throbber
ToxicoLoGxc PATHOLOGY ISSN:0192-6233
`Copyright © 1995 by the Society of Toxicologic Pathologists
`
`Volume 23, Number 2, 1995
`Printed in U.S.A.
`
`An Introduction to Drug Disposition: The Basic
`Principles of Absorption, Distribution,
`Metabolism, and Excretion*
`
`JOHN CALDWELL, IAIN GARDNER, AND NICOLA SWALES
`
`Department of Pharmacology and Toxicology, St. Mary’s Hospital Medical School,
`Imperial College of Science, Technology and Medicine, London W2 1 PG, United Kingdom
`
`ABSTRACT
`
`A knowledge of the fate of a drug, its disposition (absorption, distribution, metabolism, and excretion,
`known by the acronym ADME) and pharmacokinetics (the mathematical description of the rates of these
`processes and of concentration-time relationships), plays a central role throughout pharmaceutical research
`and development. These studies aid in the discovery and selection of new chemical entities, support safety
`assessment, and are critical in defining conditions for safe and effective use in patients. ADME studies provide
`the only basis for critical judgments from situations where the behavior of the drug is understood to those
`where it is unknown: this is most important in bridging from animal studies to the human situation. This
`presentation is intended to provide an introductory overview of the life cycle of a drug in the animal body
`and indicates the significance of such information for a full understanding of mechanisms of action and
`toxicity.
`Keywords.
`
`Xenobiotics; human and animal exposure; predictive value
`
`INTRODUCTION
`
`Humans and other animals are exposed on a daily
`basis to many xenobiotics, that is, compounds that
`are foreign to the normal energy-yielding metabo-
`lism of the body. Exposure to these xenobiotics may
`occur deliberately, as in the case of drugs and food
`additives; accidentally, as in the case of food con-
`taminants and pesticides, or coincidentally, as in
`the case of industrial chemicals and environmental
`
`pollutants. In this paper, the terms drug, xenobiotic,
`and foreign compound will be used interchangeably.
`In the present context, the importance of ADME
`(absorption, distribution, metabolism, and excre-
`tion) principles in drug development will be em-
`phasized, but it should be appreciated that these
`have comparable applicability in the safety assess-
`ment of all types of chemicals to which humans
`might be exposed.
`To achieve its effect, whether therapeutic or toxic,
`a drug and/or its metabolites must be present in
`appropriate concentrations at its sites of action. The
`
`* Address correspondence to: Professor J. Caldwell, Depart-
`ment ofPharmacology and Toxicology. St. Mary’s Hospital Med-
`ical School, Imperial College of Science, Technology and Med-
`icine. Norfolk Place. London W2 IPG, United Kingdom.
`
`concentration of xenobiotic attained will depend on
`the dose, formulation, and route of administration,
`the rate and extent of absorption, its distribution
`through the body and binding to tissues, biotrans-
`formation, and excretion. It is the purpose of this
`presentation to give an overview of these processes
`and to comment upon the factors influencing them
`and their biological significance.
`
`ABSORPTION
`
`The processes of absorption are those that lead to
`the entry of a xenobiotic into the systemic circula-
`tion of the body. The most important site of ab-
`sorption is the gastrointestinal tract, although ab-
`sorption through the skin, the main barrier between
`the internal milieu and the external environment,
`and the respiratory tract, which is important for
`volatile compounds and materials present in aero-
`sols and dust particles, can also occur. Regardless
`of the site of absorption, xenobiotics must cross cell
`membranes to enter the systemic circulation. Mech-
`anistically this can occur in 1 of 2 ways (4). Small,
`lipophilic compounds can cross the cell membrane
`by passive diffusion along a concentration gradient.
`This transfer is directly proportional to the mag-
`nitude of the concentration gradient across the
`0192-6233/95$3.00+$0.00
`
`102
`
`|nnoPharma Exhibit 1025.0001
`
`

`

`Vol. 23, No. 2. 1995
`
`BASIC PRINCIPLES OF ADME
`
`103
`
`membrane and the lipid : water partition coefficient
`of the drug (3). Large. highly polar or charged xe-
`nobiotics cannot cross the cell membranes by simple
`diffusion and. hence. are dependent on the presence
`of active carrier-mediated transport mechanisms.
`
`The Efleet of pH and pKa on Absorption
`from the Gastrointestinal Tract
`
`Many xenobiotics are weak acids or bases and are
`thus present in solution in both non-ionized and
`ionized forms. The non-ionized molecules tend to
`
`be lipid-soluble and cross membranes by passive
`diffusion. whereas the ionized forms have low lipid
`solubility and cannot cross the cell membrane (3).
`The partition of weak electrolytes across mem-
`branes will thus be a function of the pKa of the
`xenobiotic and the pH gradient across the mem-
`brane.
`
`The low pH in the stomach favors absorption of
`weak acids. Weak bases are ionized and, thus, gen-
`erally not absorbed from the stomach. In the intes-
`tine, absorption is rapid for weak acids (pK > 3) or
`weak bases (pK < 7.8). The longer transit time and
`increased surface area of the intestine mean that,
`for the majority of drugs, intestinal absorption is
`quantitatively more important even if it would be
`predicted to be less favorable on pH grounds (7).
`
`First—Pass Elimination
`
`Following absorption, drugs can be metabolized
`in the gut wall, prior to being transported to the liver
`via the hepatic portal vein (78). The hepatocytes of
`the liver are the major site of metabolism for the
`majority of drugs, and compounds can be exten-
`sively metabolized in the liver before reaching the
`systemic circulation. That portion of the dose that
`is absorbed from the lumen of the gastrointestinal
`tract but eliminated by metabolism in the gut wall
`and/or the liver on the way to the heart is said to
`have undergone f1rst—pass elimination (3). The ex-
`tent to which xenobiotics undergo first—pass elimi-
`nation will have a major influence on the exposure
`to the compound following oral administration. The
`enzymes contributing to the metabolism of xeno-
`biotics are also found in organs other than the liver,
`such as the lung and skin. albeit usually at a lower
`level. Thus. xenobiotics entering the body by routes
`other than the gastrointestinal tract can also be sub-
`ject to first-pass metabolism.
`
`DISTRIBUTION
`
`Following entry of a xenobiotic to the systemic
`circulation. its distribution into the various tissues
`
`of the body will be influenced by tissue hemody-
`namics. passive diffusion across lipid membranes.
`the presence of carrier-mediated active transport
`
`processes recognizing the xenobiotic. and protein
`binding in the blood and tissues.
`The majority oftissue membranes behave as typ-
`ical
`lipid barriers allowing small lipophilic mole-
`cules to cross cell membranes. Equilibrium drug
`concentration ratios are maintained by diffusion of
`drugs into and out oftissues. Drugs can accumulate
`in tissues at a higher concentration than predicted
`by simple diffusion under the influence of pH gra-
`dients. binding to intracellular constituents. or par-
`titioning into lipid depots. Larger or more polar
`substances do not cross lipid membranes by passive
`diffusion and require specific transporters to enter
`the tissues (44). If a drug does enter a tissue by an
`active transport mechanism. its concentration in the
`tissue may be many times greater than its plasma
`concentration.
`
`Active uptake processes tend to show stereosc-
`lectivity and can be particularly important for xe-
`nobiotics that may be analogs of nutrients (5 l). The
`operation of specific uptake mechanisms for xeno-
`biotics may play an important role in the toxicity
`ofsome compounds. For example. amantanide and
`phalloidin are toxic cyclopeptides of the fungus Am-
`anita phalloides (21). The toxins enter the liver via
`an active transport system involved in the transport
`of bile acids (23). Once inside the cells. the toxins
`bind to microfilamentous F-actin and destroy the
`mechanical stability ofthe liver cell membrane. This
`results in hemorrhagic liver swelling and animals
`die within 2-3 hr of intravenous dosing with the
`peptides (22). Co-administration of bile salts with
`the toxins reduces their hepatic uptake by this active
`transport mechanism and thereby limits the toxicity
`of the compounds. Distribution of xenobiotics can
`also be limited by binding to plasma proteins. Acidic
`drugs tend to bind to albumin, and basic drugs tend
`to bind to a,—acid glycoprotein. As only unbound
`drug is in equilibrium across membranes. a drug
`that is extensively and strongly bound to plasma
`proteins has only limited access to the tissues.
`
`Drug Reservoirs
`
`Accumulation ofa drug within a tissue can act as
`a reservoir serving to prolong its duration of action.
`Ifthe stored xenobiotic is in equilibrium with that
`in plasma and is released as its plasma concentration
`falls. then the concentration ofxenobiotic in plasma
`will be sustained and the pharmacological effect of
`the xenobiotic will be prolonged (3). Thus. the stor-
`age of a drug can prolong its action either within
`the tissue where the drug is held or at a distant site
`reached following redilfusion into the systemic cir-
`culation (29).
`The concepts of drug reservoirs and how they
`influence the concentration of a xenobiotic at
`its
`
`|nnoPharma Exhibit 1025.0002
`
`

`

`104
`
`CALDWELL ET AL
`
`TOXICOLOGIC PATHOLOGY
`
`target tissue are well illustrated by the behavior of
`the lipophilic anesthetic thiopental, which is given
`by bolus intravenous injection (2). As a consequence
`of the high blood flow to the brain and its lipid
`solubility, thiopental reaches its maximum concen-
`tration in its target tissue within 1 min of intrave-
`nous injection. When the injection is stopped, the
`plasma concentration falls as the drug distributes
`into tissues such as muscle. As thiopental is not
`tightly bound to brain lipid, its concentration in the
`brain changes in parallel with changes in the plasma
`concentration, leading to a rapid termination of an-
`esthesia by redistribution rather than elimination.
`A third distributive phase for thiopental occurs as
`the result of a slow, blood flow—limited uptake into
`poorly perfused tissues such as fat (3).
`On repeated administration, fat and other poorly
`perfused tissues can accumulate large amounts of
`thiopental. These reservoirs are then capable of
`maintaining plasma and, hence, brain concentra-
`tions of thiopental at levels above those needed for
`anesthesia. Thus, a compound whose duration of
`action is limited by rapid redistribution from its site
`of action to storage sites can become long acting if
`storage deposits of suflicient size are established. At
`this point, termination of drug action becomes de-
`pendent on biotransforrnation and excretion of drug.
`The pharmacological consequence of these changes
`in tissue distribution is that the sleeping time after
`dosing of thiopental is changed from a few minutes
`following a single administration to a few hours fol-
`lowing multiple dosing (29).
`Toxicity testing is often performed using much
`higher doses of xenobiotics than humans are ex-
`posed to. As well as leading to saturation of meta-
`bolic pathways, it must be appreciated that these
`high doses can lead to changes in tissue distribution
`similar to those seen following multiple dosing of
`thiopentone.
`
`METABOLISM
`
`Drugs and other xenobiotics that gain access to
`the body may undergo 1 or more of 4 distinct fates,
`as follows (12):
`
`l. Elimination unchanged
`2. Retention unchanged
`3. Spontaneous chemical transformation
`4. Enzymic metabolism
`
`Each of these fates are of importance but, in quan-
`titative terms it is enzymic metabolism, often also
`referred to as biotransformation, that predominates.
`The main site of metabolism of foreign com-
`pounds is the liver, although extrahepatic tissues,
`frequently the site of entry to or excretion from the
`body (e.g., lungs, kidneys, gastrointestinal mucosa),
`
`also play a role in the metabolism of xenobiotics
`(24 and references therein).
`Compounds eliminated unchanged are generally
`either (a) highly polar such as strong carboxylic or
`sulfonic acids (e.g., sodium cromoglycate) or qua-
`ternary amines (e.g., pancuronium), which if ab-
`sorbed are rapidly cleared into the urine or bile, or
`(b) volatile and hence readily lost via the lungs. In
`contrast, nonpolar, highly lipophilic compounds may
`be retained for long periods in tissue lipids, as occurs
`with chlorophenothane and many polyhalogenated
`aromatics. For a small number ofcompounds, spon-
`taneous chemical transformation within the tissues
`
`of the body can be important: this may involve
`hydrolysis at the appropriate pH (e.g., thalidomide
`with its numerous breakdown products) or reaction
`with nucleophilic or electrophilic centers in tissue
`macromolecules, most notably the nucleophilic — SH
`of glutathione (71).
`The scope of drug metabolism is immense, and
`this is reflected in the range of chemical reactions
`that are involved in the metabolism of substrates,
`including oxidation, reduction, hydrolysis, hydra-
`tion, conjugation, and condensation. Typically, the
`process of metabolism of xenobiotics is biphasic,
`whereby the compound first undergoes a function-
`alization reaction (oxidation, reduction, or hydro-
`lysis), which introduces or uncovers a functional
`group (—OH,—NH2,—SH) suitable for subsequent
`conjugation with an endogenous conjugating agent.
`By far the most important enzyme system in-
`volved in Phase 1 metabolism is cytochrome P-450,
`the terminal oxidase component of the microsomal
`electron transfer system, which is responsible for
`the oxidation of many xenobiotics. The required
`electrons are supplied by the closely associated en-
`zyme NADPH cytochrome P-450 reductase, a fla-
`voprotein that transfers 2 electrons to cytochrome
`P—450 from NAD(P)H.
`The cytochromes P-450 are an enzyme superfam—
`ily consisting of a number of related isoenzymes, all
`of which possess an iron protoporphyrin IX as pros-
`thetic group. The enzymes are named for the Soret
`band around 450 nm exhibited by the CO complex
`of the reduced form. The P-450 enzymes have been
`grouped together into families that share sequence
`identity. There are 10 mammalian gene families
`comprised of 18 subfamilies (52, 53). The most im-
`portant enzymes involved in xenobiotic metabolism
`belong to the 1A, 2B, 2C, 2D, and 3A subfamilies.
`Although the individual enzymes are thought to me-
`tabolize substrates via the same catalytic mecha-
`nism (27), they tend to show selectivity toward sub-
`strates. For individual isoforrns of P-450, the extent
`of this selectivity is highly variable with overlap of
`substrates and regio— and stereospecificities being
`
`|nnoPharma Exhibit 10250003
`
`

`

`Vol. 23. No. 2. 1995
`
`BASIC PRINCIPLES OF ADME
`
`105
`
`observed (26). In addition. substrates are often me-
`tabolized at more than I position. as in the case of
`testosterone. This is believed to be a function of
`
`hydroxylation of warfarin and phenytoin. Whereas
`CYPEC9 is involved in the metabolism of tolbu-
`tamide and a number of acidic nonsteroidal anti-
`
`both the binding characteristics ofthe enzyme and
`the ease with which the functional groups of the
`substrate undergo oxidation (66). A number of ac-
`tive site models have been proposed to explain the
`different substrate specificities of various P-450 iso-
`enzymes.
`
`CYPIAI
`
`The substrate binding site of CYP1A1 has been
`proposed to consist of a hydrophobic cleft asym-
`metrically disposed to the heme iron atom. The
`asymmetric position ofthe binding site restricts the
`number of faces of the substrate that can be exposed
`to the active oxygen species (32). CYP1A1 has been
`implicated in the metabolism ofa number of poly-
`cyclic aromatic hydrocarbon (PAH) compounds such
`as benzo(a)pyrene. The substrates tend to be large,
`rigid, planar molecules containing fused (het-
`ero)aromatic rings that are good electron acceptors.
`Lewis et al (42) proposed that the binding site of
`CYP1A1 contains a number of aromatic amino ac-
`
`ids that form a planar pocket to complement the
`(hetero)aromatic rings of the substrates. The me-
`tabolism of benzo(a)pyrene results in the preferen-
`tial production of the bay region 7,8—diol-9, lO—epox-
`ide, which is a potent DNA-reactive ultimate car-
`cinogen (33).
`In addition to PAH metabolism,
`CYP1A1 can metabolize a number of smaller non-
`
`PAH compounds in a regio- and stereoselective
`manner (62). It has been suggested that these sub-
`strates are positioned in the active site via hydrogen-
`bonding interactions between the substrate and an
`active site residue of CYP1A1 (38).
`
`CYPZBI/2
`
`The P-450 2B isozymes are involved in a number
`ofbiotransformations in the rat and are induced by
`phenobarbital. The substrates for CYPZB tend to
`be bulky, nonplanar molecules with greater confor-
`mational flexibility than CYPIA substrates (39). The
`substrates tend to have functional groups of similar
`size and hydrophobicity to isopropyl and to be poor
`electron acceptors (30, 39). It has been proposed
`that the binding site ofCYP2B contains hydrophilic
`amino acids that are capable of forming hydrogen
`bonds with carbonyl and/or amine groupings ofthe
`substrate and hydrophobic nonaromatic residues
`that complement the isopropyl function (42).
`
`CYPBC
`
`The CYPZC subfamily appears to be important
`in metabolizing a number of xenobiotics particu-
`larly in humans (66). CYPZC8 effects the aromatic
`
`inflammatory drugs and is potently inhibited by sul-
`faphenazole. CYPZCIS is subject to a genetic poly-
`morphism manifest in the hydroxylation of S-me-
`phenytoin and is not inhibited by sulfaphenazole
`(26). CYPZC substrates tend to have areas ofstrong
`hydrogen bond—forming potential positioned 5-10
`A from the site of oxidation. and a number are also
`charged at physiological pH (66). This has lead to
`the suggestion that hydrogen-bonding potential and
`possibly ion pair interactions are important in de-
`termining the substrate structure activity relation-
`ships of the P-4502C isozymes (66).
`
`C YPZD
`
`The CYPZD isozymes have been extensively in-
`vestigated, as they are involved in the genetic poly-
`morphic metabolism ofdebrisoquine. spaneine. and
`some 30 other substrates (17. 48). CYP2Dl (rat)
`and ZD6 (human) have similar substrate selectivi-
`ties, but inhibition studies with quinidine (more po-
`tent in humans than rats) and its diastereoisomer
`quinine (more potent in rats than humans) dem-
`onstrate that differences in the enzyme active site
`must exist (65).
`Substrates for CYPZD enzymes possess a basic
`nitrogen grouping that is mainly ionized at physi-
`ological pH. a hydrophobic region and a functional
`group capable of P-450 oxidation 5-7 A from the
`basic nitrogen (69). Reactions catalyzed include ar-
`omatic hydroxylation (propranolol). aliphatic hy-
`droxylation (metoprolol). and N—dealkylation (ami-
`fiamine) (17). The substrate binding site of CYPZD
`appears to contain a carboxyl group that binds and
`neutralizes the basic nitrogen of the substrate and a
`hydrophobic domain. The carboxylate group is as-
`sumed to serve as an anchoring site on the protein.
`Substrates can interact with either of the oxygen
`atoms of the carboxylate group (which are 2.2 A
`apart). explaining why for some substrates the dis-
`tance between basic nitrogen and site of oxidation
`is 5 A. typified by debrisoquine. and for other sub-
`strates it is 7 A. 1} pified by dextromethorphan (38).
`The ionic bonding between substrate and enzyme
`means that the en/_\ me tends to have a high affinity
`for substrates and. thus. a low Km (65). Many sub-
`strates also exhibit a coplanar conformation near
`the oxidation site and have a negative molecular
`electrostatic potential in a part ofthis planar domain
`approximately 3 A away from the oxidation site
`(38).
`The predictive value of the model was assessed
`by measuring the CYPZD6-mediated metabolism
`of-1 compounds. showing among them at least 14
`
`|nnoPharma Exhibit 1025.0004
`
`

`

`106
`
`CALDWELL ET AL
`
`Tox1coLoG1c PATHOLOGY
`
`TABLE I.—The 8 classical conjugation reactions.
`
`Reaction
`
`Conjugating agent
`
`A. Reactions involving activated conjugating agents
`Glucuronidation
`Glucose conjugation
`Sulfation
`Methylation
`Acetylation
`Cyanide detoxication
`B. Reactions involving activated foreign compounds
`Glutathione conjugation
`Amino acid conjugation
`
`UDP glucuronic acid
`UDP-glucose
`3’-Phosphoadenosine-5 ’-phosphosulfate
`S-adenosyl methionine
`Acetyl coenzyme A
`Sulfane SU.lf1lI'
`
`Glutathione
`Glycine, ornithine, taurine
`
`oxidative metabolic routes. From the model, 4 routes
`were predicted to be 2D6—mediated. In vivo and in
`vitro data from humans demonstrated that 3 of the
`
`4 predicted metabolic routes were in fact mediated
`by CYP2D6 (40).
`
`C YP3A
`
`The CYP3A family tends to be involved in the
`metabolism of large, structurally diverse, fairly li-
`pophilic compounds. Although substrates are bulky,
`metabolism tends to occur in small exposed func-
`tional groups that undergo reactions such as N-deal-
`kylation and aliphatic hydroxylation. Substrates in-
`clude the immunosuppressant cyclosporin A, nifed-
`ipine, and verapamil (66).
`It has been suggested that the binding site for
`CYP3A is dominated by hydrophobic interactions
`and that, in contrast to CYP2D, which is governed
`by ionic bonding, this allows for a degree of flexi-
`bility in the position of substrate binding (67).
`
`Conjugation Reactions
`
`Phase 2 conjugation reactions may be divided into
`2 distinct groups, depending on the source of energy
`for the process (10). In most instances, the energy
`is derived from the activated endogenous conjugat-
`ing agent, as is the case for the glucuronic acid,
`sulfate, methylation, and acetylation reactions. In
`other examples, the energy is derived by prior met-
`abolic activation of the xenobiotic, as is the case for
`glutathione and amino acid conjugations. Of the
`Phase 2 conjugation reactions listed in Table I, gluc-
`uronic acid conjugation ranks as highest importance,
`and many drugs (e.g., indomethacin, paracetamol,
`dapsone, clofibrate, morphine) are metabolized via
`this pathway. The conjugations are performed by a
`family of glucuronyl transferase enzymes located
`within the endoplasmic reticulum of the cells of the
`liver. intestine and kidney. These enzymes catalyze
`the conjugation of uridine diphosphate—a-l-gluc-
`uronic acid with nucleophilic O, N, C, and S atoms:
`during the reaction, C-1 of the sugar ring is inverted
`
`so that the products are 1-0-substituted B-D-glu-
`copyranosiduronic acids. The enzymes have a mo-
`lecular weight of between 50 and 60 kDa and exist
`as oligomers of between 1 and 4 subunits in vivo
`(60). At least 9 difierent isozymes in 2 different sub-
`families are known to exist (8). Glucuronidation oc-
`curs in most mammalian species with the cat and
`related felines and the Gunn rat being notable ex-
`ceptions.
`Glutathione-S-transferases catalyze the conjuga-
`tion of a number of functional groups (aryl and alkyl
`halides, lactones, epoxides, and quinones) with glu-
`tathione, the tripeptide 7-glutamylcysteinylglycine.
`The glutathione-S-transferases have a very exten-
`sive tissue distribution and are principally found in
`the cytosol of the cell. The proteins have a molecular
`weight of 24-28 kDa and exist as dimers in vivo
`(77). The dimeric proteins possess binding sites for
`glutathione and the electrophilic substrate, which
`brings the reactants close together (47). The mam-
`malian glutathione transferase enzymes have been
`divided into 5 evolutionary classes: oz, u, 7:-, 0, and
`microsomal (77). Typical substrates include para-
`thion, urethane, ethacrynic acid, and 1-chloro-2,4-
`dinitrobenzene. The glutathione transferase en-
`zymes are also very abundant in the liver cytosol
`(4—5°/o of total cytosolic protein). Thus, as well as
`having major significance in drug metabolism, these
`enzymes are also important in intracellular binding.
`This is particularly true for glutathione-S-transfer-
`ase B (Ligandin). Compounds that bind to gluta-
`thione-S-transferases include bilirubin, estradiol,
`cortisol, testosterone, tetracycline, penicillin, and
`indocyanine green (43).
`A number of catechol, phenol, and alcohol com-
`pounds are excreted as sulfate conjugates. This re-
`action between substrate and sulfate donor, 3’—phos-
`phoadenosine-5’-phosphosulfate, is catalyzed by a
`family of sulfotransferase enzymes (18). The sul-
`fotransferases have a cytosolic location and are found
`in many tissues including the liver, adrenals, lung,
`brain, jejenum, and blood platelets. The proteins
`
`|nnoPharma Exhibit 1025.0005
`
`

`

`Vol. 23, No. 2. 1995
`
`BASIC PRINCIPLES OF ADME
`
`107
`
`have molecular weights between 32 and 34 kDa and
`exist as homodimers in viva. The sulfotransferase
`
`These are important in determining the biological
`effect of a xenobiotic. The intracellular concentra-
`
`enzymes are normally classified into subfamilies
`based on their substrate specificity. although there
`is overlap among the different isozymes (18). Six
`different phenol transferases and 7 different steroid.
`bile acid sulfotransferases have been characterized
`
`in the rat. In humans. only 2 phenol transferases
`(M-PST and P-PST) and l steroid/bile acid sulfo-
`transferase (DHEA-ST) have been characterized.
`Typical substrates of the human liver cytosolic
`transferases are dehydroepiandrosterone. pregnen-
`olone, and testosterone (DHEA-ST), dopamine and
`acetaminophen (M-PST), and phenol and minoxidil
`(P-PST). As well as exhibiting species differences. it
`is known that phenol sulfotransferase activity varies
`among individuals over a 15-fold range (35).
`N-acetyltransferase catalyzes the addition of an
`acetyl group to the amino group of amine, amino
`acid, and sulfonamide compounds using acetyl co-
`enzyme A as a cosubstrate. N-acetyltransferase is a
`26.5 kDa cytosolic enzyme found in the liver and
`intestine. Typical substrates include isoniazid and
`sulfanilamide. A well-defined polymorphism for
`N-acetyltransferase exists in many species including
`humans (61), which can have profound effects on
`toxicity. For example, isoniazid accumulates in slow
`acetylators and so may predispose these individuals
`to drug-induced neuropathy. On the other hand, the
`toxicity of isoniazid is related to the formation of
`its N-acetyl derivative, which is further metabolized
`to a reactive intermediate. Rapid acetylators are
`more susceptible to this type of side effect. The dog
`and related canine species do not possess this poly-
`morphic enzyme.
`Epoxide hydrolase enzymes catalyze the trans ad-
`dition of water to a variety of epoxide compounds.
`Microsomal epoxide hydrolase is found in the liver,
`testes, kidney, ovary, and lung. It
`is an approxi-
`mately 50-kDa protein, and typical substrates in-
`clude styrene oxide, vinyl chloride epoxide. ben-
`zo(a)pyrene-4,5-oxide, phenytoin epoxide, and car-
`bamazepine epoxide (63). Cytosolic epoxide hydro-
`lase is a 60-kDa protein that exists as a homodimer
`of 120 kDa in viva. Typical substrates ofthis enzyme
`include trans-stilbene oxide. epoxymethyl stearate.
`and arachadonic acid epoxide. Species differences
`occur in the activity of cytosolic epoxide hydrolase
`in that the mouse has high activity. the rabbit. guin-
`ea pig. and humans have intermediate activity. and
`the rat has very low activity (46).
`
`Factors .-lfliecting .lIerab0li.sm ofX'en0bi0lics
`
`Factors influencing the rate and extent of metab-
`‘V
`olism via Phase 1 or _ reactions m vim can be
`
`physiological. endogenous. or exogenous (Table ll).
`
`tion of a chemical is primarily dependent on dose
`size and its physicochemical and structural prop-
`erties. Because the metabolism of most compounds
`is enzymatic. any factor that can influence the ac-
`tivity of these enzymes can alter metabolism.
`
`Species Dt'[ferem'cs In llelabo/1fs‘nz and 7'0.\‘1't‘iIy
`
`Species differences in metabolism are of most sig-
`nificance. but variability can also occur in absorp-
`tion, distribution. and excretion of foreign com-
`pounds. Species differences in drug metabolism
`reflect differences in the activities of the enzymes
`responsible for the various transformations. Such
`variations arise primarily from differences in the
`absolute activities of the enzymes. but the amounts
`of any endogenous inhibitors present, or the extent
`of any reverse reactions, are also relevant. Species
`differences occur in both Phase I and 2 metabolism
`
`and can be either quantitative (same metabolic route
`at different rates) or qualitative (different metabolic
`routes) (12). Given that species differences do occur,
`it is possible that they arise from 1 or more of 3
`origins:
`
`1. Deficiencies in certain enzymes that lead to a
`defect in a metabolism reaction that is otherwise
`
`widespread in occurrence. For example, the glu-
`curonidation deficiency in the cat leads to in-
`creased toxicity ofglucuronidogenic compounds
`in this and related species. The lack ofacetylation
`ofaromatic amines in dogs may explain why they
`are more susceptible to p-aminobenzoic acid and
`various hydrazines. The guinea pig has a defi-
`ciency in N-acetylation and is unable to N-acety-
`late S-substituted cysteines to form acid conju-
`gates (10). This is not due to defective formation
`of glutathione conjugates (the first step in the
`mercapturic acid pathway), indicating that the
`defect lies in the final step of transformation of
`the glutathione conjugate to a mercapturic acid
`(15). The pig and opossum are defective in their
`ability to conjugate phenolic compounds with
`sulfate (10).
`Although most ofthe deficiencies involve con-
`jugation reactions (13). there are a few examples
`of defects in oxidative metabolism. These in-
`
`clude the inability ofthe guinea pig and steppe
`lemming to .\'-hydroxylate the ;ir_\l-acetamide,
`2—acet_\lamidolluorene. and the inability of the
`rat and marmoset to .\'-hydroxylate the aliphatic
`amine chlorphentermine (l0).
`. Restricted species occurrences. For instance. these
`are seen with the particular amino acid (glycine,
`glutamine. taurine. or ornithine) utilized in the
`
`Ix)
`
`|nnoPharma Exhibit 1025.0006
`
`

`

`108
`
`CALDWELL ET AL
`
`TOXICOLOGIC PATHOLOGY
`
`TABLE II.—Physicochemical, endogenous, and exogenous factors affecting the rate and extent of metabolism of xe-
`nobiotic chemicals in viva.
`
`Physicochemical
`
`Electrophilicity
`Nucleophilicity
`Lipophilicity
`Polarity
`Protein binding
`
`Endogenous
`
`Age
`Sex
`Species
`Strain
`Pathology
`Genetic deficiencies
`Cofactor availability
`
`Exogenous
`
`Dose
`Nutrition
`Route of administration
`Time of day
`Enzyme inhibition
`Enzyme induction
`
`conjugation ofacids. Most species conjugate ben-
`zoic acid and heterocyclic and cinnamic acids
`with glycine, whereas bird species use ornithine.
`Of special interest are the reactions that are re-
`stricted to primates such as the glutamine con-
`jugation of phenyl acetic acid (10).
`3. Most common, the relative rates of competing
`pathways of metabolism rather than the route of
`metabolism. Amphetamine undergoes either ar-
`omatic hydroxylation, producing 4’-hydroxyam-
`phetamine, or side-chain degradation to benzoic
`acid. The compound is metabolized extensively
`in most species, but the nature of metabolites
`formed is highly variable. In the rat, ring hy-
`droxylation predominates, in the guinea pig chain
`breakdown is the major route, and in other spe-
`cies both routes are significant (10, 11, 13). Di-
`azepam is extensively metabolized in humans,
`dog, and rat. In humans, the major metabolites
`are 3-hydroxydiazepam, N'-desmethyldiaze-
`pam, and 3—hydroxydesmethyldiazepam (oxa-
`zepam). In the dog, oxazepam is the major prod-
`uct, and in the rat ring hydroxylations predom-
`inate with 4’—hydroxy—3-hydroxydiazepam, 4’-
`hydroxy—N‘ —desmethyldiazepam, oxazepam, and
`3—hydroxydiazepam all being formed. Oxazepam
`is further metabolized to the 0-glucuronide in
`humans and dog but to a number of ring hy-
`droxylated compounds in the rat (10). Phenol
`can be conjugated on the hydroxyl group with
`either sulfate or glucuronide. Humans and Old
`World monkeys excrete phenol principally as the
`sulfate, whereas New World monkeys excrete
`phenol principally as the glucuronide and the rat
`and mouse excrete approximately equal amounts
`of both conjugates (10).
`
`Sex Dzflerences in Metabolism and Toxicity
`
`Sex differences have been noted in the absorption
`(cephradine). protein binding (diazepam, warfarin),
`and biliary excretion (indocyanine green, tanrazine)
`of drugs and xenobiotics (9). However, the sex dif-
`ferences found in metabolic pathways have proba-
`bly the most significance with regard to xenobiotic
`
`toxicity. Sex differences have been reported in both
`Phase 1
`(e.g., N—demethylation of morphine and
`oxidation of pentobarbital) and Phase 2 metabolism
`(e.g., glutathione conjugation of l,2-dichlorodini-
`trobenzene, glucuronic acid conjugation of p-nitro-
`phenol, sulfation of N-hydroxy-2-acetylaminoflu-
`orene). The exact nature of

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