throbber
Current Topics in Medicinal Chemistry, 2007, 7, 557-568
`
`557
`
`Discovery of JANUVIA™ (Sitagliptin), a Selective Dipeptidyl Peptidase IV
`Inhibitor for the Treatment of Type2 Diabetes
`
`Nancy A. Thornberry and Ann E. Weber*
`
`Departments of Metabolic Disorders and Medicinal Chemistry, Merck Research Laboratories, P.O. Box 2000, Rahway,
`NJ 07065
`
`Abstract: The emergence of glucagon-like peptide 1 (GLP-1) as a well validated approach to the treatment of type 2
`diabetes and preclinical validation of dipeptidyl peptidase IV (DPP-4) inhibition as an alternate, oral approach to GLP-1
`therapy prompted the initiation of a DPP-4 inhibitor program at Merck in 1999. DPP-4 inhibitors threo- and allo-isoleucyl
`thiazolidide were in-licensed to jump start the program; however, development was discontinued due to profound toxicity
`in rat and dog safety studies. The observation that both compounds inhibit the related proline peptidases DPP8 and DPP9
`led to the hypothesis that inhibition of DPP8 and/or DPP9 could evoke severe toxicities in preclinical species. Indeed, the
`observed toxicities were recapitulated with a selective dual DPP8/9 inhibitor but not with an inhibitor selective for DPP-4.
`Thus, medicinal chemistry efforts focused on identifying a highly selective DPP-4 inibitor for clinical development. Initial
`work in an a-amino acid series related to isoleucyl thiazolidide was discontinued due to lack of selectivity; however, SAR
`studies on two screening leads led to the identification of a highly selective b-amino acid piperazine series. In an effort to
`stabilize the piperazine moiety, which was extensively metabolized in vivo, a series of bicyclic derivatives were prepared,
`culminating in the identification of a potent and selective triazolopiperazine series. Unlike their monocyclic counterparts,
`these analogs typically showed excellent pharmacokinetic properties in preclinical species. Optimization of this series led
`to the discovery of JANUVIA™ (sitagliptin), a highly selective DPP-4 inhibitor for the treatment of type 2 diabetes.
`
`INTRODUCTION
`
`TARGET SELECTION
`
`The pathogenesis of type 2 diabetes (T2DM) involves a
`set of three primary defects: insulin resistance, b cell dys-
`function, and hepatic glucose overproduction. These defects
`are the principal targets of both current and future therapy.
`Currently available classes of oral antihyperglycemic agents
`include PPARg agonists, sulfonyureas/meglitinides, and
`biguanides. These agents are used either in monotherapy or,
`increasingly, in combinations to lower glucose levels.
`Despite the availability of a range of agents for T2DM, many
`diabetic patients fail to achieve or to maintain glycemic
`targets. In addition, current therapies have limited durability
`and/or are associated with significant side effects (GI
`intolerance, hypoglycemia, weight gain, lactic acidosis and
`edema). Thus, there remain critical unmet medical needs in
`the treatment of this disorder. With an increasing understan-
`ding of the molecular pathways involved in glucose control,
`a range of new potential targets have emerged for treatment
`of the key areas of pathogenesis. In particular, there has been
`increased emphasis on new therapies that increase the
`circulating concentrations of insulin in a glucose dependent
`manner, most notably, glucagon-like peptide 1 (GLP-1)
`analogs [1] and dipeptidyl peptidase 4 (DPP-4) inhibitors
`[2]. In this review we describe Merck’s DPP-4 inhibitor
`program, which was initiated in 1999 and culminated with
`the discovery of JANUVIA™ (sitagliptin), a potent and
`highly selective inhibitor of DPP-4 that shows excellent
`promise for the treatment of T2DM.
`
`*Address correspondence to this author at Merck Research Laboratories,
`P.O. Box 2000, Rahway, NJ 07065; Tel: (732) 594-5796; E-mail:
`ann_weber@merck.com
`
`Over the last decade, GLP-1 receptor agonism has
`emerged as one of the best validated approaches for the
`treatment of T2DM. For example, in 1997 it was reported
`that continuous infusion of GLP-1 to diabetic humans
`resulted in normalization of both postprandial and fasting
`glucose [3]. More recently, sub-chronic (6 wk) continuous
`infusion of GLP-1 was shown to result in profound and
`significant decreases in fasting plasma glucose (14.1 to 10.1
`mM) and HbA1c (9.2 to 7.9 %) [4]. It is generally accepted
`that the key mechanisms responsible for glucose lowering by
`GLP-1 receptor agonism are: (i) stimulation of glucose-
`dependent insulin biosynthesis and secretion, (ii) glucose-
`dependent inhibition of glucagon release, and (iii) delayed
`gastric emptying.
`Also in the 1990s, it became increasingly clear that GLP1
`in vivo, specifically via
`was very
`tightly regulated
`proteolysis at the N-terminus to produce an inactive peptide,
`and that the key enzyme responsible for this inactivation was
`DPP-4, a proline specific dipeptidyl aminopeptidase [5,6].
`These findings resulted in the initiation of programs at
`several companies
`to
`identify DPP-4 resistant GLP-1
`analogs, and also led to the testing of DPP-4 inhibitors in
`animal models of diabetes, where increased levels of GLP-1,
`enhanced insulin secretion, and improved glucose tolerance
`were observed [7-9].
`The human validation of GLP-1, together with preclinical
`validation of DPP-4 inhibition as an alternate oral approach
`to GLP-1, prompted Merck to initiate a project on this
`enzyme in 1999. Our enthusiasm for this mechanism was
`based primarily on the view that this approach would have at
`least three potential advantages over currently available
`agents. First, because GLP-1 stimulates insulin release in a
`
` 1568-0266/07 $50.00+.00
`
`© 2007 Bentham Science Publishers Ltd.
`
`Merck Exhibit 2114, Page 1
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`558 Current Topics in Medicinal Chemistry, 2007, Vol. 7, No. 6
`
`Thornberry and Weber
`
`Probiodrug had evaluated the safety of L-threo-isoleucyl
`thiazolidide in 4-week toxicity studies in rats and dogs [20].
`In rats, toxicities were limited to the presence of lung
`histiocytosis and thrombocytopenia at relatively high doses
`(77.5 and 698 mg/kg, respectively). In dogs, acute central
`nervous system (CNS) toxicities, characterized by ataxia,
`seizures, convulsions, and tremor, were observed at 75
`mg/kg, and bloody diarrhea was also observed at 225 mg/kg
`upon acute dosing. No additional toxicities were noted in
`these 4 week studies. However, in subsequent chronic
`toxicity studies at MRL, upon 5-6 weeks of treatment with
`this compound in dogs, mortality and profound toxicities
`occurred at doses ‡ 25 mg/kg/day. These toxicities included
`anemia,
`thrombocytopenia, splenomegaly, and multiple
`organ pathology mainly affecting the lymphoid system and
`gastrointestinal tract.
`As noted above, we also in-licensed the allo isomer of
`isoleucyl thiazolidide, which, when compared to the threo
`isomer, has virtually identical affinity to DPP-4, similar
`pharmacokinetic and metabolic profiles, and similar in vivo
`efficacy in an oral glucose tolerance test in diet induced
`obese mice [20]. In parallel with the chronic toxicity studies
`with L-threo-isoleucyl thiazolidide, this compound was
`evaluated in an acute tolerability study in dogs and in 4-week
`toxicity studies in rats. As with the threo isomer, bloody
`diarrhea was observed in dogs, but the allo isomer was > 10-
`fold more toxic when compared on either a dose level or
`plasma exposure basis. In the rat studies, lung histiocytosis
`and thrombocytopenia were observed as had been seen with
`the threo compound, with the allo compound toxic at > 10-
`fold lower dosage. In addition, the other profound toxicities
`that were observed in dogs with the threo compound (e.g.,
`anemia, splenomegaly, and mortality with multiple organ
`pathology) were observed with the allo compound in rats. As
`a result of these findings, development of both compounds
`was discontinued in early 2001.
`
`DPP8/9 TOXICITY STUDIES
`
`The toxicities observed with the threo and allo comp-
`ounds deepened our concern about the potential safety of this
`mechanism. However, the finding that the allo isomer was
`approximately 10-fold more toxic in rats and dogs, despite
`having comparable pharmacodynamic activity and pharma-
`cokinetics in both species, suggested that these toxicities
`were likely not due to DPP-4 inhibition, but instead were
`potentially due
`to off-target activity. In
`this regard,
`subsequent to the initiation of our program, it had become
`increasingly clear that DPP-4 was a member of larger family
`of ‘DPP-4 activity- and/or structure-homologues’ (DASH)
`proteins, enzymes that are unified by their common post-
`proline cleaving serine dipeptidyl peptidase mechanism [21].
`Enzymes that had recently been described included quiescent
`cell proline dipeptidase (QPP) (aka DPP7) [22], DPP8 [23],
`DPP9 [24], and fibroblast activation protein (FAP) [25]. As
`the functions of these enzymes were unknown, determining
`the selectivity of our inhibitors was a key element of our
`medicinal chemistry program, and thus counterscreens for
`these enzymes were developed.
`The selectivity of the allo and threo compounds was
`determined in the DASH family counterscreens, as well as in
`
`strictly glucose-dependent manner, little or no risk of
`hypoglycemia was anticipated. Second, no weight gain was
`expected with DPP-4 inhibitors. Finally, rodent studies with
`GLP-1 analogs had demonstrated a role for this peptide in
`the regulation of b -cell mass [10]; if these findings translated
`to the clinic, there was the potential that DPP-4 inhibitors
`could have long-term beneficial effects on b-cell function.
`At the onset of this program, there were also several
`concerns regarding potential safety issues for this class.
`DPP-4 is a type II membrane bound cell surface protein that
`is ubiquitously expressed, and like many other cell surface
`molecules, DPP-4 had been implicated in a wide range of
`biological functions. Two potential issues were of most
`concern: first, DPP-4 is identical to the T cell activation
`marker CD26, and data in model systems suggested a
`potential co-stimulatory role for this enzyme in T cell
`activation [11]. Moreover, there were reports that some DPP-
`4 inhibitors (Lys [Z(NO2)] pyrrolidide and related comp-
`ounds) had several effects on immune cells, including
`inhibition of proliferation [12]. Second, DPP-4 had been
`shown to cleave a number of immunoregulatory, endocrine,
`and neurological peptides in vitro [13]. While some comfort
`was later provided by the report that DPP-4 deficient mice
`develop normally, and are healthy [14], a finding that we
`subsequently confirmed [15], we were acutely aware of the
`potential for mechanism-based toxicities, and, when pos-
`sible, exploited opportunities to address these issues as the
`medicinal chemistry program progressed, as described
`below.
`
`PROBIODRUG LICENSING EXPERIENCE
`
`When we initiated our internal screening and medicinal
`chemistry program, two compounds were already advancing
`through human clinical
`trials, Probiodrug’s
`isoleucyl
`thiazolidide (1) and NVP-DPP728 (3) from Novartis (Fig. 1)
`[16,17]. Thus, in order to “jump start” our internal program,
`in late 2000 we elected to in-license L-threo-isoleucyl
`thiazolidide (P32/98) and its allo stereoisomer (L-allo-
`isoleucyl thiazolidide, 2). In single dose pharmacodynamic
`studies, P32/98 had been shown to be well tolerated,
`increased active GLP-1, and reduced glycemic excursion
`following food or glucose intake in normal volunteers [18].
`In addition, Probiodrug reported enhanced insulin secretion
`and improved glucose tolerance in single dose studies in a
`small number of diabetic patients [19].
`
`M e
`
`O
`
`Me
`
`O
`
`Me
`
`Me
`
`N
`
`NH2
`
`S
`
`N
`
`NH2
`
`S
`
`allo-Ile-thiazolidide (2)
`
`O
`
`CN
`
`N
`
`HN
`
`NH
`
`N
`
`P32/98 (1)
`
`NC
`
`NVP-DPP728 (3)
`Fig (1). Early DPP-4 inhibitors.
`
`Merck Exhibit 2114, Page 2
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`Discovery of JANUVIA™ (Sitagliptin), a Selective Dipeptidyl Peptidase IV
`
`Current Topics in Medicinal Chemistry, 2007, Vol. 7, No. 6 559
`
`an in-house panel of other proteases and by MDS Pharma
`Services (PanLabs) in a panel of 170 receptor and enzyme
`assays. No significant activity (IC50 < 100 m M) was observed
`in any of the in-house protease and PanLabs assays, with the
`exception of the sigma s 1 receptor for the allo compound (Ki
`= 42 mM). However, for DPP-4 related dipeptidyl peptidases,
`inhibition was not only observed for DPP-4, but also for the
`closely related dipeptidyl peptidases, QPP, DPP8, and DPP9.
`Both the allo and threo isomers showed comparable QPP
`inhibition activity (IC50 = 18 mM and 14 mM, respectively);
`however, the potency for inhibition of DPP8 and DPP9
`differed by 5- to 10-fold, with the allo isomer being more
`potent in each case (220 nM vs. 2200 nM for DPP8 and 320
`nM vs. 1600 nM for DPP9) [20]. Since these differences in
`inhibition of DPP8/DPP9 were consistent with the observed
`differences in dose necessary to produce toxicity, we
`hypothesized that inhibition of DPP8 and/or DPP9 was
`responsible for the observed toxicities of both compounds in
`preclinical species.
`To obtain evidence that DPP8/9 inhibition was respon-
`sible for the toxicities observed with the allo and threo
`isomers, DPP-4, QPP, and DPP8/9 selective compounds 4, 5,
`and 6 (Fig. 2), respectively, were identified and evaluated in
`2 week rat toxicity studies and in acute dog tolerability
`studies [20]. The results from these studies showed a
`remarkable similarity between the effects produced by the
`DPP8/DPP9 selective inhibitor and the allo compound
`
`(Table 1). In rats, the DPP8/9 inhibitor produced alopecia,
`thrombocytopenia, reticulocytopenia, enlarged spleen, multi-
`organ histopathological changes, and mortality. In dogs, the
`DPP8/9 inhibitor produced gastrointestinal toxicity. The
`QPP inhibitor produced reticulocytopenia in rats only, and
`no toxicities were noted in either species for the selective
`DPP-4 inhibitor. These results provided compelling evidence
`that inhibition of DPP8/9, but not selective DPP-4 inhibition,
`is associated with multi-organ toxicities in preclinical
`species.
`There were two major reasons that we had a high level of
`confidence in this conclusion. First, at least two structurally
`distinct compounds that inhibit DPP8/9 showed remarkably
`similar toxicities in rats and dogs. As noted above, the
`DPP8/9 inhibitor is highly selective over all other proline
`specific enzymes, and inhibition of the allo compound is
`limited to DPP-4, DPP8/9, and weak inhibition of QPP. We
`also showed that the DPP8/9 inhibitors produce similar
`toxicities in DPP-4-deficient mice and wild type mice,
`establishing that the observed toxicities were not due to
`inhibition of DPP-4 [20]. Second, the degree of toxicity
`observed with the allo and threo compounds correlated with
`their affinity for DPP8/9.
`With the finding that DPP8/9 could produce a variety of
`toxicities in vivo, we hypothesized that some of immune
`effects that had been observed with Lys[Z(NO2)]–pyrrolidide
`and related compounds [13] could instead be due to
`
`F
`
`F
`
`NH2
`
`O
`
`N
`
`N
`
`N
`
`N
`
`CF3
`
`4, DPP-4 selective
`DPP-4 IC50 = 27 nM
`
`M e
`
`Me
`
`O
`
`N
`
`NH2
`
`5, DPP8/9 selective
`DPP8 IC 50 = 38 nM
`DPP9 IC 50 = 55 nM
`
`O
`
`NS
`O
`
`O
`
`N
`
`NH2
`
`S
`
`I
`
`6, QPP selective
`QPP IC 50 = 19 nM
`
`Fig. (2). DPP-4, DPP8/9 and QPP selective inhibitors used in comparative toxicity studies.
`
`Table 1. Comparative Toxicity Studies in Rats (2 Weeks of Treatment at Doses of 10, 30, 100 mg/kg/Day) and Dogs (Single Dose, 10
`mg/kg PO) with Selective Inhibitors 4, 5, and 6. A Check ((cid:214)) Indicates the Toxicity was Observed. Historical Data from
`threo and allo Isoleucyl Thiazolidide Safety Studies are Shown for Comparison
`
`Species
`
`Toxicity
`
`DPP-4 Selective (4)
`
`DPP8/9 Selective (5)
`
`QPP Selective (6)
`
`threo-Ile thia (1)
`
`allo-Ile thia (2)
`
`Rat
`
`Alopecia
`
`Thrombocytopenia
`
`Anemia
`
`Reticulocytopenia
`
`Splenomegaly
`
`Mortality
`
`Dog
`
`Bloody diarrhea
`
`anot determined
`
`(cid:214) (cid:214)
`
`(cid:214) (cid:214) (cid:214)
`
`(cid:214) (cid:214)
`
`n.d.a
`n.d.
`(cid:214) (cid:214)
`
`(cid:214) (cid:214)
`
`(cid:214) (cid:214) (cid:214)
`
`Merck Exhibit 2114, Page 3
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`(cid:214)
`

`

`560 Current Topics in Medicinal Chemistry, 2007, Vol. 7, No. 6
`
`Thornberry and Weber
`
`inhibition of DPP-8/9, and we proceeded to assess the
`selectivity of these compounds. We found that they have
`greater intrinsic potency against DPP8 and DPP9 than DPP-4
`[20]. Moreover, we discovered that the DPP8/9 selective
`inhibitor, but not the selective DPP-4 inhibitor, attenuated
`proliferation and IL-2 release in human in vitro models of T
`cell activation [20]. These results strongly suggest that
`proteolytic activity is not required for the putative co-
`stimulatory function of DPP-4/CD26, and that immunolo-
`gical effects previously observed with several DPP-4
`inhibitors compounds in preclinical models were likely due
`to off-target inhibition of DPP8/9. This result provided a
`greater level of confidence that inhibition of DPP-4 would
`not result in compromised immune function.
`While the significance to human safety is unknown, the
`finding that DPP8/9 inhibition produces profound toxicity in
`preclinical species, and is also likely responsible for effects
`on immune function that have been previously attributed to
`DPP-4, prompted us to refocus our program on the discovery
`of highly selective DPP-4 inhibitors.
`
`INITIAL MEDICINAL CHEMISTRY EFFORTS:
`CYCLOHEXYLGLYCINE LEAD
`
`We initiated our medicinal chemistry program prior to
`the discovery of the preclinical toxicity associated with
`inhibition of DPP8/9. Thus, we focused initially on identify-
`ing a “Best in Class” compound by improving upon the
`potency of isoleucyl thiazolidide (IC50 = 420 nM in our
`hands) and the short half-life of NVP-DPP728, which we
`found to be only ~15 min in rats. We thought the latter issue
`might be due to chemical instability. This compound, like
`many of the most potent DPP-4 inhibitors, contained a
`reactive electrophile (a nitrile in this case) that was believed,
`and a close analog later shown, to form a covalent bond with
`the active site serine [26]. This electrophile is six atoms
`away from an amine, perfectly set up to cyclize. In order to
`improve upon the half-life, we chose from the onset to focus
`on structures which lacked this electrophile, even though we
`knew these structures had been generally shown to be less
`potent.
`While we were waiting for results from our internal
`screening efforts, we initiated SAR studies based on the
`known a-amino acid derived inhibitors. The most potent
`inhibitor reported in the literature that did not contain an
`electrophile was cyclohexylglycyl thiazolidide (7, Table 1),
`discovered by chemists at Ferring [27]. With an IC50 of 89
`nM in our hands, this compound was already 4-fold more
`potent than Probiodrug’s related clinical candidate. In order
`to further improve the potency and identify proprietary
`compounds, substitution on the cyclohexyl ring was explored
`[28,29]. In particular, amides, carbamates and sulfonamides
`at the 4-position on the cyclohexyl ring provided compounds
`such as 8, 9 and 10 (Table 2) with improved potency and
`oral bioavailabilities in rats of 81%, 76% and 46%,
`respectively [27].
`Once the toxicity of the thiazolidide derivatives emerged,
`but before we had assays in hand for DPP8 and DPP9, we
`wondered whether the thiazolidine ring might be responsible
`for the observed toxicity. If the thiazolidine ring opened in
`
`Table 2.
`
`4-Substituted Cyclohexylglycine Analogs
`
`R
`
`O
`
`N
`
`NH2
`
`X
`
`R
`
`H
`
`(3,4-di-F-Ph)CONH-
`
`PhCH2OCONH-
`
`(4-CF3O-Ph)SO2NH-
`
`(3,4-di-F-Ph)CONH-
`
`PhCH2OCONH-
`
`(4-CF3O-Ph)SO2NH-
`
`X
`
`S
`
`S
`
`S
`
`S
`
`CH2
`
`CH2
`
`CH2
`
`(3,4-di-F-Ph)CONH-
`
`(S)-CHF
`
`PhCH2OCONH-
`
`(S)-CHF
`
`(4-CF3O-Ph)SO2NH-
`
`(S)-CHF
`
`DPP-4
`
`IC50 (nM)
`
`89
`
`54
`
`25
`
`22
`
`190
`
`94
`
`89
`
`54
`
`56
`
`36
`
`Compound
`
`7
`
`8
`
`9
`
`10
`
`11
`
`12
`
`13
`
`14
`
`15
`
`16
`
`vivo, a free thiol would be revealed, which could be reactive
`and lead to toxicity. To eliminate this possibility, we shifted
`our focus to non-sulfur containing heterocycles. The simple
`pyrrolidide derivatives were generally less potent (compare,
`for example, compounds 11 vs. 8, 12 vs. 9, and 13 vs. 10,
`Table 2); however, the addition of fluorine to the ring
`resulted compounds with increased potency. Indeed the (3S)-
`3-fluoropyrrolidine amides 14, 15, and 16 (Table 2) are
`nearly equi-potent to the corresponding thiazolidide analogs
`[30].
`The 4-trifluoromethoxybenzenesulfonamide derivative
`16 has excellent pharmacokinetic properties across species,
`with a half-life of 4 h, 12 h and 5 h in rats, dogs and rhesus
`monkeys, respectively, and oral bioavailability of 37% to
`89% [30]. It was profiled extensively as a potential
`preclinical development candidate. Only reticulocytopenia at
`100 mg/kg/day was observed in a 2-week toxicity study in
`rats, and it was clean in acute tolerability studies in dogs
`[Lankas, G.; unpublished results]. Despite an attractive
`profile, further work on this compound was discontinued due
`to unacceptable levels of DPP8 and DPP9 inhibition (IC50 =
`1400 nM and 1700 nM, respectively).
`Once the potential toxicity associated with inhibition of
`the DPP8 and DPP9 enzymes was discovered, our goal was
`to identify an inhibitor with a >1000-fold window for DPP-4
`inhibition over inhibition of these enzymes. A clue for how
`to achieve specificity came from a pair of positional scan-
`ning libraries that were developed for these aminopeptidases
`[31]. Each library consisted of dipeptidyl aminomethyl-
`coumarin substrates. In the first “P1” sublibrary, each well
`contained a spatially addressed amino acid at the P1 position
`coupled to an isokinetic mixture of amino acids at P2. In the
`
`Merck Exhibit 2114, Page 4
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`Discovery of JANUVIA™ (Sitagliptin), a Selective Dipeptidyl Peptidase IV
`
`Current Topics in Medicinal Chemistry, 2007, Vol. 7, No. 6 561
`
`O
`
`N
`
`H
`
`NH2
`
`HN
`
`O
`
`S O
`
`R
`
`17, R = COOH
`DPP-4 IC 50 = 15 nM
`DPP8 IC50 = 2800 nM
`
`18, R = CF3CH2SO2NH-
`DPP-4 IC50 = 2.6 nM
`DPP8 IC50 = 15,000 nM
`
`Fig. (4). DPP-4 inhibitors containing acidic functionality.
`
`This compound was >100-fold selective over both DPP8 and
`DPP9. Potency could be enhanced by appending a second
`phenyl ring. The 4-fluorobiphenyl derivative 20 is a 64 nM
`DPP-4 inhibitor with excellent selectivity over both DPP8
`and DPP9, though both 19 and 20 show decreased selectivity
`over QPP [32]. By incorporating polar functionality in place
`of the methyl group to give N,N-dimethylamide 21, both
`potency and selectivity over QPP were improved [33].
`inhibitor 21 has excellent pharmacokinetic
`While
`properties across species, its efficacy in an oral glucose
`tolerance test in mice was less than anticipated based on the
`exposures obtained and its inhibition of mouse DPP-4. This
`observation was attributed to its high plasma protein binding
`[33]. Replacement of the terminal phenyl ring with a
`heterocycle provided compounds with reduced serum shift,
`leading to triazolopyridine 22 Fig. (5) which was chosen for
`extensive preclinical evaluation [34]
`FROM SCREENING HITS TO b-AMINO ACID LEAD
`While medicinal chemistry efforts in the a-amino acid
`series were ongoing, screening of the Merck sample
`
`second “P2” sublibrary, each well contained a spatially
`addressed amino acid at the P2 position coupled to an
`isokinetic mixture of amino acids at P1. Results for cleavage
`of the libraries by DPP-4 and DPP8 are summarized in Fig.
`(3). While both enzymes showed a strong preference for
`cleavage of substrates with a proline at the P1 position, DPP-
`4 was much more promiscuous at P2. In particular, dipeptides
`containing acidic amino acids such as glutamic acid were
`readily cleaved by DPP-4 whereas the rate of cleavage of
`these dipeptides by DPP8 was greatly reduced. We reasoned
`that incorporation of such acidic functionality at the P2
`position of our inhibitors could provide analogs with
`improved selectivity.
`A variety of acidic derivatives were prepared. The two
`most selective compounds in this series, 17 [E. Parmee,
`unpublished results] and 18 [28], are illustrated in Fig. (4).
`Both are >100-fold selective for DPP-4 over DPP8; how-
`ever, both suffer from poor oral bioavailability in rats (<1%).
`Further optimization did not yield selective compounds with
`improved pharmacokinetic properties and this series was put
`on hold.
`a-AMINO ACID SERIES REVISITED
`The a-amino acid series was ultimately re-examined
`following the discovery of sitagliptin. Because the allo
`isomer of isoleucyl thiazolidide was a more potent inhibitor
`of DPP8 and DPP9 than the threo isomer, we wondered
`whether we could improve selectivity by incorporating a
`“threo” bias into this series, an approach that was not
`possible with the symmetrical cyclohexylglycine derivatives.
`As shown in Fig. (5), when the ethyl sidechain of isoleucyl
`thiazolide was replaced with phenyl to provide b-methyl
`phenylalanine analog 19, potency decreased by ~2-fold but
`selectivity was greatly improved [J. Xu, unpublished results].
`
`(a)
`
`(b)
`
`Fig. (3). Cleavage of peptide scanning libraries by (a) DPP-4 and (b) DPP8.
`
`Merck Exhibit 2114, Page 5
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`562 Current Topics in Medicinal Chemistry, 2007, Vol. 7, No. 6
`
`Thornberry and Weber
`
`19, R = Ph
`DPP-4 IC50 = 970 nM
`DPP8 IC5 0 > 100,000 nM
`QPP IC5 0 = 12,000 nM
`
`21, R = CONMe2
`DPP-4 IC5 0 = 12 nM
`DPP8 IC50 > 100,000 nM
`QPP IC50 = 45,000 nM
`
`Me
`
`O
`
`R
`
`N
`
`NH2
`
`S
`
`R
`
`O
`
`N
`
`NH2
`
`F
`
`Me
`
`N
`
`Me
`
`O
`
`O
`
`NH2
`
`N
`
`F
`
`F
`
`1, R = Et
`DPP-4 IC5 0 = 420 nM
`DPP8 IC 50 = 2200 nM
`QPP IC 50 = 14,000 nM
`
`20, R = Me
`DPP-4 IC5 0 = 64 nM
`DPP8 IC 50 = 88,000 nM
`QPP IC 50 = 2700 nM
`
`22
`DPP-4 IC5 0 = 8.8 nM
`DPP8 IC 50 > 100,000 nM
`QPP IC 50 > 100,000 nM
`
`F
`
`N
`
`N N
`
`N
`
`NH
`
`NH
`
`O
`
`Cl
`
`O
`
`NHSO2Me
`
`NN
`
`O
`
`HN
`
`O
`
`N
`
`CH3
`
`23, IC5 0 = 1700 nM
`
`HN
`
`O
`
`N
`
`NH2
`
`O
`
`24, IC5 0 = 1900 nM
`
`NH
`
`N
`
`O
`
`N
`
`25, IC5 0 = 11,000 nM
`
`H2 N
`
`Fig. (6). DPP-4 inhibitor screening hits.
`
`Fig (5). b-Substituted a
`
`-amino acid derived DPP-4 inhibitors.
`
`collection was completed. Surprisingly few hits were
`identified. Only three leads, xanthine derivative 23, proline
`amide 24, and piperizine 25, were deemed suitable for
`follow-up (Fig. 6). Xanthine lead 23 and several related
`micromolar hits originated from a commercially available
`screening library. An additional library of compounds was
`prepared in-house for initial optimization; however, no
`increase in potency was seen so this lead was not further
`pursued. It is interesting to note that other pharmaceutical
`companies have subsequently reported structurally similar
`DPP-4 inhibitors [35,36], highlighting a major issue associa-
`ted with screening non-proprietary compound libraries.
`Proline amide screening hit 24 was originally prepared
`for our thrombin inhibitor program, and was in fact a much
`more potent inhibitor of that enzyme than of DPP-4 (IC50s =
`52 nM and 1900 nM, respectively). Initially we believed,
`naively, that the proline moiety mapped to the thiazolidine or
`pyrrolidine ring in the a-amino acid series. Thus, we quickly
`b-amino acyl group with isoleucine and
`replaced the
`cyclohexylglycine side chains, but this led to a 2- to 3-fold
`decrease in activity. Because the proline amide moiety could
`be replaced by a thiazolidine ring without significant loss of
`activity, much of the early left-hand side SAR was
`conducted in this truncated series [37]. Shortening or
`lengthening the distance between the left-hand side phenyl
`ring and
`the amino group
`led
`to compounds with
`dramatically decreased potency, as did replacement of the
`phenyl ring with cyclopenyl, cyclohexyl, or heterocyclic
`groups. In fact the only change that was not detrimental to
`potency was the addition of fluorine to the phenyl ring. In
`particular, the 2-fluoro derivative was 3-fold more potent,
`and the 2,5-difluoro analog showed a 10-fold improvement.
`The 2,4,5-trifluoro derivative was the most potent compound
`prepared in this truncated series (IC50 = 120 nM).
`
`Merck Exhibit 2114, Page 6
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`Discovery of JANUVIA™ (Sitagliptin), a Selective Dipeptidyl Peptidase IV
`
`Current Topics in Medicinal Chemistry, 2007, Vol. 7, No. 6 563
`
`Early SAR studies on the right hand side of the molecule
`showed
`that
`the amide could be
`replaced by
`the
`corresponding acid without loss of potency. An acetic acid
`scan on the phenyl ring provided 4-phenylacetic acid
`derivative 26 (Fig. 7, IC50 = 510 nM) [38], the first
`subnanomolar compound in this series, which also proved to
`be devoid of thrombin activity (IC50 > 500,000 nM). As we
`saw in the truncated series, the addition of a 2-fluoro
`substituent to the phenyl ring led to an increase in potency.
`Conversion of the acetic acid moiety in the resultant analog
`27 to lactic acid gave 28, a 12 nM inhibitor. More lipophilic
`groups in place of methyl on the acid moiety led to a further
`potency enhancement, with the isopropyl moiety being
`preferred. The addition of a second fluorine to the phenyl
`ring provided 29, a subnanomolar inhibitor.
`Compound 29 had a number of very desirable features.
`First, it represented a truly unique class of inhibitors so we
`anticipated no patent issues. Second, it possessed excellent
`selectivity (‡ 100,000-fold over all related peptidases
`assayed, including DPP8 and DPP9). Unfortunately, oral
`bioavailability in this series was particularly poor. In rats this
`was traced to low oral absorption.
`While work on the proline amide series was proceeding,
`piperazine lead 25, an 11 mM DPP-4 inhibitor, was also
`being explored. This hit was originally prepared for our
`melanocortin 4 receptor (MC4-R) agonist program; however,
`it proved devoid of MC4-R activity. When SAR studies on
`the right hand side of the molecule failed to lead to
`substantial improvements in potency, attention then turned to
`the
`left hand side. The phenethylamine moiety was
`reminiscent of the proline amide lead, so fluorination of the
`phenyl ring, which increased potency in that series, was
`examined. The 3,4-difluorophenyl analog (30, Fig. 8)
`showed a 2-fold increase in potency [39]. Conversion of the
`side chain, essentially a reduced phenylalanine derivative, to
`the corresponding phenylalanine amide gave an inactive
`compound; however, homophenylalanine 31 showed a 100-
`
`fold increase in potency. The 2-fluoro derivative containing
`the more active (R)-benzyl group (32) is a 14 nM DPP-4
`inhibitor. The entire right hand side moiety could be
`removed to give piperazine 33, a much lower molecular
`weight analog, with only a 10-fold loss of potency.
`Optimization of the phenyl ring paralleled that of the proline
`amide series and provided trifluorophenyl derivative 34, the
`most potent compound in this series. Like the proline amide
`series, oral bioavailability of this series was poor, even for
`the low molecular weight compounds such as 33. In this
`case, the problem was traced to extensive metabolism on the
`piperazine ring.
`
`IMPROVEMENTS IN PHARMACOKINETIC PRO-
`PERTIES: THE FUSED PIPERAZINES
`
`In order to stabilize the piperazine ring toward metabo-
`lism, we elected to focus on bicyclic piperazine replace-
`ments. Initial work was done in the simplified, unsubstituted
`piperazine series. Parent compound 35 (Fig. 9) was
`converted to the corresponding triazolopiperidine 36, leading
`to an increase in potency [40]. Ethyl analog 37 was slightly
`more potent, and more importantly, completely stable to
`metabolism in hepatocytes. Despite the improved metabolic
`stability, this compound still had poor oral bioavailability in
`rats (2%), though oral bioavailability in dogs was more
`acceptable (33%). In rats, the low %F was due to low,
`variable absorption. Hepatic extraction was low (10-20%),
`reflecting the increased hepatic stability observed in vitro.
`The solution to improved absorption proved to be quite
`simple, though entirely empirical. We discovered that
`replacing the ethyl side chain with a trifluoromethyl group
`led to an improvement in oral absorption. The oral
`bioavailability of trifluoromethyl analog 38 in rats was 44%.
`With this discovery in hand, all that remained was to adjust
`the fluoro substituents to improve potency. This provided
`difluoro- and trifluorophenyl derivatives 4 and 39 (Fig. 10).
`
`CO2 H
`
`O
`
`CO2H
`
`Me
`
`HN
`
`O
`
`N
`
`NH2
`
`O
`
`27, IC 50 = 54 nM
`
`HN
`
`O
`
`N
`
`NH2
`
`O
`
`28, IC 50 = 12 nM
`
`F
`
`F
`
`CO2H
`
`O
`
`CO2H
`
`HN
`
`O
`
`N
`
`NH2
`
`O
`
`26, IC 50 = 510 nM
`
`HN
`
`O
`
`N
`
`NH2
`
`O
`
`F
`
`F
`
`29, IC5 0 = 0.4 nM
`Fig. (7). Optimization of the b-amino acid proline amide series.
`
`Merck Exhibit 2114, Page 7
`Mylan Pharmaceuticals Inc. v. Merck Sharp & Dohme Corp.
`IPR2020-00040
`
`

`

`564 Current Topics in Medicinal Chemistry, 2007, Vol. 7, No. 6
`
`Thornberry and Weber
`
`NHSO2M e
`
`NHSO2Me
`
`

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