throbber
6450
`
`J. Med. Chem. 2007, 50, 6450–6453
`
`8-(3-(R)-Aminopiperidin-1-yl)-7-but-2-ynyl-3-
`methyl-1-(4-methyl-quinazolin-2-ylmethyl)-
`3,7-dihydropurine-2,6-dione (BI 1356), a
`Highly Potent, Selective, Long-Acting, and
`Orally Bioavailable DPP-4 Inhibitor for the
`Treatment of Type 2 Diabetes†
`
`Matthias Eckhardt,*,# Elke Langkopf,*,# Michael Mark,§
`Moh Tadayyon,§ Leo Thomas,§ Herbert Nar,|
`Waldemar Pfrengle,¶ Brian Guth,‡ Ralf Lotz,‡ Peter Sieger,‡
`Holger Fuchs,⊥ and Frank Himmelsbach*,#
`
`Boehringer Ingelheim Pharma GmbH & Co. KG,
`88400 Biberach, Germany
`
`ReceiVed October 11, 2007
`
`Abstract: A new chemical class of potent DPP-4 inhibitors structurally
`derived from the xanthine scaffold for the treatment of type 2 diabetes
`has been discovered and evaluated. Systematic structural variations have
`led to 1 (BI 1356), a highly potent, selective, long-acting, and orally
`active DPP-4 inhibitor that shows considerable blood glucose lowering
`in different animal species. 1 is currently undergoing clinical phase
`IIb trials and holds the potential for once-daily treatment of type 2
`diabetics.
`
`Chart 1
`
`Scheme 1. Syntheses of Compounds 5 and 6 a
`
`a Reagents: (a) PhCH2Cl, Me2CCHCH2Br, or MeCCCH2Br, iPr2NEt,
`DMF, room temp, 4a 75%, 4b 91%, 4c 84%; (b) piperazine (excess), THF
`or MeCN, reflux, 5a 64%, 5b 39%, 5c 98%; (c) 3-aminopiperidine · 2HCl,
`K2CO3, MeCN, 70 °C, 58%.
`
`Type 2 diabetes is establishing itself as an epidemic of the
`21st century with an estimated 5% of the adult world population
`suffering from the disease.1 The number of deaths attributable
`to diabetes is steadily growing, currently estimated at 3.8 million
`cases each year, reflecting the insufficient glycemic control
`achieved with currently available treatments. Therefore, more
`effective therapeutics for glycemic control are badly needed.
`DPP-4a is a protease that specifically cleaves dipeptides from
`proteins and oligopeptides after a penultimate N-terminal proline
`or alanine.2 DPP-4 is involved in the degradation of a number
`of neuropeptides, peptide hormones, and cytokines, including
`the incretins GLP-1 and GIP.3 GLP-1 and GIP are released from
`the gut in response to food intake and exert a potent glucose-
`dependent insulinotropic action and thereby contribute to the
`maintenance of postmeal glycemic control.4 In addition, they
`exhibit beneficial effects on pancreatic (cid:1) cells.5 Other effects
`that have been described for GLP-1 are an inhibition of glucagon
`release from pancreatic R cells, a reduction of food intake, and
`a retardation of gastric emptying.6 Consequently, inhibiting
`DPP-4 prolongs the action of GLP-1 and GIP, which in turn
`
`improves glucose homeostasis with a low risk of hypoglycemia
`and potential for disease modification. Indeed, clinical trials
`involving diabetic patients have shown improved glucose control
`by administering DPP-4 inhibitors,
`thus demonstrating the
`benefit of this promising new class of antidiabetics.7 Intense
`research in the DPP-4 field has resulted in the launch of one
`inhibitor and the advancement of others into preregistration/
`phase III.8
`Herein, we report the discovery of the novel, potent, and
`selective DPP-4 inhibitor 1 (BI 1356)9 originating from the class
`of xanthines (Chart 1).
`Compound 2, discovered through a high-throughput screening
`campaign that involved about 500 000 compounds and showing
`promising inhibitory activity in the low micromolar range, was
`our starting point in the search for an effective DPP-4 inhibitor.10
`Systematic structural modifications on the xanthine scaffold were
`carried out to study the structure–activity relationship and
`optimize inhibition of DPP-4. Various substituents on the
`xanthine core were scrutinized in a broad manner while a
`stronger emphasis was put on the residues at N-1, N-7, and
`C-8 because of their higher impact on activity. Common
`synthetic starting materials for the study were 8-chlorotheo-
`† Compound 1 has been deposited into the Protein Data Bank: PDB code
`phylline 3 and 8-bromoxanthine 7 (Schemes 1 and 2). Derivative
`2RGU.
`* To whom correspondence should be addressed. Phone: +49 7351
`syntheses starting from theophylline 3 were routinely carried
`54-5539/5013 or 7657. Fax: +49 7351 54-5181. E-mail: for M.E.,
`out first with alkylation of N-7 followed by nucleophilic
`matthias.eckhardt@boehringer-ingelheim.com; for E.L., elke.langkopf@
`displacement of the chlorine at C-8 by a diamine. The
`boehringer-ingelheim.com;
`for F.H.,
`frank.himmelsbach@boehringer-
`alkylations were conducted using an appropriate alkyl halide
`ingelheim.com.
`in the presence of a base. Displacement of the chlorine was
`# Department of Chemical Research.
`§ Department of Metabolic Diseases Research.
`performed employing an excess of the diamine to be introduced
`| Department of Lead Discovery.
`or the diamine in combination with potassium carbonate. In the
`¶ Department of Chemical Development.
`case of an asymmetric diamine, the nitrogen not to be reacted
`‡ Department of Drug Discovery Support.
`was preferentially embedded in tert-butyl carbamate, with
`⊥ Department of Drug Metabolism and Pharmacokinetics.
`subsequent release by treatment with acid. Employing 7 as the
`aAbbreviations: DPP-4, dipeptidyl peptidase 4; GLP-1, glucagon-like
`starting material followed a similar reaction sequence to
`peptide 1; GIP, glucose-dependent insulinotropic peptide.
`10.1021/jm701280z CCC: $37.00  2007 American Chemical Society
`Published on Web 12/01/2007
`
`Mylan EX 1005, Page 1
`
`

`
`Letters
`
`Journal of Medicinal Chemistry, 2007, Vol. 50, No. 26 6451
`
`Scheme 2. Syntheses of Compounds 6 and 1 a
`
`Table 1. DPP-4 Inhibitory Activity of Xanthines
`
`compd
`2
`5a
`5b
`5c
`6
`6ac
`6bc
`6ad
`6ae
`6af
`
`R1
`
`R7
`H
`CH2Ph
`CH2Ph
`Me
`CH2CHCMe2 Me
`CH2CCMe
`Me
`CH2Ph
`Me
`CH2CHCMe2 Me
`CH2CCMe
`Me
`CH2CHCMe2
`CH2Ph
`CH2CHCMe2
`(CH2)2Ph
`CH2CHCMe2
`CH2COPh
`
`X/Y
`NH/H
`NH/H
`NH/H
`NH/H
`CH2/NH2
`CH2/NH2
`CH2/NH2
`CH2/NH2
`CH2/NH2
`CH2/NH2
`
`DPP-4 IC50 (nM)
`3900
`2800
`580
`200
`82
`35
`88
`284
`56
`5
`
`Table 2. Effects of 8-(3-Aminopiperidin-1-yl)xanthines on DPP-4,
`hERG, and M1
`
`M1 IC50
`hERG
`DPP-4 IC50
`R1
`R7
`(%)a
`(nM)
`(nM)
`compd
`f
`(R)-6af
`25
`31
`6
`CH2CHCMe2
`(S)-6af
`f
`CH2CHCMe2
`50
`23
`3
`(R)-6ag
`g
`CH2CHCMe2
`5
`51
`4
`(S)-6ag
`g
`CH2CHCMe2
`26
`51
`2
`88b
`(R)-6bf
`f
`CH2CCMe
`6161
`4
`88b
`(S)-6bf
`f
`CH2CCMe
`1129
`9
`(R)-6bh
`h
`CH2CCMe
`518
`51
`8
`(R)-6bi
`i
`CH2CCMe
`1174
`88
`1
`(R)-6bj
`j
`CH2CCMe
`430
`78
`3
`1 [(R)]
`k
`CH2CCMe
`295
`97
`1
`a hERG current remaining at a test concentration of 1 µM. b Value of
`the racemic compound.
`
`Table 3. Selected Basic PK Data of Compound 1 in Rat and
`Cynomolgus Monkey
`
`species
`
`MRTtot,oral
`CL
`Foral
`t1/2,
`Vss
`(%)
`oral (h)
`(h)
`(L/kg)
`((mL/min)/kg)
`rata
`50.7
`35.9
`14.3
`5.4
`37.3
`monkeyb
`50.1
`41.4
`17.4
`15.8
`15.8
`a 5 mg/kg oral and intravenous dose. b 5 mg/kg oral and 1.5 mg/kg
`intravenous dose.
`
`enantiopure compounds bearing a dimethylallyl group on N-7.
`Unfortunately, both compounds show an unacceptably high
`inhibition of the hERG channel and affinity for the muscarinic
`receptor M1. By variation of the residue on N-1 based on the
`dimethylallylated xanthine, both side effects could not con-
`comitantly be reduced sufficiently while retaining the desired
`high inhibitory activity. These shortcomings also extended to
`compounds with bicyclic aryl- or heteroarylmethyl residues
`attached to N-1 such as in 6ag that were otherwise highly potent
`DPP-4 inhibitors. Switching to the 7-butynyl derivatized scaffold
`proved to be a very effective measure to essentially abolish
`hERG channel inhibition and reduce interaction with receptor
`M1 to a high degree. Various phenacyl and bicyclic aryl- and
`heteroarylmethyl residues attached to N-1 of this scaffold
`exhibited high DPP-4 inhibition with no perturbing interactions
`with the hERG channel or M1 receptor. Curiously, in the
`7-butynyl series the (R)-configured compounds were signifi-
`cantly more active in all cases assayed, contrasting with the
`results obtained from the 7-dimethylallyl series.
`
`a Reagents: (a) Me2CCHCH2Br or MeCCCH2Br, iPr2NEt, DMF, room
`temp, 8a 86%, 8b 86%; (b) R1-Hal, K2CO3, DMF, room temp, 77–98%;
`(c) (I) racemic, (R)- or (S)-3-Boc-aminopiperidine, K2CO3, DMF, 75 °C,
`67–96%; (II) TFA, DCM or HCl, iPrOH, 69–98%.
`b Compound bears chlorine instead of bromine at C-8.
`
`assemble the fully substituted xanthines. The preferred order
`of attachment of the three substituents was N-7 followed by
`N-1 and C-8, though the order of introduction of the last two
`substituents could be reversed to streamline the examination of
`the residue on N-1. Accordingly, treatment of xanthine 7 with
`different alkyl halides was conducted in the presence of a mild
`base, such as triethylamine or ethyldiisopropylamine, furnishing
`the N-7 derivatized xanthine 8 selectively; competing N-1
`alkylation was usually not observed. The next step, alkylation
`at N-1, was regularly performed using the stronger base
`potassium carbonate leading to 9. The ensuing nucleophilic
`substitution of the bromine at C-8 in xanthine 9 for the diamine,
`3-aminopiperidine, was preferably carried out using the N-tert-
`butyloxycarbonyl protected 3-aminopiperidine in the presence
`of potassium carbonate. The synthesis was concluded by the
`release of the amino functionality from its protection by
`treatment with acid. The enantiomerically pure compounds were
`obtained by reaction with the commercial enantiopure N-tert-
`butyloxycarbonyl protected 3-aminopiperidines.
`The DPP-4 inhibitory activity of the compounds was tested
`using a preparation of human DPP-4 derived from Caco-2 cells.
`The results obtained for the racemic xanthine derivatives in
`Schemes 1 and 2 are compiled in Table 1. Variations at N-7 or
`C-8 of 5a yielded 5b, 5c, and 6, all showing significantly
`increased DPP-4 inhibition. In particular, replacement of the
`piperazine at C-8 for the 3-aminopiperidine in 5a resulted in a
`tremendous increase in potency. Combining the structural
`features of these more potent compounds led to compounds 6ac
`and 6bc, which exhibited approximately a 100-fold and 50-
`fold higher inhibitory activity, respectively, than the original 2.
`Further optimization at N-1 based on the structure of 6ac
`resulted in an additional increase of potency when attaching
`the phenacyl group (6af), while the benzylated and the phen-
`ethylated 6ad and 6ae showed inferior activity.
`Further profiling was conducted using the pure enantiomers
`(R)-6af and (S)-6af (Table 2). DPP-4 inhibition of the (S)-
`configured compound was about twice as potent as of the (R)-
`configured one, a trend that was generally observed for
`
`Mylan EX 1005, Page 2
`
`

`
`6452 Journal of Medicinal Chemistry, 2007, Vol. 50, No. 26
`
`Letters
`
`Figure 1. Compound 1 (light-blue carbon atoms) bound to DPP-4.
`Active site residues Ser630, His740, and Asp708 are shown in orange.
`Three hydrogen bonds (shown by black dashes) are formed by the
`amino function on the piperidine ring with acceptor groups on the
`protein Glu205, Glu206, and Tyr662. A fourth hydrogen bond is formed
`between the C-6 carbonyl of the xanthine scaffold and the backbone
`amide of residue Tyr631. Aromatic stacking interactions are formed
`between the xanthine ring system and Tyr547 as well as between the
`quinazoline ring and Trp629.
`
`Figure 2. Inhibition of plasma DPP-4 activity after oral administration
`of 1 at 1 mg/kg in Wistar rats, Beagle dogs, and Rhesus monkeys.
`Data are represented as the mean ( SEM (n ) 5 for rats, n ) 3 for
`dogs and monkeys).
`
`Compound 1, showing potent DPP-4 inhibition in vitro and
`a low affinity for hERG channel and M1 receptor, has been
`further examined. A favorable crystalline modification of the
`free base of 1 has been produced that is characterized by a high
`melting point (202 °C) and high aqueous solubility at physi-
`ological pH value (pH 7.4, >5 g/L). Compound 1 displays a
`log D of 0.4 at pH 7.4 and pK a of 1.9 and 8.6 corresponding
`to the protonation of the quinazoline and the primary amino
`group, respectively.
`The X-ray crystal structure of 1 in complex with human
`DPP-4 allows one to depict the main interactions of the inhibitor
`within the enzyme active site and to rationalize the observed
`SAR (Figure 1). The aminopiperidine substituent at C-8 of the
`xanthine scaffold occupies the S2 subsite. Its primary amine
`forms a network of charge-reinforced hydrogen bonds to
`Glu205, Glu206, and Tyr662, amino acid residues that constitute
`the recognition site for the amino terminus of peptide substrates
`of DPP-4. The butynyl substituent at N-7 occupies the hydro-
`phobic S1 pocket of the enzyme. The xanthine moiety is
`positioned such that its uracil moiety lies on top of Tyr547,
`forming aromatic π-stacking interactions with the phenol of
`Tyr547. Thereby, the side chain of Tyr547 is pushed from its
`relaxed position in the uncomplexed enzyme.11 A similar
`
`Figure 3. Effect of 1 on plasma glucose levels in an oral glucose
`tolerance test in db/db mice (top). Compound or vehicle was admin-
`istered 45 min before an oral glucose load. Reactive plasma glucose
`AUC was calculated from 0 to 120 min (middle). Inhibition of plasma
`DPP-4 activity was measured 30 min after the glucose load at the peak
`of the glucose excursion. Data are represented as the mean ( SEM (n
`) 7/group).
`
`conformational change has been reported for related xanthine
`based inhibitors and for
`inhibitors from other structural
`classes.12–14 The C-6 carbonyl function of the xanthine scaffold
`forms a hydrogen bond to the backbone NH of Tyr631. Finally,
`the quinazoline subsituent at N-1 is placed on a hydrophobic
`surface patch of the protein and interacts with Trp629 by
`π-stacking its phenyl ring with the pyrrol ring of the amino
`acid side chain.
`The observed boost in affinity upon the introduction of the
`aminopiperidine group at C-8 is due to the very intimate
`interaction of the positively charged terminal ammonium that
`can form three strong hydrogen bonds with the protein. Further,
`the observed bound chair conformation of the piperidine ring
`is a low-energy conformation. The original piperazine derivative
`2 can only form two hydrogen bonds and needs to adopt an
`unfavorable conformation upon binding.12
`The strong DPP-4 inhibition of 1 has been confirmed in
`various species in vitro and in vivo. In male Wistar rats, Beagle
`dogs, and Rhesus monkeys, xanthine 1 proved to be a highly
`efficacious, long-lasting, and potent DPP-4 inhibitor providing
`>70% inhibition for >7 h for all three species after oral
`administration of 1 mg/kg (Figure 2).
`Pharmacokinetic parameters including oral bioavailability,
`clearance, mean residence time, and volume of distribution of
`1 in rat and Cynomolgus monkey are summarized in Table 3.
`The long terminal half-life and high volume of distribution are
`
`Mylan EX 1005, Page 3
`
`

`
`Letters
`
`Journal of Medicinal Chemistry, 2007, Vol. 50, No. 26 6453
`
`key characteristics of 1 and, in combination with its high potency
`and good oral bioavailability, are thought to contribute to the
`strong and long-lasting inhibitory effect on DPP-4 observed in
`vivo.
`Compound 1 was further characterized in vivo in diabetic
`mice (Figure 3). Single oral administration of 1 to db/db mice
`45 min prior to an oral glucose tolerance test reduced plasma
`glucose excursion in a dose-dependent manner from 0.1 mg/kg
`(15% inhibition) to 1 mg/kg (66% inhibition). The improvement
`of oral glucose tolerance correlated with the DPP-4 activity in
`plasma, which was inhibited by 76% with the 1 mg/kg dose 30
`min after the glucose load was administered.
`Compound 1 exhibits no interaction with CYP-450 enzymes
`up to 50 µM. Because inhibition of DPP-8 and DPP-9, which
`are closely related to DPP-4, has been associated with toxicities
`in animals, it is important to note that 1 displays a more than
`10000-fold selectivity against both of these enzymes.15
`In summary, a new chemical class of highly potent DPP-4
`inhibitors structurally based on the xanthine scaffold has been
`discovered. The 3-aminopiperidine attached to C-8 proved to
`be a crucial constituent for high inhibitory activity, and but-2-
`ynyl on N-7 was essential to eliminate interaction with the hERG
`channel and M1 receptor. Further optimization led to 1 bearing
`a quinazolin-2-ylmethyl at N-1. 1 represents a highly potent,
`selective, and long-acting DPP-4 inhibitor of a novel chemotype
`that shows promise for once-daily treatment of type 2 diabetic
`patients. Compound 1 is currently undergoing clinical phase
`IIb trials.
`
`Acknowledgment. The authors are indebted to their associ-
`ates for their dedicated and excellent technical assistance.
`
`Supporting Information Available: Experimental details, ana-
`lytical data of the compounds, and X-ray crystallograpic data of 1.
`This material is available free of charge via the Internet at http://
`pubs.acs.org.
`
`References
`(1) http://www.eatlas.idf.org.
`(2) (a) Lambeir, A. M.; Durinx, C.; Scharpe, S.; de Meester, I. Dipeptidyl-
`peptidase IV from bench to bedside: an update on structural properties,
`functions, and clinical aspects of the enzyme DPP IV. Crit. ReV. Clin.
`Lab. Sci. 2003, 40, 209–294. (b) Augustyns, K.; Van der Veken, P.;
`Senten, K.; Haemers, A. The therapeutic potential of inhibitors of
`dipeptidyl peptidase IV (DPP IV) and related proline specific dipeptidyl
`aminopeptidases. Curr. Med. Chem. 2005, 12, 971–998.
`(3) (a) Mentlein, R.; Gallwitz, B.; Schmidt, W. E. Dipeptidyl-peptidase
`IV hydrolyzes gastric inhibitory polypeptide, glucagon-like peptide-1
`(7-36)amide, peptide histidine methionine and is responsible for their
`degradation in human serum. Eur. J. Biochem. 1993, 214, 829–835.
`(b) Kieffer, T. J.; McIntosh, C. H. S.; Pederson, R. A. Degradation of
`glucose-dependant insulinotropic polypeptide and truncated glucagon-
`like peptide-1 in vitro and in vivo by dipeptidyl peptidase IV.
`Endocrinology 1995, 136, 3585–3596.
`(4) (a) Thorens, B. Glucagon-like peptide-1 and control of insulin
`secretion. Diabetes Metab. 1995, 21, 311–318. (b) Meier, J. J.; Nauck,
`M. A.; Schmidt, W. E.; Gallwitz, B. Gastric inhibitory polypeptide:
`the neglected incretin revisited. Regul. Pept. 2002, 107, 1–13.
`(5) (a) Pospisilik, J. A.; Stafford, S. G.; Demuth, H.-U.; Brownsey, R.;
`Parkhouse, W.; Finegood, D. T.; McIntosh, C. H. S.; Pederson, R. A.
`Long-term treatment with the dipeptidyl peptidase IV inhibitor P32/
`98 causes sustained improvements in glucose tolerance,
`insulin
`sensitivity, hyperinsulinemia, and (cid:1)-cell glucose responsiveness in
`VDF (fa/fa) Zucker rats. Diabetes 2002, 51, 943–950. (b) Drucker,
`D. J. Glucagon-like peptide-1 and the islet (cid:1)-cell: augmentation of
`
`cell proliferation and inhibition of apoptosis. Endocrinology 2003, 144,
`5145–5148.
`(6) (a) Lim, G. E.; Brubaker, P. L. Glucagon-like peptide 1 secretion by
`the L-cell. The view from within. Diabetes 2006, 55, S70–S77. (b)
`Murphy, K. G.; Dhillo, W. S.; Bloom, S. R. Gut peptides in the
`regulation of food intake and energy homeostasis. Endocrine ReV.
`2006, 27, 719–727.
`(7) (a) Idris, I.; Donnelly, R. Dipeptidyl peptidase-IV inhibitors: a major
`new class of oral antidiabetic drug. Diabetes, Obes. Metab. 2007, 9,
`153–165. (b) Barnett, A. DPP-4 inhibitors and their potential role in
`the management of type 2 diabetes. Int. J. Clin. Pract. 2006, 60, 1454–
`1470.
`(8) (a) Villhauer, E. B.; Brinkman, J. A.; Naderi, G. B.; Burkey, B. F.;
`Dunning, B. E.; Prasad, K.; Mangold, B. L.; Russel, M. E.; Hughes,
`T. E. 1-[[(3-Hydroxy-1-adamantyl)amino]acetyl]-2-cyano-(S)-pyrro-
`lidine: a potent, selective, and orally bioavailable dipeptidyl peptidase
`IV inhibitor with antihyperglycemic properties. J. Med. Chem. 2003,
`46, 2774–2789. (b) Kim, D.; Wang, L.; Beconi, M.; Eiermann, G. J.;
`Fisher, M. H.; He, H.; Hickey, G. J.; Kowalchick, J. E.; Leiting, B.;
`Lyons, K.; Marsilio, F.; McCann, M. E.; Patel, R. A.; Petrov, A.;
`Scapin, G.; Patel, S. B.; Roy, R. S.; Wu, J. K.; Wyvratt, M. J.; Zhang,
`B. B.; Zhu, L.; Thornberry, N. A.; Weber, A. E. (2R)-4-Oxo-4-[3-
`(trifluoromethyl)-5,6-dihydro[1,2,4]triazolo[4,3-R]pyrazin-7(8H)-yl]-
`1-(2,4,5-trifluorophenyl)butan-2-amine: a potent, orally active dipep-
`tidyl peptidase IV inhibitor for the treatment of type 2 diabetes. J. Med.
`Chem. 2005, 48, 141–151. (c) Augeri, D. J.; Robl, J. A.; Betebenner,
`D. A.; Magnin, D. R.; Khanna, A.; Robertson, J. G.; Wang, A.;
`Simpkins, L. M.; Taunk, P.; Huang, Q.; Han, S.-P.; Abboa-Offei, B.;
`Cap, M.; Xin, L.; Tao, L.; Tozzo, E.; Welzel, G. E.; Egan, D. M.;
`Marcinkeviciene, J.; Chang, S. Y.; Biller, S. A.; Kirby, M. S.; Parker,
`R. A.; Hamann, L. G. 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. J. Med.
`Chem. 2005, 48, 5025–5037. (d) Feng, J.; Zhang, Z.; Wallace, M. B.;
`Strafford, J. A.; Kaldor, S. W.; Kassel, D. B.; Navre, M.; Shi, L.;
`Skene, R. J.; Asakawa, T.; Takeuchi, K.; Xu, R.; Webb, D. R.;
`Gwaltney, S. L. Discovery of alogliptin: a potent, selective, bioavail-
`able, and efficacious inhibitor of dipeptidyl peptidase IV. J. Med.
`Chem. 2007, 50, 2297–2300.
`(9) Himmelsbach, F.; Langkopf, E.; Eckhardt, M.; Mark, M.; Maier, R.;
`Lotz, R.; Tadayyon, M. WO 2004018468, 2004.
`(10) Compound 2 has also been found and further elaborated in the
`following: Kanstrup, A. B.; Christiansen, L. B.; Lundbeck, J. M.; Sams,
`C. K.; Kristiansen, M. WO 2002002560, 2002.
`(11) Thoma, R.; Loeffler, B.; Stihle, M.; Huber, W.; Ruf, A.; Hennig, M.
`Structural basis of proline-specific exopeptidase activity as observed
`in human dipeptidyl peptidase-IV. Structure 2003, 11, 947–959.
`(12) Engel, M.; Hoffmann, T.; Manhart, S.; Heiser, U.; Chambre, S.; Huber,
`R.; Demuth, H.-U.; Bode, W. Rigidity and flexibility of dipeptidyl
`peptidase IV: crystal structures of and docking experiments with DPIV.
`J. Mol. Biol. 2006, 355, 768–783.
`(13) Longenecker, K. L.; Stewart, K. D.; Madar, D. J.; Jakob, C. G.; Fry,
`E. H.; Wilk, S.; Lin, C. W.; Ballaron, S. J.; Stashko, M. A.; Lubben,
`T. H.; Yong, H.; Pireh, D.; Pei, Z.; Basha, F.; Wiedeman, P. E.; von
`Geldern, T. W.; Trevillyan, J. M.; Stoll, V. S. Crystal structures of
`DPP-IV (CD26) from rat kidney exhibit flexible accommodation of
`peptidase-selective inhibitors. Biochemistry 2006, 45, 7474–7482.
`(14) Sheehan, S. M.; Mest, H.-J.; Watson, B. M.; Klimkowski, V. J.; Timm,
`D. E.; Cauvin, A.; Parsons, S. H.; Shi, Q.; Canada, E. J.; Wiley, M. R.;
`Ruehter, G.; Evers, B.; Petersen, S.; Blaszczak, L. C.; Pulley, S. R.;
`Margolis, B. J.; Wishart, G. N.; Renson, B.; Hankotius, D.; Mohr,
`M.; Zechel, J.-C.; Kalbfleisch, J. M.; Dingess-Hammond, E. A.;
`Boelke, A.; Weichert, A. G. Discovery of non-covalent dipeptidyl
`peptidase IV inhibitors which induce a conformational change in the
`active site. Bioorg. Med. Chem. Lett. 2007, 17, 1765–1768.
`(15) Lankas, G. R.; Leiting, B.; Roy, R. S.; Eiermann, G. J.; Beconi, M. G.;
`Biftu, T.; Chan, C.-C.; Edmondson, S.; Feeney, W. P.; He, H.; Ippolito,
`D. E.; Kim, D.; Lyons, K. A.; Ok, H. O.; Patel, R. A.; Petrov, A. N.;
`Pryor, K. A.; Qian, X.; Reigle, L.; Woods, A.; Wu, J. K.; Zaller, D.;
`Zhang, X.; Zhu, L.; Weber, A. E.; Thornberry, N. A. Dipeptidyl
`peptidase IV inhibition for the treatment of type 2 diabetes: potential
`importance of selectivity over dipeptidyl peptidases 8 and 9. Diabetes
`2005, 54, 2988–2994.
`
`JM701280Z
`
`Mylan EX 1005, Page 4

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