throbber
WILSON AND G/SVOLD’S
`
`Textbook of Organic Medicinal
`and Pharmaceutical Chemistry
`
`N/nth Edit/on
`
`
`
`EDITED BY
`
`Jaime N. Delgado, Ph.D.
`Division of Medicinal Chemistry, College of Pharmacy
`University of Texas at Austin, Austin, Texas
`
`AND
`
`William A. Fiemers, Ph. D.
`
`Department of Pharmaceutical Sciences, College of Pharmacy
`University of Arizona, Tucson, Arizona
`
`17 Contributors
`
` J. B. Lippincott Company
`
`New York
`
`London Hagerstown
`
`Philadelphia
`
`EXI-||B|T
`ACTAVIS, AMNEAL,
`AUROBWDO,
`BRECKENRIDGE,
`VENNOOT,
`SANDOZ, SUN
`
`|PR2014-01126-1028 p. 1
`
`IPR2014-01126-Exhibit 1028 p. 1
`
`

`
`Production Manager: Janet Greenwood
`Acquisitions Editor: Lisa McAllister
`Manuscript Editor. Marguerite Hague
`Production: Till 8. Till, lnc.
`Compositor: Science Typographers, inc
`Printer/Binder: The Murray Printing Company
`
`Ninth Edition
`
`Copyright © 1991, by J. B. Lippincott Company
`Copyright © 1982 by J. B Lippincott Company
`Copyright © 1977, 1971, 1966, 1962, 1956 by J. B. Lippincott Company
`Copyright © 1954 by J. B. Lippincott Company
`All rights reserved. No part of this book may be used or reproduced in any manner whatsoever without written
`permission except
`for brief quotations embodied in critical articles and reviews Printed in the United States of
`America. For information write J. B. Lippincott Company, East Washington Square, Philadelphia, Pennsylvania 19105.
`654321
`
`Library of Congress Cataloging in Publication Data
`Wilson and Gisvold‘s textbook of organic medicinal and pharmaceutical
`chemistry. ~~ 9th ed. / edited by Jaime N. Delgado and William A.
`Flemers : 17 contributors.
`p.
`cm.
`Includes bibliographical references.
`includes index.
`lSBN 0-397-50877~8
`2. Chemistry, Organic.
`1. Chemistry, Pharmaceutical.
`Charles Owens, 1911 —
`.
`ll. Gisvold, Ole, 1904-
`Jaime N.
`lV. Remers, William A. (William Alan), 1932—
`V. Title: Textbook of organic medicinal and pharmaceutical
`chemistry.
`1. Chemistry, Pharmaceutical. QV 744 W754]
`[DNLM.
`1991
`RS403.T43
`615'.19~dc2O
`DNLM / DLC
`for Library of Congress
`
`I. Wilson,
`.
`Ill. Delgado,
`
`90-13652
`ClF’
`
`The authors and publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are
`in accord with current recommendations and practice at the time of publication. However, in view of ongoing research,
`changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions,
`the reader is urged to check the package insert for each drug for any change in indications and dosage and for
`added warnings and precautions. This is particularly important when the recommended agent is a new or infrequently
`employed drug.
`
`|PR2014—01126—EXhibit 1028 p. 2
`
`IPR2014-01126-Exhibit 1028 p. 2
`
`

`
`
`
`CHAPTER’ 2
`
`Physicochemical Properties
`in Relation to Biologic Action
`
`John H. B/ock
`
`INTRODUCTION
`
`Modern drug design, as compared with, Let’s make
`a change on an existing compound or synthesize a
`new structure and see what happens,
`is a fairly
`recent discipline still in its infancy. It is based on
`modern chemical techniques utilizing recent knowl-
`edge of disease mechanisms and receptor properties.
`A good understanding of how the drug is trans-
`ported into the body, distributed throughout the
`body compartments, metabolically altered by the
`liver and other organs, and excreted from the pa-
`tient is required, along with the structural charac-
`teristics of the receptor. Acid—base chemistry is uti-
`lized to aid in formulation and biodistribution. Those
`
`structural attributes and substituent patterns re-
`sponsible for optimum pharmacologic activity can be
`predicted many times by statistical techniques, such
`as regression analysis. Conformational analysis per-
`mits the medicinal chemist
`to predict the drug’s
`three-dimensional shape that is seen by the recep-
`tor. With the isolation and structural determination
`
`of specific receptors and the availability of computer
`software that can estimate the three-dimensional
`
`is possible to design
`it
`shape of the receptor,
`molecules that will show an optimum fit
`to the
`receptor.
`
`HISTORY
`
`Initially, drugs were extracted from plant sources to
`obtain agents such as digitalis, quinine, and mor-
`phine, medicinal agents that are still in use today.
`Specific plants are selected by the chemist because
`the crude preparations were being used for treat-
`ment of medical conditions by the local population
`where the plant grew.
`
`Early drug design started with elucidation of the
`structure of the natural product, followed by selec-
`tive changes in the molecule. The latter was done
`for many reasons,
`including the reduction of an
`undesirable pharmacologic response (side effect); ob-
`taining a better pharmacokinetic response; altering
`the drug’s metabolism; securing a more plentiful,
`less costly supply; and producing a competing prod-
`uct. Let us use the morphine alkaloid as an example.
`Literally thousands of compounds have been synthe-
`sized in an attempt to separate the desired analgesia
`from the undesirable addiction liability. This
`tremendous effort in numerous research laborato-
`
`ries, over many years and involving many scientists,
`had been minimally successful until a better un-
`derstanding of the opiate receptor developed (see
`Chap. 9).
`In other examples, there has been good success at
`this empirical approach. Alteration of the cocaine
`structure has led to the very successful local anes-
`thetics that lack cocaine’s undesirable central effects
`
`(see Chap. 15). In contrast with this success story,
`there have been no significant commercial synthetic
`replacements for digitalis or colchicine.
`Synthetic medicinal chemistry as a discipline be-
`came more intense in the 19005, but many of the
`so—called principal compounds still were based on a
`natural product, a fortuitous observation, or an un-
`suspected chemical reaction. The phenothiazines (see
`Chap. 9) were first synthesized as antihistamines,
`but a careful pharmacologic evaluation led to their
`use as major tranquilizing agents that revolution-
`ized the care of the severely mentally ill patient. The
`benzodiazepines (see Chap. 9) originated from an
`unexpected ring enlargement and resulted in a very
`important group of central nervous system relax-
`ants.
`
`Economic factors have stimulated the type of sci-
`entific investigations required to carry out focused
`
`3
`
`IPRZO14-01126-Exhibit 1028 p. 3
`
`IPR2014-01126-Exhibit 1028 p. 3
`
`

`
`4 |
`
`PHYSICOCHEMICAL PROPERTIES IN RELATION TO BIOLOGICAL ACTION
`
`new drug development. It has become increasingly
`costly to develop a new drug that will be approved
`by the U. S. Food and Drug Administration (FDA).
`At one time, safety was the main criterion for FDA
`approval. Today, demonstration of efficacy is an es-
`sential requirement, along with safety considera-
`tions. This has led to (1) increased basic research on
`
`the disease process for which a drug treatment is
`sought, (2) mathematically modeling the pharma-
`cokinetics of the drug’s distribution, (3) elucidation
`of the biochemistry of the pharmacologic response
`from the drug, (4) learning the metabolic fate of the
`drug, (5) defining those specific structural character-
`istics of the drug responsible for the desired phar-
`macologic response, and (6), where possible, visualiz-
`ing the structural characteristics of the receptor.
`Although the number of new compounds introduced
`annually has decreased from earlier years, the prod-
`ucts now coming into use are showing dramatic
`effects in the treatment of disease. More impor-
`tantly, because of the intensive background investi-
`
`gation that has led to the design of today’s new
`agents, a better understanding of the drug’s mecha-
`nism of action is known. Indeed, this is an exciting
`time to practice pharmacy.
`
`OVERVIEW
`
`A drug is a chemical molecule. Following introduc-
`tion into the body, a drug must pass through many
`barriers, survive alternate sites of attachment and
`storage, and avoid significant metabolic destruction
`before it reaches the site of action, usually a recep-
`tor on or in a cell (Fig. 2-1). At the receptor, the
`following equilibrium usually holds.
`
`Drug + Receptor : Drug—Receptor Complex
`l
`Pharmacologic Response
`(Rx. 2-1)
`
`Oral
`Administration
`
`Gastrointestinal
`Tract
`
`Intramuscular
`or
`Subcutaneous
`
`Injection
`
`Intravenous
`Injection
`
`Receptors
`for
`Desired
`Effects
`
`
`
`DRUG
`
`DRUG
`
`DRUG
`
`DRUG-DRUG METABOLITES
`
`onus ——¢_~__.——-
`
`svsremc cmcuumou
`
`DRUG
`
`DRUG METABOLITES
`
`DRUG-DRUG HETABOLITES
`
`DRUG-DRUG METABOLITES
`
`DRUG-DRUG HETABOLITES
`
`
`
`i‘Liver:
`
`
`bile
`site of most drug metabolism l —-~—>
`Intestinal
`Receptors
`duct
`Tract
`for
`Undesired
`
`Feces
`
`Excretion of DRUG—DRUG
`METABOLITES
`
`Effects
`
`
`
`—— Drug administered directly into systemic circulation.
`
`FIG. 2-1. Summa/y of drug d/smbunon: So/id bars: Drug must pass lhrough membranes. Broken /mes: Drug adm/nis[ered directly /nlo
`systemic circulation.
`
`|PR201401126EXhmH1028p.4
`
`IPR2014-01126-Exhibit 1028 p. 4
`
`

`
`The ideal drug molecule will show favorable-binding
`characteristics to the receptor, such that the equilib-
`rium lies to the right. At the same time the drug will
`be expected to dissociate from the receptor and reen-
`ter systemic circulation to be excreted. The major
`exceptions include the alkylating agents used in can-
`cer chemotherapy (see Chap. 8) and a few inhibitors
`of the enzyme, acetylcholinesterase (see Chap. 12).
`Both of these subclasses of pharmacologic agents
`form covalent bonds with the receptor.
`In these
`cases the cell must destroy the receptor or, as with
`the alkylating agents,
`the cell would be replaced,
`ideally with a normal cell. In other words, the usual
`use of drugs in medical treatment call for the drug’s
`effect to last for only a finite period. Then, if it is to
`be repeated, the drug will be administered again. If
`the patient does not tolerate the drug well, it is even
`more important that the agent dissociate from the
`receptor and be excreted from the body.
`
`DRUG DISTRIBUTION
`
`ORAL ADMINISTRATION
`
`An examination of the obstacle course (see Fig. 2-1)
`faced by the drug will give a better understanding of
`what is involved in developing a commercially feasi-
`ble product. Assume that the drug is administered
`orally. The drug must go into solution for it to pass
`through the gastrointestinal mucosa. Even drugs
`administered as true solutions may not remain in
`solution as they enter the acidic stomach and then
`pass into the alkaline intestinal tract. (This will be
`explained further in the discussion on acid—base
`chemistry.) The ability of the drug to dissolve is
`governed by several factors, including its chemical
`structure, variation in particle size and particle sur-
`face area, nature of the crystal form, type of coating,
`and type of tablet matrix. By varying the formula-
`tion containing the drug and physical characteristics
`of the drug, it is possible to have a drug dissolve
`quickly or slowly, the latter being the situation for
`many of the sustained-action products. An example
`is orally administered sodium phenytoin; for which
`variation of both the crystal form and tablet adju-
`vants can significantly alter the bioavailability of
`this drug, which is widely used in the treatment of
`epilepsy.
`Chemical modification is also used to a limited
`
`extent. For example, sulfasalazine, used in the treat-
`ment of ulcerative colitis, passes through a substan-
`tial portion of the intestinal tract before being me-
`tabolized to sulfapyridine and 5-aminosalicylic acid.
`The latter compound is believed to be the active
`agent for the treatment of ulcerative colitis.
`
`DRUG DISTRIBUTION I
`
`5
`
`O
`
`N
`Cl?
`HOCHJ
`HOICINNQ/H<n>
`
`__
`
`‘T
`
`\
`
`Sulfasalazine
`
`O
`
`Any compound passing through the gastrointesti-
`nal tract will encounter the many and various diges-
`tive enzymes that, in theory, can degrade the drug
`molecule. In practice, when a new drug entity is
`under investigation, it will probably be dropped from
`further consideration if it is found unable to survive
`
`in the intestinal tract. An exception would be a drug
`for which there is no other effective product avail-
`able, or one that provides a more effective treatment
`over existing products and can be administered by
`an alternate route, usually parenteral.
`In contrast, these same digestive enzymes can be
`used to advantage. Chloramphenicol is water-soluble
`enough that it comes in contact with the taste recep-
`tors on the tongue, producing an unpalatable bitter-
`ness. To mask this intense bitter taste, the palmitic
`acid moiety is added as an ester of the chloram-
`phenicol’s primary alcohol. This reduces the parent
`drug’s water solubility so much that it can be for-
`mulated as a suspension that passes over the bitter
`taste receptors on the tongue. Once in the intestinal
`tract, the ester linkage is hydrolyzed by the digestive
`esterases to the active antibiotic, chloramphenicol,
`and the very common dietary fatty acid, palmitic
`acid.
`
`NHCOCHC5
`I
`
`o2N—«<::>»-cHcHcH2oR
`
`OH
`
`Chloramphenicol: R = H;
`
`‘P
`Chloramphenicol Palmitale: R = C(CH2),4CH3
`
`Sulfasalazine and chloramphenicol palmitate are
`examples of prodrugs. Most prodrugs are com-
`pounds that are inactive in their native form, but
`are easily metabolized to the active agent. Sulfasala-
`zine and chloramphenicol palmitate are examples of
`prodrugs that are cleaved to smaller compounds, one
`of which will be the active drug. Others are metabolic
`precursors to the active form. An example of this
`type of prodrug is menadione, a simple naph-
`thoquinone, which is converted in the liver
`to
`vitamin K290).
`
`IPRZO14-01126-Exhibit 1028 p. 5
`
`
`IPR2014-01126-Exhibit 1028 p. 5
`
`

`
`6
`
`I
`
`PHYSICOCHEMICAL PROPERTIES IN RELATION TO BIOLOGICAL ACTION
`
`0 0
`
`CH3
`
`CH3
`
`CH3
`Vitamin K 2,20)
`
`CH3
`
`Occasionally the prodrug approach is used to en-
`hance the absorption of a drug that is poorly ab-
`sorbed from the gastrointestinal tract. Enalapril is
`the ethyl ester of enalaprilic acid, an active inhibitor
`of angiotensin—converting enzyme. The ester pro-
`drug is much more readily absorbed orally than the
`carboxylic acid.
`
`OOH
`O\\/
`H
`i
`NT N
`C\ ::
`CH3
`
`O
`EnaIapriIiC Acid: R -— H
`
`R\
`
`,C\\
`
`O
`‘ Enalapril: R = CZHS;
`
`Unless the drug is intended to act locally in the
`gastrointestinal tract, it will have to pass through
`the gastrointestinal mucosa barrier into the venous
`circulation to reach the receptor site. This involves
`distribution, or partitioning, between the aqueous
`environment of the gastrointestinal tract, the lipid
`bilayer cell membrane of the mucosa cells, possibly
`the aqueous interior of the mucosa cells, the lipid
`bilayer membranes on the venous side of the gas-
`trointestinal tract, and the aqueous environment of
`venous circulation. Some very lipid—soluble drugs
`may follow the route of dietary lipids by becoming
`part of the mixed micelles, passing through the
`mucosa cells into the thoracic duct of the lymphatic
`system and then into the venous circulation.
`The drug’s passage through the mucosa cells can
`be passive or active. As will be discussed later in this
`chapter, the lipid membranes are very complex, with
`a highly ordered structure. Part of this membrane is
`a series of channels or tunnels that form, disappear,
`and reform. There are receptors that move com-
`pounds into the cell by a process called pinocytosis.
`
`Most drug molecules are too large to enter the cell
`by an active transport mechanism through the pas-
`sages. On the other hand, some drugs do resemble a
`normal metabolic precursor or intermediate, and
`they can be actively transported into the cell.
`
`PAFIENTEFIAL ADMINISTRATION
`
`Many times there will be therapeutic advantages to
`bypass the intestinal barrier by using parenteral
`(injectable) dosage forms; for example, because of
`illness, the patient cannot tolerate or is incapable of
`accepting drugs orally. Some drugs are so rapidly
`and completely metabolized to inactive products in
`the liver (first-pass effect) that oral administration is
`precluded. But that does not mean that the drug
`administered by injection is not confronted by obsta-
`cles (see Fig. 2-1). Intravenous administration places
`the drug directly into the circulatory system, from
`which it will be rapidly distributed throughout the
`body, including tissue depots and the liver, in which
`most biotransformations occur,
`in addition to the
`receptors.
`It is possible to inject the drug directly into spe-
`cific organs or areas of the body. Intraspinal and
`intracerebral routes will place the drug directly into
`the spinal fluid or brain, respectively, bypassing a
`specialized tissue,
`the blood-brain barrier, which
`protects the brain from exposure to diverse metabo-
`lites and chemicals. The blood—brain barrier is com-
`posed of membranes of tightly joined epithelial cells
`lining the cerebral capillaries. The net result is that
`the brain is not exposed to the same variety of
`compounds that other organs are. Local anesthetics
`are examples of administration of a drug directly on
`the desired nerve. A spinal block is a form of anes-
`thesia performed by injecting a local anesthetic di-
`rectly into the spinal cord at a specific location to
`block transmission along specific neurons.
`
`IPR2014—01126-Exhibit1028 p. 6
`
`IPR2014-01126-Exhibit 1028 p. 6
`
`

`
`Many of the injections a patient will experience in
`a lifetime will be subcutaneous or intramuscular.
`
`These parenteral routes produce a depot in the tis-
`sues (see Fig. 2.1) from which the drug must reach
`the blood or lymph to produce systematic effects.
`Once in systemic circulation, the drug will undergo
`the same distributive phenomena as orally and in-
`travenously administered agents.
`In general,
`the
`same factors that control the drug’s passage through
`the gastrointestinal mucosa will also determine the
`rate of movement out of the tissue depot.
`The prodrug approach also can be used to alter
`the solubility characteristics which,
`in turn, can
`increase the flexibility in possible dosage forms. The
`solubility of methylprednisolone can be altered from
`essentially water-insoluble methylprednisolone ac-
`etate,
`to slightly water—insoluble methylpred-
`nisolone,
`to water-soluble methylprednisolone
`sodium succinate. The water-soluble sodium succi-
`
`nate salt is used in oral, intravenous, and intramus-
`cular dosage forms. Methylprednisolone,
`itself,
`is
`normally found in tablets. The acetate ester is found
`in topical ointments and sterile aqueous suspensions
`for intramuscular injection. Both the succinate and
`acetate esters are hydrolyzed to the active methyl-
`prednisolone by the patient’s own systemic hydro-
`lytic enzymes.
`
`
`
`(EH,
`Methylprednisolone (R = H)
`Ester Available:
`
`Methylprednisolone Acetate: R = COCH3
`Salt Available:
`Methylprednisolone Sodium Succinate:
`
`R = COCH2CH2COO‘Na*
`
`PROTEIN BINDING
`
`Once the drug enters the systemic circulation (see
`Fig. 2-1) it can undergo several events. It may stay
`in solution, but many drugs will be bound to the
`serum proteins, usually albumin. Thus, a new equi-
`librium must be considered. Depending on the equi-
`librium constant, the drug can remain in systemic
`circulation bound to albumin for a considerable pe-
`riod and be unavailable to the sites of biotransfor-
`
`mation, the pharmacologic receptors, and excretion.
`
`onus DISTRIBUTION I
`
`7
`
`Drug + Albumin : Drug—Albumin Complex
`
`(Rx. 2-2)
`
`The effect of protein binding can have a profound
`result on the drug’s effective solubility, biodistribu-
`tion, half-life in the body, and interaction with other
`drugs. A drug with such poor water solubility that
`therapeutic concentrations of the unbound (active)
`drug normally cannot be maintained, still can be a
`very effective agent. The albumin—drug complex acts
`as a reservoir by providing concentrations of free
`drug large enough to cause a pharmacologic re-
`sponse.
`
`Protein binding may also limit access to certain
`body compartments. The placenta is able to block
`passage of proteins from maternal to fetal circula-
`tion. Consequently, drugs that normally would be
`expected to cross the placenta barrier and possibly
`harm the fetus are retained in the maternal circula-
`
`tion bound to the mother’s serum proteins.
`Protein binding also can prolong the drug’s dura-
`tion of action. The drug—protein complex is too large
`to pass through the renal glomerular membranes,
`preventing rapid excretion of the drug. Protein bind-
`ing limits the amount of drug available for‘biotrans—
`formation (see later and Chap. 3) and for interaction
`with specific receptor sites. For example the try-
`panocide, suramin,
`remains in the body in the
`protein-bound form as long as three months. The
`maintenance dose for this drug is based on weekly
`administration. At first this might seem to be an
`advantage to the patient. It can be, but it also means
`that should the patient have serious adverse reac-
`tions,
`it will require a substantial length of time
`before the concentration of drug falls below toxic
`levels.
`
`The drug~protein-binding phenomenon can lead
`to some interesting drug—drug interactions, result-
`ing when one drug displaces another from the bind-
`ing site on albumin. Diverse drugs can displace the
`anticoagulant, warfarin, from its albumin-binding
`sites. This increases the effective concentration of
`
`leading to an increased
`warfarin at the receptor,
`prothrombin time (increased time for clot forma-
`tion) and potential hemorrhage.
`
`TISSUE DEPOTS
`
`The drug also can be stored in tissue depots. Neutral
`fat constitutes some 20% to 50% of body weight and
`constitutes a depot of considerable importance. The
`more lipophilic the drug is, the more likely it will
`concentrate in these pharmacologically inert depots.
`The short-acting, lipophilic barbiturate, thiopental,
`reportedly disappears
`into tissue protein,
`redis-
`
`|PR2014-01 126-Exhibit 1028 p. 7
`
`
`IPR2014-01126-Exhibit 1028 p. 7
`
`

`
`3
`
`I
`
`PHYSICOCHEMICAL PROPERTIES IN RELATION TO BIOLOGICAL ACTION
`
`tributes into body fat and, then, slowly diffuses back
`out of the tissue depots, but in concentrations too
`low for any pharmacologic response. Hence, only the
`initially administered thiopental is present in high
`enough concentrations to combine with its recep-
`tors. In general, structural changes in the barbitu-
`rate series (see Chap. 9) that favor partitioning into
`the lipid tissue stores decreases the duration of
`action, but increases central nervous system depres-
`sion. Conversely, the barbiturates with the slowest
`onset and longest duration of action contain the
`more polar side chains. This latter group of barbitu-
`rates both enters and leaves the central nervous
`
`system very slowly, as compared with the more
`lipophilic thiopental.
`
`DRUG METABOLISM
`
`including drugs, metabolites, and
`All substances,
`nutrients that are in the circulatory system, will
`pass through the liver. Most molecules absorbed
`from the gastrointestinal tract will enter the portal
`vein and be transported to the liver. A large propor-
`tion of a drug will partition or be transported into
`the hepatocyte, where it may be metabolized by
`
`hepatic enzymes to inactive chemicals during the
`initial trip through the liver by what is known as the
`first—pass effect. Over 60% of the local anesthetic-
`antiarrhythmic agent, lidocaine, is metabolized dur-
`ing its initial passage through the liver, resulting in
`it being impractical to administer orally. When used
`for cardiac arrhythmias,
`it
`is administered intra-
`venously. This rapid metabolism of lidocajne is used
`to advantage when stabilizing a patient with cardiac
`arrhythmias. Should too much lidocaine be adminis-
`
`0
`
`CH3
`
`/
`NH
`
`/C2H5
`(j\/NH"
`Cl“
`\C2H5
`
`CH3
`
`Lidocaine
`
`0
`CH3
`u
`/CYNH
`
`NH3+ CI‘
`
`CH3
`
`CH3
`
`Tocainide
`
`R
`
`S /
`
`/
`
`N
`
`”AI >
`K-N
`
`NH
`
`Sutindac: R = CH3SO"; Active Sulfide Metabolite: R = CH3S“
`
`o2N
`
`Azathioprene R =
`
`N
`
`;
`
`6—Mercaptopurine: R = H
`
`‘F
`CH3
`
`
`
`CH3
`/
`CH2—CH2—CH2—N\ ~HCt
`R
`
`lmipramine R = CH3
`Desipramine: R = H
`
`Hc—cH42—cH2—N\
`
`-HCt
`
`R
`
`Amitriptyltne: R = CH3
`Nortriptytine: R = H
`
`Phenacetin: R, = C2H5O', R2 = CH3CO‘. R3 = H;
`Acetaminophen: R1 = HO‘ ,R2 = CH3CO', R3: H;
`
`|PR2014—01126—EXhibit 1028 p. 8
`
`IPR2014-01126-Exhibit 1028 p. 8
`
`

`
`tend to
`toxic responses will
`tered intravenously,
`abate because of the rapid biotransformation to in-
`active metabolites. An understanding of
`the
`metabolic labile site on lidocaine led to the develop-
`ment of the primary amine analogue, tocainide. In
`contrast with lidocaine’s half—life of less than two
`hours,
`tocainide’s half-life is about 15 hours with
`40% of the drug excreted unchanged. The develop-
`ment of orally active antiarrhythmic agents is dis-
`cussed in more detail in Chapter 14.
`A study of the metabolic fate of a drug is a
`requirement for all new products. Many times it is
`found that the metabolites also are active. Indeed,
`sometimes the metabolite is the pharmacologically
`active molecule. These drug metabolites can provide
`the leads for additional investigations of potentially
`new products. Examples of where an inactive parent
`drug is converted to an active metabolite include the
`nonsteroidal anti-inflammatory agent, sulindac, be-
`ing reduced to the active sulfide metabolite;
`the
`immunosuppressant, azathioprine, being cleaved to
`the purine antimetabolite, 6—mercaptopurine; and
`purine and pyrimidine antimetabolites and antiviral
`agents being conjugated to their nucleotide form.
`Many times both the parent drug and its metabolite
`are active, which has led to two commercial prod-
`ucts, instead of just one, being marketed. About 75%
`to 80% of phenacetin (now withdrawn from the
`U. S. market) is converted to acetaminophen. In the
`tricyclic antidepressant
`series
`(see Chap.
`10)
`imipramine and amitriptyline, are N-demethylated
`to desipramine and nortriptyline, respectively. All
`four compounds have been marketed in the United
`States. The topic of drug metabolism is more fully
`discussed in Chapter 3.
`Although a drug’s metabolism can be a source of
`frustration for the medicinal chemist, pharmacist,
`and physician, and can lead to inconvenience and
`compliance problems for the patient, it is fortunate
`that the body has the ability to metabolize foreign
`molecules (xenobiotics). Otherwise, many of these
`substances could remain in the body for years. This
`has been the complaint against certain lipophilic
`chemical pollutants, including the once, very popu-
`lar insecticide, DDT. After entering the body, these
`chemicals sit in body tissues, slowly diffusing out of
`the depots and potentially harming the individual on
`a prolonged basis for several years.
`
`EXCRETION
`
`The main route of excretion of a drug and its
`metabolites is through the kidney. For some drugs,
`
`DRUG DISTRIBUTION l
`
`9
`
`enterohepatic circulation (see Fig. 2-1), in which the
`drug reenters the intestinal
`tract from the liver
`
`through the bile duct, can be an important part of
`the agent’s distribution in the body and route of
`excretion. The drug or drug metabolite can reenter
`systemic circulation by passing once again through
`the intestinal mucosa. A portion of it also may be
`excreted in the feces. Nursing mothers must be
`concerned because drugs and their metabolites can
`be excreted in human milk and be ingested by the
`nursing infant. Usually, the end products of drug
`metabolism are very water-soluble relative to the
`parent molecule. Obviously, drugs that are bound to
`serum protein or show favorable partitioning into
`tissue depots are going to be excreted more slowly
`for the reasons already discussed.
`This does not mean that for those drugs that
`remain in the body for longer periods, lower doses
`can be administered or the drug can be taken fewer
`times per day by the patient. Several variables deter-
`mine dosing regimens, of which the affinity of the
`drug for the receptor is crucial. Reexamine Reaction
`2-1 and Figure 2-1. If the equilibrium does not favor
`formation of the drug—receptor complex, higher and
`usually more frequent doses will have to be adminis-
`tered. If the partitioning into tissue stores, metabolic
`degradation, or excretion are favored,
`it will take
`more drug, and usually more frequent administra-
`tion, to maintain therapeutic concentrations at the
`receptor.
`
`RECEPTOR
`
`With the exception of general anesthetics (see Chap.
`9), the working model for a pharmacologic response
`consists of a drug binding to a specific receptor.
`Many drug receptors actually are used by endoge-
`nously produced ligands. Cholinergic agents interact
`with the same receptors as the neurotransmitter
`acetylcholine. Synthetic corticosteroids bind to the
`same receptors as cortisone and hydrocortisone.
`Many times, receptors for the same ligand will be
`found in a variety of tissues throughout the body.
`The nonsteroidal anti-inflammatory agents
`(see
`Chap. 17) inhibit the prostaglandin-forming enzyme
`cyclooxygenase which is found in nearly every tis-
`sue. This class of drugs has a long list of side effects,
`with many patients’ complaints. Note in Figure 2-1
`that, depending on which receptors contain bound
`drug, there may be desired or undesired effects. This
`is because there are a variety of receptors with
`similar structural requirements found in several
`organs and tissues. Thus,
`the nonsteroidal anti-
`
`|PR2014—01126—EXhibit 1028 p. 9
`
`IPR2014-01126-Exhibit 1028 p. 9
`
`

`
`10l
`
`PHYSICOCHEMICAL PROPERTIES IN RELATION TO BIOLOGICAL ACTION
`
`inflammatory drugs combine with the desired cy-
`cloxygenase receptors at the site of the inflammation
`and the receptors in the gastrointestinal mucosa
`causing severe discomfort and sometimes ulceration.
`One of the newer antihistamines,
`terfenadine,
`is
`claimed to cause less sedation because it does not
`
`readily penetrate the blood~brain barrier. The ratio-
`nale is that less of this antihistamine is available for
`
`the receptors in the central nervous system that are
`responsible for the sedation response characteristic
`of antihistamines. In contrast, some antihistamines
`are used for their central nervous system depressant
`activity, which would imply that a considerable
`amount of the administered dose is crossing the
`blood—brain barrier relative to binding to the his-
`tamine—1 (H1) receptors in the periphery.
`Although it is normal to think of side effects as
`undesirable, they sometimes can be beneficial and
`lead to new products. The successful development of
`oral hypoglycemic agents, used in the treatment of
`diabetes, began when it was found that certain sul-
`fonamides had a hypoglycemic effect (see Chap. 5).
`Nevertheless, a real problem in drug therapy is
`patient compliance to take the drug a directed. Drugs
`that cause serious problems and discomfort tend to
`be avoided by the patient.
`
`SUMMARY
`
`One of the goals is to design drugs that will interact
`with receptors at specific tissues. There are several
`ways to do this, including (1) altering the molecule
`which, in turn, can change the biodistribution, (2)
`increasing the specificity for the desired receptor
`(desired pharmacologic response) while decreasing
`the affinity for undesired receptor (producer of side
`effects), and (3) the still experimental approach of
`attaching the drug to a monoclonal antibody that
`will bind to a specific tissue that is antigenic for the
`antibody. Alteration of biodistribution can be done
`by changing the drug’s solubility, enhancing its abil-
`ity to resist being metabolized (usually in the liver),
`altering the formulation or physical characteristics
`of the drug, and changing the route of administra-
`tion. If a drug molecule can be designed in such a
`way that its binding to the desired receptor is en-
`hanced,
`relative to the undesired receptor, and
`biodistribution remains favorable, smaller doses of
`the drug can be administered. This, in turn, reduces
`the amount of drug available for binding to those
`receptors responsible for its side effects.
`Thus, the medicinal chemist is confronted with
`several challenges in designing a bioactive molecule.
`A good fit to a specific receptor is desirable, but the
`
`drug would normally be expected to eventually dis-
`sociate from the receptor. The specificity for the
`receptor would be such that side effects would be
`minimal. The drug would be expected to clear the
`body within a reasonable time. Its rate of metabolic
`degradation should allow reasonable dosing sched-
`ules and,
`ideally, oral administration. Many times
`the drug chosen for commercial sales has been se-
`lected from the hundreds of compounds that have
`been screened. It usually is a compromise product
`that meets a medical need, while demonstrating
`good patient acceptance.
`
`AC|D—BASE PROPERTIES
`
`INTRODUCTION
`
`Most drugs used today can be classified as acids or
`bases. As will be noted shortly, many drugs can
`behave as either acids or bases as they begin their
`journey into the patient in different dosage forms
`and end up in systemic circulation. A drug’s
`acid—base properties can influence greatly its biodis-
`tribution and partitioning characteristics.
`Over the years, at least four major definitions of
`acids or bases have been developed. The model com-
`m

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