throbber
2. DPPIV Inhibition: Promising Therapy
`for the Treatment of Type 2 Diabetes
`
`PAUL E. WIEDEMAN
`
`Abbott Laboratories, Department R4CP, Building AP9B, 100 Abbott Park
`Road, Abbott Park, IL 60064-6113, USA
`
`INTRODUCTION
`Type 2 Diabetes Pandemic
`Inhibition of DPPIV as a Strategy to Enhance the Incretin Effect
`Dipeptidyl Peptidase IV
`Potential for Disease Modification with DPPIV Inhibitors
`Importance of DPPIV Selectivity
`Structural Biology
`
`MEDICINAL CHEMISTRY AND PRECLINICAL STUDIES
`Early DPPIV Inhibitors
`Covalent DPPIV Inhibitors: P2 Secondary Amine Analogues
`Covalent DPPIV Inhibitors: P2 Primary Amine Analogues
`Non-Covalent DPPIV Inhibitors: P2 Primary Amine Analogues
`Peptidic DPPIV Inhibitors that Extend into P10
`Non-Peptidic/Non-Covalent DPPIV Inhibitors
`
`CLINICAL DATA FOR ORAL DPPIV INHIBITORS
`
`CONCLUSIONS
`
`AUTHORS NOTE
`
`REFERENCES
`
`64
`64
`64
`66
`68
`68
`69
`
`71
`71
`74
`80
`84
`89
`90
`
`96
`
`101
`
`102
`
`102
`
`Progress in Medicinal Chemistry – Vol. 45
`Edited by F.D. King and G. Lawton
`DOI: 10.1016/S0079-6468(06)45502-8
`
`63
`
`r 2007 Elsevier B.V.
`All rights reserved.
`
`AstraZeneca Exhibit 2162
`Mylan v. AstraZeneca
`IPR2015-01340
`
`Page 1 of 47
`
`

`
`64
`
`DPPIV INHIBITION
`
`INTRODUCTION
`
`TYPE 2 DIABETES PANDEMIC
`
`Diabetes is a major health problem encountered across the globe. Nearly
`200 million individuals worldwide suffer from diabetes of which 90–95% are
`type 2 diabetics [1]. Some staggering figures include India, home to 35.5
`million diabetics, China with 23.8 million, the United States with 16 million,
`Russia with 9.7 million and Japan with 6.7 million. The incidence of di-
`abetes has not peaked. By 2025, the diabetic population of Africa, the
`Eastern Mediterranean, the Middle East and Southeast Asia is expected to
`increase by 100%. Rises are anticipated in other regions as well, including
`Central and South America (85%), the Western Pacific (75%), North
`America (50%) and Europe (20%). Already diabetes is the fourth main
`cause of mortality in the majority of developed countries. Healthcare
`systems will certainly be strained to meet the growing demand of this pan-
`demic, as even now 50% of patients are undiagnosed. The growing diabetic
`population requires additional therapies with alternative mechanisms of
`action and improved tolerability. There are currently five main classes of
`oral antidiabetics, each limited in one way or other by the degree of efficacy
`and side effects [2]. Concerns with sulfonylureas centre on hypoglycemia
`and weight gain. Non-sulfonylurea secretagogues have the same issues along
`with a more complex dosing schedule. Patients on thiazolidinediones are
`prone to weight gain and oedema. Biguanides and a-glucosidase inhibitors
`often produce significant gastrointestinal distress. Inhibition of dipeptidyl
`peptidase IV (DPPIV) cleavage of glucagon-like peptide-1 (GLP-1) is a
`highly validated target for the treatment of type 2 diabetes. Several DPPIV
`inhibitors are in clinical development, and the first requests for regulatory
`review have been filed. The reported clinical data have established proof of
`concept in man, confirming the possibility that DPPIV inhibitors will be the
`next major new class of oral antidiabetic drug.
`
`INHIBITION OF DPPIV AS A STRATEGY TO ENHANCE THE INCRETIN EFFECT
`
`The phenomenon of increased insulin secretion following oral administra-
`tion of glucose compared to intravenous administration is known as the
`incretin effect. An agent responsible for this effect is the incretin hormone,
`GLP-1. In response to the oral ingestion of nutrients, proglucagon is proc-
`essed and GLP-1 is released from enteroendocrine L-cells in the distal small
`intestine and colon. Binding of GLP-1 to its G-protein-coupled receptor on
`pancreatic b-cells increases glucose-stimulated insulin secretion [3, 4].
`Additional desirable effects of GLP-1 include increased insulin gene
`
`Page 2 of 47
`
`

`
`P. E. WIEDEMAN
`
`65
`
`expression [5], and increased pancreatic b-cell proliferation and islet neo-
`genesis [6]. By contrast and also of benefit are the inhibition of glucagon
`secretion [7, 8] and decreased gastric emptying that results in the slowed rate
`of nutrient absorption [9,10]. GLP-1 receptors are also expressed in hypo-
`thalamic nuclei responsible for modulating feeding behaviour, and periph-
`eral administration of GLP-1 promotes satiety and inhibits food intake in
`man [11, 12]. In total these effects demonstrate that GLP-1 (7–36) amide has
`multiple biological effects that contribute to glucose homeostasis and pro-
`motes normalization of post-meal glucose levels. It is important to recognize
`that GLP-1 augments insulin secretion in a glucose-dependent manner.
`Unlike sulfonylurea drugs or insulin, enhancing endogenous GLP-1 levels
`does not increase the risk of hypoglycemia.
`Infusion of active GLP-1 and GLP-1 (7–36) amide reduces post-meal and
`fasting glycemia in patients with non-insulin-dependent diabetes mellitus;
`thus establishing the potential of GLP-1-based therapy for the treatment of
`type 2 diabetes [13–15]. This effect occurs despite a blunting of the incretin
`effect in type 2 diabetics [16]. The key problem, however, is that active GLP-
`1 (7–36) amide is rapidly converted to inactive GLP-1 (9–36) amide by the
`action of DPPIV via the cleavage of the N-terminal dipeptide (His-Ala) of
`GLP-1 (7–36) amide [17, 18]. The short half-life of GLP-1 (7–36) amide in
`the circulation (o2 min) makes it impractical as a therapeutic agent and has
`led to the development of alternative strategies to enhance the anti-
`diabetogenic activity of GLP-1. One successful approach that will not be
`covered in this chapter is the development of GLP-1 receptor agonists that
`are resistant to DPPIV cleavage [19]. This approach remains an active area
`of research and development [20]. Exenatide (Byetta) is the first marketed
`GLP-1 receptor agonist. It is effective in reducing glycosylated haemo-
`globin, HbA1c (biomarker for glycemic control) levels in type 2 diabetics but
`is a twice-a-day injectable peptide with nausea as a prominent side effect
`[21]. Another strategy that is the focus of this chapter is to increase the
`circulating half-life of endogenous GLP-1 by inhibiting its enzymatic de-
`gradation by DPPIV [17] (Fig. 2.1).
`Another incretin hormone, glucose-dependent insulinotropic polypeptide
`(GIP), is also degraded by DPPIV [18]. Similar to GLP-1, GIP is a 42-amino
`acid peptide secreted by endocrine K cells of the duodenum in response to
`ingestion of nutrients [22]. The physiological actions of GIP include glucose-
`dependent potentiation of insulin secretion and regulation of insulin gene
`transcription. In contrast to GLP-1, glucose tolerance is not improved in
`type 2 diabetics treated with exogenous GIP [23]. However, it has been
`reported that the response to GIP improves in diabetic patients treated with
`glyburide to reduce fasting glucose levels [22]. Studies with hyperglycemic
`diabetic rats or normal rats made hyperglycemic by glucose clamp have
`
`Page 3 of 47
`
`

`
`66
`
`DPPIV INHIBITION
`
`Fig. 2.1 Inhibition of DPPIV sustains endogenous GLP-1 and modulates the incretin effect.
`
`decreased expression of pancreatic GIP receptor, which in turn causes a loss
`of insulinotropic response to GIP [24]. Taken together, these data support
`the notion that the initial effect of DPPIV inhibition in human diabetes is
`primarily by GLP-1, but raises the possibility that in the long run, when
`glucose levels fall, treatment with DPPIV inhibitors could also improve the
`insulinotropic action of GIP.
`
`DIPEPTIDYL PEPTIDASE IV
`
`DPPIV (EC 3.4.14.5; also known as lymphocyte cell surface protein CD26)
`was first described in 1967 [25]. DPPIV is a post-proline cleaving serine
`protease with a catalytic triad of Ser-Asp-His oriented inversely to classical
`serine proteases and with significant homology to other a,b-hydroxylases.
`DPPIV is expressed as a 110 kDa glycoprotein on the surface of cells of most
`tissues including kidney, liver, intestine, placenta, prostate, skin, lymph-
`ocytes and endothelial cells. DPPIV is catalytically active as a dimer. Pro-
`teolytic cleavage of DPPIV from cell surfaces results in a soluble circulating
`form with a monomeric mass of approximately 100 kDa. In addition to
`cleaving GLP-1, DPPIV may play a role in the cleavage of other substrates
`with accessible amino-terminal Xaa-Pro- or Xaa-Ala-dipeptide sequences,
`resulting in their inactivation or alteration in their biological activities. Po-
`tential DPPIV substrates include growth hormone releasing hormone, GIP,
`pituitary adenylate cyclase-activating polypeptide 38 (PACAP38), substance
`P, bradykinin, gastrin releasing peptide, neuropeptide Y, peptide YY,
`
`Page 4 of 47
`
`

`
`P. E. WIEDEMAN
`
`67
`
`certain chemokines such as RANTES (regulated on activation normal T cell
`expressed and secreted), stromal cell-derived factor, eotaxin and macroph-
`age-derived chemokines [26]. Long-term safety concerns have arisen because
`these possible DPPIV substrates include chemokines, vasoactive peptides,
`neuropeptides and gastrointestinal peptides. Despite in vitro cleavage of
`these peptides by DPPIV, many of the activities associated with these pep-
`tides appear not to be physiologically regulated in vivo by DPPIV action.
`Those that appear to be regulated by DPPIV peptidase activity are described
`below.
`GLP-1 and GIP have been validated as in vivo substrates of DPPIV in
`DPPIV knockout (DPPIV KO) mice and DPPIV-deficient Fisher rats [27,
`28]. The importance of GLP-1 and GIP in the gluco-regulatory action of
`DPPIV inhibitors has been shown in double incretin receptor (i.e., GLP-1R
`and GIPR) knockout mice where the glucose-lowering effect of DPPIV
`inhibitors is abolished [29]. DPPIV-deficient mice are healthy, have normal
`blood glucose levels in the fasted state but reduced glucose excursion after a
`glucose challenge [27]. The active, insulinotropic form of GLP-1 and glu-
`cose-dependent insulin levels are both increased in DPPIV KO mice com-
`pared to wild-type littermates. Similarly, DPPIV-deficient Fisher rats have a
`phenotype of improved glucose tolerance and enhanced glucose-dependent
`insulin secretion [28].
`Similarly, PACAP38 has been validated as an additional target of in vivo
`DPPIV peptidase activity. PACAP38 is a neuropeptide that is involved with
`signalling to pancreatic nerves and is therefore associated with neural reg-
`ulation of islet function. Preservation of endogenous levels of PACAP38
`with a DPPIV inhibitor may be an additional way that the inhibitors en-
`hance antidiabetic effects. PACAP38 was administered exogenously to both
`wild-type and DPPIV-deficient mice [30]. In the DPPIV-deficient mice, the
`rate of PACAP38 clearance was reduced and little of the DPPIV metabolite,
`PACAP(3–38) was observed. In another study in mice, PACAP38 was ad-
`ministered intravenously with glucose following previous administration of
`a DPPIV inhibitor [31]. As was observed in the same study with GLP-1, the
`PACAP38-treated animals showed increased insulin levels and a greater rate
`of glucose elimination.
`Although DPPIV clearly regulates GLP-1 (and probably GIP and
`PACAP38) action in vivo, the DPPIV enzyme may have a broader role in
`metabolic control. In this regard, DPPIV KO mice show resistance to diet-
`induced obesity with a concomitant reduction in adiposity compared to
`wild-type mice [32]. Consequently, long-term inhibition of DPPIV may have
`the potential to decrease the body weight. However, DPPIV inhibitors cur-
`rently in clinical development have not demonstrated weight reduction, but
`several trials have shown weight neutrality.
`
`Page 5 of 47
`
`

`
`68
`
`DPPIV INHIBITION
`
`POTENTIAL FOR DISEASE MODIFICATION WITH DPPIV INHIBITORS
`
`GLP-1 has trophic effects on b-cells [33]. Not only does it stimulate b-cell
`proliferation but it also inhibits apoptosis of b-cells [34]. GLP-1 is capable of
`enhancing the differentiation of new b-cells from pancreatic ductal epithelium
`[6, 35]. DPPIV-mediated preservation of endogenous GLP-1 may also
`enhance the effects of GLP-1 on b-cell rescue/prevention of apoptosis.
`Indeed, chronic treatment with DPPIV inhibitors has been shown to preserve
`islet function in diabetic mice and to improve b-cell survival and islet cell
`neogenesis in streptozotocin-induced diabetic rats [36, 37]. It has been reported
`that humans with type 2 diabetes have a b-cell deficit most likely due to
`increased b-cell apoptosis [38]. Consequently, by increasing GLP-1 levels,
`DPPIV inhibitors may be capable of providing new b-cells in patients with
`type 2 diabetes and thus, prevent the worsening of the disease. Furthermore,
`the treatment of
`insulin-resistant
`individuals (impaired glucose-tolerant
`pre-diabetics) with DPPIV inhibitors may delay or prevent the onset of
`diabetes. This would be a major medical breakthrough in the treatment of type
`2 diabetes and impaired glucose tolerance. However, clinical studies to date
`have not provided conclusive evidence that the b-cell sparing activity observed
`with DPPIV inhibitors or GLP-1 analogues in diabetic animal models will also
`occur in man. The diagnostics to directly measure b-cell mass in the clinical
`setting simply do not exist yet. However, improvements in b-cell function can
`and have been assessed. On an optimistic note, an apparent decrease in the
`rate of progression of type 2 diabetes has been observed with a DPPIV in-
`hibitor in combination with metformin in a long-term clinical study [39].
`
`IMPORTANCE OF DPPIV SELECTIVITY
`
`DPPIV is a member of a family of closely related enzymes that share DPP-
`IV-like catalytic activity [40]. Consequently, there is potential for adverse
`events or toxicity associated with non-selective DPPIV inhibitors. Two
`members of the DPPIV family, DPP8 [41] and DPP9 [42, 43], were recently
`discovered and characterized. Their exact physiologic role remains under
`investigation. However, profound toxicities in rodent and adverse side
`effects in dog have been reported with a DPP8/9 inhibitor [44]. Merck sci-
`entists reported in 2-week rat toxicology studies that a selective dual DPP8/
`DPP9 inhibitor (1), IC50 ¼ 38 and 55 nM, respectively, caused alopecia,
`thrombocytopenia, anaemia, enlarged spleen, multiple histological path-
`ologies and death. In an acute dog study, the DPP8/9 inhibitor produced
`bloody diarrhea, emesis and tenesmus. The same toxicity was observed in
`both models with a DPPIV inhibitor (2) non-selective against DPP8/9, but
`was not observed with a selective DPPIV inhibitor MK-0431 (3) (DPPIV
`
`Page 6 of 47
`
`

`
`P. E. WIEDEMAN
`
`69
`
`F
`
`F
`
`NH2
`
`O
`
`N
`
`N
`
`N
`
`F
`(3)
`DPPIV IC50 = 27 nM
`DPP8 IC50 > 69000 nM
`DPP9 IC50 > 100000 nM
`
`N
`
`CF3
`
`O
`
`N
`
`NH2
`
`HN
`
`O
`
`O
`
`(5)
`DPPIV IC50 = 1300 nM
`DPP8 IC50 = 154 nM
`DPP9 IC50 = 165 nM
`
`Me
`
`Me
`
`O
`
`N
`
`NH2
`
`(1)
`DPPIV IC50 = 30000 nM
`DPP8 IC50 > 38 nM
`DPP9 IC50 > 55 nM
`Me
`
`O
`
`N
`
`S
`
`Me
`
`N
`
`NH2
`
`Me
`
`Me
`
`O
`
`NH2
`
`B(OH)2
`
`O2N
`
`(2) (allo-isomer)
`DPPIV IC50 = 460 nM
`DPP8 IC50 = 220 nM
`DPP9 IC50 = 320 nM
`
`(4)
`DPPIV IC50 < 4 nM
`DPP8 IC50 =4 nM
`DPP9 IC50 = 11 nM
`
`Fig. 2.2 Selectivity for DPPIV over related peptidases.
`
`IC50 ¼ 27 nM, DPP8 ¼ 69mM, DPP9>100 mM). Importantly, several DPP-
`IV inhibitors reported in earlier studies were found to lack selectivity for
`DPPIV over DPP8/9. Some of the compounds such as (4) and (5) with poor
`selectivity were active in in vitro models of T-cell activation while DPPIV
`selective compounds exhibited no response. This data supports the premise
`that non-selective DPPIV inhibitors likely exhibit off-target
`toxicities.
`Additionally, skin lesions have been observed in monkeys treated with DPP-
`IV inhibitors. Although the mechanism of action has yet to be determined, the
`U.S. Food and Drug Administration has insisted that all DPPIV inhibitors be
`tested in monkeys [45]. This data is relatively recent and a significant amount
`of the medicinal chemistry efforts described herein occurred prior to this
`understanding. It has been placed here to put the subsequent research in
`context and because the selectivity between prolylpeptidases may be a differ-
`entiating factor for drug candidates. Regardless of the outcome of all these
`evaluations,
`it would be prudent to remain cautious as development of
`DPPIV inhibitors for the treatment of type 2 diabetes proceeds (Fig. 2.2).
`
`STRUCTURAL BIOLOGY
`
`DPPIV has proven to be a drug target that has benefited from structural
`biology guidance, in particular crystallography. Initially, several homology
`models were constructed that provided preliminary insight into possible
`structural features [46–48]. Following was an extensive effort that provided
`
`Page 7 of 47
`
`

`
`70
`
`DPPIV INHIBITION
`
`X-ray crystal structures of recombinant human DPPIV [49–53], natural
`source porcine DPPIV [54], natural source rat DPPIV [55] and bacterial
`DPPIV [56]. Both the apo structure and those with bound inhibitors were
`examined. Many features were found as previously predicted by comparison
`with related peptidases. However, there were certainly some surprises. To
`summarize, DPPIV exists as a dimer with two domains, an a/b hydrolase
`domain and a rather rare eight-bladed propeller domain [57] with a 30–45 A˚
`cavity between them. Inhibitors bind within the cavity next to the catalytic
`triad. Two openings access the cavity, a funnel-shaped opening through the
`b-propeller and a larger opening between the hydrolase and propeller do-
`mains that is believed to be the entry route of substrates. Both are negatively
`charged, which attract the positively charged amine moiety found in all
`inhibitors. Glutamic acid residues 205 and 206 form a salt bridge to the
`amine group in the P2 portion of inhibitors [58] and this interaction is
`responsible for orienting the N-terminal portion of peptide substrates for
`cleavage. Steric constraints limit the enzyme to dipeptide cleavage. Aspara-
`gine 710 and arginine 125 hydrogen bond to the carbonyl of a P2 amino
`acid. A small hydrophobic pocket is optimally filled with small flat rings,
`such as pyrrolidine or phenyl of the inhibitor, and the oxyanion hole is
`formed by two tyrosines. Tyr547 was found to be as critical as any of the
`catalytic triad for full enzymatic activity for its ability to stablilize the oxy-
`anion intermediate [59]. Interactions for specific inhibitors will be elaborated
`in the medicinal chemistry discussions of those compounds (Fig. 2.3).
`As previously mentioned, the active form of the enzyme is a dimer and once
`formed, there is no equilibration back to the monomeric species [60]. Hydro-
`phobic forces were determined to be responsible for dimer formation with
`residues Phe713, Trp734 and Tyr735, all found to be critical. The extent of
`glycosylation was determined not to be a factor affecting dimer formation [61].
`DPPIV is an amazingly dynamic enzyme. As mentioned above, there are
`two channels which access the catalytic region, and analogy to the similar
`enzymes tricorn protease and prolyl oligopeptidase suggested that substrate
`entrance and cleavage products’ exit could be through the b-propeller tunnel
`
`HN
`
`O
`
`P1'
`
`R3
`
`NH
`
`O
`
`P1
`
`R2
`
`X
`
`N
`
`O
`
`R1
`
`N
`H
`
`P2
`
`Fig. 2.3 Schematic of DPPIV substrate.
`
`Page 8 of 47
`
`

`
`P. E. WIEDEMAN
`
`71
`
`[54]. More recent studies have determined that the side channel more likely
`acts as both entrance and exit, with an expansion of the channel to allow
`both ready access and egress [62]. Conformational change was also noted
`for Tyr547 (Tyr548 rat) in the active site that accommodated various in-
`hibitor types [55, 62]. The flexibility of this tyrosine is specific to DPPIV and
`is likely a selectivity determinant over the similar enzymes DPP8 and
`DPP9. Further selectivity may be achieved with polar residues in the P10
`position [63].
`The firm footing on structural understanding of DPPIV has allowed vir-
`tual docking exercises to suggest novel inhibitors [64, 65]. Subsequent eval-
`uation in in vitro assays established which chemotypes were suitable starting
`points for future structure-based design efforts.
`
`MEDICINAL CHEMISTRY AND PRECLINICAL STUDIES
`
`Inhibition DPPIV has proven to be a drug target that has successfully
`brought together all the drug discovery disciplines. Many institutions and
`companies have contributed to the wealth of information both in the pub-
`lished and patent literature. A quick search of the patent literature will find
`in excess of 300 patents filed by more than 60 companies and institutions.
`That body of information is too extensive to be included in this chapter. The
`study of DPPIV inhibition has been quite active over the past decade and
`the medicinal chemistry effort has been reviewed at time points throughout
`[66–71]. Here we will concentrate on journal publications, some meeting
`abstracts and the occasional patent application to illustrate the drug dis-
`covery story of DPPIV inhibition. A brief section will describe some of the
`early ground breaking studies that set the stage for the explosive research
`development in this field. Structural class will loosely collect the more con-
`temporary medicinal chemistry and preclinical information.
`
`EARLY DPPIV INHIBITORS
`
`Inhibitors of DPPIV have long been sought as tools to elucidate the func-
`tional significance of the enzyme. The first inhibitors were characterized in
`the late 1980s and 1990s. Each was important in establishing an early
`structure–activity relationship (SAR) for subsequent investigation. It should
`be noted that the inhibitors fall into two main classes, those that interact
`covalently with DPPIV and those that do not.
`Since DPPIV is a protease, it is not unexpected that inhibitors would
`likely have a peptidic nature and this theme has carried through to
`
`Page 9 of 47
`
`

`
`72
`
`DPPIV INHIBITION
`
`contemporary research. A number of non-covalent inhibitors were charac-
`terized in the same paper [72]. The tripeptides diprotin A (Ile-Pro-Ile (6))
`and diprotin B (Val-Pro-Leu (7)) were identified as modest inhibitors with
`IC50s of 8 and 920 mM, respectively. Although these simple peptides were
`not anticipated to be metabolically stable, the recognition element of proline
`in the penultimate position was established. Lys[Z-4-NO2]-pyrrolidide (5)
`indicated that a simple, rather flat heterocycle was sufficient to occupy the
`P1 site where DPPIV favours proline and alanine. The IC50 was again
`modest at 2 mM, and the compound showed significant toxicity. Two other
`compounds with five-membered heterocycles mimicking proline in the P1
`pocket were Val-pyrrolidide (9) and Ile-thiazolidide ((10), P32/98) with
`IC50s of 6.0 and 2.8 mM, respectively. These compounds would become two
`of the more extensively studied inhibitors.
`A natural product isolate, TMC-2 (11), from Aspergillus orzae A374 was
`identified as a specific inhibitor (7.7 mM) of DPPIV [73]. A solid-phase
`combinatorial chemistry effort reduced the size and determined the critical
`core structure, TSL-225 (12), that maintained equivalent potency (5.7 mM)
`(Fig. 2.4).
`
`O
`
`OH
`
`iPr
`
`HN
`
`O
`
`O
`
`N
`
`H2N
`
`iPr
`
`O
`
`H2N
`
`N
`
`S
`
`Et
`
`Me
`
`P32/98 (10)
`
`diprotin B (7)
`
`O
`
`iPr
`
`N
`
`(9)
`
`H2N
`
`H
`N
`
`H2N
`
`N
`
`O
`
`O
`
`OH
`
`OH
`
`O
`
`HN
`
`Et
`
`Me
`
`O
`
`N
`
`H2N
`
`Et
`
`Me
`
`O
`
`diprotin A (6)
`
`O
`
`N
`
`NH2
`
`HN
`
`O
`
`O
`
`(5)
`
`OMe
`
`HO
`
`OH
`
`H
`N
`
`OH
`
`OH
`
`OH
`
`O
`
`NH
`
`H2N
`
`N
`
`O
`
`O
`
`O2N
`
`TMC-2 (11)
`
`TSL-225 (12)
`
`Fig. 2.4 Peptidic and non-covalent inhibitors.
`
`Page 10 of 47
`
`

`
`P. E. WIEDEMAN
`
`73
`
`Inhibitors that interacted covalently with DPPIV were also investigated at
`an early stage. N-Peptidyl-O-aroyl hydroxylamines exemplified by (13) were
`weak, irreversible inhibitors found to suffer from hydrolytic instability [74].
`A series of dipeptide phosphonates were likewise irreversible inhibitors [75].
`Although, the potency was weak, Ki ¼ 236 mM for (14), the compounds
`addressed the issue faced by many transition-state inhibitors, the intramo-
`lecular cyclization of the N-terminus to the electrophilic moiety requisite for
`the active site serine. Phosphonate esters react poorly with nitrogen
`nucleophiles and the members of this series were stable in pH 7.8 buffer
`for hours to days. Selectivity was observed for DPPIV over the proteases
`and esterases chymotrypsin, trypsin, human leukocyte elastase, porcine
`pancreatic elastase, acetylcholinesterase, papain and cathepsin B.
`Applying work established previously with inhibitors of another class of
`post-proline-cleaving serine proteases, the IgA proteases, dipeptides incor-
`porating the a-amino boronic acid analogue boroPro were the first truly
`potent inhibitors with Ki values in the low nanomolar range [76]. The SAR
`established in this work confirmed the requirement of a basic primary or
`secondary amine at the N-terminus. The boronic acid electrophile exhibited
`slow binding kinetics characteristic of many transition state DPPIV inhib-
`itors. Unfortunately, the compounds were extremely unstable to intramo-
`lecular cyclization in a neutral aqueous environment with Pro-boroPro (15)
`having a t1/2 of 1.5 h. There has been some recent work extending the
`boroPro motif into a P2 N-alkyl-glycine that becomes a common group to
`be described below [77].
`A real breakthrough occurred with the discovery of 2-cyanopyrrolidines
`as the first potent, selective and chemically stable inhibitors [78–80]. These
`competitive inhibitors were presumed to react with the active site serine,
`thus
`forming an imidate adduct. 2-Cyanopyrrolidine
`(16) had a
`Ki ¼ 200 nM while showing weak inhibition against the related enzymes
`DPPII (110 mM) and prolyl oligopeptidase (22% inhibition at 400 mM).
`Little loss of activity was observed after incubation of solutions at 37 1C for
`20 h. The cyanopyrrolidine was to become the sought serine hook that pro-
`vided sufficient selectivity and solution stability for drug development.
`
`Ferring
`
`There were two papers by this Research Institute in 1996 that firmly es-
`tablished the 2S-cyanopyrrolidide as a potent pharmacophore for DPPIV
`inhibition. These studies were directed at finding modulators of T-cell
`activity, a reflection of most DPPIV research at that time. The first paper
`established the superiority of the cycloalkyl side chain in the L-configuration
`in the P2 position delivering both potency and stability, as exemplified by
`
`Page 11 of 47
`
`

`
`74
`
`DPPIV INHIBITION
`
`O
`
`H
`N
`
`O
`
`O
`
`O
`
`N
`
`NH2
`
`Cl
`
`O
`P
`
`O
`
`H2N
`
`O
`
`O
`
`N
`
`Me
`
`NO2
`
`Cl
`
`(14)
`
`(13)
`
`HN
`
`O
`
`Ph
`
`O
`
`O
`
`CN
`
`O
`
`CN
`
`O
`
`CN
`
`H2N
`
`N
`
`(17)
`
`H2N
`
`N
`
`Et
`
`Me
`
`S
`
`(18)
`
`N
`
`Me
`
`(16)
`
`N
`
`H2N
`
`O
`
`HO
`
`B OH
`
`NH
`
`Pro-boroPro (15)
`
`Fig. 2.5 Covalent inhibitors.
`
`(17) [79]. The second provided an exploration of the SAR associated with
`the P1 position. Various five- and six-membered saturated heterocycles
`containing a combination of nitrogen, sulfur and oxygen were prepared,
`each having a nitrile at the 2-position [80]. The most potent compound was
`the thiazolidide (18).
`To reinforce a key difference, the DPPIV inhibitors discussed herein are
`divisible into two classes, those that form a covalent interaction with the
`enzyme and those that do not. This distinction will persist throughout the
`remaining part of this chapter (Fig. 2.5).
`
`COVALENT DPPIV INHIBITORS: P2 SECONDARY AMINE ANALOGUES
`
`Novartis
`
`These investigators were aware of the potency and selectivity achieved with
`the dipeptide mimetic cyanopyrrolidides having a primary a-amino acid in
`the P2 binding site described above. They also knew that N-methylglycine
`was accepted by DPPIV in the P2 site [81]. Combining these two features
`established the N-alkylated-glycylcyanopyrrolidides as a novel DPPIV in-
`hibitor chemotype and established DPPIV as a drug target for large pharma
`[82]. Libraries of 2-(S)-pyrrolidinecarbonitriles were produced efficiently
`with either five-step solid-phase or three-step solution-phase sequences.
`When the P2 glycine was N-alkylated with 2-aminoethyl-5-nitropyridine, a
`potent (IC50 ¼ 8 nM) inhibitor (19) in this new chemotype was achieved.
`SAR exploration led to NVP-DPP728 (20) (IC50 ¼ 22 nM). It proved to
`
`Page 12 of 47
`
`

`
`P. E. WIEDEMAN
`
`75
`
`be chemically stable against intramolecular cyclization and selective against
`other closely related peptidases known at the time. Oral administration of
`(20) (1 mmol/kg) produced a successful result in an oral glucose-tolerance
`test (OGTT) in cynomolgus monkey. DPPIV activity was inhibited by
`89% while both peak plasma glucose levels and glucose area under the curve
`(AUC) were reduced. Oral bioavailability was >70% in both rat and mon-
`key. Steady-state volumes of distribution suggested that the drug was
`primarily distributed to body fluids. Clearance was moderate, and the half-
`life of 0.85 h in both monkey and man suggested that (20) might be best
`dosed in conjunction with a meal.
`Early published results had reported several key SAR and biochemical
`features of NVP-DPP728 (20) [83]. The D-antipode (Ki ¼ 5.6 mM), des-
`cyano (Ki ¼ 15.6 mM) and amide (Ki ¼ 320 mM) analogues of NVP-DPP728
`(Ki ¼ 0.11 mM) were compared. There was 3.9 kcal/mol of binding energy
`associated with the cyano group in the L-configuration. The inhibition
`kinetics was consistent with a two-step mechanism where an initial loose
`inhibitor–enzyme complex slowly isomerizes to a tight complex. SAR
`features included an understanding of hydrophobic or van der Waals in-
`teractions of the pyrrolidine in the P1 pocket. H-bonding and ionic inter-
`actions stabilized the P2 carbonyl and amine groups, respectively. P2 site
`side-chain interactions were hydrophobic in the S2 pocket. The nitrile was
`either considered to form a transient imidate with catalytic site serine or
`form some type of dipole–hydrogen bond interaction, either explained the
`binding energy.
`Additional SAR of NVP-DPP728 and the path to LAF237 (21) was
`combined in one paper [84]. Within NVP-DPP728, the P2 glycine was
`greatly preferred over the b-alanine analogue. The diamine chain length of
`two carbons was optimal. In addition to the nitro group, chlorine and nitrile
`were accepted on the terminal pyridine. Replacement of the 2-aminopyri-
`dine with pyrimidine, aniline, or phenyl was deleterious to binding potency.
`Based on selectivity considerations, NVP-DPP728 became the first devel-
`opment candidate.
`SAR continued with larger aliphatic side chains and rings in the P2 site,
`with good potency maintained with a fully substituted carbon adjacent to
`the P2 amine. This progressed to the investigation of multiple cyclic systems
`and culminated with the discovery that adamantyl groups furnished
`low nanomolar inhibitors. Studies on metabolites indicated that hydro-
`xylation of the adamantyl groups was well tolerated. This led to the syn-
`thesis of the 3-hydroxylated-1-aminoadamantane analogue (21) (LAF237,
`IC50 ¼ 3.5 nM), which avoided the introduction of a chiral centre. LAF237
`had the greatest solution stability to intramolecular cyclization of any tran-
`sition-state mimetic synthesized at that time. Further functionalization of
`
`Page 13 of 47
`
`

`
`76
`
`DPPIV INHIBITION
`
`Table 2.1 PEPTIDASE SELECTIVITY OF P2 PRIMARY AND SECONDARY AMINE
`INHIBITORS
`
`Compound
`
`DPPIV IC50 (nM)
`
`DPP8 IC50 (nM)
`
`DPPII IC50 (nM)
`
`68,985
`39,873
`26,520
`>100,000
`
`O
`
`CN
`
`N
`
`HN
`
`NH
`
`N
`
`NVP-DPP728 (20)
`DPPIV IC50 = 22 nM
`
`MeO
`
`Me
`
`N
`
`O
`
`CN
`
`NH
`
`N
`
`(22)
`DPPIV IC50 = 49 nM
`DPP8 IC50 >100000 nM
`
`2,283
`27
`4,573
`14,219
`
`NC
`
`P32/98 (10)
`(17)
`NVP-DPP728 (20)
`LAF237 (21)
`
`1,660
`12
`53
`51
`
`O
`
`CN
`
`N
`
`HN
`
`NH
`
`N
`
`O2N
`
`(19)
`DPPIV IC50 = 8 nM
`
`O
`
`CN
`
`N
`
`HN
`
`LAF237, vildagliptin (21)
`DPPIV IC50 = 3.5 nM
`
`HO
`
`Fig. 2.6 N-alkylated glycyl-2-cyanopyrrolidides.
`
`the adamantyl hydroxyl group decreased potency. LAF237 was selective
`over similar peptidases screened at the time and a recent report stated
`LAF237 to be a 9 mM (IC50) inhibitor of DPP8 [85]. [Note: The IC50 value
`reported here for DPPIV and DPP8 inhibition differs from that reported in
`Table 2.1. as a reflection of variance in procedures between laboratories.
`The constant, Ki, is preferred for comparison of data when available.] The
`pharmacokinetics of LAF237 was similar to NVP-DPP728, except the half-
`life was 2.6-fold longer. Using obese Zuckerfa/fa rats as a model of type 2
`diabetes in OGTT, and administration of LAF237 (10 mmol/kg) resulted in
`90% DPPIV inhibition over the 90-min study, a 60% higher level of GLP-1,
`a decrease in glucose excursion and a doubling of peak insulin levels. Com-
`parable studies in cynomologus monkey dosed with LAF237 (1 mmol/kg)
`showed >50% DPPIV inhibition for at least 10 h compared to 4–5 h for
`NVP-DPP728. LAF237 was brought forward as a development candidate
`suitable for once daily dosing (Fig. 2.6).
`
`Page 14 of 47
`
`

`
`P. E. WIEDEMAN
`
`77
`
`National Health Research Institutes, Taiwan
`
`These researchers used a selectivity comparison of known DPPIV inhibitors
`as their starting premise. The key comparison was between DPPIV and
`DPP8, as other proteases were not inhibited to a significant degree. It was
`found that P2 primary amino analogues (P32/98 (10) and (17)) were not
`selective over DPP8 while two P2 N-substituted glycine analogues were
`(Table 2.1) [86]. Based on these results, several

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