throbber
SAXA-DEF-00324
`
`AstraZeneca Exhibit 2003
`Mylan v. AstraZeneca
`IPR2015-01340
`
`Page 1 of 26
`
`

`
`SAXA-DEF-00325
`
`Page 2 of 26
`
`Page 2 of 26
`
`

`
`CHAPTER 1
`
`The Discovery of the Dipeptidyl
`Peptidase-4 (DPP4) Inhibitor
`Onglyzat: From Concept to
`Market
`
`JEFFREY A. ROBL AND LAWRENCE G. HAMANN
`
`Bristol-Myers Squibb Research & Development, Department of Discovery
`Chemistry – Metabolic Diseases, P.O. Box 5400, Princeton, NJ 08543, USA
`
`1.1 Introduction
`
`The prevalence of diabetes in developed and now emerging countries represents
`a significant health burden to a large portion of the world’s population. Type-2
`diabetic patients, characterized in part by elevated fasting plasma glucose of
`1 (7.0 mmol L
`1) and glycosylated hemoglobin (HbA1c) Z 6%,
`4125 mg dL
`are at increased risk for the development of both microvascular (retinopathy,
`neuropathy, nephropathy) as well as macrovascular complications (myocardial
`infarction, stroke). As such, diabetes is the leading cause of blindness, kidney
`failure, and limb amputation worldwide.1 Diabetes is a progressive disease,
`with morbidity and mortality risk increasing with both duration and severity of
`hyperglycemia. Additionally, diabetes is also now impacting different popula-
`tion sectors (adolescents, developing countries) not typically associated with
`the disease 30 years ago. Consequently, the continually increasing diabetes
`prevalence is placing greater strain on both health care systems and economies
`on a global scale. In 2007 alone, studies have shown that diabetes cost the US
`
`RSC Drug Discovery Series No. 4
`Accounts in Drug Discovery: Case Studies in Medicinal Chemistry
`Edited by Joel C. Barrish, Percy H. Carter, Peter T. W. Cheng and Robert Zahler
`r Royal Society of Chemistry 2011
`Published by the Royal Society of Chemistry, www.rsc.org
`
`1
`
`Downloaded on 05/02/2015 20:03:18.
`
`Published on 30 September 2010 on http://pubs.rsc.org | doi:10.1039/9781849731980-00001
`
`SAXA-DEF-00326
`
`Page 3 of 26
`
`Page 3 of 26
`
`

`
`Cl
`
`OEt
`
`Chapter 1
`
`HO
`
`H2N
`
`3
`
`N
`
`O
`
`CN
`
`H
`
`H O
`
`2
`
`O
`
`OH
`
`HO
`
`HO
`
`N
`
`CO2H
`
`O
`
`O
`
`O
`
`N
`
`e
`
`O M
`
`1
`
`OMe
`
`2
`
`Figure 1.1 Late-stage development candidates from the Bristol-Myers Squibb dia-
`betes research portfolio.
`
`economy $174 billion in medical expenses and lost productivity.2 While death
`rates related to heart disease, stroke, and cancer have all decreased since 1987,
`the death rate due to diabetes has increased by 45% during this same period.3
`Thus, the discovery and development of new therapies for treating and pre-
`venting diabetes continue to be a major emphasis of health care companies.
`In response to this landscape, the Discovery organization at Bristol-Myers
`Squibb (BMS) made the strategic decision to refocus efforts in the late 1990’s
`towards identifying and progressing novel targets for the treatment of diabetes.
`This was in part aimed at building upon BMS’s already established presence in
`the anti-diabetes market through the Glucophaget franchise and in recognition
`of the significant unmet medical need for novel, more efficacious, and well
`tolerated treatments for the disease. It was from these efforts that advanced
`clinical candidates such as muraglitazar (1, Pargluvat, dual PPAR agonist),4
`dapagliflozin (2, SGLT2 inhibitor),5 and saxagliptin (3, Onglyzat, DPP4 inhi-
`bitor)6 were discovered within the BMS Discovery organization (Figure 1.1).
`
`1.2 Modulation of GLP-1 in the Treatment of
`Diabetes
`
`At the start of this effort, several oral anti-diabetic agents (OADs) were
`available to patients suffering from type-2 diabetes. These included hepatic
`glucose suppressors (e.g. metformin), insulin secretagogues (e.g. sulfonylureas),
`glucose absorption inhibitors (e.g. acarbose), and insulin sensitizers (e.g. thia-
`zolidinediones or TZDs such as rosiglitazone and pioglitazone). While all have
`shown utility in lowering HbA1c levels in diabetic patients, current OADs come
`with a variety of safety and/or tolerability issues. The biguanindes such as
`metformin, currently the most widely prescribed therapy for diabetes, have
`issues related to gastrointestinal (GI) tolerability and lactic acidosis.7 Sulfo-
`nylurea treatment is often accompanied by higher incidences of hypoglycemia
`and weight gain,8 while glucose absorption inhibitors exhibit modest efficacy
`and GI disturbance.9 Finally, TZDs have been associated with edema, wor-
`sening of congestive heart failure, negative effects on bone fracture rate, and, in
`recent studies, mixed results regarding cardiovascular (CV) safety profiles.10
`
`Downloaded on 05/02/2015 20:03:18.
`
`Published on 30 September 2010 on http://pubs.rsc.org | doi:10.1039/9781849731980-00001
`
`View Online
`
`SAXA-DEF-00327
`
`Page 4 of 26
`
`Page 4 of 26
`
`

`
`The Discovery of the Dipeptidyl Peptidase-4 (DPP4) Inhibitor Onglyzat
`
`3
`
`With this background in mind, we sought to identify new targets which would
`not only provide an efficacious alternative mechanism for lowering blood
`glucose and HbA1c levels, but would also present an opportunity for achieving
`a superior safety and tolerability profile when compared to current standards of
`care. Ideally, such a drug would be suitable for combination with existing
`agents, as poly-pharmacology with multiple OADs is emerging as the standard
`treatment paradigm for type-2 diabetes therapy.
`Glucagon like peptide-1 (GLP-1) is a 30-amino acid peptide incretin hor-
`mone derived from processing of pro-glucagon and is secreted by the L-cells of
`the intestinal mucosa in response to glucose stimulation. Since the early 1990’s,
`GLP-1 had been known to be a potent insulin secretagogue and glucagon
`suppressor, with robust anti-diabetic and pro-satiety effects in diabetic
`humans,11,12 but efforts to advance GLP-1 itself as a pharmaceutical agent
`were hampered by its extremely short pharmacokinetic half-life in vivo (plasma
`t1/2E2 min). As a result, considerable effort in the drug discovery community
`was expended toward the identification of small-molecule GLP-1 receptor
`agonists that would capture the beneficial effects of GLP-1 while exhibiting oral
`bioavailability and a superior pharmacokinetic duration of action. Unfortu-
`nately, efforts to identify such small-molecule agonists have to date been
`unsuccessful, due in part to a dearth of viable bona fide screening hits.13 In light
`of this shortcoming, a number of pharmaceutical and biotech companies have
`advanced subcutaneously administered, peptide GLP-1 receptor agonists with
`superior duration of action in vivo. Among the most advanced agents are
`exenatide (Byettat)14 and liraglutide (Victozat),15 both of which have been
`approved by regulatory agencies for the treatment of type-2 diabetes. While
`these drugs are effective in lowering HbA1c and demonstrate a net beneficial
`effect on weight gain and other CV risk factors, they require parenteral
`administration (once or twice daily dosing), and patient uptake of these agents
`has been limited despite their robust efficacy and promising safety profile.
`
`1.3 Dipeptidyl Peptidase-4 as a Target for Diabetes
`Treatment
`
`While the advancement of orally active, small-molecule GLP-1 receptor ago-
`nists remains elusive, another opportunity to modulate GLP-1 receptor activity
`in vivo focused on preventing the degradation of endogenous GLP-1 with small-
`molecule inhibitors of the primary peptidase responsible for the in vivo
`degradation of GLP-1, dipeptidyl peptidase-4 (DPP4), a non-classical serine
`protease.16 Our initial interest in DPP4 inhibitors was piqued by a report from
`Holst and Deacon, wherein the authors outlined a compelling argument for the
`utility of DPP4 inhibition in the treatment of diabetes, primarily via the
`potentiation of endogenous GLP-1.17 DPP4 belongs to a family of aminodi-
`peptidases and is both a cell surface and circulating enzyme. Historically, it had
`also been identified as the lymphocyte cell surface marker CD26, and as such
`DPP4/CD26 exhibits pharmacology related to cell membrane-associated
`
`Downloaded on 05/02/2015 20:03:18.
`
`Published on 30 September 2010 on http://pubs.rsc.org | doi:10.1039/9781849731980-00001
`
`View Online
`
`SAXA-DEF-00328
`
`Page 5 of 26
`
`Page 5 of 26
`
`

`
`4
`
`Chapter 1
`
`activation of intracellular signal transduction pathways and cell–cell interac-
`tion in addition to its peptidase enzymatic activity.18 It is widely believed that
`the signaling function of DPP4/CD26 is distinct from its enzymatic function.
`The concept of generating targeted protease/peptidase inhibitors as thera-
`peutic agents is well documented in the literature.19 In the majority of cases,
`selective enzyme inhibitors have been used to prevent the conversion of an
`endogenous ‘‘non-functional’’ peptide/protein precursor (e.g. angiotensin I) to
`a physiologically active peptide/protein (e.g. angiotensin II), thereby attenu-
`ating formation of the protagonist bio-active enzyme product to effect ameli-
`oration of the disease state. Use of such approaches has led to marketed drugs
`for numerous indications, including ACE and renin inhibitors for hyperten-
`sion,20 HIV protease inhibitors for AIDS,21 and thrombin inhibitors for
`DVT.22 Less prevalent are approaches targeting proteases/peptidases that
`degrade endogenous substrates which are known to exert a beneficial effect. For
`example, neutral endopeptidase (NEP) proteolytically degrades the endogen-
`ous vasodilator atrial natriuretic peptide (ANP) to inactive fragments. By
`retarding this degradation, NEP inhibitors have found use in the treatment of
`hypertension.23 In common with NEP, where inhibition of protease mediated
`protein degradation was the pharmacological objective, BMS and several other
`research groups engaged in the search for DPP4 inhibitors to maximize the
`beneficial effects of endogenously released GLP-1.
`
`1.4 Early Inhibitors of DPP4
`
`To jump-start the BMS DPP4 chemistry program, the group was able to
`capitalize on groundwork laid in the mid-late 1990’s when several potent
`inhibitors of DPP4/CD26 were reported that could be classified as either
`‘‘irreversible’’ or ‘‘reversible’’, depending on the mechanism of inhibition
`(Figures 1.2 and 1.3). Hydroxamates such as 4 were proposed to be both
`substrates and inhibitors of DPP4, presumably via direct covalent modification
`of the enzyme through the active-site serine residue (Ser630).24 Phosphate-
`based inhibitors such as 5 were also reported to undergo covalent addition to
`DPP4 but exhibited weak potency.25 The interesting boronate-based inhibitors
`(e.g. 6), originally advanced by the Tufts University and Boehringer Ingelheim
`groups, exhibited exceptionally high inhibitory activity in vitro, presenting
`
`N N
`
`7
`
`B
`
`OH
`OH
`
`H H
`
`Me
`
`N
`
`O
`
`P
`
`O
`O
`
`O
`
`H2N
`NO2
`
`5
`
`Cl
`
`O
`
`H2N
`
`Cl
`
`N
`
`HO
`
`O
`
`6
`
`B
`
`OH
`
`O
`
`O
`
`NH
`
`4
`
`H2N
`
`Me
`
`N
`
`O
`
`O
`
`Figure 1.2 Early examples of ‘‘irreversible’’ DPP4 inhibitors.
`
`Downloaded on 05/02/2015 20:03:18.
`
`Published on 30 September 2010 on http://pubs.rsc.org | doi:10.1039/9781849731980-00001
`
`View Online
`
`SAXA-DEF-00329
`
`Page 6 of 26
`
`Page 6 of 26
`
`

`
`The Discovery of the Dipeptidyl Peptidase-4 (DPP4) Inhibitor Onglyzat
`
`5
`
`N
`
`O
`
`CN
`
`NH
`
`HN
`
`N
`
`11 (X = CF3)
`12 (X = CN)
`
`N
`
`H2N
`
`H2N
`
`O
`
`8
`
`S
`
`N
`
`O
`
`9
`
`H2N
`
`N
`
`O
`
`10
`
`CN
`
`X
`
`O
`
`N
`
`O
`
`N
`
`O
`
`NH
`
`14
`
`NH
`
`NH
`
`13
`
`Figure 1.3 Early examples of ‘‘reversible’’ DPP4 inhibitors
`
`themselves as ‘‘transition-state’’ inhibitors which presumably form tetrahedral
`boronate esters involving the Ser630 hydroxyl group.26–28 As such, many of
`these compounds exhibited slow tight-binding kinetics with Koff rates of several
`days (versus seconds/minutes with non-covalently bound inhibitors). However,
`these compounds also suffered from poor solution stability arising from
`intramolecular cyclization of the terminal primary amine with the boronate,
`affording an inactive product (e.g. 7). The propensity of compounds of this
`chemotype to undergo internal cyclization limited their viability as drug
`candidates.
`Due to the uncertain risks associated with advancing irreversible inhibitors as
`drug candidates, the team viewed the reversible inhibitors exploited by the
`Mount Sinai, Probiodrug, Ferring Research, and Novartis scientific teams as
`more attractive starting points for a lead finding effort (Figure 1.3). Probiodrug
`had described simple Ile-pyrrolidides (8) and Ile-thiazolidides (9, later advanced
`to the clinic by Merck and Probiodrug as P32/98) which exhibited in vitro
`potency in the nanomolar range, were chemically stable, and, in the case of 9,
`demonstrated glucose area under the curve (AUC) reductions in Zuckerfa/fa rats
`in an oral glucose tolerance test (oGTT).29–31 Reports from Li et al. at Mount
`Sinai highlighted early examples of nitrilo-pyrrolidines specifically designed as
`inhibitors of DPP4.32 Equally intriguing was the work described by Ferring in
`which nitrilo-pyrrolidines such as 10 were identified as exceptionally potent
`inhibitors of the enzyme.33,34 Initially targeted as agents for immunomodula-
`tion (via CD26 inhibition), these compounds represented ‘‘drug-like’’ scaffolds
`and exhibited exceptional
`inhibitory potency. Additionally, a contempo-
`raneous patent application from Novartis35 described the structure of com-
`pound 11 (a related analogue 12 would later be disclosed as Novartis’ first
`clinical compound, DPP-728)36 and its ability to increase plasma insulin in
`
`Downloaded on 05/02/2015 20:03:18.
`
`Published on 30 September 2010 on http://pubs.rsc.org | doi:10.1039/9781849731980-00001
`
`View Online
`
`SAXA-DEF-00330
`
`Page 7 of 26
`
`Page 7 of 26
`
`

`
`6
`
`Chapter 1
`
`fasted, high-fat fed rats in an oGTT. The inhibitors represented by compounds
`8–12 provided useful insights for the design of DPP4 inhibitors at BMS.
`At the initiation of the program, there were still many unknown factors
`related to the pharmacology and safety of DPP4 inhibition. It was clear from
`earlier work that Fischer 344 rats possessing a naturally occurring loss-of-
`function mutation of the DPP4 gene were healthy, viable, and free of serious
`immunological complications.37 It was later shown that these rats also exhib-
`ited a favorable metabolic phenotype on a high-fat diet and demonstrated
`improved glucose tolerance and GLP-1 secretion.38 Thus, complete ablation of
`DPP4 did not appear to represent a serious safety concern in rats, but rather the
`Fischer rat provided support for the concept that inhibition of DPP4 could be
`both safe and efficacious. Others questions still remained. Was high selectivity
`for DPP4 versus other related peptidases (e.g. DPP8, DPP9, FAP, etc.) an
`absolute requirement for this target? Would inhibition of DPP4 potentiate
`other endogenous peptides, leading to unintended deleterious (or beneficial)
`consequences? Would potentiating endogenous GLP-1 (versus exogenous
`administration) be sufficient to affect a robust anti-diabetic response in
`humans? Finally, what potential mechanism-based toxicological effects, if any,
`would be seen upon chronic administration of a DPP4 inhibitor?
`In light of limited literature in the field, and with no reports of a compound
`having advanced to clinical trials, these questions would ultimately need to be
`addressed during the execution of our discovery and subsequent clinical
`development programs. Despite the unknowns, the positive aspects of this
`target were numerous. Potent small-molecule inhibitors with systemic exposure
`upon oral dosing were known. Although limited, DPP4 inhibitors had
`demonstrated pharmacodynamic efficacy in genetic animal models of type 2
`diabetes in preliminary pharmacological studies. Preclinical proof-of-concept
`for the anti-diabetic actions of GLP-1 was already established and suggested a
`low potential for hypoglycemia. In vitro assays and several in vivo models were
`already described in the literature, enabling rapid program initiation. It was
`against
`this backdrop that significant medicinal chemistry and biology
`resources at BMS were deployed on this newly emerging target in the early
`months of 1999.
`
`1.5 Design of BMS’s DPP4 Medicinal Chemistry
`Program
`
`Given the attractiveness of DPP4 as a therapeutic target, it was anticipated that
`this field would soon become highly competitive, and we therefore sought to
`accelerate the program. High-throughput screening (HTS), routinely a key
`component of drug discovery programs, was deemed as too time consuming to
`rapidly afford a chemical starting point. Thus, we decided to initially adopt a
`design optimization approach,
`improving upon the leads reported by the
`Probiodrug, Ferring, and Novartis research groups; HTS would later be used
`to provide leads for a second-generation effort. From a potency perspective, the
`
`Downloaded on 05/02/2015 20:03:18.
`
`Published on 30 September 2010 on http://pubs.rsc.org | doi:10.1039/9781849731980-00001
`
`View Online
`
`SAXA-DEF-00331
`
`Page 8 of 26
`
`Page 8 of 26
`
`

`
`The Discovery of the Dipeptidyl Peptidase-4 (DPP4) Inhibitor Onglyzat
`
`7
`
`Figure 1.4 Rationale for generation of conformationally restricted DPP4 inhibitors.
`
`nitrilo-pyrrolidines were deemed to be highly attractive, but were reported to
`exhibit modest pharmacokinetic duration of action and suffered from chemical
`instability.33,34 In solution, the proximal amino group attacks the nitrile
`functionality (see Figure 1.3), eventually leading to the intermediate cyclic
`imidate 13 and ultimately the diketopiperzine 14, both of which are inactive
`versus DPP4. Addressing this issue was viewed as a critical component of the
`medicinal chemistry effort due to considerations regarding both the half-life of
`the compound in vivo, and for high purity processing of the active pharma-
`ceutical ingredient (API) on large scale in a drug manufacturing setting.
`From earlier work by Lin et al.,39 it was demonstrated that replacement of
`the prolyl amide bond with a fluoroalkene isostere resulted in the generation of
`potent DPP4 inhibitor 15 (Figure 1.4). This finding was significant in that it
`suggested that the critical prolyl and amino pharmacophores in 16 may be
`conformationally locked in an extended arrangement which is favorable for
`enzyme inhibition. In addition, because incorporation of the alkene prohibits
`intramolecular attack of the amine onto the acyl hydroxamate, the finding
`suggested novel paths for inhibitor design to retard intramolecular cyclization.
`Taking a cue from earlier work performed at BMS in the design of dual
`ACE/NEP inhibitors,40 we applied the concept of conformationally restricted
`dipetide mimetics in our search for novel inhibitor chemotypes. Many of these
`cores seemed to possess the critical elements required for DPP4 inhibition,
`including a prolyl amide group and a charged amino functionality at the P2
`position. It was hoped that locking the inhibitor conformation by this approach
`would not only enhance binding affinity, but also prevent inactivating cycli-
`zation in compounds possessing an electrophilic pharmacophore (e.g. nitrile,
`phosphate, etc.) on the proline ring. Unguided by the availability of a DPP4
`X-ray crystal structure at that time, our design efforts led to a variety of
`different bi- and monocyclic dipetide mimetics, generically represented in
`Figure 1.4. Unfortunately, all of the compounds generated in this series were
`inactive against DPP4, which we attributed to either incorrect conformational
`geometry required for inhibition, or to steric intolerance for substitution on the
`proline ring. The latter hypothesis was supported by the poor activity exhibited
`by the simple methyl-substituted prolyl derivative 17 as compared to its
`unsubstituted counterpart 8. Interestingly, a recent report from Phenomix
`disclosing 5,5-fused bicyclic lactams such as 18 as potent DPP4 inhibitors
`provides validation for this initial approach.41
`
`Downloaded on 05/02/2015 20:03:18.
`
`Published on 30 September 2010 on http://pubs.rsc.org | doi:10.1039/9781849731980-00001
`
`View Online
`
`SAXA-DEF-00332
`
`Page 9 of 26
`
`Page 9 of 26
`
`

`
`Chapter 1
`8
`1.6 Design of Cyclopropyl-fused Nitrilo-pyrrolidines
`
`In concert with the effort described above, the discovery team examined
`alternative approaches to the generation of novel inhibitors that might mini-
`mize or obviate the undesired cyclization reaction. Cognizant of the impact of
`pyrrolidine substitution on DPP4 activity (e.g. 17), we proposed that cyclo-
`propyl fusion to the prolyl ring, represented by 19,42 might represent a new and
`viable approach (Figure 1.5). Incorporation of a fused cyclopropane ring
`constituted a minimal steric burden at P1, would impact the planarity of the
`prolyl ring, and importantly have the potential to conformationally retard
`intramolecular attack of the amine to the nitrile. Hence, a series of cyclopro-
`pane-fused nitrilo-pyrrolidines 20–23 were synthesized and assessed for both
`DPP4 inhibition and solution stability (pH 7.2 phosphate buffer, 39.5 1C).43 As
`compared to the unsubstituted prototype 10, the trans-4,5- and trans-2,3-iso-
`mers 22 and 23, respectively, were not well tolerated by the enzyme, negating
`further evaluation of these as viable program leads. In contrast, both the cis-
`4,5- (20) and the cis-3,4- (21) analogues exhibited acceptable, though slightly
`diminished, activity as compared to 10. More interestingly, 20 exhibited a
`significant enhancement in solution stability as compared to either 10 or 21,
`confirming that cis-4,5-cyclopropyl fusion did indeed retard the intramolecular
`cyclization process. Expanding upon this finding, compounds which incorpo-
`rated a corresponding tert-leucine at P2 as in 24–26 minimized potency dif-
`ferences between the respective cores, though solution stability was still
`
`Figure 1.5 Genesis of cyclopropyl-fused nitrilo-pyrrolidine chemotype.
`
`Downloaded on 05/02/2015 20:03:18.
`
`Published on 30 September 2010 on http://pubs.rsc.org | doi:10.1039/9781849731980-00001
`
`View Online
`
`SAXA-DEF-00333
`
`Page 10 of 26
`
`Page 10 of 26
`
`

`
`The Discovery of the Dipeptidyl Peptidase-4 (DPP4) Inhibitor Onglyzat
`
`9
`
`O
`
`O
`
`N
`
`NH
`
`30
`
`R
`
`NH
`
`N
`
`NH
`
`29
`
`O
`
`R
`
`O
`
`R
`
`N
`
`CN
`
`NH2
`
`28 (anti)
`
`R
`
`N
`
`H2N
`
`O
`
`CN
`
`27 (syn)
`
`Syn oreintation favored with:
`
`- increased steric bulk at R
`- cis-4,5-fused cyclopropyl substitution
`
`Figure 1.6 Factors determining syn versus anti orientation of the methano-nitrilo-
`pyrrolidine chemotype.
`
`favorably preserved in the cis-4,5-methano core (e.g. 24). These and other data
`suggested that increased steric and lipophilic bulk at the P2 position of the
`inhibitor (e.g. ethyl vs. isobutyl vs. tert-butyl, etc.) enhanced both in vitro
`potency as well as solution stability. A similar trend was also observed by the
`Ferring Research group in the simple nitrilo-pyrrolidine series.34
`Clearly both the steric bulk of the P2 side-chain and the cyclopropyl fusion
`on the pyrrolidine ring cooperatively enhanced comformational stability.
`Computational analyses later demonstrated that, primarily because of van der
`Waals interactions,
`increasing the size of the side-chain R (Figure 1.6)
`disfavored the anti orientation 28 required for irreversible intramolecular
`cyclization to the inactive products 29 and 30. For example, where R is Me
`versus tert-butyl, the DDH between the anti (28) and syn (27) orientations
`1. Furthermore, addition of the cis-4,5-
`were calculated to be 0.8 kcal mol
`cyclopropyl moiety also disfavors adoption of the anti orientation by an
`1 as compared to the unsubstituted nitrilo-pyrrolidine
`additional 0.6 kcal mol
`ring. Thus, by combining these conformational elements, we were able to
`identify novel inhibitors of DPP4 with high in vitro potency and enhanced
`solution stability.
`Compound 24 represented a major breakthrough in the chemistry program.
`Profiling of this lead demonstrated a low potential for off-target liabilities
`[hERG inhibition, cytochrome P450 (CYP450) inhibition, broad receptor
`screening, etc.], as well as favorable pharmacokinetic properties in the rat
`(F¼ 77%, t1/2¼ 2.8 h). Additionally, compound 24 was efficacious in Zuckerfa/fa
`rats, reducing glucose AUC in an acute oGTT (ED50¼ 3.3 mg kg
`1, glucose
`challenge 30 min post-dose) when compared to vehicle control. While the
`duration of action of 24 was not particularly long (ED50¼ 92 mg kg
`1, glucose
`challenge 5 h post-dose), the in vivo potency was still significantly greater
`than that of the literature thiazolidine lead 9 (ED50¼ 38 mg kg
`1, glucose chal-
`lenge 30 min post-dose). On the basis of these findings, additional staff were
`assigned to the program to permit a detailed and robust exploration of the
`structure–activity relationship (SAR), primarily focusing on further modifica-
`tions at P2.
`
`Downloaded on 05/02/2015 20:03:18.
`
`Published on 30 September 2010 on http://pubs.rsc.org | doi:10.1039/9781849731980-00001
`
`View Online
`
`SAXA-DEF-00334
`
`Page 11 of 26
`
`Page 11 of 26
`
`

`
`Chapter 1
`10
`1.7 SAR Optimization Leading to the Discovery of
`Saxagliptin
`
`Building upon the cis-4,5-methano-2-nitrilopyrrolidine core, a wide variety of
`substituents, represented generically in 31, were explored (Figure 1.7). As
`highlighted earlier, greater potency and stability were realized via introduction
`of highly b-branched P2 side-chains. Due to the very limited commercial
`availability of b-quaternary a-amino acids, the team devised an efficient
`strategy to generate novel P2 units by several complementary paths, each
`deriving from cyclic and/or symmetrical ketones to avoid unnecessary intro-
`duction of additional stereocenters.6,43 In one arm of this approach, ketones
`underwent Knoevenagel condensation with a malonate diester, and the
`resulting Michael acceptor was subjected to conjugate addition to introduce
`the alkyl-substituted quaternary center. Mono-hydrolysis and subsequent
`Curtius rearrangement yielded the desired b-branched amino acids. A parallel
`approach began with Horner–Emmons condensation of the ketone to give an
`a,b-unsaturated ester. Reduction of the ester to the primary allylic alcohol
`and esterification with Boc-Gly set up a zinc-mediated ester enolate Claisen
`rearrangement, which provided the desired amino acids with a vinyl functional
`handle at the b-position. Alternatively, a standard Strecker synthesis could
`be used on available aldehydes to ultimately afford the desired amino acid
`building blocks. In all cases, the racemic amino acids were then coupled to the
`
`R1
`
`R3
`
`R1
`
`R3
`
`R1
`
`R3
`
`OH
`
`CO2R
`
`O
`
`O
`
`NH
`
`PG
`
`R1
`
`R3
`
`CO2H
`
`NH
`
`PG
`
`R1
`
`R3
`
`O
`
`R2
`
`R3
`
`R1
`
`R2
`
`R3
`
`R1
`
`R1
`
`R3
`
`CO2H
`
`HO2C
`
`CO2H
`
`RO2C
`
`CO2R
`
`NH
`
`PG
`
`R2
`
`R3
`
`R1
`
`CHO
`
`R2 R3
`
`R1
`
`N
`
`CN
`
`O
`
`31
`
`NH
`
`R
`
`HN
`
`CONH2
`
`Figure 1.7 Various synthetic routes for the generation of highly b-branched amino
`acids.
`
`Downloaded on 05/02/2015 20:03:18.
`
`Published on 30 September 2010 on http://pubs.rsc.org | doi:10.1039/9781849731980-00001
`
`View Online
`
`SAXA-DEF-00335
`
`Page 12 of 26
`
`Page 12 of 26
`
`

`
`The Discovery of the Dipeptidyl Peptidase-4 (DPP4) Inhibitor Onglyzat
`
`11
`
`N
`
`H2N
`
`O
`
`CN
`
`32
`DPP4 Ki = 3.9 nM
`
`in vivo
`conversion
`
`N
`
`H2N
`
`O
`
`CN
`
`34
`DPP4 Ki = 0.9 nM
`
`HO
`
`H2N
`
`N
`
`O
`
`CN
`
`33
`DPP4 Ki = 7.4 nM
`
`HO
`
`H2N
`
`N
`
`O
`
`CN
`
`3
`DPP4 Ki = 1.3 nM
`
`R2
`
`R3
`
`R1
`
`N
`
`O
`
`CN
`
`NH
`
`R
`
`31
`
`- High in vitro potency
`- Excellent in vivo efficacy & duration
`of action
`- Poor oral exposure and short half life
`
`- High in vitro potency
`- Excellentin vivo efficacy & duration
`of action
`- Good oral exposue and half life
`
`Figure 1.8 Classical PK/PD disconnect in rats leads to hypothesis of active meta-
`bolite generation in vivo, resulting in the discovery of saxagliptin.
`
`enantiomerically pure P1 nitrilo-methanopyrrolidine fragment and the result-
`ing diastereomers were chromatographically resolved.
`From this effort emerged two compounds of particular interest, inhibitors 32
`and 34 (Figure 1.8). In the case of 32, this compound demonstrated superior in
`vivo efficacy and a sustained duration of response for inhibition of plasma
`1 (71% @ 30 min post-
`DPP4 in normal fasted SD rats at a dose of 4 mmol kg
`dose and 64% @ 4 h post-dose). Duration of the pharmacodynamic (PD)
`response was particularly long when compared to compounds generated earlier
`in the program. Intriguingly, this extended PD response was most often
`observed in analogues containing a vinyl substituent (e.g. 32) at P2. Despite its
`robust in vivo activity, compound 32 demonstrated low oral bioavailability
`(5%) and a short half-life (t1/2¼ 1.2 h) in rats. In contrast, the related saturated
`analogue of 32 (vinyl replaced with ethyl) exhibited significantly higher bio-
`availability (31%) and greater in vitro stability in rat and human liver micro-
`somes, but a weaker response in the rat plasma DPP4 inhibition assay (despite
`equivalent in vitro potency). The disconnect between the pharmacokinetic and
`pharmacodynamic profile of 32 immediately suggested in vivo conversion to an
`active metabolite possessing superior target potency and/or reduced clearance
`profile. Based on this hypothesis, a variety of putative or surrogate hydr-
`oxylated metabolites related to 32 were prepared. While unequivocal char-
`acterization of 33 as an active metabolite of 32 was never established, the
`activity of 33 mirrored that of 32 in the aforementioned PD model. While the
`rat half-life of 33 (t1/2¼ 1.3 h) was not significantly longer when compared to
`that of 32, the absolute bioavailability increased 10-fold to 53%. A more
`
`Downloaded on 05/02/2015 20:03:18.
`
`Published on 30 September 2010 on http://pubs.rsc.org | doi:10.1039/9781849731980-00001
`
`View Online
`
`SAXA-DEF-00336
`
`Page 13 of 26
`
`Page 13 of 26
`
`

`
`12
`
`Chapter 1
`
`striking finding was observed with compound 34. In concordance with estab-
`lished SAR, incorporation of the bulkier adamantyl hydrophobic group in the
`P2 side-chain enhanced in vitro potency (Ki¼ 0.9 nM) as compared to com-
`pounds with smaller fragments in this position. In the normal rat, this com-
`1, po) afforded robust plasma DPP4 inhibition (84% @ 0.5 h
`pound (4 mmol kg
`post-dose and 83% @ 4 h post-dose) despite exceptionally low bioavilability
`(2%) and modest half-life (t1/2¼ 1.4 h). Capitalizing on the learnings from
`compound 33, and considering the propensity of adamantyl groups to undergo
`CYP-mediated hydroxylation, the corresponding metabolite 3 (saxaglipitin)
`was synthesized. The compound proved to be highly potent in vitro, exhibited
`good pharmacokinetic properties in the rat (F¼ 75%, t1/2¼ 2.1 h) and effected
`near maximal (B87%) plasma DPP4 inhibition in the rat at both 0.5 and 4 h
`1.
`post-dose when administered at 4 mmol kg
`In addition to saxagliptin, a number of other inhibitors incorporating
`P2 side-chains with various substituted adamantanes were prepared and
`evaluated, including other positional isomers of hydroxyadamantane, dihy-
`droxyadamantane, and fluoroadamantane. While these other adamantane-
`derived compounds also exhibited potent DPP4 inhibition in vitro, saxagliptin
`provided a comparatively superior PK and PD profile, and was ultimately
`chosen for development.
`
`1.8 Binding of Saxagliptin to Human DPP4: A Slow
`Tight-binding Inhibitor
`
`Through more extensive characterization of the binding kinetics of saxagliptin
`(and select analogs) to DPP4, it was noted that several compounds showed
`evidence of slow tight-binding. Further analysis of SAR patterns relating to this
`slow on/off-rate feature revealed correlations with both steric bulk in the P2
`side-chain (specifically b-quaternary substitution) and the presence of a nitrile
`functionality. Prior to our undertaking of detailed kinetic studies, it had been
`speculated by several groups that a transient covalent bond was formed
`between the hydroxyl group of the active-site Ser630 of the DPP4 catalytic triad
`and the nitrile carbon of nitrile-based inhibitors, yet early X-ray co-crystal
`structures with such compounds lacked adequate resolution to confirm
`appropriate electron density where such a bond would exist.44 Our findings
`were consistent with a hypothesis whereby the bulky P2 side-chain’s displace-
`ment of water in the S2 pocket drove entropic aspects governing on-rate (for 3,
`Kon¼ 4.6105 M
`1 s
`1), and, once anchoring the inhibitor in the active site,
`covalent interaction with the nitrile drove enthalpic aspects governing off-rate
`(for 3, Koff¼ 2310
`5 s
`1), resulting in overall Ki enhancement for certain
`inhibitors possessing these features.45 As a result, the t1/2 for dissociation of
`saxagliptin from DPP4 was determined to be B50 min at 37 1C (B250 min at
`room temperature). In comparison to the rapid off rates of non-nitrilo-pyrro-
`lidines such as sitagliptin, the slow off-rates exhibited by saxagliptin and, to a
`lesser extent, other nitrilo-pyrrolidines such as 12,45b,46 proved to be a unique
`
`Downloaded on 05/02/2015 20:03:18.
`
`Published on 30 September 2010 on http://pubs.rsc.org | doi:10.1039/9781849731980-00001
`
`View Online
`
`SAXA-DEF-00337
`
`Page 14 of 26
`
`Page 14 of 26
`
`

`
`The Discovery of the Dipeptidyl Peptidase-4 (DPP4) Inhibitor Onglyzat
`
`13
`
`attribute of saxagliptin among the clinically advanced inhibitors. The slow off-
`rate kinetics exhibited by saxagliptin likely

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