throbber
P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`Annu. Rev. Pharmacol. Toxicol. 2001. 41:443–70
`Copyright c(cid:176) 2001 by Annual Reviews. All rights reserved
`
`METABOLISM OF FLUORINE-CONTAINING DRUGS
`
`B Kevin Park, Neil R Kitteringham, and Paul M O’Neill
`Department of Pharmacology and Therapeutics, University of Liverpool,
`New Medical Building, Liverpool, United Kingdom; e-mail: bkpark@liverpool.ac.uk,
`neilk@liverpool.ac.uk, p.m.oneill01@liv.ac.uk
`
`Key Words drug metabolism, drug toxicity, drug design, drug safety,
`defluorination, metabolic stability
`n Abstract This article reviews current knowledge of the metabolism of drugs that
`contain fluorine. The strategic value of fluorine substitution in drug design is discussed
`in terms of chemical structure and basic concepts in drug metabolism and drug toxicity.
`
`INTRODUCTION
`
`Fluorine substitution can alter the chemical properties, disposition, and biological
`activity of drugs (1). Many fluorinated compounds are currently widely used in
`the treatment of disease. These include antidepressants, antiinflammatory agents,
`antimalarial drugs, antipsychotics, antiviral agents, steroids, and general anaesthet-
`ics (2). The chemistry and medicinal chemistry of fluoro-organic compounds and
`drugs have been reviewed (1, 3–5). The development of new fluorinating agents
`has vastly increased the potential for synthesis of novel fluorinated drugs. In ad-
`dition, the development of sophisticated noninvasive analytical techniques based
`on fluorine nuclear magnetic resonance (NMR) and positron emission topography
`has transformed the study of fluorinated drugs in man and animals (6–8).
`The inclusion of a fluorine atom in a drug molecule can influence both the
`disposition of the drug and the interaction of the drug with its pharmacological
`target (Figure 1). For example, the effects of fluorine substitution on the inter-
`and intramolecular forces that affect binding of ligands, and thus introduce re-
`ceptor subtype selectivity, at cholinergic and adrenergic receptors are now well
`understood (9–11). Fluorine substitution can also have a profound effect on drug
`disposition, in terms of distribution, drug clearance, route(s), and extent of drug
`metabolism (12). Such changes can be used constructively by medicinal chemists
`to improve both the safety and the efficacy of a drug. Therefore, the purpose of
`this review is twofold. First, to outline the chemical basis of changes in drug dis-
`position that can be achieved by the introduction of fluorine. Second, to consider
`the pharmacological and toxicological implications of such changes with respect
`to drug response.
`
`0362-1642/01/0421-0443$14.00
`
`443
`
`FUSTIBAL Ex. 1004
`
`

`
`P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`444
`
`PARK ¥ KITTERINGHAM ¥ O’NEILL
`
`Figure 1 Flow diagram illustrating the
`effect of fluorine substitution on drug re-
`sponse.
`
`PHYSICOCHEMICAL PROPERTIES
`OF FLUORINATED DRUGS
`
`The replacement of a hydrogen atom or hydroxyl group by a fluorine atom is a
`strategy widely used in drug development to alter biological function. Although it
`is generally thought that fluorine for hydrogen substitution causes minimal steric
`effects at receptor sites, the actual van der Waals radius of fluorine (1.47 ˚A) lies
`between that of oxygen (1.57 ˚A) and hydrogen (1.2 ˚A) (Table 1). Despite the fact
`that fluorine has a greater size than hydrogen, several studies have demonstrated
`that it is a reasonable hydrogen mimic and exerts only a minor steric demand at
`receptor sites, at least for monofunctional analogues (3).
`In contrast to their slight differences in size, hydrogen and fluorine have quite
`different electronic properties. Fluorine is the most electronegative element in the
`periodic table (Table 1). The resulting change in the electron distribution in a
`molecule, following the replacement of a hydrogen atom for fluorine, can alter
`the pKa, the dipole moments, and even the chemical reactivity and stability of
`neighboring functional groups. The magnitude of the change in these electronic
`
`

`
`P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`METABOLISM OF FLUORINATED DRUGS
`
`445
`
`TABLE 1 Physiochemical properties of the carbon-fluorine bond
`
`Element
`
`H
`F
`O(OH)
`
`Electro-
`negativity
`
`Bond length Van der Waals Bond energy
`radius ( ˚A)
`(CH2X, ˚A)
`(kcal/mol)
`
`2.1
`4.0
`3.5
`
`1.09
`1.39
`1.43
`
`1.20
`1.35
`1.40
`
`99
`116
`85
`
`properties is often determined by the bonding between the fluorine atom and the
`functional group. Thus, the presence of a fluorine atom ortho to a phenolic group
`is associated with a reduced pKa of 1.2, whereas meta and para fluoro substitutions
`have much less effect. The incorporation of two fluorine atoms at the 2- and 6-
`positions of phenol leads to a reduction of pKa of 2.7 U. Based on this effect,
`the 2,6-difluorophenol group has been used as an isostere of a carboxylic acid
`in a series of GABA aminotransferase inhibitors (13). These compounds were
`shown to inhibit the aminotransferase enzyme demonstrating the potential of this
`bioisosteric replacement.
`The presence of a single fluorine, adjacent to a carboxylic acid function in
`aliphatic systems, can also have pronounced effect on the pKa. This fact was
`used to rationalize the decrease in toxicity of new monofluorinated analogues
`of methotrexate. The incorporation of a fluorine atom adjacent to the glutamic
`carboxyl acid function results in an increase in the acidity. As a result, these
`new analogues are less toxic than methotrexate because they do not form polyg-
`lutamates, metabolites associated with undesirable prolonged cellular retention
`(14).
`Fluorine forms a strong bond with carbon (bond energy C-F D 116 kcal/mol),
`which has an increased oxidative and thermal stability compared with the carbon-
`hydrogen bond (C-H D 99 kcal/mol). The carbon-fluorine bond is one of the
`strongest known in organic chemistry. In addition to the formation of covalent
`bonds, a fluorine atom present in a molecule can also form reversible, electrostatic
`bonds with certain functional groups.
`The isosteric replacement of the hydroxyl group is a commonly used strategy in
`medicinal chemistry. This substitution is usually based on the premise that the fluo-
`rine can hydrogen bond accept in a manner similar to the oxygen of a hydroxyl func-
`tion. However, the higher electronegativity and lower polarizability of fluorine over
`oxygen has a major influence on the ability of fluorine to mimic a hydroxyl group
`(15, 16). Recent calculations have measured the strength of an optimum F...H bond
`¡1 in an adduct between fluoromethane and water. There-
`(1.9 ˚A) to be 2.38 kcal mol
`fore, the F...H bond is clearly much weaker than the corresponding O...H (conserva-
`¡1). The carbon-fluorine bond also has a strong
`tively estimated to be ca 5 kcal mol
`dipole, and this may interact, either positively or negatively, with other dipoles. For
`example, it is thought that in fluorinated derivatives of noradrenaline, interactions
`between a ring carbon-fluorine bond and the hydroxyl group on the beta-carbon
`in the side chain determine the conformation of the molecule, and hence, the
`
`

`
`P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`446
`
`PARK ¥ KITTERINGHAM ¥ O’NEILL
`
`position of the fluorine atom in the aromatic ring can determine receptor selectivity
`(17). Fluorine also has a number of specific stereoelectronic effects, such as
`the fluorine anomeric effect in carbohydrates, the Anh-Eisenstein stabilization
`effect in lipase-mediated kinetic resolutions, and the cis effect in difluorinated
`alkanes (3).
`In contrast to the single replacement of a hydrogen for fluorine, replacement
`of a methylene function with a difluoromethylene function (CF2 for CH2) can
`have a significant effect on both conformation and physical properties (3). The
`difluoromethylene moiety has in fact been used as an electronic mimic of labile
`2¡
`2¡
`). This functional
`oxygen atoms in phosphate esters (R-CF2-PO3
`vs R-OPO3
`group has found extensive use in the design of inhibitors of enzymes that hydrolyze
`or bind phosphate esters (18). The CF2 has been proposed as a reasonable isosteric
`and isopolar replacement for the hydroxyl group because of their size, electron
`distribution, and ability to act as a hydrogen bond acceptor (19–21). The CF2H
`group is particularly favored because of its ability to act as a hydrogen donor (22),
`potentially allowing interaction with solvent and biological molecules. Further
`introduction of fluorine causes even greater steric restrictions. The frequently used
`trifluoromethyl group (-CF3) is closer in size to an isopropyl group (23). Indeed,
`several workers have suggested that the CF3 group can exert an effect comparable
`to a phenyl ring or even a tert-butyl function (24).
`The presence of a fluorine atom can influence the lipophilicity of a molecule
`and hence affect the partitioning of the drug into membranes, and also facilitate
`hydrophobic interactions of the drug molecule with specific binding sites, on either
`receptors or enzymes. The replacement of a single aromatic hydrogen atom usually
`results only in a modest increase in lipophilicity, whereas the CF3 group is among
`the most lipophilic of all substituents.
`The fluoride ion is a good leaving group, being the conjugate base of a strong
`acid. Therefore, the fluoride ion can be lost, in both displacement and elimination
`reactions, and this aspect of fluorine chemistry can be utilized in the design of drugs
`or chemical agents that form stable covalent bonds with target receptors or enzymes
`as part of their pharmacological response. This is the basis of the “lethal synthesis”
`concept (25). More recently, fluorinated inhibitors of GABA aminotransferase have
`been synthesized as potential mechanism-based inhibitors (13).
`The presence of fluorine can alter the oxidation potential of an aromatic system,
`and thus alter the rate of autoxidation and formation of quinones and quinoneimines.
`Sequential introduction of fluorine atoms into the nucleus of paracetamol produced
`an increase in the oxidation potential of the molecule, as measured by cyclic
`voltammetry (26).
`Before embarking on fluorination as a synthetic strategy to alter drug dispo-
`sition, it is imperative to determine whether the changes in the physicochemical
`properties will diminish the inherent pharmacological activity of the drug. Molecu-
`lar modeling techniques can, in theory, be used to examine (a) the importance of the
`group to be replaced in the drug-receptor interaction and (b) whether the resulting
`C-F bond can provide the same chemical interaction with the receptor. However,
`it must be stressed that modeling of the C-F bond in drug-receptor interactions
`
`

`
`P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`METABOLISM OF FLUORINATED DRUGS
`
`447
`
`is not a trivial task, even for very simple molecules in an aqueous environment
`(27).
`
`STRATEGIES FOR THE USE OF FLUORINE
`SUBSTITUTION TO ALTER DRUG DISPOSITION
`
`The introduction of fluorine into a molecule can be used to alter the rate, route, and
`extent of drug metabolism. Fluorine substitution can also be used to dissociate the
`pharmacological and toxicological properties of a drug when a toxic metabolite
`has been identified.
`Such alterations are most commonly achieved by fluorine substitution at the site
`of metabolic attack, based on the premise that the carbon-fluorine bond is much
`more resistant to direct chemical attack by cytochrome P450, in comparison to the
`carbon-hydrogen bond. In addition, substitution at sites adjacent to and, in some
`instances, distal to the site of metabolic attack can also affect drug metabolism,
`by either inductive/resonance (through bond) effects or conformational and elec-
`trostatic (through space) effects. The presence of a fluorine atom adjacent to a
`site of metabolic attack could, in theory, either increase or decrease the rate of
`biotransformation, depending on (a) whether the metabolic attack is nucleophilic
`or electrophilic in nature and (b) inductive or resonance effects of the fluorine
`atom predominate in the reaction. For example, in a simple saturated system, the
`inductive effect of fluorine should reduce the susceptibility of adjacent groups to
`attack by P450 enzymes. In contrast, it might be anticipated that the presence of
`fluorine ortho to a phenolic group might increase its reactivity as a nucleophile in
`methylation and glucuronidation reactions, and there is some evidence to support
`this hypothesis (28, 29).
`Fluorine substitution can therefore have complex effects on drug metabolism.
`The framework outlined in Figure 1 is used to consider the importance of the role of
`fluorinated subgroups on various aspects of drug disposition, and the consequent
`impact on drug efficacy and drug toxicity.
`
`THE EFFECT OF FLUORINE SUBSTITUTION
`ON DRUG DISTRIBUTION
`
`The inclusion of fluorine in a molecule has two benefits with respect to drug distri-
`bution. First, certain fluorine-containing functional groups enhance lipophilicity
`and therefore passive diffusion of drug across membranes. Second, noninvasive
`techniques can be used to assess penetration of the drug to the site of action,
`whether brain, tumor, or site of infection (30–32).
`Centrally acting drugs must pass through the blood brain barrier in sufficient
`concentration to elicit their pharmacological effect. For example, there are three
`categories of neuroleptics that act by blocking dopamine receptors in the cen-
`tral nervous system (CNS): tricyclics, butyrophenones, and diarylbutylamines.
`Many of these drugs contain either a CF3 group or a fluoro-phenyl group, which
`
`

`
`P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`448
`
`PARK ¥ KITTERINGHAM ¥ O’NEILL
`
`contribute to the overall pharmacological activity of the compounds by enhanc-
`ing CNS penetration and retarding metabolic degradation. Several clinically use-
`ful phenothiazines containing fluorine have been introduced (4, 5). Trifluoropro-
`mazine, trifluperazine, and fluphenazine are all more active than chlorpromazine
`(5-, 50-, and 100-fold, respectively).
`The most widely used butyrophenone is haloperidol. It has been established
`that the para-fluorophenyl group is optimal for neuroleptic activity of the bu-
`tyrophenones. In the search for more potent neuroleptics, it was found that the
`diarylbutylamine, pimozide, which contains two fluorophenyl groups, was longer
`acting than haloperidol (12).
`Another important class of centrally acting drugs containing fluorine that have
`gained clinical prominence over the past few years is the selective serotonin up-
`take inhibitors. Of the most widely used agents in this class (fluoxetine, fluvoxa-
`mine, paroxetine sertraline, and citalopram), only sertraline does not contain a
`fluorine atom in its structure. Sertraline has the shortest half-life of the drugs men-
`tioned (26 h), is the most slowly absorbed, and undergoes almost total metabolic
`conversion, principally through presystemic elimination: all properties that may
`be associated with the lack of fluorine (2).
`Quinolone antibacterial agents act by inhibition of bacterial DNA synthesis.
`Fluoroquinolones were first introduced in the early 1980s (norfloxacin) and include
`ofloxacin, enoxacin, ciprofloxacin, tosufloxacin, sparfloxacin, grepafloxacin, lev-
`ofloxacin, and, most recently, trovafloxacin. This class of drugs is active against a
`wide range of gram-positive and gram-negative pathogens, possesses improved
`oral absorption and systemic distribution, and therefore has extended clinical
`applications that include urinary tract infections, respiratory tract infections, skin
`infections, and soft tissue infections. The structural features, which determine tis-
`sue distribution and cellular uptake, are complicated by specific efflux mechanisms
`(33). Nevertheless, it has been proposed that the 5-fluoro group is important for
`both cell penetration and gyrase affinity (34).
`
`THE EFFECT OF FLUORINE SUBSTITUTION
`ON THE RATE OF DRUG METABOLISM
`
`Fluorine substitution has been used to extend the biological half-life of endogenous
`compounds and synthetic compounds. Fluorinated analogues are not only useful
`tools for physiological investigations (Figure 2), they may also be potential thera-
`peutic agents. For example, the prostanoids, prostacyclin and thromboxane, play
`an essential physiological role in the regulation of the cardiovascular system and
`platelet function. Both compounds have short (<5 min) half-lives in vivo. Conse-
`quently, there has been a concerted effort to synthesize analogues with enhanced
`stability and, thus, extended activity.
`The potent platelet aggregating agent thromboxane A2 has a half-life of only
`32 s under physiological conditions. Incorporation of fluorine into the oxetane ring
`
`

`
`P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`METABOLISM OF FLUORINATED DRUGS
`
`449
`
`Figure 2 Examples of how fluorination
`of endogenous compounds can enhance
`metabolic stability.
`
`alpha to the acetalic linkage reduces the rate of carbonium ion formation and acid
`hydrolysis. Thus, model compounds related to 7,7-difluoro-TxA2 have a rate of
`hydrolysis that is 108-fold slower than that of TXA2 (35), whereas 10,10-difluoro-
`TxA2 is also a stable analogue and retains thromboxane-like activity (36).
`Prostacyclin, which is an inhibitor of platelet aggregation, contains an acid-
`labile enol-ether group, which is responsible for its short biological half-life.
`Electrophilic attack of a hydroxonium ion at the enol-ether double bond is the
`rate-limiting step in the metabolic degradation of the compound. Fluorination
`alpha to the labile group reduces the electron density on the enol-ether group
`and, thus, improves the stability of the molecule toward acid hydrolysis. Thus
`10,10-difluoro-13-dehydro-prostacyclin exhibits a half-life 150 times greater than
`
`

`
`P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`450
`
`PARK ¥ KITTERINGHAM ¥ O’NEILL
`
`that of PGI2 and has equal potency to the natural compound (37). Similarly, the int-
`roduction of a 7-fluoro group into PGI2 stabilized the enol by reducing the
`electron density of the double bond at C-5 through the inductive effects of flu-
`orine (38).
`Fluorination of natural hormones can lead to molecules with enhanced efficacy,
`and also to qualitative differences in activity. The changes in biological activity
`can be complex (pharmacodynamic and pharmacokinetic) and involve several
`chemical factors, including conformational changes in the steroid nucleus and
`altered receptor binding.
`The introduction of fluorine into the 9fi-position, of corticosteroids has a dra-
`matic reduction in some of the major routes of metabolism of cortisol. The most
`striking finding is the lack of oxidation of the 11fl-hydroxyl group to a ketone,
`which occurs rapidly with cortisol and leads to a loss of biological activity. Hence,
`in vivo, there is a shift in the enzymatic equilibrium between the biologically
`inactive 11-oxo compounds and the active reduced 11fl-hydroxy form (39).
`In vitro studies have shown that although cortisone and A-ring–reduced metabolites
`are the major products of cortisol metabolism in liver, 9fi-fluorocortisol undergoes
`preferential 6fl-hydroxylation and 20fl-reduction (40).
`We have investigated how introduction of fluorine may alter the balance be-
`tween metabolism of the A- and D-rings of estrogens (Figure 3). Introduction
`of fluorine into the 2-position can block 2-hydroxylation and bioactivation to a
`quinone (41, 42). Introduction of fluorine into the 16-position blocked not only
`C-16 hydroxylation but also dehydrogenation of the C-17 hydroxyl group (43).
`The restriction on D-ring metabolism was partly compensated for by enhancement
`of glucuronylation and A-ring hydroxylation.
`The 4-fluorophenyl group is usually resistant to aromatic hydroxylation, es-
`pecially at the 4-position, although aromatic hydroxylation at the unsubstituted
`positions does occur. Defluorination of the fluoromethyl group is extremely rare,
`although 5-trifluoromethyluracil is converted into 5-carboxyuracil in man with
`concomitant excretion of inorganic fluoride (44). Metabolism by cytochrome
`P450 enzymes does occur at the ortho and para positions. In molecules such as
`fluphenazine, which contain two rings that are chemically equivalent apart from the
`one being substituted with a CF3 group, hydroxylation occurs in the unsubstituted
`ring because of the deactivating effect of the CF3 group (45).
`The presence of the fluorine group in aromatic systems serves two purposes.
`First, the presence of fluorine per se can block oxidation at a specific position.
`Second, fluorine decreases the rate of reaction of the …-system of the benzene
`ring with activated cytochrome P450(FeO)3C
`(46). Nevertheless, cytochrome P450
`enzymes can attack at positions adjacent to the carbon-fluorine bond. Direct evi-
`dence for such a process comes from the demonstration that 1,4-difluorobenzene
`forms a significant amount of the NIH-shift product 2,4-difluorophenol (Figure 4).
`However, molecular orbital calculations show that increasing the extent of fluorine
`substitution further decreases nucleophilicity and, thus, reduces further the rate of
`reaction of di-, tri-, and tetrafluorobenzenes (46, 47). Evidence for such a pro-
`cess occurring in man is the detection of an NIH-shift metabolite of the novel
`
`

`
`P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`METABOLISM OF FLUORINATED DRUGS
`
`451
`
`Figure 3 Differential effects of A- and D-ring fluorination on the regioselective
`metabolism of estradiol (29, 41).
`
`quinoxazoline reverse trancriptase inhibitor GW420867X in human urine, using
`NMR and tandem mass spectrometry (48).
`The blockade of aromatic hydroxylation by the CF3 group may have undesirable
`consequences. Endoperoxides are a new class of antimalarials that destroy para-
`sites by selective bioactivation of the peroxide group, to carbon-centered radi-
`cals, within the parasite. In contrast, bioactivation in mammalian systems is
`extremely limited (49). Arteflene is a synthetic derivative of the natural product,
`which has a promising biological and pharmacokinetic profile. However, blockade
`of aromatic hydroxylation by fluorination promotes extensive bioactivation of the
`peroxide system to potentially toxic metabolites in mammalian systems (50).
`The strategic value of fluorine substitution in rational drug design is neatly il-
`lustrated by the development of the orally active inhibitor of cholesterol absorption
`SCH58235 from SCH48461 (51). Fluorine was introduced to block undesirable
`metabolic transformations and produce a lead compound with 50-fold greater po-
`tency in vivo (Figure 5).
`
`

`
`P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`452
`
`PARK ¥ KITTERINGHAM ¥ O’NEILL
`
`Figure 4 Mechanism of aromatic hydroxylation of fluorobenzenes. The NIH shift of
`fluorine (47, 48).
`
`THE USE OF FLUORINE SUBSTITUTION TO PREVENT
`METABOLIC BIOACTIVATION
`
`The physiological role of drug metabolism is that of detoxication. However, cer-
`tain biotransformations, and in particular those catalyzed by cytochrome P450,
`can produce chemically reactive metabolites. Normally enzymes such as epoxide
`hydrolase and glutathione transferase rapidly detoxify such metabolites. However,
`if such reactive metabolites interact with cellular macromolecules, they may cause
`carcinogenicity, apoptosis, necrosis, or hypersensitivity (52). Fluorine substitution
`has been used as a tool to investigate mechanisms of chemical toxicity and in the
`development of safer drugs.
`
`

`
`P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`METABOLISM OF FLUORINATED DRUGS
`
`453
`
`Figure 5 Examples of fluorine substitution in rational drug design (51, 64, 65, 67).
`
`Polycyclic aromatic hydrocarbons undergo metabolic bioactivation to arene ox-
`ides, which bind covalently to DNA and thus initiate carcinogenesis. Metabolic ac-
`tivation by the cytochrome P450 system is generally blocked at the carbon to which
`fluorine is attached. Miller & Miller (53) used fluorine substitution as a tool in the
`early mechanistic investigations of carcinogenicity and found that substitution of
`fluorine in the 3-position of 10-methyl-1,2-benzanthracene virtually abolished the
`carcinogenic activity of this hydrocarbon toward mouse skin. This technique has
`been widely used to define the chemistry of carcinogen activation (Figure 6).
`
`

`
`P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`454
`
`PARK ¥ KITTERINGHAM ¥ O’NEILL
`
`Figure 6 Use of fluorine substitu-
`tion to define the role of metabolic
`activation in chemical carcinogenic-
`ity (54, 55).
`
`Benzpyrene was the first member of the polycyclic aromatic hydrocarbon class
`of compounds for which the structures of both proximate and ultimate carcino-
`gens were defined. The metabolic pathway is catalyzed by CYP1A enzymes and
`epoxide hydrolysis leading to the ultimate carcinogen, 7.8-dihydroxy-9,10-epoxy-
`tetra-hydrobenzpyrene. Accordingly, it was found that 7-, 8-, 9-, and 10-fluorobenz
`(a)pyrenes were not tumorigenic in mice and that fluorine substitution, at each po-
`sition, had blocked formation of the respective diol-epoxides (54).
`The metabolic activation of 5-methylchrysene has been of interest because this
`carcinogen is typical of the class of methylated polynuclear aromatic hydrocarbons
`and is the most carcinogenic of all the methylchrysene isomers. Hecht et al (55)
`investigated the comparative carcinogenicity of seven derivatives fluorinated at
`the 1-, 3-, 6-, 7-, 9-, 11-, and 12-positions, respectively. Investigation of mouse
`skin tumor formation showed that the 6-, 7-, 9-, and 11-fluorinated derivatives
`were as carcinogenic as 5-methylchrysene, 12-fluoro was significantly less potent,
`and both 1-F and 3-F were inactive as complete carcinogens. The results are
`entirely consistent with the characterization of 5-methylchrysene-1,2-dihydrodiol-
`3,4-epoxide-DNA adducts (56, 57).
`Synthetic and natural estrogens have been associated in humans with a variety
`of vaginal, breast, hepatic, and cervical cancers. It has been suggested that both
`hormonal potency and oxidative metabolism may play a role in the carcinogenic-
`ity of steroid estrogens. The natural estrogens, estrone and estradiol, undergo
`extensive oxidation in the 2- and 4-positions by cytochrome P450 (CYP) en-
`zymes. The resulting catechols are readily oxidized to chemically reactive quinones
`
`

`
`P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`METABOLISM OF FLUORINATED DRUGS
`
`455
`
`and semiquinones, which form covalent bonds with proteins and DNA. Liehr (58)
`found that 2-fluoro-estradiol was as estrogenic as estradiol but noncarcinogenic
`in the Syrian hamster. In vivo studies (42, 43) showed that the presence of a 2-
`fluoro substituent diverted the metabolism of the steroid from 2-hydroxylation to
`glucuronidation, thus providing direct evidence for the role of bioactivation in the
`model of oestrogen carcinogenicity used by Liehr (58).
`The widely used analgesic paracetamol serves as a paradigm for the role of
`metabolic bioactivation in drug toxicity. Studies in knockout mice have clearly
`shown that formation of a reactive metabolite, N-acetylbenzoquinoneimine, is res-
`ponsible for the hepatic necrosis seen when the drug is taken in overdose (59, 60).
`Introduction of fluorine into the paracetamol molecule alters the oxidation poten-
`tial in a manner dependent on the number and position of the fluorine atoms (26).
`The presence of fluorine at the 2- and 6-positions increased the oxidation potential
`of paracetamol sufficiently to reduce the propensity of the molecule to undergo
`oxidative bioactivation in vivo and thereby reduced hepatotoxicity (61). Introduc-
`tion of fluorine did not affect the important detoxication pathways of glucuroni-
`dation or sulphation. It is interesting that 2,6-fluorine substitution also resulted in
`a loss of analgesic activity, indicating a role for oxidation in the analgesic activity
`of the drug (61).
`The 4-aminoquinoline antimalarial amodiaquine also contains the 4-aminophe-
`nol function. It is thought that the hemotoxicity and hepatotoxicity of this drug
`may be a result of oxidative metabolism to a chemically reactive quinoneimine
`metabolite (62, 63). The effect of systematic fluorine substitution of the aminop-
`henol ring was investigated with respect to (a) increasing the oxidative stability of
`the ring system and (b) reducing in vivo oxidation to potentially toxic quinoneimine
`metabolites. As was the case for paracetamol, 6-difluorination of the aminophe-
`nol ring of amodiaquine produced an analogue with significantly raised oxidation
`potential and reduced bioactivation in vivo. Isosteric replacement of the hydroxyl
`function with fluorine provided an analogue, fluoroamodiaquine, which had similar
`antimalarial potency to the parent drug but was not bioactivated to toxic metabolites
`in vivo. In contrast to the reduction in biological activity observed in the fluori-
`nated paracetamol series, fluorinated analogues of amodiaquine retained pharma-
`cological activity, which suggested that oxidation to quinonimine metabolites is
`not required for antimalarial activity (64). Fluorinated 4-aminoquinolines and
`8-aminoquinolines are being assessed as lead compounds for drug development
`(65, 66) (Figure 5).
`The effect of fluorine substitution on the metabolism and toxicity of drugs and
`use of such knowledge for the development of safer therapeutic agents is neatly
`illustrated by the flurane group of anaesthetics (68) (Figure 7). Methoxyflurane
`was widely used in clinical anaesthesia during the 1960s, until it was discovered
`that there was an association with nephrotoxicity. A high urine output syndrome
`leading to dehydration, and in some cases fatal renal failure, was related to the
`extensive (40%) metabolism of methoxyflurane and high serum concentrations of
`
`

`
`P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`456
`
`PARK ¥ KITTERINGHAM ¥ O’NEILL
`
`Figure 7 The toxicity and metabolism of general anesthetics.
`
`

`
`P2: FXY/GBC
`P1: FUM/GBC
`February 22, 2001
`12:24
`
`QC: aaa
`Annual Reviews
`
`AR126-18
`
`METABOLISM OF FLUORINATED DRUGS
`
`457
`
`inorganic fluoride. This theory has been questioned in recent years, since the intro-
`duction of sevoflurane, which also produces high serum fluoride but is not reported
`to cause nephrotoxicity (69). Methoxyflurane has been shown to be metabolized
`in man and animals to oxalic acid and free fluoride (70). Studies to date have shown
`only modest inorganic fluoride levels with enflurane and isoflurane, and there are
`only occasional reports of nephrotoxicity for enflurane and none at all for isoflu-
`rane (68, 71, 72). Increasing fluorine substitution results in a reduction in overall
`metabolism, and in specifically biotransformations leading to defluorination.
`In man, enflurane is metabolized to the fluoride ion, difluoromethoxydifluo-
`roacetic acid, and an unidentified acid metabolite. Metabolism is catalyzed by
`CYP2E1 and is stereoselective (73). The predicted products of oxidation of the
`difluoromethyl group, chlorofluoroacetic acid and oxalic acid, have not been
`detected (74). Inorganic fluoride and trifluoroacetic acid have been identified as
`end products of isoflurane metabolism (75). These products are thought to arise
`by a sequence that begins with insertion of an active oxygen atom into the bond
`connecting hydrogen to the ethyl fi-carbon and which is catalyzed by CYP2E1 in
`man (76, 77). Desflurane, in which the chlorine atom in isoflurane is replaced by
`a further fluorine atom, is excreted by the lungs and appears resistant to biotrans-
`formation. The oil gas partition coefficient for desflurane (18.7) is considerably
`less than that of isoflurane (91), which explains the more rapid rates of onset and
`offset of anesthesia of the former (78, 79).
`The first of the modern fluorinated anesthetics was halothane; however, its
`clinical use is now limited because of metabolism-associated liver toxicity. The
`National Halothane Study (80) identified two distinct forms of hepatotoxicity: The
`first, a mild form of transaminitis, occurred in 20% of cases but was reversible.
`In contrast, a small group (approximately 1 in 35,000) suffered severe hepatitis,
`characterized by massive liver necrosis, which could be fatal. The incidence of
`hepatitis increased on reexposure (to 1 in 3,700), indicative of an immunological
`aetiology. Two pathways of halothane metabolism occur (Figure 8): a reductive
`pathway, mediated by CYP3A4 and CYP2A6 (81), and oxidation, principally
`involving CYP2E1 (82). The reductive pathway generates free radicals, which,
`following further reduction and fluorine elimination, gives

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