throbber
DRUG-DRUG
`
`INTERACTIONS:
`
`SCIENTIFIC AND
`
`REGULATORY
`
`PERSPECTIVES
`
`Albert P. Li
`In Vitro Technologies Inc.
`University of Ivlarylan
`echnology Center
`"more, Maryland
`
`’
`
`ACADEMIC PRESS
`
`San Diego London Boston New York Sydney Tokyo Toronto
`
`Vanda Exhibit 2043 - Page 1
`
`Vanda Exhibit 2043 - Page 1
`
`

`
`This book is printed on acid—free paper.
`
`Copyright © 1997 by ACADEMIC PRESS
`
`All Rights Reserved.
`No part of this publication may be reproduced or transmitted in any form or by any
`means, electronic or mechanical, including photocopy, recording. or any information
`storage and retrieval system. without permission in writing from the Publisher.
`The appearance of the code at the bottom of the first page of a chapter in this book
`indicates the Publisher‘s consent that copies of the chapter may be made for
`personal or internal use of specific clients. This consent is given on the condition.
`however, that the copier pay the stated per copy fee through the Copyright Clearance
`Center, Inc. (222 Rosewood Drive. Danvers. Massachusetts 01923), for copying
`beyond that permitted by Sections 107 or 108 of the U.S. Copyright Law. This consent
`does not extend to other kinds of copying, such as copying for general distribution. for
`advertising or promotional purposes. for creating new collective works, or for resale.
`Copy fees for pre-1997 chapters are as shown on the title pages. if no fee code
`appears on the title page. the copy fee is the same as for current chapters.
`1054-3589/97 $25.00
`
`Academic Press
`a division 0fHarcom'I Bl'(1(‘(’ & C(}H1[J(lI1V\'
`525 B Street. Suite 1900. San Diego. California 92101—4495. USA
`http://\v\v\v.apnet.com
`
`Academic Press Limited
`24-28 Oval Road. London NW1 7DX. UK
`http://\v\v\v.hbuk.co.uk/ap/
`
`International Standard Book Number: 0-12-03L’94'~1—1
`
`Vanda Exhibit 2043 - Page 2
`
`

`
`Vanda Exhibit 2043 - Page 3
`
`This material may be protected by Copyright law (Title 17 U.S. Code)
`
`

`
`F. Peter Guengerich
`
`such as lung and small intestine, where an appreciable contribution to overall
`metabolism of a drug can occur depending on the route of administration.
`Fortunately, it appears that the metabolism of most drugs can be ac-
`counted for by a relatively small subset of the P4505. One estimate is that
`290% of human drug oxidation can be attributed to six enzymes: P4505
`1A2, 2C9/10, ZC19, ZD6, 2E1, and 3A4 (Guengerich, 1995; Wrigliton and
`Stevens, 1992). Further, most could probably be attributed to P4505 1A2,
`2C9/10, ZD6, and 3A4 and, by some estimates, half can be attributed to
`P450 3A4 (Guengerich, 1995; Guengerich et £11., 1994a). This view is based
`primarily on (in uitro) microsomal studies done with drugs studied to date
`and may change somewhat with time. For instance, the fraction of drugs
`oxidized by P450 ZD6 may be too high in current estimates because of the
`ease of identifying these and the attention that has been given to this particu-
`lar enzyme. Nevertheless, the concept that most drug oxidations are cata-
`lyzed primarily by a small number of P450 enzymes is important in that
`the approaches to identifying drug—drug interactions are feasible, both in
`z/itro and in viva.
`I
`This chapter operates from the premise that many significant drug—drug
`interactions can be understood in terms of P4505. However, drug—drug
`interactions are more complex for at least two reasons. First, some drug-
`drug interactions can be attributed to pharmacokinetic differences due to
`other enzymes such as monoamine oxidases, flavin-containing monooxygen—
`ases, UDP—glucuronosyl transferases, and sulfotransferases. These and other
`so—called “drug-metabolizing” (or “xenobiotic—metabolizing”) enzymes also
`show the characteristics of induction and inhibition by drugs that are associ-
`ated with P4505, although most have not yet been studied as extensively.
`The other aspect of drug—drug interactions is that some of these are probably
`pharmacodynamic instead of pharmacokinetic. For instance, drugs can com-
`pete for binding to a receptor directly related to the pharmacological re-
`sponse.
`
`ll. Potential Consequences of Drug—Drug Interactions
`
`the major effects of drug—drug
`From a pharmacokinetic standpoint,
`interactions can be understood in terms of causing the disposition of a durg
`to be unusually slow or fast. The major consequence is a high or low plasma
`and tissue level of the drug.
`If the metabolism of a drug is impeded due to enzyme inhibition, then
`a high plasma level may follow (Fig. 1). One of the major effects will be
`increased pharmacological activity, and this may or may not be a problem,
`depending on the therapeutic window. Of course, not only the desired effect
`
`Vanda Exhibit 2043 - Page 4
`
`

`
`Plasma
`level of
`
`drug
`
`FIGURE I
`
`Time (arrows show repeated doses)
`Effect of enzyme inhibition on drug metabolism and plasma drug levels.
`
`be anticipated. Another possibility is that when the major pathway of metab-
`olism of a drug is blocked, secondary pathways may become more favorable.
`This can be a problem if the secondary pathway leads to a toxic product.
`An example of this is seen with the analgesic phenacetin (no longer on the
`U.S. market). If O-deethylation (P450 1A2) is slow, then other pathways
`are favored that lead to quinoneimine formation and methemoglobinemia
`(Fischbach and Lenk, 1985; Klehr et ((1., 1987). Another possibility is that
`the increased level of a drug due to inhibition of the P450 involved in its
`oxidation may lead to inhibition of another P450. Although direct evidence
`for such a situation has not been presented, one could postulate that accumu-
`lation of quinidine due to P450 3A4 inhibition might lead to inhibition of
`P450 ZD6, an enzyme for which quinidine is an inhibitor but not a substrate
`(Guengerich er £11., 1986b; Otton at al., 1984).
`When levels of P450 (or, for that matter, another enzyme) are induced,
`the major consequence is a lack of therapeutic effectiveness. Although this
`might seem to be a common event, the number of real clinical situations in
`which this has been a problem are rather limited. Two of the best documented
`examples are cyclosporin and 17oz-ethynylestradiol (uide infra). Another
`possibility with a pro-drug is that activation may be too rapid and a seriously
`high level of active drug could result. This could be a problem, as one of
`the primary reasons for developing pro—drugs is to avoid a transiently high
`level of the active drug. However, no good examples of clinical problems
`resulting from a phenomenon of this type are known yet.
`There are two other possibilities that can be considered in regard to
`issues of drug—drug interactions. One involves metabolism of chemical car-
`cinogens. Most of the P4505 that transform drugs can also oxidize chemical
`carcinogens (Guengerich and Shimada, 1991; Guengerich, 1995). The possi-
`bility exists that a P450 induced by a drug could lead to enhanced levels
`
`Vanda Exhibit 2043 - Page 5
`
`Vanda Exhibit 2043 - Page 5
`
`

`
`F. Peter Guengerich
`
`of DNA—carcinogen adducts due to increased carcinogen activation. The
`induction of human P450s 1A1 and 1A2 by omeprazole was postulated to
`present a risk due to such considerations (Diaz er a/., 1990). Whether or
`not this is a serious issue is unknown, as levels of P450 1A2 are only one
`of many factors linked to cancer risk from known carcinogenic substrates
`for the enzyme (Lang et a1., 1994). Nevertheless, most pharmaceutical com-
`panies and the Food and Drug Administration (FDA) would rather avoid
`drugs that induce P450 1A subfamily enzymes, which have been suggested
`to be related to cancer development (Ioannides and Parke, 1990, 1993).
`Again, it should be emphasized that increased cancer risk due to P450 (1A
`or other) induction is still a hypothesis. Another matter to consider is that
`some P4505 are involved in the detoxication of potential carcinogens and
`that induction or inhibition might have an impact on this process, as well
`as the activation (Guengerich and Shimada, 1991; Richardson et a1., 1952;
`Nebert, 1989).
`The other possibility involves the influence of drugs on P450s involved
`in the transformation of endogenous compounds, i.e., those normally found
`in the body. This matter has not been extensively investigated, but some
`possibilities exist. Depending on the tissue, 17B—estradiol
`is oxidized by
`P450 3A4 (Brian et 511., 1990; Guengerich et ((1., 1986a), P450 1A2 (Guo
`et (11,, 1994), or P450 1B1 (Liehr et a1., 1995). It is not known what the
`impact of changes in these enzymes is on total body levels. Testosterone is
`a substrate for P450 3A4 (Guengerich et £11., 1986a; Waxman et (11,, 1988).
`Another case to consider is animals devoid of bilirubin UDP-glucuronosyl
`transferase activity, who can be administered P450 1A inducers to lower
`their levels of bilirubin to nontoxic levels (Kapitulnik and Gonzalez, 1993;
`Kapitulnik and Ostrow, 1978).
`A final point to consider is that some drugs may affect the disposition
`of chemicals in foods and beverages through P450 interactions. For instance,
`the drug disulfiram (Antabuse) inhibits P450 2E1. This would affect ethanol
`oxidation by P450 2E1, although the more serious effect is on aldehyde
`dehydrogenase (Guengerich et ((1., 1991). The P450 1A2 inhibitor furafylline
`blocks caffeine N3-demethylation to the point where severe insomnia is
`associated with drinking coffee (Sesardic er ((1., 1990; Kunze and Trager,
`1993).
`
`III. Use of Information about Human P450s
`
`Much of the information abot drug metabolism by human P450s has
`been acquired in the past decade. Induction was first seen in clinical settings
`in the 1950s (Remmer, 1959) and there were many in I/itro studies with
`
`Vanda Exhibit 2043 - Page 6
`
`

`
`and CDNA cloning methods were used to obtain DNA sequences for the
`human P4505 (Gonzalez, 1989).
`In recent years the access to human tissue samples in the United States
`and Europe has facilitated characterization of P450 reactions catalyzed by
`human P4505. The availability of the recombinant human P4505 expressed
`in various systems has also facilitated studies on their catalytic selectivity
`(Gonzalez et al., 1991a,b; Guengerich et ((1., 1996). Thus, it is now relatively
`straightforward to use in uitro studies to determine which P4503 oxidize a
`particular drug and which drugs can inhibit oxidations catalyzed by this
`P450. The in uitro determination of inducibility is not as easily done, but
`a number ofpossibilities exist with cultures ofhuman hepatocytes (Guillouzo
`et £11., 1993; Loretz et (11,, 1989) (also see chapter by A. P. Li).
`It is also possible to do logical in viva studies to test the relevance of
`in uitro findings. For instance, individuals known to be high or low in a
`particular P450 from the use of other noninvasive assays can be examined
`with regard to the pharmacokinetics of the new drug to see if there is a
`match. In some cases, inducers or inhibitors of a specific P450 can be given
`safely to people to verify that a P450 is involved in the oxidation of a drug.
`Also, the drug under consideration can be given to people to determine if
`it affects the pharmacokinetics of other drugs through enzyme induction.
`The acquisition of the in L/itro information about a new drug can be
`extremely useful. In many cases, the FDA now expects in uitro information
`on the P4505 involved in the oxidation of a drug early in the registration
`process. The in 1/itro information can be used to guide the more expensive
`and time—consuming in viva studies. In particular, potential adverse drug
`interactions due to pharmacokinetics can be predicted and the number of
`in uiz/0 interaction studies can be restricted, as some of those historically
`done with all new drugs may be found irrelevant. The in 1/[tro procedures,
`if used early in the drug development process, may be used to select from
`a series of potential candidates, in terms of which will be least likely to
`cause problems with drug—drug interactions. Another point is that the in
`uitro studies can be used as a guide in predicting bioavailability, simply by
`screening candidate drugs for resistance to oxidation by the major known
`human P4505.
`
`IV. Mechanisms of Drug Interactions Attributable
`
`to P4505
`
`A. Induction
`
`This is a phenomenon first identified a halfcentury ago in in viva studies
`with humans, primarily in the laboratories of Remmer and Brodie (Remmer,
`
`Vanda Exhibit 2043 - Page 7
`
`Vanda Exhibit 2043 - Page 7
`
`

`
`F. Peter Guengerich
`
`1959; Brodie et a/., 1958). Individuals who were administered certain drugs
`developed a certain “tolerance,” in that increasing doses were needed to
`produce the same effect. Work with experimental animals demonstrated
`that the effect could be reproduced. For instance, animals treated with
`barbiturates decreased their “sleeping time,” a parameter indicating how
`long a certain dose of a barbiturate would keep animals sedated (Burke,
`1981). Other studies on chemical carcinogenesis reinforced the concept of
`enzyme induction (Conney et ((1., 1956; Conney, 1967), particularly with
`what are now termed the P450 1A subfamily genes.
`The mechanism of P450 1A induction is perhaps the most well character-
`ized in this field (for reviews, see Hankinson, 1993; Denison and Wliitlock,
`1995). Although barbiturate induction was also discovered early, mechanis-
`tic studies on this phenomenon are not as well developed and there is not
`general agreement regarding observations in different laboratories (Rangara—
`jan and Padmanaban, 1989; Liang er a/., 1995; Ramsden er a/., 1993).
`Nevertheless, there seems to be a rather general agreement that most human
`P450 2C and 3A subfamily proteins are induced by barbiturates (Zilly et
`a/., 1975; Morel et al., 1990). Studies with experimental animals indicate that
`subfamily 2B proteins are induced by barbiturates (Burke, 1981; Guengerich,
`1987), but direct information on inducibility in humans is not available.
`Evidence indicates that human P450 2E1 is inducible by ethanol and isonia—
`zid, although the mechanism of the process is complex (Perrot et al., 1989;
`Kim et a/., 1994). Compounds (including drugs) that cause peroxisomal
`proliferation induce P4505 in the 4A subfamily in experimental animals
`(Muerhoff et (11,, 1992; Rao and Reddy, 1991; Gibson, 1993); presumably
`this can also happen in humans, although the system is suspected to be less
`responsive (Bell et a/., 1993). The mechanism involves the interactions of
`ligand—bound peroxisomal proliferation activation receptor (PPAR): retinoid
`X receptor (RXR) heterodimers with upstream recognition sequences (Lee
`et al., 1993), and compounds such as fatty acids and retinoids may be
`involved in this response.
`The effect of induction is simply to increase the amount of the P450
`present and make oxidation and clearance of a drug faster. As mentioned
`earlier, details of mechanisms remain to be understood. The overall situation
`is complicated because even in situations where a response element can be
`identified, there are probably interactions with other response systems that
`must be considered. However, knowledge of such phenomena will be useful
`in the further development of in uitro systems that can be used to screen new
`drug candidates for their potential as inducers of P4505 and other enzymes.
`
`B. Inhibition
`
`Vanda Exhibit 2043 - Page 8
`
`

`
`that can be attributed to inhibition rather than induction. There are different
`
`types of enzyme inhibition, and the clinical effects are influenced by the
`basic mechanisms.
`V
`The first type of inhibition is competitive, where the inhibitor and sub-
`strate compete for the same binding site on an enzyme. In the situations
`under consideration here, the inhibitor and the substrate would be drugs,
`competing for the binding site of a P450. Insofar as is currently known,
`P4505 are thought to have a single substrate—binding site (aside from some
`possible allosteric situations, z/ide infra) (Raag and Poulos, 1991; Cupp-
`Vickery and Poulos, 1995), although the sizes and flexibility of the micro-
`somal P4505 we are concerned with here are not known. The inhibitor may
`be a substrate itself. For examples, see the section on P450 ZD6 (uide infra).
`This type of inhibition is easily identified by the classic intersecting plots
`seen in in z/itro studies (Kuby, 1991).
`Another type of inhibition has precedent in the classical studies of
`enzymology. The two situations are called nozzcompetitiz/e inhibition, where
`the inhibitor binds at a site on the enzyme distinct from the substrate, and
`zmcompetitiz/e inhibition, where the inhibitor binds only to the enzyme-
`substrate complex (Kuby, 1991 ). Actually, neither of these have many clear
`examples in the literature of drug—drug interactions or in drug metabolism
`in general. (An example of a noncompetitive inhibitor would be a reagent
`that modifies sulfhydryl groups remote from the substrate—binding site to
`attenuate the activity of an enzyme.)
`A fairly common mechanism of inhibition related to drug—drug interac-
`tions is mec/mzzism-based, or suicide, inhibition (Silverman, 1988, 1995;
`Ortiz de Montellano and Correia, 1983; Ortiz de Montellano and Reich,
`1986; Halpert and Guengerich, 1997). In the strict definition of the mecha-
`nism, a substrate (the inhibitor) is transformed by the enzyme in the normal
`course used for other substrates and an intermediate is formed, which usually
`has a fleeting but finite half-life. This intermediate can partition between
`reaction with the enzyme (at the active site), to inactivate the enzyme, or
`undergo a different transformation (e.g., reaction with water or proton loss)
`to yield a stable product. The ratio of the two processes (latter : former) is
`termed the partition ratio and is used to compare the efficiencies of different
`_mechanism-based inactivators. Mechanism—based inactivators are character-
`ized in uitro by a number of properties, including time—dependent loss of
`enzyme activity, requirement for normal enzyme cofactors, blockage by
`noninhibitory substrates, saturation kinetics, and (usually) single irreversible
`modification of the protein or prosthetic group (Silverman, 1988). Examples
`of this type are seen in the P450 drug metabolism literature with compounds
`such as secobarbital (Levin et al., 1973), gestodene (Guengerich, 1990a),
`
`Vanda Exhibit 2043 - Page 9
`
`Vanda Exhibit 2043 - Page 9
`
`

`
`F. Peter Guengerich
`
`furafylline (Kunze and Trager, 1993), and disulfiram (Guengerich et a/.,
`1991; Brady et al., 1991), and many clinical interactions may be understood
`in these terms. The inhibition of specific enzymes by mechanism—based inacti-
`vators is an approach used in the design of new drugs. In principle, a
`substrate can be designed as a mechanism—based inactivator of a single
`enzyme. This approach has been used to attenuate monoamine oxidase
`(Thull and Testa, 1994). The only good examples of development of drugs
`to specifically inhibit P450s deal with P450 19, the steroid aromatase, which
`is a target in breast and ovarian tumors because of its role in estrogen
`production (Brodie, 1994). Nevertheless, there are many examples of experi-
`mental compounds that are selective inactivators of individual P450 enzymes
`in uitro and in experimental animals (Ortiz de Montellano and Reich, 1986).
`These can be used in a diagnostic manner (to help identify P4505 involved
`in various reactions) (Guengerich and Shimada, 1991) to label enzyme active
`sites (Roberts et al., 1994; Yun et al., 1992) and to identify a drug target
`in a complex mixture of proteins (Rando, 1984).
`Several other types of irreversible enzyme inhibition are related to
`mechanism—based inactivation but can be distinguished. In one case, there
`is time-dependent inhibition at the active site by reaction of a substrate (or
`analog) with the protein, unrelated to the normal enzyme mechanism. A
`good example is not available for P450, except perhaps a substrate such as
`acrylonitrile that reacts rather nonselectively with all protein sulfhydryls
`but is oxidized by P450 2E1. A slow—binding inhibitor of testosterone 50¢-
`reductase is the prostate growth inhibitor finasteride (Proscar) in which the
`enzyme bonds with the drugs at a slow rate, competitive with normal ste-
`roids, and irreversibly inactivates the enzyme (Tian et a/., 1995).
`Another case involves the conversion of a substrate to a product that
`is reactive enough to modify the protein. An example of this latter case is
`chloramphenicol, which is oxidized by P450 to an acyl chloride (Halpert
`et 111., 1985). The acyl chloride is not an enzyme intermediate in the strict
`sense. The product can be readily hydrolyzed by water. It would also leave the
`protein and modify other proteins; however, the similarity of the molecule to
`the substrate seems to keep it in the active site so that it will label groups
`there. Distinguishing inhibitors of this type from true mechanism—based
`inactivators may not be easy; one test is to determine if a scavenger such
`as glutathione (which would not enter the active site of the enzyme) can
`block inhibition. Another test is to find a certain P450 enzyme (“P450 1”)
`that is not inactivated when incubated with the drug (plus normal cofactors).
`This P450 (“P450 1”) can be mixed with the drug, cofactors, and another
`P450 known to be inactivated (“P450 2”). If P450 1 is now inactivated,
`then the most direct explanation is that a reactive product has migrated
`from P450 2 to P450 1. Although the mechanistic distinction may seem
`
`Vanda Exhibit 2043 - Page 10
`
`

`
`the heme iron instead of covalent modification of amino acid residues. For
`
`instance, many amines are oxidized to nitroso compounds that form spectral
`complexes with absorbance maxima at 455 nm (jonsson and Lindeke, 1992;
`Mansuy et a/., 1983). A classical case in pesticide biochemistry is the synergist
`piperonyl butoxide, which is oxidized to a carbene that binds the heme
`(Ortiz de Montellano and Reich, 1986). Evidence shows that mechanisms
`of this type may be important in inhibition under physiological conditions
`(Bensoussan et a/., 1995).
`
`C. Stimulation
`
`Enzyme stimulation refers to the process by which direct addition of
`one compound to an enzyme enhances the rate of reaction of the substrate.
`This phenomenon has been observed in a number of cases with P4505
`(Halpert and Guengerich, 1997; Huang et al., 1981).
`Distinguishing enzyme induction and stimulation in viva is not easy
`because some of the compounds that seem most effective in P450 stimulation
`are also enzyme inducers, e.g., flavonoids. One approach used was the
`treatment of rats with a substrate in which product formation was accompa-
`nied by the release of tritiated water, for a short period of time (15 min),
`in the absence or presence of flavone (Lasker er ((1., 1982). The increase in
`product formation observed (in total body radioactive water) in the presence
`of flavone provides evidence that stimulation occurred in a time frame before
`significant enzyme induction could have occurred.
`In our laboratory we have been studying the effect of a—naphthoflavone
`on the oxidation of the carcinogen aflatoxin B, by P450 3A4 (Raney et al.,
`1992; Ueng et a1., 1995). oz-Naphthoflavone has the interesting effect of
`inhibiting the 3oz—hydroxylation of aflatoxin B, but stimulating the 8,9-
`epoxidation, and our current working hypothesis is that an allosteric mecha-
`nism is involved (Ueng et al., 1995; Guengerich et al., 1994b). In line with
`this view, plots of rates of these reactions versus substrate concentration
`are sigmoidal
`in the absence of wnaphthoflavone but hyperbolic in the
`presence of oz-naphthoflavone (Ueng et a/., 1995). There is also evidence in
`the literature that sigmoidal kinetics are observed in the in uitro oxidation
`of drugs [e.g., carbamazepine (Kerr et a/., 1994) and possibly acetaminophen
`(Lee et al., 1991)] and steroids [e.g., progesterone and 17B—estradiol (Schwab
`et al., 1988)], usually with P450 3A subfamily enzymes.
`The in viz/o relevance of these phenomena to drug metabolism remains
`to be established, as does the mechanism(s).
`
`Vanda Exhibit 2043 - Page 11
`
`Vanda Exhibit 2043 - Page 11
`
`

`
`F. Peter Guengerich
`
`V. Examples of P450-Based Interactions
`
`A. Cimetidine
`
`Cimetidine (Tagemet, Fig. 2) is a drug that inhibits antihistamine H1
`receptor binding and is used in the treatment of gastric ulcers. There is
`considerable literature on the inhibition of drug metabolism by cimetidine
`in both animal and human models (Gerber er al., 1985). A similar H3
`receptor antagonist, ratinidine (Zantac), was developed by another company
`and was devoid of the inhibitory properties, a point that was exploited
`in marketing.
`Analysis of the scientific literature indicates thatcimetidine is a relatively
`weak P450 inhibitor (Knodell et £11., 1991). No serious acute episodes of
`adverse health have been attributed to cimetidine despite long use in many
`patients, many of whom are undoubtedly using other drugs.
`The mechanism of inhibition appears to involve the imidazole ring of
`cimetidine, which is not present in ranitidine. Cimetidine shows selectivity
`for inhibiting reactions catalyzed by P4505 ZD6 and 3A4 (Knodell et ((1.,
`1991). The inhibition has generally been regarded as due to competitive
`binding of cimetidine, possibly through interaction of the imidazole with
`the P450 heme. However, some evidence for mechanism—based inactivation
`of P450 has also been published (Coleman et a/., 1991), although a chemical
`basis has not been established.
`
`B. P450 2D6
`
`P450 2D6 inhibitors and substrates have attracted considerable concern.
`In the early 1970s Smith personally experienced an adverse response in a
`clinical trial of the antihypertensive agent debrisoquine. This episode led
`him to study the basis in more detail, and the work led to the identification
`of a subset of the population (~7% Caucasians) as “poor metabolizers,”
`who hydroxylated the drug at a much slower rate than the rest of the
`population (Mahgoub et al., 1977).
`Subsequent work led to characterization of this enzyme, P450 ZD6, by
`purification, CDNA cloning, and genetic analysis (Gonzalez et a/., 1988;
`Gonzalez and 1\/Ieyer, 1991). P450 ZD6 is now recognized to be involved
`in the oxidation of >30 drugs. Some of these show relatively narrow thera-
`
`CN
`
`H3C\N N/\/SWé<
`
`H
`
`H
`
`NH
`
`N§/
`
`Vanda Exhibit 2043 - Page 12
`
`

`
`(Shah et a/., 1982). However, P450 2D6—deficient individuals do not convert
`the pro-drug encainide to its active form as effectively as the rest of the
`population (\X/oosley ez‘ a/., 1981). A number of drugs are also potent
`inhibitors of P450 ZD6 (Fig. 3) (Strobl et a/., 1993). Prominent among these
`are alkaloids such as quinidine and the ajmalicine derivatives (Strobl er a/.,
`1993; Fonne—Pfister and Meyer, 1988).
`It is now relatively easy to identify P450 ZD6 substrates and inhibitors
`in 1/itro early in the development process. Strong P450 ZD6 inhibitors are
`generally avoided. An issue can be raised, though, as to how serious a P450
`ZD6 inhibitor really is. Because ~5% of the population (depending on the
`country) is already deficient in P450 ZD6, the effect of the inhibitor is to
`extend this group of individuals. The problem would be slow metabolism
`of P450 2D6 substrates, but this may not be a serious issue.
`
`
`
`H‘-,CO,C
`
`Caihanamina
`
`R0
`
`Sempervirlne
`
`R5
`
`
`
`lndoia derivatives
`
`R2
`
`H
`
`H
`
`HO
`
`N
`R3
`
`H
`
`Fl:
`
`/ \
`
`N
`Oulnldine/quinine and
`derivatives
`
`
`
`Ouinlnone
`
`R4
`
`\
`/
`N
`Ouinoiine and derivatives
`
`R
`
`/
`
`N
`H
`Harman and derivatives
`
`FIGURE 3
`
`Some inhibitors of P450 ZD6 (Strobl at a/., 1993).
`
`Vanda Exhibit 2043 - Page 13
`
`Vanda Exhibit 2043 - Page 13
`
`

`
`F. Peter Guengerich
`
`The issue of development of P450 ZD6 substrates has been a more
`serious matter, and some pharmaceutical companies had developed policies
`of dropping these from development. A realistic way of addressing the issue
`is to test candidate drugs in 1/itro to determine if they are substrates and
`then proceed to examine them in viva to establish the pharmacokinetics
`and the therapeutic window. The majority of P450 ZD6 substrates can
`probably be tolerated reasonably well even by P450 ZD6-deficient indi-
`
`The molecular basis of the P450 ZD6 polymorphism has been described
`in detail, and there are a number of alleles that contribute to cause both
`unusually slow and also unusually rapid oxidation (Broly et 41., 1991;
`johansson et (11,, 1993). The P450 ZD6 substrates and inhibitors all seem
`to share a basic nitrogen group, which is positioned 5-7 A away from the
`site of hydroxylation. The carboxylate anionic moiety of Asp 301 has been
`suggested to interact with the basic nitrogen of the substrate, on the basis
`of modeling and site-directed mutagenesis work (Ellis et al., 1995). Sub-
`strates and inhibitors have been used to develop pharmacophore models of
`P450 ZD6 (Strobl et al., 1993; Islam et (IL, 1991; Koymans et £11., 1992).
`The strong inhibitors of P450 ZD6 (e.g., quinidine) are not readily oxidized
`(Strobl et al., 1993; Guengerich et al., 1986b), and the conclusion has been
`reached that the basic nitrogen in these binds to the same protein anion as
`the substrates (Asp 301) but no atoms that can be oxidized are accessible
`to the FeO complex (Islam er al., 1991). The model does not explain the
`oxidation of deprenyl by P450 ZD6 (Grace et al., 1994). A modification
`involves the transient deprotonation of the amine and electron transfer
`(Grace et al., 1994; Guengerich, 1995).
`
`I701-Ethynylestradiol
`
`This is a classic example of a drug—drug interaction and one of the few
`attributed to induction, instead of inhibition. In the early 1970s several
`German reports indicated that women who were using oral contraceptives
`began spotting or became pregnant after using rifampicin or barbiturates
`(Reimers and jezek, 1971; Nocl<e—Fincl< et £11., 1973; Janz and Schmidt,
`1974). The major estrogen in oral contraceptives is 17a—ethynylestradiol
`(Fig. 4), which is metabolized via 2-hydroxylation, plus other pathways
`(Bolt et al., 1973; Guengerich, 1990b). Administration of rifampicin resulted
`in the faster elimination of 17oz—ethynylestradiol in volunteers (Bolt 61‘ al.,
`
`Subsequently, P450 3A4 was shown to be a major enzyme involved in
`the (2—)hydroxylation of 17oz—ethynylestradiol (Guengerich, 1988). P450
`3A4 can also be induced by rifampicin or barbiturates in cultured human
`
`Vanda Exhibit 2043 - Page 14
`
`

`
`} |
`
`OH
`§.“\\'_-E
`
`.
`
`S OH
`.\‘\\—-_
`
`170:-Ethynylestradiol
`
`D°3°9°5"°'
`
`0H
`| am“:
`
`Norethlsterone
`
`OH
`.\‘\\:_
`
`3-Ketodesogestrel
`
`OH
`
`r 39“:
`
`O
`
`O
`
`O
`
`HO
`
`0
`
`o
`
`,
`
`; oH
`“E
`
`Gestodene
`
`OH
`.\‘\\'——
`
`Levonorgestrel
`
`OH
`
`‘F
`
`Q4 3 E
`
`11.cH2.A15.N°ret|-fistefone
`11'CH2-N0l'ethlStef0ne
`Structures of 17a—ethynylestradiol and several progestins used in oral Contracep-
`FIGURE 4
`tives (Gucngerich, l990a,l)).
`
`The ineffectiveness of oral contraceptives due to P450 3A4 induction
`can be explained in these terms. There could also be contributions of induced
`conjugating enzymes (e.g., UDP-glucuronosyltransferases), but these have
`not been documented. This phenomenon of lack of efficacy of oral contracep-
`tives is still a problem because of the low doses 0f17oz-ethynylestradiol used
`(to prevent unwanted effects of estrogens) and the sensitivity to changes due
`to variations in P450 3A4.
`
`In the course of work with 17oz—ethynylestradiol and oral contraceptives,
`the progestin gestodene (Fig. 4) was found to be a relatively effective and
`
`Vanda Exhibit 2043 - Page 15
`
`Vanda Exhibit 2043 - Page 15
`
`

`
`F. Peter Guengerich
`
`selective mechanism—based inactivator of P450 3A4 in in z/itro experiments
`(Guengerich, 1990a). This inactivation is due in part to the presence of an
`ethynyl moiety, which is also a part of many P450 inactivators (Ortiz de
`Montellano et a/., 1979; Gan et al., 1984). However, most of the pro-
`gestins used in oral contraceptives have 1701-ethynyl groups (Fig. 4), and
`other features of gestodene are apparently responsible for the inactivation
`(Guengerich, 1990a). This inactivation phenomenon has been postulated to
`account for the increased levels of estradiol and cortisol in women using
`oral contraceptives (]ung—Hoffmann and Kuhl, 1990), although it
`is not
`clear that the dose of gestodene is sufficient to inhibit a large fraction of
`hepatic or intestinal P450 3A4 (Guengerich, 1990a).
`
`D. Terfenadine
`
`Terfenadine is a component of the antihistamine formulation Seldane.
`It is rapidly oxidized by P450 3A4 to two products, acyclinol and an alcohol
`derived from oxidation of a t-butyl methyl group (Fig. 5) (Yun et al., 1993).
`Acyclinol is inactive. The alcohol is further oxidized to a carboxylic acid
`by either P450 3A4 (Rodrigues ez‘ al., 1995) or by dehydrogenases; the
`relative contributions of the two enzyme systems are not known (Fig. 6). This
`carboxylic acid, like terfenadine itself, binds to the H, histamine receptor and
`should produce relief of allergy symptoms. However, the acid is a zwitterion

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