throbber
ROLE OF PHARMACOKINETICS AND METABOLISM IN DRUG RESEARCH
`
`433
`
`required to displace the drug binding from the binding
`sites. Thus, in vitro studies designed to assess the pos-
`sibility of in vivo binding displacement must use undi-
`luted plasma and clinically relevant drug concentra-
`tions.
`
`The use of supratherapeutic drug concentrations or
`unusually low protein concentrations may produce bind-
`ing displacement in Vitro, but not in vivo. Zini et al.
`(1979) showed that indomethacin markedly decreased
`warfarin binding to human serum albumin in vitro at an
`indomethacin concentration of 100 ,u.M. However, Vesell
`et al. (1975) found no clinically significant displacement
`interaction between indomethacin and warfarin in vivo
`
`where the indomethacin concentration ranged from 0.08
`to 1.0 ,uM. Bupivacaine caused a 109% increase in the
`free fraction of mepivacaine in a solution of 011-acid gly-
`coprotein, but only a 9% increase in the free fraction of
`mepivacaine in plasma containing the same :11-acid gly-
`coprotein concentration (Hartrick et al., 1984). Both bu-
`pivacaine and inepivacaine are highly bound to high-
`aiiinity and low-capacity 0:1-acid glycoprotein and low-
`affinity and high-capacity albumin in plasma.
`Similar to metabo1ism—based drug interaction, the in-
`terpretation and extrapolation of in vitro displacement
`interaction data requires a good understanding of phar-
`macokinetic principles. Rowland and Aarons (Rowland,
`1980; Aarons and Rowland, 1981) have reviewed the
`theoretical and clinically relevant issues regarding drug
`displacement interactions. Depending on whether it is a
`low- or high-clearance drug, displacement interaction
`will cause different alterations in pharmacokinetics. As
`shown in equation [10], changes in the free fraction (fp)
`in plasma caused by displacement binding will affect
`drug distribution. As seen in equations [6] and [10], an
`increase in the fp of high—clearance drugs caused by
`binding displacement interaction will have little change
`in the clearance (cL), but will lead to an increase in the
`volume of distribution (Vd); hence, the elimination ti/4
`will increase. The tv; is related to both the cL and Vd as
`follows:
`
`0.693 X Vd
`CL
`
`.
`
`to,
`
`[12]
`
`For low—clearance drugs, both CL and Vd will increase
`with an increase in fp as shown in equations [5] and [10].
`Although the changes in cL and Vd may not exactly
`balance, the tn/1 will be affected to a much smaller degree
`compared with that of highly cleared drugs.
`Because only unbound drug is responsible for phar-
`macological effect, it is important to make a clear dis-
`tinction of the effects of displacement interaction on
`unbound and total drug concentrations in plasma. The
`simplest way of considering the effect of protein binding
`on the unbound and total drug concentration profiles is
`to examine the AUC. For low—clearance drugs, the AUC
`
`of unbound and total drug after intravenous dosing can
`be expressed as:
`
`AUC
`
`dose
`dose
`‘°“*‘ ‘ cL ‘ £1,-cL,,,,
`
`13
`
`1
`
`[
`
`and
`
`AUCunbound : AUCtotal ' fp 7
`— CI-‘int.
`
`dose
`
`On the other hand, the AUC of unbound and total
`drug of high-clearance drugs after intravenous adminis-
`tration can be expressed as:
`
`AUC
`
`_ dose _ dose
`total — CL — Qh
`
`[15]
`
`and
`
`dose - fp
`AUCunbound = AUCtotal ‘ fp = F
`Qh
`
`From equations [13] and [14], it is evident that the
`AUC of unbound drug for low—clearance drugs is inde-
`pendent of any change in fp if cLim is unaffected by
`displacement interaction, whereas an increase in the fp
`caused by binding displacement interactions will result
`in a decrease in the AUC of total drug. On the other
`hand, exactly the opposite situation occurs with a high-
`clearance drug in which the clearance and, hence, total
`drug concentration is unaffected by changes in plasma
`protein binding, whereas the unbound drug concentra-
`tion increases as a result of increased fp as shown in
`equations [15] and [16]. Figure 7 depicts the effects of
`displacement from protein binding sites on the steady-
`state unbound and total drug concentrations of low— and
`high-clearance drugs during intravenous
`infusion
`(Aarons, 1986).
`After oral administration, the AUC of unbound and
`total drug, regardless of whether it is a high— or low—
`clearance drug, can be expressed as equation [17], which
`
`C
`
`3%
`'23:

`
`(8)
`
`T _
`
`Total
`
`(bl
`
`\.
`
`./
`
`T Total
`
`T
`
`_,-—--"--Unbound
`
`l‘-._L,|_n_b_ound _____
`.. ._,
`
`Time
`
`Time
`
`FIG. 7. The effect of displacing a low—clearance drug ( a) or high-
`clearance drug (bl, given chronically, from plasma protein binding
`sites. Displacement is produced by infusing a drug that displaces the
`first drug, starting from the arrowed point. Reproduced with permis-
`sion from Aarons (1986).
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2028 - 0031
`
`

`
`434
`
`LIN AND LU
`
`is similar to equation [8]:
`
`AUCt0ta1 =
`
`F ' dose
`CL
`
`dose
`= [17]
`
`and
`
`A-Ucunbound = AI-lctotal ' fp =
`
`dose
`
`From equations [17] and [18], the AUC of unbound
`drug after oral dosing is insensitive to the changes in the
`fp, whereas the AUC of total drug will decrease when the
`fp increases as a result of displacement interactions.
`Because a significant change in the unbound AUC of
`drugs after oral dosing is not expected, and because most
`drugs are given orally, the displacement interactions
`rarely have significant clinical effects
`(Mackichan,
`1984,1989; Sellers, 1979). When changes in binding are
`associated with clinical effects, it has almost always
`been found that this is the result of a change in the cLm,
`caused by a mechanism quite independent of plasma
`protein binding as indicated in equation [18]. Warfarin—
`phenylbutazone interaction is a good example. When
`concomitantly administered with warfarin, phenylbuta-
`zone caused profound potentiation of a hypoprothrom—
`binemic response (Sellers, 1986). Although phenylbuta-
`zone is known to displace warfarin from plasma
`proteins, it is clear from equation [18] that the hypopro-
`thrombinemic effect was not caused by binding displace-
`ment of phenylbutazone, because the unbound concen-
`tration of warfarin should not be changed. Later, it was
`found that phenylbutazone stereoselectively inhibited
`the metabolism of S—warfarin (Lewis et a1., 1974;
`O’Reilly et al., 1980). Thus, the metabolism inhibition,
`rather than binding displacement, causes the serious
`hemorrhagic complications of warfarin-phenylbutazone
`interaction. Similarly,
`although sulfaphenazole
`is
`known to displace tolbutamide from plasma proteins,
`the inhibitory effect of sulfaphenazole on the metabo-
`lism of tolbutamide is responsible for the serious hypo-
`glycemic reactions (Christensen et al., 1963).
`Whereas the unbound concentration after oral dosing
`is unaffected by displacement interaction, the transient
`increase in the unbound drug concentration occurring
`immediately after introduction of the displacing drug
`sometimes may be of clinical significance (Levy, 1976).
`Elie and Levy (1979a,b) reported that rapid intravenous
`infusion of salicylic acid or sulfisoxazole resulted in a
`transitory increase of unbound bilirubin concentration
`in rats. This suggests that the fatal kernicterus seen in
`the newborn after administration of sulfonamides may
`be due to a transitory increase in unbound bilirubin in
`the brain. In addition, the displacement interactions will
`be of clinical significance for high-clearance drugs after
`intravenous dosing. As shown in figure 7, a substantial
`increase in the unbound concentration may occur.
`
`V. Interindividual Variability: A Critical Issue in
`Drug Development
`
`From the market point of view, it is desirable that the
`dosage can be generalized to provide drugs for the treat-
`ment of a large number of patients. In reality, the gen-
`eralization may work for most patients, but not for all.
`The standard dosage regimen of a drug may prove to be
`therapeutically effective in most patients, ineffective in
`some patients, and toxic in others. Variability in drug
`response becomes an important problem in drug therapy
`for drugs that have a narrow therapeutic window. War-
`farin is a good example. There is a wide range of daily
`dose requirements (<2 mg—>11 mg‘) of warfarin needed
`to produce a similar prothrombin time (Koch-Weser,
`1975). Variability in drug response can be broadly di-
`vided into pharmacokinetic and pharmacodynamic
`bases. Sources of pharmacokinetic variability include
`genetics, disease, age, and environmental factors (Brei-
`mer, 1983).
`
`A. Pharmacokinetic Variability
`
`The patient’s exposure to drug is a crucial determi-
`nant of the drug’s actions, and therefore its efficacy and
`safety. The term “drug exposure” is defined as the time
`course of the concentration of the drug and its active
`metabolites in plasma. The time course of drug concen-
`tration is governed by absorption, distribution, metabo-
`lism, and excretion. All these processes can contribute to
`pharmacokinetic variability.
`I. Variability in absorption. Variation in absorption is
`one of the major sources of pharmacokinetic variability.
`An impression prevails that the degree of variability in
`the amount of drug reaching the systemic blood circula-
`tion is minimized if a drug‘ has high bioavailability,
`whereas the risk of greater variation in the amount
`taken up is increased if a drug has low bioavailability.
`However, all too often, the degree of variability in ab-
`sorption is similar for drugs of high and low bioavail-
`ability. The causes of absorption variability include
`pharmaceutical formulation, gastrointestinal physiol-
`ogy, and first—pass metabolism.
`Being absorbed primarily from the upper part of the
`small intestine, oral absorption of drugs is often affected
`by the gastric emptying time and small intestinal motil-
`ity, which vary considerably between individuals
`(Meyer, 1987; Weisbrodt, 1987). Usually, rapid gastric
`emptying results in rapid drug absorption. Changes in
`gastric emptying normally affect the rate of absorption
`but do not affect the amount of drug absorbed unless the
`drug is chemically unstable in the stomach or associated
`with saturable first-pass metabolism (Nimmo, 1976).
`Dietary factors are also important sources of absorp-
`tion variability that can be accounted for. The influence
`of food on the absorption of drugs is largely unpredict-
`able. Food may enhance or reduce the absorption of some
`drugs while having no effect on others, depending not
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2028 - 0032
`
`

`
`ROLE OF‘ PHARMACOKINETICS AND METABOLISM IN DRUG RESEARCH
`
`435
`
`only on the composition and volume of the meal or the
`drink, but also on the physicochemical properties of
`drugs. For example, absorption of the lipophilic drugs
`griseofulvin and sulfamethoxydiazine increased consid-
`erably when given with a high-fat meal (Crouse, 1961;
`Kaumeier, 1979). Amoxicillin, a poorly soluble antibi-
`otic, was absorbed to a greater extent when swallowed
`with 250 mL water (Welling et al., 1977). In addition,
`dietary factors have been shown to alter drug-metabo-
`lizing enzyme activity, leading to changes in first—pass
`metabolism and bioavailability. Both charcoal—broiled
`beef and a high-protein, low-carbohydrate diet cause an
`increase in theophylline and antipyrine metabolism
`(Kappas et al., 1978, 1976). Certain vegetables, includ-
`ing brussel sprouts, cabbage, broccoli, and cauliflower,
`contain chemicals that induce drug-metabolizing en-
`zyme activities (Pantuck, 1979). Because the diet is so
`different among patients, it is conceivable that the ef-
`fects of food account for a substantial part of the absorp-
`tion variability.
`Ironically, most clinical studies de-
`signed to address the question as to whether food intake
`affects drug absorption were conducted in healthy vol-
`unteers with or without a more or less standardized
`
`meal. Thus, such information may not be meaningful,
`sometimes even misleading.
`The problem of absorption variability is complicated
`further by diseases. Hepatic disease may influence the
`oral bioavailability of drugs highly metabolized by the
`liver. The bioavailability of propranolol was increased
`significantly from 35% in normal subjects to 54% in
`cirrhotic patients, and the steady-state unbound pro-
`pranolol concentration increased from 7.5 ng/mL to 22
`ng/mL (Wood et al., 1978). The increased bioavailability
`was due mainly to a decrease in hepatic first—pass me-
`tabolism.
`
`2. Variability in binding. As discussed earlier, plasma
`protein binding is an important determinant of the
`drug’s disposition and actions. The fp varies widely
`among drugs, and often (for highly bound drugs) among
`individuals. Differences in binding among drugs arise
`primarily from differences in their affinities for binding
`proteins, whereas differences in binding among individ-
`uals are due mainly to qualitative or quantitative differ-
`ences in binding proteins. Nevertheless, interindividual
`variability in drug binding is generally less as compared
`with that in other pharmacokinetic processes such as
`absorption and metabolism (Yacobi and Levy, 1977;
`Barth et al., 1976).
`a1-Acid glycoprotein is a major determinant for the
`binding of basic drugs in plasma (Piafsky and Borga,
`1977; Piafsky et al., 1978). Several inflammatory states
`(infections, rheumatic disorders, and surgical injury)
`and pathological conditions (myocardial infarction, ma-
`lignancies, and nephritis) elevate the plasma concentra-
`tion of al-acid glycoprotein (Abramson, 1982; Freilich
`and Giardini, 1984). Furthermore, ac,-acid glycoprotein
`is known to be inducible. Treatment with phenobarbital
`
`resulted in a substantial increase in plasma concentra-
`tion of oz1—acid glycoprotein (Abramson, 1991). Because
`there is a strong correlation between the binding of basic
`drugs and the plasma levels of 0:1-acid glycoprotein
`(Lunde et al., 1986; Sjtiqvist and Koike, 1986), an eleva-
`tion of this protein will increase the binding of basic
`drugs.
`In contrast to the elevation of 041-acid glycoprotein,
`hypoalbuminemia is always associated with a large va-
`riety of pathological conditions, including liver cirrhosis,
`renal failure, nephrotic syndrome, chronic inflamma-
`tion, malignancies, and sepsis (Gugler and Jensen,
`1986). In hypoalbuminemia, the binding of acidic drugs
`is reduced, and the decrease is related to a decrease in
`the plasma albumin concentration. Although normal
`subjects have a plasma albumin concentration of at least
`35 mg/mL, plasma albumin concentrations can be as low
`as 10 mg/mL in patients with nephrotic syndrome.
`In addition to the quantitative changes in plasma
`protein concentrations, qualitative structural changes of
`plasma proteins also alter the binding of drugs. High
`doses of acetylsalicylic acid can acetylate serum albumin
`and modify its binding sites (Hawkins et al., 1968).
`Cyanate, spontaneously formed from urea, carbamy-
`lates lysine residues on the albumin molecules and de-
`creases the binding of acidic drugs in uremic patients
`(Erill et al., 1980). Furthermore,
`in uremic patients,
`retained endogenous acids that are highly protein bound
`can displace the binding of drugs from proteins. Collier
`et al. (1986) have identified one of these acids, 3—car—
`boxy—4-methyl-5-propyl-2-furanpropanic acid, as a po-
`tent displacer of drug binding. From these data, it is
`clear that disease states also are the main sources of
`
`binding variability.
`Genetically determined variations in amino acid se-
`quences of serum albumin and 0:1-acid glycoprotein also
`can contribute to binding variability. To date, more than
`30 apparently different genetic variants of human se-
`rum albumin have been identified. Only approximately
`half of these variants have been absolutely character-
`ized by peptide mapping and sequence determination
`(Eap and Baumann, 1991). Kragh—Hansen et al. (1990)
`have compared the binding affinities (association con-
`stants) of warfarin, salicylate, and diazepam to five vari-
`ants of human serum albumin with known mutations.
`
`The association constants of all three drugs to albumin
`Canterbury (313 Lys—>Asn) and to albumin Parkland
`(365 Asp—>His) decreased substantially by a factor of 4-
`to 10-fold, whereas the binding affinity to albumin Ve-
`rona (570 Glu—>Lys) was unchanged. These results sug-
`gest that the region 313-365 seems to exert important
`effects on the binding of drugs, whereas the mutation
`570 near the C-terminus does not affect drug binding.
`Three main variants of a1—acid glycoprotein, namely
`ORM1 F1, ORM1 S, and ORM2 A, have been fully char-
`acterized (Eap and Baumann, 1991). Among the three
`variants, ORM2 A is the most important variant associ-
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2028 - 0033
`
`

`
`436
`
`LIN AND LU
`
`ated with the binding of basic drugs. Eap et al. (1990)
`have determined the in vitro binding of d-methadone,
`L—methadone and dl—methadone in plasma samples from
`45 healthy subjects. The concentrations of oil-acid glyco-
`protein variants also were measured. Using multiple
`stepwise regression analysis, significant correlations
`were obtained between the binding of methadone and
`the total 0:1-acid glycoprotein or ORM2 A concentrations,
`but only a weak correlation between the binding and
`ORM1 S concentrations, and no correlation between the
`binding and ORM1 F1 concentrations were found. The
`frequencies for the three phenotypes, i.e., ORM1 Fll
`ORM2 A, ORM1 FUORM1 S/ORM2 A, and ORM1
`S/ORM2 A were found to be 33.7, 50.5, and 15.2%, re-
`spectively,
`in a Swiss population (Eap et al., 1988).
`These results suggest genetically determined variations
`in O!1'3.C1d glycoprotein could be a major source of vari-
`ability in the binding of basic drugs.
`3. Variability in excretion. Although metabolism is the
`major route of elimination for most drugs, some drugs
`are excreted mainly as unchanged drug via the kidneys
`and liver. Both biliary and renal excretion correlate to
`their function. Ceftazidime, a cephalosporin antibiotic,
`is excreted mainly by the kidneys. The total clearance of
`ceftazidime correlated linearly with creatinine renal
`clearance in patients with varying degrees of renal func-
`tion (Van Dalen et al., 1986). Similarly, a strong corre-
`lation should exist between the clearance and hepatic
`function if a drug is excreted mainly by the liver. The
`biliary excretion of indocyanine green correlated well
`with hepatic function in cirrhotic patients (Kawasaki et
`al., 1985).
`Many endogenous organic acids are accumulated in
`the plasma of patients with renal dysfunction. These
`endogenous organic acids may inhibit the transport of
`certain drugs in the liver. The hepatic uptake and biliary
`excretion of bromosulfophthalein and dibromosulfoph-
`thalein are decreased in rats with acute renal failure
`
`(Silberstein et al., 1988). These data demonstrate that
`variations in hepatic and renal function, particularly in
`patients with hepatic and renal disorders, contribute
`significantly to pharmacokinetic variability.
`Reabsorption is one of the important factors governing
`renal clearance of drugs. Lipophilic drugs tend to be
`extensively reabsorbed, whereas hydrophilic drugs do
`not. Urine flow and pH have a substantial effect on the
`renal clearance of a drug that is mostly reabsorbed. An
`increase in the urine flow will result in a decrease in
`
`reabsorption, leading to an increase in renal clearance.
`The renal clearance of theophylline increases with in-
`creasing urine flow rate (Tan-Liu et al., 1982). Similarly,
`the renal clearance of phenobarbital is also dependent
`on the urine flow rate (Linton et al., 1967).
`Unlike plasma that has a narrow pH range of 7.3 to
`7.5, urine pH ranges from 4.5 to 8.5. Thus, the urine pH
`is an additional factor that influences the reabsorption
`of drugs that are weak acids and bases. The renal ex-
`
`cretion of salicylic acid is markedly pH-dependent. Re-
`nal excretion of salicylate increases more than ten-fold
`as the urinary pH increases from 5 to 8 (Macpherson et
`al., 1955). In contrast, the renal clearance of quinidine
`has been shown to diminish with increasing urinary pH
`(Gerhardt et al., 1969). Drugs that show pH—sensitive
`reabsorption also generally Show flow-rate dependence.
`Clearly, variations in urine flow and pH also contribute
`significantly to excretion variability.
`
`B. Pharmacogenetics of Drug Metabolism
`
`All enzymes involved in the metabolism of drugs are
`regulated by genes and gene products. Because of evo-
`lutionary and environmental factors, there is a remark-
`able degree of genetic variability built into the popula-
`tion. Thus, the genetic factor represents an important
`source of interindividual variation in drug metabolism.
`Mutations in the gene for a drug—metabolizing enzyme
`result in enzyme variants with higher, lower, or no ac-
`tivity or may lead to a total absence of the enzyme.
`Therefore, it is not unusual to find a ten-fold or as much
`as a 50-fold difference in the rate of drug metabolism
`among patients.
`With the technological breakthroughs in molecular
`biology, significant progress has been made in under-
`standing the role of genetic polymorphisms in drug me-
`tabolism. The major polymorphisms that have clinical
`implications are those related to the oxidation of drugs
`by CYP2D6 and CYP2C19 (Meyer et al., 1990b, 1992;
`Meyer, 1994; Wilkinson et al., 1989; Broly and Meyer,
`1993; Alvan et al., 1990), acetylation by N-acetyltrans-
`ferase (Evans, 1992), and S-methylation by thiopurine
`methyltransferase (Weinshiboum, 1992; Creveling and
`Thakker, 1994). Individuals who inherit an impaired
`ability to catalyze one or more of these enzymatic reac-
`tions may be at an increased risk of concentration-re-
`lated adverse effects and toxicity.
`1. Polymorphism in drug oxidation. CYPZD6 polymor-
`phism is perhaps the most studied genetic polymor-
`phism in drug metabolism. Since its discovery in 1977
`(Mahgoub et al., 1977), hundreds of studies have been
`carried out to investigate the nature of CYP2D6 poly-
`morphism,
`the mode of inheritance, and the conse-
`quences of the deficient trait on drug disposition and
`pharmacological effects. This polymorphism divides the
`populations into two phenotypes: EM and PM. Approx-
`imately 5 to 10% of individuals in Caucasian populations
`are the PM phenotype, compared with only 1 to 2% of
`individuals in Asian populations. To date, more than 50
`drugs,
`including antidepressants, antipsychotics, and
`cardiovascular drugs, are known to be catalyzed primar-
`ily by CYP2D6 (Parkinson, 1996).
`Clinical studies have demonstrated that the PMs of
`
`CYP2D6—mediated drugs represent a high—risk group
`with a propensity to develop adverse effects. The dispo-
`sition of haloperidol, a potent neuroleptic, was studied in
`a panel of six EMS and six PMs of debrisoquine (Llerena
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2028 - 0034
`
`

`
`ROLE OF‘ PHARMACOKINETICS AND METABOLISM IN DRUG RESEARCH
`
`437
`
`et al., 1992). The PMS that received 4 mg of haloperidol
`developed neurological side effects, whereas at the same
`dose, the EMS experienced only mild side effects, such as
`tiredness, difficulty concentrating, and some restless-
`ness. The PMS eliminated haloperidol significantly
`slower than the EMS, and the high plasma concentra-
`tions of haloperidol might, therefore, be associated with
`the side effects observed in the PMS. Similarly, an in-
`creased risk of side effects also was observed in the PMS
`
`of debrisoquine when taking other neuroleptics, such as
`perphenazine (Dahl—PuuStinen et al., 1989) and thiorid—
`azine (Meyer et al., 1990a). Both drugs also are metab-
`olized by CYP2D6.
`I antiarrhythrnic
`Similarly, propafenone, a class
`agent, is metabolized by CYP2D6. The relationship be—
`tween debrisoquine phenotype and pharmacokinetics
`and pharmacodynamics of propafenone was studied in
`28 patients (22 EMS and 6 PMS) with chronic ventricular
`arrhythmias (Siddoway et al., 1987). Steady—state con-
`centrations of propafenone in plasma were found to be
`significantly higher in PMS than EMS. These higher
`concentrations were associated with a greater incidence
`of CNS side effects in the PMS (67%), relative to the EMS
`(14%).
`
`The effects of CYP2D6 polymorphism on pharmaco-
`logical responses can be quite complex, depending on
`whether the parent drug or metabolites, or both, are
`pharmacologically active. Encanide, a class I antiar-
`rhythmic, is a good example. CYP2D6 O-demethylates
`encanide to a metabolite that is 6 to 10 times more
`
`potent than the parent drug in blocking sodium chan-
`nels. In both PMS and EMS, standard doses of this drug
`tend to produce similar therapeutic responses, because
`relatively high parent drug concentrations in the former
`are matched by relatively high active metabolite concen-
`trations in the latter (Buchert and Woosley, 1992). Sim-
`ilarly, both propafenone and its 5-hydroxy propafenone
`metabolite are pharmacologically active. The metabo-
`lism of propafenone to 5-hydroxy propafenone is grossly
`impaired in the PMS, resulting in very low or no levels of
`this active metabolite. However, as with encanide, there
`were no significant differences between EMS and PMs in
`an effective propafenone dosage or frequency of antiar-
`rhythmic response (Siddoway et al., 1987). This again
`can be explained by the compensatory effect of the active
`metabolite of 5—hydroxy propafenone, present in the
`plasma of EMS but not in that of PMS.
`Codeine is metabolized extensively by glucuronida-
`tion; the O-demethylation of codeine to morphine is a
`minor pathway that is mediated by CYP2D6 (Chen et
`al., 1988). As only a small fraction of the drug is metab-
`olized by the O-demethylated pathway, PMS are not
`expected to have an altered disposition of codeine rela-
`tive to EMS. As anticipated, plasma concentrations of
`codeine were similar in PMS and EMS, but measurable
`concentrations of morphine, its more analgesic O-dem-
`ethylation product, were only detected in EMS (Sindrup
`
`et al., 1991). Consequently, codeine increased the pain
`thresholds to copper vapor laser stimuli in EMS, but not
`in PMs, affirming the functional importance of the co-
`deine-morphine biotransformation for codeine analge-
`sia.
`
`CYP2C19 also exhibits genetic polymorphism in drug
`metabolism. The incidence of the PM phenotype in pop-
`ulations of different racial origin varies; approximately 2
`to 6% of individuals in the Caucasian populations are
`the PM phenotype, as are 14 to 22% in the Asian popu-
`lations (Wilkinson et al., 1992; Kalow and Bertilsson,
`1994). Although it is expected that PMS will have higher
`plasma
`concentrations
`of drugs metabolized by
`CYP2C19 than EMS and experience an increase in ad-
`verse effects, the clinical implications of CYP2C19 poly-
`morphism have not been thoroughly characterized. Con-
`trary to CYP2D6, CYP2C19 has been studied far less,
`which is reflected by the much shorter list of known
`drugs characterized by CYP2C19 than by CYP2D6 (Par-
`kinson, 1996).
`Diazepam is demethylated by CYP2C19 in humans
`(Anderson et al., 1990). The disposition of diazepam has
`been Studied in 13 Caucasians of the EM phenotype and
`3 Caucasians of the PM phenotype (Bertilsson et al.,
`1989). The plasma clearance of diazepam in the EMS
`was more than 2 times that in the PMS (11.0 and 5.0
`
`mL/min, respectively), whereas the iv; in the EMS was
`shorter than that in the PMS (59 and 128 h, respective-
`ly). The difference in the plasma clearance appeared to
`be related to formation of the desmethyl metabolite.
`Omeprazole, a proton pump inhibitor, is metabolized
`(by CYP2C19) by hydroxylation and oxidation of the
`sulfoxide group to a sulfone (Anderson et al., 1990). The
`metabolism of omeprazole has been studied in the EMS
`and PMS of S-mephenytoin selected from phenotyped
`healthy Swedes and Chinese (Andersson et al., 1992).
`The plasma concentrations of omeprazole and its metab-
`olites were determined after a single oral dose (20 mg).
`The AUC of omeprazole was substantially higher in PMS
`than in EMS in both Swedes (11.1 and 0.94 p'.M'h) and
`Chinese (13.3 and 2.6 MM-h). Although the AUC was not
`different between Swedish and Chinese PMS, there was
`a significant interethnic difference in EMS. The fact that
`the AUCS in Chinese EMS were 3 times higher than
`those of the Swedish EMS might be due to the higher
`proportion of heterozygotes in the Chinese.
`From a genetic point of view, the different enzyme
`polymorphisms in drug metabolism are inherited inde-
`pendently. However, an inherited deficiency of different
`drug—metabolizing enzymes could occur simultaneously
`on the basis of probability. A population study of mephe-
`nytoin hydroxylation and debrisoquine hydroxylation
`was carried out in 221 unrelated normal volunteers
`
`(Kiipfer and Preisig, 1984). Twelve (5%) of them exhib-
`ited defective hydroxylation of mephenytoin, and 23
`(10%) could be identified as PMS of debrisoquine. Among
`these 35 subjects, 3 (1 female and 2 males) displayed
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2028 - 0035
`
`

`
`438
`
`LIN AND LU
`
`simultaneously both defects of mephenytoin and de-
`brisoquine hydroxylation.
`Propranolol is hydroxylated by CYP2D6 and N—deal—
`kylated by CYP2C19. The relative contributions of these
`two isoforms to propranolol metabolism have been stud-
`ied in a panel of phenotyped normal volunteers (Ward et
`al., 1989). Six subjects were EMS of both mephenytoin
`and debrisoquine. Four subjects were PMs of debriso-
`quine but rapid metabolizers of mephenytoin. Five sub-
`jects were PMs of mephenytoin but rapid metabolizers of
`debrisoquine, and one subject had a deficiency for both
`debrisoquine and mephenytoin. PMs of either mepheny-
`toin or debrisoquine had a similar disposition of pro-
`pranolol to that of EMs, whereas the subject with both
`mutations had a tie 2 times longer than the other sub-
`jects’.
`In view of the examples presented above, it is clear
`that genetic polymorphism in drug metabolism could
`lead to clinically significant differences in pharmacoki—
`netics and pharmacological responses of some patients
`and therefore might result in adverse effects or thera-
`peutic failure. Thus, drugs metabolized by enzymes ex-
`hibiting genetic polymorphism are considered to be un-
`desirable. However,
`the development of
`a drug
`sometimes is prematurely terminated based solely on
`the fact that its metabolism is polymorphic. To avoid
`premature termination, the clinical relevance of genetic
`polymorphism must be assessed carefully. Pharmacoki-
`netic differences between phenotypes are most relevant
`for drugs with narrow therapeutic indices. For com-
`pounds with a variability of plasma concentrations out-
`side the therapeutic range that is not associated with
`adverse effects, polymorphic metabolism will be of less
`or little concern. Propranolol is a typical example. De-
`spite the critical involvement of CYP2D6 and CYP2C19
`polymorphism in the metabolism of propranolol, this
`drug is quite safe clinically. Another important factor in
`determining the go/no—go decision is the overall benefit-
`to-risk ratio. If the benefit of a drug is significantly
`greater than its risk, and dosage can be titrated by direct
`clinical monitoring, then polymorphic metabolism is of
`less consequence.
`2. N~Acetylo;tion polymorphism. Acetylation is an im-
`portant route of elimination for a large number of hydr-
`azine and arylamine drugs (Weber et al., 1990). The
`N—acety1transferase (NAT) polymorphism in humans
`was discovered as a result of studying the rate of isoni-
`azid elimination in tuberculous patients in 1960 (Evans
`et al., 1960). The patients could be classified as slow and
`rapid acetylators based on their plasma concentrations
`of isoniazid. In addition to isoniazid, sulfamethazine,
`hydralazine, procainamide, dapsone, and nitrazepam
`also are polymorphically acetylated (Evans, 1992, 1989).
`The proportions of rapid and slow acetylators vary con-
`siderably between ethnic groups. For example, the per-
`centage of slow acetylators in Egyptians and Mideast-
`erners is 80 to 90%, whereas in Asian populations, it is
`
`only 10 to 20%, with European and North American
`Caucasians having an intermediate value of 40 to 70%
`(Evans, 1989). On the other hand, other N-acetylated
`compounds, such as p-aminobenzoic acid and p-amin-
`osalicylic acid, were unable to distinguish rapid and slow
`acetylators in vivo and in vitro (Evans, 1989). These
`compounds are,
`therefore, classified as monomorphic
`substrates.
`
`Although the acetylation polymorphism was sus-
`pected for nearly 40 years, the molecular mechanics
`underlying this polymorphism were not known until
`recently. Meyer and his colleagues (Blum et al., 1990;
`Grant et al., 1991) have successfully cloned three human
`genes: NAT1, NAT2, and a related pseudogene, NATP.
`The discovery of two separate genes encoding NAT1 and
`NAT2 resolved the old question on monomorphic and
`polymorphic substrates. NAT2 has a high affinity for
`polymorphic substrates, whereas NAT1 has a high af-
`finity for monomorphic substrates. Mutations of the
`NAT2 gene result in slow acetylation. The most common
`acetylator allele in Ca

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