throbber
Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on September 1, 2015
`
`Supplemental material to this article can be found at:
`http://dmd.aspetjournals.org/content/suppl/2012/04/10/dmd.112.045450.DC1.html
`
`1521-009X/12/4007-1345–1356$25.00
`DRUG METABOLISM AND DISPOSITION
`Copyright © 2012 by The American Society for Pharmacology and Experimental Therapeutics
`DMD 40:1345–1356, 2012
`
`Vol. 40, No. 7
`45450/3776821
`
`Characterization of the In Vitro and In Vivo Metabolism and
`Disposition and Cytochrome P450 Inhibition/Induction Profile of
`Saxagliptin in Human□S
`
`Hong Su, David W. Boulton, Anthony Barros Jr., Lifei Wang, Kai Cao, Samuel J. Bonacorsi Jr.,
`Ramaswamy A. Iyer, W. Griffith Humphreys, and Lisa J. Christopher
`
`Departments of Pharmaceutical Candidate Optimization (H.S., A.B., L.W., R.A.I., W.G.H., L.J.C.), Discovery Medicine and
`Clinical Pharmacology (D.W.B.), and Radiochemistry (K.C., S.J.B.), Bristol-Myers Squibb Research, Princeton, New Jersey
`
`Received March 2, 2012; accepted April 10, 2012
`
`ABSTRACT:
`
`Saxagliptin is a potent dipeptidyl peptidase-4 inhibitor approved
`for the treatment of type 2 diabetes mellitus. The pharmacokinetics
`and disposition of [14C]saxagliptin were investigated in healthy
`male subjects after a single 50-mg (91.5 ␮Ci) oral dose. Saxagliptin
`was rapidly absorbed (Tmax, 0.5 h). Unchanged saxagliptin and
`5-hydroxy saxagliptin (M2), a major, active metabolite, were the
`prominent drug-related components in the plasma, together ac-
`counting for most of the circulating radioactivity. Approximately
`97% of the administered radioactivity was recovered in the excreta
`within 7 days postdose, of which 74.9% was eliminated in the urine
`and 22.1% was excreted in the feces. The parent compound and
`M2 represented 24.0 and 44.1%, respectively, of the radioactivity
`recovered in the urine and feces combined. Taken together, the
`excretion data suggest that saxagliptin was well absorbed and was
`
`subsequently cleared by both urinary excretion and metabolism;
`the formation of M2 was the major metabolic pathway. Additional
`minor metabolic pathways included hydroxylation at other posi-
`tions and glucuronide or sulfate conjugation. Cytochrome P450
`(P450) enzymes CYP3A4 and CYP3A5 metabolized saxagliptin and
`formed M2. Kinetic experiments indicated that the catalytic effi-
`ciency (Vmax/Km) for CYP3A4 was approximately 4-fold higher than
`that for CYP3A5. Therefore, it is unlikely that variability in expres-
`sion levels of CYP3A5 due to genetic polymorphism will impact
`clearance of saxagliptin. Saxagliptin and M2 each showed little
`potential to inhibit or induce important P450 enzymes, suggesting
`that saxagliptin is unlikely to affect the metabolic clearance of
`coadministered drugs that are substrates for these enzymes.
`
`Introduction
`The dipeptidyl peptidase-4 (DPP4) inhibitors are promising re-
`cent additions to the arsenal of therapies available for the treatment
`of type 2 diabetes mellitus (Scheen, 2012). The DPP4 enzyme is
`
`Parts of this work were previously presented as follows: Christopher LJ, Su H,
`Barros A Jr, Wang L, Cao K, Bonacorsi S Jr, Iyer RA, and Humphreys WG (2009)
`Identification of the enzymes involved in the oxidative metabolism of saxagliptin
`and kinetics of formation of its major hydroxylated metabolite; Abstract 289. 16th
`North American Regional International Society for the Study of Xenobiotics Meet-
`ing; 2009 Oct 18–22; Baltimore, MD.
`International Society for the Study of
`Xenobiotics, Washington, DC; Su H, Christopher LJ, Iyer RA, Cao K, Bonacorsi S
`Jr, and Humphreys W (2011) In vivo disposition and pharmacokinetics and in vitro
`inhibition and induction profiles of [14C]saxagliptin, a potent inhibitor of dipeptidyl
`peptidase 4, in human. Abstract P236. 17th North American Regional Interna-
`tional Society for the Study of Xenobiotics Meeting; 2011 Oct 16–20; Atlanta, GA;
`International Society for the Study of Xenobiotics, Washington, DC.
`Article, publication date, and citation information can be found at
`http://dmd.aspetjournals.org.
`http://dx.doi.org/10.1124/dmd.112.045450.
`□S The online version of this article (available at http://dmd.aspetjournals.org)
`contains supplemental material.
`
`responsible for degrading and inactivating glucagon-like peptide-1
`(GLP-1) and glucose-dependent insulinotropic peptide, incretins
`that regulate blood glucose levels. GLP-1 is released postprandi-
`ally and stimulates meal-induced insulin secretion and contributes
`to glucose homeostasis (Kieffer and Habener, 1999; Gorrell,
`2005). By inhibiting the DPP4 enzyme, GLP-1 is sustained,
`thereby leading to increased activity and improved glycemic con-
`trol
`in patients with type 2 diabetes (McIntosh et al., 2005).
`Because this mechanism results in a glucose-dependent release of
`insulin, DPP4 inhibitors are expected to offer important advantages
`over traditional diabetes treatments including low risk for hypo-
`glycemia and weight gain (Gallwitz, 2008).
`Saxagliptin (Onglyza; Bristol-Myers Squibb, Princeton NJ and
`AstraZeneca, Wilmington, DE) (Fig. 1) is an orally administered,
`small molecule, reversible DPP4 inhibitor approved for the treatment
`of type 2 diabetes mellitus. It was specifically designed for enhanced
`potency and selectivity and to provide extended inhibition of the
`DPP4 enzyme (Augeri et al., 2005). The ability of saxagliptin to affect
`reductions in glycosylated hemoglobin (HbA1C) and fasting plasma
`glucose in type 2 diabetes patients has been demonstrated in multiple
`
`ABBREVIATIONS: DPP4, dipeptidyl peptidase-4; GLP-1, glucagon-like peptide-1; P450, cytochrome P450; HPLC, high-performance liquid
`chromatography; HLM, human liver microsomes; LSC, liquid scintillation counting; LC-MS/MS, liquid chromatography with tandem mass
`spectrometry; MRM, multiple reaction monitoring; AUC, area under plasma concentration-time curve; T-HALF, terminal phase half-life; CLR, renal
`clearance; DMSO, dimethyl sulfoxide; 3-MC, 3-methylcholanthrene; PB, phenobarbital; RIF, rifampicin; CT, threshold cycle.
`
`1345
`
`Page 1 of 12
`
`AstraZeneca Exhibit 2045
` Mylan v. AstraZeneca
` IPR2015-01340
`
`

`
`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on September 1, 2015
`
`1346
`
`SU ET AL.
`
`FIG. 1. Primary pathways for biotransformation of
`[14C]saxagliptin in humans. The C-14 label was
`evenly distributed between the carbonyl carbon and
`the adjacent carbon, as indicated by asterisk (ⴱ) on
`the saxagliptin structure. The percentage of admin-
`istered radioactivity recovered in urine and feces as
`saxagliptin and M2 is indicated underneath the struc-
`tures. The estimated flux through each pathway (also
`reported as percentage of administered radioactivity)
`is indicated next to the corresponding arrow. D1, a
`degradant known to form in solution, was found in
`small quantities in all samples.
`
`Phase III clinical trials, both as a single agent and in combination
`regimens with metformin, a sulfonylurea or a thiazolidinedione (Ka-
`nia et al., 2011). The most commonly used clinical dose of saxagliptin
`in adults is 5 mg, once daily (United States prescribing information for
`Onglyza, http://www.packageinserts.bms.com/pi/pi_onglyza.pdf).
`In nonclinical pharmacokinetic studies, saxagliptin was rapidly
`absorbed and showed good oral bioavailability in rats (75%), dogs
`(76%), and monkeys (51%). A significant portion (33– 60%) of the
`administered dose was excreted as unchanged drug in the urine in
`these species. Formation of 5-hydroxy saxagliptin (M2) was a major
`metabolic pathway, and this metabolite was a major circulating me-
`tabolite in all species (Fura et al., 2009). Metabolite M2 was phar-
`macologically active, with an in vitro DPP4 inhibitory activity ap-
`proximately half that of saxagliptin (Augeri et al., 2005; Fura et al.,
`2009).
`The purpose of the current study was to investigate the in vivo
`disposition of saxagliptin and to determine its major metabolic path-
`ways in healthy male subjects after administration of a single 50-mg
`(91.5 ␮Ci) p.o. dose of [14C]saxagliptin. In addition, a series of in
`vitro studies were conducted to gain insight regarding possible cyto-
`chrome P450 (P450)-based drug-drug interactions between saxaglip-
`tin and potential comedications. These included the identification of
`enzymes involved in the metabolism of saxagliptin and formation of
`M2 and the determination of the potential of saxagliptin and M2 to
`inhibit or induce P450 enzymes.
`
`Materials and Methods
`Chemicals. [14C]Saxagliptin (radiochemical purity 99.86%, specific activity
`1.83 ␮Ci/mg) with the C-14 label distributed between the carbonyl carbon and the
`adjacent carbon (Fig. 1) and stable-labeled 13C4,15N-saxagliptin, and 13C4,15N-5-
`hydroxy saxagliptin [internal standards for high-performance liquid chromatogra-
`phy (HPLC) analysis] were synthesized by the Radiochemistry Group of the
`Department of Chemical Synthesis, Bristol-Myers Squibb Research (Princeton,
`
`NJ) (Cao et al., 2007). Unlabeled saxagliptin (P, (1S,3S,5S)-2-((S)-2-amino-2-(-3-
`hydroxyadamantan-1-yl)acetyl)-2-azabicyclo[3.1.0]hexane-3-carbonitrile); and
`reference standards for 5-hydroxy saxagliptin, (M2, (1S,3S,5S)-2-((S)-2-amino-2-
`((1r,3R,5S,7S)-3,5-dihydroxyadamantan-1-yl)acetyl)-2-azabicyclo[3.1.0]hexane-
`3-carbonitrile); degradant (D1, (1aS,4S,6aR,7aS)-4-(-3-hydroxyadamantan-1-yl)-
`6-iminohexahydro-1H-cyclopropa[4,5]pyrrolo[1,2-a]pyrazin-3(1aH)-one); the
`S,R,S,S and S,S,S,R diastereomers of saxagliptin (Supplemental Fig. S1) were
`supplied by the Departments of Chemical Development or Chemical Synthesis
`(Bristol-Myers Squibb).
`Selective chemical inhibitors of P450 enzymes for reaction phenotyping
`experiments were obtained from Sigma-Aldrich (St. Louis, MO), with the
`exception of montelukast, which was purchased from Sequoia Research Prod-
`ucts (Pangbourne, UK), and benzylnirvanol (BD Biosciences, Woburn, MA).
`Chemical inducers, inhibitors, substrates, and metabolites of P450 enzymes
`and internal standards used in experiments to evaluate whether saxagliptin and
`M2 were inhibitors or inducers of P450 enzymes were procured by CellzDirect
`(Pittsboro, NC). All chemicals were of the highest purity available.
`Human liver microsomes (HLM; 19 donors male/female) and individual
`human cDNA-expressed cytochrome P450 enzymes were purchased from BD
`Biosciences. Individual lots (n ⫽ 16) of HLM, for which the vendor had
`determined the activities of various P450 enzymes, were purchased as a
`Reaction Phenotyping kit (version 7) from XenoTech, LLC (Lenexa, KS).
`Monoclonal antibodies with inhibitory activity for specific P450 enzymes were
`obtained from Kristopher W. Krausz at the Laboratory of Metabolism, Na-
`tional Institutes of Health (Bethesda, MD).
`Ecolite scintillation cocktail was obtained from MP Biomedicals (Irvine,
`CA), and Emulsifier-Safe and Permofluor E⫹ scintillation fluid were obtained
`from PerkinElmer Life and Analytical Sciences (Waltham, MA). Deionized
`water was prepared using a MilliQ ultrapure water system (Millipore Corpo-
`ration, Billerica, MA). All organic solvents were HPLC grade, and other
`regents were reagent grade or better.
`Clinical Study Design, Dosing, and Sample Collection. The clinical phase
`of the study was conducted at the Bristol-Myers Squibb Clinical Research
`Center (Hamilton, NJ). This was an open-label, nonrandomized single dose
`study. Six healthy males participated in the study. The mean age was 29 years
`
`Page 2 of 12
`
`

`
`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on September 1, 2015
`
`METABOLISM AND DISPOSITION OF SAXAGLIPTIN IN HUMANS
`
`1347
`
`(range, 25 to 37 years), and the mean body mass index was 23.6 kg/m2 (range,
`19.7 to 28.1 kg/m2). The study was conducted in accordance with the Decla-
`ration of Helsinki and guidelines on Good Clinical Practice. Before study
`initiation, the study protocol and informed consent documents were approved
`by the Institutional Review Board of the New England Institutional Review
`Board (Wellesley, MA). All study participants provided written informed
`consent before the initiation of study-specific procedures.
`After a 10-h fast, each subject received a single 50-mg p.o. solution dose of
`[14C]saxagliptin containing 91.5 ␮Ci of radioactivity, immediately followed
`by 240 ml of water on day 1. Blood samples were collected at selected time
`points via an indwelling catheter or direct venipuncture into Vacutainers (BD
`Biosciences Medical Supplies, Franklin Lakes, NJ) containing K3EDTA and
`were centrifuged to obtain plasma for pharmacokinetic and biotransformation
`analysis. The total urine and fecal output was collected for the duration of the
`study (0 –168 h). On the morning of day 6, a single 30-ml oral dose of Milk of
`Magnesia was administered to each subject to facilitate defecation before
`release from the clinical facility. All subjects were released on the morning of
`day 8.
`Blood samples (6 ml total per time point) for the plasma pharmacokinetic
`analysis of saxagliptin, M2, and radioactivity were collected predose and at
`0.25, 0.5, 0.75, 1, 1.5, 2.0, 2.5, 3, 4, 6, 8, 12, 24, 36, 48, 72, 96, 120, 144, and
`168 h postdose. Additional aliquots (10 ml per time point) for metabolite
`profiling were collected in conjunction with the pharmacokinetic samples at
`predose and at 1, 2, 4, 8, 12, 24, 48, 96, 144, and 168 h postdose.
`Cumulative urine was collected predose and over 0 to 12 h, 12 to 24 h, and
`thereafter in 24-h intervals through 168 h for determining saxagliptin, M2, and
`radioactivity concentrations and for metabolite profiling. Feces were collected
`predose and over 24-h intervals postdose for the measurement of radioactivity
`concentrations and for metabolite profiling. All samples were stored at ⫺20°C
`or below until analysis.
`Radioactivity Analysis. Radioactivity in plasma, urine, and feces was
`measured by liquid scintillation counting (LSC) on a Model LS 6500 liquid
`scintillation counter (Beckman Coulter, Inc., Fullerton, CA). Plasma and urine
`were mixed with Emulsifier-Safe scintillation fluid (PerkinElmer Life and
`Analytical Sciences) and counted directly. Water was added to each fecal
`sample to form an approximately 20% (w/w) feces mixture, which was
`homogenized using a probe-type homogenizer (Kinematica Polytron model no.
`PT 45-80; Brinkman Instruments, Westbury, NY). Aliquots of fecal homoge-
`nate were then combusted using a sample oxidizer before counting by LSC, as
`described previously (Christopher et al., 2008).
`Quantification of Saxagliptin and M2 in Plasma and Urine Samples.
`The concentrations of saxagliptin and M2 in individual plasma and urine were
`determined with validated liquid chromatography/tandem mass spectrometry
`(LC-MS/MS) methods. In brief, after the addition of stable-labeled internal
`standards (13C4,15N-saxagliptin and 13C4,15N-5-hydroxy saxagliptin) to each
`plasma or urine sample, the analytes were isolated by solid-phase extraction
`(Waters Oasis HLB, 10 mg; Waters, Milford, MA). The eluates were evapo-
`rated to dryness, reconstituted in mobile phase, and then applied to an Atlantis
`dC18, 2.1 ⫻ 50 mm, 5-␮m HPLC column (Waters). The LC-MS system used
`for plasma samples consisted of LC10AD delivery pumps (Shimadzu Corpo-
`ration, Columbia, MD) and a Series 200 Autosampler (PerkinElmer Life and
`Analytical Sciences). The HPLC system was interfaced to either a Quattro
`Premier mass spectrometer (Waters Corporation, Manchester, UK) for the
`plasma method or an API3000 mass spectrometer (AB Sciex, Foster City, CA)
`for the urine method. The mass spectrometers were operated in positive ion
`electrospray mode, and analytes were monitored by multiple reaction moni-
`toring (MRM) with transitions that were characteristic for each analyte. For
`saxagliptin and M2, the standard curve ranges were 5 to 1000 and 10 to 2000
`ng/ml, respectively, for the plasma method and 25 to 5000 and 50 to 10,000
`ng/ml, respectively, for the urine method.
`Pharmacokinetic Analysis of Saxagliptin, M2, and Total Radioactivity.
`The noncompartmental pharmacokinetic parameters of saxagliptin, M2, and
`total radioactivity were determined from plasma concentration versus time
`profiles and urine concentrations with cumulative urinary excretion volumes
`using noncompartmental methods with Kinetica 4.2 in eToolbox (Thermo
`Fisher Scientific, Waltham, MA). The single-dose pharmacokinetic parameters
`determined included the following: maximum observed concentration (Cmax);
`time of maximum concentration (Tmax); area under the plasma concentration-
`
`time curve (AUC) between time 0 and the last quantifiable concentration
`[AUC(0-T)]; and AUC between time 0 to infinity AUC(INF), terminal phase
`half-life (T-HALF), renal clearance (CLR), and percentage of urinary excre-
`tion. The percentage of fecal excretion was determined for total radioactivity
`only, and the calculation was based on cumulative fecal weights and fecal total
`radioactivity concentrations. For the percentage of dose excreted in urine and
`feces, the actual dose of saxagliptin administered to each subject was deter-
`mined by subtracting the weight of the dosing syringe (in grams) after dosing
`from the weight of the dosing syringe (in grams) before dosing and multiplying
`by the density of the dosing solution (1.0 g/ml) and the concentration of the
`dosing solution (5 mg/ml).
`Preparation of Samples for Biotransformation Profiling and Identifi-
`cation of Metabolites. Representative pools of plasma, urine, and feces were
`prepared for metabolite profiling and identification experiments. Plasma sam-
`ples were segregated by collection time (i.e., 1, 2, 4, and 8 h), and equal
`volumes from all subjects were combined. Plasma samples collected after 8 h
`were not analyzed because the radioactivity in these samples was too low to
`produce meaningful profiles. Urine and fecal homogenate pools (0 –168 h)
`were prepared across all subjects by combining a percentage of the volume
`(urine) or weight (fecal homogenate) proportional to the total amount excreted
`over each interval.
`Pooled plasma and fecal homogenate samples were each extracted with
`three volumes of methanol/acetonitrile 50:50 (v/v). After centrifugation at
`2500g for 40 min, the pellets were extracted an additional two times with
`methanol/acetonitrile/water (25:25:50, v/v/v). The supernatants from each ex-
`traction step were combined and evaporated to dryness under nitrogen. The
`dried residues were reconstituted in methanol/acetonitrile/water (⬃10:20:70,
`v/v/v), and the resulting supernatants were analyzed by HPLC with offline
`radioactivity detection or LC-MS/MS. The recovery of radioactivity from
`extracted plasma and fecal samples was approximately 100%. Pooled urine
`samples were centrifuged at 11,000g to remove any particulates and analyzed
`without additional processing.
`LC-Radiochromatographic and LC-MS/MS Methods for Metabolite
`Profiling of In Vivo Samples and Identification of Metabolites. Samples for
`metabolite profiling were analyzed on a Shimadzu LC-10AD HPLC system
`(Shimadzu Corporation), equipped with two 10AD VP pumps, a SIL-10AD
`autoinjector, a model SCL-10A system controller, and an SPD-M10A photo-
`diode array detector. A Zorbax 4.6 ⫻ 250 mm, 5-␮m, RX-C8 column (Agilent
`Technologies, Santa Clara, CA) maintained at 30°C was used to separate
`drug-related components. The mobile phase consisted of two solvents:
`1) mobile phase (A) 0.1% formic acid and 1% acetonitrile in water and
`2) mobile phase (B) 0.1% formic acid in acetonitrile. The mobile phase flow
`rate was 0.5 ml/min. The gradient program used for sample elution was as
`follows: hold isocratic at 0% B (0 –5 min); linear gradient from 0 to 20% B
`(5–35 min); hold isocratic at 20% B (35– 42 min); linear gradient from 20 to
`30% B (42– 45 min); hold isocratic at 30% B (45–50 min); linear gradient from
`30 to 40% B (50 –52 min); linear gradient from 40 to 80% B (52–55 min); hold
`isocratic at 80% B (55– 60 min); return to 0% B (60 – 62 min); re-equilibrate
`at 0% B for 10 min before the next injection.
`For quantification of metabolites by radioactivity, the HPLC eluate was
`collected in 0.25-min intervals on Wallac ScintiPlate-96-well plates with a
`Gilson Model FC 204 fraction collector (Gilson, Middleton, WI). The
`plates were evaporated to dryness on a Savant Speed-Vac (Savant Instru-
`ments Inc., Holbrook, NY) and counted for 10 min/well with a PerkinElmer
`1450 MicroBeta Wallac TRILUX Liquid Scintillation and Luminescence
`Counter (PerkinElmer Life Sciences, Turku, Finland) to quantify radioac-
`tivity. Radioprofiles were prepared by plotting the net counts per minute
`values obtained from the MicroBeta versus time after injection using
`Microsoft Excel (Microsoft Corporation, Redmond, WA). The metabolites
`were quantified based on the percentage of total radioactivity in each peak
`relative to the entire radiochromatogram.
`Mass spectral analysis was performed on a Finnigan LCQ Deca XP ion trap
`mass spectrometer equipped with an electrospray ionization probe (Thermo
`Fisher Scientific). Analyses were performed in the positive ion mode. Samples
`were introduced into the mass-spectrometer after chromatographic separation,
`using the same HPLC method used for radioprofiling. High purity nitrogen was
`used as the sheath and the auxiliary gas with levels at 60 and 10 (relative flow
`rate), respectively. The capillary temperature was 350°C. The nitrogen gas
`
`Page 3 of 12
`
`

`
`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on September 1, 2015
`
`1348
`
`SU ET AL.
`
`flow rate, spray current, and voltages were adjusted as required to achieve
`maximum sensitivity or optimal fragmentation of drug-related components.
`Reference standards, available for saxagliptin, M2, D1, M13, and the S,R,S,S
`and S,S,S,R epimers of saxagliptin were used to confirm the retention time and
`mass-spectral fragmentation patterns of these analytes.
`Identification of Enzymes Involved in the Metabolism of Saxagliptin
`and in the Formation of M2. [14C]Saxagliptin (10 ␮M) was incubated at
`37°C with pooled HLM (1 mg/ml protein) and individually expressed human
`P450 enzymes (500 pmol/ml each, including CYP1A2, CYP2A6, CYP2B6,
`CYP2C8, CYP2C9, CYP2C18, CYP2C19, CYP2D6, CYP2E1, CYP3A4, and
`CYP3A5) in 100 mM phosphate buffer (pH 7.4), fortified with 1 mM NADPH
`to evaluate which enzymes were capable of metabolizing the compound and
`forming the major metabolite, M2. After the incubation period (30 min for
`expressed enzymes; 60 min for HLM), the reactions were terminated by adding
`1 to 2 volumes of ice-cold acetonitrile. The samples were then centrifuged, and
`the resulting supernatants were analyzed by LC-MS/MS and off-line radio-
`analysis. The analytical methodology was similar to that described for bio-
`transformation profiling and metabolite identification of in vivo samples, with
`the exception that drug-related components were separated on a YMC
`ODS-AQ S-3 120A column, maintained at 30°C, and the mobile phase gradient
`was modified, with a shorter run time (48 min).
`A correlation analysis between the formation of M2 and P450 activity was
`conducted by incubating saxagliptin, at concentrations of 1 and 10 ␮M, in
`singlicate with individual lots of HLM from 16 different donors. For each lot,
`the activities of CYP1A2, -2A6, -2B6, -2C8, -2C9, -2C19, -2D6, -2E1, -3A4/5,
`and -4A11 had been determined by the vendor using marker substrates specific
`for each enzyme (Technical Information for Reaction Phenotyping Kit version
`7, ref. 0510189; XenoTech, LLC). The reaction mixtures contained 0.25 mg/ml
`of HLM, 1 or 10 ␮M saxagliptin, 1 mM NADPH, and 0.1 M phosphate buffer
`with 2 mM MgCl2 (pH 7.4). Incubations (final volume, 0.5 ml) were conducted
`for 30 min at 37°C in a shaking water bath. Reactions were stopped with the
`addition of ice-cold acetonitrile. The concentration of M2 in each sample was
`then determined by analyzing an aliquot of the resulting supernatant by
`LC-MS/MS with multiple reaction monitoring as described below. Plots of M2
`versus marker substrate activity were prepared, and r values were calculated
`with Microsoft Excel (Office 2003; Microsoft Corporation).
`The metabolism of saxagliptin to M2 by specific P450 enzymes was also
`investigated with HLM in the presence of specific chemical or monoclonal
`antibody inhibitors of P450 enzymes. Chemical inhibitors included direct
`inhibitors, tranylcypromine (2 ␮M, CYP2A6), montelukast (3 ␮M, CYP2C8),
`sulfaphenazole (10 ␮M, CYP2C9), benzylnirvanol (1 ␮M, CYP2C19), quin-
`idine (1 ␮M, CYP2D6), ketoconazole (1 ␮M, CYP3A4/5) and time-dependent
`inhibitors, furafylline (10 ␮M, CYP1A2), orphenadrinie (50 ␮M, CYP2B6),
`diethyldithiocarbamate (50 ␮M, CYP2E1), troleandomycin (20 ␮M, CYP3A4/
`5), and 1-aminobenzotriazole (1000 ␮M, all P450s). Anti-P450 monoclonal
`antibodies included anti-CYP1A2, anti-CYP2B6, anti-CYP2C8, anti-
`CYP2C19, anti-CYP2D6, and anti-CYP3A4/5. A final concentration of 5 to 7
`␮l of antibody mixture was used per incubation. The antibody solutions were
`used as received; the concentration of each of the antibodies was not provided.
`Saxagliptin, at concentrations of 1 and 10 ␮M, was incubated in triplicate
`with pooled HLM (0.25 mg/ml), 1 to 2 mM NADPH, and 2 mM MgCl2 in 0.1
`M phosphate buffer (pH 7.4), in the presence or absence of chemical or
`anti-P450 monoclonal antibody inhibitors. The final volume of the reaction
`mixtures was 1 ml for the chemical inhibitor experiments and 0.25 ml for the
`anti-P450 antibody experiments. For incubations with direct chemical inhibi-
`tors, all ingredients except NADPH were added to the incubation tubes, and the
`samples were equilibrated at 37°C for 2 to 3 min before incubation. Then,
`NADPH was added to initiate the reactions. For incubations with time-
`dependent chemical inhibitors, all ingredients except saxagliptin were added to
`the incubation tubes. The samples were equilibrated at 37°C for 2 to 3 min, and
`then 1 mM NADPH was added to initiate a 15-min preincubation with the
`inhibitors. After the preincubation period, saxagliptin and an additional 1 mM
`NADPH were added to initiate the reactions. For incubations with anti-P450
`antibodies, the antibodies were preincubated with HLM in phosphate buffer on
`ice for 20 min, and then warmed at 37°C for 10 min. Saxagliptin and NADPH
`were added to the incubation mixtures to initiate the reactions. To establish the
`initial rate of metabolism of saxagliptin to M2 in HLM, incubations without
`chemical inhibitors or with antibodies against egg lysozyme (Hy-Hei-9) rather
`
`than anti-P450 antibodies were conducted. The rate of M2 formation in other
`incubations was normalized to the appropriate control incubation. Negative
`control incubations were carried out in the same manner, but they either lacked
`NADPH or contained heat-inactivated microsomes (boiled for 5 min).
`After the appropriate equilibrations and preincubation periods, HLM incu-
`bations were carried out for 30 min at 37°C. An equivalent volume of ice-cold
`acetonitrile was added to stop each reaction. Samples were vortex mixed and
`centrifuged to precipitate proteins. The concentration of M2 in each sample
`was then determined by analyzing an aliquot of the resulting supernatant by
`LC-MS/MS.
`Concentration-Dependent Metabolism of Saxagliptin to M2. The kinet-
`ics for the formation of M2 were determined in pooled HLM and expressed
`CYP3A4 and CYP3A5. Incubations (0.25 ml total volume, in triplicate)
`contained 1 mM NADPH, 2 mM MgCl2, 0.1 mM phosphate buffer (pH 7.4),
`saxagliptin, and HLM (0.25 mg protein/ml), or expressed CYP3A4 or
`CYP3A5 (10 pmol P450 enzyme/ml). Twelve concentrations of saxagliptin
`from 1 to 800 ␮M were evaluated. The HLM incubations were conducted at
`37°C. After the designated incubation period (30 min for HLM, 10 min for
`CYP3A4 and CYP3A5), reactions were quenched by adding an equal volume
`of ice-cold acetonitrile (0.25 ml). The quenched reaction mixtures were vor-
`texed to mix and centrifuged. The concentration of M2 in each sample was
`then determined by analyzing an aliquot of the resulting supernatant by
`LC-MS/MS.
`LC-MS/MS Method for Quantification of M2 in In Vitro Samples.
`Internal standard, 13C4,15N-5-hydroxy saxagliptin, was added to the quenched
`reaction mixtures from the in vitro incubations before LC-MS/MS analysis.
`The LC/MS system used for quantitation of M2 in in vitro samples consisted
`of two 10AD-VP pumps, a model SCL system controller and a degasser
`(Shimadzu Corporation), a LEAP HTC PAL autosampler equipped with a
`cooling stack maintained at 10°C (CTC Analytics, Carrboro, NC), and a
`Micromass Quattro Ultima triple quadrupole mass spectrometer (Waters).
`MassLynx software (version 4.0 or 4.1; Waters) was used to control the
`instrumentation and acquire data. Chromatographic separation of M2 from
`other mono-hydroxylated metabolites was achieved on an Agilent Zorbax
`SB-C8 column (4.6 ⫻ 75 mm, 3.5-␮m particle size) (Agilent, Wilmington,
`DE) maintained at ambient temperature. The mobile phase consisted of two
`solvents: mobile phase (A), 0.1% formic acid in water; and mobile phase (B),
`0.1% formic acid in acetonitrile. The mobile phase flow rate was 0.3 ml/min.
`The linear gradient program used for elution of the sample components was as
`follows: hold isocratic at 15% B (0 – 0.1 min); from 15 to 30% B (0.1–3 min);
`from 30 to 38% B (3– 4 min); from 38 to 40% B (4 –5 min); from 40 to 80%
`B (5–5.5 min); hold isocratic at 80% B (5.5– 6.5 min); return to 15% B (6.5–7
`min); re-equilibrate at 0% B for 5 min before the next injection.
`The Micromass Quattro Ultima mass spectrometer was operated in positive
`electrospray ionization mode. Ultrahigh purity nitrogen was used for the
`nebulizing and desolvation gases at flow rates of approximately 85 and 1000
`l/h, respectively. The capillary voltage was 3.5 kV, the cone voltage was 36V,
`and the collision energy was 45 eV. The desolvation temperature was 300°C,
`and the source temperature was 150°C. Detection of 5-hydroxysaxagliptin and
`its internal standard were achieved through MRM. The individual selected
`reaction monitoring transitions were 332 3 196 for M2 and 335 3 196 for the
`internal standard.
`Assessment of Potential of Saxagliptin and M2 to Inhibit P450 En-
`zymes. The potential for saxagliptin and M2 to inhibit P450 enzymes in a
`direct or time-dependent manner was assessed with HLM (n ⫽ 15 donors,
`mixed gender pool; CellzDirect, Durham, NC). IC50 values for nine enzymes
`were determined using probe substrates specific for each of the enzymes
`evaluated. The metabolic reactions monitored and probe substrate concentra-
`tions used were phenacetin O-deethylation (50 ␮M, CYP1A2), coumarin
`7-hydroxylation (1 ␮M, CYP2A6), bupropion hydroxylation (20 ␮M,
`CYP2B6), paclitaxel 6-hydroxylation (5 ␮M, CYP2C8), tolbutamide hydroxy-
`lation (140 ␮M, CYP2C9), S-mephenytoin 4⬘-hydroxylation (50 ␮M,
`CYP2C19), bufuralol 1⬘-hydroxylation (40 ␮M, CYP2D6), chlorzoxazone
`6-hydroxylation (50 ␮M, CYP2E1), midazolam 1⬘-hydroxylation and testos-
`terone 6␤-hydroxylation (5 and 50 ␮M, respectively, CYP3A). The final
`concentration of each probe substrate was near the experimentally determined
`Km value for the indicated enzyme.
`
`Page 4 of 12
`
`

`
`Downloaded from
`
`dmd.aspetjournals.org
`
` at ASPET Journals on September 1, 2015
`
`METABOLISM AND DISPOSITION OF SAXAGLIPTIN IN HUMANS
`
`1349
`
`To evaluate whether saxagliptin or M2 were competitive inhibitors of P450
`enzymes, saxagliptin (at concentrations of 0, 0.1, 1, 5, 20, and 50 ␮M), M2 (at
`concentrations of 0, 0.1, 1, 10, 50, and 200 ␮M), or prototypical P450
`inhibitors (positive controls) were mixed with HLM, and the probe substrates
`in 100 mM phosphate buffer (pH 7.4) in a total volume of approximately 0.5
`ml. After a 3-min equilibration at 37°C, 1 mM NADPH was added to initiate
`the reactions. The reactions were carried out using previously established
`conditions to ensure linearity with respect to protein concentration and incu-
`bation time. Incubations were stopped with addition of organic solvents. To
`assess the time-dependent inhibition, saxagliptin, M2, or positive control
`time-dependent inhibitors were preincubated for 15 min at 37°C with pooled
`human liver microsomes in the presence and absence of 1 mM
`NADPH. After the preincubation, P450-specific probe substrates were added
`to the incubation mixtures at the same concentrations used above. Metabolite
`formation in incubations with test compounds and control inhibitors was
`assessed with validated LC-MS/MS methods for each of the reaction products
`as described in Supplemental Table S1. Then,
`the percentage remaining
`activity was determined by comparison of probe substrate metabolism in
`incubations containing NADPH but without test compounds or control inhib-
`itors. If inhibition reached significant levels (i.e., the percentage remaining
`activity was ⬍50%), IC50 values were reported.
`Assessment of Potential of Saxagliptin and M2

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