throbber
Inhibitors of Dipeptidyl Peptidase 4
`
`Stephen L. Gwaltney, II and Jeffrey A. Stafford
`
`Takeda San Diego, Inc., 10410 Science Center Drive, San Diego, CA 92121
`
`Contents
`1. Introduction
`1.1. Function of DPP4
`1.2. Structure of DPP4
`1.3. Therapeutic significance
`2. Preclinical DPP4 inhibitors
`3. DPP4 inhibitors in clinical development
`4. Alternative indications for DPP4 inhibitors
`References
`
`1. INTRODUCTION
`
`149
`149
`150
`150
`151
`156
`160
`161
`
`Dipeptidyl peptidase 4 (EC 3.4.14.5, DPP-IV, DPP4, CD26) is a ubiquitous serine
`protease that modulates the biological activities of numerous peptides, including
`glucagon-like peptide-1 (GLP-1). GLP-1 plays an important role in the control of
`post-prandial glucose levels by potentiating glucose-stimulated insulin release and
`inhibiting the release of glucagon. Other actions of GLP-1 include delaying gastric
`emptying, inducing satiety and increasing beta cell mass. GLP-1 has shown efficacy
`in diabetics, but suffers from a very short physiological half-life (t1/2 2 min) due to
`DPP4-mediated cleavage of the active peptide (7-36 amide or 7-37) to an inactive
`form (9-36 amide or 9-37). Intense research in the pharmaceutical industry aims to
`discover and develop stable GLP-1 analogs, exogenous agonists of the GLP-1 re-
`ceptor or small-molecule inhibitors of DPP4. This research has been buoyed re-
`cently by positive clinical trial data on GLP-1 analogs and DPP4 inhibitors. The
`field of DPP4 inhibition has been reviewed extensively [1–12]. This review attempts
`to provide an update to the previous ARMC article on DPP4 inhibitors [13] cov-
`ering the primary literature from 2001 through the end of March 2005. It is not the
`intent of the authors to provide another review of the pharmacology of DPP4, but
`to concentrate on the medicinal chemistry in the field.
`
`1.1. Function of DPP4
`
`DPP4 functions as a serine protease and cleaves the amino-terminal dipeptide from
`oligopeptides with a proline or alanine at the penultimate position. Peptides with
`residues other than Pro or Ala at the penultimate position may also be low-affinity
`substrates for DPP4. In contrast, DPP4 is not selective with respect to the N-terminal
`
`ANNUAL REPORTS IN MEDICINAL CHEMISTRY, VOLUME 40
`ISSN: 0065-7743 DOI 10.1016/S0065-7743(05)40010-X
`
`r 2005 Elsevier Inc.
`All rights reserved
`
`MYLAN Ex. 1011, Page 1
`
`

`
`150
`
`S.L. Gwaltney, II and J.A. Stafford
`
`residue [14] and shows little discrimination of various prime-side residues [15,16]. A
`number of biologically important peptides are substrates for DPP4 in vitro [17,18].
`
`1.2. Structure of DPP4
`
`DPP4 is a 110-kDa glycoprotein expressed on the cell surface and widely distributed
`throughout the body. Cleavage of the extracellular portion of DPP4 from the
`22-residue transmembrane section results in a soluble, circulating form of approxi-
`mately 100 kDa. Functional DPP4 is a homodimer, although an active heterodimer
`with fibroblast activation protein has been observed [19]. The consensus sequence for
`DPP4 is G-W-S-Y-G and the catalytic triad is made up of Ser630, Asp708 and
`His740. It has been shown that the glycosylation state of the enzyme is not important
`for enzyme activity, dimerization, and adenosine deaminase binding [20].
`Several groups have reported crystal structures of human DPP4 [15,21–24], and
`one group has reported the structure of porcine DPP4 [25]. These structures show
`the dimeric nature of the enzyme and reveal that the catalytic site is located in a
`cavity between the a/b hydrolase domain and an eight-bladed propeller domain.
`Also revealed is the oxyanion hole, which is composed of the backbone NH of
`Tyr631 and the OH of Tyr47. A co-complex of DPP4 and the inhibitor Val-
`pyrrolidide demonstrates that two glutamates in the active site play an important
`role in substrate binding by forming a salt bridge with the N-terminus of a peptide
`substrate. The pyrrolidine of the inhibitor effectively fills a hydrophobic pocket that
`will only accommodate small residues. This pocket engenders DPP4’s selectivity for
`proline at P1. This work also revealed that two openings in the enzyme may provide
`access to and egress from the catalytic site for some substrates and products [21].
`The importance of Tyr547 in the stabilization of the intermediate oxyanion was
`confirmed through site-directed mutagenesis [26]. Most authors agree that peptides
`enter the larger side opening to access the active site [15]. It has been postulated that
`the dipeptide product is expelled through the narrow b-propeller opening [21,24].
`The co-complex of DPP4 and a compound related to NVP-DPP728 [23] confirms
`that cyanopyrrolidine inhibitors form an imidate with the active site serine, con-
`sistent with a model proposed earlier [27]. Two groups have observed the trapping
`of tetrahedral intermediates in co-complexes of peptides with DPP4 [15,24].
`
`1.3. Therapeutic significance
`
`Relative to wild-type controls, DPP4-deficient mice are resistant to the development
`of obesity and hyperinsulinemia when fed a high-fat diet [28]. DPP4 knockout mice
`also show elevated GLP-1 levels and improved metabolic control. Relative to DPP4
`positive controls, DPP4-deficient Fischer rats show improved glucose tolerance
`following an oral glucose challenge due to enhanced insulin release mediated by
`high levels of active GLP-1 [29,30]. In these studies, the authors note that fasting
`and post-challenge glucose levels in both strains are similar, supporting previous
`assertions that hypoglycemia is unlikely during treatment with DPP4 inhibitors.
`
`MYLAN Ex. 1011, Page 2
`
`

`
`Inhibitors of Dipeptidyl Peptidase 4
`
`151
`
`The use of GLP-1 and its analogs in the treatment of diabetes has been reviewed
`recently [31,32]. It has been shown that DPP4 inhibition prevents the degradation of
`endogenous GLP-1 and glucose-dependent insulinotropic polypeptide (GIP) in dogs,
`thereby preserving the insulinotropic effects of these peptides [33]. In the same study, it
`was noted that total incretin secretion was reduced, suggesting that feedback mech-
`anisms restrict the secretion of incretins when levels of active peptide are elevated. It
`has been demonstrated that agonism of the GLP-1 receptor results in growth and
`differentiation of pancreatic islet beta cells [34–36]. If realized in humans, such an
`effect may result in preservation or restoration of b-cell function in diabetics. In
`human clinical trials, infusion of GLP-1 led to such beneficial effects as decreases in
`post-prandial glucose excursions, increases in post-prandial insulin, reductions in
`HbA1c, weight loss, enhanced insulin sensitivity and improved b-cell function [37,38].
`Administration of the GLP-1 analogs exendin-4, CJC-1131 and NN2211 resulted in
`similar beneficial effects [31,32]. Notably, DPP4 inhibition has been shown to augment
`the insulin secretion effects of not only GLP-1 and GIP, but also pituitary adenylate
`cyclase-activating polypeptide (PACAP) and gastrin-releasing peptide (GRP) [39].
`
`2. PRECLINICAL DPP4 INHIBITORS
`
`Early DPP4 inhibitors closely mimicked DPP4 substrates, as exemplified by valine-
`pyrrolidide (Val-Pyr, 1), P32/98 (2) and FE 999011 (3). A large body of data has
`been reported for these compounds and provided early biological validation for the
`use of DPP4 inhibitors as an approach to the treatment of diabetes.
`
`N
`
`CN
`
`O 3
`
`H2N
`
`S
`
`N
`
`H2N
`
`O
`
`2
`
`N
`
`O 1
`
`H2N
`
`Treatment of six-week-old db/db mice with Val-Pyr resulted in increased endo-
`genous GLP-1 levels, potentiated insulin secretion and improved glucose tolerance;
`however, while the effects on GLP-1 and insulin were maintained in mice at 23
`weeks of age, the improved glucose control was lost [40]. Studies in rats demon-
`strated that combining Val-Pyr with metformin leads to reduced food intake and
`body weight gain, improved glucose tolerance and increases in active plasma GLP-1
`and that these effects are absent or less significant when using either drug as
`monotherapy [41,42]. In related work, treatment of rats with metformin or piog-
`litazone resulted in reduced serum DPP4 activity. Since the authors found that these
`agents are not inhibitors of DPP4 in vitro, they suggested that the effect resulted
`from reduced DPP4 secretion [43].
`Double incretin receptor knockout (DIRKO) mice are genetically altered to
`lack both the GLP-1 receptor and the GIP receptor. A study in these animals with
`
`MYLAN Ex. 1011, Page 3
`
`

`
`152
`
`S.L. Gwaltney, II and J.A. Stafford
`
`Val-Pyr and a structurally unrelated inhibitor, SYR106124, showed that while these
`inhibitors provide improved glucose tolerance and increased insulin levels in wild-
`type and single incretin receptor knock out mice, these effects were lost in the
`DIRKO mice. This result points to the essential nature of the incretin receptors in
`the actions of DPP4 inhibitors [44].
`While inhibitors such as 4 (Ki ¼ 6.03 mM) and 5 (IC50 ¼ 12 mM) are related to
`the cyanopyrrolidine DPP4 inhibitors through the use of the fluoroolefin amide
`isostere, these compounds are only weak inhibitors of the enzyme [45–47].
`
`CN
`
`F
`
`5
`
`NH
`
`H2N
`
`CN
`
`F
`
`4
`
`Several recent papers have examined the effects of long-term treatment with P32/
`98 (2) in rodent models of diabetes. A three-month treatment regimen provided
`sustained improvements in glucose tolerance, increased b-cell responsiveness and
`improved peripheral insulin sensitivity in Zucker fa/fa rats [48,49]. The same in-
`vestigators have shown that 7 weeks of treatment with 2 enhances b-cell survival
`and islet neogenesis in a streptozotocin-induced diabetes model [50]. A study de-
`signed to compare the effects of 2 with those of rosiglitazone and to the effects of
`the combination of the two agents found that the DPP4 inhibitor provided im-
`proved glucose tolerance in both prediabetic and diabetic animals. While rosiglita-
`zone resulted in increased body weight, 2 was body-weight neutral. However,
`neither agent was very effective at improving the diabetic condition of older ZDF
`rats [51]. Studies have shown that the metabolism of 2 is dominated by oxidation of
`the sulfur atom and glucuronidation of the primary amine [52].
`In rodent models of diabetes, chronic treatment with FE 999011 (3) provided
`improved glucose tolerance, postponed the progression to hyperglycemia by 21
`days, reduced hypertrigylyceridemia and prevented a rise in circulating free fatty
`acids [53].
`Rodent studies using NVP-DPP728 (6, IC50 ¼ 7 nM) [54] and the structurally
`related K579 (7, IC50 ¼ 5 nM) have demonstrated similar pharmacological effects
`as those seen with the inhibitors discussed above. In a comparative study, 7 ap-
`peared to provide better control of DPP4 activity and glucose excursions than did 6
`[55]. Combination of 7 with glibenclamide further enhanced the glucose control
`without significant hypoglycemia [56].
`
`N
`
`O
`
`CN
`
`NH
`
`7
`
`N
`
`N
`
`N
`
`N
`
`O
`
`CN
`
`NH
`
`6
`
`HN
`
`N
`
`NC
`
`MYLAN Ex. 1011, Page 4
`
`

`
`Inhibitors of Dipeptidyl Peptidase 4
`
`153
`
`The 2-CN pyrrolidine present in 6 can be substituted by a cyanopyrazoline, but
`this results in a less potent compound (8, IC50 ¼ 360 nM) [57]. A pyrazolidine
`heterocycle has also been examined (9, IC50 ¼ 1.56 mM) [58].
`
`NO2
`
`HN
`
`N
`
`N
`
`N
`
`N
`
`O
`
`CN
`
`H2N
`
`O
`
`O
`
`9
`
`NH
`
`8
`
`HN
`
`N
`
`NC
`
`Several groups have examined substituted pyrrolidines in an effort to improve
`potency or stability of the inhibitors. Attempted incorporation of hydroxy or met-
`hoxy substituents at various positions on the ring led to reduced potency, but
`fluorination at
`the 4-position gave increased potency as in compound 10
`(IC50 ¼ 0.6 nM). This compound also displayed increased plasma drug concentra-
`tions relative to the unsubstituted inhibitor [59]. In an examination of pyrrolidines
`cyclopropanated at either the 3,4 or 4,5 positions, it was found that while intro-
`duction of the cyclopropane on the face of the pyrrolidine trans to the cyano group
`’ s, the cis-3,4-methano and cis-4,5-methano
`led to compounds with micromolar IC50
`moieties were well tolerated. One goal of this work was to reduce the intramolecular
`amine-nitrile cyclization that plagues many cyanopyrrolidine DPP4 inhibitors.
`Bulky substituents on the amino acid and the cyclopropane moiety provided im-
`pressive improvements in solution stability. Compound 11 (IC50 ¼ 1.5 nM) has a
`half-life of 5 hours, while compound 12 (Ki ¼ 8 nM) has one of 27 hours and com-
`pound 13 (Ki ¼ 7 nM), 42 hours. Compound 13 reduced glucose excursions fol-
`lowing an oral glucose tolerance test (OGTT) in Zucker fa/fa rats [60].
`
`F
`
`N
`
`O
`
`10
`
`CN
`
`H2N
`
`CN
`
`N
`
`O
`
`11
`
`CN
`
`N
`
`O
`
`12
`
`H2N
`
`CN
`
`N
`
`O
`
`13
`
`H2N
`
`H2N
`
`Ketopyrrolidines and ketoazetidines, which replace the cyano group with a
`heteroaryl ketone, have also been examined as DPP4 inhibitors. Heteroaryl ketones
`have been used extensively as reversible serine protease inhibitors and act by pro-
`viding an electrophilic carbonyl that can form a tetrahedral species with the active
`site serine. An examination of rings from four to six atoms revealed that only the
`piperidine derivatives were not inhibitors of the enzyme. 2-Thiazolyl and 2-ben-
`zothiazolyl substituents provided sufficient activation of the carbonyl to give low
`nanomolar inhibitors such as 14 (IC50 ¼ 30–42 nM). These compounds suffer from
`an internal cyclization followed by oxidation to give dihydroketopyrazines such as
`15 [61].
`
`MYLAN Ex. 1011, Page 5
`
`

`
`154
`
`S.L. Gwaltney, II and J.A. Stafford
`
`S
`
`N
`
`N
`
`O
`
`O
`
`N
`
`N
`
`S
`
`N
`
`H2N
`
`O
`15
`14
`Substituted cycloalkylglycine thiazolidides and pyrrolidides are potent DPP4 in-
`hibitors. Compound 16 (IC50 ¼ 88 nM) demonstrated good PK in both the rat and
`dog with bioavailabilities of 36% and 100%, respectively [62]. An extensive exam-
`ination of the SAR surrounding cyclopentyl and cyclohexylglycine derived pyrro-
`lidides and thiazolidides has been reported. The cyclopentylglycine derivatives were
`found to be more potent than their cyclohexyl counterparts. While the thiazolidides
`provided greater potency, these compounds suffered from reduced metabolic sta-
`bility. Compound 17 (IC50 ¼ 13 nM) was found to be a potent inhibitor selective for
`DPP4 over QPP and PEP [63]. In a series of mono or disubstituted pyrrolidides with
`fluorine at the 3 and 4 positions, the monofluorinated compounds were more potent
`than the difluoro analogs. Compound 18 (IC50 ¼ 48 nM) was bioavailable in rat
`and dog and gave a 42% reduction in glucose excursion following an OGTT in lean
`mice [64]. This compound undergoes metabolic activation and subsequent conju-
`gation with biological nucleophiles. This is believed to occur through oxidation and
`defluorination events, which produce an enal that acts as a Michael acceptor [65].
`Compounds 19 (IC50 ¼ 6 nM) and 20 (IC50 ¼ 6 nM) are potent inhibitors of DPP4
`that also incorporate the monofluorinated pyrrolidine [66].
`
`F
`
`N
`
`O
`
`HN
`
`S
`
`F
`
`O O
`
`F
`
`HN
`
`S
`
`O O
`
`O
`
`O
`
`S
`
`HN
`
`S
`
`F
`
`O O
`
`F
`
`H
`H2N
`
`18
`
`F
`
`N
`
`O
`
`H2N
`
`N
`
`HN
`
`O
`
`S
`
`N
`
`O
`
`H
`H2N
`
`17
`
`F
`
`F3C
`
`N
`
`O
`
`H
`H2N
`
`16
`
`F3C
`
`N
`
`S
`
`F
`
`N
`
`O
`
`H2N
`
`19
`20
`Starting from high-throughput screening (HTS) hit 21 (IC50 ¼ 1.9 mM), a series of
`b-homophenylalanine thiazolidides was developed [67]. Substitution of fluorine at the
`
`MYLAN Ex. 1011, Page 6
`
`

`
`Inhibitors of Dipeptidyl Peptidase 4
`
`155
`
`2-position of the phenyl ring was found to provide an approximately 3-fold improvement
`in potency. The most potent compound reported in this series was 22 (IC50 ¼ 119 nM).
`This work was extended to a series of proline and thiazolidine amides such as 23
`(IC50 ¼ 0.48 nM). While very potent, these analogs demonstrate poor PK properties
`[68]. Investigation of the SAR in a series of related piperazines represented by 24
`(IC50 ¼ 19 nM) revealed that the R-benzyl group was important for potency [69]. These
`analogs also suffer from short metabolic half-lives due to oxidation of the piperazine ring
`and poor pharmacokinetics. These liabilities were addressed through the discovery of
`MK-0431, which will be discussed in the section on clinical DPP4 inhibitors.
`
`F
`
`CO2H
`
`F
`
`F
`
`F
`
`F
`
`F
`
`NH2
`
`O
`
`22
`
`N
`
`S
`
`NH2
`
`O
`
`N
`
`24
`
`Ph
`
`NH
`
`Cl
`
`O
`
`NH
`
`HN
`
`O
`
`NH2
`
`O
`
`N
`
`21
`
`O
`
`HN
`
`O
`
`NH2
`
`O
`
`N
`
`23
`
`Ph
`
`F
`
`F
`
`Sulphostin (25) is a natural product with an IC50 of 6.0 ng/mL, which corres-
`ponds to approximately 20 nM [70,71]. In an examination of the structure-activity
`relationships for analogs of sulphostin, it was found that the carbonyl and C-3
`amino group of the parent structure were important for maintaining potency, as
`was the absolute configuration at phosphorus. Heterocycle ring sizes of 5–7 atoms
`were well tolerated. The sulfonic acid moiety could be removed while maintaining
`potency, but deletion of this group negatively impacts the stability of these analogs.
`Compound 26 is an 11 nM inhibitor of DPP4 [72].
`
`NH2
`
`NH2
`
`NH2
`
`NH2
`
`N
`
`N
`
`Cl
`
`Ar
`
`N
`
`Cl
`
`28: Ar = phenyl
`29: Ar = 3,5-dimethoxyphenyl
`
`OO
`
`N
`
`27
`
`NH2
`
`N
`
`O
`
`PO
`
`NHSO3H
`
`NH2
`
`25
`
`N
`PO
`
`H2N
`
`NH2
`
`O
`
`NH2
`
`26
`
`Starting from HTS hit 27 (IC50 ¼ 10 mM), the potency in a series of amino-
`methylpyrimidines was improved 100,000 fold through modification of the two aryl
`
`MYLAN Ex. 1011, Page 7
`
`

`
`156
`
`S.L. Gwaltney, II and J.A. Stafford
`
`substituents [73]. Ortho and para substituents on the C-6 phenyl ring were tolerated,
`whereas meta substitution generally led to loss of potency. A breakthrough was
`realized when the 2,4-dichlorophenyl derivative 28 (IC50 ¼ 10 nM) was prepared.
`Optimization of 28 through modification of the C-2 phenyl group led to compound
`29, reported to be a 100-picomolar inhibitor. An X-ray crystal structure of 29 in
`DPP4 reveals that the dichlorophenyl group effectively fills S1, the pyrimidine ring
`forms a cation-p interaction with Arg125, the aminomethyl group interacts with
`Tyr662 and the two active-site glutamates, and the anilino nitrogen forms an ad-
`ditional H-bond with the backbone carbonyl of Glu205.
`Analogs of these compounds where the pyrimidine is replaced with a pyridine
`(e.g. 30) were explored. It was found that potency is improved by reducing the
`torsion angle between the phenyl ring and the pyridine core. Optimization in this
`series led to compound 34 [74].
`
`Cmpd
`
`30
`31
`32
`33
`34
`
`n
`
`0
`3
`2
`1
`
`NH2 NH2
`
`N
`
`Cl
`
`(CH2)n
`
`30-33
`
`MeO
`
`Cl
`
`MeO
`
`Torsion (calc.)
`
`IC50 (mM)
`
`321
`221
`01
`
`NH2 NH2
`
`N
`
`Cl
`
`0.92
`0.24
`0.045
`0.039
`0.007
`
`NH2 NH2
`
`N
`
`Cl
`
`O
`
`Cl
`
`N
`
`N
`
`35
`
`34
`
`Cl
`
`Compound 28 is an inhibitor of CYP450 3A4 with an IC50 of 5.4 mM. This
`compound also caused phospholipidosis in cultured fibroblasts. It was anticipated
`that by reducing the lipophilicity of these compounds, improved properties would
`be realized. While replacing the 2-phenyl group with small groups such as Me, OMe
`and NH2 led to significant decreases in potency, groups such as 4-thiomorpholinyl
`and N-hydroxyethyl, N-methylamino were well tolerated. Compound 35 has an
`IC50 of 9 nM versus DPP4, does not induce phospholipidosis and has an IC50 of 30
`mM versus CYP450 3A4 [75].
`
`3. DPP4 INHIBITORS IN CLINICAL DEVELOPMENT
`
`Small-molecule DPP4 inhibitors have advanced into clinical trials. First-generation
`inhibitors P32/98 (2) [76] and NVP-DPP728 (6) [77] were important tools to vali-
`date the concept that DPP4 inhibition is an effective method to improve glucose
`control in diabetic patients through an increase in active GLP-1 levels. In a 4-week
`study evaluating ninety-three type 2 diabetic patients, NVP-DPP728 (6), at doses of
`100 mg three times daily and 150 mg twice daily, demonstrated meaningful reduc-
`tions of plasma glucose, insulin, and the glycohemoglobin, HbA1c [78].
`
`MYLAN Ex. 1011, Page 8
`
`

`
`Inhibitors of Dipeptidyl Peptidase 4
`
`157
`
`The early agents, however, have been replaced by a second generation of DPP4
`inhibitors, which have improved potency, selectivity, and pharmacokinetics over
`their pioneering predecessors. These include LAF237 (36, vildagliptin), MK-0431
`(37, Sitagliptin), BMS-477118 (38, saxagliptin), and GSK23A (39). Other DPP4
`inhibitors that are known to have entered clinical trials are P93/01 and SYR322,
`though the chemical structures of these compounds have not been disclosed [79,80].
`
`NH2
`
`O
`
`N
`
`37
`
`N
`
`N
`
`N
`
`CF3
`
`O
`
`CN
`
`N
`
`NH2
`
`S
`
`O O
`
`39
`
`F
`
`F
`
`F
`
`F
`
`NC
`
`O
`
`N
`
`NH
`
`36
`
`O
`
`CN
`
`HO
`
`HO
`
`N
`
`NH2
`
`38
`
`CH3O
`
`The chemical architecture of the DPP4 inhibitors that have been advanced into
`clinical trials is interesting both for their common structural features and those
`features that make them unique. A cyanopyrrolidine amide is a frequently repeating
`motif in small-molecule DPP4 inhibitors. In each of the compounds possessing this
`functionality, the cyano group undergoes nucleophilic addition by the catalytic se-
`rine, resulting in covalent modification of the DPP4 enzyme. Introductions to the
`pyrrolidine moiety of fluorine substitution (e.g., 39) or a fused ring (e.g., 38) are
`reported to provide improved in vivo and/or stability properties. An a-amino acid
`fragment is also common among DPP4 inhibitors, and it has been revealed from
`crystallography studies that the basic, protonated amino group interacts with a pair
`of Glu residues in the DPP4 active site. Like the modifications on the pyrrolidine
`ring, the installation of a quaternary center adjacent to this essential amino group
`serves the purpose of inhibiting the internal cyclization reaction, which gives rise to a
`bicyclic amidine that undergoes further hydrolysis to a diketopiperazine. In the case
`of NVP-DPP728, the observed by-product of this decomposition pathway is 40 [81].
`
`O
`
`H
`
`N
`
`N
`
`O
`
`40
`
`NH
`
`N
`
`NC
`
`MYLAN Ex. 1011, Page 9
`
`

`
`158
`
`S.L. Gwaltney, II and J.A. Stafford
`
`LAF237 (36) is a 4 nM inhibitor of DPP4 with 410,000-fold selectivity over post-
`proline converting enzyme (PPCE) and DPP-II [81]. The identification of the
`1-amino-3-hydroxyadamantane ring system was the result of careful and systematic
`SAR studies on the P-2 site, wherein a loss of enzymatic potency was observed with
`both carbamate and ester derivatives of the 3-hydroxy group. As expected, the steric
`encumbrance of the adamantane retards the rate of intramolecular cyclization by
`about 30-fold. Compound 36 demonstrated oral efficacy in a standard OGTT
`model using obese Zucker fa/fa rats.
`In a 12-week, placebo-controlled phase II trial, 36 was effective at improving
`glycemic control when given as monotherapy to drug-naı¨ ve patients [82]. The mean
`change in HbA1c was 0.6% from a baseline of 8.0. The most common adverse event
`was hypoglycaemia, which was observed in 7 patients (10%) and was considered
`mild.
`In a double-blinded trial, 107 patients with type 2 diabetes being treated with
`metformin, patients were randomized to receive 36 or placebo [83]. After 12 weeks
`of combination treatment, patients completing the 12-week therapy study were
`eligible to extend treatment to 52 weeks. In the 56 patients randomized to 36 (50 mg
`once daily), HbA1c levels after 12 weeks were reduced by 0.6% from an average
`baseline of 7.7%. In contrast, HbA1c levels were unchanged in patients (n ¼ 107)
`receiving placebo. In the 42 patients that progressed to the extended study, HbA1c
`levels in the 36-treated groups were unchanged from 12 weeks to 52 weeks. Patients
`in the metformin-plus-placebo group (n ¼ 29) showed a gradual increase in HbA1c
`over the 40-week extension, resulting in a between-group average difference of
`–1.1% HbA1c levels. These 52-week data on 36 in combination with metformin
`provide compelling evidence that DPP4 inhibition represents a robust method for
`longer-term glycemic control.
`The discovery of MK-0431 (37, Sitagliptin), and the incorporation of a b-amino
`acid moiety, represented a notable departure from the characteristic a-amino acid
`fragment featured in most reported DPP4 inhibitors [84]. Indeed, X-ray evidence
`suggests a binding orientation of the amide carbonyl that is opposite to its a-amino
`acid progenitors, with the b-amino group retaining the predicted interactions to the
`Glu 205/206 pair. Compound 37 is a non-covalent, 18 nM inhibitor of DPP4 that
`possesses excellent selectivity (2000 to 5000-fold) over related peptidases DPP2,
`DPP8, and DPP9. The desired animal pharmacokinetics of 37 came from modi-
`fications of piperazine amides such as 24, which undergo extensive metabolism yet
`are also potent DPP4 inhibitors [69]. Interestingly, and perhaps generally pertinent
`to the development of safe and effective DPP4 inhibitors for chronic administra-
`tion, DPP8 and DPP9 were specifically highlighted for cross-reactivity as it has
`been reported that the DPP8/9 selective inhibitor 41 is associated with ‘‘multi-organ
`pathology and mortality’’ when administered to rats for 2 weeks at a dose of
`100 mg/kg/day. Moreover, these effects were observed in both wild-type and DPP4-
`deficient mice, suggesting that the toxicities were independent of DPP4 inhibition
`[85]. The preclinical oral efficacy of 37 was demonstrated in an OGTT model using
`C57BL/6N male mice.
`
`MYLAN Ex. 1011, Page 10
`
`

`
`Inhibitors of Dipeptidyl Peptidase 4
`
`159
`
`H3C
`
`CH3
`
`O
`
`N
`
`NH2
`
`41
`
`A randomized, placebo-controlled OGTT in fifty-six type 2 diabetics was con-
`ducted to assess the glucose-lowering activity and safety/tolerability of 37 following
`a single oral dose of 25- or 200-mg. As compared to placebo, incremental glucose
`AUC was reduced by approximately 22% and 26%, for the 25- and 200-mg
`doses respectively [86]. Dose-responsive plasma increases in insulin, c-peptide, and
`GLP-1, and reductions in glucagon were also noted, leading to the conclusion that
`pharmacologic proof-of-concept had been achieved.
`GSK23A (39) is a penicillinamine-based inhibitor of DPP4 with a Ki value of
`53 nM [87]. The compound contains a 4-fluoro substituent on the cyanopyrrolidine
`ring, which confers unique biochemical and physical properties versus its des-fluoro
`analog 42. For example, 39 has a half-time to onset of DPP4 inhibition of 120 min,
`as measured in human plasma, compared to o20 min for compound 42. In ad-
`dition, 39 has a half-time for internal cyclization (37 1C, pH 7.2) of 1733 hr versus
`360 hr for 42. In a standard OGTT in ob/ob mice 39 showed an expected lowering
`of plasma glucose with an increase in both GLP-1 and insulin. By contrast, in the
`db/db mouse model, serum levels of glucose were unchanged following 8 weeks of
`treatment with 39, presumably due to the severe insulin resistance of these animals.
`
`O
`
`CN
`
`S
`
`O O
`
`N
`
`NH2
`
`42
`
`CH3O
`
`BMS-477118 (38) is a methanoproline-based DPP4 inhibitor [88,89]. The cis-
`fused, 4,5-methano bridge on the pyrrolidine ring appears to have been borrowed
`from the existing ACE inhibitor literature, wherein it has been shown that captopril
`analogs such as 43 having a fused cyclopropane ring retain the full ACE inhibitory
`potency of captopril (44) itself [90]. This modification is highlighted as a structural
`tool to enhance the chemical stability of the compound by retarding the rate of the
`internal cyclization reaction. BMS-477118 is a potent inhibitor of DPP4 with a
`reported IC50 value of 0.45 nM. It is reported to be selective over the related pep-
`tidases DPP2, DPP8, DPP9, and fibroblast-activating protein (FAP). In preclinical
`OGTT studies using both Zucker fa/fa rats and ob/ob mice, BMS-477118 displays
`the phenotypic profiles characteristics of DPP4 inhibition including transient lowe-
`ring of glucose with concomitant increases in plasma insulin.
`
`MYLAN Ex. 1011, Page 11
`
`

`
`160
`
`S.L. Gwaltney, II and J.A. Stafford
`
`O
`
`CO2H
`
`O
`
`CO2H
`
`HS
`
`N
`
`HS
`
`N
`
`CH3
`
`43
`
`CH3
`
`44
`
`It is widely accepted that the level of glycohemoglobin, HbA1c, is the best mea-
`sure of long-term glycemic control and should be a primary endpoint for assessing
`the effectiveness of diabetes therapy [91]. A key topic that remains unanswered is
`the relationship between in vivo DPP4 inhibition over time and its effects on both
`HbA1c and safety, though the early reports are highly encouraging that sustained
`DPP4 inhibition is both efficacious and well tolerated. Based on the large volume of
`primary journal literature, patent literature, and reports from scientific meetings, it
`is fair to speculate that there are a number of other small-molecule DPP4 inhibitors
`that are undergoing preclinical and clinical evaluation.
`
`4. ALTERNATIVE INDICATIONS FOR DPP4 INHIBITORS
`
`Numerous studies suggest DPP4 inhibition for pharmacological uses other than the
`restoration of glycemic control and type 2 diabetes. The common underlying hy-
`potheses of these studies are divided into two categories that describe fundamental
`properties of DPP4. CD26, a membrane-associated peptidase that has DPP4 ac-
`tivity has been extensively studied in relation to its role in regulating T-cell phys-
`iology. As such, DPP4 activity has been studied in the context of T-cell activation
`and immune function [92]. Activated T-cells are known to have increased cell-
`surface expression of DPP4 [93]. Furthermore, cytokines such as RANTES,
`SDF-1a, MCP-2, and TNF-a have all been characterized as substrates for DPP4, so
`it is a reasonable hypothesis that DPP4 plays an immunomodulatory role [94].
`However, recent studies have raised questions on the dependence of DPP4’s pro-
`teolytic activity to T-cell activation and other functions such as proliferation and
`cytokine release. In a study using compounds with varying selectivity profiles for
`DPP4 and related peptidases, such as QPP, DPP8, and DPP9, it was found that
`both a DPP4 and a QPP inhibitor had no effects in in vitro assays measuring the
`immune responses of T-cell proliferation and IL-2 release [95]. By contrast, less
`selective compounds, such as Val-boro-Pro (45) and Lys[Z(NO2)]-pyrrolidide (46),
`which also show inhibitory activity against DPP8 and DPP9, were effective in these
`assays. The authors conclude that the T-cell-mediated effects previously assigned to
`the inhibition of DPP4 might actually be a consequence of DPP8 and/or DPP9
`activity. In addition, the uncompetitive DPP4 inhibitor, TMC-2A (47), [96] has
`been shown to suppress paw swelling in a rat adjuvant model for arthritis [97]. The
`authors of this study also note that DPP4 inhibition, per se, does not affect T-cell
`function, and that mice with mutated DPP4 lacking enzymatic activity still show a
`normal immune response. It is suggested that the binding of TMC-2A (47) may
`
`MYLAN Ex. 1011, Page 12
`
`

`
`Inhibitors of Dipeptidyl Peptidase 4
`
`161
`
`affect the function of other proteins that associate with CD26, specifically the PTPase
`activity of CD45 [98]. Compound 45 is also known as talabostat or PT-100 and is in
`clinical trials for hematological malignancies and hematopoiesis [99]. The efficacy of
`45 in these therapeutic indications may be derived from the compound’s inhibition of
`FAP [100]. Numerous other reports have focused on the involvement of DPP4 and/
`or use of DPP4 inhibitors in various inflammatory and autoimmune diseases and
`pathologies, such as Crohn’s disease, [92] and organ transplantation [101].
`
`OMe
`
`HO
`
`OH
`
`OH
`
`OH
`
`OH
`
`O
`
`NH
`
`47
`
`N
`
`O
`
`O
`
`HN
`
`H2N
`
`O
`
`O
`
`NH
`
`H2N
`
`46
`
`N
`
`O
`
`–O
`
`N+
`O
`
`H2N
`
`N
`
`O
`
`45
`
`B(OH)2
`
`In addition to its identity with the membrane-associated protein CD26, DPP4 is
`ubiquitously distributed, and many important biomolecules other than GLP-1, in-
`cluding hormones, neuropeptides, chemokines, and cytokines, have been charac-
`terized as DPP4 substrates [3]. Among these substrates for DPP4, the peptide
`hormone GLP-2 has received considerable attention recently for its activity as an
`intestinal growth factor [102]. This activity has led to the hypothesis that a DPP4
`inhibitor could show intestinotrophic effects that may be useful in the treatment of
`inflammatory bowel disease (IBD). Indeed GLP-2 itself has shown efficacy in a
`rodent model of IBD [103].
`
`REFERENCES
`
`[1] D. J. Drucker, Exp. Opin. Investig. Drugs, 2003, 12, 87.
`[2] P. E. Wiedeman and J. M. Trevillyan, Curr. Opin. Investig. Drugs, 2003, 4, 412.
`[3] A.-M. Lambeir, C. Durinx, S. Scharpe´ and I. De Meester, Crit. Rev. Clin. Lab. Sci.,
`2003, 40, 209.
`[4] J. J. Holst, Exp. Opin. Emerg. Drugs, 2004, 9, 155.
`[5] C. F. Deacon, B. Ahre´ n and J. J. Holst, Exp. Opin. Investig. Drugs, 2004, 13, 1091.
`[6] J. J. Holst and C. F. Deacon, Curr. Opin. Pharmacol., 2004, 4, 589.
`[7] A. E. Weber, J. Med. Chem., 2004, 47, 4135.
`[8] B. Ahre´ n, Current Enzyme Inhibition, 2005, 1, 65.
`[9] C.H.S. McIntosh, H.-U. Demuth, J.A. Pospisilik and R. Pederson, Regul. Pept., 2005,
`159.
`[10] E. M. Sinclair and D. J. Drucker, Curr. Opin. Endocrinol. Diabet., 2005, 12, 146.
`[11] R. Mentlein, Exp. Opin. Investig. Drugs, 2005, 14, 57.
`[12] T. Hoffman and H.-U. Demuth, in Ectopeptidases, (eds Langner and Ansorge), Kluwer
`Academic/Plenum Publishers, New York, 2002, p. 259.
`[13] E. B. Villhauer, G. M. Coppola and T. E. Hughes, Annu. Rep. Med. Chem., 2001, 36,
`191.
`[14] B. Leiting, K. D. Pryor, J. K. Wu, F. Marsilio, R. A. Patel, C. S. Craik, J. A. Ellman,
`R. T. Cummings and N. A. Thornberry, Biochem. J., 2003, 371, 525.
`
`MYLAN Ex. 1011, Page 13
`
`

`
`162
`
`S.L. Gwaltne

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