throbber
J. Med. Chem. 2007, 50, 5339-5356
`
`5339
`
`Discovery of 1-(4-Methoxyphenyl)-7-oxo-6-(4-(2-oxopiperidin-1-yl)phenyl)-4,5,6,7-tetrahydro-
`1H-pyrazolo[3,4-c]pyridine-3-carboxamide (Apixaban, BMS-562247), a Highly Potent, Selective,
`Efficacious, and Orally Bioavailable Inhibitor of Blood Coagulation Factor Xa
`
`Donald J. P. Pinto,* Michael J. Orwat, Stephanie Koch, Karen A. Rossi, Richard S. Alexander, Angela Smallwood,
`Pancras C. Wong, Alan R. Rendina, Joseph M. Luettgen, Robert M. Knabb, Kan He, Baomin Xin, Ruth R. Wexler, and
`Patrick Y. S. Lam
`DiscoVery Chemistry, Research and DeVelopment, Bristol-Myers Squibb Company, 31 Pennington-Rocky Hill Road,
`Pennington, New Jersey 08534
`
`ReceiVed May 1, 2007
`
`Efforts to identify a suitable follow-on compound to razaxaban (compound 4) focused on modification of
`the carboxamido linker to eliminate potential in vivo hydrolysis to a primary aniline. Cyclization of the
`carboxamido linker to the novel bicyclic tetrahydropyrazolopyridinone scaffold retained the potent fXa binding
`activity. Exceptional potency of the series prompted an investigation of the neutral P1 moieties that resulted
`in the identification of the p-methoxyphenyl P1, which retained factor Xa binding affinity and good oral
`bioavailability. Further optimization of the C-3 pyrazole position and replacement of the terminal P4 ring
`with a neutral heterocycle culminated in the discovery of 1-(4-methoxyphenyl)-7-oxo-6-(4-(2-oxopiperidin-
`1-yl)phenyl)-4,5,6,7-tetrahydro-1H-pyrazolo[3,4-c]pyridine-3-carboxamide (apixaban, compound 40). Com-
`pound 40 exhibits a high degree of fXa potency, selectivity, and efficacy and has an improved pharmacokinetic
`profile relative to 4.
`
`Introduction
`Thrombotic diseases remain the leading cause of death in
`developed countries despite the availability of anticoagulants
`such as warfarin,1a-c heparin and low molecular weight hep-
`arins,2,3 and antiplatelet agents such as aspirin and clopidogrel.
`The oral anticoagulant warfarin inhibits the post-translational
`maturation of coagulation factors VII, IX, and X and prothrom-
`bin and has proven effective in both venous and arterial
`thrombosis. However, warfarin’s usage is limited because of
`its narrow therapeutic index, slow onset of therapeutic effect,
`numerous dietary and drug interactions, and a need for monitor-
`ing and dose adjustment.4a,b This not withstanding, warfarin
`remains the standard orally administered anticoagulant available
`in the United States. Patients on warfarin therapy require regular
`monitoring in part because of its narrow therapeutic index and
`interactions with food and other drugs. Injectable agents that
`are also widely used include low molecular weight heparins
`and the synthetic pentasaccharide fondaparinux.5 Thus, discov-
`ering and developing safe and efficacious oral anticoagulants
`for the prevention and treatment of a wider range of thrombotic
`diseases has become increasingly important.
`A key strategy for the discovery and development of new
`anticoagulants has been the targeting of specific enzymes within
`the blood coagulation cascade. One approach is to inhibit
`thrombin generation by targeting the inhibition of coagulation
`factor Xa (fXa).5,6a-h Factor Xa, a trypsin-like serine protease,
`is crucial to the conversion of prothrombin to thrombin, the
`final enzyme in the coagulation cascade that is responsible for
`fibrin clot formation. Preclinical animal models have suggested
`that inhibiting fXa has the potential for providing excellent
`antithrombotic efficacy with minimal bleeding risk when
`compared to direct thrombin inhibitors.6a-g Recent disclosures
`from clinical studies with direct fXa inhibitors such as compound
`
`* To whom correspondence should be addressed. Phone:
`5295. Fax (609) 818-3460. E-mail: donald.pinto@bms.com.
`
`(609) 818-
`
`Figure 1. Schematic of important pyrazole fXa compounds.
`
`4,7a-c rivaroxaban (BAY 59-7939),8a,b 1H-indole-5-carboxylic
`acid {(R)-2-[4-(4-methylpiperazin-1-yl)-piperidin-1-yl]-2-oxo-
`1-phenylethyl}amide (LY-517717)9 and the indirect parenteral
`fXa inhibitor fondaparinux5 have confirmed the preclinical
`findings.10
`The discovery of the pyrazole scaffold, illustrated by SN429
`(compound 1, Figure 1, fXa Ki ) 13 pM, trypsin Ki ) 16 nM),11
`was a significant milestone in our search for molecules targeting
`coagulation fXa and proved to be crucial in the evolution of
`orally bioavailable fXa inhibitors such as DPC423 (compound
`2, fXa Ki ) 0.15 nM,
`trypsin Ki ) 60 nM),11 DPC602
`(compound 3, fXa Ki ) 0.87 nM, trypsin Ki ) 1500 nM),12a
`and razaxaban (compound 4, fXa Ki ) 0.15 nM, trypsin Ki >
`
`10.1021/jm070245n CCC: $37.00 © 2007 American Chemical Society
`Published on Web 10/03/2007
`
`MYLAN EXHIBIT 1005
`
`

`

`5340 Journal of Medicinal Chemistry, 2007, Vol. 50, No. 22
`
`Pinto et al.
`
`Scheme 1. Syntheses of C-3-carboxypyrazolo-pyridinone Analoguesa
`
`a (a) NaNO2, HCl, 0 (cid:176) C, NaOAc, EtOH, ethyl 2-chloroacetoacetate; (b) Et3N/toluene, reflux; (c) 3 N HCl or TFA, DCM; (d) 2-formylphenylboronic acid,
`(Ph3P)4Pd, toluene/EtOH or DME/water (4:1), Na2CO3 (2 N), reflux; (e) 3-(R)-OH-pyrrolidine (2 equiv), NaCNBH3, ZnCl2 (0.5 N, in THF), MeOH; (f)
`LiOH or NaOH (1 N), MeOH/water; (g) NH4OH, EtOH, 80 (cid:176) C; (h) amine, NaCNBH3, ZnCl2 (0.5 N, in THF), MeOH; (i) oxalyl chloride, DMF; (j) MeNH2
`or NHMe2, trimethylaluminum (1 N), DCM, 0 (cid:176) C to room temp; (k) ammonia/MeOH, 50 (cid:176) C; (l) DMAP, TFAA; (m) ether, 20% aq. HCl; (n)
`p-methoxyphenylhydrazine, MeOH reflux.
`
`5000 nM).7a Compounds 2 and 4 were advanced to clinical trials.
`Subsequently, compound 4 was further advanced to a phase II
`trial for the prevention of venous thromboembolism (VTE) after
`knee replacement surgery and was shown to be highly effica-
`cious when compared to enoxaparin.7c
`Consistent with our strategy of developing and advancing key
`follow-on candidates, our focus was directed toward the
`identification of novel entities that would be significantly
`differentiated from previous candidates in terms of improving
`on potential
`liabilities of earlier compounds. A common
`structural feature that is present with compound 4 and its
`predecessor candidates was the presence of the 5-carboxamido
`linker that connects the pyrazole scaffold to the P4 moiety. In
`the advancement of potential candidates for preclinical evalu-
`ations, it was necessary to determine the susceptibility of the
`amide linker to metabolic cleavage because this could potentially
`liberate a aniline fragment. Fortunately, for compound 4 and
`its predecessor clinical compound 2 the amide linker was stable
`to metabolic hydrolysis; however, this was not the case with
`our preclinical compound 3, which liberated the biarylamino
`group at a higher pH. In the bacterial reverse mutation (AMES)13
`assay, the aniline moiety of 3 tested positive, which was further
`
`confirmed in follow-up assays for mutagenicity.14 Therefore,
`as part of our optimization strategy, we sought to modify the
`carboxamido portion of the molecule to obviate the need for
`mutagenicity studies on potential aniline degradants. Toward
`this end, we recently disclosed several series of bicyclic pyrazole
`scaffolds15a-c,16a in which the carboxamido linker was cyclized
`into the pyrazole ring, some of which showed similar or better
`fXa potency compared with the previously disclosed monocyclic
`pyrazole analogues.7a,b,11 The optimization strategy with the
`bicyclic pyrazole scaffold led to the identification of BMS-
`740808 (compound 5, fXa Ki ) 0.03 nM, trypsin Ki > 5000
`nM, Figure 1),15a which was advanced to preclinical safety
`evaluation. Importantly, the discovery of the potent bicyclic
`scaffold set the stage for exploratory work employing additional
`P1 moieties,7c,16 many of which demonstrated subnanomolar fXa
`binding affinities and moderate to high clearance (Cl) and
`volume of distribution (Vdss) in dogs. However, the lack of
`adequate differentiation from compound 4 in terms of improve-
`ment in the overall pharmacokinetic profile made them less
`attractive for further development. In this paper, we report an
`optimization strategy that resulted in the identification of
`compound 40, a structurally novel and neutral bicyclic pyrazole
`
`

`

`Apixaban as Inhibitor
`
`Journal of Medicinal Chemistry, 2007, Vol. 50, No. 22 5341
`
`fXa inhibitor (currently in phase III trials) with a superior
`pharmacokinetic profile (low clearance and volume of distribu-
`tion) compared to compound 4.
`
`Chemistry
`The synthesis of the C-3 trifluoromethylpyrazole analogue 6
`was accomplished via the cyclization methodology previously
`described.15,16a Scheme 1 illustrates the general synthetic
`methodology utilized to prepare diversified pyrazole C-3 ana-
`logues. Commercially available 4-methoxyaniline was diazotized
`(NaNO2, concentrated HCl, 0 (cid:176) C) and condensed in situ with
`either 1-chloro-1-(methylsulfonyl)propan-2-one or ethyl 2-chlo-
`roacetoacetate in the presence of sodium acetate17 to provide
`the requisite p-methoxyphenylchlorohydrazones 7a in 57% yield
`and 7b in 90% yield. Treatment of the chlorohydrazones 7a
`and 7b with compound 815 using excess triethylamine afforded
`the requisite [3 + 2] cycloadducts which, when treated with
`TFA in dichloromethane, led to compounds 9a (80% yield) and
`9b (71% yield) respectively. Suzuki coupling of 9a,b with
`2-formylbenzeneboronic acid as illustrated for compound 515
`afforded the biaryl o-carboxaldehyde intermediates 10a in 71%
`yield and 10b in 80% yield, respectively. Subsequent reductive
`amination with 3-(R)-hydroxypyrrolidine15,16a provided the
`bicyclic pyrazole compounds 11a (45% yield) and 11b (69%
`yield). Hydrolysis (LiOH in THF and water) of the ester group
`in 11b gave the desired C-3 carboxylic acid compound 12 in
`51% yield. Compounds 13a-h were prepared in a two-step
`sequence by the reductive amination of 10b followed by
`carboxamide formation as described above in yields that ranged
`between 80% and 90%. Alternatively, treatment of compound
`11b with ammonium hydroxide in ethanol at 80 (cid:176) C for 4 days
`provided the carboxamidopyrazole analogue 13f in 45% yield.
`Hydrolysis (NaOH (1 N) in THF/water) of 9b gave carboxylic
`acid intermediate 14a (90% yield). Treatment of the pyrazole
`ester 9b under the Weinreb amide conditions (methylamine or
`dimethylamine, trimethylaluminum (1 N) in DCM at 0 (cid:176) C to
`room temperature)18 provided 14b (92% yield) and 14c (88%
`yield). The compounds were subsequently converted to 15a,b
`in 55% and 46% yield, respectively, following the Suzuki and
`reductive amination procedures. To prepare the cyanopyrazole
`compound 18, compound 10b was first converted to the
`carboxamidobiarylcarboxaldehyde 16 in 66% yield by treatment
`with ammonia in methanol at 80 (cid:176) C. Dehydration (oxalyl
`chloride in DMF) to 17 (42% yield) followed by reductive
`amination gave the desired cyano compound 18 (27% yield).
`The aminopyrazole compounds 20-25 were accessed ac-
`cording to the methodologies outlined in Scheme 2. Curtius
`rearrangement19 of the pyrazolecarboxylic intermediate 14a
`provided the Boc protected aminopyrazole intermediate 19a in
`22% yield. Biarylcarboxaldehyde formation (20, 93% yield)
`followed by reductive amination with 3-(R)-hydroxypyrrolidine
`afforded compound 21 in 77% yield. Treatment of compound
`21 with TFA provided compound 22 in 10% yield. Alternatively,
`compound 21 was alkylated with sodium hydride and iodo-
`methane in anhydrous DMF to afford 23 in 42% yield. Treat-
`ment of 23 with TFA in dichloromethane afforded compound
`24 in 99% yield. To prepare compound 25, pyrazole derivative
`19a was deprotected with TFA and reductively aminated with
`formaldehyde (37%) and sodium cyanoborohydride in the
`presence of zinc chloride (0.5 M in THF) to afford the
`dimethylaminopyrazole compound 19b in 51% yield. Biaryl-
`carboxaldehyde formation followed by reductive amination with
`3-(R)-hydroxypyrrolidine afforded compound 25 in 34% yield.
`Tetrazolyl compounds 27 and 28 were prepared according
`to Scheme 3. Treatment of 14b with lutidine and triflic
`
`Scheme 2. Syntheses of 3-Aminopyrazole Analoguesa
`
`a (a) Oxalylchloride, DCM, catalyst DMF; (b) NaN3, water, acetone 0
`(cid:176) C; (c) toluene, 80 (cid:176) C, tBuOH; (d) TFA, DCM; (e) formaldehyde (37%,
`excess), ZnCl2 (0.5 M/THF), NaBH3CN, MeOH; (f) 2-formylphenylboronic
`acid, (Ph3P)4Pd, Na2CO3 (2 N), 4:1 toluene/EtOH, reflux; (g) 3-(R)-OH-
`pyrrolidine, NaCNBH3, ZnCl2 (0.5 N, in THF), MeOH; (h) NaH, DMF,
`MeI, room temp.
`
`anhydride generated the iminotriflate, which was directly treated
`with excess sodium azide to give the tetrazole derivative 26
`in 48% yield. Suzuki coupling with 2-formylboronic acid
`and reductive amination with 3-(R)-hydroxypyrrolidine led to
`27 in 47% yield. The tetrazole compound 28 was prepared
`in 52% yield by heating compound 18 with sodium azide in
`DMF.
`Heteroarylalkyl compounds 33a-e were synthesized accord-
`ing to procedures outlined in Scheme 4. Borane reduction of
`carboxylic acid20 intermediate 14a afforded the alcohol inter-
`mediate 29 in 89% yield, which was subsequently converted to
`the bromomethyl intermediate 30 by treatment with phosphorus
`tribromide (PBr3) in dichloromethane in 94% yield. Displace-
`ment of the crude bromide 30 with 1,2,3-triazole, 1,2,4-triazole,
`or 1H-tetrazole afforded mixtures of regioisomeric triazole-
`methyl or tetrazolylmethyl compounds 31a-e, which were
`subsequently converted to biarylcarboxaldehyde compounds
`32a-e and later to the desired compounds 33a-e.
`Variably substituted P4 anilino compounds 34, 35, and 36a-e
`were prepared according to the methods outlined in Scheme 5.
`Aryl amination of compound 9c according to the Buchwald
`amination methodology21 afforded compound 34 in 97% yield.
`Acetylation of 34 with acetic anhydride and triethylamine gave
`the acetyl derivative 35 in 97% yield. Alternatively, aniline 34
`was converted to the Boc protected derivative 36a by treatment
`with Boc anhydride (neat) at 80 (cid:176) C in 84% yield. Alkylation
`with iodomethane provided 36b in quantitative yield (100%).
`Removal of the Boc protecting group afforded 36c was
`acetylated to afford compound 36d. Alkylation of 36c with
`idodomethane and potassium carbonate provided compound 36e
`in 47% yield.
`
`

`

`5342 Journal of Medicinal Chemistry, 2007, Vol. 50, No. 22
`
`Scheme 3. Syntheses of C-3-Cyano, 3-Tetrazole Analoguesa
`
`Scheme 4. Syntheses of Substituted C-3 Heteroalkyl
`Analoguesa
`
`Pinto et al.
`
`a (a) Triflic anhydride, lutidine, NaN3, DMF; (b) 2-formylphenylboronic
`acid, (Ph3P)4Pd, Na2CO3 (2 N), 4:1 toluene/EtOH, Na2CO3 (2 N), reflux;
`(c) 3-(R)-OH-pyrrolidine (2 equiv), NaCNBH3, ZnCl2 (0.5 N, in THF),
`MeOH; (d) NaN3, DMF, heat.
`
`Analogues in which the P4 moiety is either the phenylpip-
`eridinyl or the corresponding phenyllactam groups were accessed
`according to the methods outlined in Scheme 6. Ullmann
`coupling22
`(K2CO3, CuI, 1,10-phenanthroline in DMSO,
`130 (cid:176) C) of compound 9c with excess piperidine in a sealed tube
`provided compound 37 in 5% yield. In a similar manner, the
`Ullmann coupling of 9c with (cid:228)-valerolactam or caprolactam led
`to the P4 phenyllactam analogues 38a,b in 20-25% yield.
`Likewise, treatment of pyrazole 9b with (cid:228)-valerolactam under
`similar Ullmann conditions provided compound 39 in 21% yield,
`which on aminolysis with ammonia in ethylene glycol at
`120 (cid:176) C provided compound 40 in 76% yield.
`The preparation of compound 47 is outlined in Scheme 7.
`Cycloaddition of chlorohydrazone compound 7b and morpholine
`derivative 42 (prepared in 65% yield from lactam 41) with
`triethylamine in toluene under reflux conditions followed by
`treatment with TFA afforded the bicyclic pyrazole 43 in 75%
`yield. Hydrogenation (palladium on carbon in methanol)
`provided aniline 44 in 96% yield. Boc protection of 44 (Boc2O,
`NaH in THF) followed by alkylation (NaH and iodomethane)
`and removal of
`the Boc group with TFA provided the
`N-methylaniline derivative 45 in 56% yield. Aminolysis of 45
`with ammonia in ethylene glycol at 120 (cid:176) C led to compound
`46, which was acetylated (acetyl chloride in the presence of
`sodium hydroxide (1 N) in DCM) to compound 47 in 30% yield.
`
`Results and Discussion
`in potency seen with the
`Because of the enhancement
`tetrahydropyrazolopyridone scaffold, efforts to extend the SAR
`to include neutral P1 groups such as the p-methoxyphenyl that
`previously showed reduced fXa binding in the monocyclic
`pyrazole series proved to be successful.16a-b Although the
`compounds with this P1 group showed potent fXa inhibition in
`
`a (a) BH3. THF, room temp; (b) PBr3, DCM, room temp; (c) NaH, 1,2,3-
`triazole or 1,2,4-triazole or 1H-tetrazole, DMF; (d) 2-formylphenylboronic
`acid, (Ph3P)4Pd, Na2CO3 (2 N), 4:1 toluene/EtOH, reflux; (e) 3-(R)-
`hydroxypyrrolidine, NaCNBH3, ZnCl2 (0.5 N in THF), MeOH.
`
`Scheme 5. Syntheses of Substituted P4 Amino Analoguesa
`
`a (a) Diphenylmethanimine, BINAP, NaOtBu, Pd2(dba)3, toluene, reflux;
`(b) hydroxylamine hydrochloride, NaOAc, MeOH; (c) Boc2O, neat, 80 (cid:176) C;
`(d) NaH, MeI, DMF; (e) TFA, DCM; (f) MeI, DMF, K2CO3, room temp;
`(g) Ac2O, TEA, DCM, room temp.
`
`the binding assay, the in vitro clotting activity as measured by
`the prothrombin time (PT) assay of these compounds was
`moderate to high. Further optimization of the p-methoxyphenyl
`
`

`

`Apixaban as Inhibitor
`
`Journal of Medicinal Chemistry, 2007, Vol. 50, No. 22 5343
`
`Scheme 6. Syntheses of P4 Lactam Analogues and Compound
`37a
`
`Table 1. In Vitro Activity for Substituted C-3 Pyrazolopyridinonesa
`
`a (a) 1.5 equiv of piperidine, K2CO3, catalyst CuI, DMSO, sealed tube,
`130 (cid:176) C, 24 h; (b) (cid:228)-valerolactam or azepan-2-one, K2CO3, catalyst CuI,
`catalyst 1,10-phenanthroline, DMSO, 130 (cid:176) C 24 h; (c) ammonia in ethylene
`glycol, 120 (cid:176) C, 4 h.
`
`Scheme 7. Synthesis of Compound 47a
`
`a (a) 3 equiv of PCl5, CHCl3, reflux; (b) morpholine reflux; (c) TEA,
`toluene, reflux; (d) TFA, DCM; (e) H2, Pd/C (10%), MeOH; (f) Boc2O,
`NaH, THF; (g) NaH, THF, MeI; (h) TFA, DCM; (i) ammonia, MeOH/
`ethylene glycol, 120 (cid:176) C, 4 h; (j) acetyl chloride, NaOH (1 N), DMC.
`
`bicyclic pyrazole series required careful adjustment for potency
`and polarity at the C-3 pyrazole position for possible alternatives
`to the lipophilic trifluoromethyl substituent (Table 1). In the
`course of our investigation of the C-3 pyrazole position, we
`
`a Ki values were obtained from purified human enzymes and are averaged
`from two experiments (n ) 2).28,29 PT values are measured according to
`refs 7a and 11. Human trypsin Ki values for all compounds above are >3000
`nM. NT indicates “not tested”.
`
`were gratified to see the breadth of substitutions that were readily
`accommodated in this region of the fXa active site. For example,
`in addition to the trifluoromethyl analogue 6,16a subnanomolar
`inhibitory activity was seen for the methylsulfonyl compound
`11a (fXa Ki ) 0.25 nM), the carboxamido compound 13f
`
`

`

`5344 Journal of Medicinal Chemistry, 2007, Vol. 50, No. 22
`
`Pinto et al.
`
`Table 2. Comparative Permeability and Dog Pharmacokinetic
`Parametersa
`
`assay. Amongst the tetrazole analogs, the tetrazole compound
`27 showed moderate clotting activity, though less potent in the
`binding assay, and the reverse was true for tetrazole 28.
`Triazolylmethyl and tetrazolylmethyl analogues 33a-d exhib-
`ited good potencies in both assays. Overall, the compounds
`shown in Table 1 were highly selective (>1000-fold) for fXa
`relative to other serine proteases such as thrombin and trypsin.
`Table 2 lists the pharmacokinetic profile in dogs of a repre-
`sentative set of the most optimized C-3 substituted compounds.
`The carboxamide 13f demonstrated an excellent pharmacokinetic
`profile, with low clearance (Cl ) 0.32 L kg-1 h-1), moderate
`volume of distribution (Vdss ) 1.6 L kg-1), and a half-life (T1/2)
`of 5.6 h. The high oral bioavailability (F ) 100%) exhibited
`by 13f was consistent with the high apparent permeability (Papp
`) 2.3 (cid:2) 10-6 cm s-1) of this compound in the Caco-2 assay.23
`In contrast, the pharmacokinetic profiles of the triazole analogue
`33a and the 1,2,3,4-tetrazole analogue 33d were poor with high
`clearance, moderate volume of distribution, and poor oral
`bioavailability.
`Given the excellent fXa activity exhibited by compound 13f
`and its high oral bioavailability, we shifted the focus on further
`P4 optimization (Table 3). In general, the compounds retained
`subnanomolar fXa binding affinity and potent clotting activity,
`good selectivity (trypsin/thrombin, >100-fold), and showed
`good permeability (Papp) in the Caco-2 assay. The unsubstituted
`amino compound 13a (fXa Ki ) 0.97 nM) was the least potent,
`whereas the substituted amino compounds 13b-d exhibited
`Table 3. C-3 Carboxamido Pyrazoles: In Vitro and in Vivo Profile of the P4 Biarylmethylamino Moietiesa
`
`a Compounds were dosed (po/iv) as TFA salts in a cassette dosing N-in-
`one format7a,34a-c at 0.4 mg/kg iv and at 0.2 mg/kg po (n ) 2).10 Caco-2
`and dog PK parameters were measured according to refs 7a and 11.
`(fXa Ki ) 0.07 nM), the nitrile compound 18 (fXa Ki )
`0.33 nM), and the dimethylamino compound 25 (fXa Ki )
`0.31 nM). The binding affinity and clotting activity of the
`carboxamide analog 13f showed significant improvement when
`compared to the corresponding trifluoro-methyl compound 6.
`Compared to the parent carboxamide 13f,
`the substituted
`carboxamides 15a and 15b, though significantly less potent in
`the binding assay, were only about 2- to 3-fold less potent in
`the clotting (PT) assay. The ester analogue 11b and its
`corresponding carboxylic acid 12 were less potent in both assays.
`Among the C-3 amino analogues investigated, the order of fXa
`potency was NMe2 > NHMe g N(Me)Boc g NH2, NHBoc.
`The unsubstituted amino analogue 22 and the N-methylamino
`compound 24 demonstrated acceptable activity in the clotting
`
`a Ki values were obtained from purified human enzymes and are averaged from two experiments (n ) 2).28,29 Prothrombin time (PT) values are measured
`according to refs 7a and 11. Human trypsin Ki values for all compounds listed in Table 2 are >3000 nM. Caco-2 and dog PK parameters were measured
`according to refs 7a and 11. b Compounds were dosed (po/iv) as TFA salts in a cassette dosing N-in-one format at 0.4 mg/kg iv and 0.2 mg/kg po (n )
`2).34a-c NT indicates “not tested.”
`
`

`

`Apixaban as Inhibitor
`potent fXa inhibitory activity (fXa Ki < 0.3 nM) and good
`clotting activity (PT EC2(cid:2) < 1.5 (cid:237)M). As was observed with
`the 3-(R)-hydroxypyrrolidine compound 13f, the 4-hydroxypi-
`peridinyl analogue 13g also demonstrated low clearance, moder-
`ate volume of distribution, and half-life in the same range as
`observed with 13f. Again, the high dog oral bioavailability seen
`for 13g (F ) 55%) correlated well with the observed Caco-2
`(Papp) permeability value. In the rabbit arteriovenous shunt
`(AVShunt) thrombosis model,6g compounds 13b, 13c, 13f, and
`13g inhibited thrombus formation in a dose-dependent manner
`with IC50 values of 445, 175, 120, and 180 nM, respectively,
`and with the exception of 13b were slightly more potent than
`compound 4 (AVShunt IC50 ) 340 nM)7a and were in the same
`range when compared to compound 5 (AVShunt IC50 ) 140
`nM).15a Of the compounds in this series, 13b had the longest
`half-life in dogs, albeit with relatively high clearance and high
`volume of distribution. Interestingly, these data did not correlate
`well with the observed 13b half-life in the human liver
`microsome (HLM) assay24 (T1/2 > 100 min). In the same assay,
`the HLM half-life for compounds 4 and 5 was 38 and 42 min,
`respectively. Taken together therefore, the carboxamide pyrazole
`analogue 13f emerged as a potent alternative to compound 4,
`with excellent potency both in vitro and in vivo and a good
`pharmacokinetic profile in dogs.
`In a parallel effort, the compounds containing P4 nitrogen
`atom (as the point of attachment) were also explored (Table
`4).12c This strategy proved to be highly successful in that a potent
`compound 36d bearing a N-methylacetyl group was quickly
`identified. This discovery was significant in that it differed in
`structure from all our previous pendent P4 groups we had
`explored. The compound, though more potent than the aniline
`derivatives 34, 36c, and 36e, was weak in the clotting assay,
`suggesting high protein binding. The high level of potency
`exhibited by compound 36d (fXa Ki ) 0.50 nM) suggested that
`the orientation of the P4 N-methylacetyl substituent in the S4
`region of the fXa active site is very important. This type of
`observation was unique in the fXa literature at the time it was
`discovered. To explain this finding, a closer look at the model
`of 36d in the active site of fXa clearly showed the N-methyl P4
`group forming a lipophilic (cid:240) interaction with the bottom S4
`Trp215 residue,12d and thus positioning the acetyl carbonyl
`functionality perpendicular to the inner P4 phenyl ring, thereby
`forming a hydrophobic interaction with the other residues in
`this region. The importance of the orientation of the N-methyl
`group was confirmed by the loss in fXa affinity with the
`acetamide analogue 35 (fXa Ki ) 180 nM) where the planarity
`of this group positioned it in an unfavorable orientiation in the
`S4 region of the enzyme. The dimethylamino analogue 36e (fXa
`Ki ) 6.0 nM) and piperidinyl 37 (fXa Ki ) 2.1 nM) were also
`less potent, suggesting a planar orientation with these moieties
`in the S4 region as well. Cyclization of the P4 N-methyl acetyl
`group in 36d to form lactam analogues 38a and 38b retains the
`subnanomolar fXa binding affinity. Unfortunately,
`lactam
`analogues 38a (PT ) 23 (cid:237)M) and 38b (PT ) 26 (cid:237)M) exhibited
`poor anticoagulant activity. This could be explained by the high
`lipophilicity (cLogP > 7) and high human serum protein binding
`(>99%)25 exhibited by these compounds.
`In order to modulate the lipophilicity of 36d and 38a, we
`reintroduced the polar C-3 carboxamido moiety (Table 5) that
`was shown to be important in compounds 13a-h. The car-
`boxamidopyrazole analogue 40 (fXa Ki ) 0.08 nM, PT ) 3.8
`(cid:237)M) and 47 (fXa Ki ) 0.61 nM, PT ) 3.1 (cid:237)M) not only
`maintained subnanomolar fXa binding affinity but also dem-
`onstrated much improved potency in the clotting (PT) assay,
`
`Journal of Medicinal Chemistry, 2007, Vol. 50, No. 22 5345
`
`Table 4. In Vitro Profile of P4 Substituents with Imbedded Nitrogena
`
`a Ki values were obtained from purified human enzymes and are averaged
`from two experiments (n ) 2).28,29 Prothrombin time (PT) values are
`measured according to refs 7a and 11. Human trypsin Ki values for all
`compounds listed in Table 2 are >3000 nM. NT indicates “not tested”.
`
`especially when compared to the related trifluoromethylpyrazole
`analogues 38a and 36d.
`Selectivity and Liability Profiling. Compound 40 shows a
`high degree of selectivity versus other proteases (see supple-
`mental section), even compared to compounds 47a and 5.15a
`Additionally, the compound shows weak activity against various
`P450 isozymes (IC50 > 25(cid:237)M) and weak activity against the
`hERG potassium channel (IC50 > 25 (cid:237)M, patch clamp assay).26a-e
`The solubility of compound 40 was shown to be approximately
`40-50 (cid:237)g/mL.27 In the human liver microsome assay, com-
`pound 40 was very stable with a T1/2 of >100 min (the HLM
`T1/2 of 47 was not measured). The Caco-2 permeability values
`for compounds 40 (Papp ) 0.9 (cid:2) 10-6 cm s-1) and 47 (Papp )
`2.5 (cid:2) 10-6 cm s-1) were moderate to high.
`Dog Pharmacokinetics and Rabbit Antithrombotic Ef-
`ficacy. As a result of the excellent
`in vitro potency and
`selectivity of compounds 40 and 47, the pharmacokinetic profiles
`of both compounds were studied in dogs using a cassette dosing
`paradigm (“N-in-one” study, Table 6).7a,34a,b The acetylated
`N-methyl analogue 47 was orally bioavailable; but showed high
`clearance (Cl ) 2.8 L kg-1 h-1), moderate volume of distribu-
`tion (Vdss ) 1.7 L kg-1), and unacceptable half life. The dog
`pharmacokinetics for compound 40 was outstanding with very
`low clearance (Cl ) 0.02 L kg-1 h-1), and low volume of
`distribution (Vdss ) 0.2 L kg-1). These values were significantly
`
`

`

`5346 Journal of Medicinal Chemistry, 2007, Vol. 50, No. 22
`
`Table 5. Optimization of the Amido and Lactam P4 Moietiesa
`
`Pinto et al.
`
`a Ki values were obtained from purified human enzymes and are averaged from two experiments (n ) 2).28,29 PT and APTT values, Caco-2, and solubilities
`were measured according to refs 7a and 11. Human trypsin Ki values for all compounds listed in Table 2 are >3000 nM. NT indicates “not tested.”
`
`Table 6. Comparative in Vitro and in Vivo Profiles of Compounds 40, 47, 4, and 5a
`
`a “h” and “r” refer to human and rabbit species, respectively. Ki values were obtained from purified human enzymes and are averaged from two experiments
`(n ) 2). Compounds were dosed in a cassette dosing (po/iv) N-in-one format7a,34a-c a refers to the cassette dosing (po/iv) dog pharmacokinetic parameters.
`P.B. refers to serum protein binding. NT indicates “not tested”.
`
`lower than those observed with compounds 4 and 5. FXa being
`a vascular target, the pharmacokinetic profile for compound 40
`was viewed as highly desirable and less likely to have nontarget-
`related adverse effects. Importantly, compound 40 had a
`moderate half-life (T1/2 ) 5.8 h) and good oral bioavailability
`(F ) 58%). The human serum protein binding as measured by
`equilibrium dialysis for 40 was 87%.25 In the rabbit AVShunt
`thrombosis model (Figure 4), compound 40 inhibited thrombus
`formation in a dose-dependent manner with an IC50 value of
`329 nM.15d,e This is comparable to the antithrombotic potency
`obtained for compound 4 (IC50 ) 340 nM) in the same model.
`X-ray Crystallography of Compound 40. The X-ray
`structure for compound 40 bound to fXa (Figure 2, 2.3 Å
`resolution with an R value of 0.229 and an Rfree of 0.277) shows
`a tight inhibitor-enzyme complex30-33 and shows a similar
`binding mode compared to compound 211 and 4.7a The p-
`methoxy group in the S1 specificity pocket does not appear to
`interact with any specific residue in this region of the enzyme,
`and is oriented in a planar manner relative to the phenyl P1
`moiety. Other interactions that were found to be similar to those
`observed for compounds 47a and 515a included the pyrazole N-2
`nitrogen atom interaction with the backbone of Gln192 (3.2 Å)
`and the carbonyl oxygen (scaffold carboxamide) interaction with
`the NH of Gly216 (2.9 Å). The pyrazole C-3 carboxamido
`moiety shows the NH within bonding distance to the Glu146
`carbonyl oxygen (3.1 Å) with the carbonyl oxygen solvent
`exposed. The orientation of the pendant P4 phenyllactam of 40
`in the S4 region shows an edge to face interaction with Trp215
`and is appropriately positioned between the Tyr99 and Phe174
`
`Figure 2. X-ray structure of fXa bound to compound 40. Atomic
`coloring is used with cyan for fXa cartoon and carbon atoms and gray
`for compound 40 carbon atoms. Water molecules are shown as red
`spheres. Initial electron density (2Fo - Fc contoured at 1(cid:243)) is shown
`in magenta. Hydrogen bonds between protein and ligand are shown as
`dashed black lines. The figure was created using PyMol.35
`
`residues. The X-ray structure does not appear to show the
`carbonyl oxygen group of the pendent lactam moiety directly
`interacting with any residues in the S4 pocket but appears to
`show it interacting with a water molecule. Importantly, the P4
`lactam carbonyl brings about a conformational bias toward
`
`

`

`Apixaban as Inhibitor
`
`Journal of Medicinal Chemistry, 2007, Vol. 50, No. 22 5347
`
`Table 7. Diffraction Data for Compound 40 (BMS-562247)
`
`(A) Data Collection Statistics
`P212121
`
`space group
`unit cell parameters
`a, Å
`b, Å
`c, Å
`
`diffraction limits, Å
`no. of unique reflections
`data completeness
`
`56.4
`72.8
`79.4
`overall
`19.4-2.3
`14544
`96.8
`
`highest shell
`2.4-2.3
`1645
`94.9
`
`2238
`34
`122
`0.008
`1.5
`0.229
`0.277
`5.9
`
`(B) Refinement Statistics
`no. of non-H atoms in protein
`no. of non-H atoms in inhibitor
`no. of refined water molecules
`rmsd bond distances from ideal, Å
`rmsd angles from ideal, deg
`crystallographic residual R value
`crystallographic residual Rfree
`test set, % of all reflections
`
`Figure 3. Superposition X-ray structures of fXa bound to compounds
`40 and 4. Atomic coloring is used with cyan for fXa cartoon and carbon
`atoms, gray for compound 40 carbon atoms, magenta for compound 4
`carbon atoms, and orange for compound 4 fluorine atoms. The
`orientation is the same as in Figure 2. The figure was created using
`PyMol.35
`
`Figure 4. Rabbit arteriovenous shunt (AVShunt) profile of compound
`40 (BMS-562247).
`
`orthogonality to optimally position itself between the S4 enzyme
`residues Trp215, Tyr99, and Phe174. Overall, compound 40 fits
`into the fXa enzyme active site in a highly complementary
`manner. Figure 3 shows an overlay

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