throbber
Bioorganic & Medicinal Chemistry 23 (2015) 3831–3842
`
`Contents lists available at ScienceDirect
`
`Bioorganic & Medicinal Chemistry
`
`j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / b m c
`
`Effect of deuteration on metabolism and clearance of Nerispirdine
`(HP184) and AVE5638
`Joseph Schofield d,⇑
`, Volker Derdau b, Jens Atzrodt b, Patricia Zane c, Zuyu Guo c, Robert van Horn c,
`Valérie Czepczor a, Axelle Stoltz a, Magalie Pardon d
`a Sanofi R&D, DSAR-DD, 13, Quai Jules Guesde, 94400 Vitry-sur-Seine, France
`b Sanofi R&D, DSAR-DD, Industriepark Höchst, 65926 Frankfurt am Main, Germany
`c Sanofi R&D, DSAR-DD, 55 Corporate Drive, Bridgewater, NJ 08807, USA
`d Sanofi R&D, DSAR-DD, 1, Avenue Pierre Brossolette, 91385 Chilly-Mazarin, France
`
`a r t i c l e
`
`i n f o
`
`a b s t r a c t
`
`Replacing hydrogen with deuterium as a means of altering ADME properties of drug molecules has
`recently enjoyed a renaissance, such that at least two deuterated chemical entities are currently in clini-
`cal development. Although most research in this area aims to increase the metabolic stability, and hence
`half-life of the active species, experience has shown that prediction of the in vivo behaviour of deuterated
`molecules is difficult and depends on multiple factors including the complexity of the metabolic scheme,
`the enzymes involved and hence the mechanism of the rate-determining step in the biotransformation. In
`an effort to elucidate some of these factors we examined the metabolic behaviour of two molecules from
`the Sanofi portfolio in a range of in vitro and in vivo systems. Although some key metabolic reactions of
`the acetylcholine release stimulator HP184 4 were slowed in vitro and in vivo when deuterium was
`present at the sites of metabolism, this did not translate to an increase in overall metabolic stability.
`By contrast, the tryptase inhibitor AVE5638 13 was much more metabolically stable in vitro in its deuter-
`ated form than when unlabelled. These results indicate that it could be of value to concentrate efforts in
`this area to molecules which are metabolised by a major pathway that involves enzymes of the amine
`oxidase family or other low-capacity enzyme families.
`
`Ó 2015 Elsevier Ltd. All rights reserved.
`
`Article history:
`Received 13 March 2014
`Revised 23 March 2015
`Accepted 24 March 2015
`Available online 2 April 2015
`
`Keywords:
`Stable isotopes
`Deuterium
`Isotope effects
`Metabolism
`Cytochrome P450
`SSAO
`Monoamine oxidase
`H/D exchange
`
`1. Introduction
`
`D3CO
`
`H
`
`N
`
`O
`
`1: SD809
`⇑ Corresponding author.
`
`The theoretical principle underlying these attempts, that of the
`kinetic isotope effect (KIE), relies on the assumption that a meta-
`bolic reaction which breaks a C–H bond in the rate-determining
`step will be slowed down when hydrogen is substituted for deu-
`terium.6,7 Depending on the mode of action, this could translate
`to increased exposure to the parent, decreased formation of a toxic
`or reactive metabolite, or switching to a metabolic pathway which
`enhances formation of a species with increased activity.7 It has
`become increasingly clear that this simple rationale is difficult to
`put into practice in vivo: the vast majority of metabolic reactions
`responsible for clearance are mediated by one or several enzymes
`of the cytochrome P450 family which present such a wide range of
`reactivities, that simply slowing one pathway may not result in
`appreciable alteration of the ADME property being targeted.7
`Additional complicating factors have also been discussed by
`authors in the field.8
`In order to better understand the scope and limitations of
`applying the deuterium isotope effect to real cases in pharmaceu-
`tical development and to attempt to understand which structural
`and biochemical factors are important in translating the theory
`into practical examples, we have undertaken a selective study of
`
`Of the various chemical modifications used in medicinal
`chemistry to alter ADME properties of drug candidates, replace-
`ment of hydrogen by deuterium has recently received much atten-
`tion as a way to prolong half-lives of rapidly metabolised drug
`molecules.1,2 Although these attempts have a long history,3 no
`deuterated molecule has yet made it to market. The recent upsurge
`of activity in this area has, however, resulted in a candidate
`molecule, SD-809 1 reaching Phase III clinical trials,4 with further
`examples, including CTP-499, 2,5 also in the clinic.
`D3CO
`
`O
`
`N N
`
`N
`
`O
`
`N
`
`2: CTP-499
`
`OH
`
`D
`
`D
`
`D
`
`D D
`
`E-mail address: joseph.schofield@sanofi.com (J. Schofield).
`
`http://dx.doi.org/10.1016/j.bmc.2015.03.065
`0968-0896/Ó 2015 Elsevier Ltd. All rights reserved.
`
`Auspex Exhibit 2003
`Apotex v. Auspex
`IPR2021-01507
`Page 1
`
`

`

`3832
`
`J. Schofield et al. / Bioorg. Med. Chem. 23 (2015) 3831–3842
`
`In vitro experiments using 14C-labelled 4 incubated with human
`liver microsomes have shown that the N-dealkylation pathway is
`primarily mediated by CYP3A4, but that isoforms 1A2 and 2D6
`are implicated to a minor extent. Additionally, the methyl hydrox-
`ylation which begins the pathway to the acids is mediated by many
`different isoforms including 1A2, 2C9, 2C19 and 2D6, although it
`was not possible to quantify the individual contributions.13
`In order to study the effect of deuteration on the rate of meta-
`bolism, we targeted a derivative carrying deuterium atoms at each
`of these three major sites of metabolism, namely the fused phenyl
`ring, the propyl side chain and the indole 3-methyl group. 4a was
`thus synthesised and its metabolic behaviour studied in vitro using
`human hepatocytes and microsomes, and in vivo in rats and com-
`pared to that of 4, by following the rate of clearance of 4 versus 4a
`and the comparative kinetics of formation of metabolite 5 versus
`5a and 9 versus 9a.
`D
`
`O
`
`OH
`
`D
`
`N
`N H
`
`F
`
`D
`
`N
`
`D D
`
`D
`
`D
`
`D
`
`N
`N
`
`D
`
`H
`
`F
`
`D
`
`N
`
`D
`
`D D
`
`D
`
`D
`
`D
`
`D
`
`D
`
`D
`
`D
`
`D
`
`F
`
`D
`
`N
`N
`
`D
`
`N
`
`D D
`
`some compounds from the Sanofi portfolio and attempted to gauge
`the effect that site-specific deuteration has on their metabolism
`and clearance. Our recently published work on dronedarone, 3,9 a
`compound whose in vitro metabolism is dominated by CYP3A4-
`mediated N-debutylation10 showed that deuteration in a variety
`of metabolised positions had essentially no effect on the metabolic
`stability of the compound. This confirmed previous findings that
`deuteration can often have little effect on the kinetics of N-dealky-
`lation with this enzyme.11
`
`O
`
`N
`
`O
`
`O
`
`N
`N
`
`N
`
`F
`
`3: dronedarone
`
`4: HP184, nerispirdine
`
`HP184 (Nerispirdine 4) has been developed as an acetylcholine
`release stimulator for treatment of spinal cord injury.12 It is exten-
`sively metabolised in vitro, in animals, and in humans. 4 is metabo-
`lised principally by N-dealkylation to secondary amine HP183 5,
`and thence by oxidation to the carboxylic acid HP185 9 which is
`recovered in part as its glucuronide, 12. Carboxylic acid 8, with
`the propyl group intact, is also recovered as its glucuronide 11.13
`Furthermore, formation of both carboxylic acids occurs after prior
`hydroxylation of the methyl group. In addition to these major
`pathways, several other metabolic routes have been observed in
`the different experiments, the most important of which begins
`with aromatic hydroxylations to the phenols 10a and 10b (see
`Scheme 1).13
`
`4a: HP184-d14
`
`5a: HP183-d7
`
`9a: HP185-d4
`
`AVE5638,14 13 was developed as a tryptase inhibitor, and its
`metabolism is dominated by oxidative deamination to the car-
`boxylic acid 14 via aldehyde 15. In common with other compounds
`of this class15 and b tryptase itself, this transformation is mediated
`by semicarbazide sensitive amine oxidase (SSAO)16 (see Scheme 2).
`
`N
`
`H
`
`OH
`
`N
`
`N
`
`N
`
`4: HP184,
`nerispirdine
`
`F
`
`OH
`
`7
`
`N
`
`N
`
`F
`
`5: HP183
`
`N
`
`O
`
`OH
`
`F
`
`N
`
`N
`
`6
`
`OH
`
`O
`
`N
`
`N
`
`N
`
`F
`
`N
`
`N
`
`N
`
`F
`
`8
`
`N
`
`N
`
`N
`
`9: HP185
`
`O
`
`GluO
`
`N
`
`N
`
`F
`
`OH
`
`N
`
`10a, 10b
`
`Other minor
` metabolites
`
`N
`
`N
`
`F
`
`11
`
`N
`
`OGlu
`
`O
`
`N
`
`N
`
`12
`
`N
`
`Scheme 1. Major in vitro and in vivo metabolic pathways of HP184 4.13
`
`F
`
`F
`
`Auspex Exhibit 2003
`Apotex v. Auspex
`IPR2021-01507
`Page 2
`
`

`

`J. Schofield et al. / Bioorg. Med. Chem. 23 (2015) 3831–3842
`
`3833
`
`O
`
`N
`
`O
`
`NH2
`
`NH2
`
`D
`
`D
`
`13: AVE5638
`
`O
`
`N
`
`O
`
`13a: AVE5638-d2
`
`F
`
`F
`
`H
`
`O
`
`15
`
`OH
`
`O
`
`F
`
`F
`
`O
`
`N
`
`O
`
`O
`
`N
`
`O
`
`14
`
`Scheme 2. Oxidative metabolism of AVE5638, 13 to carboxylic acid, 14 and structure of deuterated analogue, 13a.
`
`To investigate the effect of deuteration on this single, non-CYP
`metabolic clearance pathway, we prepared a deuterated derivative,
`13a carrying D atoms at the site of oxidative deamination. Its rate
`of metabolism was compared to that of its unlabelled parent, 13 in
`human liver subcellular fractions (S9 and microsomes) and in
`human hepatocytes.
`
`2. Methods and results
`
`2.1. Chemistry
`
`2.1.1. HP184
`Recent work in our group17 and others active in the field18 has
`shown the versatility of hydrogen-deuterium exchange reactions
`to prepare deuterated derivatives, and in the case of 3-methylindole
`16, use of a mixed platinum–palladium catalyst system under
`microwave enhanced conditions,
`led to excellent deuterium
`
`incorporation levels at most positions in 16a. For the purpose of this
`study we aimed to exchange as many H atoms for D as possible, but
`the aromatic protons at the 4 and 7 positions were resistant to these
`conditions. For convenience, any positions exchanged to greater
`than 50% are shown as deuterated (Scheme 3). Using an adaptation
`of conditions described in the literature, the deuterated inter-
`mediate 16a was then N-aminated using methodology developed
`in our laboratories19 and the resultant N-amino compound 17
`reacted with 3-chloro-2-fluoropyridine 18 giving a good yield of
`5a. N-alkylation with commercially available n-1-bromopropane-
`d7 under microwave irradiation, followed by reverse-phase HPLC
`gave the target compound 4a in 28% overall yield, using previously
`described conditions.20
`
`2.1.2. AVE5638
`Surprisingly, the major challenge in the synthesis of 13a was
`the selective deuteration at the benzylic position. Attempts to
`
`D
`
`D
`
`DD
`
`N
`NH2
`
`D
`
`17
`
`D D
`
`(iii)
`
`Cl
`
`N
`
`F
`
`18
`
`(ii)
`
`[91]
`
`[99]
`
`NH
`
`16a
`
`[97]
`[97]
`
`[14]
`
`[53]
`
`(i)
`
`NH
`
`16
`
`DD
`
`D
`
`D
`
`HCl
`
`N
`
`F
`
`N
`
`NH
`
`D
`
`D D
`
`D
`D
`
`D D
`
`Br
`
`D D
`
`D
`
`(iv)
`
`DD
`
`D
`
`D
`
`FD
`
`D
`
`D
`D
`
`N
`
`N
`
`N
`
`HCl
`
`D
`
`D
`
`D
`
`D
`
`D
`
`D
`
`5a:HP183-d7
`Scheme 3. Synthesis of HP184-d14, 4a via deuterated metabolite HP183-d7 5a. Reagents (i) NaBD4/D2O, Pd/Pt–C, 92%; (ii) H2NOSO3H, NaH, DMF, 82%; (iii) 18 HCl; (iv) n-1-
`bromopropane-d7, Cs2CO3, KI, DMF then HCl, 56%.
`
`4a, HP184-d14
`
`Auspex Exhibit 2003
`Apotex v. Auspex
`IPR2021-01507
`Page 3
`
`

`

`O
`
`O
`
`NH
`
`19
`
`3834
`
`J. Schofield et al. / Bioorg. Med. Chem. 23 (2015) 3831–3842
`
`D D
`
`O
`
`O
`
`NH
`
`N
`
`(i)
`
`D D
`NH2
`
`(ii)
`
`N
`
`20
`
`N
`
`19a
`
`N
`
`(iii)
`
`N
`
`D D
`
`O
`
`O
`
`NH
`
`O
`
`Cl
`
`O
`
`Br
`
`22
`
`(iv)
`
`NH
`
`21
`
`O
`
`N
`
`O
`
`F
`
`13a: AVE5638-d2
`
`x CF3CO2H
`
`O
`
`O
`
`D
`D
`
`NH
`
`O
`
`O
`
`Br
`
`N
`
`23
`
`(v)
`
`(vi)
`
`Me3Si
`
`F
`
`24
`
`NH2
`
`D
`D
`
`Scheme 4. Synthesis of AVE5638-d2 13a. Reagents (i) LiAlD4/AlCl3; (ii) Boc2O, 30%, 2 steps; (iii) H2, Pt–C, HCl, EtOH, 77%; (iv) 22, Et3N, toluene, 66%; (v) 24, CuI, Pd(PPh3)2Cl2,
`Et3N, THF; (vi) MeSO3H, followed by preparative HPLC with TFA, 78%.
`
`follow the procedure of Greene et al.21 via formation of the ben-
`zylic dianion of the N-Boc benzylamine 19 followed by a D2O
`quench, yielded mixtures of products in which deuterium
`incorporation was incomplete. A second approach, in which nitrile
`20 was reduced using LiAlD4 under standard conditions22 also
`failed to give workable amounts of the required deuterated amine.
`24 or variations using
`Attempts with NaBD4/CoCl2,23 NaBD4/NiCl2
`
`LiAlD425 were likewise disappointing. Finally, we succeeded in
`selectively reducing 20 with lithium aluminium deuteride in pres-
`ence of aluminium chloride,26 followed immediately by Boc pro-
`tection, to give the deuterated carbamate 19a. 19a was converted
`to the target compound using a modification of published
`methods.27 Thus, partial reduction to the tetrahydropyridine, 21
`followed by reaction with substituted furoic acid chloride, 22 gave
`amide 23 in good yield. 23 was reacted under modified
`Sonogashira conditions with the acetylene, 24 to give 13a as its
`free base, which was converted to its trifluoroacetate after
`
`purification (see Scheme 4). The overall yield of the synthesis
`was 10%.
`
`2.2. Metabolism studies
`
`2.2.1. HP184 in vitro
`The in vitro metabolism of 4a was examined and compared to
`that of the unlabelled compound 4 following incubation with two
`different preparations of plated human hepatocytes at concentra-
`tions of 2 and 10 lM. Aliquots were withdrawn at various time
`points up to 24 h and, analysed by LC–MS/MS for parent
`compounds (4/4a), the N-depropylated metabolites (5/5a) and
`carboxylic acid metabolites (9/9a). There was no significant
`difference in the results observed between the two preparations.
`The time-course of the metabolism experiment with respect to the
`concentration of the parent compound and the two metabolites
`are shown in Graphs 1 and 2 for one hepatocyte preparation.
`
`Graph 1. Time versus concentration of HP184, 4 and HP184-d14, 4a incubated at (a) 2 lM and (b) 10 lM concentration in presence of plated human hepatocytes.
`
`Auspex Exhibit 2003
`Apotex v. Auspex
`IPR2021-01507
`Page 4
`
`

`

`J. Schofield et al. / Bioorg. Med. Chem. 23 (2015) 3831–3842
`
`3835
`
`Graph 2. Time versus concentration of (a) 5/5a and (b) 9/9a in incubation of 10 lM 4/4a in presence of plated human hepatocytes.
`
`Additionally, similar comparative experiments were con-
`ducted in human liver microsomes at a substrate concentration
`of 5 lM, at protein concentrations of 0.25, 0.5 and 1 mg of
`protein/mL. Aliquots were withdrawn at various time points up
`to 30 min and, analysed by LC–MS/MS for parent compounds
`(4/4a), the N-depropylated metabolites (5/5a). Note that the
`
`carboxylic acid metabolites (9/9a) were not formed in these
`incubations.
`The time-course of the metabolism experiments with respect to
`the concentration of the substrate and the metabolites of the three
`compounds at the various protein concentrations are shown in
`Graphs 3 and 4.
`
`Graph 3. Time versus concentration of 4 and 4a incubated at 5 lM concentration in presence of human liver microsomes at (a) 0.25, (b) 0.5 and (c) 1 mg protein/mL.
`
`Auspex Exhibit 2003
`Apotex v. Auspex
`IPR2021-01507
`Page 5
`
`

`

`3836
`
`J. Schofield et al. / Bioorg. Med. Chem. 23 (2015) 3831–3842
`
`Graph 4. Time versus concentration of 5 and 5a after incubation of 4 and 4a at 5 lM concentration in presence of human liver microsomes at (a) 0.25, (b) 0.5 and (c) 1 mg
`protein/mL.
`
`2.2.2. HP184 in vivo
`The in vivo metabolism of 4a was examined and compared to
`that of the unlabelled compound 4 following separate treatments
`of 5 male Sprague–Dawley rats orally at a dose of 20 mg/kg each
`with 4 or 4a. Aliquots of animal plasma were withdrawn at various
`
`time points up to 48 h and analysed by LC–MS/MS for parent
`compounds (4/4a), the N-depropylated metabolites (5/5a) and
`carboxylic acid metabolites (9/9a). The time-course of
`the
`metabolism experiment with respect
`to the concentration
`of the parent compounds and the metabolites are shown in
`Graphs 5 and 6.
`
`2.2.3. AVE5638
`The in vitro metabolism of 13a was examined and compared to
`that of 13, following incubation with human S9 fractions, human
`liver microsomes and human hepatocytes at both 1 and 10 lM
`concentrations. Graphs of drug concentration over time are shown
`below for the three in vitro systems (see Graphs 7–9).
`No difference in reaction rate was observed between incubates
`prepared in the presence or absence of an NADPH-generating
`system, confirming that CYP enzymes are not involved in the
`metabolism of 13/13a.
`
`3. Discussion
`
`3.1. HP184
`
`Graph 5. Time versus plasma concentration plot for 4 and 4a after oral administra-
`tion in Sprague–Dawley rats.
`
`In vivo, the overall clearance of 4 was practically unaffected
`(less than 10% difference in AUC) by the presence of deuterium
`atoms at the major sites of metabolism, as shown by the near
`overlap of the curves in Graph 5 and the following calculated
`
`Auspex Exhibit 2003
`Apotex v. Auspex
`IPR2021-01507
`Page 6
`
`

`

`J. Schofield et al. / Bioorg. Med. Chem. 23 (2015) 3831–3842
`
`3837
`
`Graph 6. Time versus plasma concentration plot for (a) 5/5a and (b) 9/9a after oral administration of 4 or 4a in Sprague–Dawley rats.
`
`Graph 7. Time versus concentration of 13 and 13a at (a) 1 lM and (b) 10 lM concentration in presence of human liver S9 fractions.
`
`Graph 8. Time versus concentration of 13 and 13a at (a) 1 lM and (b) 10 lM concentration in presence of human liver microsomes.
`
`pharmacokinetic parameters for the substrate compounds 4 and 4a
`(Table 1).
`Using plated hepatocytes the result was broadly similar.
`There was some saturation of metabolism for both 4 and 4a under
`the experimental condition, but at both concentrations the
`
`concentration/time curves for 4 and 4a practically overlap (Graph
`1a and b). At a non-saturating dose (2 lM), the calculated mean
`intrinsic clearance was slightly higher for 4a (38%) but the
`relevance of this difference for such a high clearance was consid-
`ered insignificant (Table 2).
`
`Auspex Exhibit 2003
`Apotex v. Auspex
`IPR2021-01507
`Page 7
`
`

`

`3838
`
`J. Schofield et al. / Bioorg. Med. Chem. 23 (2015) 3831–3842
`
`Graph 9. Time versus concentration of 13 and 13a at (a) 1 lM and (b) 10 lM concentration in presence of human hepatocytes.
`
`Table 1
`Pharmacokinetic parameters after oral administration of 4 or 4a in rats
`
`Compound
`
`4
`4a
`
`tmax (h)
`1.0 (1.0–1.0)
`0.5 (0.5–1.0)
`
`Cmax (ng/mL)
`822 ± 275
`971 ± 578
`
`AUClast (ng h/mL)
`4073 ± 1450
`3668 ± 1503
`
`AUC (ng h/mL)
`4176 ± 1527
`3801 ± 1626
`
`t1/2 (h)
`10.2 ± 1.91
`11.1 ± 1.94
`
`Table 2
`Clearance values after incubation of 4 or 4a in plated human hepatocytes
`
`Compound
`
`4
`4a
`
`Clearance
`At 10 lM (mL/h/106 cells)
`At 2 lM (mL/h/106 cells)
`0.415
`0.200
`0.572
`0.196
`
`The situation with human liver microsomes was more complex
`and depended on the concentration of the microsomes in the
`incubation medium—at 0.25 mg/mL the initial rate of metabolism
`of 4a was considerably less than that for 4, at 0.5 mg/mL 4a was
`metabolised more quickly than 4, while at 1 mg/mL metabolism
`rates for the two analogues were similar (Table 3).
`Despite these negative results concerning metabolic stability of
`the parent compound, it is interesting to compare the kinetics of
`formation for the two major metabolites, 5/5a and 9/9a. In all
`cases, the concentration of 5/5a was significantly reduced when
`the deuterated derivative, 4a was administered/incubated (see
`Graphs 2, 4 and 6). These results could indicate that either, (i) 5a
`was formed less readily, or (ii) it was metabolised more quickly
`in a second step. Since the acid metabolites 9/9a are formed, at
`least partly, from it (see Scheme 1) and it is also less abundantly
`formed, it is plausible to rule out (i) and to state that deuteration
`has indeed inhibited the N-dealkylation step.
`
`Table 3
`Clearance values after incubation of 4 or 4a with human liver microsomes at various
`protein concentrations
`
`Protein concentration (mg/mL)
`
`Initial mean metabolism rate
`(nmol/min/mg proteins)
`
`0.25
`0.5
`1
`
`With 4
`
`1.152
`0.346
`0.253
`
`With 4a
`
`0.420
`0.638
`0.217
`
`Our recent work on dronedarone 39 for which a similar cyto-
`chrome P450 mediated N-dealkylation is the major metabolism
`pathway, concurs with this conclusion. The overall clearance of a
`series of derivatives was unaffected by deuteration at the metabo-
`lised sites9 although individual metabolic steps were slowed
`down.13 In the present case, N-dealkylation was attenuated, but
`not blocked completely, an observation which is in broad agree-
`ment with previous observations which state that kinetic isotope
`effects in CYP-mediated N-dealkylations are generally quite
`small.11
`Although none of our experiments followed the formation of
`other, minor metabolites, it is reasonable to assume that the
`decrease in the amounts of 5a and 9a which formed with
`the deuterated substrate was compensated by an increase in the
`amount of these other metabolite(s). This phenomenon, known
`as ‘metabolic switching’ is well documented28 and although it
`can thwart attempts to increase drug half-life by deuteration, it
`can be very beneficial in cases where the formation of inactive or
`toxic metabolites is reduced or stopped.7,34
`The unusual behavior of the compound pair 4/4a during incuba-
`tion with human liver microsomes is not readily explainable, but
`may reflect a lower affinity of an enzyme’s active site for the
`deuterated form of the molecule. As protein concentration is
`increased, other metabolising enzymes, less sensitive to the pres-
`ence of deuterium are sufficiently abundant that this affinity differ-
`ence is no longer significant in the overall metabolic clearance. It
`has been documented that
`in vitro kinetics of cytochrome
`mediated reactions can be complex and that isotope effects can
`be used to study them.35 Since, in our case, the molecules 4/4a
`can present multiple metabolic sites to the enzymes, rigorous
`analysis becomes impossible.
`
`3.2. AVE5638
`
`From the Graphs 7–9 above, the rates of metabolism of 13 and
`13a were calculated, and are shown in Table 4. Reaction rates
`are significantly lower (60–80%) for 13a in both isolated enzyme
`
`Auspex Exhibit 2003
`Apotex v. Auspex
`IPR2021-01507
`Page 8
`
`

`

`J. Schofield et al. / Bioorg. Med. Chem. 23 (2015) 3831–3842
`
`3839
`
`Table 4
`Comparison of rates of metabolism of 13 and 13a in various matrices at 1 and 10 lM
`Reaction rate (nmol/min/mg for S9 and microsomes, nmol/min/1  106 cells for hepatocytes)
`At 10 lM
`At 1 lM
`
`Matrix
`
`Human S9
`Human liver microsomes
`Human hepatocytes
`
`With 13
`
`0.0219
`0.0201
`0.0143
`
`With 13a
`
`0.0081
`0.0048
`0.0143
`
`With 13
`
`0.0238
`0.0215
`0.016
`
`With 13a
`
`0.0043
`0.0045
`0.0081
`
`systems (microsomes and S9) while the effect is less pronounced in
`hepatocytes, with only a 50% rate reduction observed, and only at
`the higher concentration (Table 4). Unlike the case of 5/5a, no sat-
`uration of the metabolism was observed under our experimental
`conditions.
`The origin of this lower effect in hepatocytes is unclear, but
`could be related to a lower enzyme content in this system, or lower
`non-saturating concentrations of 13 and 13a within the hep-
`atocyte, and ultimately at the enzyme active site.
`Since the oxidative deamination step (Scheme 2) catalysed by
`SSAO is essentially the only metabolic pathway operating13 it
`appears that deuteration has a marked effect on the rate of reaction
`with this enzyme system.
`SSAO is a member of the amine oxidase family of enzymes
`which includes monoamine oxidases A and B and polyamine oxi-
`dase in addition to SSAO.29,30 The observation that site selective
`deuteration can have a marked effect on the metabolic stability
`of drug molecules metabolized by enzymes of this family is quite
`well known. For example, the deuterated analogue of the MAO-
`inhibitor phenelzine 25 had increased biological activity in rats
`compared to the non-deuterated parent.31 It was further shown
`that 25 is catabolized by the same enzyme. Similarly,
`it has
`recently been proposed32 that administration of deuterated lysine
`26 could potentially be beneficial
`in pathologies where lysyl
`oxidase (LOX), a similar enzyme to SSAO, is implicated. The basis
`of this hypothesis is that the high observed KIE for oxidative deam-
`ination of 26, a key step involved in LOX biosynthesis in vivo, will
`modulate the amount of LOX present in the organism, reducing its
`nefarious effects.
`
`D
`
`D
`
`D
`
`D
`
`NHNH2
`
`D
`
`NH2
`
`D
`
`O
`
`OH
`
`NH2
`
`25: Phenelzine-d4
`
`26: Lysine-d2
`
`Thus, our observation of a marked in vitro effect of deuteration
`on the rate of oxidative deamination of 13a is in accordance with
`these observations for the amine oxidase family as a whole. This
`has been explained by a common mechanism33 in which H
`abstraction by the enzyme of an amine’s a hydrogen atom is the
`rate limiting step. Unfortunately, a shift of the project focus did
`not permit us to investigate the effect of deuteration of 13 on
`metabolic clearance in vivo.
`
`4. Conclusions
`
`The work presented here, and other results published by us and
`other workers in the field11 tend to conclude that, compounds for
`which CYP450 mediated N-dealkylation is a major metabolic route
`are unlikely to exhibit significantly slower clearance when deuter-
`ated at the site of metabolism. There is generally only a small
`deuterium isotope effect for this step. Several of the CYP isoforms,
`especially CYP3A4,36 are versatile catalysts which can metabolise a
`
`very wide range of compound types—probably due to the presence
`of multiple binding sites with varying substrate affinities. In the
`HP184, 4 system presented above,
`the correlation between
`in vitro systems, especially hepatocytes, and animal experiments
`was good and showed the value of conducting such screening prior
`to in vivo experimentation, a conclusion which is strengthened by
`other examples reported in the literature.11 Building on this idea,
`the in vitro results for 13/13a suggest that there would be a chance
`of prolonging the parent half-life in vivo. Overall, it appears that
`efforts to increase metabolic stability of drugs by deuteration
`would best be targeted on molecules with single, and/or lower-
`capacity metabolic pathways, although this correlation has yet to
`be proved.
`
`5. Experimental
`
`5.1. Synthesis
`
`5.1.1. General
`1H (300, 500 MHz) and 13C (75, 125 MHz) NMR spectra were
`obtained on Bruker Advance spectrometers in the solvents indi-
`cated. Column chromatography was performed using Merck silica
`gel 60 (particle size: 63–200 lm). Product purity was determined
`by a LC–MS system with a Symmetry Shield RP18 column,
`3.9  150 mm with gradient program; conditions: mobile phase:
`A: water (900 mL), CH3CN (100 mL), TFA (1 mL) mobile phase B:
`water (100 mL), CH3CN (900 mL), TFA (0.75 mL), flow 0.6 mL/min,
`detection UV 254 nm and UV 210 nm. Commercially available
`chemicals and solvents were used as received. Deuterated water
`(D2O) was purchased from Aldrich. Dry Pd/C (10% on charcoal)
`was purchased from Degussa. Depending on the batch or supplier
`of the catalyst significant changes of catalyst reactivity were
`observed. Dry Pt/C was purchased from Hereaus. The percentage
`of deuteration at each CH-position was determined by using 1H
`NMR with tartaric acid as internal standard.
`
`5.1.2. HP184
`5.1.2.1. Methylindole-d7 (16a). An argon-filled pressure tube was
`charged with 3-methylindole (16; 500 mg, 3.8 mmol), 25 mg 10%
`Pd/C catalyst (25 mg), 50 mg 5% Pt/C catalyst (50 mg), sodium
`borodeuteride (23 mg, 0.57 mmol), and D2O (12 mL). The mixture
`was stirred for approximately 30 s and the tube sealed (note: the
`reaction vessel was not closed until effervescence had stopped)
`and heated to 140 °C for 2 h. The mixture was cooled to room
`temperature. Eight similar reaction runs were combined and
`EtOAc (50 mL) was added. The catalyst was separated by filtration,
`the filter cake washed with EtOAc (20 mL) and the phases sepa-
`rated. The aqueous phase was extracted with EtOAc (2  50 mL)
`and the combined organic layers dried over Na2SO4. The solvent
`was removed in vacuo to give 16a as a brown solid (3.89 g, 92%).
`MS (ESI+) m/z (%): 137 (8) [d5+H]+, 138 (39) [d6+H]+, 139 (43)
`[d7+H]+, 140 (9) [d8+H]+. HPLC purity (UV, 254 nm) 97.2%.
`
`5.1.2.2. 3-Methyl-indol-1-ylamine-d7 (17). Under argon, sodium
`hydride (60% dispersion in mineral oil, 4.83 g, 121 mmol) was
`
`Auspex Exhibit 2003
`Apotex v. Auspex
`IPR2021-01507
`Page 9
`
`

`

`3840
`suspended in DMF (60 mL) and cooled to 5 °C. 16a (1.20 g,
`8.69 mmol) was added in small portions keeping the internal
`temperature below 5 °C. After stirring for 1 h, hydroxylamine-O-
`sulfonic acid (4.55 g, 40.2 mmol) was added in portions and the
`temperature was again kept below 5 °C (attention:
`internal
`exothermic reaction !). After complete addition, the cooling bath
`was removed and the reaction mixture was stirred at room
`temperature for 2 h (reaction followed by LC–MS). The reaction
`mixture was poured into ice water (80 mL) and after 15 min
`extracted with EtOAc (3  80 mL). The combined organic layers
`were dried over Na2SO4 and the solvent was removed in vacuo to
`give a brown oil which was purified by chromatography (SiO2, hep-
`tane/EtOAc 5:1 v/v) to give 17 as a colourless solid (1.10 g, 82%).
`MS (ESI+) m/z (%): 152 (6) [d4+H]+, 153 (13) [d5+H]+, 154 (34)
`[d6+H]+, 155 (39) [d7+H]+, 156 (8) [d8+H]+. HPLC purity (UV,
`254 nm) 95.3%.
`
`5.1.2.3. (3-Fluoro-pyridin-4-yl)-(3-methyl-indol-1-yl)-amine-d7
`hydrochloride (5a). 4-Chloro-3-fluoro-pyridine (18) (1.11 g, 7.85
`mmol) was dissolved in N-methylpyrrolidone (NMP) (1.5 mL) and
`methyl tert-butyl ether (MTBE) (0.5 mL) and heated to 80 °C.
`Concentrated HCl (0.93 mL) was added slowly and then a solution
`of 17 (1.10 g, 7.14 mmol) in NMP (3.5 mL) was added dropwise.
`The reaction mixture was kept at 90 °C for 4 h (reaction followed
`by LC–MS). After complete conversion, MTBE (5 mL) was added
`and the reaction mixture allowed to cool to room temperature.
`The precipitated product was isolated by centrifugation, washed
`twice with MeOH/MTBE (1:1), filtered and the filter cake washed
`twice with MeOH/Et2O (1:1) to give, after drying, 5a as a slightly
`orange solid (1.36 g, 67%). 1H NMR (500 MHz, DMSO-d6) d 11.81
`(br s, 1H), 8.92 (d, 5.6 Hz, 1H), 8.22 (d, 5.6 Hz, 1H), 7.64 (s,
`0.47H), 7.48 (s, 0.01H), 7.29 (s, 0.86H), 7.22 (s, 0.03H), 7.19 (s,
`0.03H), 6.22 (dd, 5.6/5.6 Hz, 1H), 2.28 (s, 0.09H). 13C NMR
`(125 MHz, DMSO-d6) d 149.7, 147.7, 145.7, 140.7, 135.5, 129.6,
`129.4, 127.4, 125.8, 119.8, 111.4, 107.9. MS (ESI+) m/z (%): 247
`(0.5) [d4+H]+, 248 (8) [d5+H]+, 249 (39) [d6+H]+, 250 (42) [d7+H]+,
`251(9) [d8+H]+, 252 (1). HPLC purity (UV, 254 nm) 98.7%.
`
`(3-Fluoro-pyridin-4-yl)-(3-methyl-indol-1-yl)-propyl-
`5.1.2.4.
`amine-d14, HP184-d14 (4a). 5a (150 mg, 0.53 mmol), caesium
`carbonate (359 mg, 1.10 mmol), potassium iodide (8.80 mg,
`0.05 mmol) and DMF (12 mL) were charged to a 20 mL microwave
`tube. After addition of propyl bromide-d7 (75 lL, 0.77 mmol) the
`flask was sealed and heated in the microwave at 100 °C for 2 h
`(reaction followed by LC–MS). The reaction was repeated three
`times and the combined reaction mixtures were poured into a
`mixture of 1:1 water/EtOAc (100 mL). The layers were separated
`and the aqueous phase extracted with EtOAc (2  50 mL). The
`combined organic layers were dried over Na2SO4 and the solvent
`removed in vacuo to give a brown oil which was purified by
`prep. HPLC (Synergy 10 lm C18 column (250  15 mm)) eluent
`CH3CN/water 10:90 (v/v) for 4 min, then in 8 min to 80:20 (v/v),
`8 min at 80/20 (v/v) and in 1 min back to 10:90 (v/v); flow:
`15 mL/min; UV detection at 220 and 250 nm. The combined
`product fractions were evaporated to dryness in vacuo and after
`addition of EtOAc (2 mL) the compound crystallized to give 4a as
`a beige solid (298 mg, 56%). 1H NMR (500 MHz, DMSO-d6): d 8.90
`(d, 7.7 Hz, 1H), 8.23 (d, 6.7 Hz, 1H), 7.64 (s, 0.47H), 7.48 (s,
`0.01H), 7.40 (s, 0.86H), 7.22 (s, 0.03H), 7.19 (s, 0.03H), 6.31 (dd,
`7.7/6.7 Hz, 1H), 4.05 (s, 0.03H), 3.88 (s, 0.03H), 2.28 (s, 0.09H),
`0.86 (s, 0.04H). 13C NMR (125 MHz, DMSO-d6): d 148.5, 146.6,
`144.6, 139.1, 133.8, 131.3, 131.0, 126.5, 124.2, 119.4, 111.2,
`110.7, 56.5 (br), 20.1 (br), 9.6 (br). MS (ESI+) m/z (%): 296 (12)
`[d12+H]+, 297 (45) [d13+H]+, 298 (35) [d14+H]+, 299 (8) [d15+H]+.
`HPLC purity (UV, 254 nm) 99.9%.
`
`J. Schofield et al. / Bioorg. Med. Chem. 23 (2015) 3831–3842
`
`5.1.3. AVE5638
`5.1.3.1. N-Boc-3-(4-pyridyl)benzylamine-d2 (19a). A mixture of
`lithium aluminum deuteride (1.14 g, 30 mmol) and aluminum
`chloride (4.0 g, 30 mmol) was stirred in Et2O (40 mL) at room
`temperature for 30 min. The mixture was
`treated with
`3-(4-pyridyl)benzonitrile (20) (2.06 g, 10 mmol) at rt and the reac-
`tion mixture stirred for 2 h. After the reaction was complete, as
`shown by LC–MS, 0.1 N NaOH aqueous solution (80 mL) was added
`to the reaction mixture. This mixture was extracted with CH2Cl2
`(3  50 mL). The combined organic layers were dried over
`Na2SO4, filtered, and concentrated in vacuo to give the crude free
`amine, which was used without further purification in the next
`reaction step. MS (ESI+) m/z (%): 187 (71) [d2+H]+, 188 (24), 189
`(3). HPLC purity (UV, 254 nm) 85.6%.
`The crude product was dissolved in MeOH (30 mL) and NaHCO3
`(941 mg, 11.2 mmol) was added. Then a solution of Boc2O (2.45 g,
`11.1 mmol) in MeOH (10 mL) was added dropwise and the reaction
`mixture stirred at rt for 4 h (LC–MS control). Finally the reaction
`mixture was filtered and evaporated to dryness. The crude product
`was purified by chromatography (SiO2, heptane/EtOAc/triethy-
`lamine 50:50:1 v/v/v) to give 19a as a colourless solid (858 mg,
`30%). 1H NMR (300 MHz, DMSO-d6) d 8.62–8.60 (m, 2H), 7.70–
`7.68 (m, 3H), 7.49–7.32 (m, 3H), 4.02 (s, 0.03H), 1.35 (s, 9H). 13C
`NMR (100 MHz, DMSO-d6) d 155.8, 150.4, 147.0, 141.0, 137.0,
`129.1, 127.8, 125

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