throbber
2994
`J. Med. Chem.2002, 45,2994-3008
`Pyrazole Urea-Based Inhibitors of p38 MAP Kinase: From Lead Compound to
`Clinical Candidate
`John Regan,*,† Steffen Breitfelder,† Pier Cirillo,† Thomas Gilmore,† Anne G. Graham,‡ Eugene Hickey,†
`Bernhard Klaus,† Jeffrey Madwed,§ Monica Moriak,† Neil Moss,† Chris Pargellis,‡ Sue Pav,‡ Alfred Proto,‡
`Alan Swinamer,† Liang Tong,† and Carol Torcellini§
`Departments of Medicinal Chemistry, Biology, and Pharmacology, Boehringer Ingelheim Pharmaceuticals,
`Research and Development Center, 900 Ridgebury Road, Ridgefield, Connecticut 06877
`
`Received February 6, 2002
`
`We report on a series of N-pyrazole, N′-aryl ureas and their mode of binding to p38 mitogen
`activated protein kinase. Importantly, a key binding domain that is distinct from the adenosine
`5′-triphoshate (ATP) binding site is exposed when the conserved activation loop, consisting in
`part of Asp168-Phe169-Gly170, adopts a conformation permitting lipophilic and hydrogen
`bonding interactions between this class of inhibitors and the protein. We describe the correlation
`of the structure-activity relationships and crystallographic structures of these inhibitors with
`p38. In addition, we incorporated another binding pharmacophore that forms a hydrogen bond
`at the ATP binding site. This modification affords significant improvements in binding, cellular,
`and in vivo potencies resulting in the selection of 45 (BIRB 796) as a clinical candidate for the
`treatment of inflammatory diseases.
`In addition to the discovery of this important sig-
`Introduction
`nal
`transduction pathway, pyridinyl
`imidazole 1
`The proinflammatory cytokines tumor necrosis fac-
`(SB 203580)11 and analogues18-22 have been identified
`tor-R (TNF-R) and interleukin-1â (IL-1â) help regulate
`as potent and selective inhibitors of p38 MAP kinase.
`the body’s response to infections and cellular stresses.1
`Compound 1 was shown to be an effective orally active
`However, the pathophysiological consequences resulting
`agent in several animal models of acute and chronic
`from chronic and excessive production of TNF-R and IL-
`inflammation.23 Recently, an analogue of 1, compound
`1â are believed to underlie the progression of many
`2 (SB 242235), inhibited endotoxin-induced ex vivo
`inflammatory diseases such as rheumatoid arthritis
`production of TNF-R and IL-1â in human clinical
`(RA),2 Crohn’s disease, inflammatory bowel disease, and
`trials.24 The interest in p38 as a viable target for drug
`psoriasis.3 Recent data from clinical trials have secured
`intervention has escalated as a result of these early
`the continued use of the soluble TNF-R receptor fusion
`disclosures. In addition to a plethora of patent applica-
`protein, etanercept, or the chimeric TNF-R antibody,
`tions on imidazole-based compounds,25-27 several jour-
`infliximab, in the treatment of RA4-8 and Crohn’s
`nal papers have described strategies for the modification
`disease.9,10 The signal transduction pathway leading to
`of 1, by either the addition of other substituents to the
`the production of TNF-R from stimulated inflammatory
`imidazole or its replacement with different heterocycles.
`cells, while not fully understood, has been shown to be,
`in part, regulated by p38 mitogen activated protein
`These endeavors have produced imidazoles 328 and 4
`(MAP) kinase.11 p38 MAP kinase belongs to a group of
`(RPR200765A),29 a pyrrole analogue of 1,30 oxazole
`serine/threonine kinases that includes c-Jun NH2-
`5,31and pyrrolo[2,3-b]pyridine 6 (RWJ 68354).32 Imid-
`terminal kinase (JNK) and extracellular-regulated pro-
`azole 7 (RWJ 67657)33 was described to inhibit LPS-
`tein kinase (ERK).12 Upon extracellular stimulation by
`stimulated TNF-R production in human clinical
`a variety of conditions and agents,13 p38 is activated
`trials.34 Also, compounds with different structural fea-
`through bis-phosphorylation on a Thr-Gly-Tyr motif
`tures as compared to 1 have been reported as p38
`located in the activation loop. Activation is achieved by
`inhibitors. These include, among others, 8 (VX-745)35
`dual-specificity serine/threonine MAPK kinases, MKK3
`and N,N-diaryl urea 8a36 as well as pyrazole ketone 9
`and MKK6. Once activated, p38 can phosphorylate and
`(RO3201195)37 and pyrimido[4,5-d]pyrimidinone 10.38
`activate other kinases or transcription factors leading
`Indole amide 11 represents another group of p38 inhibi-
`to stabilized mRNA and an increase or decrease in the
`tors.39 A benzophenone class of p38 inhibitors, an
`expression of certain target genes.14-17
`example shown as 12 (EO1428), has recently been
`described.40 Diamides (13) are disclosed as p38 inhibi-
`* To whom correspondence
`should
`be
`tors41 (Chart 1).
`(203)798-4768.
`Fax:
`(203)791-6072.
`Our focus in cytokine-regulated approaches to inflam-
`rdg.boehringer-ingelheim.com.
`† Department of Medicinal Chemistry.
`matory diseases prompted us to evaluate the potential
`‡ Department of Biology.
`of p38 MAP kinase as a therapeutic target. Toward this
`§ Department of Pharmacology.
`10.1021/jm020057r CCC: $22.00 © 2002 American Chemical Society
`Published on Web 05/25/2002
`
`addressesd. Tel.:
`E-mail:
`jregan@
`
`FUSTIBAL Ex. 1010
`
`

`
`Pyrazole Urea-Based Inhibitors of p38 MAP Kinase
`Chart 1. Structural Classes of p38 MAP Kinase Inhibitors
`
`
`
`Journal of Medicinal Chemistry, 2002, Vol. 45, No. 142995
`
`Table 1. Substitution at Pyrazole N-2
`
`Scheme 1a
`
`(a) Phenylhydrazine, toluene, reflux or aqueous
`aReagents:
`HCl, ethanol, reflux. (b) 4-Chlorophenyl
`isocyanate, THF or
`CH2Cl2, 25 °C.
`14, coupled with its distinction as a new structural type
`of inhibitor vs others (e.g., 1-13) prompted us to under-
`take a systematic evaluation of its pharmacophores. The
`structure-activity relationships (SAR) for this class of
`compounds and their correlation to structural data,
`which led to the discovery of the clinical candidate BIRB
`796,45 are the subjects of this paper.
`Chemistry
`Modifications to the 2-position of the pyrazole nucleus
`were prepared as shown in Scheme 1 using 16 as a
`representative example of the compounds in Table 1.
`The assembly of pyrazole nucleus 15 involved the
`condensation of phenylhydrazine and 4,4-dimethyl-3-
`oxopentanenitrile in either toluene or aqueous HCl in
`ethanol at reflux. Urea formation was accomplished
`with 15 and 4-chlorophenyl isocyanate to produce 16.
`For examples in Table 1 requiring noncommercially
`available aryl hydrazines, the method of Demers46 was
`used to convert aryl halides to aryl hydrazines. The
`cyclohexylhydrazine that was used in the synthesis of
`
`end, compound 14 was identified from high throughput
`screening. Reports on utilizing this compound as a lead
`have been disclosed.42-44 While 14 showed only a modest
`binding affinity for human p38 MAP kinase (Kd ) 350
`nM) (Table 1), our interest in this molecule further
`increased upon obtaining a cocrystal structure with
`recombinant human p38. The unique binding mode of
`
`

`
`2996 Journal of Medicinal Chemistry, 2002, Vol. 45, No. 14
`Scheme 3a
`Table 2. Substitution at Pyrazole C-5
`
`Regan et al.
`
`Scheme 2a
`
`aReagents: (a) CNCH2CO2H, n-BuLi, THF, CH2Cl2, -70 °C and
`then 25 °C. (b) Phenylhydrazine, toluene, reflux. (c) THF, 25 °C.
`(d) LDA, THF, MeI, -78 °C. (e) NaH, CH3CN, THF, 75 °C.
`target compound 47 was obtained from the sodium
`cyanoborohydride-mediated reductive hydrazination of
`cyclohexanone with hydrazine.47
`Compound 46 was used as a frame of reference for
`probing the SAR at the 5-position of pyrazole by re-
`placing the t-butyl moiety (Table 2). This effort required
`the construction of a diverse set of oxopentanenitrile
`subunits. Scheme 2 outlines two general procedures to
`prepare oxopentanenitrile derivatives. Briefly, the ad-
`dition of the dianion of cyanoacetic acid48 to acid
`chlorides (e.g., 17) or the anion of acetonitrile49 to esters
`(e.g., 22) supplied the â-keto nitrile components. Fol-
`lowing the chemistry described in Scheme 1, pyrazole
`formation and urea couplings were completed for target
`20 as well as the other compounds in Table 2.
`To evaluate the role of the urea linkage to the binding
`of p38, the synthesis of several urea analogues was
`undertaken. The biological role of each of the urea N-H
`groups in 16 was examined by replacement with CH2
`(25 and 31) and N-methyl (34 and 37). Amides 25 and
`31 were prepared as shown in Scheme 3. EDC-mediated
`condensation of aminopyrazole 15 and 4-chlorophenyl-
`acetic acid (24) furnished amide 25. Amide 31, however,
`required the construction of pyrazole acetic acid 30.
`Thus, pyrazolidinone 26 was converted to its O-triflate
`derivative 27, which underwent Stille cross coupling
`with tributyl(vinyl)tin to give vinyl pyrazole 28. Regio-
`selective hydroboration of 28 produced alcohol 29, which
`was converted to the desired carboxylic acid 30 with
`Jones reagent. Amide bond formation between 30 and
`4-chloroaniline with DCC furnished 31. The syntheses
`of N-methyl urea analogues 34 and 37 were undertaken
`as follows. Exposure of aminopyrazole 15 to phosgene50
`
`aReagents: (a) EDC, CH2Cl2. (b) Tf2O, DTBMP, CH2Cl2, -78
`to 0 °C. (c) Tributyl(vinyl)tin, Pd[P(Ph)3]4, LiCl, dioxane, 100 °C.
`(d) (i) 9-BBN, THF, reflux. (ii) NaOH, H2O2. (e) Jones reagent. (f)
`4-Chloroaniline, DCC, DMAP, CH2Cl2. (g) COCl2, CH2Cl2, aqueous
`NaHCO3. (h) CH2Cl2, 25 °C. (i) HCO2H, reflux. (j) BH3-DMS, THF,
`25 °C. (k) 4-Chlorophenyl isocyanate, CH2Cl2, 25 °C. (l) 4-Chloro-
`phenyl isothiocyanate, CH2Cl2, 25 °C.
`produced pyrazole isocyanate 32, which was coupled
`with N-methyl-4-chloraniline (33) to provide N-methyl
`urea 34. Alternatively, aminopyrazole 15 was heated
`with formic acid to produce N-formyl aminopyrazole 35,
`which upon reduction with borane51 yielded N-methyl-
`aminopyrazole 36. Treatment of 36 with 4-chlorophenyl
`isocyanate produced N-methyl urea analogue 37. Thio-
`urea 38 served as a basis to understand the part that
`the O-atom plays in p38 binding, and its preparation
`was accomplished by treatment of aminopyrazole 15
`with 4-chlorophenyl isothiocyanate.
`The compounds designed to explore the region of the
`urea phenyl of 46 are summarized in Table 4. They were
`conveniently obtained by the treatment of pyrazole
`isocyanate 32 with aniline derivatives or alkylamines.
`For example, as shown in Scheme 4, exposure of
`isocyanate 32 to 2-aminoindan (39) furnished urea 40.
`Other target ureas were prepared according to Scheme
`1 wherein amine 15 was coupled to aryl isocyanates.
`To access target compounds with groups attached
`to the 4-position of the urea naphthalene, the route
`shown in Scheme 5 was utilized. Alkylation of N-Boc
`naphthol 41, prepared from 4-amino-1-naphthol with
`4-(2-chloroethyl)morpholine, gave ether 42. Removal of
`the Boc protecting group (43) and urea formation, as
`described above with the isocyanate derived from 44,
`gave 45.
`
`

`
`Pyrazole Urea-Based Inhibitors of p38 MAP Kinase
`Table 3. Urea Modifications
`
`Scheme 4a
`
`
`
`Journal of Medicinal Chemistry, 2002, Vol. 45, No. 142997
`
`aReagents: (a) CH2Cl2, 25 °C.
`Gly170 (DFG) of the kinase is required for the observed
`binding mode of the diaryl urea inhibitor (Figure 2). In
`all of the currently known protein Ser/Thr kinase
`structures, the residues assume a conformation such
`that the Phe side chain is buried in a hydrophobic pocket
`in the groove between the two lobes of the kinase (DFG-
`in conformation). In the structure of the complex with
`compound 14, however, the Phe side chain has moved
`by about 10 Å to a new position (DFG-out conformation).
`In this position, one face of the Phe side chain and the
`urea phenyl ring are involved in hydrophobic interac-
`tions whereas the other face is exposed to solvent. This
`movement of the Phe side chain reveals a large hydro-
`phobic domain in the kinase, and the tert-butyl group
`of 14 inserts deep into this pocket (Figure 2). Neither
`nitrogen atom on the pyrazole ring participates in
`specific hydrogen-bonding interactions with the kinase.
`As shown in Figure 3, the urea of 14 establishes a
`bidentate hydrogen bond with the conserved side chain
`of Glu71.
`Most protein kinase inhibitors use the ATP binding
`pocket and inhibit the kinase by directly competing with
`the binding of ATP. In contrast, compound 14 does not
`compete directly with ATP binding, as it has no struc-
`tural overlap with the ATP molecule (Figure 2). How-
`ever, our structure shows that the DFG-out conforma-
`tion impedes ATP binding, as the side chain of the Phe
`residue would be sterically incompatible with the phos-
`phate groups of ATP (Figure 2). This is supported by
`our observation that compound 14 interferes with the
`inactivation of p38 MAP kinase activity by the fluores-
`cent ATP analogue 5′-p-fluorosulfonyl benzoyl adenosine
`(data not shown). Therefore, the diaryl urea compounds
`inhibit p38 MAP kinase by stabilizing a conformation
`of the kinase that is incompatible with ATP binding.
`The data in Tables 1 and 2 highlight the binding roles
`of the groups appended to the pyrazole nucleus of 14s
`methyl at N-2 and tert-butyl at C-5. Fortuitously, our
`first modification, replacement of the methyl of 14 with
`a phenyl group (16), improved binding potency 40-fold
`(Table 1) as measured in a fluorescent binding assay.
`The crystal structure of 16 and the recombinant human
`p38 complex (Figure 4) help rationalize this result. The
`phenyl ring at N-2 of the pyrazole participates in
`lipophilic interactions with the alkyl portion of the side
`chain of the Glu71 residue. In addition, the phenyl ring
`may serve as a water shield for the hydrogen bond
`network of the urea and the Glu71 carboxylate. The
`presence of the phenyl ring causes this Glu residue to
`adopt a side chain conformation that results in a
`monodentate hydrogen-bonding interaction with the
`urea moiety of the inhibitor. This alignment of Glu71
`is in contrast to the bidentate interactions in the
`complex with 14 (Figure 3).
`Further profiling of this key region in the inhibitor
`helped establish the preferred substitution at N-2 of the
`
`Table 4. Modification of Urea-Phenyl Ring
`
`Results and Discussion
`We recently reported the crystal structure of recom-
`binant human p38 MAP kinase in complex with com-
`pound 14 at 2.5 Å resolution (Figure 1).45 Interestingly,
`the crystal structure reveals that this compound utilizes
`binding interactions on the kinase that are spatially
`distinct from the adenosine 5′-triphosphate (ATP) pocket.
`There is no structural overlap between the atoms of
`compound 14 and the ATP (Figure 2). Similarly, there
`is only limited spatial overlap between 14 and an iodo
`analogue of SB203580 (1a)52 and this occurs in a
`lipophilic pocket commonly referred to in the kinase field
`as the specificity pocket (Figure 3). A large conforma-
`tional change for conserved residues Asp168-Phe169-
`
`

`
`2998 Journal of Medicinal Chemistry, 2002, Vol. 45, No. 14
`Scheme 5a
`
`Regan et al.
`
`aReagents: (a) Di-tert-butyl dicarbonate, THF, 25 °C. (b) 4-(2-Chloroethyl)morpholine, K2CO3, acetonitrile, heat. (c) HCl, dioxane. (d)
`Compound 44, phosgene, THF.
`
`Figure 1. Crystal structure of human p38 MAP kinase and
`14 at 2.5 Å resolution.
`pyrazole and confirmed our hypothesis regarding bind-
`ing interactions at this domain. The diminished potency
`of saturated derivative 47 highlighted the necessity for
`an aromatic ring to achieve optimal hydrophobic inter-
`actions (Table 1). Addition of methyl groups to the 3-
`and 4-position of the phenyl ring of 46 provided modest
`improvements in binding (49-51). However, 2-methyl
`derivative 48 displayed a substantial loss of binding
`affinity possibly due to an increase in the torsional angle
`favored between the phenyl and the pyrazole rings
`beyond the observed angle of 54° for 16 (Figure 4). This
`position tolerates bulkier groups as judged by the
`binding potency of 2-naphthyl analogue 52. In addi-
`tion, heteroatoms (53-57) can be accommodated at this
`site. The close proximity of the 3- and 4-positions of
`the phenyl ring of 46 to solvent may explain these
`results.
`In Figure 4, the tert-butyl group at C-5 of pyrazole
`16 is embedded deep into a hydrophobic pocket formed
`by the reorganization of Phe169 in the DFG-out con-
`formation. In an effort to understand the binding role
`of the tert-butyl moiety in this class of compounds, we
`investigated the size and electronic requirements of this
`group. As can be seen in Table 2, removal of one methyl
`group lowered potency over 20-fold (cf. 46 vs 59).
`Further reduction to a methyl resulted in an inactive
`
`Figure 2. Overlap of 14 (blue) and ATP (red). The urea
`hydrogen atoms are shown for clarity. Phe169 is shown in red
`when occupying the DFG-in conformation (ATP bound) and
`in blue in the DFG-out conformation when 14 is bound to p38.
`compound (58). This lipophilic binding pocket tolerated
`bulkier tert-alkyl groups such as dimethylethyl (60) and
`methylcyclohexyl (20). However, the 50-fold loss of
`binding observed with dimethylbenzyl analogue 64 may
`indicate a size limitation for this domain. A comparison
`of cyclohexyl derivatives 62 and 20 further exemplifies
`the strong preference for a tertiary group. The relatively
`poor activity of compounds 61 and 63 as compared to
`60 and 20 suggest that lipophilic substitution at C-5 of
`the pyrazole is favored. Taken together, these results
`are rationalizable based on the crystal structure of 16
`and p38 (Figure 4) that indicate a lipophilic group at
`C-5 of the pyrazole has important hydrophobic binding
`interactions with the protein in the DFG-out conforma-
`tion. The tert-butyl group was incorporated into all
`subsequent target molecules since it offered the best
`balance of potency and physicochemical properties.
`The X-ray crystallographic structure of 16 with p38
`reveals a hydrogen bond network consisting of a urea
`hydrogen and the carboxylate oxygen of Glu71 and also
`the urea oxygen and N-H of Asp168. The data in Table
`3 highlight the significance of these interactions on
`binding affinity. Replacement of either N-H in the urea
`with a methylene group (compounds 25 and 31) or
`introduction of N-methyl (34 and 37) results in signifi-
`cant loss of activity. Likewise, the thiourea analogue 38
`shows a 60-fold decrease in binding potency to p38 as
`compared to 16. These findings underscore the crucial
`contribution that the urea makes to binding with p38
`through extensive hydrogen bonding and, also likely,
`to establishing the correct geometric relationships of the
`other pharmacophores of the inhibitor.
`
`

`
`Pyrazole Urea-Based Inhibitors of p38 MAP Kinase
`
`
`
`Journal of Medicinal Chemistry, 2002, Vol. 45, No. 142999
`
`Figure 3. Two views of the overlap of 14 and 1a. The urea hydrogen atoms are shown for clarity. The urea phenyl group of 14
`occupies the same kinase specificity pocket in p38 as the phenyl ring of 1a. The bindentate hydrogen bond interaction between
`the urea hydrogen atoms and the carboxylate oxygens of Glu71 is shown.
`can also provide good binding affinity as seen with
`bicyclic indan derivatives (40 and 77). Thus, these data
`demonstrate that the kinase specificity pocket of p38
`favors lipophilic pharamacophores that are not limited
`to phenyl rings.
`We examined several compounds from this series for
`oral activity in a mouse model of LPS-stimulated TNF-R
`synthesis. Compound 50 (Table 5) furnished an inter-
`esting and important result. The tolyl group on 50
`provided a 100-fold increase in plasma concentration in
`the mouse vs 46, which lacks this substitution. The
`increased plasma levels in combination with a modest
`improvement in cellular activity provided our first orally
`active compound. Of the 4-methylphenyl derivatives
`examined in this model, analogue 78 showed the best
`in vivo profile. This compound, with even higher plasma
`concentrations than 50, suppressed TNF-R production
`by 90% when dosed at 100 mg/kg and also was active
`at 30 mg/kg (53% inhibition).
`Unfortunately, we were unable to improve the in vitro
`and in vivo activities of the phenyl-based urea inhibitors
`beyond that of compound 78. Despite available crystal-
`lographic data, obvious solutions to achieve additional
`binding interactions were not realized. A breakthrough
`arrived upon establishing a binding assay having more
`sensitivity for compounds whose binding activity was
`near the limit of the fluorescence assay.45 One observa-
`tion from this new assay suggested that 79 was more
`potent than initially thought. Table 6 shows selected
`examples of the Kd values from the fluorescence assay
`vs the exchange curve assay. Remarkably, naphthyl
`compound 79 binds 20-fold more tightly to p38 than
`phenyl analogue 50 despite similar cellular potencies.
`The overlap of the X-ray crystal structures of phenyl
`analogue 16 and naphthyl 75 (Figure 5) provides a
`possible explanation for this increased binding. The
`naphthyl moiety of 75 resides deep in the kinase
`specificity pocket and achieves substantial hydrophobic
`binding interactions with the protein that are not
`possible with the phenyl ring of 16. Thus, it seemed
`reasonable that the lack of improvement in cellular
`
`Figure 4. X-ray crystallographic structure of human p38 with
`16. The urea hydrogen atoms are shown for clarity. The
`hydrophobic effects of the pyrazole phenyl ring and the
`monodendate hydrogen bond of the urea N-H atoms with
`Glu71 are seen.
`As seen in Figure 3, the phenyl ring attached to the
`urea of 14 fits into the specificity pocket of p38 in a
`manner similar to the pyridinyl imidazole inhibitor 1a.52
`The importance of binding in this pocket to potency53
`and kinase specificity28 has been described for the
`imidazole-based group of inhibitors. Highlights of the
`prominent role that the phenyl ring plays in binding
`with this series of compounds are shown in Table 4.
`Removal of the urea phenyl ring results in complete loss
`of binding potency (cf. 46 and 65). Saturation of the
`phenyl ring (66) or separation of the ring from the urea
`by either one or two carbon atoms (73 and 74) resulted
`in decreased binding affinity. Incorporation of polar
`groups, through either pyridine (67-69) or aniline
`lowers potency. However,
`derivatives (70 and 71),
`lipophilic groups appended to the phenyl nucleus can
`improve potency (cf. 72 and 75). Other lipophilic groups
`
`

`
`3000 Journal of Medicinal Chemistry, 2002, Vol. 45, No. 14
`Table 5. In Vivo Activity of Selected Pyrazole-Phenyl Ureas
`
`Regan et al.
`
`Table 6. Comparison of Phenyl vs Naphthyl Ureas with ATP
`Site Binding Pharmacophore
`
`Figure 5. Overlap of 16 (yellow) and 75 (green) with human
`p38 MAP kinase. The urea hydrogen atoms are shown for
`clarity. The naphthyl group of 75 fits deeper into the kinase
`specificity pocket as compared to the phenyl ring of 16. The
`4-position of the naphthalene ring points toward the hinge
`region and solvent. Phe169 is removed for clarity.
`activity of 79 might be a consequence of its higher
`lipophilicity as compared to 50.
`The crystal structure of 75, in addition to providing
`a rationale to the improved binding affinity of the
`naphthalene group, offered an opportunity to explore
`modifications from this platform that would be unavail-
`
`able from the phenyl ring of 16. That is, groups
`appended to the 4-position of the naphthalene could
`much more readily access the ATP binding region of p38
`than the 4-positon of the phenyl ring. Hence, pharma-
`cophores attached to the 4-position could have ad-
`ditional binding interactions or be used to improve
`physicochemical properties. The ethoxy morpholine
`group proved to be a very effective group for achieving
`both of these goals. This moiety improved binding
`potency 10-fold and, more significantly, increased cell
`activity over 20-fold. A crystal structure of 45 with
`recombinant human p38 (Figure 6a) provided an ex-
`planation for the enhanced potency.45 The gauche
`conformation of the ethoxy linker of 4554,55 effectively
`orients the morpholine group so that the morpholine
`oxygen can achieve a strong hydrogen bond with the
`N-H of Met109. This hydrogen bond is the same one
`used by the adenine base of ATP and the pyridine
`nitrogen of the pyridinyl imidazole class of compounds
`such as 1a.52 The favorable edge to π hydrophilic
`interactions of the phenyl ring of Phe169 and the
`naphthalene group of 45 is revealed in Figure 6b and
`likely further contributes to binding potency.
`To highlight the contribution of the naphthalene
`group to 45, we prepared phenyl derivative 80. Despite
`the inclusion of the ethoxy morpholine unit in phenyl
`analogue 80, it exhibited a dramatic loss in potency vs
`45 (Table 6). This result reinforces the importance of
`the naphthalene ring in providing better lipophilic
`interactions with the specificity pocket and properly
`aligning the ethoxy morpholine unit for productive
`binding with the ATP binding region.
`In addition to superior in vitro and cellular activities,
`compound 45 demonstrated enhanced in vivo potency.
`For example, in a mouse model of LPS-stimulated
`TNF-R synthesis, a 65% inhibition of TNF-R synthesis
`was observed when 45 was dosed orally at 10 mg/kg.
`In a 5 week model of established collagen-induced
`arthritis using B10.RIII mice, 45 produced a 63%
`inhibition of arthritis severity when dosed orally at 30
`mg/kg qd.56 Some pharmacokinetic data of 45 in mice
`and cynomolgous monkeys are summarized in Table 7.
`The selectivity profile against a panel of protein kinases
`for 45 was determined and is shown in Table 8.45 On
`the basis of these and other data, compound 45 (BIRB
`796) was selected for human clinical trials.
`
`

`
`Pyrazole Urea-Based Inhibitors of p38 MAP Kinase
`
`
`
`Journal of Medicinal Chemistry, 2002, Vol. 45, No. 143001
`
`(a) X-ray crystallographic complex of human p38 and 45. Phe169 is removed for clarity. (b) X-ray crystallographic
`Figure 6.
`complex of human p38 and 45 with Phe169.
`Table 7. Pharmacokinetic Properties of Compound 45
`
`Table 8. Selectivity Profile of Compound 45
`
`Conclusion
`We have shown that a series of N-pyrazole, N′-aryl
`ureas occupy a binding domain on p38 that is exposed
`when the conserved binding loop, consisting in part of
`Asp168-Phe169-Gly170, adopts a conformation (DFG-
`out) not previously noted in other protein Ser/Thr
`kinases. A 40-fold improvement in binding was achieved
`by the replacement of the methyl group in the original
`screening lead (14) with phenyl. The urea atoms, shown
`to be involved in an extensive hydrogen bond network
`with Glu71 and Asp168 (Figures 3 and 4), proved critical
`for binding activity. A toluene ring attached to the
`pyrazole nucleus was necessary to secure high plasma
`levels in the mouse. The naphthalene was a preferred
`pharmacophore as compared to phenyl to bind in the
`kinase specificity pocket. We added an ethoxy morpho-
`line pharmacophore, which successfully extended the
`binding of the inhibitor to also include a hydrogen-
`bonding interaction in the ATP binding region of p38.
`This modification afforded significant improvements in
`binding affinity, cellular activity, and in vivo reduction
`of TNF-R production and arthritis severity that resulted
`
`in the selection of 45 (BIRB 796) as a clinical candidate
`for the treatment of inflammatory diseases.45,57
`Experimental Section
`All solvents and reagents were obtained from commercial
`sources and used without further purification unless indicated
`otherwise. Melting points were obtained from a Mel-temp 3.0
`or Fisher-Johns melting point apparatus and are uncorrected.
`1H nuclear magnetic resonance (NMR) spectrum were recorded
`on either a Bruker AC-F-270 spectrometer or Bruker Avance
`DPX 400 spectrometer. Chemical shifts are reported in parts
`per million (δ) from the tetramethylsilane resonance in the
`indicated solvent. Mass spectra were obtained from a Finni-
`gan-SSQ7000 spectrometer. Samples were generally intro-
`duced by particle beam and ionized with NH4Cl. Thin-layer
`chromatography (TLC) analytical separations were conducted
`with E. Merck silica gel F-254 plates of 0.25 mm thickness
`and were visualized with UV or I2. Flash chromatographies
`were performed according to the procedure of Still et al. (EM
`Science Kieselgel 60, 70-230 mesh). Elemental analysis were
`performed at Quantitative Technologies, Inc., Whitehouse, NJ.
`1-(5-tert-Butyl-2-phenyl-2H-pyrazol-3-yl)-3-(4-chloro-
`phenyl)urea (16). A solution of phenyl hydrazine (0.83 mL,
`8.39 mmol) and 4,4-dimethyl-3-oxo-pentanenitrile (1.0 g, 8.0
`mmol) in toluene (3 mL) was heated to reflux overnight.
`Removal of the volatiles in vacuo provided a residue, which
`was purified by silica gel chromatography using 50% ethyl
`acetate in hexanes as the eluent. Concentration in vacuo of
`the product-rich fractions provided 3-amino-5-tert-butyl-2-
`phenyl-2H-pyrazole (15) as a light orange solid (1.53 g, 89%).
`A solution of 15 (0.058 g 0.27 mmol) and 4-chlorophenyl
`isocyanate (0.038 g, 0.25 mmol) in CH2Cl2 (1 mL) was stirred
`overnight at room temperature under inert atmosphere.
`Removal of the volatiles in vacuo provided a residue, which
`was triturated with 50% dichloromethane in hexanes (2 mL).
`The urea (16) was filtered and dried in vacuo to afford 0.078
`g (85%) and was then recrystallized from methanol to afford
`analytically pure material; mp 202-203 °C. 1H NMR (400
`
`

`
`3002 Journal of Medicinal Chemistry, 2002, Vol. 45, No. 14
`Regan et al.
`MHz, dimethyl sulfoxide (DMSO)-d6): δ 1.27 (s, 9H, tert-butyl),
`Trifluoromethanesulfonic Acid 5-tert-Butyl-2-phenyl-
`6.36 (s, 1H, pyrazole), 7.28-7.30 (m, 2H, aromatic), 7.39-7.43
`2H-pyrazol-3-yl Ester (27). To a solution of 5-tert-butyl-2-
`(m, 3H, aromatic), 7.50-7.52 (m, 4H, aromatic), 8.42 (s, 1H,
`phenyl-2,4-dihydro-pyrazol-3-one (26, 5.12 g, 23.7 mmol) and
`urea), 9.12 (s, 1H, urea). MS (NH3-CI): m/e369 (MH+). Anal.
`2,6-di-tert-butyl-4-methyl pyridine (6.12 g, 29.8 mmol) in
`(C20H21ClN4O‚CH3OH) C, H, N.
`CH2Cl2 (50 mL) was added dropwise trifluoromethanesulfonic
`anhydride (4.4 mL, 7.4 g, 26 mmol) at -78 °C. The resulting
`1-[5-(1-Methylcyclohexyl)-2-phenyl-2H-pyrazol-3-yl]-3-
`solution was warmed to 0 °C, saturated NaHCO3 solution (100
`phenyl-urea (20). A solution of cyclohexane-1-methyl-1-
`mL) was added, and the mixture was stirred vigorously for 10
`carboxylic acid (1.31 g, 9.21 mmol), oxalyl chloride (5.5 mL of
`min. The layers were separated, and the aqueous layer was
`a 2.0 M solution in CH2Cl2, 11.05 mmol), and a drop of
`extracted with CH2Cl2 (3×). The combined organic layers were
`anhydrous dimethylformamide (DMF) in CH2Cl2 (5 mL) was
`washed with brine and dried (Na2SO4). Removal of the volatiles
`heated to reflux for 3 h and cooled to ambient temperature to
`in vacuo provided a residue, which was purified by flash
`give 17. In a separate flask to a solution of cyanoacetic acid
`chromatography eluting unpolar impurities with hexanes and
`(1.57 g, 18.4 mmol, freshly dried with MgSO4) and a catalytic
`subsequently eluting the product with hexanes:ethyl acetate
`amount of 2,2′-bipyridine in anhydrous tetrahydrofuran (THF)
`(20:1). Concentration in vacuo of the product-rich fractions
`(68 mL) at -70 °C under an inert atmosphere was added
`gave 8.19 g (99%) of yellow oil 27. 1H NMR (400 MHz,
`dropwise n-butyllithium (15 mL of a 2.5 M solution in hexanes,
`CDCl3): δ 1.34 (s, 9H, tert-butyl), 6.19 (s, 1H, pyrazole), 7.35
`37.2 mmol). The mixture was slowly warmed to 0 °C until a
`(dd, 3J1 ) 3J2 ) 7.4 Hz, 1H, phenyl), 7.4 (dd, 3J1 ) 3J2 ) 7.8
`persistent red-colored slurry was obtained. The mixture was
`Hz, 2H, phenyl), 7.54 (d, 7.96 Hz, 2H, phenyl).
`cooled to -70 °C, and 17 in CH2Cl2 was slowly added. The
`3-tert-Butyl-1-phenyl-5-vinyl-1H-pyrazole (28). A solu-
`mixture was slowly warmed to room temperature, stirred for
`1 h, and quenched with 2 N aqueous HCl. The aqueous layer
`tion of 27 (3.74 g, 10.7 mmol) in dioxane (90 mL) in a sealable
`was extracted twice with CHCl3. The combined organic layers
`tube was degassed under vacuum and charged with nitrogen.
`LiCl (2.91 g, 68.8 mmol) was added, and the mixture was
`were washed with saturated aqueous NaHCO3 and brine and
`degassed and charged with nitrogen again. Pd(PPh3)4 (0.491
`dried (MgSO4). Removal of the volatiles in vacuo provided a
`g, 0.425 mmol) was added, and the mixture was degassed and
`residue, which was purified by silica gel chromatography using
`ethyl acetate in hexanes as the eluent. Concentration in vacuo
`charged with nitrogen again. Tributyl(vinyl) tin (4.0 mL, 4.3
`of the product-rich fractions provided 1.26 g of 18. A mixture
`g, 14 mmol) was added, and the mixture was degassed and
`of 18 (0.80 g, 4.8 mmol) and phenylhydrazine (0.48 mL, 4.8
`charged with nitrogen again. The tube was sealed, and the
`mmol) in dry toluene (6 mL) was heated to reflux overnight.
`mixture was heated to 100 °C overnight. After it was cooled
`Removal of the volatiles in vacuo provided a residue, which
`to room temperature, the volatiles were removed in vacuo and
`was purified by silica gel chromatography. Concentration in
`the residue was purified by flash chromatography eluting
`vacuo of the product-rich fractions provided 1.10 g (90%) of
`unpolar im

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