throbber
0031-699779714904-0403$03.00.'0
`PHARMACOLOGICAL REVIEWS
`Copyright © 1997 by The American Society for Pharmacology and Experimental Therapeutics
`
`Vol. 49, No. 4
`Printed in U.S.A.
`
`Role of Pharmacokinetics and Metabolism in Drug
`Discovery and Development
`JIUNN H. LIN“ AND ANTHONY Y. H. LU
`
`Department of Drug Metabolism, Merck Research Laboratories, West Point, Pennsylvania
`
`V)
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`. 404
`
`I. Introduction .
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`II. Role of pharmacokinetics and metabolism in drug design .
`A. Metabolism and drug design .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`1. Hard drugs .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2. Soft drugs .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3. Active metabolites .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`B. Pharmacokinetics and drug design .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2. Prodrugs.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`1. Absorption .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`
`. 405
`. 405
`. 406
`. 407
`. 407
`. 407
`. 410
`. 408
`
`M
`|—‘
`I
`>
`E
`
`1
`
`U
`2
`
`§‘
`
`5.}
`3
`3
`g*
`
`I—]
`<
`U
`.
`"1
`O
`0
`V-1
`O
`U
`
`B. Species- and tissue—specific toxicity .
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`. 418
`
`. 411
`. 412
`. 412
`. 413
`. 413
`. 414
`. 415
`. 416
`. 417
`. 418
`. 418
`. 420
`. 420
`. 421
`. 423
`. 423
`. 423
`. 424
`. 425
`. 427
`

`
`$-
`
`w
`Z}

`3
`:3‘
`‘E
`9.1
`3


`E3

`°‘
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3. Distribution .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4. Plasma half~life .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`5. Stereoselectivity .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`III. Role of metabolism in drug toxicity .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`A. Species differences in metabolism .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`1. Oxidation and conjugation .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2. Induction .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3. Inhibition .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4. Sexual dimorphism .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`_
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`1. Species-specific toxicity.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2. Site-specific toxicity .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`C. Stereoselectivity and toxicity .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`1. Stereoselective metabolism .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2. Stereoselective toxicity .
`.
`.
`.
`.
`.
`.
`.
`IV. Role of pharmacokinetics and metabolism in drug development .
`A. In vitro studies of drug metabolism .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`1. Determination of metabolic pathways .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2. Identification of drug-metabolizing enzymes .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3. Drug-drug interactions .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`4. Prediction of in vivo metabolic clearance .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`B. In vitro studies of drug absorption .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`l. Extrapolation of in vitro absorption data .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`2. Extrapolation of animal absorption data .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`C. In vitro studies of protein binding .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`1. In vitro/in vivo protein binding .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`_
`.
`.
`.
`.
`.
`.
`.
`.
`_
`2. Plasma and tissue protein binding .
`.
`.
`.
`.
`.
`_
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3. Protein binding displacement interactions.
`V. Interindividual variability: a critical issue in drug development .
`A. Pharmacokinetic variability .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`1. Variability in absorption .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`_
`_
`.
`.
`.
`.
`.
`.
`.
`_
`_
`_
`.
`.
`.
`2. Variability in binding .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`3. Variability in excretion .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`B. Pharmacogenetics of drug‘ metabolism .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`1. Polymorphism in drug oxidation .
`.
`.
`.
`.
`.
`.
`.
`_
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`_
`.
`.
`.
`_
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`_
`.
`.
`.
`_
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`_
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`. 428
`. 428
`. 429
`. 430
`. 430
`. 431
`. 432
`. 434
`. 434
`. 434
`. 435
`. 436
`. 436
`. 436
`
`M
`
`L‘
`
`“ Address for correspondence: Jiunn H. Lin, WP26A-2044, Department of Drug Metabolism, Merck Research Laboratories, West Point, PA
`19486.
`403
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2028 - 0001
`
`

`
`404
`
`LIN AND LU
`
`.
`.
`2. N-acetylation polymorphism .
`.
`3. S—methylation polymorphism .
`4. Atypical butyrylcholinesterase .
`VI. Conclusions .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`VII. References .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`. ..
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`
`. .. 438
`. .. 439
`. .. 439
`. .. 439
`. .. 440
`
`I. Introduction
`
`Drug research encompasses several diverse disci-
`plines united by a common goal, namely the develop-
`ment of novel therapeutic agents. The search for new
`drugs can be divided functionally into two stages: dis-
`covery and development. The former consists of setting
`up a working hypothesis of the target enzyme or recep-
`tor for a particular disease, establishing suitable models
`(or surrogate markers) to test biological activities, and
`screening the new drug molecules for in vitro and/or in
`vivo biological activities. In the development stage, ef-
`forts are focused on evaluation of the toxicity and effi-
`cacy of new drug candidates. Recent surveys indicate
`that the average new chemical entity taken to market in
`the United States requires 10 to 15 years of research and
`costs more than $300 million.
`Once the target enzyme or receptor is identified, me-
`dicinal chemists use a variety of empirical and seIniem—
`pirical structure-activity relationships to modify the
`chemical structure of a compound to maximize its in
`vitro activity. However, good in vitro activity cannot be
`extrapolated to good in vivo activity unless a drug has
`good bioavailability and a desirable duration of action. A
`growing awareness of the key roles that pharmacokinet-
`ics and drug metabolism play as determinants of in vivo
`drug action has led many drug companies to include
`examination of pharmacokinetics and drug metabolism
`properties as part of their screening processes in the
`selection of drug candidates. Consequently, industrial
`drug metabolism scientists have emerged from their
`traditional supportive role in drug development to pro-
`vide valuable support in the drug discovery efforts.
`To aid in a discovery program, accurate pharmacoki-
`netic and metabolic data must be available almost as
`
`early as the results of the in vitro biological screening.
`Early pharmacokinetic and metabolic evaluation with
`rapid information feedback is crucial to obtain optimal
`pharmacokinetic and pharmacological properties. To be
`effective, the turnover rate needs to be at least three to
`five compounds per week for the support of each pro-
`gram. Due to time constraints and the availability of
`only small quantities of each compound in the discovery
`stage, studies are often limited to one or two animal
`species. Therefore, the selection of animal species and
`the experimental design of studies are important in pro-
`viding a reliable prediction of drug absorption and elim-
`ination in humans. A good compound could be excluded
`on the basis of results from an inappropriate animal
`species or poor experimental design.
`
`After a drug candidate is selected for further develop-
`ment, detailed information on the metabolic processes
`and pharmacokinetics of the new drug is required by
`regulatory agencies. The rationale for the regulatory
`requirement is best illustrated by the case of active
`metabolite formation. Many of the currently available
`psychotropic drugs form one or more metabolites that
`have their own biological activity (Baldessarini, 1990).
`Pharmacokinetically, the active metabolites may differ
`in distribution and clearance from that of the parent
`drug. Pharmacologically, the parent drug and its metab-
`olites may act by similar mechanisms, different mecha-
`nisms, or even by antagonism. An understanding of the
`kinetics of active metabolite formation is important not
`only for predicting therapeutic outcome, but also for
`explaining the toxicity of specific drugs.
`Conventionally, the metabolism of new drugs in hu-
`mans is studied in vivo using radiotracer techniques as
`part of clinical absorption and disposition studies. How-
`ever, this approach often occurs relatively late in the
`development stage. Ideally, the metabolism of new drugs
`should be studied in vitro before the initiation of clinical
`
`studies. Early information on in vitro metabolic pro-
`cesses in humans, such as the identification of the en-
`zymes responsible for drug metabolism and sources of
`potential enzyme polymorphism, can be useful in the
`design of clinical studies, particularly those that exam-
`ine drug-drug interactions. It is also desirable that the
`comparison of metabolism between animals and humans
`be performed in the early stage of the drug development
`process to provide information for the appropriate selec-
`tion of animal species for toxicity studies before these
`toxicity studies begin.
`The advance of in vitro enzyme systems used for drug
`metabolism studies (Wrighton and Stevens, 1992; Guil-
`louzo et al., 1993; Berry et al_, 1992; Remmel and Burch-
`ell, 1993; Brendel et al., 1990; Chapman et al., 1993),
`together with the explosion of our knowledge of various
`drug-metabolizing enzymes including uridine-diphos-
`phate-glucuronosyl-transferases (Cougletrie, 1992), cy-
`tochrome P-450s (Henderson and Wolf, 1992; Gonzalez
`and Nebert, 1990) and carboxylesterases (Wang, 1994;
`Hosokawa, 1990), allows us to obtain early information
`on the metabolic processes of new drug candidates well
`before the initial clinical studies. In addition, the advent
`of commercial liquid chromatography—mass spectrome-
`try instrumentation and the development of high-field
`nuclear magnetic resonance as well as liquid chromatog-
`raphy-nuclear magnetic resonance techniques have fur-
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2028 - 0002
`
`

`
`ROLE OF PHARMACOKINETICS AND METABOLISM IN DRUG RESEARCH
`
`405
`
`ther strengthened our capability to study the metabo-
`lism of new drugs in the early drug discovery stage
`(Fenselau, 1992; Baillie and Davis, 1993). However, the
`role of drug metabolism scientists in drug discovery is
`more than just screening compounds in vitro and in vivo.
`It really entails a good understanding of the basic mech-
`anisms of the events involved in absorption, distribu-
`tion, metabolism and excretion; the interaction of chem-
`icals with the drug-metabolizing enzymes, particularly
`cytochrome P-450; sources of pharmacokinetic and phar-
`macodynamic interindividual variability; and the conse-
`quences of metabolism on potential drug toxicities.
`The purpose of this paper is to review the role of
`pharmacokinetics and drug metabolism in drug discov-
`ery and development from an industrial perspective.
`The intent is to provide a comprehensive, rather than
`exhaustive, overview of the pertinent literature in the
`field. Several excellent review articles on individual top-
`ics are available and the reader is referred to the most
`
`recent articles in the text. It is hoped that with a better
`understanding of the fate of the drugs, a balanced in
`vitro/in vivo approach and an intelligent application of
`sound principles in pharmacokinetics and enzymology,
`drug metabolism scientists can contribute significantly
`to the development of safe and more efiicacious drugs.
`
`II. Role of Pharmacokinetics and Metabolism in
`
`Drug Design
`
`The history of the pharmaceutical industry shows that
`many important drugs have been discovered by a com-
`bination of fortuity and luck. This serendipity is best
`exemplified by the discovery of isoniazid. Isoniazid was
`first synthesized by Meyer and Mally (1912). Its antitu-
`berculosis properties were not found until 40 years later,
`when Robitzek et al.
`(1952) gave isoniazid to 92
`“hopeless” patients with progressive caseous—pneumonic
`pulmonary tuberculosis that had failed to show im-
`provement after any therapy. Furthermore, both indo-
`methacin and ibuprofen compounds were developed as
`antirheumatic agents even without any knowledge of
`their mode of action (Shen, 1972; Adams et al., 1969,
`1970). The mode of action was established several years
`after the drugs were on the market when Vane (1971)
`showed that these nonsteroidal anti-inflammatory
`drugs acted by inhibiting the synthesis of prostag1an—
`dins.
`
`Another example of serendipity is the discovery of
`anxiolytics. Diazepam and chlordiazepoxide, the most
`widely used henzodiazepines, were found to have anxio-
`lytic activity in 1958 and were marketed in 1960. Efforts
`to determine the mechanism of benzodiazepine action
`were initiated only after their introduction into the
`clinic. It was not until 1974 that convincing evidence
`from behavioral, electrophysiological, and biochemical
`experiments was accumulated to demonstrate that ben-
`zodiazepines act specifically at synapses in which y-ami-
`
`nobutyric acid (GABA)h functions as a neurotransmitter
`(Baldessarini, 1990; Haefely et al., 1985; Williams and
`Olsen, 1989).
`Over the past decades, through a better understand-
`ing of disease processes, mechanism-based drug design
`has evolved and produced drugs that interrupt specific
`biochemical pathways by targeting certain enzymes or
`receptors. This approach does not require a knowledge of
`the three-dimensional environment in which drugs act.
`Recent advances in molecular biology and protein chem-
`istry have provided pure protein in sufficient quantities
`to allow structural studies to be carried out. Visualiza-
`
`tion of these structures by sophisticated computer
`graphics has made structure-based drug design feasible.
`These rational approaches of drug design have been
`successful historically in the fields of HIV protease in-
`hibitors (Vacca et al., 1994), hepatic hydroxymethylglu-
`taryl coenzyme A reductase inhibitors (Alberts et al.,
`1980) and angiotensin—converting enzyme (ACE) inhibi-
`tors (Patchett et al., 1980).
`Today, the design of new drugs is still received by
`many medicinal chemists to mean maximization of the
`desired drug activity within certain structural limits.
`Sometimes, however, compounds that show very high
`activity in vitro may prove later to have no in vivo
`activity, or to be highly toxic in in vivo models. Lack of in
`vivo activity may be attributed to undesirable pharma-
`cokinetic properties, and the toxicity may result from
`the formation of reactive metabolites. Therefore, ratio-
`nal drug design should also take both pharmacokinetic
`and metabolic information into consideration, and the
`information should be incorporated with molecular bio-
`chemical and pharmacological data to provide well-
`rounded drug design.
`
`A. Metabolism and Drug Design
`
`From toxicological and pharmacological points of
`view, it is desirable to design a “safer” drug that under-
`goes predictable metabolic inactivation or even under-
`
`" Abbreviations: 3-MC, 3-methylcholanthrene; 6-TGN, 6-thiogua-
`nine nucleotide; ACE, angiotensin-converting enzyme; AFB, alfa-
`toxin B1; Ah, aromatic hydrocarbon; AUC, area under the curve;
`AZT, zidovudine; BBB, blood-brain barrier; CCKB cholecystokinin;
`cL, clearance; CL“ hepatic clearance; cL,,,,, intrinsic clearance; CNS,
`central nervous system; CSF, cerebrospinal fluid; DMBA, 7,12-di-
`methylbenzlalanthracene; DMBB, 5-(1,3-dimetliylbutyl)-5-ethyl bar-
`bituric acid; EM, extensive metabolize:-; fp, fraction of unbound drug
`in plasma; f,, free fraction in tissue; GABA, y-aminobutyric acid;
`GSH, glutathione; Ki, dissociation constant of an inhibitor; Kimcc,
`maximum inactivation rate constant; K,,,, Michaelis constant; K”,
`ratio of drug concentration in tissue to that in plasma after drug
`administration; L-dopa, levodopa; MPH, rnethylphenidate; NAT, N-
`acetyltransferase; NSAID, nonsteroidal anti-inflammatory agent;
`PEG, polyethelene glycol; PFDA, perfluorodecanoic acid; PM, poor
`metabolizers; PPAR, peroxisome proliferator-activated receptors;
`TMT, thio methyltransferase; TPMT, thiopurine methyltransferase;
`UDPGT, uridine diphosphoglucose transferase; Vd, volume of distri-
`bution; V,—, velocity of an enzyrnic reaction in the presence of of
`inhibitor extensive metabolizers; V,,,,,,,, maximum velocity; V0, veloc-
`ity of an enzymic reaction in the absence of inhibitor.
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2028 - 0003
`
`

`
`406
`
`LIN AND LU
`
`goes no metabolism. Several approaches have been used
`for the design of safer drugs.
`1. Hard drugs. The concept of nonmetabolizable
`drugs, or so-called hard drugs, was proposed by Ariéns
`(1972) and Ariéns and Simonis (1982). The hard drug
`design is quite attractive. Not only does it solve the
`problem of toxicity due to reactive intermediates or ac-
`tive metabolites, but the pharmacokinetics also are sim-
`plified because the drugs are excreted primarily through
`either the bile or kidney. If a drug is excreted mainly by
`the kidney, the differences in the elimination between
`animal species and humans will be dependent primarily
`on the renal function of the corresponding species giving
`highly predictable pharmacokinetic Profiles using the
`allometric approach (Lin, 1995; Mordenti, 1985). A few
`successful examples of such hard drugs include bisphos—
`phonates and certain ACE inhibitors.
`Bisphosphonates are a unique class of drugs. As a
`class, they are characterized pharmacologically by their
`ability to inhibit bone resorption, whereas pharmacoki-
`netically, they are classified by their similarity in ab-
`sorption, distribution and elimination. In the clinic,
`these drugs are used in patients as antiosteolytic agents
`for the treatment of a broad range of bone disorders
`characterized by excessive bone resorption. These in-
`clude hypercalcemia of malignancy, metastatic bone dis-
`ease, Paget’s disease, and osteoporosis.
`The discovery of bisphosphonates was based on earlier
`studies of inorganic pyrophosphate by Fleisch and his
`coworkers (Fleisch et al., 1966, 1968, 1969; Fleisch and
`Russell, 1970). They found that pyrophosphate bound
`very strongly to calcium phosphate and inhibited not
`only the formation of calcium phosphate crystals, but
`also the crystal dissolution in vitro. However, pyrophos-
`phate exhibited no effect on bone resorption in vivo. This
`was later explained by the observation that pyrophos-
`phate is hydrolyzed before it reaches the site of bone
`resorption. These findings led to a search for analogs
`that would display the activities similar to pyrophos-
`phate, but would also resist enzymatic hydrolysis. It
`was found that the bisphosphonates, characterized by a
`P—O—P bond rather than the P—O—P bond of pyrophosphate,
`iiilfilled these criteria. As hard drugs, bisphosphonates are
`not metabolized in animals or humans, and the only route
`of elimination is renal excretion (Lin et al., 1991c; Lin,
`1996a). In general, these compounds are very safe with no
`significant systemic toxicity (Fleisch, 1993).
`Similarly, enalaprilat and lisinopril are considered
`hard drugs. These two ACE inhibitors undergo very
`limited metabolism and are exclusively excreted by the
`kidney (Ulm et al., 1982; Tocco et al., 1982; Lin et al.,
`1988). Unlike sulfhydryl-containing ACE inhibitors,
`such as captopril and its analogs, neither enalaprilat nor
`lisinopril exhibits significant side effects (Kelly and
`0’Malley, 1990). The most common side effects accom-
`panying the clinical use of captopril are rashes and taste
`dysfunction (Atkinson and Robertson, 1979; Atkinson et
`
`al., 1980). Similar side effects are observed with penicil-
`lamine, which is a sulflriydryl-containing heavy metal
`antagonist used extensively in the treatment of Wilson’s
`disease (Levine, 1975; Suda et al., 1993). It is therefore
`speculated that captopril
`interacts with endogenous
`sulfhydryl-containing proteins to form disulfides that
`may act as haptens, resulting in immunological reactiv-
`ity, which may be responsible for these side effects
`(Patchett et al., 1980). Enalaprilat and lisinopril were
`designed to avoid these undesirable side effects by re-
`moval of the sulfhydryl group (Patchett et al., 1980).
`Due to their poor lipophilicity, the bisphosphonates,
`enalaprilat and lisinopril, are not metabolized in vivo.
`Ironically, the poor lipophilicity of these compounds re-
`sults in poor oral absorption. For the bisphosphonate
`alendronate, the octanol/buffer partition coefficient is
`0.0017 (Lin, 1996a). As a result of its poor lipophilicity,
`alendronate has very poor oral bioavailability in hu-
`mans (< 1%) (Lin, 1996a). To our knowledge, bisphos—
`phonates are the only class of drugs being developed for
`oral dosage in spite of their poor bioavailability (Lin,
`1996a). This is because the systemically available
`bisphosphonates are largely taken up by the target
`(bone) tissues, where their elimination is very slow (Lin,
`1996a, 1992, 1993b). The half-life of alendronate in bone
`was estimated to be at least 10 years in humans.
`Like bisphosphonates, both enalaprilat and lisinopril
`have low lipophilicity. The octanol-to-water partition co-
`efficient is approximately 0.003 for both drugs (Ondetti,
`1988). Interestingly, enalaprilat, a diacid compound
`with a net negative charge, is poorly absorbed (<10%),
`whereas lisinopril, a zwitterionic compound, has accept-
`able oral absorption (~30%) (Ulm et al., 1982; Tocco et
`al., 1982). Consequently, enalaprilat was developed as
`its ethyl ester prodrug (enalapril) to increase its bio-
`availability, whereas the prodrug approach was not em-
`ployed for lisinopril.
`Bisphosphonates and these two carboxyalkyldipeptide
`ACE inhibitors were not intentionally designed as hard
`drugs. The “hardness” came about only as a result of
`structural improvement. It so happens that the newer
`ACE inhibitors, such as benazepril, perindopril, and
`fosinopril, undergo significant metabolism (Kelly and
`O’Ma1ley, 1990).
`Although metabolically inert compounds are highly
`desirable candidates for drug design, the versatility of
`the drug-metabolizing enzymes presents quite a chal-
`lenge to achieve this goal. For example, cytochrome
`P-450s are known to catalyze numerous oxidative reac-
`tions involving carbon, oxygen, nitrogen, and sulfur at-
`oms in thousands of substrates with diverse structures.
`
`In addition, cytochrome P-4503 are unique in that met-
`abolic switchings can occur when the primary metabolic
`site of a compound is blocked. Thus, considering the
`broad substrate specificities and the versatilities of cy-
`tochrome P-450s and other drug-metabolizing enzymes,
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2028 - 0004
`
`

`
`ROLE OF‘ PHARMACOKINETICS AND METABOLISM IN DRUG RESEARCH
`
`407
`
`designing drug candidates that are metabolically inert
`may not always be feasible.
`2. Soft drugs. In contrast to the concept of hard drugs,
`Bodor (1984, 1982) and Bodor et al. (1980) have proposed
`the approach of soft drugs. A soft drug is pharmacolog-
`ically active as such, and it undergoes a predictable and
`controllable metabolism to nontoxic and inactive metab-
`
`olites. The main concept of soft drug design is to avoid
`oxidative metabolism as much as possible and to use
`hydrolytic enzymes to achieve predictable and control-
`lable drug metabolism. Most oxidative reactions of drugs
`are mediated by hepatic cytochrome P—450 enzyme sys-
`tems that are often affected by age, sex, disease, and
`environmental factors, resulting in complex biotransfor-
`mation and pharmacokinetic variability (Hunt et al.,
`1992; Soons et al., 1992). In addition, P-450 oxidative
`reactions have the potential to form reactive intermedi-
`ates and active metabolites that can mediate toxicity
`(Guengerich and Shimada, 1993). These undesirable ef-
`fects attributed to oxidative metabolism may be circum-
`vented to some extent by incorporating metabolic struc-
`tural “softness.”
`
`Bodor and his colleagues (Bodor, 1984, 1982; Bodor et
`al., 1980) have designed soft quaternary—type drugs con-
`taining three structural components: an acidic group, an
`aldehyde, and a tertiary amine. Upon absorption, the
`soft quaternary drugs are hydrolyzed to three nontoxic
`components that are rapidly eliminated from the body.
`Atracurium, a nondepolarizing muscle relaxant, can
`be considered a soft drug. This drug contains quaternary
`N—functions and ester groups. Atracurium is metabo-
`lized in vivo by two nonoxidative processes: a nonenzy—
`matic metabolism by Hofmann-degradation to form a
`tertiary amine and an alkene, and hydrolysis of the ester
`groups by esterases (Mutschler and Derendorf, 1995;
`Hughes and Chapple, 1981).
`Remifentanil, a novel short-acting p.—opioid receptor
`agonist, may also be considered a soft drug. This drug is
`a methyl ester and is metabolized extensively by ester-
`ases to an inactive acid metabolite, GI—90291, of which
`over 90% is subsequently recovered in urine. To a much
`lesser extent, the drug also is metabolized by N-dealky-
`lation to a second metabolite, GI-94219 (Feldman et al.,
`1991; Biirkle et al., 1996; Glass, 1995). The major me-
`tabolite GI—90291 is approximately 2000- to 4000—fold
`less potent compared with remifentanil. Although both
`hard and soft drug designs are of academic interest,
`there are only a few successful examples in the drug
`market.
`
`3. Active metabolites. For many years, the process of
`biotransformation was considered synonymous with the
`inactivation of pharmacologically active compounds.
`There is increasing evidence, however, that the metab-
`olites of some drugs are pharmacologically active. Nu-
`merous examples of pharmacologically active metabo-
`lites being used as a source of new drug candidates exist
`
`because these metabolites often are subject to phase II
`reactions and have better safety profiles.
`Perhaps the best known example is acetaminophen,
`which is an O-deethylated metabolite of phenacetin.
`Acetaminophen shows superior analgesic activity when
`compared with phenacetin. The main advantage of acet-
`aminophen over phenacetin is that it does not produce
`methemoglobinemia and hemolytic anemia (Flower et
`al., 1985). Phenacetin is converted to at least 1 dozen
`metabolites by O-deethylation, N—deacetylation, and hy-
`droxylation processes. N—hydroxyphenatidine, a metab-
`olite of phenacetin, has been shown to be responsible for
`the formation of methemoglobin and hemolysis of red
`blood cells (Jensen and Jollow, 1991). Conversely, acet-
`aminophen primarily undergoes glucuronidation and
`sulfation exclusively and is quite safe clinically at the
`recommended dose. Similarly, the analgesic oxyphen-
`butazone is an active para—hydroxy metabolite of phe-
`nylbutazone. Similar to acetaminophen, this active me-
`tabolite also shows better analgesic activity than
`phenylbutazone and causes less gastric irritation (Flow-
`er et al., 1985).
`Although pharmacologically active metabolites are
`generally formed by phase I oxidative reactions, phase II
`conjugation reactions also can produce biologically ac-
`tive metabolites. Morphine 6—glucuronide is more potent
`as a pxopioid receptor agonist than morphine itself (Paul
`et al., 1989; Mulder, 1992). Recent clinical studies in
`cancer patients given morphine 6-glucuronide indicated
`that useful analgesic effects are achieved without the
`side effects of nausea and vomiting that are often asso-
`ciated with morphine (Osborne et al., 1992). These find-
`ings have led to the commercial marketing of morphine
`6—glucuronide. Sulfation also produces biologically ac-
`tive metabolites. Minoxidil, a potent vasodilator,
`is a
`good example. Studies concerning the action of minoxidil
`revealed that the therapeutic activities were mediated
`by its sulfate conjugate (Bray and Quast, 1991).
`In addition to the advantages that active metabolites
`may have in terms of efficacy with fewer unwanted side
`effects, active metabolites can also be preferred over the
`parent drugs for kinetic reasons. Many benzodiazepines
`form active metabolites with similar pharmacological
`properties. Oxazepam is the common active metabolite
`of chlordiazepoxide, halazepam, chlorazepate, and diaz-
`epam (Caccia and Garattini, 1990). Unlike other benze-
`diazepines, oxazepam undergoes only glucuronidation
`and has a shorter half-life than any of its precursors.
`This kinetic advantage has led to the marketing of ox-
`azepam as a short—acting benzodiazepine in the treat-
`ment of sleeping disorders (Baldessarini, 1990).
`
`B. Pharmacokinetics and Drug Design
`
`Many of the failures of drug candidates in develop-
`ment programs are attributed to their undesirable phar-
`macokinetic properties, such as too long or too short tn/2,
`poor absorption, and extensive first-pass metabolism. In
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2028 - 0005
`
`

`
`408
`
`LIN AND LU
`
`a survey, Prentis et al. (1988) reported that of 319 new
`drug candidates investigated in humans, 77 (40%) of the
`198 candidates were withdrawn due to serious pharma-
`cokinetic problems. This high failure rate illustrates the
`importance of pharmacokinetics in drug discovery and
`development.
`To ensure the success of a drug’s development, it is
`essential that a drug candidate has good bioavailability
`and a desirable tie. Therefore, an acc

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