throbber
J. Med. Chem. 2005, 48, 5025-5037
`
`5025
`
`Discovery and Preclinical Profile of Saxagliptin (BMS-477118): A Highly Potent,
`Long-Acting, Orally Active Dipeptidyl Peptidase IV Inhibitor for the Treatment
`of Type 2 Diabetes
`
`David J. Augeri,*tt Jeffrey A. Robl,t David A. Betebenner,t David R. Magnint Ashish Khanna,
`James G. Robertson, Aiying Wang,' Ligaya M. Simpkins,t Prakash Taunk,t Qi Huang,- Song-Ping Han,-
`Benoni Abboa-Offei,'I Michael Cap, Li Xin,- Li Tao,# Efflie Tozzo,- Gustav E. Welzel, Donald M. Egan,'
`Jovita Marcinkeviciene," Shu Y. Chang," Scott A. Biller,t Mark S. Kirby,' Rex A. Parker,' and
`Lawrence G. Hamaim*
`
`Departments of Discovery Chemistry, Metabolic Diseases, Pharmaceutical Candidate Optimization,
`Exploratory Pharmiaceutics, Chemical Enzymologv, Bristol-Myers Squibb, Phacrmaceutical Research Institute,
`P.O. Box 5400, Princeton, New Jersey 08543-5400
`
`Received March 22, 2005
`
`Efforts to further elucidate structure -activity relationships (SAR) within our previously
`disclosed series of 3-quaternary amino acid linked L-cis-4,5-methanoprolinenitrile dipeptidyl
`peptidase IV (DPP-IV) inhibitors led to the investigation of vinyl substitution at the /-position
`of t.-cycloalkyl-substituted glycines. Despite poor systemic exposure, vinyl-substituted com-
`pounds showed extended duration of action in acute rat ex vivo plasma DPP-IV inhibition
`models. Oxygenated putative metabolites were prepared and were shown to exhibit the potency
`and extended duration of action of their precursors in efficacy models measuring glucose
`clearance in Zuckerf"'f rats. Extension of this approach to adamantylglycine-derived inhibitors
`led to the discovery of highly potent inhibitors, including hydroxyadamantyl compound BMS-
`477118 (saxagliptin), a highly efficacious, stable, and long-acting DPP-IV inhibitor, which is
`currently undergoing clinical trials for treatment of type 2 diabetes.
`
`Introduction
`Primary defects in insulin secretion, along with
`development of insulin resistance, contribute to the
`etiology of type 2 diabetes mellitus. Diminished post-
`prandial insulin secretion resulting from both functional
`defects and
`loss of survival of pancreatic
`/3-cells
`progresses into hyperglycemia and declining insulin
`sensitivity. As lifestyle trends and dietary factors have
`contributed to an alarming rise in the incidence of type
`2 diabetes,' the search for novel mechanistic approaches
`to control this chronic metabolic disease has intensified
`in parallel. To complement the currently available
`diabetes treatments,2 approaches operating within the
`enteroinsular axis through the incretin hormone glu-
`cagon-like peptide 1 (GLP-1), alone or in combination
`with other agents, are beginning to show promise in the
`treatment of diabetes.3 GLP-1 is a major component of
`the prandial nutrient-sensing mechanism regulating
`insulin secretion
`following meals.4 Intact, active
`GLP-1(7-36) amide is secreted into the circulation from
`intestinal L-cells in response to dietary signals. Con-
`centrations of GLP-1(7-36) amide sufficient to activate
`
`fax:
`
`609-818-5526
`* Corresponding
`Telephone:
`author.
`609-818-3550. E-mail: lawrenice.hamannbm s-com.
`I Discovery Chemistry.
`Present address: Lexicon Pharmaceuticals, 350 Carter Road,
`Princeton, NJ 08540.
`Present address: Pharmaceutical Discovery Division, Abbott
`LaboraLories, Abbott Park, IL 60064
`1 Pharmaceutical Candidate Optimization.
`IMetabolic Diseases,
`4 Exploratory Pharmaceutics.
`Chemical Enzymology.
`Present address: Novartis tnstitute for BioMedical Research,
`250 Massachusetts Avenue, Cambridge, MA 02139
`
`the GLP-1 receptor expressed on pancreatic /3-cells
`result in increased insulin secretion, delayed glucose
`absorption, and reduced hepatic glucose production. All
`of these components work in concert to modulate blood
`glucose levels. Because GLP-1 release is nutrient stimu-
`lated, this mechanism promotes insulin secretion under
`prandial glycemia conditions, minimizing the potential
`for hypoglycemia. Recent reports have further demon-
`strated a beneficial effect of agents acting through the
`GLP-1 axis on the preservation and/or restoration of
`/3-cell function in animals,' suggesting the exciting
`possibility that emerging drugs acting in this pathway
`may lead to improvement of the diabetic condition.
`GLP-1 is rapidly truncated during its secretion in the
`ileum by the dipeptidyl peptidase IV (DPP-IV, EC
`3.4.14.5) located on the capillary endothelium proximal
`to the L-cells where GLP-1 is secreted The efficient
`cleavage by DPP-IV of the N-terminal dipeptide His-
`Ala from GLP-1(7-36) amide yields GLP-1(9-36) amide,
`a weak antagonist of the receptor, and this cleavage
`has been demonstrated to be the primary physiological
`route of degradation of GLP-1(7-36) amide in both
`humans and animals.7 The rapid cleavage by DPP-IV
`results in an apparent elimination half-life of only 60-
`90 s for GLP-1(7-36) amide, and peak circulating levels
`of intact GLP-1(7-36) amide typically do not exceed
`5-10 pM, a range bracketing its K. as GLP-1 receptor
`agonist. Inhibition of DPP-IV prevents the degradation
`of the incretin hormones GLP- 1 and glucose-dependent
`insulinotropic peptide (GIP) and has been demonstrated
`to potentiate the levels of these peptides in multiple
`species."
`
`10.1021/jm050261p CCC: $30.25 @ 2005 Ameican Chemical Society
`Published on Web 06/24/2005
`
`AstraZeneca Exhibit 2171
`Mylan v. AstraZeneca
`IPR2015-01340
`
`Page 1 of 13
`
`

`
`5026 Journal of Medicinal Chemistry, 2005, Vol. 48, No. 15
`
`Augeri et al.
`
`Scheme 1
`
`R2
`
`Bo N
`
`b, c
`
`H2
`
`0
`7a-g
`
`CONH2
`
`2
`R
`
`a, b^
`
`OH + 5
`
`CN
`
`0
`Sa-g
`
`R2
`2 ]
`
`H2N
`
`N
`
`CN
`10a, b, d, g
`
`Ri
`
`BocHN
`
`o
`9a, b, d, g
`
`BocHN mrOH
`
`6a-g
`
`TFAa
`CONH 2
`
`d
`
`a. R, = R 2 = Me
`b. R, = R2 - Et
`c. R 1,R 2 = -9tH 2)3-
`d. R,R 2 = -(CH24-
`e. R1,R2 =-H2)5-
`f. RiR 2 -
`-(CH) 6 -
`g. R1 ,R2 = -(CH 2CH 2OCH 2CH 2)-
`DM'; 'b) POC13 , pyridine, imidazole, -20
`
`I (a) EDAC, HOBT,
`
`DPP-IV is a 240 kDa, 766 residue N-terminal dipep-
`tidyl exopeptidase that is composed of two 110 kDa
`subunits' and exists as both a membrane-bound protein
`and as a soluble protein in plasma. It is a nonclassical
`serine protease that exhibits high specificity for peptides
`with proline or alanine in the P1 position. Any amino
`acid can occupy the P2 position so long as the P2 -P1
`peptide bond can adopt a trans configuration."0 The
`membrane-bound form of DPP-TV is expressed in several
`tissues, including kidney, liver, the brush border mem-
`branes of intestinal enterocytes, on the pancreatic duct
`epithelia, and in vascular endothelial cells. In these
`tissues DPP-IV is N-terminally bound to the membrane
`with its catalytic activity located in the extracellular
`domain. The soluble, circulating form of DPP-IV is shed
`from cell surfaces by proteolytic cleavage releasing a
`fully active soluble form minus the 29 amino acids of
`the N-terminus.
`Clinical evidence has shown that small molecule
`inhibitors of DPP-IV lower blood glucose levels, increase
`glucose tolerance, and improve insulin response to oral
`glucose in patients with type 2 diabetes-" Reversible
`small-molecule inhibitors of DPP-IV have been studied
`for the past several years, and a large body of structure-
`activity relationship (SAR) data has been generated. 3b,12
`Until the very recent disclosures of several nonpeptidic
`chemotypes," 14 the known inhibitors had all been
`dipeptidomimetic in nature, bearing structural resem-
`blance to the N-terminal dipeptide of the enzyme
`substrates. For this class of inhibitors, the penultimate
`N-terminal proline or proline mimetic, generally a
`thiazolidine (1),ls a C-substituted or N-substituted
`
`R
`
`SR
`
`H2N
`
`'
`0
`1
`
`H2N
`
`'
`0
`2
`
`qN
`
`RN
`
`CN
`
`CN
`
`H
`3
`
`R,
`
`R2 R
`
`H2N
`
`N
`0 CN
`4
`
`cyanopyrrolidine (2, 3),16,17 or a cyclopropanated cyan-
`opyrrolidine (4)," is appended to an amino acid or an
`amino acid surrogate. Many inhibitors in this cyano-
`pyrrolidine structural class have suffered from varying
`degrees of chemical instability which have hampered
`formulation efforts. In addition, many examples of this
`class exhibit limited pharmacodynamic duration of
`action. We report herein the discovery of highly effica-
`cious long-acting inhibitors of DPP-IV that have led to
`the identification of compound 26 (BMS-477 118, saxa-
`
`'C; (c) TFA, CH2C1 2, rt; (d) 5% Pd/C, 112 1 atm, MeOHl.
`Scheme 2"
`R1
`
`a
`
`0
`
`R2
`
`11a-g
`
`R1
`R2
`O 2Et
`12a-g
`
`b
`
`b 2 R
`13a-g
`
`OH
`
`Rj
`R2
`
`0
`
`NHBoc
`
`R2
`
`OH
`
`d
`
`BocHN
`
`0
`14a-g
`6a-g
`'C
`triethylphosphcnoacetate, NaH, THF 0
`I (a)
`to rt;
`(b) DIBAL-H, toluene, -78
`'C to rt; (c) N-Boc glycine, DCC,
`DMAP, CH 2C12, Irt; (d) ZnC12, THY, LDA, -78
`-C to rt.
`
`gliptin), which is currently undergoing clinical evalua-
`tion for the treatment of type 2 diabetes.
`
`Chemistry
`To further our understanding of the SAR surrounding
`/3-quaternary N-terminal amino acid-containing inhibi-
`tors, we focused on elaboration of our previously dis-
`closed cyanomethanopyrrolidine-based scaffold 3 to pro-
`duce long-acting inhibitors structurally related to the
`prototype scaffold 4. A general synthesis route was
`chosen that incorporated at the
`-position a vinyl
`substituent amenable to functionalization for further
`elucidation of SAR. Standard peptide coupling condi-
`tions were employed to link enantiomerically pure
`L,-methanoprolinamide core fragment 5 with various
`racemic vinyl-substituted amino acids Ca-g to give
`dipeptides 7a-g in yields of 85-95% (Scheme 1).
`Dehydration of the resultant amides using TFAA or
`POCI3 gave the corresponding nitriles.20 Chromato-
`graphic isolation of the bioactive L-isomer was generally
`carried out at the stage of the Boc-protected nitrile. 21
`Finally, removal of the N-terminal Boc using TFA gave
`inhibitors 8a-g in high yield. The vinyl groups of Ca-f
`could be reduced (Pd/C, H2 ) to afford the corresponding
`ethyl compounds, which were similarly elaborated to
`dipeptides lOa,b,d,g.
`Amino acids possessing a J-quaternary vinyl group
`were prepared in a manner complementary to the
`malonate Knoevenagel/Michael addition sequence used
`previously." Lewis acid-mediated ester enolate Claisen
`rearrangement of substituted glycinyl allylic esters2 led
`directly to /-vinyl amino acids 6a-g in 58-85% overall
`yields (Scheme 2). The requisite Claisen precursors were
`readily prepared in three steps from the appropriate
`ketones 11a-g. Horner-Enmons olefination ofketones
`
`Page 2 of 13
`
`

`
`Discovery and Preclin ical Profile of Saxagliptin
`
`Journal of Medicinal Chemistry, 2005, Vol. 48, No. 15 5027
`
`Scheme 30
`
`BocHN
`
`)n
`
`N
`
`O
`15c-e
`
`CN
`
`1. aorbc-
`
`2. d
`
`Hoc 0
`H N
`Bo/
`
`N
`CN
`
`If
`O
`
`17d
`18d
`to 0 'C, 60-79%; (b) Os04, NMNO, THF/H20 1:1, rt, 47-63%; (c) NaIO 4;
`'C; then NaBH 4, -78
`(a) 03, MeOH/CH 2C2 10:4, -78
`rt, 56%; (d) TFAICH2Cla1:2, O 'C to rt-
`workup, then NaBH 4, MeO
`
`d.n=1
`d. n = 1
`e. ni = 2
`
`H2N
`
`I/
`
`N.
`
`O
`16c-e
`
`CN
`
`d
`
`CN
`
`N
`
`r 0
`
`HO
`HO
`H 2N
`
`Scheme 40
`
`H3 CO 2 C
`19
`
`a, bc
`
`HO
`
`g, h, i
`
`20
`
`21
`
`H2N
`
`N
`H
`
`C0 2H
`
`-tN_
`0
`CN
`23
`22
`a (a) LAH, THF, 0 'C to rt, 96%; (b) (CICO)2 , DMSO, CH2 C12,
`'C, 98%; c (-(-)-2-phenylgycinol, NaHSO 3, KCN, 65%;
`-78
`(d) 12 M HCI, HOAc, 80 'C, 16 h, 78%; (c) 20% Pd(OH)2, 50 psi
`H2 , MeOH/HOAc 5:1; (f) (Boc20, KXCO, DMF, 92%, two steps;
`(g) 5, EDAC, HOBT, DMF, 92%; (h) POC13, pyridine, imidazole,
`'C;
`i) TFA, CH9 C12, r, quanL.
`-20
`
`11a-g with the ylide generated from triethylphospho-
`noacetate gave the a,3-unsaturated esters 12a-g in
`92-98% yield. Esters 12a-g were then reduced with
`DIBAL to the corresponding allylic alcohols 13a-g and
`condensed with N-Boc glycine using DCC/DMAP to give
`esters 14a-g in 79-87% yield over two steps. ZnClz-
`mediated Claisen rearrangement of the LIDA-generated
`enolate of glycine esters 14a-g proceeded at low tem-
`perature to give the desired /3-vinyl amino acids 6a-g
`in 65-90% yield.
`Further elaboration of vinyl-containing dipeptides
`7c-e was accomplished at the stage of the dehydrated
`cyano-containing compounds 15c-e (Scheme 3). Oidda-
`tive cleavage of the vinyl substituent to prepare hy-
`droxymethyl compounds 16c-e was achieved either by
`ozonolysis/NaBH 4 reduction or by catalytic 0s04-
`NMNOINaIO4/NaBH 4 conditions, followed by acidic
`deprotection of the Boc group. Additionally, 15d was
`converted to the corresponding diol and deprotected to
`give 18d.
`A logical extension of our previously observed SAR
`trends favoring f-branched P2 units led us to explore
`rigidly bridged polycyclic systems such as adamantyl.
`Analogues bearing an adamantyl ring at the N-terminal
`a-carbon were synthetically derived from a common
`homochiral adamantylglycine intermediate prepared
`using asymmetric Strecker chemistry (Scheme 4).3
`Reduction of commercially available adamantane car-
`boxylic acid methyl ester 19 by LAH, followed by Sworn
`
`oxidation, afforded the requisite aldehyde. which was
`then subjected to asymmetric Strecker conditions (con-
`densation with (R)-(-)-2-phenylglycinol with addition
`of KCN) to give the desired homochiral R,S diastere-
`omer 20 in 65% yield. Hydrolysis of the nitrile group to
`give acid 21, followed by hydrogenolysis of the chiral
`auxiliary, afforded the enantiomerically pure amino acid
`22. Boc protection of the resulting primary amine,
`followed by coupling to methanoprolinamide core 5,
`dehydration of the amide to nitrile, and deprotection,
`afforded the adamantylglycine containing inhibitor 23
`in good overall yield-
`Hydroxylation of N-Boc-adamantylglycine 22 at the
`bridgehead was accomplished using KMnO 4 in 2%
`aqueous KOH at elevated temperature to give N-Boc
`hydroxyadamantyl glycinc 24 in 51% yield (Scheme 5).24
`Standard acylation conditions were used to couple 24
`to methanoprolinamide core 5, furnishing anide 25 in
`high yield. Amide 25 was subsequently elaborated to
`provide two additional analogues. Dehydration of amide
`25 with TFAA, followed by in situ basic hydrolysis of
`the resulting trifluoroacetate and deprotection of the
`N-terminus, gave hydroxy derivative 26 in 87q yield
`over three steps. The hydroxy group of 25 was subjected
`to fluoride substitution using DAST, 24 and subsequent
`dehydration using POC 3 in pyridine, followed by depro-
`tection of the terminal nitrogen, provided fluoroada-
`mantylglycine analogue 30 in 73% overall yield for three
`steps. Prolonged exposure of protected adamantylglycine
`22 to KMnO4 in 2% aqueous KOH provided the dihy-
`droxyadainantylglycine derivative 27. Coupling of 27 to
`5, followed by dehydration of the resultant prolineamide
`with TFAA, in situ basic hydrolysis of the bis-trifluoro-
`acetate, and removal of the terminal Boc group using
`TFA, afforded dihydroxyadamantyl analogue 28 in 74%
`overall yield.
`In Vitro and in Vivo Biological Activity. DPP-
`IV Inhibitory Activity in Vitro and ex Vivo. The
`DPP-IV inhibitory activity of analogues in the present
`series was measured against human DPP-IV using
`standard assays as described in the Experimental
`Section (Table 1). Many of the compounds in this series
`were potent inhibitors of DPP-IV in vitro, several with
`Ki's in the sub-nanomolar range. Additionally, several
`inhibitors in this series exhibited significant slow, tight-
`binding kinetics. 5
`A finer discrimination between the most potent
`compounds within this structurally related series of
`inhibitors with respect to pharmacodynamic effects and
`
`Page 3 of 13
`
`

`
`5028 Journal of Medicinal Chemistry, 2005, Vol. 48, No. 15
`
`Augeri et al.
`
`Scheme 50
`
`/a
`
`HO
`
`b
`
`c, d
`
`BOC-HN
`
`COH
`
`BOC-HN
`
`CO 2H
`
`BOC
`
`22
`
`24
`
`a, 90 oin
`
`HO
`
`JOH
`
`HO
`
`,OH
`
`BOC-HN CO2H
`
`H2 N
`
`Ie
`
`f,d
`
`F
`
`H
`2
`
`CN
`
`O0
`27
`30
`(-f>NH2
`28
`29
`(a) KMnO 4, 2% aq KOH, 60 to 90 0C, 60 min, 51%, (b) 5, EDAC, HOBT, DMF, 77-85%, (c) (CF 3COhO, pyridine, THF 0 'C to rt, then
`10% aq K 2C0 3 in MeOH, 89-92%; (d) TFA, CH2C12 , rt, 89-95/; (e) DAST, CH 2C12, _78 C, 94%; f) POC1:, pyridine, imidazole, 82%.
`
`Table 1. In Vitro Inhibition Constants for Human DPP-IV and
`cx Vive Plasma DPP-IV Inhibition in Normal Rats
`
`Table 2. Potency and Duration of Effect of Compounds 16d
`and 26 in the ex Vive Rat Plasma DPP-IV Inhibition Model
`
`ED, emol/kg at time postdosel
`
`compd
`
`0.5 h
`
`2 h
`
`l6d
`26
`
`0.4 + 0.15
`0.12 + 0.04
`
`3.2 z 1.2
`0.2 + 0.07
`
`4 h
`5.0 & 1.9
`0.3 + 0.10
`
`6 h
`
`11 + 4.2
`0.5 + 0.15
`
`' Compounds dosed po to fasted normal SD rats at the indicated
`times postdose, plasma aliquots were isolated, and DPP-IV inhibi-
`tion was assayed using the fluorogenic peptide assay. EDso is the
`50%. inhibitory dose calculated from the plot s of percent inhibition
`vs dose aL each time point.
`
`postdose to assay plasma (prepared with EDTA) DPP-
`IV activity in vitro using the fluorogenic DPP-IV-specific
`substrate Ala-Pro-AFC. Plasma DPP-IV activity deter-
`minations were calculated by linear regression from
`plots of product vs time (initial 20 min). Data were
`calculated as mean percent inhibition vs controls receiv-
`ing water vehicle. Maximal inhibition of plasma DPP-
`IV under the conditions of this assay reached 85 -90%
`(Table 1). In a dose- relationship mode, ED 5o's were
`determined for select compounds at multiple time points
`of 0.5, 2, 4, and 6 h postdose (Table 2).
`
`Results and Discussion
`The SAR described in our previous account culminat-
`ing in 4,5-methanoprolinenitrile analogues 4 revealed
`a strong preference for compounds with lipophilic
`N-terminal fl-quaternary amino acids." In the course
`of further studies exploring SAR around fl-quaternary
`cycloalkylglycine-based inhibitors, we encountered un-
`expectedly potent activity and extended duration of
`action in ex vivo DPP-IV inhibition studies with com-
`pound 8d, which contains a (vinylcyclopentyl)glycine
`amino acid fragment. However, metabolism and phar-
`macokinetic studies with 8d revealed uncharacteristi-
`cally poor oral bioavailability (F = 5.3%) and high rat
`liver microsomal turnover rate [0.55 nmol/inin/mg pro-
`tein for 8d vs 0.32 for compound 4 (where R1 and R 2
`taken together - cyclopentyl, and R3 - Me)]. Similar
`observations were made for other vinyl-containing
`analogues 8c,c,g, and these results suggested conver-
`sion to an active metabolite in vivo. As the vinyl
`substituent seemed a likely site of metabolism, synthe-
`sis of oxygenated analogues (16d and 18d) derived from
`chemical modification of the olEfin moiety was under-
`
`% plasma DPP-IV inhibn at
`4 jimol/kg po, normal rats
`
`4 h
`10
`20
`32
`64
`60
`66
`nd
`
`0n
`
`d
`44
`nd
`17
`56
`
`8n
`
`d
`83
`87
`57
`61
`
`30 min
`13
`39
`42
`71
`76
`77
`nd5
`
`0n
`
`d
`40
`nd
`36
`69
`17
`nd
`84
`87
`62
`80
`
`human
`DPP-IV
`K, (nM
`
`57+8
`25 + 4
`12 + 0.9
`3.9
`0.6
`1.4 + 0.06
`10 + 3
`10 ± 2
`7.1
`0.7
`31 ± 2
`5.5 - 0.7
`21 ± 0.6
`42 ± 4
`7.4 ± 1.1
`8.0 + 0.4
`143 ± 15
`0.9 + 0.32
`0.6 + 0.06,
`2.1 ± 0.3
`1.8 + 0.5
`
`compd
`
`8a
`8b
`Sc
`8d
`8e
`Sf
`8g
`10a
`10b
`10d
`log
`16c
`16d
`16e,
`l8d
`23
`26
`28
`30
`
`' Values represent the mean - SEM and are at least triplicate
`determinations. nd = not determined. ' Compound 26 did not
`show any significant inhibition of dipeptidyl peptidase II
`(DPP-I) at concentrations up to 30 pM.
`
`duration of action required utilization of a medium-
`throughput acute efficacy model measuring a surrogate
`biomarker expected to be predictive of downstream
`antihyperglycemic effects. As DPP-IV is found in plasma
`and on the surfaces of blood and tissue cells, it was
`reasoned that measurement of inhibition of the circulat-
`ing enzyme in plasma might provide a convenient
`biomarker for the degree of preservation of plasma
`incretin hormone levels. Though the relative contribu-
`tion of these enzyme loci to the physiological degrada-
`tion of GLP-1(7 -36) amide important for antihyper-
`glycemic effects is not fully understood, it was further
`envisioned that plasma enzyme inhibition measured cx
`vivo after an oral dose of test compound might be used
`to develop pharrmacokinctic- pharmacodynamic rela-
`tionships and provide information regarding duration
`of action. Compounds were administered orally in water
`vehicle at 4 jmol/kg to normal Sprague-Dawley rats,
`and blood samples were taken at 30 min and 4 h
`
`Page 4 of 13
`
`

`
`Discov~ery and Preclin ical Profile of Saxagliptin
`
`Journal of Medicinal Chemistry, 2005, Vol. 48, No. 15 5029
`
`taken. Dio1 1Sd showed only weak inhibitory activity;
`however, hydroxymethyl analogue 16d exhibited po-
`tency similar to that of the vinyl analogue in both in
`vitro and ex vivo assays, restored rat liver micros omal
`turnover rate to a more moderate level (0.16 nmollmin/
`mg protein), and restored oral bioavailability to within
`the range characteristic for other structurally related
`analogues in the series (F = 59%). Unequivocal char-
`acterization of 16d as the active metabolite of Sd was
`never established, though the behavior of 16d mirrored
`that achieved upon administration of Sd. Accordingly,
`DPP-IV inhibitors 16c and 16e were prepared. A similar
`trend toward reconnection of pharmacokinetic proper-
`ties with pharmacodynamic measurements was ob-
`served for the homologous pairs of inhibitors Sc16c and
`8e/16e. Despite this latter observation, the five-mem-
`bered ring compound 16d stood out as significantly more
`effective in the rat ex vivo plasma DPP-IV inhibition
`assay.
`A more striking observation of metabolic conversion
`was seen with the highly potent adamantylglycine-
`containing analogue 23 (Ki = 0.9 nM). Although this
`compound afforded potent plasma DPP-IV inhibition
`after oral administration to rats (84% at 0 5 h, 83% at
`4 h), it exhibited poor absolute bioavailability (F = 2%)
`after oral dosing and rapid turnover in rat liver micro-
`somes. Interestingly, compound 23 also weakly inhibited
`CYP3A4 with an IC 5 0 of 20 pM, where previous closely
`related analogues were devoid of any CYP inhibitory
`activity. Preparation of the bridgehead-hydroxylated
`analogue 26 gave a compound with a virtually identical
`in vitro (Ki = 0.6 nM) and ex vivo (87% inhibition of
`plasma DPP-IV at 0.5 and 4 h) profile, a slow rat liver
`microsomal turnover rate, no CYP3A4 inhibition up to
`100 pM, and good oral exposure (F = 75%, tl/2 = 2.1 h).
`Two other substituted adamantyl-derived compounds
`were also synthesized and investigated. Dihydroxyada-
`mantyl compound 28, while still reasonably active in
`the ex vivo assay, exhibited extremely high aqueous
`solubility but exhibited low oral exposure in rats,
`presumably resulting from very poor absorption. Though
`fluoroadamantyl compound 30 was also effective ex vivo,
`it exhibited very low oral exposure and had a rat liver
`microsomal turnover rate indicative of extensive me-
`tabolism, similar to that of compound 23. Due to its
`exceptional plasma inhibitory potency and pharmaco-
`dynamic duration of action in this preliminary ex vivo
`assay (ED5 0 for 26 at 6 h = 0.5 ,imol/kg vs ED5 0 for 16d
`at 6 h = 11 pmol/kg, Table 2, compound 26 was chosen
`for further study in acute efficacy models.
`Oral Glucose Tolerance in Zuckerfafa Rats.
`Zuckert f" rats are a well-established genetically modi-
`fled rodent model of obesity-induced insulin resistance 26
`and provide a background to measure the effects of
`DPP-IV inhibitors in a prediabetic animal.2 7 The
`nutrient-induced incretin secretion component of the
`GLP-1-dependent mechanism makes this a suitable
`model with which to study postprandial glucose excur-
`sions after administration of an oral glucose tolerance
`test (oGTT). DPP-IV inhibitor 26 was chosen for futher
`study in this animal model by virtue of its highly potent
`effects in vitro and ex vivo. Compound 26 was admin-
`istered orally to Zuckerfait
`rats at 0.5 h pnor to oGTT,
`consisting of a glucose challenge (2.0 g/kg), followed by
`
`300
`
`-3,
`
`qehic. 0.3
`
`1
`
`3.
`
`Figure 1. Effects of inhibitor 26 dosed at 0.3, 1, and 3 umoli
`kg po versus vehicle control on plasma glucose clearance after
`an oGTT given 4h postdose in 5ucker'
`[a rats.
`
`blood sampling at intervals over the next 2 h for plasma
`glucose measurenents (data not shown). Maximal re-
`sponses in glucose excursion in this model were associ-
`ated with plasma DPP-IV inhibition of approximately
`60% vs control, and no additional antihvperglycemic
`effects were seen at higher percent inhibition On the
`basis of these preliminary findings at a single dose,
`compound 26 was further evaluated in the Zuckerftuit
`rat model with an oGTT performed 4 h after oral
`administration of test compound in a dose-response
`format. Postprandial plasma glucose and insulin levels
`were again measured at intervals over 2 h following the
`glucose challenge. Compound 26 was highly effective at
`eliciting marked dose-dependent enhancements in glu-
`the dose range 0.3-3 ymol/kg
`cose clearance
`in
`(0.13-1.3 mg/kg) in this model relative to controls
`(Figure 1).
`Oral Glucose Tolerance in ob/ob Mice. Evidence
`from both inhibitor studies and knock-out animals
`support that the mouse is also a suitable species in
`which to study the effects of DPP-IV inhibition on
`glucose clearance and insulin potentiation. 2 s To this end,
`the effects of compound 26 on glucose clearance and
`enhancement of insulin secretion was studied in the
`ob/ob mouse. In this model the oGTT was performed at
`1 h after oral administration of 26 at 1, 3, or 10 ymoll
`kg (Figure 2). The data show that compound 26 dose-
`dependently elevated plasma insulin significantly at
`15 min post-oGTT, with concomitant improvement in
`the glucose clearance curves at 60 min post-oGTT.
`Compound 26 exhibited robust glucose-lowering ef-
`fects in a dose-relational manner in the Zuckert Ir rat
`oGTT model, even when the glucose challenge was
`administered 4 h postdose of compound. Similarly
`outstanding efficacy was observed in reducing postpran-
`dial glucose AUC in ob lob mice. This compound also
`proved quite effective in elevating insulin levels after
`an oGTT
`in ob/ob mice, further demonstrating the
`effectiveness of potentiating GLP-1-induced insulin
`secretion as a key component mediating the antihyper-
`glycemic actions of this potent DPP-IV inhibitor. It is
`anticipated that compound 26, given its extended phar-
`macodynamic response, will be amenable to once daily
`dosing in humans.
`
`Conclusion
`A series of f-quaternary cycloalkylglycine amino acid
`residues were incorporated into our previously disclosed
`4,5-mothanoprolinenitrile scaffold, and many of these
`compounds showed potent DPP-IV inhibitory activity.
`Several compounds containing a vinyl functionality also
`
`Page 5 of 13
`
`

`
`5030 Journal of Medicinal Chemistry, 2005, Vol. 48, No. 15
`
`Augeri et al.
`
`~60C
`-.-
`--.-v.--. 261,06510
`---. 3.---..
`0 0,ooASo
`26 0~m51~
`
`1506
`
`1400i),0
`
`-.50
`
`II
`
`Figure 2. Effects of inhibitor 26 dosed at 1, 3, and 10 pmol/kg po versus vehicle control on plasma insulin (left panel) and
`plasma glucose (right panel) after an oGTT in oblob mice. Compound 26 significantly lowered plasma glucose levels
`(vs vehicle) at the 60 min time point at 3 and 10 jmol/kg (p < 0.05) and significantly increased plasma insulin levels (vs vehicle)
`at the 15 min time point at 10 umol/kg (p < 0.05).
`
`exhibited extended duration of action in an ex vivo
`plasma DPP-IV inhibition model in normal rats relative
`to closely related analogues lacking this moiety. These
`analogues, however, also showed markedly reduced
`systemic exposure after oral dosing and rapid rat liver
`microsomal turnover rates where related small alkyl-
`substituted analogues did not. Efforts to define the role
`of suspected metabolites resulted in the synthesis of
`several hydroxymethyleycloalkyl-based analogues that
`maintained in vitro and, for some, in vivo activity. These
`hydroxymethyl analogues also displayed favorable phar-
`macokinetic properties with a tighter correlation of
`pharmacokinctics to pharmacodynamics. Analogously,
`hydroxylation of a similarly disposed adamantylglycme-
`based inhibitor yielded a compound (26) with in vivo
`potency and duration of action superior to that of any
`compound from this series. Consequently, this com-
`pound was chosen for development and is currently
`under clinical investigation for the treatment of type 2
`diabetes. The basis for the enhanced efficacy observed
`for the present compounds in animal models relative
`to other agents may be due to contributions from
`multiple factors, including exquisite enzyme inhibitory
`potency and compound distribution to the tissue com-
`partment potentially critical for maximal antihyper-
`glycemic effects (V_ = 5.2 L/kg for compound 26 in the
`rat). Further studies are underway in these laboratores
`to more fully understand and quantitatively character-
`ize the physicochemical basis for the observed findings.
`
`Experimental Section
`All reactions were carried out under a static atmosphere of
`argon or nitrogen and stirred magnetically unless otherwise
`noted. All reagents used were of commercial quality and were
`obtained from Aldrich Chemical Co., Sigma Chemical Co.,
`Lancaster Chemical Co__ or Acros Chemical Co- 'H (400 MHz)
`and 53C (100 MHz) NMR spectra were recorded on a JEOL
`GSX400 spectrometer using Me 4Si as an internal standard
`unless otherwise noted. 'H (500 MHz) and "C (125 MHz) NMR
`spectra were recorded on a JEOL JNM-ECPoO spectrometer.
`Chemical shifts are given in parts per million (ppm) downfield
`from internal reference tetramethylsilane
`in 5-units, and
`coupling constants (J-values) are given in hertz (Hz). Selected
`data are reported in the following manner: chemical shift,
`multiplicity, coupling constants, and assignment. All reactions
`were carried out using commercially available anhydrous
`solvents from Aldrich Chemical Co. or EM Science Chemical
`Co. unless otherwise noted. All flash chromatographic separa-
`tions were performed using E. Merck silica gel (particle size,
`0.040-0.063 mm). Reactions were monitored by TLC using
`0 25 mm E. Merck silica gel gel plates (60 F2 51) and were
`visualized with UV light, with 5% phosphomolybdic acid in
`
`95% EtOH, or by a sequential treatment with I N HCI/MeOH
`followed by ninhydrin staining. LCIMS data were recorded on
`a Shimadzu LC-1OAT equipped with a SIL-10A injector, a
`SPD-IOAV detector, normally operating at 220 am, and
`interfaced with a Micromass ZMD mass spectrometer. LCIMS
`or HPLC retention times, unless otherwise noted, are reported
`using a Phenomenex Luna C-18 4.6 mm x 50 mm column
`eluted with a 4 min gradient from 0 to 100% B, where A =
`10% MeOH-90% H2 0-0.1% TFA and B = 90% MeOH-10%
`H 2 0-0.1% TFA. All solvents were removed by rotary evapora-
`tion under vacuum using a standard rotovap equipped with a
`dry ice condenser- All filtrations were performed with a
`vacuum unless otherwise noted.
`General Method A. Peptide Coupling to Enantiomeri-
`eally Pure L-cis-4,5-Methanoprolinamide 5, Amide De-
`hydration, and Deprotection. Methanoprolinamide 5 was
`coupled to a variety of racemic quaternary protected amino
`acids using HOBT/EDC in DMF at room temperature to give
`a D/L mixture of diastereomers at the N-terminal amino acid.
`The desired L-diastercomer was most often chromatographi-
`cally isolated as the N-Boc-protected nitrile, obtained by
`treatment of amide 7 with POCI/imidazole in pyridine at
`'C. The final target compounds Sa-g were obtained by
`-20
`deproection using TFA in CH 2 C12.
`(S)-2-(l-Ethenylcyclopent-1-yl)glycine-L-cis-4,5-metha-
`noprolinamide (7d). 4,5-Methanoprolinamide 5 (877 mg,
`3.65 mmol ) and N-Boc cyclopentylvinylamino acid Gd (1.13 g,
`4.20 mmol), described in general method B, were dissolved in
`20 mL of DMF and cooled to 0 'C and to this mixture were
`added EDAC (1.62 g, 8.4 mmol), HOET hydrate (2.54 g,
`12.6 mmol), and TEA (1.27 g, 12.6 mmol). The reaction was
`allowed
`to warm to room temperature and stirred for 24 h.
`The reaction mixture was taken up in 100 mL of EtOAc,
`washed with H 20 (3 x 20 mL), dried (NaSO4], and purified
`by flash chromatography (100% EtOAc) to give 1.38 g (86%)
`1H NMR (500 MHz,
`of 7d as a mixture of diastereomers
`CDC13) 1.02-108 (m, 2H), 143 (s, 9H), 1.54-1.80 (in, 7H),
`184-L94 (m, 2H), 236 (dd, J - 13.6, 2.6, 1H), 258 (m, 1H),
`3.90
`(in,
`(d, J = 9.7,
`1H), 4.63
`1H), 5.04
`(dd,
`J= 10.5, 2.2, 1H), 514 (d, J= 17.6, 1H), 5.23 (d, J= 11, 1H),
`5.97 (dd, J = 17.6, 11, 1H,, MS niz 378 [M + H]
`.
`(S)-2-(1-Ethenylcyclopent-1-yl)glycine-L-cis-4,5-metha-
`noprolinenitrile (8d). Diastereomeric amide 7d (68 mg,
`0.18 mmol, 1 equiv) and imidazole (26 mg, 0.38 mmol, 2.1
`equiv) were dissolved in 2 mL of pyridine at -30
`'C, to which
`POC 3 (0.070 mL, 0.739 mmol, 4.10 cquiv) was added. After
`stirring at -30
`'C for 40 min, the solvent was removed and
`the residue thoroughly dried under vacuum. Purification by
`flash chromatography (10% EtOAclCH2C 2) afforded the de-
`sired slower eluting diastereomerically pure N-Buc-protected
`mtrile as a white solid (34 mg, 0.0946 mmol, 53%): 1H NMR
`(500 MHz, CDC 3 ) L02-1.07 (m, 2H), 142 (s, 9H), 1.55-1.75
`(m, 7H), 1.83-1.93 (in, 2H), 2.36 (dd, J = 13.6, 2.6, IH), 2.58
`(m, iH), 3.90 (in, 1H), 4.62 (d, J= 9.2, 1H), 5.03 (dd, J= 10.5,
`2.2, 1H), 513 (d, J= 17,6, 1H), 5,23 (d, J= 11, 1H1), 5 96 (dd,
`J= 17.6, 11, 1H); MS rm/z 360 [M + H]. The N-Boc protected
`
`Page 6 of 13
`
`

`
`Discovery and Preclinical Profil

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