throbber
0090-9556/03/3112-1481–1498$7.00
`DRUG METABOLISM AND DISPOSITION
`Copyright © 2003 by The American Society for Pharmacology and Experimental Therapeutics
`DMD 31:1481–1498, 2003
`
`Vol. 31, No. 12
`1140/1106831
`Printed in U.S.A.
`
`THE USE OF DEUTERIUM ISOTOPE EFFECTS TO PROBE THE ACTIVE SITE
`PROPERTIES, MECHANISM OF CYTOCHROME P450-CATALYZED REACTIONS, AND
`MECHANISMS OF METABOLICALLY DEPENDENT TOXICITY
`
`SIDNEY D. NELSON AND WILLIAM F. TRAGER
`
`Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, Washington
`
`(Received April 2, 2003; accepted July 1, 2003)
`
`This article is available online at http://dmd.aspetjournals.org
`
`ABSTRACT:
`
`Critical elements from studies that have led to our current under-
`standing of the factors that cause the observed primary deuterium
`isotope effect, (kH/kD)obs, of most enzymatically mediated reac-
`tions to be much smaller than the “true” or intrinsic primary deu-
`terium isotope effect, kH/kD, for the reaction are presented. This
`new understanding has provided a unique and powerful tool for
`probing the catalytic and active site properties of enzymes, partic-
`ularly the cytochromes P450 (P450). Examples are presented that
`
`illustrate how the technique has been used to determine kH/kD, and
`properties such as the catalytic nature of the reactive oxenoid
`intermediate, prochiral selectivity, the chemical and enzymatic
`mechanisms of cytochrome P450-catalyzed reactions, and the rel-
`ative active site size of different P450 isoforms. Examples are also
`presented of how deuterium isotope effects have been used to
`probe mechanisms of the formation of reactive metabolites that
`can cause toxic effects.
`
`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on April 30, 2021
`
`substrate concentrations. The disparity between kH/kD and (kH/kD)obs
`is a consequence of the multistep nature (substrate binding, debinding,
`product release, etc.) of the catalytic cycle of enzymatically mediated
`reactions. If deuterium isotope effects are to be successfully applied to
`such reactions, then the relationship between kH/kD and (kH/kD)obs
`must be known, since experiment can only yield (kH/kD)obs. Fortu-
`nately,
`the relationship between kH/kD and (kH/kD)obs has been
`largely clarified (Northrop, 1975, 1978, 1981a; Cleland, 1982); and
`how partially rate-limiting steps exclusive of the bond-breaking step
`can mask kH/kD by lowering the magnitude of (kH/kD)obs is now
`clearly understood.
`Even the simplest of enzymatic reactions is composed of three
`components (Northrop, 1981b). The first involves substrate combin-
`ing with enzyme to form an enzyme-substrate complex. Rate con-
`stants k12 and k21 govern the process. The second component is the
`catalytic component that transforms the enzyme-substrate complex
`into an enzyme-product complex with rate constant k23 (Reaction 1).
`In this simplest of cases the catalytic step is considered to be irre-
`versible. The third component is the product release step that entails
`dissociation of the complex to free enzyme and product with rate
`constant k31. Equations 1 to 3 define the kinetic expressions for the
`maximum velocity, V, the Michaelis constant, K, and the ratio of the
`two, V/K (Northrop, 1975, 1981b). In analyzing the kinetic
`
`Historically, the determination of deuterium isotope effects has
`been a powerful tool to help unravel the intricacies of carbon-hydro-
`gen bond cleavage and define the mechanism of specific chemical
`reactions. The intrinsic primary isotope effect, kH/kD, for a reaction is
`the magnitude of the isotope effect on the rate constant for the
`bond-breaking step (C–H versus C–D) of the reaction and is related to
`the symmetry of the transition state for that step. The larger the
`isotope effect, the more symmetrical the transition state, with the
`theoretical limit being 9 at 37°C in the absence of tunneling effects
`(Bell, 1974). The chemical events leading to the transition state and
`subsequent formation of products are the descriptors of a chemical
`mechanism. It is the relationship of kH/kD to transition state that
`provides mechanistic insight. Thus, kH/kD is the quantity that needs to
`be known, and for homogenous chemical reactions, the experimen-
`tally observed deuterium isotope effect, (kH/kD)obs, where (kH/kD)obs
`is defined as the ratio of a kinetic parameter such as Vmax or Vmax/Km
`obtained from a nondeuterated substrate to a deuterated substrate, is
`identical to kH/kD.
`This is generally not true of enzymatically mediated reactions and
`is the reason for the much less successful application of deuterium
`isotope effects to such reactions until the system was better under-
`stood. The experimentally observed isotope effect, (kH/kD)obs, was
`invariably found to be much smaller than kH/kD, the intrinsic isotope
`effect for that reaction, thereby obscuring both meaning and mecha-
`nism. Even more perplexing was the observation that (kH/kD)obs for
`the same enzymatic reaction could vary with different experimental
`designs. For example, (kH/kD)obs determined at saturating substrate
`concentrations could differ from values determined at decreasing
`
`Address correspondence to: William Trager, Department of Medicinal Chem-
`istry, University of Washington, 1959 NE Pacific Street, Health Sciences Bldg.,
`H-364C, Seattle, WA 98195-7631. E-mail: trager@u.washington.edu
`
`1481
`
`V ⫽
`
`K ⫽
`
`k23k31Et
`k23 ⫹ k31
`k31(k12 ⫹ k23)
`k12(k23 ⫹ k31)
`k12k23Et
`k21 ⫹ k23
`
`V/K ⫽
`
`(1)
`
`(2)
`
`(3)
`
`Auspex Exhibit 2006
`Apotex v. Auspex
`IPR2021-01507
`Page 1
`
`

`

`DV ⫽ VH/VD ⫽
`
`k23H/k23D ⫹ k23H/k31
`1 ⫹ k23H/k31
`k23H/k23D ⫹ k23H/k21
`D(V/K) ⫽ (V/K)H/(V/K)D ⫽
`1 ⫹ k23H/k21
`DV and D(V/K) are short-form terms introduced by Northrop (1975)
`for VH/VD and (V/K)H/(V/K)D, respectively. If eqs. 4 and 5 are
`rewritten in general form as eqs. 6 and 7, as proposed by Northrop
`(1975), the factors
`
`(4)
`
`(5)
`
`(6)
`
`(7)
`
`1482
`NELSON AND TRAGER
`“commitment to catalysis,” is a measure of the enzyme-substrate
`the expression of
`parameters, Northrop (1975) demonstrates that
`complex tendency to proceed through the first irreversible step versus
`deuterium isotope effects on V and V/K should be the most revealing
`reversing to free enzyme and substrate. The value of D(V/K) [the
`and important as they depend on the fewest variables and on rate
`constants for different parts of the kinetic scheme. Since V is deter-
`value of (kH/kD)obs for V/K conditions] will only be close to Dk if the
`mined at saturating concentrations, the binding component is elimi-
`first irreversible step is also the catalytic step and the reverse step to
`nated and its value reflects the catalytic and product release compo-
`free enzyme and substrate is fast relative to the catalytic step. What
`nents. V/K, which is reflective of the kinetics at
`low substrate
`makes Northrop’s presentation of isotope effects in this way so
`concentration, is dependent on all rate constants up to and including
`powerful is that steady-state equations for much more complex en-
`the first irreversible step. In this case it is also the catalytic step.
`zyme mechanisms can all be reduced to this form. R and C simply
`Isotope effects on V and V/K for this example are described by eqs.
`become more complex expressions of a collection of rate constants. In
`4 and 5.
`more complicated schemes involving a greater number of reversible
`steps, an additional grouping of rate constants, Cr, termed the “com-
`mitment to reverse catalysis,” can be factored from both DV and
`D(V/K) experiments. Cr is a measure of the substrates tendency to
`return to E ⫹ S. In such schemes DV experiments now contain both
`R and Cr, while D(V/K) experiments will contain Cr and Cf, where Cf
`is a collection of rate constants termed the “commitment to forward
`catalysis” that measures the substrate’s tendency to move forward to
`E ⫹ P. Despite the greater complexity, the general forms of the
`equations still indicate that modification of (kH/kD)obs relative to
`kH/kD can be readily understood in the interplay of a collection of
`specific rate constants.
`The simplest formulation for a cytochrome P450 (P4501)-catalyzed
`reaction involves one extra step over the general scheme presented
`above. This is the single irreversible step prior to substrate transfor-
`mation in which the enzyme-substrate complex, ES, is irreversibly
`transformed into the substrate-bound active oxygenating perferryl
`oxene enzymatic species, EOS, which then proceeds to the enzyme-
`product complex, EP, with the rate constant k34 (the isotopically
`sensitive step) (Korzekwa et al., 1989) (Reaction 2). The kinetics of
`the system have been described (Northrop, 1978, 1981; Cleland,
`1982) and the following equations for the isotope effect derived. The
`term k34H(1/k23 ⫹ 1/k41) in
`k34H/k34D ⫹ k34H(1/k23 ⫹ 1/k41)
`1 ⫹ k34H(1/k23 ⫹ 1/k41)
`(V/K)H/(V/K)D ⫽ DV/K ⫽ 1
`(9)
`eq. 8 is termed the V ratio and is the factor that will increasingly mask
`the intrinsic isotope effect, k34H/k34D; the larger k34 is relative to k23
`and k41. So if product formation is faster than product release, the
`formation of the perferryl oxene species, the magnitude of the intrinsic
`isotope effect, will be suppressed; i.e., (kH/kD)obs will be smaller than
`kH/kD. Thus, the faster product formation is, the greater the suppres-
`sion of kH/kD. Equation 9 indicates that for V/K conditions [(DV/K)],
`no isotope effect should be observed, i.e., kH/kDobs must equal 1. This
`is because the first irreversible step in the scheme, conversion of ES
`to EOS, is not isotopically sensitive. Once EOS is formed, it is
`committed to continue on to product irrespective of whether or not
`substrate contains deuterium. Under V/K conditions, when an irre-
`versible step precedes the isotopically sensitive step, the decreased
`rate constant, k34D, generated by deuterium substitution, will be
`compensated for by an equal and opposite increase in the concentra-
`tion of EOSD, i.e., (EOSH)k34H ⫽ (EOSD)k34D. Thus, for cytochrome
`P450 reactions, deuterium isotope effects on V/K should in general
`never be observed (Korzekwa et al., 1989). Isotope effects can only be
`
`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on April 30, 2021
`
`VH/VD ⫽ DV ⫽
`
`(8)
`
`DV ⫽
`
`D(V/K) ⫽
`
`Dk ⫹ R
`1 ⫹ R
`Dk ⫹ C
`1 ⫹ C
`that can lead to the observed isotope effect, DV or D(V/K), having a
`much lower value than the intrinsic isotope effect, Dk (Northrop’s
`nomenclature), become clear. In the equation for the isotope effect on
`V, R, termed the “ratio of catalysis,” is a measure of rate of the
`catalytic step relative to the rate of the other forward steps contribut-
`ing to maximal velocity. In the present example, the other forward
`step would be the product release step. The value of DV [the value of
`(kH/kD)obs for Vmax conditions] will only be close to Dk (intrinsic
`isotope effect, kH/kD) when product release is fast relative to the
`catalytic step. In the equation for the isotope effect on V/K, C, termed
`
`REACTION 1.
`
`REACTION 2.
`
`REACTION 3.
`
`1 Abbreviations used are: P450, cytochrome P450; NDMA, N-nitrosodimeth-
`ylamine; EDB, ethylene dibromide; GSH, glutathione; Tris-BP, tris(2,3-dibro-
`mopropyl)phosphate; DBCP, 1,2-dibromo-3-chloropropane; NMF, N-methylfor-
`mamide; NDPS, N-(3,5-dichlorophenyl)succinimide; 3-MC, 3-methylcholan-
`threne; BHT, butylated hydroxytoluene; VPA, valproic acid; 3MI, 3-methylindole.
`
`Auspex Exhibit 2006
`Apotex v. Auspex
`IPR2021-01507
`Page 2
`
`

`

`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on April 30, 2021
`
`USE OF DEUTERIUM ISOTOPE EFFECTS TO PROBE P450 REACTIONS
`
`1483
`
`FIG. 1. The (kH/kD)obs value for the O-demethylation of p-methoxyanisole determined from an experiment of intermolecular design (a) and intramolecular design (b).
`
`observed if some mechanism or some special experimental condition
`exists that diminishes the increase in EOSD that occurs in response to
`the decreased value of k34D relative to k34H. Fortunately, there are
`both mechanisms and experimental conditions that will tend to equal-
`ize (EOSH) and (EOSD) and thereby unmask the intrinsic isotope
`effect, i.e., cause (kH/kD)obs to approach kH/kD. One mechanism that
`can achieve this goal is the presence of a branched reaction pathway
`from the substrate-bound active oxygenating perferryl oxene species,
`EOSD, to a nonisotopically sensitive alternate product and free en-
`zyme. A second and similar mechanism is reduction of the perferryl
`oxene EOSD to free substrate, free enzyme, and water (Atkins and
`Sligar, 1987, 1988; Korzekwa et al., 1989). A special experimental
`condition that can achieve the same end is an isotope effect experi-
`ment of symmetrical intramolecular design (Hjelmeland et al., 1977;
`Miwa et al., 1980; Gelb et al., 1982; Lindsay Smith et al., 1984).
`Symmetrical Intramolecular Design
`In an isotope effect experiment of intramolecular design, a substrate
`is chosen that is susceptible to enzymatic attack at either of two
`symmetrically equivalent sites (Hjelmeland et al., 1977; Miwa et al.,
`1980; Gelb et al., 1982; Lindsay Smith et al., 1984). One site contains
`deuterium and the other retains its normal complement of hydrogen.
`The enzyme then has the choice of attacking a deuterated or an
`
`REACTION 4.
`
`equivalent protio site within the same molecule. It can be viewed as
`a special case of branching and can be modeled as shown. The
`observed isotope effect, (kH/kD)obs, measured as the ratio of product
`resulting from attack at the protio site versus product resulting from
`attack at the deutero site, PH/PD, reflects the intramolecular competi-
`tion between these two sites, i.e., PH/PD ⫽ kH[EOSH]/kD[EOSD]
`(Reaction 3). Kinetically, the experimental conditions correspond to a
`V/K isotope effect (Korzekwa et al., 1989). Thus, (kH/kD)obs is
`independent of all kinetic steps following branching of EOS and can
`be described by eq. 10. The equation reveals that the faster the rate of
`reorientation of the substrate is in the active site of the enzyme, k43,
`relative to the
`
`(10)
`
`PH/PD ⫽ (kH/kD)obs ⫽
`
`k45H/k45D ⫹ k45H/k43
`1 ⫹ k45H/k43
`commitment to catalysis (catalytic step), k45H, i.e., k43 ⬎ k45H, the more
`the concentrations of EOSD and EOSH will equalize and the closer
`(kH/kD)obs will be to k45H/k45D. Conversely, the higher the commitment
`to catalysis relative to the rate of reorientation, the more the intrinsic
`isotope effect will be suppressed. An early, if the not first, example of the
`power of an intramolecular isotope effect experiment to unmask the
`intrinsic isotope effect is provided by the work of Foster et al. (1974).
`These investigators measured the deuterium isotope effect associated
`with the oxidative O-demethylation of p-methoxyanisole using two dif-
`ferent experimental designs. The first was the traditional experiment of
`intermolecular design in which the rate of demethylation of the hydrogen-
`containing substrate was measured and compared with the rate of de-
`methylation of the deuterium-containing substrate determined in a sepa-
`rate experiment (Fig. 1a). The observed isotope, (kH/kD)obs, was found to
`be 2. The second experiment was of intramolecular design in which the
`hydrogens of one of the O-methyl groups were replaced with deuterium.
`Incubation of the substrate with a microsomal preparation of cytochrome
`P450 followed by measurement of the p-methoxyphenol to p-trideutero-
`methoxyphenol product ratio gave a (kH/kD)obs of 10 (Fig. 1b). The
`observed products, p-methoxyphenol and formaldehyde, are consistent
`with at least two distinct mechanisms that might be operative. The first
`would be an insertion mechanism in which an oxygen atom inserts
`between a carbon-hydrogen bond of one of the methoxy groups to
`generate a hemiacetal intermediate. The hemiacetal is then hydrolyzed to
`generate the observed products. This mechanism should be accompanied
`
`Auspex Exhibit 2006
`Apotex v. Auspex
`IPR2021-01507
`Page 3
`
`

`

`1484
`
`NELSON AND TRAGER
`
`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on April 30, 2021
`
`FIG. 2. The (kH/kD)obs values for the formation of n-octanol from n-octane-1,8-2H2, n-octane-1-2H3, and n-octane-1,2,3-2H7.
`
`by a primary deuterium isotope effect of low magnitude, approximately
`2, since the transition state for oxygen insertion would necessarily be
`nonlinear (Shea et al., 1983). The second mechanism would involve
`initial hydrogen atom abstraction from the methyl group to form meth-
`ylene and hydroxy radicals followed by recombination to generate the
`intermediate hemiacetal. This is the mechanism first postulated by
`Groves et al. (1978) to account for the cytochrome P450-catalyzed
`hydroxylation of norbornane, a mechanism that is now generally ac-
`cepted as the mechanism for all cytochrome P450-catalyzed hydroxyla-
`tions of aliphatic carbon-hydrogen bonds. Since direct hydrogen abstrac-
`
`tion would, if possible, involve a linear transition state, the magnitude of
`the primary deuterium isotope effect could approach the theoretical value
`of approximately 9, depending upon the symmetry of the transition state.
`The (kH/kD)obs of 2 given by the intermolecular isotope effect experi-
`ment is supportive of a mechanism involving oxygen insertion. In con-
`trast, the (kH/kD)obs of 10 from the intramolecular isotope effect exper-
`iment is only consistent with the abstraction recombination mechanism.
`Since (kH/kD)obs from the intramolecular isotope effect experiment is
`much closer to kH/kD than is (kH/kD)obs from the intermolecular exper-
`iment, the choice between possible mechanisms is clear. Indeed the
`
`FIG. 3. The intrinsic isotope effect, kH/kD, for the cytochrome P450-catalyzed hydroxylation of (1,2)-dideuteromethyl-o-xylene (a) and (1,2*)-dideuteromethyl-p-xylene
`(b).
`
`Auspex Exhibit 2006
`Apotex v. Auspex
`IPR2021-01507
`Page 4
`
`

`

`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on April 30, 2021
`
`1485
`
`(11)
`
`(kH/kD)obs ⫽
`
`USE OF DEUTERIUM ISOTOPE EFFECTS TO PROBE P450 REACTIONS
`k45H/k45D ⫹ k45H/(k43 ⫹ k46)
`1 ⫹ k45H/(k43 ⫹ k46)
`The values of (kH/kD)obs found for cytochrome CYP2B1-catalyzed
`␻-hydroxylation of the dideuterated analog of n-octane, n-octane-1,8-
`2H2, the heptadeuterated analog, n-octane-1,2,3-2H7, and the trideu-
`terated analog, n-octane-1-2H3, of 16.11, 4.0, and 11.77, respectively,
`nicely illustrate the effects of k43 and k46 (Fig. 2). Before comparing
`values, it is first necessary to determine kH/kD for the reaction. Of the
`three substrates, the ␻-hydroxylation of n-octane-1,8-2H2 can best be
`expected to provide kH/kD. Both terminal methyl groups are isotopi-
`cally equivalent, and it can be assumed that the inherent rate of
`rotation of a methyl group is a much faster process than the rate of
`cleavage of a carbon-hydrogen bond via hydrogen atom abstraction.
`Thus, the enzyme always has the choice of oxidizing a carbon-
`hydrogen or carbon-deuterium bond irrespective of which methyl
`group is oriented for catalysis. The value of (kH/kD)obs ⫽ 16.11 for
`this substrate corresponds to an intrinsic primary isotope effect of 9.18
`once it has been corrected for the contribution of secondary isotope
`effects and the fact that the methyl group contains two hydrogens but
`only one deuterium (Jones and Trager, 1987). As indicated above, an
`intrinsic primary isotope effect of 9 is the theoretical limit for cleav-
`age of a carbon-hydrogen bond in the absence of tunneling effects
`(Bell, 1974). This provides compelling evidence that the cytochrome
`CYP2B1-catalyzed ␻-hydroxylation of n-octane involves a highly
`symmetrical transition state and is consistent with the abstraction-
`recombination mechanism for aliphatic hydroxylation (Groves et al.,
`1978). In the case of n-octane-1,2,3-2H7, the enzyme has the choice of
`oxidizing either a carbon-hydrogen bond or a carbon-deuterium bond,
`depending on which terminal methyl group of the substrate is properly
`oriented for catalysis. According to eq. 11, if (kH/kD)obs is to be close
`to kH/kD, the rate of methyl group interchange, k43, must be much
`faster than the rate of bond breaking, k45H. The (kH/kD)obs of 4
`indicates that this is clearly not true for this substrate and suggests that
`the distance between terminal methyl groups is large enough so that
`the rate of interchange is slow enough to prevent the concentrations of
`[EOSH] and [EOSD] from equalizing. For the case of n-octane-1-2H3,
`the enzyme also has the choice of oxidizing either a carbon-hydrogen
`bond or a carbon-deuterium bond, depending on which terminal
`methyl group of the substrate is properly oriented for catalysis. What
`is different about this substrate relative to n-octane-1,2,3-2H7 is that it
`also has the choice of a branched reaction pathway, (␻-1)-hydroxy-
`lation, that is blocked by deuteration in the case of n-octane-1,2,3-2H7.
`
`FIG. 4. The cytochrome P450-catalyzed N-demethylation of various (X ⫽ H, Cl,
`CN, and NO2) p-substituted N,N-dimethylaniline analogs.
`(kH/kD)obs of 10, which is a composite value for one primary and two
`secondary isotope effects, i.e., PS2 (Hanzlik et al., 1985) is still large
`enough to suggest that the transition state is close to symmetrical and that
`the primary isotope effect is not too far from its maximum value.
`Although the ratio of the concentration of [EOSH] to the concen-
`tration of [EOSD] must equal 1 for (kH/kD)obs to be equal to kH/kD,
`rapid equilibration of equivalent but isotopically distinct intramolec-
`ular oxidation sites is not the only means by which this condition can
`be met. As indicated above, the presence of a branched reaction
`pathway from the substrate-bound active oxygenating perferryl oxene
`species, EOSD, to a nonisotopically sensitive alternate product and
`free enzyme or reduction of the perferryl oxene EOSD to free sub-
`strate, free enzyme, and water can also equalize [EOSH] and [EOSD]
`and lead to (kH/kD)obs being equal to kH/kD. A model for a branched
`reaction pathway in competition with the isotopically sensitive step
`follows (Harada et al., 1984; Jones et al., 1986). In the model,
`substrate can reversibly reorient in the active site of the enzyme with
`rate constants k34 and k43 (Reaction 4). This allows it to present either
`the deutero site, EOSD, or the protio site, EOSH, for catalysis. EOSH
`and EOSD then go on to form products. P1 is formed from EOSH and
`EOSD with rate constants, k45H or k45D, respectively, whereas a single
`rate constant, k46, characterizes the formation of P2 from EOSH and
`EOSD. This is because P2 arises from oxidative attack at a molecular
`site remote from the site of deuterium substitution. The observed
`isotope effect for the model is given by eq. 11. Equation 11 reveals
`that the rates of substrate reorientation in the active site, k43, and
`formation of P2, k46, relative to the rate of formation of isotopically
`sensitive P1 are the factors that govern the magnitude of (kH/kD)obs
`and define the degree of masking of k45H/k45D. The larger k43 and/or
`k46 are relative to k45H, the closer (kH/kD)obs will be to k45H/k45D.
`
`FIG. 5. The fixed intramolecular distance between methyl and trideuteromethyl groups of o-xylene-␣-2H3, p-xylene-␣-2H3, 2-2H3-6-dimethylnaphthalene, and 4-2H3-4⬘-
`dimethylbiphenyl.
`
`Auspex Exhibit 2006
`Apotex v. Auspex
`IPR2021-01507
`Page 5
`
`

`

`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on April 30, 2021
`
`1486
`
`REACTION 5.
`
`FIG. 6. The cytochrome P450-catalyzed formation of (S)- and (R)-2-methyl-2-
`phenylethanol.
`
`TABLE 1
`Value of the isotope effect expected for the cytochrome P450 hydroxylation of a
`substrate at a nondeuterated site by parallel pathway, nondissociative, and
`dissociative mechanisms determined from competitive and noncompetitive
`experiments
`
`NELSON AND TRAGER
`carbon-hydrogen bond. This is because the magnitude of kH/kD varies
`as a bell-shaped curve and correlates with transition state symmetry.
`The magnitude of kH/kD is maximum when the transition state is most
`symmetrical and declines when the transition state is either more
`reactant-like or more product-like (Melander, 1960; Westheimer,
`1961; Hammond, 1965). It is therefore not surprising that the intrinsic
`primary isotope effect associated with the benzylic hydroxylation of
`o- and p-xylene by various microsomal preparations and purified
`P450s is less than maximal, falling within the range of 5.2 to 7.4 (Fig.
`3) (Hanzlik and Ling, 1993; Iyer et al., 1997). Because the benzylic
`radical can be resonance stabilized, the less than maximal value for
`the intrinsic isotope effect for benzylic hydroxylation of xylene indi-
`cates that the transition state for the reaction is more reactant-like, and
`the value of the isotope effect lies on the ascending portion of the
`bell-shaped curve.
`Another factor, besides substrate structure, that could affect the
`symmetry of the transition state for hydrogen atom abstraction is the
`active site structure of the enzyme itself. This raises the question of
`whether or not different P450s can catalyze the same oxidative reac-
`tion with different kH/kD values. If they can, it must mean that either
`different P450s can catalyze the same reaction with different mecha-
`nisms or the symmetry of the transition state can vary as a function of
`different active site topographies by modulating the stability of the
`activated oxygen-substrate complex. The activated oxygen atom of
`cytochromes P450 is an extremely reactive species that is capable of
`oxidizing virtually any carbon-hydrogen bond with which it comes in
`contact. These enzymes are different from most other enzymes in that
`the energy cost for catalysis is in generating the active oxygen species
`rather than in orienting the substrate toward a transition state-like
`structure by specific binding. This suggests that the mechanism of a
`given reaction catalyzed by different P450s is likely to be unchanged.
`In this regard, considerable evidence exists indicating that aliphatic
`hydroxylation reactions, irrespective of enzyme, proceed by a hydro-
`gen atom abstraction-recombination mechanism. Thus, if differences
`are found, they are likely to be caused by differences in active site
`topographies. To probe this question, the isotope effects for the
`␻-hydroxylation of n-octane by three very different P450s, CYP1A1,
`CYP2B1, and CYP2B4, were determined (Jones et al., 1990). All
`three enzymes gave virtually identical intrinsic primary isotope effects
`of approximately 9, even though the ␻- to (␻-1)-hydroxylation prod-
`uct ratio was very different for each isoform and the individual P450s
`were from different species (rat and rabbit) and different enzyme
`families. The N-demethylation of a series of para substituted (H, Cl,
`CN, NO2) dimethylanilines (Fig. 4) catalyzed by CYP1A2, CYP2B1,
`CYP4B1, and CYP101 gave similar findings (Karki et al., 1995).
`Whereas the magnitude of the intrinsic isotope effect varied from
`substrate to substrate, it was virtually identical across enzymes. The
`variation in the magnitude of the intrinsic isotope effect between
`substrates is expected because the transition state for the N-demeth-
`ylation of this series of substrates would fall on the ascending slope of
`the bell-shaped curve that defines the relationship between transition
`state and the magnitude of the isotope effect. Different aromatic
`substituents would differ in their ability to stabilize the radical inter-
`mediate. The picture that emerges from these data is that any given
`cytochrome P450-catalyzed reaction is likely to proceed by the same
`mechanism with the same value for the intrinsic isotope effect, irre-
`spective of the isoform catalyzing the reaction.
`The lack of sensitivity to individual P450s and different active
`site architectures of the intrinsic primary isotope effect for ali-
`phatic hydroxylation suggests that the masking effect might be
`used to explore effective active site dimensions. As has been
`indicated above, the concentrations of [EOSH] and [EOSD] for
`
`Isotope Effect
`
`Competitive VH/VD
`Noncompetitive
`(V/K)H/(V/K)D
`
`Parallel
`Pathway
`1.0
`1.0
`
`Mechanism
`
`Nondissociative
`⬍1.0
`⬍1.0
`
`Dissociative
`1.0
`⬍1.0 to
`⬎1.0
`
`Although both substrates will form the (␻-1)-product, it will form at
`a much faster rate when n-octane-1-2H3 is the substrate. The value of
`11.77 for (kH/kD)obs for n-octane-1-2H3 corresponds to a primary
`isotope effect of 9.14, when the secondary isotope effect contribution
`is removed, indicating that k46 is much larger than k45H, so that the
`sum (k43 ⫹ k46) is now big enough to overwhelm k45H and allow full
`expression of the intrinsic isotope effect. The dominance of k46 over
`k45H is substantiated by the product ratio. 2-Octanol is formed 23
`times faster than 1-octanol.
`Establishing that the ␻-hydroxylation of n-octane proceeds with a
`maximum intrinsic isotope effect of 9, and therefore a highly sym-
`metrical transition state, provides an important limit for the mecha-
`nistic interpretation of all cytochrome P450 reactions that involve
`cleavage of a carbon-hydrogen bond. It means that cleavage of a
`carbon-hydrogen bond of any saturated carbon atom having a differ-
`ent substitution pattern (secondary, tertiary, substituted with other
`atoms in addition to hydrogen) must proceed with a lower intrinsic
`isotope effect and a less symmetrical transition state. The degree to
`which the intrinsic isotope effect departs from maximum and the
`transition state from symmetrical depends on the difference that that
`specific substitution pattern engenders relative to a primary aliphatic
`
`Auspex Exhibit 2006
`Apotex v. Auspex
`IPR2021-01507
`Page 6
`
`

`

`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on April 30, 2021
`
`USE OF DEUTERIUM ISOTOPE EFFECTS TO PROBE P450 REACTIONS
`
`1487
`
`FIG. 7. The CYP2C11-catalyzed hydroxylation of testosterone to 2␣-hydroxytestosterone and 16␣-hydroxytestosterone.
`
`that F87 in the wild-type enzyme occupies active site space that
`effectively leads to a smaller active site that is more restrictive
`toward larger substrates (Rock et al., 2002).
`Nonsymmetrical Intramolecular Design
`More than a single product can sometimes be formed by the action
`of a single P450 on a single substrate. If one of the intramolecular
`product-forming sites is deuterated, the possibility of formation of an
`alternate product from a different site could be in competition with the
`deuterated site and thereby modulate the value of (kH/kD)obs. A
`model describing this possibility is shown below (Korzekwa et al.,
`1989, 1995). The model assumes that the substrate is free to reorient
`within the enzyme-substrate complex, EOS, rapidly, relative to the
`rate of substrate oxidation, k34 and k35 (Reaction 5). When P1 is the
`isotopically sensitive step while P2 is in competition with P1 but is not
`itself isotopically sensitive, DVP1 and D(V/K)P1 are given by eqs. 12
`and 13, respectively. It is apparent from the model that an isotope
`effect on k34, k34D, will alter the ratio of products, P1 and P2.
`Equation 12 also indicates that the larger k35 is relative to k34, the
`closer (kH/kD)obs will approach the intrinsic isotope effect, k34H/k34D.
`Furthermore, if product release from the isotopically sensitive path-
`way, k41, is slow, k34H/k34D will tend to be masked. In contrast, if
`product release from the alternate pathway, k51, is slow, k34H/k34D
`will tend to be unmasked.
`
`isotopically nonequivalent but otherwise identical catalytic sites
`within a substrate must be equal for (kH/kD)obs to be equal to
`kH/kD. The larger and less impeded an active site and/or the
`smaller the intramolecular distance between protium and deute-
`rium sites, the less likely the catalytic event will occur before
`equilibration of [EOSH] and [EOSD]. A set of substrates for ali-
`phatic hydroxylation with a range of known fixed distances be-
`tween protium and deuterium sites might be expected to display a
`range of (kH/kD)obs values for different P450 isoforms reflecting
`different active site topographies. The concept was tested and
`verified with CYP101, a P450 with a known active site, using the
`benzylic hydroxylation of o- and p-xylene-␣-2H3 and 4-2H3,4⬘-
`dimethylbiphenyl, in which the protio and deuterio catalytic sites
`vary by 2.5 Å, 6.6 Å, and 11 Å, respectively (Audergon et al.,
`1999). Results from CYP2B1, several microsomal preparations,
`and the same set of substrates indicated that an intramolecular
`distance of less than 7 Å between catalytic sites allowed [EOSH]
`and [EOSD] to equalize prior to catalysis, whereas a distance
`greater than 11 Å led to complete suppression of kH/kD (Iyer et al.,
`1997). In a subsequent study, 2-2H3, 6-dimethylnaphthalene (Fig.
`5), was added to the set of additional fixed distance probes (methyl
`versus trideuteromethyl groups) to explore the active site topog-
`raphy of CYP4B1 relative to CYP2B1 and CYP102 (Henne et al.,
`2001). The results of the study led to the conclusion that the active
`site of CYP4B1 is considerably restricted relative to CYP2B1. In
`a similar study, p-xylene-␣-2H3 an

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