throbber
Biochemistry 1998, 37, 7039-7046
`
`7039
`
`Evaluation of Cytochrome P450 Mechanism and Kinetics Using Kinetic Deuterium
`Isotope Effects†
`LeeAnn Higgins, Grace A. Bennett, Miyuki Shimoji, and Jeffrey P. Jones*
`Department of Pharmacology and Physiology, UniVersity of Rochester, Rochester, New York 14642
`ReceiVed December 4, 1997; ReVised Manuscript ReceiVed March 11, 1998
`
`ABSTRACT: In this paper two hypotheses are tested: (i) the active oxygen species is similar in energetics
`for all cytochrome P450 (CYP) enzymes and (ii) linear free-energy relationships can be used to evaluate
`the mechanism of the reaction of these enzymes. A series of intramolecular isotope effects were determined
`and compared for CYPs 1A2, 2B1, 2C9, 2E1, and P450cam. The results indicate that the isotope effects
`are very similar for each of these isoforms of P450 and that the first hypothesis is likely to be true.
`Attempts to establish a linear free-energy relationship were only moderately successful: log Vmax ) 0.11(cid:243)+
`p
`+ 1.73; r2 ) 0.588. It was determined, through the use of intermolecular isotope effects, that the rates
`of hydrogen atom abstraction are masked. Thus, the second hypothesis is found to be false. This is
`likely to be a general result for CYP reactions, and linear free-energy relationships can only be used to
`determine the mechanism under very special circumstances. In all cases, the rate-limiting step should be
`evaluated with isotope effect experiments before any mechanistic conclusions can be drawn.
`If the
`intermolecular isotope effects are found to be masked, no mechanistic conclusion can be drawn from the
`linear free-energy relationship study.
`
`The cytochrome P450 (CYP)1 enzymes comprise a su-
`perfamily of heme-containing enzymes that consists of more
`than 300 individual isoforms, and the CYP enzymes are
`found in plant, bacterial, and mammalian species (1). These
`enzymes function mainly as monooxygenases (2). In mam-
`mals, they are responsible for the metabolism of certain
`endogenous as well as exogenous compounds (3). Because
`these enzymes play an important role in xenobiotic metabo-
`lism, the abilities to predict reaction rates, identify possible
`metabolites, and understand CYP mechanisms are important
`goals. Electronic models have been developed that use
`computational (4, 5) and chemical (6, 7) approaches for the
`successful prediction of relative reaction rates and isotope
`effect profiles for some classes of small compounds.
`One of the most important considerations for a potential
`CYP model is whether the model will apply to relative rate
`predictions for several (or all) CYP isoforms or for a limited
`number of isoforms.
`In this study, we use isotope effect
`profiles (IEPs) to test the hypothesis that the active oxygen
`is conserved in multiple CYP isoforms. An IEP consists of
`
`† This research was supported by the National Institutes of Health,
`Grants ES06062 and ES09122.
`* To whom correspondence should be addressed. Department of
`Pharmacology and Physiology, Box 711, University of Rochester, 601
`Elmwood Avenue, Rochester, NY 14642. Phone:
`(716) 275-5371.
`Fax: (716) 244-9283. E-mail:
`jpj@cyp.medicine.rochester.edu.
`1 Abbreviations: CYP, cytochrome P450; LFER, linear free-energy
`relationship; IEP, isotope effect profile; KIE, kinetic isotope effect(s);
`LAD, lithium aluminum deuteride; CH2Cl2, dichloromethane; TLC,
`thin-layer chromatography; NMR, nuclear magnetic resonance; GC-
`MS, gas chromatography-mass spectroscopy; TK-, thymidine kinase
`deficient; KPi, potassium phosphate; Pd, putidaredoxin; PdR, putidare-
`doxin reductase; E. coli, Escherichia coli; NADP+, (cid:226)-nicotinamide
`adenine dinucleotide phosphate; NADH, (cid:226)-nicotinamide adenine di-
`nucleotide, reduced; MTBSTFA, N-methyl-N-(tert-butylimethylsilyl)-
`trifluoroacetamide; HP, Hewlett-Packard; D0, nondeuterated; D3, tri-
`deuterated; DV and D(V/K), kinetic deuterium isotope effects on the
`enzymatic rate constants Vmax and V/K, respectively.
`
`isotope effect measurements for a series of structurally related
`compounds that most nearly approximate intrinsic isotope
`effects. When intrinsic KIEs are available, comparable
`trends in IEPs between different systems or different isoforms
`support similar mechanisms (6-8).
`If the hypothesis that the active oxygen is very similar in
`multiple CYP isoforms is proven to be true, a single
`electronic model can be used to describe (the corresponding)
`multiple P450 enzymes. Linear free-energy relationships are
`utilized in chemistry and enzymology as a predictive tool
`and as a probe of the electronic characteristics of the
`transition state of a reaction. We test here the second
`hypothesis that LFERs can be used to predict relative rates
`of metabolism by CYP and to evaluate physical character-
`istics of CYP reactions. If relative rates of product formation
`are masked by other steps in an enzymatic cycle, LFERs
`must be interpreted with caution. Kinetic information, but
`not mechanistic information, can be deduced, if relative
`reaction rates are masked. Therefore, it is important to assess
`whether masking occurs with the substrates chosen for the
`analysis of a LFER.
`Herein, we report IEP results for a series of para-
`substituted toluenes. The data support the hypothesis of a
`conserved active oxygen species for multiple CYPs. Our
`attempts to establish LFERs for a series of para-substituted
`toluenes were unsuccessful. From isotope effect studies, we
`conclude that relative reaction rates for para-substituted
`toluenes in this study are partially masked.
`MATERIALS AND METHODS
`Chemicals and Reagents. All solvents were purchased
`from J. T. Baker, Inc. (Phillipsburg, NJ). Chemicals were
`purchased from Aldrich Chemical Co. (Milwaukee, WI) with
`the following exceptions: MTBSTFA (+1% tert-butyldi-
`methylchlorosilane) was purchased from Regis Technologies,
`
`S0006-2960(97)02986-3 CCC: $15.00 © 1998 American Chemical Society
`Published on Web 04/23/1998
`
`Downloaded via WASHINGTON STATE UNIV on April 14, 2020 at 18:45:46 (UTC).
`
`See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
`
`Apotex Ex. 1026
`
`Apotex v. Auspex
`IPR2021-01507
`
`

`

`7040 Biochemistry, Vol. 37, No. 19, 1998
`
`Inc. (Morton Grove, IL), and potassium phosphate dibasic
`and monobasic and magnesium sulfate were from EM
`Science (associate of E. Merck, Darmstadt, Germany).
`p-Chlorotoluene, p-methylanisole, and p-xylene were purified
`by distillation before use. p-Bromotoluene was crystallized
`from ethanol, and p-tolunitrile was crystallized from benzene:
`pet ether. Gibco BRL products (Gaithersburg, MD) were
`used for all cell culture methods. Biochemicals were
`purchased from Sigma (St. Louis, MO). p-Xylene-R, R, R,
`R¢, R¢, R¢-d 6 (99.8% pure by GC) was purchased from
`Aldrich Chemical Co.
`Synthesis of Compounds. Para-Substituted Benzyl-R-2H2
`Alcohols. Methyl 4-bromobenzoate, methyl 4-chloroben-
`zoate, methyl 4-cyanobenzoate, methyl 4-methylbenzoate,
`and methyl 4-methoxybenzoate were used as starting materi-
`als for the synthesis of the corresponding para-substituted
`benzyl alcohols according to the following procedure.
`Lithium aluminum deuteride (1.2 mol) was suspended in
`ether and cooled to 0 (cid:176)C in an ice bath while under N 2. The
`substituted benzoate (1 mol) was dissolved in a small amount
`of ether and added dropwise to the LAD suspension while
`keeping the temperature at 0 (cid:176) C. After the addition, the
`mixture was stirred at 0 (cid:176)C for an additional hour, or until
`complete by TLC (silica gel: 80% hexane, 20% ethyl
`acetate). The unreacted LAD was decomposed, and the
`mixture was filtered, dried over MgSO4, and evaporated to
`dryness. The crude product was purified by recrystallization
`and/or distillation, and purity was assessed by NMR and
`GC-MS.
`Para-Substituted Toluenes-R,R,R-2H3. p-Bromotoluene,
`p-chlorotoluene, and p-methylanisole were synthesized from
`the corresponding para-substituted benzyl-d2 alcohols ac-
`cording to the following method. Methane sulfonyl chloride
`(0.11 mol) was dissolved in a small amount of CH2Cl2 and
`added dropwise to a solution of the para-substituted benzyl-
`d2 alcohol (0.10 mol) and triethylamine (0.15 mol) in CH2-
`Cl2 that was cooled to -78 (cid:176)C in a dry ice -acetone bath.
`The reaction was stirred for another 30 min at -78 (cid:176)C.
`Because the mesylates are generally unstable, they were not
`isolated, and the procedure was continued by adding ice
`water, separating the layers, and extracting the aqueous layer
`with CH2Cl2. The organic layers were combined, washed
`with saturated sodium bicarbonate, and dried over MgSO4,
`and the solvent was removed under reduced pressure. The
`oil that remained was reconstituted in ether and reduced with
`LAD using the same procedure as described for the reduction
`of the benzoates. The products were purified by chroma-
`tography (silica gel: 100% CH2Cl2), distilled under vacuum
`using a Kugelrohr, and, when necessary, purified again with
`a neutral alumina column and 100% hexane to remove any
`residual benzyl alcohol. Any residual benzyl alcohol was
`detected by select ion monitoring using GC-MS as described
`below.
`Substituted Toluene-R-2H1 and -R-2H1-R¢ -2H1. Monodeu-
`terated analogues of p-bromotoluene, p-chlorotoluene, p-
`methylanisole, and toluene were synthesized by the mesyl-
`ation of the corresponding nondeuterated benzyl alcohols
`according to the same procedure described above for the
`trideuterated compounds, and p-xylene-R-2H1-R¢- 2H1 was
`synthesized with the same method from benzene dimethanol,
`except that the mole ratio of benzene dimethanol to the other
`reactants was 1:2.
`
`Higgins et al.
`For p-tolunitrile-R-2H1, the benzyl alcohol was first syn-
`thesized by the reduction of p-cyanobenzaldehyde using sod-
`ium borohydride. For this procedure, the sodium borohydride
`(0.05 mol) was mixed with about 100 mL of ethanol and
`stirred under N2. The aldehyde (0.025 mol) was dissolved
`in a small amount of ethanol and added dropwise to the
`NaBH4 solution. The reaction was monitored by TLC (silica
`gel: 50% ethyl acetate, 50% hexane). After about 1.5 h,
`the mixture was filtered and the solvent was evaporated. The
`oil that remained was mixed with water and then extracted
`with CH2Cl2. The organic layer was dried over MgSO4 and
`evaporated to yield a white solid, which was recrystallized
`with hexane and assessed for purity by NMR and GC-MS.
`Preparation of HepG2-Expressed CYP. Individual CYP
`isoforms (1A2, 2B1, 2E1, and 2C9) were obtained using the
`HepG2 vaccinia expression system described previously (9).
`Briefly, the recombinant vaccinia viruses containing cDNAs
`encoding a single CYP were propagated in human TK- 143
`(human embryoblast) cells, which were then used to infect
`confluent flasks of hepatoma HepG2 cells (American Type
`Culture Collection, Rockville, MD). For each isoform, cells
`were scraped from 50 flasks, pooled, divided into 1-mL
`aliquots, and stored at -78 (cid:176)C. Microsomal preparations
`from crude cell lysates were obtained immediately before
`use according to the following procedure: aliquots were
`thawed, mixed with 1 mL of 50 mM KPi buffer pH 7.4,
`sonicated with a Branson Sonifier Model 450, and centri-
`fuged for 15 min at 300000g in a cold (4 (cid:176)C) Beckman TL-
`100 ultracentrifuge; pellets were reconstituted with 2 mL of
`KPi buffer pH 7.4, pooled, and homogenized in a cold room.
`The total protein concentration of the final enzyme-buffer
`mixture was typically 3-4 mg mL-1 by Bradford assay (10)
`with bovine serum albumin as the standard. The typical low
`levels of P450 obtained using this expression system were
`undetectable by CO difference spectra.
`Cytochrome P450cam, Putidaredoxin, and Putidaredoxin
`Reductase. Construction of expression plasmids for P450cam,
`Pd, and PdR as well as protein expression and purification
`have been described previously (11).
`Incubation Conditions and Isolation of Products. All
`incubations using HepG2-expressed CYP were run for 20
`min in a shaking water bath at 37 (cid:176)C and contained the
`following: 300-400 (cid:237)g of protein suspension, depending
`on the isoform; 10 mM glucose-6-phosphate, 1 mM NADP+,
`and 1 U glucose-6-phosphate dehydrogenase as an NADPH
`generating system; and 1400 U catalase in a total volume of
`5 mL of 50 mM KPi buffer, pH 7.4, 2 mM MgCl2. For
`P450cam, the assays were carried out at 37 (cid:176)C for 20 min
`in a 1-mL reaction mixture in 50 mM KPi buffer, pH 7.4,
`200 mM KCl, 1.5 (cid:237)M P450cam, 3 (cid:237)M Pd, 1.5 (cid:237)M PdR,
`and 0.3 mM NADH. Reactions were initiated by the addition
`of 50 (cid:237)L (HepG2-expressed CYPs) or 10 (cid:237)L (P450cam) of
`a substrate solution in CH3CN, terminated upon addition of
`3 mL of CH2Cl2, and 10 (cid:237)L of internal standard in CH3CN
`was added,
`if applicable (see below). Samples were
`extracted three times with CH2Cl2; the organic fractions were
`pooled, dried with anhydrous MgSO4, and concentrated
`gently under a stream of N2 to a volume of approximately
`100 (cid:237)L. After reconstitution with 300 (cid:237)L (HepG2-expressed
`CYPs) or 100 (cid:237)L (P450cam) of CH3CN, 50 (cid:237)L of MTB-
`STFA + 1% t-BDMCS was added, and samples were heated
`overnight at 70 (cid:176)C.
`
`Apotex Ex. 1026
`
`

`

`P450 Mechanism, Kinetics, and Isotope Effects
`
`Biochemistry, Vol. 37, No. 19, 1998 7041
`
`Enzyme incubations with monodeuterated toluenes and
`p-xylene-R-2H1-R¢ -2H1 were performed in triplicate at a single
`substrate concentration of 0.25 mM. Determinations of Vmax
`and Km for D0 or D3 substrates were made using at least six
`samples, with six different substrate concentrations in a range
`between 0.5 and 100 (cid:237)M, and 10 nmol of the appropriate
`para-substituted benzyl alcohol as the internal standard.
`Product formation from D0 substrates was measured with
`the corresponding para-substituted benzyl alcohol dideuter-
`ated at the benzylic carbon as the internal standard; for D3
`substrates the internal standard was the corresponding
`nondeuterated benzyl alcohol. Competitive experiments
`were performed in triplicate with mixtures of D0 and D3
`substrates, each at a final concentration of 0.25 mM.
`Linearity studies using low and high concentrations of select
`D0 substrates showed linear product formation over 30 min.
`Two types of controls were run for each substrate with the
`identical procedure used for the complete samples except
`that in one case substrate was omitted, and in the other case,
`NADP+ and the NADPH-generating system or NADH was
`omitted. Controls for toluene-R-2H1 showed a small peak
`at the same retention time as product that originated from
`the derivatizing agent, which was quantified and subtracted
`from the observed amount of alcohol product. The kinetic
`parameters for the metabolism of the substituted toluenes
`were determined by GC-MS analysis of the M-(dimethyl-
`silyl) derivative of the product benzyl alcohols (see below).
`Control experiments using a mixture of deuterated and
`nondeuterated products showed that there was no isotope
`effect on product workup.
`Product Analysis by GC-MS. All samples were analyzed
`with an HP (Palo Alto, CA) GC/MS 5890/5972 system,
`equipped with an HP 7673A automatic injector. An HP-1
`capillary column (25 m, 0.2-mm ID, 0.5-(cid:237)m film thickness)
`was used for product analysis from all substrates except
`toluene and p-xylene, which were analyzed with an HPWax
`capillary column (30 m, 0.25-mm ID, 0.5-(cid:237)m film thickness).
`For the HP-1 column, the injector and detector temperatures
`were both set at 250 (cid:176) C; for the HPWax column, the injector
`and detector temperatures were 230 and 240 (cid:176) C, respectively.
`All samples were injected in the splitless mode. Oven
`conditions for the HP-1 column were 50 (cid:176)C for 0.5 min, 10
`(cid:176)C/min to 250 (cid:176)C, and 2 min at 250 (cid:176)C except when
`p-tolunitrile was used as the substrate, in which case a
`gradient of 15 (cid:176)C/min was used. For the HPWax column
`the oven conditions were 50 (cid:176)C for 0.5 min, 10 (cid:176)C/min to
`240 (cid:176)C, and 3 min at 240 (cid:176)C. The MS was operated at an
`ionizing voltage of -70 eV and a 20-ms dwell time. Scans
`of authentic standards of the derivatized benzyl alcohols were
`used to find peak retention times and to verify products
`formed from enzyme incubations. Selected ion recording
`of
`the M-(dimethylsilyl)
`ion of products and internal
`standards was used to determine the amount of metabolism.
`The ratio of the protio and deuterio products or of the product
`and internal standard was corrected for ion overlap and
`percent deuterium incorporation with Brauman’s least-
`squares approach (12). Deuterium incorporation of substrates
`was measured with an HP GC-MS 5890-5970 system with
`an HP-1 column using the same operating conditions as
`above except the MS ionizing voltage was adjusted to -10.0
`eV.
`
`Scheme 1
`
`DV )
`
`+ k5H
`k7
`+ k5H
`k7
`
`+ k5H
`k5H
`k5D
`k3
`1 + k5H
`k3
`D(V/K) ) 1
`
`(1)
`
`(2)
`
`Data Analysis. Kinetic parameters Vmax and Km were
`determined by nonlinear regression using GraphPad Prism
`v2.01 (San Diego, CA) data analysis software. Kinetic
`expressions were solved using the program Mathematica
`(Wolfram Research, Champain, IL).
`THEORY
`Linear Free-Energy Relationships and KIE. When LFERs
`are sought, experiments should be performed that definitively
`answer the following question:
`Is the rate of product
`formation masked (is product formation limited) by a rate-
`determining step in the catalytic cycle that is not the chemical
`step of interest? Fortunately, innovative methods for the
`investigation of masking in enzymatic systems were devel-
`oped by Cleland and Northrop (13, 14). With this approach,
`KIE are measured on the enzymatic parameters Vmax and V/K.
`Isotope effects on Vmax and V/K are expressions for the
`isotope effects on rates of product formation with the limiting
`conditions of high and low substrate concentrations, respec-
`tively. In this study we report deuterium isotope effects on
`Vmax and V/K which are designated DV and D(V/K). The
`expressions for both DV and D(V/K) contain the rate constant
`for the isotopically sensitive step, but they differ in the
`following ways: only DV contains the rate constant for the
`dissociation of product from enzyme, and only D(V/K)
`contains the rate constants for the binding of free substrate
`and enzyme, and dissociation of the first ES complex to free
`substrate and enzyme. The relative magnitudes of these steps
`as well as others to the isotopically sensitive step determine
`the degree to which the intrinsic KIE (the KIE associated
`solely with the bond breaking step, i.e., kH/kD) will be
`expressed in DV or D(V/K). Therefore, comparisons of DV
`and D(V/K) isotope effects to an intrinsic KIE can provide
`information regarding which steps, if any, are involved in
`masking the relative rates of product formation. These types
`of studies can reveal
`if LFERs can be used to make
`mechanistic conclusions. It should be noted that even if
`a linear relationship is seen between the natural log of
`the rates and the descriptors for a series of compounds,
`it does not mean that the relative rates are unmasked.
`Intermolecular Isotope Effects. Intermolecular deuterium
`isotope effects are often used for kinetic and mechanistic
`studies on enzyme systems. While this type of experiment
`usually does not provide significant mechanistic information
`since the intrinsic isotope effect is not observed, it can
`provide information about
`the kinetic mechanism. An
`intermolecular noncompetitive KIE experiment involves the
`
`Apotex Ex. 1026
`
`

`

`7042 Biochemistry, Vol. 37, No. 19, 1998
`
`independent determination of Vmax and V/K using protonated
`
`Scheme 2
`
`D(V/K)P1
`
`)
`
`+ k7Hk6
`k7H
`k7D
`k5k9
`1 + k7Hk6
`k5k9
`
`+ k7H
`k5
`+ k7H
`k5
`
`(3)
`
`DVP1
`
`)
`
`k7H
`k7D
`
`k5k9
`
`k5k9
`
`+[k7Hk6
`1 +[k7Hk6
`
`] +[2k3k7H
`] +[2k3
`
`k13
`
`+ k7H
`k5
`+ k7H
`k5
`
`+ 2k3k6k7H
`+ k3k7H
`+ k3k7H
`k11k5k9
`k13k5
`k11k5
`k13k7D
`+ 2k3k6k7H
`+ k3k7H
`+ k3k7H
`k11k5
`k13k5
`k11k5k9
`
`]
`
`]
`
`(4)
`
`and isotopically labeled substrates, for example, R1-CH3 and
`R1-CD3 (15). For the calculation of a KIE, the kinetic
`constant (Vmax or V/K) pertaining to the unlabeled substrate
`is divided by the corresponding constant for the labeled
`substrate. Scheme 1 shows a potential kinetic mechanism
`that describes an intermolecular noncompetitive KIE, and
`eqs 1 and 2 are the expressions for DV and D(V/K) obtained
`for this scheme. Substrate (SH or SD) combines with enzyme
`(E) reversibly to form the corresponding ES complex (ESH
`(Subscript “H” or “D” signifies the involvement
`or ESD).
`of a compound or a complex in the pathway for the
`abstraction of hydrogen or deuterium, respectively.) Forma-
`tion of an “activated” complex ESH* or ESD*, from ESH or
`ESD, respectively, is depicted here as an irreversible step
`that occurs prior to the isotopically sensitive step. This
`irreversible step is consistent with our understanding of the
`kinetic mechanism of CYP (16, 17).
`It
`is a kinetic
`representation of a collection of steps that
`include the
`following: the first electron reduction (Fe3+ f Fe2+); oxygen
`binding and reduction; the second electron reduction of
`oxygen; and the heterolytic cleavage of the peroxy anion, a
`postulated step which is presumed to be irreversible. Product
`is formed at the isotopically sensitive step, producing an
`enzyme-product complex (EPH or EPD), which is followed
`by the dissociation of the corresponding product from the
`enzyme. Rate constants k1, k2, k3, and k7 may each actually
`represent more than one step, and inclusion of these
`additional steps increases the complexity of the KIE equa-
`tions but does not modify this analysis. When there is no
`pathway for alternate product formation, which is typical for
`most enzymes, the value of DV depends on the relative
`magnitudes of k3 and k7 to k5H. As the rate constants k3 and
`k7 decrease relative to k5H, DV becomes masked (see eq 1).
`D(V/K) for the noncompetitive KIE with the kinetic mech-
`anism shown in Scheme 1, which has no branching pathway
`from ESH* or from the corresponding ESD* position, is
`completely masked (eq 2).
`
`Higgins et al.
`
`While the D(V/K) is completely masked given Scheme 1,
`a branched pathway has the potential to unmask both DV
`and D(V/K) (18). This is illustrated using Scheme 2, which
`represents the formation of two different products (Product
`1 ) P1, Product 2 ) P2) from one substrate, either deuterated
`or nondeuterated. Metabolism at an alternate, isotopically
`insensitive step, k9, is in competition with the isotopically
`sensitive step, k7. Rate constants k1 and k2 represent substrate
`binding to and debinding from the enzyme. Rate constants
`k3 and k3¢ are shown as irreversible steps that represent steps
`that occur after substrate binding, up to and including oxygen
`activation. These steps lead to the formation of “activated
`complexes” (ES1* and ES2*) between the enzyme and the
`substrate in the relative positions 1 and 2. Rate constants
`k3 and k3¢ are assumed to be equal for a given substrate. The
`active oxygen species is stable enough to permit translational
`and conformational changes of the substrate in the active
`site of the enzyme (19-21). Consequently, it is possible
`for an equilibrium to be established between ES1* and ES2*
`(see k5 and k6). This is the key to unmasking KIE. When
`deuterium is substituted for hydrogen at the site of metabo-
`lism, the rate constant for product formation is reduced. This
`can be expressed as k7D < k7H, where k7D represents the rate
`constant for oxidation at a deuterated position on a substrate,
`and k7H represents the rate constant for oxidation at the same
`position on a nondeuterated analogue of the substrate.
`Deuterium substitution will cause the concentration of ES1D*
`to increase relative to the concentration of ES1H* (which is
`the corresponding enzyme-substrate complex for a nondeu-
`terated substrate) unless ES1D* is in equilibrium with another
`complex, for example, ES2D*. (The corresponding equilib-
`rium would exist for the nondeuterated substrate as well.) If
`an alternate pathway does not exist, then [ES1D*] > [ES1H*],
`and an isotope effect is masked.
`Oxidation at an alternate, unlabeled position on a substrate
`with the formation of a second product (P2) can unmask a
`KIE. This is accomplished by the redirection of “excess”
`ES1D* toward ES2D* which allows [ES1D*] ) [ES1H*],
`which is necessary for the expression of a maximum, or
`unmasked, isotope effect. According to the convention of
`Northrop (15), a substrate with a low forward internal
`commitment to catalysis will branch from ES1 toward ES2
`(Scheme 2), and isotope effects on DV and D(V/K) will be
`unmasked, depending on the magnitude of the commitment
`terms. The extent to which D(V/K) is unmasked depends on
`the relative values of k5 and k9 to k7H (eq 3). As the rates of
`substrate rotation and branching increase relative to the rate
`of P1 formation, the amount of unmasking increases. The
`expression for a KIE on DV for Scheme 2 is quite complex
`(not shown) but can be simplified to eq 4 with the following
`two assumptions. If we assume that reduction of oxygen is
`rate-limiting (see prior discussion) and is significantly smaller
`than (i) the rotation of substrate in the active site (k3 , k5,
`k6) and (ii) the rates of product formation k7H and k9, the
`expression for DVP1 is simplified to eq 4.
`If the external
`reverse commitment to catalysis is small (k11, k13 . k3), that
`is, product release is not rate-limiting, then eq 4 reduces to
`eq 3 (DVP1 ) D(V/K)P1).
`Cytochrome P450 has the potential to function as an
`oxidase, whereby molecular oxygen undergoes a four-
`electron reduction, with the overall
`formation of
`two
`molecules of water (22). Formation of the second molecule
`
`Apotex Ex. 1026
`
`

`

`P450 Mechanism, Kinetics, and Isotope Effects
`
`Biochemistry, Vol. 37, No. 19, 1998 7043
`
`of water acts as a branch point leading from the ES complex
`in Scheme 2 and precludes substrate oxidation. As a
`consequence, if branching to water is fast relative to product
`formation, KIE (or rates of product formation) can become
`unmasked.
`Intramolecular Isotope Effects. To obtain better mecha-
`nistic information from enzyme kinetic studies, intramolecu-
`lar KIE can be used. While this type of experimental design
`cannot be used for all enzymes,
`in cases where it
`is
`appropriate it can be used to provide a closer estimate of
`the intrinsic isotope effect, since the potential for masking
`is decreased. For this experiment, a substrate is chosen that
`contains at least two symmetrical sites, with at least one site,
`but not all, isotopically labeled. An intramolecular KIE is
`a special case of branched kinetics in that the branch point
`occurs at the two symmetric sites. Numerous references
`provide discussions and schemes for intramolecular KIE and
`their relation to intrinsic KIE (19, 23, 24).
`An intramolecular KIE will always be greater than or equal
`to an intermolecular KIE. However, it has been shown that
`even intramolecular KIE can be masked (19, 25, 26),
`depending on the choice of substrate. Masking is signifi-
`cantly reduced when substrates are chosen that contain
`hydrogen and deuterium bonded to the same carbon atom
`(26).
`Isotope Effect Profiles. To determine if the mechanisms
`of two reactions are similar, IEPs are preferred over a single
`measurement since the trend in isotope effects provides
`additional information about the energetics of the reaction
`and makes fortuitous agreement between the two systems
`unlikely (27). An IEP consists of isotope effect measure-
`ments in a system (e.g., chemical or enzymatic) using a series
`of compounds that are structurally related. When intrinsic
`KIE are available, comparable trends in KIE profiles between
`different systems argue for similar mechanisms, while
`different trends are indicative of dissimilar mechanisms. The
`rationale for this inference is based on the fundamental idea
`of the Melander-Westheimer principle (28, 29) which is
`based on the Hammond postulate (30). According to the
`Melander-Westheimer principle, maximum isotope effects
`will be observed with reactions that have symmetrical
`reaction coordinates. Mechanisms with nonsymmetrical
`reaction coordinates will yield smaller isotope effects. From
`this information it can be inferred that intrinsic KIE that are
`measured for a series of compounds that differ in some
`systematic (e.g., chemical or physical) way can serve as a
`sensitive probe of the energetics of the isotopically sensitive
`step.
`RESULTS AND DISCUSSION
`Isotope Effect Profiles with Multiple CYPs. Isotope effect
`profiles for multiple CYP isoforms were measured in order
`to test the hypothesis that the active oxygen of multiple CYPs
`is conserved. The intramolecular isotope effects for six para-
`substituted toluenes (p-X-Ph-CH2D: X ) OCH3, CH2D, H,
`Cl, Br, CN; Ph ) phenyl) using expressed CYPs 1A2, 2B1,
`2E1, and 2C9 and purified P450cam are listed in Table 1.
`These experiments were performed with expressed CYP3A4
`also, but product formation was too low to accurately
`measure, most likely because these compounds are poor 3A4
`substrates.
`Isotope effects measured with substrates that
`contain hydrogen and deuterium bonded to the same carbon
`
`Table 1: Intramolecular Isotope Effects for Six Para-Substituted
`Toluenes Using Various CYP Isoforms, Where Isotopic Discrimin-
`ation Occurs at a Monodeuterated Benzylic Position (i.e., R-CH2D)a
`para substituent
`2E1
`2B1
`1A2
`2C9
`P450cam
`4.24 (1)
`3.69 (3)
`4.64 (4)
`4.3 (1)
`4.44 (1)
`OCH3
`CH2Db
`5.45 (5)
`6.23 (5)
`5.59 (1)
`5.9 (1)
`6.0 (2)
`ndc
`H
`6.1 (1)
`7 (1)
`6.1 (4)
`nd
`Cl
`6.75 (2)
`8.1 (1)
`7.06 (2)
`6.2 (1)
`6.5 (8)
`Br
`6.75 (3)
`8.02 (3)
`6.83 (8)
`6.9 (2)
`8.3 (1)
`CN
`10.1 (1)
`11.9 (3)
`10.1 (2)
`11.1 (6)
`11.6 (5)
`a CYPs 2E1, 2B1, 1A2, and 2C9 were expressed in HepG2 cells,
`and P450cam was purified from E. coli. Each result is the average of
`three determinations, and the numbers in parentheses indicate the
`standard deviation in the last significant digit of the mean isotope effect
`value. b p-Xylene-R-2H1-R¢- 2H1 was used as the substrate. c Not deter-
`mined due to low levels of product formation. For each substrate except
`toluene, a comparison of all the isotope effect values revealed that the
`means were statistically different from each other at the 0.05 signifi-
`cance level. This is explained by the fact that the standard deviations
`reflect the precision of the method only and do not take into account
`systematic and day-to-day errors.
`
`atom permit close measurement of the intrinsic isotope effects
`(see prior discussion of intramolecular isotope effects). Table
`1 shows that, for each isoform, the IEPs for the toluenes are
`practically identical: the values range from 4 to 11, and the
`magnitudes of the isotope effects have the same rank order.
`In addition, for each substrate the KIE for all five CYPs are
`in close agreement, which exemplifies the conserved nature
`of the active oxygen species.
`The IEPs presented in this paper provide the first evidence
`for a common reaction mechanism for aliphatic hydroxylation
`at the benzylic position in four mammalian CYP isoforms
`as well as one bacterial isoform. Inclusion of P450cam into
`our series of IEPs is the first extension of this application to
`a P450 outside of the Class II P450s. P450cam belongs to
`Class I, which consists of P450s that interact with two
`electron-transfer partners. Class II members differ in that
`they have only one protein as a reducing partner (31). The
`finding that the KIE profile for P450cam is not different from
`the CYPs that belong to Class II is significant because it
`indicates that the mechanism of hydrogen atom abstraction
`is independent of the steps that occur prior to the irreversible
`formation of an active oxygen species.
`Our results agree with other IEP data in the literature for
`different groups of compounds that are metabolized by CYP.
`Karki et al. measured isotope effects for a series of N,N-
`dimethylanilines using mammalian CYPs 2B1 (purified), 4B1
`and 1A2 (expressed), rat liver microsomes, and purified
`bacterial CYP102 and compared them to KIE in two different
`chemical systems (6). Practically identical IEPs for all of
`the P450s and the tert-butoxyl radical system provide strong
`evidence for a similar mechanism of N-dealkylation, that is,
`initial hydrogen atom abstraction. Furthermore, the mech-
`anism was conserved among all the CYPs that were tested,
`which includes both mammalian and bacterial members of
`the Class II P450s. Isotope effect profiles for various CYPs
`and for the tert-butoxy radical in several small molecules
`reported by Manchester et al. also support the mechanism
`of (initial) hydrogen atom abstraction for CYPs (7). This
`abundant amount of data for both mammalian and bacterial
`isoforms strongly supports a common mechanism for mul-
`tiple CYPs.
`LFER with Para-Substituted Toluenes and CYP2E1. Since
`the data above support the hypothesis that

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