throbber
Chemico-Biological Interactions 142 (2002) 135 /154
`www.elsevier.com/locate/chembiont
`
`The metabolic activation of abacavir by human
`liver cytosol and expressed human alcohol
`dehydrogenase isozymes
`
`John S. Walsh *, Melinda J. Reese, Linda M. Thurmond
`
`Division of Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Research Triangle Park,
`NC 27709-3398, USA
`
`Abstract
`
`Abacavir (ZIAGEN†) is a reverse transcriptase inhibitor marketed for the treatment of
`HIV-1 infection. A small percentage of patients experience a hypersensitivity reaction
`indicating immune system involvement and bioactivation. A major route of metabolism for
`abacavir is oxidation of a primary bg unsaturated alcohol to a carboxylic acid via an aldehyde
`intermediate. This process was shown to be mediated in vitro by human cytosol and NAD,
`and subsequently the aa and g2g2 human isoforms of alcohol dehydrogenase (ADH). The aa
`isoform effected two sequential oxidation steps to form the acid metabolite and two isomers,
`qualitatively reflective of in vitro cytosolic profiles. The g2g2 isozyme generated primarily an
`isomer of abacavir, which was minor in the aa profiles. The aldehyde intermediate could be
`trapped in incubations with both isozymes as an oxime derivative. These metabolites can be
`rationalized as arising via the aldehyde which undergoes isomerization and further oxidation
`by the aa enzyme or reduction by the g2g2 isozyme. Non-extractable abacavir protein residues
`were generated in cytosol, and with aa and g2g2 incubations in the presence of human serum
`albumin (HSA). Metabolism and residue formation were blocked by the ADH inhibitor 4-
`methyl pyrazole (4-MP). The residues generated by the aa and g2g2 incubations were analyzed
`by SDS-PAGE with immunochemical detection. The binding of rabbit anti-abacavir antibody
`to abacavir-HSA was shown to be dependent on metabolism (i.e. NAD-dependent and 4-MP
`sensitive). The mechanism of covalent binding remains to be established, but significantly less
`abacavir-protein residue was detected with an analog of abacavir in which the double bond
`was removed, suggestive of a double bond migration and 1,4 addition process.
`# 2002 Elsevier Science Ireland Ltd. All rights reserved.
`
`* Corresponding author. Tel.: /1-919-469-4719; fax: /1-919-315-6003
`E-mail address: John.S.Walsh@GSK.com (J.S. Walsh).
`
`0009-2797/02/$ - see front matter # 2002 Elsevier Science Ireland Ltd. All rights reserved.
`PII: S 0 0 0 9 - 2 7 9 7 ( 0 2 ) 0 0 0 5 9 - 5
`
`1 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`136
`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`Keywords: Abacavir; HIV-1; Carboxylic acid; Glucuronide
`
`1. Introduction
`
`Adverse drug reactions represent an area of increasing clinical concern for both
`prescribing physicians and pharmaceutical companies [1,2]. Approximately 25% of
`these are designated as idiosyncratic, or hypersensitivity reactions, in that they do
`not occur in most patients, and do not involve the known pharmacological
`properties of the drug [3]. There is substantial evidence of immune system mediation,
`which in turn may be initiated through metabolic activation of the drug to reactive
`species which covalently bind to proteins [4,5].
`Abacavir (ZIAGEN†, Fig. 1) is a nucleoside reverse transcriptase inhibitor
`marketed in 1999 for the treatment of HIV-1 infection. Approximately 4% of
`
`Fig. 1. Major routes of metabolism of abacavir in humans. Following a 600 mg oral dose of 14C-abacavir,
`83% of the radioactivity was excreted in the urine, and 16% in the feces. In urine, the acid metabolite
`(2269W93) accounted for 30% of the dose and the glucuronide for 36%, with 1% unmetabolized abacavir.
`The remaining 16% of dose was comprised of numerous minor metabolites [8]. A number of these have
`been characterized, and include two isomers of 2269W93. The fecal component was comprised primarily of
`2269W93 and abacavir.
`
`2 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`137
`
`patients experience a hypersensitivity reaction. The symptoms are varied, but fever
`and rash are the most common [6], and a small number of fatalities have also been
`reported. The clinical presentation is consistent with immune mediation, and hence
`bioactivation is likely to play a role in its origination.
`As part of broader investigations to understand the etiology of this hypersensi-
`tivity, metabolic investigations were performed to identify potential bioactivation
`pathways for abacavir, and the enzyme systems involved. While many factors are
`likely to play a role in idiosyncratic toxicities [7], genetic polymorphisms in drug
`metabolizing enzymes are factors that can potentially be identified.
`The major routes of metabolism of abacavir in humans are conjugation to an ether
`glucuronide, and oxidation to a carboxylic acid metabolite designated as 2269W93
`[8], (Fig. 1). Approximately 15% of the dose is comprised of a number of minor
`metabolites, but from structural considerations, none of those identified to date
`indicate the potential for reactive intermediates. In contrast, the metabolism of
`abacavir to 2269W93 involves a two step oxidation process via an aldehyde
`intermediate. This aldehyde has not been observed directly as a metabolite of
`abacavir, and attempts to synthesize it have been unsuccessful due to its apparent
`instability.
`A number of aldehyde metabolites have been previously implicated as reactive and
`capable of covalent binding to proteins, and this in turn has been suggested to
`underlay clinical adverse events of the parent compounds. In these cases, the
`underlying reactivity can be ascribed to one of two mechanisms: Schiff base
`formation, or a 1,4 addition process. Schiff base formation has been proposed for
`the aldehyde metabolite of Sorbinil, which shows an incidence of hypersensitivity [9],
`and for ethanol, in which acetaldyde formation is proposed to underlay the immune
`hepatitis observed with chronic alcohol consumption [10]. The reversibility of this
`Schiff base formation has been proposed to be limited either by reduction by
`ascorbate [11], or for acetaldehyde, cyclization with an amide nitrogen of the protein
`to form a stable imidazolidinone [12]. Thus mechanisms may exist in vivo for the
`stabilization of these somewhat unstable adducts. A more widely encountered
`mechanism is observed for ab unsaturated aldehydes (Michael type acceptors) and
`numerous examples exist in the literature of proposals for this type of reactive
`intermediate being associated with covalent binding and adverse clinical events [13 /
`15].
`As discussed below, both mechanisms might be possible for abacavir. The
`purposes of the studies described here were to utilize in vitro methods to determine
`if oxidation of abacavir to the carboxylic acid via an aldehyde intermediate could
`lead to protein covalent binding, to investigate the mechanism, and to identify the
`enzyme systems involved.
`
`3 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`138
`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`2. Materials and methods
`
`2.1. Chemicals
`
`Human liver microsomes and cytosol were obtained from Xenotech, (Kansas City,
`KS). NADP, glucose-6-phosphate, glucose-6-phosphate dehydrogenase, NAD, 4-
`MP, methoxylamine, horse liver alcohol dehydrogenase (HLADH), HSA, triethy-
`lamine, toluene, isobutylchloroformate, and 7-ethoxycoumarin were obtained from
`Sigma-Aldrich (St Louis, MO). Laemmli sample buffer was obtained from Bio-Rad
`(Hercules, CA). Ultima-Flo M was obtained from Packard (Meriden, CT). Gelcode
`Blue Reagent, SuperBlock Blocking Buffer, SuperSignal West Pico Chemilumines-
`cent Substrate, horseradish peroxidase-conjugated goat anti-rabbit IgG (H/L
`chains), BupH Tris Buffered Saline Packs, Blue Ranger pre-stained protein
`standards, and keyhole limpet hemocyanin (KLH) were obtained from Pierce
`(Rockford, IL). Immobilized recombinant protein A IPA-400HC was purchased
`from RepliGen (Needham, MA). Abacavir, 14C- abacavir, 2269W93, and dihydro-
`abacavir were synthesized at GlaxoSmithKline.
`Human ADH isozymes were obtained as a gift from Dr.Tom Hurley, Dept of
`Biochemistry and Molecular Biology, Indiana University Medical School.
`Isozymes were analyzed by SDS-PAGE and each shown to be comprised of a
`single major protein. The identities of the aa and g2g2 isozymes were independently
`confirmed by LC/MS/MS analysis of tryptic digests using MASCOT protein
`database searching [16].
`
`2.2. Microsomal incubations
`
`14C-Abacavir (3 mg/ml; 0.5 mCi/ml) was incubated with 1 mg/ml pooled human
`liver microsomes in 100 mM potassium phosphate buffer, pH 7.4, containing 1 mM
`NADP, 5 mM MgCl2, 10 mM glucose-6-phosphate, and 0.5 Units/ml glucose-6-
`phosphate dehydrogenase. Incubations containing no NADP were used as negative
`controls. After incubating the samples in duplicate for 1-20 h in a 37 8C shaking
`incubator, 50 ml of each incubation was removed and added to 50 ml of cold
`acetontrile. The samples were vortexed and centrifuged in an Eppendorf model 5314
`microcentrifuge at 10,000 rpm for 5 min. An aliquot of 20 ml was analyzed by HPLC
`for metabolite profiling. The remainder of the incubations were analyzed for non-
`extractable residue formation as described below. Metabolic viability of
`the
`microsomes was confirmed using 7-ethoxycoumarin.
`
`2.3. Cytosol and expressed ADH incubations
`
`14C-Abacavir (3 mg/ml; 0.5 mCi/ml) was incubated with 1 mg/ml of pooled human
`liver cytosol in 50mM sodium pyrophosphate buffer, pH 7.4 or pH 8.8, containing
`7.5 mM NAD. Incubations containing no NAD were used as negative controls.
`When used, 4-MP was added as an aqueous solution at the start of the incubation, to
`give a final concentration of 0.6 mM. Incubations were run in duplicate for 2-20
`
`4 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`139
`
`hours in a 37 8C shaking incubator, and then placed on ice for 5 min prior to
`filtering a 100 ml aliquot through a 0.45 mm AcroPrep GHP filter plate using a
`Waters Alliance filtration system. Aliquots of /20 ml were analyzed by HPLC and
`LC/MS for metabolite profiling.
`Incubations with human ADH isozymes were performed as described for cytosol,
`in 50 mM sodium pyrophosphate buffer, pH 7.4 and pH 7.8 (aa isozyme), or pH 8.8.
`Screening for metabolic activity with abacavir was performed with the different
`ADH isozymes using constant units of activity (0.025 Units/ml each) with variable
`enzyme protein level, and at constant enzyme protein level (17 mg/ml) with a variable
`number of units. Incubations were analyzed at 1, 2, 3, 4 and 20 hours by HPLC.
`When present, HSA was at a final concentration of 20 mg/ml. Incubations with
`dihydro-abacavir were performed under the same conditions. ADH inhibition by 4-
`MP was performed as described for cytosolic incubations. For trapping experiments
`with methoxylamine, an aqueous solution was added at the start of each incubation
`such that the final concentration was 10 mM.
`All human ADH isozymes were assayed for viability using either ethanol (aa,
`b1b1, b2b2, g2g2, p and s) or pentanol (x) as substrates. Metabolism was assessed
`by measuring initial rates of NADH formation spectrophotometrically, and
`expressed as units of activity (mmol NADH formed/min) as previously described [17].
`
`2.4. Measurement of Non-Extractable Protein Residues
`
`Incubations were placed on ice for 5 min and the proteins precipitated by adding 3
`ml cold acetonitrile. The samples were vortexed and centrifuged at 2500 rpm for 5
`min at 4-8 8C. The supernatants were removed and the pellets resuspended in 3 ml
`cold acetonitrile, vortexed, and centrifuged as before for a total of 5 washes or until
`the radioactivity counted in the washes wereB/2 times background. The pellets were
`dissolved in 1 ml 1%SDS and 0.8 ml was counted in a liquid scintillation counter.
`Some samples were further analyzed by SDS-PAGE/Western blot analysis.
`
`2.5. HPLC and LC/MS analyses
`
`Incubation supernatants were analyzed using a Waters Alliance 2690 HPLC
`system. The eluate was monitored using a Waters 996 photo diode array detector
`(l/295nm) and a Berthold Model LB 507A radioactivity flow monitor (EG & G
`Berthold, Nashua, NH), equipped with a 1 ml flow cell. The HPLC eluate was mixed
`with 3 volumes of Ultima-Flo M scintillation cocktail using a Waters 510 pump prior
`to entering the flow cell. HPLC analyses were performed on a Kromasil C18, 5 mm
`column (3.2/150mm; Phenomenex, Torrance, CA) with a mobile phase consisting
`of 0.025 M ammonium acetate buffer (pH 4) containing 5% methanol (A) and
`acetonitrile (B) delivered as a linear 50 min gradient of 0-13% B, at a flow rate of 0.7
`ml /min.
`While optimum chromatographic resolution of metabolites was achieved with the
`chromatographic conditions described above, this gave poor mass spectral response.
`For LC/MS analyses, the same column was used with a mobile phase consisting of
`
`5 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`140
`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`5% methanol/0.1% formic acid/water (A) and 0.1% formic acid/acetonitrile (B)
`delivered as a linear 50 min gradient of 0-13% B, at a flow rate of 0.7 ml /min.
`Samples were analyzed using a Quattro II triple quadrupole mass spectrometer
`(Micromass Ltd, Manchester, UK) equipped with a Z-spray ion source operated in
`positive ion mode. Nitrogen was used as the nebulizing and drying gas. Source and
`desolvation temperatures were 100 and 350 8C, respectively. Capillary and cone
`voltages were 2.5 kV and 15 V, respectively. The multiplier voltage was 650 V. Full-
`scanning mass spectra were acquired between 100-500 amu. Data were processed
`using MassLynx 3.3 software.
`
`2.6. Production of rabbit anti-abacavir antibodies
`
`Two abacavir analogs with spacer and carboxyl groups attached to different parts
`of the molecule (analogs A and B, Fig. 9) were conjugated to KLH using the mixed
`anhydride reaction. Mixed anhydrides were formed by mixing triethylamine and
`isobutylchloroformate under nitrogen in an ice bath for 45 min, then added dropwise
`to 25 ml of 0.8 mg/ml KLH in 0.1 M sodium bicarbonate buffer. The reaction was
`allowed to proceed 3 hr at ambient room temperature, then overnight at 48C. The
`reaction mix was dialyzed against phosphate buffer saline and concentrated by
`ultrafiltration to approximately 2 mg/ml in sterile water.
`Conjugation ratios were determined by hapten absorbance at 250 nm corrected for
`KLH, and by protein assay;
`the extinction coefficient
`for each hapten was
`determined empirically. The abacavir:KLH ratios for the KLH conjugates were
`approximately 264 (analog A) and 502 (analog B). Analog A: KLH immunoconju-
`gate was selected as the immunogen, and was diluted 1:1 with either complete
`(primary immunization) or incomplete (boosting immunizations) Freund’s adjuvant;
`New Zealand rabbits were immunized over a period of 6 weeks. Serum antibodies
`were characterized for binding to abacavir analogs using surface plasmon resonance
`(SPR) on a Biacore 2000 (Fig. 9). The KLH conjugates (or KLH alone; control
`surface) were immobilized on a carboxymethyldextran-coated gold chip using amine
`coupling. Serum samples were diluted 1/10 in Hepes-buffered saline, and injected at a
`flow rate of 20 ml/min for 2 min prior to determination of resonance units (RU)
`binding signal. The biosensor surface was regenerated with a 30-sec wash of glycine-
`HCl (pH 1.5) followed by a 30-sec wash with 250 mM NaOH containing 0.1% SDS.
`Antibody specificity was further analyzed by binding to the analog B: KLH
`conjugate. As shown in Fig. 9, the antibody recognized both analog A: and analog B:
`KLH conjugates, demonstrating broad specificity of the antibody. Antibody was
`purified by affinity chromatography on immobilized recombinant protein A
`(RepliGen IPA-400 HC), eluted with 0.2 M glycine buffer (pH 2), and neutralized
`with Tris-HCl buffer (pH 8). The purified antibody was used in western blotting
`experiments to detect abacavir conjugated to HSA.
`
`6 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`141
`
`2.7. SDS-PAGE with Immunoblotting
`
`Samples were prepared for SDS-PAGE analysis by diluting 1:1 in Laemmli sample
`buffer with 0.7 M 2-mercaptoethanol and boiling for 7 min. SDS-PAGE was
`performed using a Mini-PROTEAN II gel system (Bio-Rad) with either 10% or 4 /
`15% gradient Tris-HCl pre-cast gels (Bio-Rad). For immunoblot analysis, the
`resolved proteins were transferred electrophoretically to nitrocellulose membranes
`for 1 hour at 100 V using a Mini Trans-Blot cell (Bio-Rad). The following steps were
`conducted at room temperature with constant shaking. The membranes were
`blocked for 1 hr with blocking buffer (Super Block blocking buffer containing
`0.05% Tween 20), and then incubated with the rabbit anti-abacavir antibodies
`diluted 1:2000 in blocking buffer. Unbound antibodies were removed by washing the
`membranes in wash buffer [Tris Buffered Saline (TBS) containing 0.05% Tween-20]
`(5 washes/5 min each). The membranes were then incubated for 1 hr with
`horseradish peroxidase-conjugated goat anti-rabbit IgG (H/L chains) diluted
`1:50,000 in blocking buffer, and then washed with wash buffer (6 washes/5 min
`each). The blots were developed by incubating with SuperSignal West Pico
`Chemiluminescent Substrate for 5 min, and exposing the nitrocellulose membranes
`to ECL film under safe-light conditions.
`
`3. Results
`
`3.1. Microsomal and cytosol incubations
`
`All incubations were performed at physiologically relevant concentrations (/
`clinical Cmax). No metabolites were detected in incubations of 14C-abacavir with
`human liver microsomes at pH 7.4 from 1 /20 h. In the human liver cytosolic
`incubations containing NAD, polar metabolites were detected at 20 h at pH 7.4,
`including a peak of the same retention time as the major acid metabolite observed in
`vivo, 2269W93 (Fig. 2). Cytosolic NAD-dependent oxidation of alcohols is
`indicative of ADH involvement. This enzyme system has in vitro pH optimums
`higher than 7.4 [18] and is inhibited by 4-MP. As indicated in Fig. 2, significantly
`more metabolism was observed in cytosol at pH 8.8, and metabolism at this pH was
`inhibited completely with 4-MP.
`Following incubations of abacavir with cytosol or microsomes, proteins were
`precipitated, extensively washed, and non-extractable residues determined radio-
`chemically (Fig. 3). No NADPH-dependent non-extractable residues were observed
`in microsomal incubations at 2 h (at which point CYP450 activity is typically
`decreasing). Similarly, no NAD-dependent residues were observed in cytosolic
`incubations at pH 7.4 after 2 h, but at 20 h, NAD-dependent residues were clearly
`observed, and these residues were substantially increased at pH 8.8. Both
`metabolism and residue formation were also blocked in cytosol by 4-MP. These
`results implicate ADH involvement in metabolism and residue formation, rather
`than CYP450.
`
`7 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`142
`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`Fig. 2. Radiochemical profiles of 14C-abacavir after incubating 20 h with human liver cytosol. UV
`detection was used for the 2269W93 reference standard. The retention time of 2269W93 was identical to
`the major metabolite in the cytosolic UV profile.
`
`3.2. Expressed Human ADH Incubations
`
`Some of the properties of the known human ADH isozymes are summarized in
`Table 1. All human isozymes were examined except the g1g1 and b3b3 isoforms,
`which were unavailable. While metabolic turnover with ethanol or pentanol was
`observed at pH 7.4, all isozymes showed significantly higher metabolic activity at pH
`8.8.
`14C-Abacavir was incubated with the different human ADH isoforms at pH 8.8
`and the incubations analyzed by HPLC for evidence of metabolism from 1 to 20 h.
`At 2 h, only the aa and g2g2 isoforms showed any evidence of product formation,
`and further metabolism was observed out to 20 h. No metabolites were detected with
`
`8 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`143
`
`Fig. 3. Non-extractable residues for microsomal (mx) and cytosolic incubations of 14C-abacavir.
`
`Table 1
`
`Gene Alleles Subunit Classa In vitro pH opti-
`mum
`
`Distribution Specific activity (Units/mg pro-
`teinc)
`
`ADH1 ADH1 a
`2 b1
`ADH2 ADH1
`2 b2
`ADH2
`2 b3b
`ADH3
`3 g1b
`ADH3 ADH1
`3 g2
`ADH2
`ADH4 ADH4 p
`ADH5 ADH5 x
`ADH7 ADH7 s
`
`I
`I
`I
`I
`I
`I
`II
`III
`IV
`
`10.5
`10.5
`8.5
`7.0
`10.5
`10.5
`10.5
`10.5
`10.5
`
`1.8d
`Hepatic
`0.013e
`Hepatic
`6.4d
`Hepatic
` /
`Hepatic
` /
`Hepatic
`0.34d
`Hepatic
`0.07e
`Hepatic
`Many tissues 0.37f
`9.2d
`Stomach
`
`a Only class I are sensitive to inhibition by 4-MP.
`b Isozymes not examined in this study.
`c Assayed as described previously [17], at pH 8.8. Mean of 2 /4 determinations.
`d 30 mM ethanol.
`e 600 mM ethanol.
`f 300 mM pentanol.
`
`any of the other isoforms out to 20 h at pH 8.8. For the different isozymes, specific
`activities (i.e. units of activity/mg of protein) were variable, and so incubations were
`run both at constant units of activity with variable enzyme protein levels, and at
`constant enzyme protein levels with variable units of activity. Under both sets of
`conditions, only the aa and g2g2 enzymes showed metabolic activity with abacavir.
`Fig. 4 shows the radiochemical profiles at 20 h for the aa and g2g2 incubations run
`at pH 8.8. The aa isozyme gave a profile similar to that seen in cytosol, with a peak
`of the same retention time of the acid metabolite, and at least one other polar
`product. The profile for the g2g2 isoform was quite different. No acid metabolite or
`polar peaks were observed. Instead a single slightly less polar product was generated,
`which was minor in the aa incubations. All of these peaks were NAD and enzyme
`dependent.
`
`9 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`144
`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`Fig. 4. Radiochemical profiles of 14C-abacavir after incubating 20 h with expressed human aa and g2g2
`ADH isozymes.
`
`Because high pH optimums are required for alcohol oxidation by ADH, the effect
`of pH on abacavir metabolism by the aa isozyme was examined at lower, more
`physiological pHs. Incubations at pH 7.4 showed only trace metabolism. At pH 7.8,
`metabolite peaks were clearly observed, giving a similar qualitative profile to that
`observed at pH 8.8 (Fig. 4). Insufficient quantities of the g2g2 isozyme were available
`to perform these studies with this isozyme.
`
`10 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`145
`
`Fig. 5. Selected ion traces for incubations of 14C-abacavir with expressed human aa and g2g2 isozymes.
`The aa isozyme traces (a, b) show the protonated molecular ion for the 14C isotope of the carboxylic acid
`metabolite (m/z 303) (a), and the 12C isotope (m/z 301) when co-injected with 2269W93 (b). The
`protonated molecular ion for the 14C isotope of abacavir (m/z 289) is shown for the g2g2 isozyme (d), and
`in the absence of enzyme (e).
`
`LC/MS profiles for the aa and g2g2 incubations run at pH 8.8 are shown in Fig. 5.
`Analysis was done under different chromatographic conditions than used for the
`radiochemical profiling, so relative retention times are shifted. The aa isozyme trace
`(Fig. 5, a) shows the protonated molecular ion for the 14C isotope of 2269W93 (m/z
`303) which was the major isotope in these incubations due to the high specific
`activity of abacavir used. The protonated molecular ion for reference 2269W93 is m/
`z 301 (12C isotope). In the aa incubations, three discrete peaks with apparent
`molecular ions corresponding to carboxylic acid were detected, and one of these co-
`eluted with 2269W93 in the m/z 301 trace for the incubation (Fig. 5, b). These peaks
`are absent in incubations containing abacavir but no enzyme (Fig. 5, c). For the g2g2
`
`Fig. 6. Non-extractable 14C residues of incubations of 14C-abacavir with aa and g2g2 human ADH
`isozymes, in the presence of human serum albumin.
`
`11 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`Fig.7.Radiochemicalprofilesofaaandg2g2incubationsof14C-abacavirinthepresenceof10mMmethoxylamine.
`
`146
`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`12 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`147
`
`incubations, these acid metabolites were detected in only trace amounts by LC/MS.
`In contrast, the less polar peak, which was minor in the aa incubations, was observed
`as the only major metabolite and had the same apparent molecular weight as parent
`(m/z 289 for the 14C isotope, Fig. 5, d). This peak is absent in incubations with no
`enzyme (Fig. 5, e).
`Incubations of both aa and g2g2 ADH enzymes in the presence of HSA gave rise
`to non-extractable protein residues, which were inhibited by 4-MP. For the aa
`isozyme, a lower level of 4-MP sensitive residue was detected at pH 7.4 (Fig. 6).
`While the aldehyde intermediate was not observed directly in these incubations, its
`presence was confirmed by trapping in situ with methoxylamine. Fig. 7 shows the
`radiochemical profiles of such incubations,
`in which the previously observed
`metabolites are substantially reduced, and a less polar peak is now detected as the
`major product with both isozymes. LC/MS analysis indicated this to have an
`apparent MH/ of 316/314 (for 14C/12C isotopes) consistent with an oxime
`derivative. The mass spectrum for the oxime is shown in Fig. 8.
`
`3.3. Anti-abacavir antibody characterization
`
`The specificity of the anti-abacavir antibody was evaluated by biosensor analysis
`(Fig. 9). Even in the absence of steady-state binding during the association phase,
`significant interaction with the abacavir analog A: KLH immunoconjugate can be
`seen from 170 /350 s (the dissociation phase) compared to the KLH protein alone.
`There was also significant binding to the analog B: KLH conjugate, confirming that
`
`Fig. 8. Mass spectrum of non-polar product observed in ADH incubations with methoxylamine. The
`spectrum shown was obtained from incubation using commercial horse liver alcohol dehydrogenase,
`where higher metabolic turnover was seen. The protonated molecular ions (316/314) for both carbon
`isotopes for the oxime were detected in incubations with the aa and g2g2 human isozymes.
`
`13 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`148
`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`Fig. 9. Rabbit serum antibody binding to immobilized abacavir analogs A (immunogen) and B, coupled
`to KLH. Biosensor data are shown in resonance units (RU), and convey relative antibody binding activity
`to different surfaces [35]. The antibody was suitable for immunoblotting as it was able to recognize
`abacavir when covalently bound to protein either through the purine or cyclopentene ring.
`
`the rabbit antibody was able to recognize abacavir when conjugated to KLH from
`different sites in the molecule.
`
`3.4. Western blot analyses of expressed human ADH incubations
`
`The covalent nature of the non-extractable residues was confirmed by Western
`blot analysis of the incubations. Abacavir incubations were run with the aa and g2g2
`isozymes in the presence of HSA and NAD. Fig. 10 shows immunoblots for these
`incubations together with those for incubations without NAD, and with NAD and
`4-MP. Also shown are the total protein (Coomassie) stains. While a major protein
`band corresponding to HSA was readily detected in all incubations with Coomassie
`staining, only those incubations with NAD and without 4-MP were observed as
`immunoreactive.
`
`3.5. Metabolism of dihydro-abacavir by human aa ADH isozyme
`
`The dihydro analog of abacavir was incubated with both aa and g2g2 isoforms
`using the same conditions as for abacavir. A comparison of the non-extractable
`
`14 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`149
`
`Fig. 10. Immunoblots using the rabbit serum anti-abacavir antibody and Coomassie-stained SDS-PAGE
`gels for abacavir incubations with aa and g2g2 isozymes in the presence of human serum albumin. ABC:
`Abacavir. MWS: Molecular weight standards
`
`Fig. 11. A comparison of non-extractable 14C residue levels and extent of metabolism, for abacavir and
`dihydro-abacavir with the aa human ADH isozyme. Percent metabolism was determined from
`radiochemical profiling, and refers to the percent conversion to total metabolites. Incubations were also
`analyzed by LC/MS. Two isomeric carboxylic acid metabolites were identified as the major products with
`dihydro-abacavir.
`
`15 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`150
`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`residue levels and extent of metabolism by ADH aa of abacavir and the dihydro
`analog are shown in Fig. 11. Despite the fact that the dihydro analog was
`metabolized more extensively than abacavir, the level of non-extractable residue
`was substantially reduced.
`
`4. Discussion
`
`The metabolism of primary alcohols to carboxylic acids is most commonly
`mediated by cytosolic ADH or microsomal CYP450 [19,20]. No metabolites could be
`detected radiochemically in incubations of abacavir with pooled human liver
`microsomes, but good conversion to the acid metabolite was observed with human
`liver cytosol. An aldehyde would be the initial product of such metabolism, and the
`formation of the acid directly in cytosol might involve a role for aldehyde
`dehydrogenase. However, ADH is well known to be capable of oxidizing aldehydes
`further to carboxylic acids via gem diol formation, and /or dismutation [21,22].
`Incubations of 14C-abacavir with cytosol also lead to NAD dependent non-
`extractable protein residues. The pH and 4-MP sensitivity for both metabolism and
`residue formation implicate ADH in both processes.
`The ADH system in humans has been extensively reviewed [23,24] (Table 1). While
`methodology for phenotyping with expressed human CYP450s is well established,
`previous reports of such studies with expressed human ADH isozymes are very
`limited [25]. The high in vitro pH optimums for oxidation of alcohols by ADH
`enzymes can pose a problem for these studies if pH sensitive substrates or
`intermediates are involved. Minimal oxidation of alcohols may be observed at pH
`7.4, and under the conditions used here, low activity with ethanol or pentanol as
`substrates was seen. As a balance between optimizing turnover and minimizing base
`catalyzed side reactions, ADH activity can be examined at an intermediate pH [26 /
`28]. Metabolic turnover of ethanol or pentanol was significantly enhanced at pH 8.8
`(Table 1). When abacavir was incubated with the ADH isozymes at pH 8.8,
`metabolism was observed only with the aa and g2g2 isoforms. For the aldehyde
`intermediate in abacavir oxidation, the enolizable nature of the chiral center adjacent
`to the aldehyde functionality would suggest the potential for pH dependence on
`isomerization processes at this center. To ascertain if isomers of 2269W93 were
`arising solely due to pH effects, some studies were also conducted at lower pH. As
`shown in Fig. 2, in cytosol, qualitatively similar profiles are observed at pH 7.4 and
`8.8. In addition, non-extractable, cofactor-dependent residues could also be
`measured at pH 7.4. When abacavir was incubated with the aa isoform at pH 7.4,
`only very low levels of metabolism could be detected, but a low level of NAD-
`dependent non-extractable protein residue was observed (Fig. 6). At pH 7.8, isomeric
`acid metabolites are clearly detected in incubations with the aa enzyme (Fig. 4).
`Taken collectively, these results suggest that the isomerization processes are not
`solely artifactual due to pH, but are directly related to enzyme activity. The
`observation of two isomers of 2269W93 as minor metabolites in human urine
`following dosing with abacavir supports this proposal.
`
`16 of 20
`
`PENN EX. 2039
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`J.S. Walsh et al. / Chemico-Biological Interactions 142 (2002) 135 /154
`
`151
`
`Radiochemical profiling and LC/MS analysis of the ADH incubations showed
`that the major pathways of metabolism for abacavir with the two isozymes are
`different. The aa isozyme generated a peak of the same retention time as 2269W93,
`and two apparent isomers of this. The formation of acid metabolites in these
`incubations indicates the capability of the aa isozyme to oxidize the aldehyde
`intermediate,
`in addition to abacavir itself. While this does not rule out the
`possibility of aldehyde dehydr

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