Index substrates
`
`Index inhibitors
`
`Index inducers
`
`CYPIA2
`
`Caffeine
`
`Fluvoxamine
`
`CYP2B6
`CYP2C9
`CYP2C19
`CYP2D6
`
`CYP3A
`
`Bupropion, efavirenz
`Flurbiprofen
`Omeprazole
`Desipramine,
`dextromethorphan
`Midazolam, triazolam,
`buspirone
`
`Clopidogrel
`Fluconazole
`Fluvoxamine
`Quinidine, paroxetine
`
`[Cigarette
`smoking]
`Rifampin
`Rifampin
`Rifampin
`[None known]
`
`Ritonavir, ketoconazole Rifampin
`
`*Table entries are intended to be representative, not inclusive.
`
`et ai., 2003; Zhu et aI., 2001; Chainuvati et aI., 2003; Blakey et aI., 2004; Gurley
`et aI., 2002; Chow et aI., 2006; Zhou et al., 2004; Christensen et aL, 2003). Instead
`of separate studies, volunteer subjects receive a mix of substrates concurrently, or in
`close temporal proximity, in a single study. Many possible substrate combinations
`have been proposed and utilized in cocktail DDI studies. A key piece of prelimi(cid:173)
`nary infOlmation is an unequivocal demonstration that each pairwise combination
`of substrates in the cocktail does not itself create DDIs with each other.
`
`6 Approach to Analysis of Data
`
`If the clearance of a substrate drug is not dependent on a polymorphically regu(cid:173)
`lated process, the population distribution of AUC values following a fixed dose of
`that substrate will not be consistent with a normal distribution, but rather will have
`a positive skew. Generally the skewed pattern is consistent with a log-normal dis(cid:173)
`tribution (Fig. 24.5) (Greenblatt, 2008; Friedman et al., 1986; Lacey et al., 1997;
`Greenblatt et al., 1989). In any given DDI study, the number of AUC values is
`usually not sufficient for a stable characterization of the underlying statistical dis(cid:173)
`tribution. Nonetheless a log-normal distribution is generally assumed, based on
`experience with larger population studies.
`Going "by the book," a calculation of arithmetic mean and standard deviation
`(SD) of AUC is theoretically precluded if the underlying distribution is non-norma1.
`In practice, however, the values of mean and SD calculated based on the assump(cid:173)
`tion of underlying normal or log-normal distributions are nearly identicaL Statistical
`theory reportedly justifies calculation of geometric mean AUC, along with a 90%
`confidence interval. However, the geometric mean value will underestimate the
`"real" mean value calculated using the assumption of a normal or log-normal
`distribution (Fig. 24.5).
`Statistical analysis of the significance of the DDI - that is, whether the aggre(cid:173)
`gate value of AUCo differs from AVCI -
`is most straightforwardly done via a
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2049, Page 15
`
`

`

`

`

`24 Clinical Studies of Drug-Drug Interactions: Design and Interpretation
`
`639
`
`Compare mean AUCo and AUCr using nonparametric test
`Compare arithmetic mean RAUC VS. l.00 using Student's t-test
`Determine whether 1.00 falls within 90% CI around geometric mean of RAUC
`In our experience, the different statistical options yield conclusions that are essen(cid:173)
`tially identical.
`
`24.5 Is a Drug Interaction of Clinical Importance?
`
`A key and critical point is that none of these options for data aggregation and
`statistical analysis described above provides insight on 'whether a DDI is clini(cid:173)
`cally important. That judgment must be based on supplemental knowledge of the
`exposure-response relationship for the victim drug. A quantitatively large inter(cid:173)
`action is more likely to be clinically significant, but this is not necessarily so
`(Culm-Merdek et al., 2005). Also, statistical significance is not equivalent to clinical
`significance.
`If a perpetrator drug, acting as a CYP inhibitor, causes a 50% increase in
`exposure to the substrate victim, this would be numelically evident as
`
`meanAUCr-meanAUCo - 0 5
`mean AUCo
`- .
`
`or
`mean RAUC = 1.5
`
`To judge clinical importance of the interaction, one would need another data
`source to determine whether a 50% increase in mean plasma levels of the sub(cid:173)
`strate drug was sufficient to cause a meaningful change in efficacy or the occunence
`of toxicity. The most convincing data on this question comes from clinical trials
`evaluating efficacy and adverse events in relation to dosage among patients tak(cid:173)
`ing the substrate drug for clinical purposes. If the daily dosage is increased by
`a factor of l.5 - on average, the equivalent consequence of the DDI - is there
`greater efficacy ancl!or an increased frequency of adverse events? In some DDI
`studies, pharmacodynamic endpoints are included as part of the study design, in
`which case the DDI study itself may provide data on the clinical consequences of
`the interaction (Cysneiros et al., 2007; Greenblatt et at, 1998a; Greenblatt et al.,
`2000; Culm-Merdek et al., 2005; Greenblatt et al., 1984; Greenblatt et al., 1998b;
`Greenblatt et a1., 2003; Culm-Merdek et al., 2006; von Moltke et al., 1996). The
`limitation of kinetic-dynamic studies
`healthy volunteers is that extrapolation to
`patient populations taking the substrate drug on an extended basis is not necessarily
`straightforward.
`Investigators with a pre-existing understanding of dose/concentrationiresponse
`relationships for the substrate victim may have the option of incorporating this infor(cid:173)
`mation into the statistical inference plan for the DDI study. Suppose it is established
`that RAUC can range from 0.7 to 1.4 with no evident clinical consequence. The DDI
`protocol could then adopt this range as pre-determined "no-effect boundaries." If
`
`Teva Pharmaceuticals USA, Inc. v. Corcept Therapeutics, Inc.
`PGR2019-00048
`Corcept Ex. 2049, Page 17
`
`

`

`640
`
`D.J. Greenblatt and L.L. von Moltke
`
`the 90% CI around the mean RAUC falls entirely within the 0.7-1.4 range, then the
`DDI - even if statistically significant - can be deemed as unlikely to be clinically
`important.
`
`24.6 Are Clinical Drug Interactions Predictable from In Vitro
`Models?
`
`In vitro data now is commonly used in the course of drug development to identify
`potential DDIs that may require clinical studies to rule out or confirm, and to quan(cid:173)
`titate the magnitude of the DDI if there is one. Current FDA guidelines deem that a
`DDI is possible if [1]/Ki is greater than 0.1, as discussed above (Huang et ai., 2008;
`Huang et aI., 2007; Tucker et ai., 2001). These guidelines do allow more informed
`targeting of the extensive resources needed to conduct clinical studies, but the
`guidelines nonetheless are very conservative and minimally quantitative. For more
`than a decade, there has been substantial investment of research energy into the
`question of whether in vitro models can provide more specific quantitative infor(cid:173)
`mation that can forecast not only whether or not a clinical DDI will happen, but
`also how small or big the interaction will be (Venkatakrishnan et aI., 2001, 2003;
`Greenblatt et aI., 2008; von Moltke et aI., 1994a; Brown et aI., 2006; Galetin et aI.,
`2005; Lin, 2006; Obach et aI., 2005, 2006; Ragueneau-Majlessi et aI., 2007; von
`Moltke et aI., 1998; Youdim et aI., 2008; Zhou, 2008; Williams et aI., 2004; Ito et
`a1., 2003; Rostami-Hodjegan and Tucker, 2007; Galetin and Houston, 2006; Galetin
`et aI., 2006, 2007; Ito et aI., 2004; Ohno et aI., 2007; Kanamitsu et aI., 2000; Brown
`et aI., 2005; Bachmann and Lewis, 2005; Bachmann, 2006; Lin, 2000; Volak et al.,
`2007; Farkas et aI., 2008).
`The basis for all predictive in vitro-in vivo scaling models is a link between what
`is observed in a clinical DDI study (as in Equation (24.3», what is available from
`the in vitro model (such as Ki or IC50), and some measured or assumed in vivo
`exposure to the inhibitor ([1]). The most straightforward linkage relationship is
`
`AUCr
`[1]
`RAUC = - - = 1 + -
`AUCo
`Ki
`
`(24.4)
`
`RAUC is objectively determined in a clinically DDI study, and Ki is determined in
`vitro based on generally accepted procedures. A principal uncertainty is the inhibitor
`exposure. Theoretical model validity requires that [1] be the inhibitor c

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