Discovery of AZD3147: A Potent, Selective Dual Inhibitor of mTORC1

Feb 2, 2015 - The authors thank Dr. Paul Johnson, John Bailey, Jennifer Peed, Matthew Box, Greg Carr, George Hill, Gillian Lamont, Steve Glossop, and ...
0 downloads 0 Views 3MB Size
Article pubs.acs.org/jmc

Discovery of AZD3147: A Potent, Selective Dual Inhibitor of mTORC1 and mTORC2 Kurt G. Pike,* Jeff Morris, Linette Ruston, Sarah L. Pass, Ryan Greenwood, Emma J. Williams, Julie Demeritt, Janet D. Culshaw, Kristy Gill, Martin Pass, M. Raymond V. Finlay, Catherine J. Good, Craig A. Roberts, Gordon S. Currie, Kevin Blades, Jonathan M. Eden, and Stuart E. Pearson Oncology Innovative Medicines, AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TG, U.K. ABSTRACT: High throughput screening followed by a lead generation campaign uncovered a novel series of urea containing morpholinopyrimidine compounds which act as potent and selective dual inhibitors of mTORC1 and mTORC2. We describe the continued compound optimization campaign for this series, in particular focused on identifying compounds with improved cellular potency, improved aqueous solubility, and good stability in human hepatocyte incubations. Knowledge from empirical SAR investigations was combined with an understanding of the molecular interactions in the crystal lattice to improve both cellular potency and solubility, and the composite parameters of LLE and pIC50−pSolubility were used to assess compound quality and progress. Predictive models were employed to efficiently mine the attractive chemical space identified resulting in the discovery of 42 (AZD3147), an extremely potent and selective dual inhibitor of mTORC1 and mTORC2 with physicochemical and pharmacokinetic properties suitable for development as a potential clinical candidate.



treated with rapamycin.12 This suggests that by inhibiting mTORC2 in addition to mTORC1, and thus having a direct inhibitory effect on Akt signialling, a compound may offer greater clinical benefit compared with rapalogues. In addition, dual mTORC1 and mTORC2 inhibitors may exhibit a broader spectrum of clinical activity. A number of compounds are known to inhibit mTOR and other members of the PIKK family and have been reviewed extensively elsewhere.13 Early reported ATP-competitive inhibitors of mTOR were found to inhibit the activity of a number of different PIKK enzymes although were generally selective when tested more broadly against other kinase targets, such as LY294002 which is a moderate inhibitor of PI3K, mTOR, and DNA-PK. Following significant investment from a number of groups, more potent dual PI3K/mTOR inhibitors have been discovered, such as NVP-BEZ235 and PI103. Compounds with a high degree of selectivity for mTOR over the PI3K isoforms and the other PIKK enzymes have also subsequently been reported, such as AZD8055, PP242, AZD2014, and WYE-354 (Figure 1). More recently compounds have been disclosed that show selectivity for other members of the PIKK family such as Ku-0060648, a potent and selective inhibitor of DNAPK. While NVPBEZ235 and PP242 contain classical kinase hinge binding motifs, compounds such as LY294002, PI-103, AZD8055, AZD2014, WYE-354, and Ku-0060648 are believed to bind to the kinase hinge region through the morpholine oxygen in a fashion analogous to that observed for LY294002 in PI3Kγ.14

INTRODUCTION The mammalian target of rapamycin (mTOR) is a key target in the development of antitumor therapies.1 Activated by growth factor/mitogenic stimulation activation of the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway, mTOR is a central regulator of cell growth and proliferation. This PI3K-Akt-mTOR pathway is one of the most frequently dysregulated pathways in cancer.2 mTOR, a serine/theronine kinase of approximately 289 kDa in size, is a member of the evolutionary conserved eukaryotic PI3K-related kinase (PIKK) family of atypical protein kinases which also include the protein kinases DNA-PK (DNA dependent protein kinase), ATM (ataxia-telangiectasia mutated) and ATR (ataxiatelangiectasia and Rad3 related).3−5 The known mTOR inhibitor rapamycin and its analogues (RAD001, CCI-779, AP23573) bind to the FKBP12/ rapamycin complex binding domain (FRB), resulting in suppression of signaling to the downstream targets p70S6K and 4E-BP1.6,7 The potent but nonspecific inhibitors of PI3K, LY294002 and wortmannin, have also been shown to inhibit the kinase function of mTOR; however, in this case the catalytic domain of the protein is targeted.8 Recently it has been shown that mTOR can exist in an alternative, rapamycin insensitive complex that signals to Akt.9 The existence of both a rapamycin sensitive complex (mTORC1) and a rapamycin insensitive complex (mTORC2) may provide an explanation for the differences observed in the earlier work of Brunn et al.8 and Edinger et al.10 Rapamycin and its analogues have been shown to activate AKT signaling as a consequence of inhibition of the negative feedback loop downstream of mTORC1,11 which is associated with a shorter time to progression in glioblastoma patients © XXXX American Chemical Society

Received: November 16, 2014

A

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry

Figure 1. Structures of reported ATP-competitive PIKK inhibitors.

A recent crystal structure of PI-103 bound into the mTOR protein further supports this binding mode.15 We have previously reported the identification of compound 1 following a screen of the AstraZeneca compound collection against a recombinant mTOR enzyme assay (Figure 2a).16 Although of only modest potency against mTOR (pIC50 = 5.85), compound 1 was considered of particular interest because of its significantly lower affinity for PI3Kα (pIC50 = 4.76).17 Optimization of this novel scaffold resulted in the identification of 2 showing increased mTOR affinity (pIC50 = 6.54) and selectivity over PI3Kα (pIC50 = 4.58). The importance of the indole hydrogen bond donor in 2 was established through structure−activity relationship (SAR) studies and rationalized using a homology model of the mTOR ATP binding site built upon the ATP binding site of PI3Kγ.14,18 The indole is believed to form a productive interaction with a glutamic acid residue present in mTOR but not PI3K (Figure 2b).19 Comparison of the homology model with the recently reported crystal structure of mTOR15 reveals no significant differences in protein structure and further validates the use of the homology model to help guide design. Subsequent optimization looked to maintain this key interaction and resulted in the discovery of the urea containing compound 3 in which mTOR affinity has been further increased (pIC50 = 7.55) while selectivity over PI3Kα has been maintained (pIC50 = 6.25). Compound 3 is a potent inhibitor of substrates of both mTORC1 (phosphorylation of Ser 235/236 on S6) and mTORC2 (phosphorylation of Ser 473 on Akt) in MDA-MB-468 cells (pIC50 of 6.92 and 7.06, respectively).20 However, the low aqueous solubility of 3 ( 3.2) together with the ability to form strong crystal lattices due the planar nature of the molecule and the potential to form extended hydrogen bonding networks. The identification of compounds with improved cellular potency and aqueous solubility, while maintaining or improving selectivity over PI3K, became the primary focus for the optimization campaign. To improve the chance that any compound resulting from this optimization campaign would be of sufficient quality to warrant clinical development, the team aimed to equal or surpass the cellular potency and aqueous solubility reported for the known clinical candidate AZD8055 (cellular potencies as measured against pS6(S235/236) and pAKT(S473) were pIC50 = 7.57 and pIC50 = 7.62, respectively, and the aqueous solubility was measured as 30 μM).26,27 The concept of using composite parameters as a measure of compound quality and progress within a series, such as ligand efficiency (LE), lipophilic ligand efficiency (LipE), or unbound clearance (Clu), is well established within the medicinal chemistry community.28 Considering the issues faced within this novel series of mTOR inhibitors, namely, optimizing potency and aqueous solubility, we adopted the composite parameter of cellular pIC50 − p(aqueous solubility [M]) as a measure of compound progression. A large internal data set (data not shown) has established a good correlation for cellular potency values as measured by both the pAKT(S473) and the pS6(S235/236) end points within this series, and in an effort to simplify the screening cascade, only the pAKT(S473) data were measured routinely and used to drive chemical optimization. Cellular potency against the pS6(S235/236) end point, alongside mTOR enzyme potency and PI3K enzyme potency, was generated subsequently for key compounds. Previous groups have reported how subtle changes to the morpholine hinge binding motif can have significant impact on mTOR potency, selectivity over PI3K, and physicochemical properties.29 In particular, work from our own laboratories had highlighted a switch to 3(S)-methylmorpholine as being associated with improved cellular potency and aqueous solubility for related series.16,26 Similar effects were observed in this novel series of urea containing compounds as

T (80% yield). The lower yield for the construction of intermediate R possibly reflects the less favorable nature of constructing four-membered rings compared to rings of other sizes. The tetrahydropyran ring in intermediate V was constructed by the double deprotonation of D followed by reaction with bis(bromoethyl) ether in good yield (78%). Palladium mediated cross-coupling reactions were used to convert intermediates N, P, R, T, V to intermediates O, Q, S, U, W in moderate to good yields (50−92%). These intermediates were converted to compounds 26−32 by reaction with phenyl chloroformate followed by the appropriate amine in moderate to good yields (39−75% over two steps). The synthesis of compounds 33−44 is outlined in Scheme 5 and Scheme 6.25,23 The hydroxypropylsulfone moiety in intermediate X was introduced by displacement of iodide C with 3-mercaptopropan-1-ol followed by subsequent oxidation with mCPBA (53% yield over two steps). Protection of the alcoholic funtionality as a triisopropylsilyl ether, cyclopropanation using phase transfer conditions, and subsequent fluoride mediated desilylation gave Y in an overall yield of 54%. Y was converted to Z in a 71% yield using a palladium mediated cross-coupling reaction, and final conversion to 33 was achieved in a 59% yield. The 4-pyridylsulfone moiety present in intermediate AA was also introduced through a two-step displacement/oxidation proceedure; however, in this instance sodium tungstate and hydrogen peroxide were used as the oxidants to limit N-oxide formation. Intermediate AA was prepared in 79% yield from intermediate C. Reaction of intermediate C with either 4-fluorobenzenesulfinic acid or benzenesulfinic acid furnished intermediates AB and AC in a 76% and 85% yield, respectively. Phase transfer cyclopropanation was performed on intermediates L, AA, AB, and AC to give intermediates AD, AE, AF, and AG in excellent yield (82−93%). These intermediates could be further functionalized by palladium mediated cross-coupling to furnish intermediates AH, AI, AJ, and AK, again in high yield (73−97%). Installation of the urea functionality was achieved by a two-step procedure involving the reaction of the aniline intermediates with phenyl chloroformate followed by amine displacment to give compounds 35−38, 40, and 41 in good yield (61−88% over two steps). Conversion of aniline intermediates O, Q, AH, and Z to the thioureas 34, 39, and 42−44 was achieved by initial reaction with di(imidazol-1-yl)methanethione followed by displacement with the appropriate amine in moderate to excellent yield (41−99%). F

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry exemplified by compounds 4 and 5, pAKT(S473) pIC50 of 5.90 and 6.17, respectively, and aqueous solubility 17 and 120 μM, respectively (Figure 3). It is tempting to speculate that the

Table 1. Importance of Urea Hydrogen Bond Donors

Figure 3. Structures of 4 and 5.

addition of this chiral methyl favors a more favorable binding conformation for the molecule while in addition disrupting the efficiency of crystal packing in the solid state by increasing the twist between the morpholine and pyrimidine rings. These trends are observed across a number of morpholine vs 3(S)-methylmorpholine matched pairs within this urea containing scaffold with an average increase in cellular pIC50 of 0.56 and an increase in aqueous solubility of approximately 3-fold (Figure 4). No attempt to characterize the solid state of the compounds prior to assessing the aqueous solubility was made in this analysis.

a

Data quoted are the mean pIC50 of at least three different measurements ± SEM.

of the donor is vital for adopting a favorable urea conformation for binding. A systematic exploration of the urea substituent was undertaken to understand the impact on cellular potency and physicochemical properties (Table 2). Simple alkyl substituents were tolerated (5, 6, and 9) with increasing lipophilicity, resulting in an increase in potency alongside a reduction in aqueous solubility. However, when analyzing LLE or pIC50 − pSolubility, it is clear that there is a slight reduction of compound quality as the size of the alkyl substituent increases. A significant improvement in both cellular potency and LLE is observed upon the introduction of a cyclopropyl substituent, 10, perhaps reflecting an improved interaction with the protein and the reduced lipophilicity of this substituent compared to isopropyl. However, compound quality as measured by pIC50 − pSolubility is reduced. The introduction of polar functionality is tolerated as exemplified by 11 which exhibits good potency and much improved solubility, thus demonstrating improved quality as measured by either LLE or pIC50 − pSolubility. Compound 11 retains the excellent selectivity for binding to the mTOR enzyme over the PI3Kα enzyme that is generally observed within this series (mTOR pIC50 = 8.24, PI3Kα pIC50 = 5.49). The introduction of a basic substituent, such as 12, significantly lowers the log D and improves the aqueous solubility albeit with a concomitant reduction in potency. Analyses of LLE and pIC50 − pSolubility parameters highlight 12 as a compound with improved quality despite the lower absolute potency, and this compound was therefore selected for further investigation. However, more detailed profiling revealed that both mTOR binding affinity and selectivity over PI3Kα are dramatically reduced following the introduction of the basic substituent (mTOR pIC50 = 6.12, PI3Kα pIC50 = 5.10). The reduced selectivity (as calculated from enzyme binding affinities), alongside the unexplained difference in enzyme− cell correlation for this compound, discouraged us from further investigation of basic substituents in this position. The introduction of aryl substituents to the urea is tolerated and results in potent compounds (13−15); however, the increase

Figure 4. Matched pairs analysis highlighting the impact of switching from morpholine to (3S)-3-methylmorpholine on both cell potency and solubility.

The importance of the hydrogen bond donor motif in the para-position of the 2-aryl substituent of the pyrimidine was highlighted during the conception of the urea containing scaffold and is further confirmed by the loss of potency upon methylation of this nitrogen (7 cf. 6) (Table 1). In addition, the second urea hydrogen bond donor also appears to be important with methylation again resulting in a loss of potency (8 cf. 6), suggesting that this nitrogen also forms a constructive interaction with the protein or that the presence G

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry Table 2. Impact of Urea Substituent on Cellular Potency and Physicochemical Propertiesb

a Data quoted are the mean pIC50 of at least three different measurements ± SEM. bThe asterisk (∗) indicates evidence of crystallinity as assessed by polarized light microscopy.

observed for the cyclohexyl substituent, 19. Analyses of the compound quality metrics show that despite improved potency, compound quality is eroded slightly as the size of alkyl group is increased. Polar substituents are tolerated on the sulfone with compound 20 showing a promising balance of potency and solubility, albeit with a similar quality assessment to the simple methyl substituted compound, 6. Aryl and heteroaryl substituents are tolerated, as exemplified by 21 and 22, and result in increased potency; however, the lipophilic nature of these substituents results in only modest improvements in compound quality. The SAR investigations into the urea and sulfone portions of the molecule highlighted the general positive relationship between lipophilicity and cellular potency for the series in addition to the general negative relationship between lipophilicity and aqueous solubility. The challenge to identify compounds with the desired balance of cellular potency and

in lipophilicity combined with the reduction in aqueous solubility results in a reduction in both LLE and pIC50 − pSolubility when compared with simple alkyl substituents. The electronic nature of the substituent on the aryl ring appears to have little impact on either cellular potency or solubility. Heteroaryl substituents are also tolerated with 16 showing a promising balance of potency and solubility and a compound quality similar to 6. Benzyl substituents, such as 17, are not tolerated suggesting that the conformation adopted by such groups is incompatible with efficient binding. Following the assessment of the urea substituent attention was turned to the sulfone moiety and another systematic exploration of the SAR undertaken (Table 3). Small alkyl substituents were tolerated with a general increase in potency and reduction in solubility observed upon increasing bulk, exemplified by 6 and 18; however, there does appear to be a limit to the size of the substituent with a reduction in potency H

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry Table 3. SAR of the Sulfone Substituentb

a Data quoted are the mean pIC50 of at least three different measurements ± SEM. bThe asterisk (∗) indicates evidence of crystallinity as assessed by polarized light microscopy.

that the sulfone oxygens are not essential for mTOR potency (data not shown) although it was anticipated that the removal of these oxygens would result in a significant increase in lipophilicity, thus rendering such a strategy as unattractive at this stage. The low probability of success associated with strategies targeting the disruption of the hydrogen bonding network led us to focus our efforts on disrupting the planarity of the molecules and thus their ability to efficiently stack. The introduction of substituents ortho to the urea did indeed result in improved aqueous solubility, presumably through the disruption of planarity, but a significant reduction in potency was observed, 24 and 25 (Table 4). Further inspection of the small molecule crystal structure revealed the sulfone moiety to be slightly out of plane compared to the pyrimidine ring (Figure 5). This conformation is also present in the proposed binding mode of 2 in a homology model of mTOR (Figure 2b). The apparent ability for the mTOR protein to accommodate this conformation prompted us to explore options to either stabilize this conformation or to increase the degree of nonplanarity in this region of the molecule. To explore this strategy further, compound 26 was prepared in which the methylene group linking the sulfone to the pyrimidine has been dimethylated (Figure 6). Compound 26 shows a significant improvement in cellular potency (pIC50 = 7.62) when compared to compound 6, while reasonable aqueous solubility has been maintained (46 μM) thus making this compound the first from the series to show comparable cell potency/aqueous solubility to AZD8055. The improved compound quality is also reflected in both LLE (5.3) and

aqueous solubility in light of these conflicting relationships is apparent, and it was appreciated that simple variation of these two positions was unlikely to deliver a compound of the required profile. We therefore sought alternative optimization strategies to continue to improve compound quality. The ability of a molecule to form stable lattices in the crystalline state is known to influence the solubility of the compound and thus potentially provides another avenue to modulate solubility in addition to the modulation of lipophilicity. Other groups have reported approaches targeting the ability of molecules to form hydrogen bond networks or efficient π−π staking interactions in the crystal lattice as a strategy to improve aqueous solubility.30 To understand if a similar strategy could be utilized for our series of mTOR inhibitors, a small molecule crystal structure of 15 was obtained (Figure 5). The small molecule crystal structure highlights the extended hydrogen bond network formed between the hydrogen bond donors of the urea and the sulfone oxygens on adjacent molecules. The largely planar conformation of the molecules is also apparent thus resulting in efficient stacking. The importance of the urea hydrogen bond donors for mTOR potency has already been established thus invalidating strategies that involve their removal to disrupt the hydrogen bonding network. A variety of known urea isosteres were investigated with the thiourea, 23, showing similar cellular potency to the urea analogue, 10, but with slightly improved aqueous solubility perhaps reflecting the differing hydrogen bonding strength of thiourea versus urea, Table 4.31 All other urea isosteres attempted displayed a disappointing loss of potency (data not shown). Early SAR studies had established I

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry

Figure 6. Structure of 26.

(pIC50 = 6.29) when assessed in the appropriate enzyme assays. A more systematic investigation into the consequences of alkylation on the methylene linker was performed (Table 5). Again, dimethylation results in a potency improvement and a significant improvement in the pIC50 − pSolubility quality measure (27 cf. 10). A further increase in pIC50 − pSolubility was observed upon the introduction of a cyclopropyl moiety, 28, driven mostly by an increase in aqueous solubility. Cyclobutyl and cyclopentyl groups are tolerated, 29 and 30, respectively, and exhibit excellent levels of cellular potency; however, this is offset by the increased lipophilicity resulting in a reduction in both LLE and pIC50 − pSolubility. The introduction of a tetrahydropyranyl linker, 31, shows promising cellular potency, but despite the reduction in lipophilicity compared to 29 and 30, the aqueous solubility remains low. The increase in compound quality highlighted through these SAR studies, and in particular the increase in pIC50 − pSolubility identified following alkylation of the methylene linker, has resulted in the discovery of a number of compounds possessing cellular potency and aqueous solubility profiles comparable with that of AZD8055 (e.g., 26, 27, and 28). The detailed assessment of AZD8055 in preparation for clinical development highlighted a high turnover in human hepatic incubations. The stability of compounds toward metabolism by human hepatocytes is often used as a key preclinical experiment to predict the likely human pharmacokinetics and therefore likely clinical dose, and thus,

Figure 5. Small molecule crystal structure of 15 (CCDC 1019570) highlighting (a) the packing scheme along the b-axis and (b) hydrogen bonding between the urea and sulfone functionality of neighboring molecules and the largely planar nature of the molecule.

pIC50 − pSolubility (3.3) measures. Compound 26 retains excellent selectivity for mTOR (pIC50 = 9.01) over PI3Kα Table 4. SAR of the Urea Structural Motifb

Data quoted are the mean pIC50 of at least three different measurements ± SEM. bThe asterisk (∗) indicates evidence of crystallinity as assessed by polarized light microscopy. a

J

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry Table 5. Alkylation of the Methylene Spacer

a

Data quoted are the mean pIC50 of at least three different measurements ± SEM.

compounds with high turnover in human hepatocyte incubations are often considered as high risk when trying to identify compounds with suitable properties for clinical dosing. Human hepatocyte incubation studies performed on 26, 27, and 28 highlighted high turnover for each of these compounds (CLint of 36.3, 50.0, and 37.8 (μL/min)/106 cells, respectively). The optimization of stability toward metabolism by human hepatocytes played an important role in the preclinical decision to select AZD2014 (CLint < 4.2 (μL/ min)/106 cells) as a complementary clinical candidate to AZD8055 (CLint = 36.4 (μL/min)/106 cells) but with reduced human pharmacokinetic risk and is discussed elsewhere.26 In order to identify compounds that would have a reduced human pharmacokinetic risk for clinical development, the team used the learning developed within these laboratories during the discovery of AZD2014 and decided to implement an upper limit for human hepatic stability (CLint of 100 μM).36

significant enrichment in compounds successfully meeting the criteria was observed. Approximately 4% of compounds prioritized for synthesis following this exercise met all of the following criteria: cellular pIC50 of >7.5, solubility of >30 μM, human hepatic CLint of 0.67, 0.41 78, 2.3, 0.9, 61 16, 2.0, 1.9, 73 5.6, 43

a

Units for CL, Vss, t1/2, and F are mL/min/kg, L/kg, h, and %, respectively. bUnit for Papp is ×10−6 cm/s. N

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry

J = 8.76 Hz, 2H), 8.72 (s, 1H), 8.89 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C24H26FN5O4S, 500.176 18; found 500.176 23. 1-(4-Methoxyphenyl)-3-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]urea (15). Yield 56%. 1 H NMR (DMSO-d6) δ 1.24 (d, J = 6.79 Hz, 3H), 3.20 (s, 3H), 3.21−3.25 (m, 1H), 3.49 (td, J = 3.06, 11.80, 11.81 Hz, 1H), 3.64 (dd, J = 3.08, 11.48 Hz, 1H), 3.71 (s, 3H), 3.77 (d, J = 11.41 Hz, 1H), 3.98 (dd, J = 3.69, 11.36 Hz, 1H), 4.16 (s, 1H), 4.48 (s, 3H), 6.77 (s, 1H), 6.83−6.91 (m, 2H), 7.31−7.43 (m, 2H), 7.49−7.59 (m, 2H), 8.19−8.3 (m, 2H), 8.49 (s, 1H), 8.81 (s, 1H). 13C NMR (DMSO, d6) 13.25, 38.62, 41.37, 46.35, 55.13, 61.43, 65.97, 70.12, 102.00, 113.96, 117.34, 120.12, 128.47, 130.70, 132.43, 142.18, 152.43, 154.57, 157.32, 161.69, 162.24. HRMS-ESI (m/z): [M + H]+ calcd for C25H29N5O5S, 512.196 22; found, 512.196 04. Compounds 6 and 8 were prepared from intermediate E according to the general procedure B. Compound 24 was prepared from intermediate G according to the general procedure B. General Proceedure B. A solution of the appropriate intermediate (1 equiv) in a mixture of DCM and pyridine (4:1) was added dropwise to a solution of phosgene (20% solution in toluene, 1.2 equiv) in DCM and the reaction stirred for 1 h at ambient temperature. The appropriate amine (1.2 equiv) was added and the reaction stirred for a further 1 h or until the reaction was complete by TLC or LCMS analysis. The reaction was partitioned between ethyl acetate and water, and the organics were dried over MgSO4 and concentrated in vacuo. The crude material was purified by either flash silica chromatography or HPLC. 1-Methyl-3-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]urea (6). Yield 38%. 1 H NMR (DMSO-d6) δ 1.25 (d, J = 6.72 Hz, 3H), 2.67 (d, J = 4.64 Hz, 3H), 3.21 (s, 3H), 3.23 (m, 1H), 3.44−3.6 (m, 2H), 3.66 (dd, J = 2.95, 11.52 Hz, 1H), 3.78 (d, J = 11.47 Hz, 1H), 3.99 (dd, J = 3.47, 11.40 Hz, 1H), 4.18 (d, J = 11.23 Hz, 1H), 4.49 (s, 3H), 6.08 (d, J = 4.66 Hz, 1H), 6.78 (s, 1H), 7.51 (d, J = 8.87 Hz, 2H), 8.21 (d, J = 8.84 Hz, 2H), 8.76 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C19H25N5O4S, 420.170 00; found 420.169 89. 1,1-Dimethyl-3-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]urea (8). Yield 11%. 1 H NMR (DMSO-d6) δ 1.25 (d, J = 6.73 Hz, 3H), 2.96 (s, 6H), 3.21 (s, 3H), 3.28 (s, 1H), 3.51 (td, J = 2.84, 11.76, 11.84 Hz, 1H), 3.66 (dd, J = 2.99, 11.46 Hz, 1H), 3.79 (d, J = 11.47 Hz, 1H), 4.00 (dd, J = 3.41, 11.49 Hz, 1H), 4.18 (s, 1H), 4.49 (s, 3H), 6.79 (s, 1H), 7.60 (d, J = 8.88 Hz, 2H), 8.21 (d, J = 8.84 Hz, 2H), 8.50 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C20H27N5O4S, 434.185 61; found 434.185 65. 3-[2-Methoxy-4-[4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]-1-methylurea (24). Yield 30%. 1H NMR (DMSO-d6) δ 1.23 (d, J = 6.76 Hz, 3H), 2.64 (d, J = 4.58 Hz, 3H), 3.20 (m, 4H), 3.49 (td, J = 3.04, 11.81, 11.84 Hz, 1H), 3.64 (dd, J = 3.06, 11.47 Hz, 1H), 3.76 (d, J = 11.42 Hz, 1H), 3.90 (s, 3H), 3.97 (dd, J = 3.63, 11.36 Hz, 1H), 4.17 (s, 1H), 4.45 (d, J = 15.08 Hz, 1H), 4.48 (s, 2H), 6.76 (s, 1H), 6.83 (d, J = 4.61 Hz, 1H), 7.83−7.93 (m, 2H), 8.08 (s, 1H), 8.21 (d, J = 8.40 Hz, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C20H27N5O5S, 450.180 57; found 450.180 45. Compound 7 was prepared from intermediate I according to the general procedure C. Compounds 10, 11, 12, 16, and 17 were prepared from intermediate E according to the general procedure C. Compound 18 was prepared from intermediate M according to the general procedure C. Compound 25 was prepared from intermediate H according to the general procedure C. Compounds 26−31 were prepared from intermediate O according to the general procedure C. Compounds 32, 33, 36, and 37 were prepared from intermediates R, Z, AI, and AJ, respectively, according to the general procedure C. Compounds 35 and 38 were prepared from intermediate AH according to the general procedure C. Compounds 40 and 41 were prepared from intermediate AK according to the general procedure C. General Proceedure C. Phenyl chloroformate (1 equiv) and sodium carbonate (1.5 equiv) were added to a solution of the

HPLC, and mass spectral techniques and are consistent with the proposed structures; purity was ≥95%. Electrospray mass spectral data were obtained using a Waters ZMD or Waters ZQ LC/mass spectrometer acquiring both positive and negative ion data, and generally, only ions relating to the parent structure are reported. Proton NMR chemical shift values were measured on the δ scale using a Bruker DPX300 spectrometer operating at a field strength of 300 MHz, a Bruker DRX400 operating at 400 MHz, a Bruker DRX500 operating at 500 MHz, or a Bruker AV700 operating at 700 MHz. Unless otherwise stated, NMR spectra were obtained at 400 MHz in deuterated dimethyl sulfoxide. The following abbreviations have been used: s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br, broad; qn, quintet. The following abbreviations may be used: THF = tetrahydrofuran; DIPEA = diisopropylethylamine; DMF = N,N dimethylformamide; DMSO = dimethyl sulfoxide; DMA = N,N-dimethylacetamide; DME = 1,2-dimethoxyethane; DCM = dichloromethane; MeOH = methanol; Compound 4 was prepared from intermediate F according to the general procedure A. Compounds 5, 9, 13, 14, and 15 were prepared from intermediate E according to the general procedure A. General Proceedure A. The appropriate isocyanate (approximately 5 equiv) was added to the appropriate intermediate (1 equiv) in 1,4-dioxane and heated at 70 °C for 4 h or until the reaction was complete by TLC or LCMS analysis. The reaction mixture was concentrated in vacuo and the resultant material purified by either flash silica chromatography or HPLC. 1-Ethyl-3-[4-[4-(methylsulfonylmethyl)-6-morpholin-4-ylpyrimidin-2-yl]phenyl]urea (4). Yield 44%. 1H NMR (DMSO-d6) δ 1.07 (t, J = 7.16, 7.16 Hz, 3H), 3.13 (dd, J = 5.61, 7.15 Hz, 2H), 3.21 (s, 3H), 3.66−3.75 (m, 8H), 4.48 (s, 2H), 6.19 (t, J = 5.55, 5.55 Hz, 1H), 6.81 (s, 1H), 7.50 (d, J = 8.86 Hz, 2H), 8.21 (d, J = 8.80 Hz, 2H), 8.69 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C19H25N5O4S, 420.170 00; found 420.169 92. 1-Ethyl-3-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]urea (5). Yield 94%. 1 H NMR (DMSO-d6) δ 0.98 (t, J = 7.16, 7.16 Hz, 3H), 1.25 (d, J = 6.71 Hz, 3H), 3.00 (qd, J = 5.66, 7.17, 7.20, 7.20 Hz, 2H), 3.08− 3.18 (m, 2H), 3.21 (s, 3H), 3.44−3.56 (m, 1H), 3.66 (d, J = 8.42 Hz, 1H), 3.74−3.82 (m, 1H), 3.99 (d, J = 8.11 Hz, 1H), 4.16 (s, 1H), 4.49 (s, 2H), 6.17 (t, J = 5.55, 5.55 Hz, 1H), 6.78 (s, 1H), 7.50 (d, J = 8.85 Hz, 2H), 8.21 (d, J = 8.83 Hz, 2H), 8.67 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C20H27N5O4S, 434.185 65; found 434.185 61. 3-[4-[4-[(3S)-3-Methylmorpholin-4-yl]-6-(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]-1-propan-2-ylurea (9). Yield 66%. 1H NMR (DMSO-d6) δ 1.10 (d, J = 6.52 Hz, 6H), 1.23 (d, J = 6.78 Hz, 3H), 3.19 (m, 4H), 3.48 (td, J = 3.03, 11.82, 11.84 Hz, 1H), 3.64 (d, J = 3.03 Hz, 1H), 3.71−3.82 (m, 2H), 3.97 (dd, J = 3.64, 11.35 Hz, 1H), 4.15 (s, 1H), 4.46 (s, 3H), 6.05 (d, J = 7.51 Hz, 1H), 6.75 (s, 1H), 7.41−7.52 (d, J = 8.76 Hz, 2H), 8.19 (d, J = 8.76 Hz, 2H), 8.52 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C21H29N5O4S, 448.201 30; found, 448.201 02. 1-[4-[4-[(3S)-3-Methylmorpholin-4-yl]-6-(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]-3-phenylurea (13). Yield 70%. 1H NMR (DMSO-d6) δ 1.24 (d, J = 6.80 Hz, 3H), 3.20 (s, 3H), 3.21 (m, 1H), 3.49 (td, J = 3.06, 11.77, 11.81 Hz, 1H), 3.64 (dd, J = 3.07, 11.47 Hz, 1H), 3.77 (d, J = 11.41 Hz, 1H), 3.98 (dd, J = 3.68, 11.35 Hz, 1H), 4.16 (s, 1H), 4.48 (s, 3H), 6.78 (s, 1H), 6.94−7.03 (m, 1H), 7.21−7.32 (m, 2H), 7.38−7.5 (m, 2H), 7.51−7.62 (m, 2H), 8.2− 8.31 (m, 2H), 8.69 (s, 1H), 8.89 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C24H27N5O4S, 482.185 65; found, 482.185 55. 3-(4-Fluorophenyl)-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]urea (14). Yield 65%. 1 H NMR (DMSO-d6) δ 1.24 (d, J = 6.79 Hz, 3H), 3.20 (s, 3H), 3.23 (dd, J = 3.75, 12.96 Hz, 1H), 3.43−3.55 (m, 1H), 3.64 (dd, J = 3.10, 11.47 Hz, 1H), 3.77 (d, J = 11.41 Hz, 1H), 3.98 (dd, J = 3.73, 11.36 Hz, 1H), 4.16 (s, 1H), 4.48 (s, 3H), 6.78 (s, 1H), 7.12 (t, J = 8.88, 8.88 Hz, 2H), 7.42−7.52 (m, 2H), 7.52−7.61 (m, 2H), 8.25 (d, O

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry

J = 0.63 Hz, 1H), 7.55 (d, J = 8.84 Hz, 2H), 7.76 (s, 1H), 8.18−8.31 (m, 2H), 8.39 (s, 1H), 8.84 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C22H27N7O4S, 486.191 80; found 486.191 80. 1-[4-[4-(Cyclopropylsulfonylmethyl)-6-[(3S)-3-methylmorpholin4-yl]pyrimidin-2-yl]phenyl]-3-methylurea (18). Yield 53%. 1H NMR (DMSO-d6) δ 0.93−1.02 (m, 2H), 1.05 (dd, J = 2.67, 7.98 Hz, 2H), 1.23 (d, J = 6.78 Hz, 3H), 2.65 (d, J = 4.56 Hz, 3H), 2.81−2.9 (m, 1H), 3.13−3.26 (m, 1H), 3.49 (td, J = 3.09, 11.78, 11.82 Hz, 1H), 3.64 (dd, J = 3.10, 11.48 Hz, 1H), 3.76 (d, J = 11.41 Hz, 1H), 3.97 (dd, J = 3.73, 11.38 Hz, 1H), 4.45 (s, 1H), 4.48 (s, 2H), 6.05 (d, J = 4.63 Hz, 1H), 6.75 (s, 1H), 7.48 (d, J = 8.79 Hz, 1H), 8.20 (d, J = 8.81 Hz, 1H), 8.71 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C21H27N5O4S, 446.185 65; found 446.185 58. 3-Methyl-1-[2-methyl-4-[4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]urea (25). Yield 24%. 1 H NMR (DMSO-d6) δ 1.23 (d, J = 6.76 Hz, 3H), 2.24 (s, 3H), 2.66 (d, J = 4.55 Hz, 3H), 3.13−3.25 (m, 4H), 3.45−3.52 (m, 1H), 3.64 (dd, J = 3.08, 11.49 Hz, 1H), 3.76 (d, J = 11.42 Hz, 1H), 3.97 (dd, J = 3.67, 11.37 Hz, 1H), 4.17 (s, 1H), 4.47 (s, 3H), 6.54 (d, J = 4.62 Hz, 1H), 6.75 (s, 1H), 7.77 (s, 1H), 8.00 (m, 1H), 8.05 (m, 1H), 8.07 (m, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C20H27N5O4S, 434.185 65; found 434.185 61. 3-Methyl-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6-(2-methylsulfonylpropan-2-yl)pyrimidin-2-yl]phenyl]urea (26). Yield 70%.1H NMR (DMSO-d6) δ 1.23 (d, J = 6.72 Hz, 3H), 1.77 (d, J = 2.27 Hz, 6H), 2.66 (d, J = 4.61 Hz, 3H), 3.03 (s, 3H), 3.16−3.28 (m, 1H), 3.45−3.55 (m, 1H), 3.57−3.69 (m, 1H), 3.77 (d, J = 11.44 Hz, 1H), 3.98 (dd, J = 3.41, 11.40 Hz, 1H), 4.23 (d, J = 12.74 Hz, 1H), 4.59 (s, 1H), 6.07 (q, J = 4.45, 4.45, 4.48 Hz, 1H), 6.74 (s, 1H), 7.51 (d, J = 8.84 Hz, 2H), 8.23 (d, J = 8.80 Hz, 2H), 8.74 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C21H29N5O4S, 448.201 30; found 448.201 26. 3-Cyclopropyl-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6-(2-methylsulfonylpropan-2-yl)pyrimidin-2-yl]phenyl]urea (27). Yield 68%. 1 H NMR (DMSO-d6) δ 0.40 (dd, J = 2.30, 3.64 Hz, 2H), 0.63 (dd, J = 1.94, 6.85 Hz, 2H), 1.22 (d, J = 6.77 Hz, 3H), 1.76 (d, J = 4.19 Hz, 6H), 2.55 (dd, J = 3.61, 6.88 Hz, 1H), 3.02 (s, 3H), 3.16−3.25 (m, 1H), 3.48 (td, J = 3.00, 11.82, 11.85 Hz, 1H), 3.63 (dd, J = 3.02, 11.47 Hz, 1H), 3.75 (d, J = 11.40 Hz, 1H), 3.96 (dd, J = 3.59, 11.36 Hz, 1H), 4.21 (d, J = 11.41 Hz, 1H), 4.58 (s, 1H), 6.41 (s, 1H), 6.72 (s, 1H), 7.49 (d, J = 8.74 Hz, 2H), 8.22 (d, J = 8.74 Hz, 2H), 8.52 (s, 1H). 13C NMR (DMSO-d6) δ 6.30, 13.19, 20.43, 22.34, 36.41, 38.70, 46.23, 66.02, 66.39, 70.21, 99.08, 117.12, 128.38, 130.43, 142.61, 155.67, 161.48, 161.99, 164.51. HRMS-ESI (m/z): [M + H]+ calcd for C23H31N5O4S, 474.216 95; found 474.216 89. 3-Cyclopropyl-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6-(1methylsulfonylcyclopropyl)pyrimidin-2-yl]phenyl]urea (28). Yield 75%. 1H NMR (DMSO-d6) δ 0.40 (dd, J = 2.15, 3.76 Hz, 2H), 0.63 (dd, J = 1.98, 6.86 Hz, 2H), 1.22 (d, J = 6.77 Hz, 3H), 1.54 (d, J = 2.31 Hz, 2H), 1.6−1.7 (m, 2H), 2.55 (dd, J = 3.64, 6.88 Hz, 1H), 3.14−3.24 (m, 1H), 3.32 (s, 3H), 3.4−3.52 (m, 1H), 3.62 (dd, J = 3.08, 11.48 Hz, 1H), 3.74 (d, J = 11.42 Hz, 1H), 3.95 (dd, J = 3.67, 11.38 Hz, 1H), 4.19 (s, 1H), 4.55 (s, 1H), 6.41 (d, J = 2.54 Hz, 1H), 6.75 (s, 1H), 7.49 (d, J = 8.76 Hz, 2H), 8.18 (d, J = 8.76 Hz, 2H), 8.53 (s, 1H). 13C NMR (DMSO-d6) 6.32, 12.10, 13.27, 22.34, 38.65, 39.99, 40.45, 46.02, 66.01, 70.18, 100.43, 117.12, 128.39, 130.33, 142.63, 155.68, 161.55, 161.80, 162.11. HRMS-ESI (m/z): [M + H]+ calcd for C23H29N5O4S, 472.201 30; found 472.201 20. 3-Cyclopropyl-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6-(1methylsulfonylcyclobutyl)pyrimidin-2-yl]phenyl]urea (29). Yield 50%. 1H NMR (DMSO-d6) δ 0.40 (dd, J = 2.00, 3.84 Hz, 2H), 0.63 (dd, J = 1.98, 6.89 Hz, 2H), 1.22 (d, J = 6.77 Hz, 3H), 1.83− 1.96 (m, 1H), 1.98−2.13 (m, 1H), 2.54 (dt, J = 3.42, 3.42, 6.93 Hz, 1H), 2.71−2.84 (m, 2H), 2.85 (s, 3H), 2.86−2.95 (m, 2H), 3.20 (td, J = 3.88, 12.92, 13.04 Hz, 1H), 3.48 (td, J = 3.07, 11.82, 11.83 Hz, 1H), 3.63 (dd, J = 3.09, 11.49 Hz, 1H), 3.75 (d, J = 11.42 Hz, 1H), 3.96 (dd, J = 3.70, 11.37 Hz, 1H), 4.22 (s, 1H), 4.55 (s, 1H), 6.41 (d, J = 2.64 Hz, 1H), 6.69 (s, 1H), 7.49 (d, J = 8.77 Hz, 2H), 8.20 (d, J = 8.77 Hz, 2H), 8.53 (s, 1H). 13C NMR (DMSO-d6) 6.31, 13.23, 15.16, 22.34, 27.30, 27.35, 35.36, 38.70, 46.28, 66.03, 68.60,

appropriate intermediate (1 equiv) in 1,4-dioxane, and the mixture was stirred at ambient temperature for 2 h or until the reaction was complete by TLC or LCMS analysis. The reaction mixture was partitioned between ethyl acetate and water, and the organics were dried over MgSO4 and concentrated in vacuo. The crude phenyl carbamate was used either directly in the subsequent step or following purification by flash silica chromatography. A solution of the phenyl carbamate (1 equiv), the appropriate amine (2−5 equiv), and triethylamine (3.25 equiv) in either NMP or DMF was heated between 50 and 70 °C for 2 h or until complete by TLC or LCMS analysis. The reaction mixture was purified by either flash column chromatography or HPLC. 1,3-Dimethyl-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]urea (7). Yield 4%. 1H NMR (CDCl3) δ 1.37 (d, J = 6.82 Hz, 3H), 2.76 (d, J = 4.61 Hz, 3H), 3.08 (s, 3H), 3.32 (s, 3H), 3.37 (dd, J = 3.89, 13.00 Hz, 1H), 3.60 (td, J = 3.04, 11.83, 11.96 Hz, 1H), 3.75 (dd, J = 3.09, 11.52 Hz, 1H), 3.84 (d, J = 11.53 Hz, 1H), 4.06 (dd, J = 3.75, 11.57 Hz, 1H), 4.16 (d, J = 12.33 Hz, 1H), 4.27 (s, 2H), 4.36 (d, J = 4.52 Hz, 1H), 4.50 (s, 1H), 6.52 (s, 1H), 7.32 (d, J = 8.56 Hz, 2H), 8.39 (d, J = 8.53 Hz, 2H). Mass calculated for C20H27N5O4S 433.53, found 434 (ESI, M + H). 3-Cyclopropyl-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]urea (10). Yield 39%. 1 H NMR (DMSO,d6) δ 0.41 (dd, J = 2.09, 3.75 Hz, 2H), 0.63 (dd, J = 1.98, 6.85 Hz, 2H), 1.23 (d, J = 6.77 Hz, 3H), 2.55 (dd, J = 3.67, 6.86 Hz, 1H), 3.19 (s, 3H), 3.22 (dd, J = 3.72, 12.99 Hz, 1H), 3.48 (td, J = 3.06, 11.78, 11.82 Hz, 1H), 3.64 (dd, J = 3.09, 11.49 Hz, 1H), 3.76 (d, J = 11.43 Hz, 1H), 3.97 (dd, J = 3.72, 11.38 Hz, 1H), 4.16 (s, 1H), 4.47 (m, 3H), 6.42 (d, J = 2.56 Hz, 1H), 6.76 (s, 1H), 7.49 (d, J = 8.79 Hz, 2H), 8.20 (d, J = 8.78 Hz, 2H), 8.52 (s, 1H). 13 C NMR (DMSO-d6) 6.37, 13.30, 22.40, 38.65, 41.43, 46.40, 61.50, 66.04, 70.19, 101.98, 117.14, 128.43, 130.30, 142.70, 155.73, 157.35, 161.75, 162.37. HRMS-ESI (m/z): [M + H] + calcd for C21H27N5O4S, 446.185 65; found 446.185 58. 3-(2-Hydroxyethyl)-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]urea (11). Yield 22%. 1 H NMR (DMSO-d6) δ 1.24 (d, J = 6.74 Hz, 3H), 3.13−3.24 (m, 5H), 3.48 (dd, J = 3.87, 15.61 Hz, 3H), 3.65 (dd, J = 2.92, 11.55 Hz, 1H), 3.78 (d, J = 11.28 Hz, 1H), 3.91−4.03 (m, 1H), 4.16 (s, 1H), 4.48 (s, 3H), 4.73 (s, 1H), 6.27 (t, J = 5.62, 5.62 Hz, 1H), 6.77 (s, 1H), 7.49 (d, J = 8.87 Hz, 2H), 8.21 (d, J = 8.81 Hz, 2H), 8.82 (s, 1H). Mass calculated for C20H27N5O5S 449.53, found 449.97 (ESI, M + H). 3-(2-Dimethylaminoethyl)-1-[4-[4-[(3S)-3-methylmorpholin-4yl]-6-(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]urea (12). Yield 44%. 1H NMR (DMSO-d6) δ 1.23 (d, J = 6.76 Hz, 3H), 2.17 (s, 6H), 2.32 (t, J = 6.19, 6.19 Hz, 2H), 3.14−3.23 (m, 6H), 3.48 (td, J = 3.02, 11.87, 11.89 Hz, 1H), 3.63 (dd, J = 3.01, 11.47 Hz, 1H), 3.76 (d, J = 11.41 Hz, 1H), 3.97 (dd, J = 3.56, 11.38 Hz, 1H), 4.15 (s, 1H), 4.46 (s, 2H), 6.15 (d, J = 5.27 Hz, 1H), 6.75 (s, 1H), 7.44− 7.52 (m, J = 8.76 Hz, 2H), 8.19 (d, J = 8.76 Hz, 2H), 8.88 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C22H32N6O4S, 477.227 85; found 477.227 91. 3-[(4-Methoxyphenyl)methyl]-1-[4-[4-[(3S)-3-methylmorpholin4-yl]-6-(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]urea (17). Yield 71%. 1H NMR (DMSO-d6) δ 1.23 (d, J = 6.77 Hz, 3H), 3.19 (s, 3H), 3.21 (dd, J = 3.66, 12.96 Hz, 1H), 3.44−3.53 (m, 1H), 3.63 (dd, J = 3.09, 11.50 Hz, 1H), 3.72 (s, 3H), 3.76 (d, J = 11.43 Hz, 1H), 3.97 (dd, J = 3.72, 11.37 Hz, 1H), 4.15 (s, 1H), 4.23 (d, J = 5.83 Hz, 2H), 4.46 (m, 3H), 6.58 (s, 1H), 6.75 (s, 1H), 6.89 (d, J = 8.67 Hz, 2H), 7.23 (d, J = 8.68 Hz, 2H), 7.49 (d, J = 8.84 Hz, 2H), 8.20 (d, J = 8.79 Hz, 2H), 8.74 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C26H31N5O5S, 526.211 87; found 526.212 28. 1-[4-[4-[(3S)-3-Methylmorpholin-4-yl]-6-(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]-3-(1-methylpyrazol-4-yl)urea (16). Yield 30%. 1H NMR (DMSO-d6) δ 1.25 (d, J = 6.73 Hz, 3H), 3.21 (s, 3H), 3.27 (s, 1H), 3.51 (td, J = 2.93, 11.78, 11.85 Hz, 1H), 3.66 (dd, J = 2.92, 11.51 Hz, 1H), 3.79 (m, 4H), 3.99 (dd, J = 3.38, 11.32 Hz, 1H), 4.18 (d, J = 12.17 Hz, 1H), 4.49 (s, 3H), 6.79 (s, 1H), 7.38 (d, P

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry

(d, J = 11.49 Hz, 1H), 3.96 (d, J = 8.03 Hz, 1H), 4.02−4.19 (m, 3H), 4.73 (t, J = 4.98, 4.98 Hz, 1H), 6.22 (t, J = 5.51, 5.51 Hz, 1H), 6.64 (s, 1H), 7.3−7.51 (m, 4H), 7.69−7.95 (m, 4H), 8.78 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C27H30FN5O5S, 556.202 44; found 556.202 15. 3-Methyl-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6-(1-pyridin-4ylsulfonylcyclopropyl)pyrimidin-2-yl]phenyl]urea (37). Yield 62%. 1 H NMR (DMSO-d6) δ 1.17 (d, J = 6.78 Hz, 3H), 1.65−1.7 (m, 2H), 1.95 (q, J = 4.05, 4.21, 4.21 Hz, 2H), 2.64 (d, J = 4.68 Hz, 3H), 3.11−3.19 (m, 1H), 3.44 (td, J = 3.06, 11.83, 11.85 Hz, 1H), 3.59 (dd, J = 3.10, 11.48 Hz, 1H), 3.73 (d, J = 11.42 Hz, 1H), 3.94 (dd, J = 3.72, 11.39 Hz, 1H), 4.14 (s, 1H), 4.44 (s, 1H), 6.03 (d, J = 4.66 Hz, 1H), 6.65 (s, 1H), 7.35 (d, J = 8.79 Hz, 2H), 7.64 (d, J = 8.76 Hz, 2H), 7.7−7.83 (m, 2H), 8.69 (s, 1H), 8.8−8.92 (m, 2H). HRMS-ESI (m/z): [M + H]+ calcd for C25H28N6O4S, 509.196 55; found 509.196 32. 1-[4-[4-(1-Cyclopropylsulfonylcyclopropyl)-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-2-yl]phenyl]-3-(2-hydroxyethyl)urea (38). Yield 73%. 1H NMR (DMSO-d6) δ 0.88−0.95 (m, 2H), 1.02 (m, 3H), 1.22 (d, J = 6.78 Hz, 2H), 1.51−1.6 (m, 2H), 1.6−1.69 (m, 2H), 2.93−3.01 (m, 1H), 3.13−3.19 (m, 2H), 3.19 (d, J = 3.59 Hz, 1H), 3.45 (q, J = 5.51, 5.53, 5.53 Hz, 2H), 3.47 (d, J = 3.10 Hz, 1H), 3.62 (dd, J = 3.02, 11.48 Hz, 1H), 3.75 (d, J = 11.41 Hz, 1H), 3.96 (dd, J = 3.65, 11.37 Hz, 1H), 4.16 (s, 1H), 4.51 (s, 1H), 4.72 (t, J = 5.09, 5.09 Hz, 1H), 6.23 (s, 1H), 6.82 (s, 1H), 7.47 (d, J = 8.76 Hz, 2H), 8.19 (d, J = 8.77 Hz, 2H), 8.78 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C24H31N5O5S, 502.211 87; found 502.211 98. 1-[4-[4-[1-(Benzenesulfonyl)cyclopropyl]-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-2-yl]phenyl]-3-(2-hydroxyethyl)urea (40). Yield 88%. 1H NMR (DMSO-d6) δ 1.16 (d, J = 6.77 Hz, 3H), 1.57−1.68 (m, 2H), 1.88 (d, J = 2.73 Hz, 2H), 3.15 (dd, J = 7.11, 12.08 Hz, 3H), 3.42−3.47 (m, 4H), 3.59 (dd, J = 3.05, 11.51 Hz, 1H), 3.72 (d, J = 11.43 Hz, 1H), 3.94 (dd, J = 3.69, 11.39 Hz, 1H), 4.09 (s, 1H), 4.36 (s, 1H), 6.23 (s, 1H), 6.60 (s, 1H), 7.37 (d, J = 8.69 Hz, 2H), 7.58 (t, J = 7.84, 7.84 Hz, 2H), 7.70 (d, J = 7.45 Hz, 1H), 7.75−7.8 (m, 2H), 7.83 (d, J = 8.73 Hz, 2H), 8.77 (s, 1H). 13C NMR (DMSO-d6) 13.14, 13.28, 21.29, 34.42, 37.38, 38.62, 41.74, 46.39, 47.10, 60.24, 65.95, 70.07, 101.48, 116.54, 128.43, 128.54, 129.07, 133.49, 139.04, 142.88, 154.93, 161.30, 161.71. HRMS-ESI (m/z): [M + H]+ calcd for C27H31N5O5S, 538.211 87; found, 538.211 73. 1-[4-[4-[1-(Benzenesulfonyl)cyclopropyl]-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-2-yl]phenyl]-3-(3-hydroxypropyl)urea (41). Yield 61%. 1H NMR (DMSO-d6) δ 1.15 (d, J = 6.77 Hz, 3H), 1.5−1.7 (m, 4H), 1.88 (m, 2H), 3.04−3.21 (m, 3H), 3.38−3.52 (m, 3H), 3.59 (dd, J = 3.07, 11.49 Hz, 1H), 3.72 (d, J = 11.41 Hz, 1H), 3.93 (dd, J = 3.69, 11.38 Hz, 1H), 4.08 (s, 1H), 4.36 (s, 1H), 4.45 (t, J = 5.13, 5.13 Hz, 1H), 6.16 (s, 1H), 6.60 (s, 1H), 7.3−7.41 (m, 2H), 7.57 (t, J = 7.84, 7.84 Hz, 2H), 7.66−7.73 (m, 1H), 7.73−7.88 (m, 4H), 8.65 (s, 1H). 13C NMR (DMSO-d6) 13.08, 13.20, 17.15, 28.92, 30.03, 32.75, 36.40, 38.54, 46.27, 47.27, 48.42, 58.51, 65.97, 70.11, 101.27, 116.55, 128.41, 129.04, 129.88, 133.42, 139.20, 142.76, 154.93, 160.50, 161.40, 161.92. HRMS-ESI (m/z): [M + H]+ calcd for C28H33N5O5S, 552.227 52; found 552.227 36. Compounds 19, 21, and 22 were prepared from intermediate J according to the general procedure D. Compound 20 was prepared from intermediate K according to the general procedure D. General Proceedure D for Preparing Compounds. Intermediate J or K (1 equiv) was treated with methanesulfonyl chloride (1.5 equiv) and triethylamine (1.5 equiv) in DCM at 0 °C, then allowed to come to ambient temperature and stirred until the reaction was complete by TLC or LCMS. The reaction mixture was concentrated in vacuo to yield the crude mesylate which was used without further purification. DBU (2 equiv) was added to a solution of the appropriate thiol (1.75 equiv) and the crude mesylate (1 equiv) in acetonitrile and the reaction stirred at ambient temperature until complete by TLC or LCMS analysis. The reaction mixture was concentrated in vacuo, and the crude thioether was used in the next step either directly or following purification by HPLC. 3-

70.19, 99.90, 117.10, 128.38, 130.42, 142.62, 155.68, 161.73, 161.87, 164.38. HRMS-ESI (m/z): [M + H]+ calcd for C24H31N5O4S, 486.216 95; found 486.216 89. 3-Cyclopropyl-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6-(1methylsulfonylcyclopentyl)pyrimidin-2-yl]phenyl]urea (30). Yield 39%. 1H NMR (DMSO-d6) δ 0.40 (dd, J = 2.20, 3.75 Hz, 2H), 0.63 (dd, J = 1.97, 6.87 Hz, 2H), 1.21 (d, J = 6.77 Hz, 3H), 1.56 (d, J = 7.00 Hz, 2H), 1.80 (dd, J = 3.02, 5.79 Hz, 2H), 2.36−2.46 (m, 2H), 2.55 (m, 1H), 2.67−2.72 (m, 1H), 2.75 (dt, J = 6.59, 6.59, 12.98 Hz, 1H), 2.88 (s, 3H), 3.19 (td, J = 3.86, 12.95, 13.04 Hz, 1H), 3.48 (td, J = 3.01, 11.84, 11.85 Hz, 1H), 3.63 (dd, J = 3.03, 11.48 Hz, 1H), 3.75 (d, J = 11.41 Hz, 1H), 3.96 (dd, J = 3.68, 11.36 Hz, 1H), 4.23 (d, J = 11.23 Hz, 1H), 4.54 (s, 1H), 6.42 (d, J = 2.60 Hz, 1H), 6.77 (s, 1H), 7.49 (d, J = 8.75 Hz, 2H), 8.22 (d, J = 8.76 Hz, 2H), 8.54 (s, 1H). 13C NMR (DMSO-d6) 6.31, 13.19, 22.34, 24.56, 31.67, 31.82, 37.04, 46.31, 66.03, 70.19, 77.21, 100.44, 117.11, 128.38, 130.45, 142.62, 155.68, 161.56, 161.96, 163.97. HRMS-ESI (m/z): [M + H]+ calcd for C25H33N5O4S, 500.232 60; found 500.232 76. 3-Cyclopropyl-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6-(4-methylsulfonyloxan-4-yl)pyrimidin-2-yl]phenyl]urea (31). Yield 72%. 1H NMR (CDCl3-d6) δ 0.71 (dd, J = 2.24, 3.51 Hz, 2H), 0.89 (dd, J = 1.95, 6.70 Hz, 2H), 1.35 (d, J = 6.79 Hz, 3H), 2.46−2.59 (m, 3H), 2.64 (dd, J = 2.37, 3.55 Hz, 1H), 2.71 (m, 4H), 3.28−3.5 (m, 3H), 3.56−3.67 (m, 1H), 3.73−3.88 (m, 2H), 3.97−4.13 (m, 3H), 4.20 (d, J = 12.79 Hz, 1H), 4.45 (s, 1H), 4.89 (s, 1H), 6.64 (s, 1H), 7.00 (s, 1H), 7.51 (d, J = 8.81 Hz, 2H), 8.31 (d, J = 8.79 Hz, 2H). HRMS-ESI (m/z): [M + H]+ calcd for C25H33N5O5S, 516.227 52; found 516.227 60. 3-(2-Hydroxyethyl)-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6-(1methylsulfonylcyclobutyl)pyrimidin-2-yl]phenyl]urea (32). Yield 41%. 1H NMR (DMSO-d6) δ 1.22 (d, J = 6.77 Hz, 3H), 1.85− 1.93 (m, 1H), 2.01−2.1 (m, 1H), 2.74−2.83 (m, 2H), 2.85 (s, 3H), 2.89 (ddd, J = 5.05, 9.59, 16.90 Hz, 2H), 3.16 (d, J = 5.70 Hz, 1H), 3.17−3.23 (m, 1H), 3.45 (q, J = 5.50, 5.55, 5.55 Hz, 2H), 3.46−3.51 (m, 1H), 3.63 (dd, J = 3.09, 11.47 Hz, 1H), 3.75 (d, J = 11.41 Hz, 1H), 3.96 (dd, J = 3.67, 11.36 Hz, 1H), 4.22 (s, 1H), 4.55 (s, 1H), 4.71 (t, J = 5.13, 5.13 Hz, 1H), 6.23 (s, 1H), 6.68 (s, 1H), 7.47 (d, J = 8.77 Hz, 1H), 8.20 (d, J = 8.81 Hz, 1H), 8.79 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C23H31N5O5S, 490.211 87; found 490.211 88. 3-Cyclopropyl-1-[4-[4-[1-(3-hydroxypropylsulfonyl)cyclopropyl]6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-2-yl]phenyl]urea (33). Yield 59%. 1H NMR (DMSO-d6) δ 0.41 (dd, J = 2.14, 3.74 Hz, 2H), 0.63 (dd, J = 1.96, 6.86 Hz, 2H), 1.22 (d, J = 6.76 Hz, 3H), 1.5−1.58 (m, 2H), 1.59−1.68 (m, 2H), 1.86−1.97 (m, 2H), 2.55 (dq, J = 3.24, 3.25, 3.25, 6.94 Hz, 1H), 3.13−3.26 (m, 1H), 3.43− 3.55 (m, 5H), 3.62 (dd, J = 3.09, 11.49 Hz, 1H), 3.74 (d, J = 11.42 Hz, 1H), 3.95 (dd, J = 3.65, 11.37 Hz, 1H), 4.19 (s, 1H), 4.53 (s, 1H), 4.67 (t, J = 5.28, 5.28 Hz, 1H), 6.41 (d, J = 2.43 Hz, 1H), 6.75 (s, 1H), 7.49 (d, J = 8.76 Hz, 2H), 8.15−8.23 (m, J = 8.76 Hz, 2H), 8.52 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C25H33N5O5S, 516.227 52; found 516.227 54. 3-Cyclopropyl-1-[4-[4-(1-cyclopropylsulfonylcyclopropyl)-6-[(3S)3-methylmorpholin-4-yl]pyrimidin-2-yl]phenyl]urea (35). Yield 74%. 1H NMR (DMSO-d6) δ 0.33−0.47 (m, 2H), 0.63 (td, J = 4.85, 6.74, 6.82 Hz, 2H), 0.85−0.97 (m, 2H), 1.01 (dd, J = 2.13, 8.02 Hz, 2H), 1.22 (d, J = 6.77 Hz, 3H), 1.48−1.61 (m, 2H), 1.61− 1.71 (m, 2H), 2.55 (dd, J = 3.66, 6.89 Hz, 1H), 2.98 (ddd, J = 3.21, 4.66, 8.01 Hz, 1H), 3.12−3.25 (m, 1H), 3.47 (td, J = 3.06, 11.80, 11.83 Hz, 1H), 3.62 (dd, J = 3.08, 11.47 Hz, 1H), 3.75 (d, J = 11.41 Hz, 1H), 3.96 (dd, J = 3.67, 11.38 Hz, 1H), 4.17 (s, 1H), 4.51 (s, 1H), 6.40 (d, J = 2.53 Hz, 1H), 6.83 (s, 1H), 7.48 (d, J = 8.76 Hz, 2H), 8.20 (d, J = 8.76 Hz, 2H), 8.51 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C25H31N5O4S, 498.216 95; found, 498.217 10. 1-[4-[4-[1-(4-Fluorophenyl)sulfonylcyclopropyl]-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-2-yl]phenyl]-3-(2-hydroxyethyl)urea (36). Yield 62%. 1H NMR (DMSO-d6) δ 1.18 (d, J = 6.71 Hz, 3H), 1.61 (d, J = 4.80 Hz, 2H), 1.82−1.98 (m, 2H), 3.17 (dd, J = 2.27, 5.40 Hz, 2H), 3.42−3.5 (m, 2H), 3.61 (dd, J = 2.84, 11.48 Hz, 2H), 3.74 Q

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry

Hz, 1H), 4.51 (s, 2H), 6.83 (s, 1H), 7.63 (d, J = 8.72 Hz, 2H), 8.22−8.3 (m, 2H), 9.52 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C21H27N5O3S2, 462.162 81; found 462.162 63. 3-(2-Hydroxyethyl)-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6-(2methylsulfonylpropan-2-yl)pyrimidin-2-yl]phenyl]thiourea (34). Yield 41%. 1H NMR (DMSO-d6) δ 1.24 (d, J = 6.72 Hz, 3H), 1.78 (d, J = 2.25 Hz, 6H), 3.04 (s, 3H), 3.18−3.24 (m, 1H), 3.50 (d, J = 2.85 Hz, 1H), 3.57 (m, 4H), 3.65 (dd, J = 2.93, 11.47 Hz, 1H), 3.77 (d, J = 11.45 Hz, 1H), 3.98 (dd, J = 3.36, 11.25 Hz, 1H), 4.24 (d, J = 13.50 Hz, 1H), 4.63 (s, 1H), 4.81 (s, 1H), 6.78 (s, 1H), 7.63 (d, J = 8.75 Hz, 2H), 7.87 (s, 1H), 8.30 (d, J = 8.74 Hz, 2H), 9.81 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C22H31N5O4S2, 494.189 02; found 494.188 87. 3-(2-Hydroxyethyl)-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6-(1methylsulfonylcyclopropyl)pyrimidin-2-yl]phenyl]thiourea (39). Yield 81%.1H NMR (DMSO-d6) δ 1.23 (d, J = 6.77 Hz, 3H), 1.55 (d, J = 2.29 Hz, 2H), 1.66 (t, J = 3.33, 3.33 Hz, 2H), 3.20 (td, J = 3.86, 12.92, 13.04 Hz, 1H), 3.29 (s. 3H), 3.47 (td, J = 3.07, 11.79, 11.82 Hz, 1H), 3.52−3.6 (m, 4H), 3.62 (dd, J = 3.09, 11.49 Hz, 1H), 3.75 (d, J = 11.43 Hz, 1H), 3.96 (dd, J = 3.67, 11.37 Hz, 1H), 4.20 (s, 1H), 4.57 (s, 1H), 4.79 (s, 1H), 6.79 (s, 1H), 7.62 (d, J = 8.70 Hz, 2H), 7.85 (s, 1H), 8.24 (d, J = 8.70 Hz, 2H), 9.79 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C22H29N5O4S2, 492.173 37; found 492.173 31. 1-[4-[4-(1-Cyclopropylsulfonylcyclopropyl)-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-2-yl]phenyl]-3-(2-hydroxyethyl)thiourea (42 (AZD3147)). Yield 77%. 1H NMR (DMSO-d6) δ 0.92 (dd, J = 1.63, 3.29 Hz, 2H), 1.02 (dd, J = 2.16, 8.04 Hz, 2H), 1.23 (d, J = 6.77 Hz, 3H), 1.57 (dd, J = 1.12, 6.00 Hz, 2H), 1.64 (dd, J = 2.73, 6.38 Hz, 2H), 2.92−3.01 (m, 1H), 3.21 (td, J = 3.89, 12.88, 13.00 Hz, 1H), 3.48 (td, J = 3.07, 11.83, 11.83 Hz, 1H), 3.51−3.59 (m, 4H), 3.63 (dd, J = 3.09, 11.50 Hz, 1H), 3.75 (d, J = 11.43 Hz, 1H), 3.96 (dd, J = 3.70, 11.38 Hz, 1H), 4.17 (s, 1H), 4.54 (s, 1H), 4.79 (s, 1H), 6.87 (s, 1H), 7.61 (d, J = 8.71 Hz, 2H), 7.83 (s, 1H), 8.26 (d, J = 8.68 Hz, 2H), 9.78 (s, 1H). 13C NMR (DMSO-d6) 4.44, 4.51, 12.13, 12.15, 29.29, 38.70, 45.96, 46.40, 59.10, 65.99, 70.17, 101.44, 121.53, 128.12, 132.84, 141.60, 161.84, 161.65, 161.51, 180.24. HRMS-ESI (m/z): [M + H]+ calcd for C24H31N5O4S2, 518.189 02; found 518.188 96. 3-Cyclopropyl-1-[4-[4-[1-(3-hydroxypropylsulfonyl)cyclopropyl]6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-2-yl]phenyl]thiourea (43). Yield 98%. 1H NMR (DMSO-d6) δ 0.59 (s, 2H), 0.75 (d, J = 5.36 Hz, 2H), 1.23 (d, J = 6.79 Hz, 3H), 1.53−1.59 (m, 2H), 1.6− 1.67 (m, 2H), 1.88−1.97 (m, 2H), 2.91 (s, 1H), 3.16−3.25 (m, 1H), 3.49 (m, 5H), 3.62 (dd, J = 3.07, 11.49 Hz, 1H), 3.75 (d, J = 11.43 Hz, 1H), 3.96 (dd, J = 3.65, 11.37 Hz, 1H), 4.20 (s, 1H), 4.56 (s, 1H), 4.67 (t, J = 5.26, 5.26 Hz, 1H), 6.80 (s, 1H), 7.60 (d, J = 8.62 Hz, 2H), 8.13 (s, 1H), 8.25 (d, J = 8.66 Hz, 2H), 9.48 (s, 1H). 13C NMR (DMSO-d6) 6.77, 11.63, 11.65, 13.29, 25.03, 38.69, 45.00, 46.27, 49.60, 59.03, 65.99, 70.16, 101.12, 127.85, 132.96, 141.90, 161.52, 161.79, 161.96, 181.44. HRMS-ESI (m/z): [M + H]+ calcd for C25H33N5O4S2, 532.204 67; found 532.204 77. 3-(2-Hydroxyethyl)-1-[4-[4-[1-(3-hydroxypropylsulfonyl)cyclopropyl]-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-2-yl]phenyl]thiourea (44). Yield >99%. 1H NMR (DMSO-d6) δ 1.22 (d, J = 6.78 Hz, 3H), 1.55 (d, J = 5.06 Hz, 2H), 1.58−1.69 (m, 2H), 1.85−1.98 (m, 2H), 3.20 (td, J = 3.82, 12.92, 13.03 Hz, 1H), 3.45− 3.52 (m, 5H), 3.56 (s, 4H), 3.62 (dd, J = 3.01, 11.49 Hz, 1H), 3.75 (d, J = 11.42 Hz, 1H), 3.96 (dd, J = 3.58, 11.35 Hz, 1H), 4.19 (s, 1H), 4.56 (s, 1H), 4.67 (t, J = 5.25, 5.25 Hz, 1H), 4.79 (s, 1H), 6.80 (s, 1H), 7.61 (d, J = 8.66 Hz, 2H), 7.84 (s, 1H), 8.25 (d, J = 8.62 Hz, 2H), 9.79 (s, 1H). 13C NMR (DMSO-d6) 11.65, 13.30, 25.03, 45.00, 46.27, 46.40, 49.58, 59.02, 65.99, 70.16, 101.12, 121.55, 128.13, 132.73, 141.68, 161.52, 161.78, 161.93, 180.26. HRMS-ESI (m/z): [M + H]+ calcd for C24H33N5O5S2, 536.199 59; found 536.199 58. The prepartion of the intermediates used in the above procedures are described below. Methyl 2-Chloro-6-[(3S)-3-methylmorpholin-4-yl]pyrimidine-4carboxylate (Intermediate A). A solution of (3S)-3-methylmorpholine (2.49 g) and triethylamine (3.70 mL) in DCM (10 mL) was

Chloroperbenzoic acid (75%, 2 equiv) and sodium permanganate (2.5 equiv) were added to a solution of the thioether (1 equiv) in a mixture of 1,4-dioxane and water. The reaction was stirred at ambient temperature for up to 18 h with additional 3chloroperbenzoic acid and sodium permanganate added if required to push the reaction to completion by TLC or LCMS analysis. The reaction mixture was purified by ion exchange cartridge (SCX) with the desired material eluted with 7 N ammonia in methanol. The crude material was further purified by HPLC. 1-[4-[4-(Cyclohexylsulfonylmethyl)-6-[(3S)-3-methylmorpholin-4yl]pyrimidin-2-yl]phenyl]-3-methylurea (19). Yield 43%. 1H NMR (DMSO-d6) δ 1.23 (d, J = 6.74 Hz, 5H), 1.43 (d, J = 12.48 Hz, 2H), 1.66 (d, J = 12.46 Hz, 1H), 1.87 (d, J = 12.98 Hz, 2H), 2.23 (s, 2H), 2.65 (d, J = 4.61 Hz, 3H), 3.15−3.24 (m, 1H), 3.34 (d, J = 8.68 Hz, 1H), 3.48 (td, J = 2.95, 11.84, 11.84 Hz, 1H), 3.63 (dd, J = 2.98, 11.50 Hz, 1H), 3.76 (d, J = 11.42 Hz, 1H), 3.97 (dd, J = 3.54, 11.35 Hz, 1H), 4.15 (s, 1H), 4.41 (s, 1H), 6.07 (d, J = 4.65 Hz, 1H), 6.75 (s, 1H), 7.49 (d, J = 8.76 Hz, 2H), 8.18 (d, J = 8.75 Hz, 2H), 8.73 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C24H33N5O4S, 488.232 60; found, 488.232 51. 3-Cyclopropyl-1-[4-[4-(3-hydroxypropylsulfonylmethyl)-6-[(3S)-3methylmorpholin-4-yl]pyrimidin-2-yl]phenyl]urea (20). Yield 17%. 1 H NMR (DMSO-d6) δ 0.42 (dd, J = 2.13, 3.74 Hz, 2H), 0.65 (dd, J = 2.02, 6.86 Hz, 2H), 1.24 (d, J = 6.72 Hz, 3H), 1.96 (dd, J = 5.74, 10.22 Hz, 2H), 2.53−2.59 (m, 1H), 3.22 (d, J = 3.47 Hz, 1H), 3.34−3.46 (m, 2H), 3.45−3.61 (m, 3H), 3.65 (dd, J = 2.94, 11.47 Hz, 1H), 3.78 (d, J = 11.48 Hz, 1H), 3.99 (dd, J = 3.37, 11.39 Hz, 1H), 4.16 (s, 1H), 4.46 (s, 3H), 4.72 (t, J = 5.23, 5.23 Hz, 1H), 6.43 (d, J = 2.76 Hz, 1H), 6.78 (s, 1H), 7.50 (d, J = 8.84 Hz, 2H), 8.21 (d, J = 8.82 Hz, 2H), 8.53 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C23H31N5O5S, 490.211 87; found 490.211 85. 1-[4-[4-(Benzenesulfonylmethyl)-6-[(3S)-3-methylmorpholin-4yl]pyrimidin-2-yl]phenyl]-3-methylurea (21). Yield 24%. 1H NMR (DMSO-d6) δ 1.21 (d, J = 6.73 Hz, 3H), 2.67 (d, J = 4.42 Hz, 3H), 3.07−3.26 (m, 1H), 3.49 (td, J = 2.91, 11.77, 11.83 Hz, 1H), 3.64 (dd, J = 2.97, 11.51 Hz, 1H), 3.77 (d, J = 11.45 Hz, 1H), 3.98 (dd, J = 3.42, 11.35 Hz, 1H), 4.10 (d, J = 13.03 Hz, 1H), 4.38 (s, 1H), 4.71 (s, 2H), 6.06 (d, J = 4.64 Hz, 1H), 6.61 (s, 1H), 7.39 (d, J = 8.80 Hz, 2H), 7.63 (t, J = 7.72, 7.72 Hz, 2H), 7.73−7.78 (m, 1H), 7.8−7.85 (m, 4H), 8.71 (s, 1H). HRMS-ESI (m/z): [M + H]+ calcd for C24H27N5O4S, 482.185 65; found, 482.185 55. 3-Methyl-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6-(pyridin-4ylsulfonylmethyl)pyrimidin-2-yl]phenyl]urea (22). Yield 15%. 1H NMR (DMSO-d6) δ 1.19 (d, J = 6.78 Hz, 3H), 2.64 (d, J = 4.58 Hz, 3H), 3.17 (td, J = 3.86, 12.89, 12.99 Hz, 1H), 3.46 (td, J = 3.04, 11.83, 11.84 Hz, 1H), 3.62 (dd, J = 3.08, 11.48 Hz, 1H), 3.75 (d, J = 11.41 Hz, 1H), 3.95 (dd, J = 3.67, 11.37 Hz, 1H), 4.10 (s, 1H), 4.37 (s, 1H), 4.85 (d, J = 1.48 Hz, 2H), 6.04 (d, J = 4.62 Hz, 1H), 6.69 (s, 1H), 7.35 (d, J = 8.78 Hz, 2H), 7.56−7.71 (d, J = 8.78 Hz, 2H), 7.72−7.86 (m, 2H), 8.70 (s, 1H), 8.84−8.96 (m, 2H). Mass calculated for C23H26N6O4S 482.56, found 483.45 (ESI, M + H). Compounds 23, 34, 39, and 42 were prepared from intermediates E, O, Q, and AH, respectively, according to the general procedure E. Compounds 43 and 44 were prepared from intermediate Z according to the general procedure E. General Proceedure E for Preparing Compounds. A solution of di(imidazol-1-yl)methanethione (1.3 equiv) in DCM was added to a solution of the appropriate intermediate (1 equiv) in either DCM or a mixture of DCM and THF. The reaction was stirred at ambient temperature for 3 h and then the appropriate amine (5 equiv) added followed by triethylamine (1 equiv). The reaction was stirred at ambient temperature for 1 h or until complete by TLC or LCMS analysis. The reaction mixture was concentrated in vacuo and the residue purified by HPLC. 3-Cyclopropyl-1-[4-[4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidin-2-yl]phenyl]thiourea (23). Yield 47%. 1H NMR (DMSO-d6) δ 0.60 (s, 2H), 0.71−0.86 (m, 2H), 1.26 (d, J = 6.73 Hz, 3H), 2.91 (d, J = 12.23 Hz, 1H), 3.25 (s, 1H), 3.48−3.6 (m, 1H), 3.66 (dd, J = 2.98, 11.53 Hz, 1H), 3.78 (d, J = 11.46 Hz, 2H), 3.99 (dd, J = 3.39, 11.37 Hz, 2H), 4.18 (d, J = 11.30 R

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry

3-[[2-Chloro-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-4-yl]methylsulfonyl]propan-1-ol (Intermediate X). A solution of intermediate C (12.4 g, 35.07 mmol) in DCM (50 mL) was added to a stirred solution of 3-mercapto-1-propanol (3.64 mL, 42.08 mmol) and DIPEA (9.77 mL, 56.11 mmol) in DCM (100 mL) over a period of 40 min under a nitrogen atmosphere. The resulting solution was stirred at ambient temperature for 18 h and then washed sequentially with a saturated aqueous solution of sodium hydrogen carbonate (2 × 50 mL) and saturated brine (50 mL). The organic layer was dried (MgSO4), filtered, and evaporated to afford crude product as a dark brown oil. The crude product was purified by flash silica chromatography, eluting with 0−75% ethyl acetate in DCM, to give 3-[[2-chloro-6-[(3S)-3-methylmorpholin-4yl]pyrimidin-4-yl]methylsulfanyl]propan-1-ol as a yellow gum (5.86 g, yield 53%). 1H NMR (CDCl3) δ 1.32 (3H, d), 1.84−1.90 (2H, m), 1.94 (1H, s), 2.69 (2H, t), 3.24−3.32 (1H, m), 3.51−3.58 (1H, m), 3.61 (2H, s), 3.67−3.71 (1H, m), 3.73−3.80 (3H, m), 3.98−4.04 (2H, m), 4.28−4.34 (1H, m), 6.45 (1H, s). ESI (m/z): [M + H]+ = 318. 3-Chlorobenzoperoxoic acid (4.00 g, 23.16 mmol) was added to 3-[[2-chloro-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-4-yl]methylsulfanyl]propan-1-ol (3.68 g, 11.58 mmol) in DCM (100 mL) and the resulting solution stirred at ambient temperature for 3 h. A further portion of 3-chlorobenzoperoxoic acid (2.00 g, 11.58 mmol) was added and the resulting solution stirred at ambient temperature for 1 h. The reaction mixture was washed sequentially with 10% aqueous sodium metabisulfite solution (2 × 100 mL), a saturated aqueous solution of sodium hydrogen carbonate (100 mL), and saturated brine (100 mL). The organic layer was dried (MgSO4), filtered, and evaporated to give the desired material as a gum (4.05 g, yield 100%). 1H NMR (CDCl3) δ 1.34 (3H, d), 2.12−2.18 (2H, m), 3.27 (2H, t), 3.31−3.35 (1H, m), 3.51−3.57 (1H, m), 3.67−3.70 (1H, m), 3.77−3.82 (3H, m), 3.99−4.03 (1H, m), 4.18 (2H, s), 4.26−4.37 (1H, m), 6.51 (1H, s). ESI (m/z): [M + H]+ = 350. 2-Chloro-4-[(4-fluorophenyl)sulfonylmethyl]-6-[(3S)-3-methylmorpholin-4-yl]pyrimidine (Intermediate AB). 4-Fluorobenzenesulfinic acid sodium salt (6.70 g, 36.77 mmol) was added to intermediate C (10.00 g, 28.28 mmol) in acetonitrile (250 mL) under nitrogen and the resulting solution stirred at 80 °C for 20 h. The reaction was cooled, the solvent was removed in vacuo and the residue partitioned between DCM and water. The organic layer was dried over MgSO4 and concentrated in vacuo to give crude material which was purified by flash silica chromatography, elution gradient 0−30% ethyl acetate in DCM, to give the desired material as a cream solid (8.32 g, yield 76%). 1H NMR (DMSO-d6) δ 1.17 (3H, d), 3.13−3.20 (1H, m), 3.27−3.28 (1H, m), 3.39−3.46 (1H, m), 3.57 (1H, dd), 3.72 (1H, d), 3.93 (1H, dd), 4.17 (1H, s), 4.65 (2H, s), 6.71 (1H, s), 7.48 (2H, t), 7.83−7.87 (2H, m). ESI (m/z): [M + H]+ = 386. 2-Chloro-4-[(3S)-3-methylmorpholin-4-yl]-6-(pyridin-4ylsulfonylmethyl)pyrimidine (Intermediate AA). Intermediate C (5.00 g, 14.14 mmol) was added portionwise to 4-mercaptopyridine (1.572 g, 14.14 mmol) and DIPEA (4.93 mL, 28.28 mmol) in acetonitrile (120 mL) over a period of 10 min. The resulting solution was stirred at ambient temperature for 3 h before being concentrated in vacuo and the residue partitioned between DCM and water. The organics were dried over MgSO4, concentrated in vacuo and the residue was purified by flash silica chromatography, elution gradient 0−4% methanol in DCM, to yield 2-chloro-4-[(3S)3-methylmorpholin-4-yl]-6-(pyridin-4-ylsulfanylmethyl)pyrimidine as a yellow solid (4.41 g, yield 93%). 1H NMR (DMSO-d6) δ 1.14− 1.16 (3H, d), 3.11−3.18 (1H, td), 3.37−3.44 (1H, td), 3.53−3.57 (1H, dd), 3.64−3.67 (1H, d), 3.86−3.90 (2H, dd), 4.01 (2H, s), 4.14 (1H, bs), 6.43 (1H, s), 7.04−7.06 (2H, d), 8.29−8.30 (2H, d). ESI (m/z): [M + H]+ = 337. A 30% aqueous solution of hydrogen peroxide (8.87 mL, 286.78 mmol) was added dropwise to a stirred solution of 2-chloro-4-[(3S)-3-methylmorpholin-4-yl]-6-(pyridin-4ylsulfanylmethyl)pyrimidine (4.83 g, 14.34 mmol) and sodium tungstate dihydrate (0.095 g, 0.29 mmol) in water (3.5 mL) and 2 N sulfuric acid (0.358 mL) in dioxane (48 mL) and methanol (2 mL). The resulting solution was stirred at 55 °C for 4 h and then

added dropwise to a solution of methyl 2,6-dichloropyrimidine-4carboxylate (5 g) in DCM (120 mL) over 10 min and the reaction stirred at ambient temperature for 1 h. The reaction was concentrated in vacuo, the residue dissolved in DCM and washed with water. The organics were dried (MgSO4), filtered, and evaporated and the residue chromatographed on silica, eluting with 2.5% methanol in DCM, to give the desired material as a white solid (3.15 g, yield 48%). 1H NMR (DMSO-d6) δ 1.22−1.24 (3H, m), 3.25 (1H, d), 3.41−3.48 (1H, m), 3.57−3.61 (1H, m), 3.71 (1H, d), 3.87 (3H, s), 3.91−3.95 (1H, m), 4.25 (1H, s), 4.45 (1H, s), 7.29 (1H, s). ESI (m/z): [M + H]+ = 272. [2-Chloro-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-4-yl]methanol (Intermediate B). Intermediate A (3.15 g) was dissolved in dry THF (20 mL) and cooled to 0 °C under nitrogen. A solution of lithium borohydride (2.0 M in THF, 6.09 mL) was added dropwise and the solution allowed to warm to rt and stirred for 1 h. The reaction was quenched with water (20 mL), then evaporated to dryness, the residue dissolved in ethyl acetate (150 mL) and washed with water (150 mL) followed by brine (50 mL). The organics were evaporated to dryness to give the desired material as a white solid (2.44 g, yield 66%). 1H NMR (DMSO-d6) δ 1.20−1.21 (3H, m), 3.18−3.22 (1H, m), 3.40−3.47 (1H, m), 3.56−3.60 (1H, m), 3.71 (1H, d), 3.91−3.94 (1H, m), 3.98 (1H, d), 4.35 (3H, d), 5.51 (1H, t), 6.74 (1H, s). ESI (m/z): [M + H]+ = 244. 2-Chloro-4-(iodomethyl)-6-[(3S)-3-methylmorpholin-4-yl]pyrimidine (Intermediate C). Methanesulfonyl chloride (0.245 mL, 3.14 mmol) was added dropwise over a period of 5 min to a solution of triethylamine (0.875 mL, 6.28 mmol) and intermediate B (510 mg, 2.09 mmol) in DCM (30 mL) at 0 °C under nitrogen. The resulting solution was stirred at ambient temperature for 45 min and then diluted with water (20 mL). The organic layer was dried (MgSO4) and filtered. Sodium iodide (1569 mg, 10.46 mmol) was added and the reaction heated at 50 °C for 20 h. The reaction mixture was filtered and evaporated to afford the desired material which was used crude in further reactions (761 mg, yield ∼100%). 1 H NMR (DMSO-d6) δ 1.19−1.25 (3H, m), 3.18−3.22 (1H, m), 3.40−3.47 (1H, m), 3.57−3.60 (1H, m), 3.71 (1H, d), 3.90−3.94 (1H, m), 3.96−3.98 (1H, m), 4.28−4.32 (3H, m), 6.94 (1H, s). ESI (m/z): [M + H]+ = 354. 2-Chloro-4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidine (Intermediate D). Methanesulfinic acid, sodium salt (11.75 g, 115.11 mmol) was added in one portion to intermediate C (37 g, 104.64 mmol) in acetonitrile (900 mL) and the resulting solution stirred at 85 °C for 24 h. The mixture was concentrated in vacuo and the residue dissolved in DCM. The organic layers were combined and washed with water (3 × 100 mL), dried over MgSO4, filtered, and the solvent was removed by evaporation to give the crude product as a dark brown oil, which solidifed (36 g). The crude solid was purified by flash silica chromatography, elution gradient 0−30% ethyl acetate in DCM, to give the desired material as a cream solid (22 g, yield 69%). 1H NMR (DMSO-d6) δ1.21−1.23 (m, 3H), 3.11 (s, 3H), 3.19−3.26 (m, 1H), 3.42−3.49 (m, 1H), 3.58−3.62 (1H, m), 3.73 (d, 1H), 3.92− 3.96 (m, 2H), 4.27−4.31 (m, 1H), 4.45 (s, 2H), 6.92 (s, 1H). ESI (m/z): [M + H]+ = 306. 2-Chloro-4-(cyclopropylsulfonylmethyl)-6-[(3S)-3-methylmorpholin-4-yl]pyrimidine (Intermediate L). Cyclopropanesulfinic acid, sodium salt (381 mg, 2.97 mmol) was added in one portion to intermediate C (700 mg, 1.98 mmol) in acetonitrile (20 mL) and the resulting suspension stirred at 90 °C for 3 h. The reaction mixture was evaporated to dryness and redissolved in DCM (50 mL) and washed with water (50 mL). The organic layer was dried (MgSO4), filtered, and evaporated to afford crude product. The crude product was purified by flash silica chromatography, elution gradient 0−40% ethyl acetate in DCM, to give the desired material as a white solid (458 mg, yield 70%). 1H NMR (DMSO-d6) δ 0.95−0.98 (2H, m), 1.02−1.06 (2H, m), 1.18−1.23 (3H, m), 2.77−2.83 (1H, m), 3.19− 3.25 (1H, m), 3.42−3.49 (1H, m), 3.58−3.62 (1H, m), 3.73 (1H, d), 3.92−3.96 (2H, m), 4.30 (1H, s), 4.48 (2H, s), 6.92 (1H, s). ESI (m/z): [M + H]+ = 332. S

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry

3.91 (2H, dd), 4.25 (1H, s), 6.70 (1H, s), 7.45 (2H, t), 7.79−7.84 (2H, m). ESI (m/z): [M + H]+ = 412. 2-Chloro-4-[(3S)-3-methylmorpholin-4-yl]-6-(1-pyridin-4ylsulfonylcyclopropyl)pyrimidine (Intermediate AF). Yield 82%. 1H NMR (DMSO-d6) δ 1.15−1.16 (3H, d), 1.61−1.65 (2H, m), 1.90− 1.93 (2H, m), 3.11−3.19 (1H, td), 3.37−3.44 (1H, td), 3.53−3.57 (1H, dd), 3.68−3.71 (1H, d), 3.89−3.96 (1H, dd), 3.96 (1H, bs), 4.28 (1H, bs), 6.75 (1H, s), 7.74−7.75 (2H, dd), 8.88−8.90 (2H, dd). ESI (m/z): [M + H]+ = 395. 4-[1-(Benzenesulfonyl)cyclopropyl]-2-chloro-6-[(3S)-3-methylmorpholin-4-yl]pyrimidine (Intermediate AG). Yield 93%. 1H NMR (DMSO-d6) δ 1.13−1.15 (3H, d), 1.55−1.57 (2H, m), 1.83−1.86 (2H, m), 3.09−3.16 (1H, td), 3.36−3.43 (1H, td), 3.52−3.56 (1H, dd), 3.68−3.71 (1H, d), 3.86−3.93 (2H, m), 4.20 (1H, bs), 6.67 (1H, s), 7.60−7.63 (2H, m), 7.72−7.77 (3H, m). ESI (m/z): [M + H]+ = 394. 3-[1-[2-Chloro-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-4-yl]cyclopropyl]sulfonylpropan-1-ol (Intermediate Y). To a solution of intermediate X (5.04 g, 14.41 mmol) in DMF (25 mL) was added a solution of chlorotrisopropylsilane (3.70 mL, 17.29 mmol) and imidazole (2.354 g, 34.58 mmol) in DMF (25 mL) over a period of 5 min under a nitrogen atmosphere. The resulting solution was stirred at ambient temperature for 18 h. The reaction mixture was evaporated to dryness, the residue dissolved in DCM (200 mL) and then washed sequentially with water (100 mL) and saturated brine (100 mL). The organic layer was dried (MgSO4), filtered, and evaporated to give 3-[[2-chloro-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-4-yl]methylsulfonyl]propoxytri(propan-2-yl)silane as an oil (7.29 g, yield 100%). 1H NMR (CDCl3) δ 0.99−1.07 (21H, m), 1.33 (3H, d), 2.06−2.13 (2H, m), 3.20−3.24 (2H, m), 3.26− 3.34 (1H, m), 3.50−3.57 (1H, m), 3.66−3.70 (1H, m), 3.77−3.83 (3H, m), 3.99−4.03 (2H, m), 4.16 (2H, s), 4.25−4.37 (1H, m), 6.54 (1H, s). ESI (m/z): [M + H]+ = 506. 1,2-Dibromoethane (1.723 mL, 20 mmol) was added to 3-[[2-chloro-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-4-yl]methylsulfonyl]propoxytri(propan-2-yl)silane (5.0 g, 9.88 mmol), 40% sodium hydroxide solution (10 mL, 98.78 mmol), and tetrabutylammonium bromide (0.645 g, 2 mmol) in toluene (50 mL). The resulting mixture was stirred at 60 °C for 4 h. The reaction mixture was evaporated to dryness and redissolved in ethyl acetate (100 mL) and washed sequentially with water (100 mL) and saturated brine (100 mL). The organic layer was dried (MgSO4), filtered, and evaporated to afford crude product. The crude product was purified by flash silica chromatography, eluting with 0−20% ethyl acetate in DCM, to give 3-[1-[2-chloro-6-[(3S)-3methylmorpholin-4-yl]pyrimidin-4-yl]cyclopropyl]sulfonylpropoxytri(propan-2-yl)silane as a colorless gum (2.86 g, yield 54%). 1H NMR (CDCl3) δ 1.00−1.05 (21H, m), 1.32 (3H, d), 1.49−1.52 (2H, m), 1.78−1.81 (2H, m), 2.02−2.09 (2H, m), 3.21−3.32 (3H, m), 3.50− 3.56 (1H, m), 3.65−3.69 (1H, m), 3.77−3.80 (3H, m), 3.98−4.02 (2H, m), 4.28−4.36 (1H, m), 6.90 (1H, s). ESI (m/z): [M + H]+ = 532. Tetrabutylammonium fluoride (1 M in THF, 31 mL, 31 mmol) was added to 3-[1-[2-chloro-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-4-yl]cyclopropyl]sulfonylpropoxytri(propan-2-yl)silane (3.28 g, 6.16 mmol) in THF (30 mL). The resulting solution was stirred at ambient temperature for 1 h and then concentrated in vacuo and diluted with saturated ammonium chloride (10 mL) and water. The mixture was extracted twice with ethyl acetate, and the combined organic extracts were washed with brine, dried (MgSO4), filtered, and evaporated to give the desired material as a gum (2.33 g, yield 100%). 1H NMR (CDCl3) δ 1.33 (3H, d), 1.42−1.51 (4H, m), 2.07−2.14 (2H, m), 2.40 (1H, s), 3.28−3.32 (2H, m), 3.37−3.42 (3H, m), 3.51−3.57 (1H, m), 3.66−3.70 (1H, m), 3.77−3.80 (2H, m), 3.99−4.02 (1H, m), 4.28−4.38 (1H, m), 6.84 (1H, s). ESI (m/ z): [M + H]+ = 376. 2-Chloro-4-[(3S)-3-methylmorpholin-4-yl]-6-(1methylsulfonylcyclobutyl)pyrimidine (Intermediate R). Sodium hydroxide (50%w/w solution) (36.0 g, 450 mmol) was added to intermediate D (5 g, 16.35 mmol), 1,3-dibromopropane (4.98 mL, 49.05 mmol), and tetrabutylammonium bromide (0.527 g, 1.64 mmol) in toluene (200 mL). The resulting suspension was stirred at

diluted with water and cooled. A 10% aqueous solution of sodium metabisulfite was added and the mixture extracted with DCM, and the organics were dried over MgSO4 and filtered to obtain the desired material as a solid (4.47 g, 85%). 1H NMR (DMSO-d6) δ 1.17−1.19 (3H, d), 3.14−3.22 (1H, td), 3.40−3.47 (1H, td), 3.56− 3.60 (1H, dd), 3.71−3.74 (1H, d), 3.90 (1H, bs), 3.91−3.95 (1H, dd), 4.20 (1H, bs), 4.79 (2H, s), 6.79 (1H, s), 7.77−7.79 (2H, q), 8.92−8.93 (2H, q). ESI (m/z): [M + H]+ = 369. 4-(Benzenesulfonylmethyl)-2-chloro-6-[(3S)-3-methylmorpholin4-yl]pyrimidine (Intermediate AC). Benzenesulfinic acid, sodium salt (4.22 g, 25.74 mmol) was added to intermediate C (7.0 g, 19.80 mmol) in acetonitrile (200 mL) and the resulting mixture stirred under a nitrogen atmosphere at 80 °C for 20 h. The reaction was cooled, and the solvent was removed. DCM was added, and the solution was washed with water. The organics were dried (MgSO4), filtered, and the solvent was removed. The crude product was purified by flash silica chromatography, elution gradient 0−30% ethyl acetate in DCM, to give the desired material as a cream solid (6.21 g, yield 85%). 1H NMR (DMSO-d6) δ 1.15−1.16 (3H, d), 3.11−3.18 (1H, td), 3.38−3.45 (1H, td), 3.55−3.58 (1H, dd), 3.70−3.73 (1H, d), 3.85−3.94 (2H, m), 4.15 (1H, bs), 4.64 (2H, s), 6.67 (1H, s), 7.63−7.66 (2H, m), 7.74−7.80 (3H, m). ESI (m/z): [M + H]+ = 368. 2-Chloro-4-[(3S)-3-methylmorpholin-4-yl]-6-(2-methylsulfonylpropan-2-yl)pyrimidine (Intermediate N). Sodium tert-butoxide (278 mg, 2.89 mmol) was added to intermediate D (883 mg, 2.89 mmol) in DMF (25 mL) at 0 °C under nitrogen. Iodomethane (0.180 mL, 2.89 mmol) was added, and the resulting solution was stirred at 0 °C for 15 min. Further sodium tert-butoxide (278 mg, 2.89 mmol) was added followed by iodomethane (0.180 mL, 2.89 mmol), and the resulting solution was stirred at 0 °C for 1 h. The reaction was diluted with DCM (100 mL) and washed with water (100 mL) and brine (100 mL). The organic layer was dried (MgSO4), filtered and the solvent evaporated to a gum which slowly crystallized.The crude product was purified by flash silica chromatography, eluting with 0−5% methanol in DCM, to give the desired material as a white solid (691 mg, yield 72%). 1H NMR (DMSO-d6) δ 1.26 (3H, d), 1.72 (6H, s), 2.87 (3H, s), 3.19−3.27 (1H, m), 3.44−3.51 (1H, m), 3.60−3.63 (1H, m), 3.72 (1H, d), 3.92−3.96 (2H, m), 4.23−4.32 (1H, m), 6.53 (1H, s). ESI (m/z): [M + H]+ = 334. Intermediates P, AD, AE, AF, and AG were prepared from intermediates D, L, AB, AA, and AC respectively, according to the general procedure F. General Procedure F. A mixture of the appropriate intermediate (1 equiv), tetrabutylammonium bromide (0.1−0.2 equiv) and 1,2dibromoethane (2−5 equiv) in an aqueous solution of sodium hydroxide (large excess) and toluene was heated at 60−70 °C for 4 h or until complete by either TLC or LCMS analysis. The reaction mixture was either washed directly with water or partitioned between ethyl acetate or DCM and water. The organics were dried over MgSO4, concentrated in vacuo, and purified by either flash silica chromatography or trituration with diethyl ether. 2-Chloro-4-[(3S)-3-methylmorpholin-4-yl]-6-(1methylsulfonylcyclopropyl)pyrimidine (Intermediate P). Yield 78%. 1 H NMR (DMSO-d6) δ 1.22 (d, 3H), 1.51 (m, 2H), 1.64 (m, 2H), 3.18 (s, 3H), 3.22 (m, 1H), 3.43 (m, 1H), 3.58 (m, 1H), 3.72 (d, 1H), 3.93 (m, 1H), 4.05 (d, 1H), 4.41 (s, 1H), 6.93 (s, 1H). ESI (m/z): [M + H]+ = 332. 2-Chloro-4-(1-cyclopropylsulfonylcyclopropyl)-6-[(3S)-3-methylmorpholin-4-yl]pyrimidine (Intermediate AD). Yield 93%. 1H NMR (DMSO-d6) δ 0.87−0.92 (2H, m), 0.99−1.04 (2H, m), 1.20 (3H, d), 1.50−1.54 (2H, m), 1.59−1.63 (2H, m), 2.89−2.96 (1H, m), 3.16− 3.24 (1H, m), 3.39−3.47 (1H, m), 3.58 (1H, dd), 3.72 (1H, d), 3.93 (1H, dd), 4.06 (1H, s), 4.38 (1H, s), 6.98 (1H, s). ESI (m/z): [M + H]+ = 358. 2-Chloro-4-[1-(4-fluorophenyl)sulfonylcyclopropyl]-6-[(3S)-3methylmorpholin-4-yl]pyrimidine (Intermediate AE). Yield 90%. 1H NMR (DMSO-d6) δ 1.15 (3H, d), 1.53−1.56 (2H, m), 1.82−1.85 (2H, m), 3.14 (1H, dt), 3.40 (1H, dt), 3.55 (1H, dd), 3.70 (1H, d), T

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry 45 °C for 3 h and then allowed to cool. The reaction was washed with water, dried over MgSO4, filtered, and evaporated. The crude product was purified by flash silica chromatography, elution gradient 30−50% ethyl acetate in DCM, to give the desired material as a solid (1.89 g, yield 33%). 1H NMR (DMSO-d6) δ 1.22 (3H, s), 1.86−1.93 (1H, m), 2.01−2.09 (1H, m), 2.66−2.75 (2H, m), 2.84 (2H t, J= 14.7 Hz), 2.86 (3H, s), 3.18−3.25 (1H, m), 3.42−3.49 (1H, m), 3.58−3.62 (1H, m), 3.73 (1H d, J= 11.6 Hz), 3.92−3.96 (1H, m), 4.08−4.10 (1H, m), 4.43 (1H, s), 6.83 (1H, s). ESI (m/z): [M + H]+ = 346. 2-Chloro-4-[(3S)-3-methylmorpholin-4-yl]-6-(1methylsulfonylcyclopentyl)pyrimidine (Intermediate T). Tetrabutylammonium bromide (0.637 g, 1.98 mmol) was added to intermediate D (6.04 g, 19.75 mmol), 1,4-dibromobutane (11.79 mL, 98.76 mmol), and sodium hydroxide solution (50% w/w) (35 mL, 438 mmol) in toluene (150 mL) under nitrogen. The resulting mixture was stirred at 60 °C for 1.5 h before being allowed to cool and partitioned between ethyl acetate and water. The organics were dried over Na2SO4, concentrated in vacuo, and purified by flash silica chromatography, eluting with 5−50% ethyl acetate in isohexane, to give the desired material as a white solid (5.70 g, yield 80%). 1H NMR (DMSO-d6) δ 1.20 (3H, d), 1.50−1.60 (2H, m), 1.72−1.82 (2H, m), 2.30−2.41 (2H, m,), 2.50−2.60 (2H, m), 2.88 (3H, s), 3.20 (1H, dd), 3.45 (1H, dd), 3.60 (1H, dd), 3.71 (1H, d), 3.94 (1H, dd), 4.0−4.10 (1H, m), 4.42 (1H, s), 6.89 (1H, s). ESI (m/z): [M + H]+ = 360. 2-Chloro-4-[(3S)-3-methylmorpholin-4-yl]-6-(4-methylsulfonyloxan-4-yl)pyrimidine (Intermediate V). Sodium tert-butoxide (1.38 g, 14.39 mmol) was added portionwise to a mixture of intermediate D (2.00 g, 6.54 mmol) and bis(2-bromoethyl) ether (2.055 mL, 16.35 mmol) in DMF (75 mL) at 0 °C over a period of 10 min under nitrogen. The resulting solution was allowed to warm to rt and stirred for 7 h. Further sodium tert-butoxide (629 mg, 6.54 mmol) was added portionwise, and the solution was stirred at rt for a further 45 h. The reaction mixture was concentrated, diluted with ethyl acetate (200 mL), and washed sequentially with water (2 × 200 mL) and saturated brine (100 mL). The organic layer was dried (MgSO4), filtered, and evaporated. The crude product was purified by flash silica chromatography, elution gradient 40−100% ethyl acetate in isohexane. Pure fractions were evaporated to dryness and the residue was crystallized from ethyl acetate/isohexane to afford the desired material as a white crystalline solid (1.42 g, yield 58%). 1 H NMR (CDCl3) δ 1.34 (3H, d), 2.50 (2H, m), 2.55 (2H, m), 2.73 (3H, s), 3.33 (3H, m), 3.56 (1H, ddd), 3.71 (1H, dd), 3.80 (1H, d), 4.01 (4H, m), 4.31 (1H, br s), 6.62 (1H, s). ESI (m/z): [M + H]+ = 376, 378. Intermediates E, M, O, Q, S, U, W, Z, AH, AI, AJ, and AK were prepared from intermediates D, L, N, P, R, T, V, Y, AD, AE, AF, and AG according to the general procedure G. General Procedure G. A mixture of the appropriate intermediate (1 equiv), (4-aminophenyl)boronic acid pinacol ester (1−1.5 equiv), sodium carbonate (1−5 equiv), and dichlorobis(triphenylposphine)palladium (5−10 mol %) in a solvent mixture of 18% DMF, 82% DME/water/ethanol (7:3:2) was heated at 80−95 °C under a nitrogen atmosphere for 5 h or until the reaction was complete by TLC or LCMS analysis. The reaction mixture was partitioned between ethyl acetate and water, and the organic layer was washed with water or brine, dried over MgSO4, and concentrated in vacuo. The crude material was purified by flash silica chromatography and where necessary by ion exchange chromatography (SCX column). 4-[4-[(3S)-3-Methylmorpholin-4-yl]-6-(methylsulfonylmethyl)pyrimidin-2-yl]aniline (Intermediate E). Yield 90%. 1H NMR (DMSO-d6) δ 1.23 (3H, d), 3.31 (3H, s), 3.5 (1H, m), 3.64 (1H, m), 3.78 (1H, m), 4.13 (1H, m), 4.49 (2H, m), 5.57 (2H, s), 6.61 (2H, d), 6.68 (1H, s), 8.08 (1H, d). ESI (m/z): [M + H]+ = 363. 4-[4-(Cyclopropylsulfonylmethyl)-6-[(3S)-3-methylmorpholin-4yl]pyrimidin-2-yl]aniline (Intermediate M). Yield 92%. 1H NMR (DMSO-d6) δ 0.97−1.02 (2H, m), 1.03−1.10 (2H, m), 1.23 (3H, d), 2.81−2.87 (1H, m), 3.15−3.22 (1H, m), 3.46−3.52 (1H, m), 3.62− 3.66 (1H, m), 3.77 (1H, d), 3.96−3.99 (1H, m), 4.12−4.15 (1H, m),

4.45 (3H, s), 5.53 (2H, d), 6.58−6.61 (2H, m), 6.66 (1H, s), 8.03− 8.07 (2H, m). ESI (m/z): [M + H]+ = 389. 4-[4-[(3S)-3-Methylmorpholin-4-yl]-6-(2-methylsulfonylpropan2-yl)pyrimidin-2-yl]aniline (Intermediate O). Yield 68%. 1H NMR (DMSO-d6) δ 1.33 (3H, d), 1.87 (6H, s), 2.93 (3H, s), 3.28−3.35 (1H, m), 3.56−3.63 (1H, m), 3.72−3.76 (1H, m), 3.82 (1H, d), 3.90 (2H, s), 4.01−4.05 (1H, m), 4.11−4.15 (1H, m), 4.46−4.53 (1H, m), 6.55 (1H, s), 6.71 (2H, d), 8.21 (2H, d). ESI (m/z): [M + H]+ = 391. 4-[4-[(3S)-3-Methylmorpholin-4-yl]-6-(1methylsulfonylcyclopropyl)pyrimidin-2-yl]aniline (Intermediate Q). Yield 91%. 1H NMR (DMSO-d6) δ 1.24 (3H, d), 1.55 (2H, m), 1.67 (2H, m), 3.23 (1H, m), 3.27 (3H, s), 3.47 (1H, m), 3.63 (1H, m), 3.77 (1H, d), 3.97 (1H, m), 4.24 (1H, s), 4.58 (1H, s), 5.75 (1H, s), 6.68 (2H, d), 8.04 (2H, d). ESI (m/z): [M + H]+ = 389. 4-[4-[(3S)-3-Methylmorpholin-4-yl]-6-(1methylsulfonylcyclobutyl)pyrimidin-2-yl]aniline (Intermediate S). Yield 84%. 1H NMR (DMSO-d6) δ 1.21 (3H,d), 1.85−1.95 (2H,m), 2.0−2.10 (2H,m), 2.71−2.82 (2H,m), 2.82 (3H,s), 3.18 (1H,dd), 3.50 (1H,dd), 3.62 (1H,dd), 3.75 (1H,d), 3.95 (1H,dd), 4.20 (1H,d), 4.53 (1H,s), 5.55 (2H,s), 6.60 (3H,d), 8.05 (2H,d). ESI (m/z): [M + H]+ = 403. 4-[4-[(3S)-3-Methylmorpholin-4-yl]-6-(1methylsulfonylcyclopentyl)pyrimidin-2-yl]aniline (Intermediate U). Yield 92%. 1H NMR (DMSO-d6) δ 1.21 (3H,d), 1.50−1.60 (2H,m), 1.75−1.90 (2H,m), 2.34−2.43 (2H,m), 2.62−2.78 (2H,m), 2.88 (3H,s), 3.18 (1H,dd), 3.48 (1H,dd), 3.65 (1H,dd), 3.75 (1H,dd), 3.95 (1H,dd), 4.20 (1H,d), 4.51 (1H,s), 5.55 (2H,s), 6.62 (2H,d), 6.68 (1H,s), 8.09 (1H,d). ESI (m/z): [M + H]+ = 417. 4-[4-[(3S)-3-Methylmorpholin-4-yl]-6-(4-methylsulfonyloxan-4yl)pyrimidin-2-yl]aniline (Intermediate W). Yield 50%. 1H NMR (CDCl3) δ 1.34 (3H, d), 2.53 (2H, ddd), 2.70 (3H, s), 2.72 (2H, br d), 3.33 (1H, ddd), 3.41 (2H, ddd), 3.61 (1H, ddd), 3.76 (1H, dd), 3.83 (1H, d), 3.93 (2H, s), 4.03 (3H, m), 4.18 (1H, d), 4.45 (1H, br.d), 6.58 (1H, s), 6.71 (2H, d), 8.18 (2H, d). ESI (m/z): [M + H]+ = 433.5. 3-[1-[2-(4-Aminophenyl)-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-4-yl]cyclopropyl]sulfonylpropan-1-ol (Intermediate Z). Yield 71%. 1H NMR (DMSO-d6) δ 1.22 (3H, d), 1.52−1.54 (2H, m), 1.60−1.62 (2H, m), 1.90−1.97 (2H, m), 3.14−3.21 (1H, m), 3.44−3.52 (5H, m), 3.60−3.64 (1H, m), 3.75 (1H, d), 3.94−3.98 (1H, m), 4.16−4.19 (1H, m), 4.48−4.55 (1H, m), 4.67 (1H, t), 5.56 (2H, s), 6.60 (2H, d), 6.67 (1H, s), 8.04 (2H, d). ESI (m/z): [M + H]+ = 433. 4-[4-(1-Cyclopropylsulfonylcyclopropyl)-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-2-yl]aniline (Intermediate AH). Yield 78%. 1H NMR (DMSO-d6) δ 0.90−0.98 (2H, m), 0.98−1.05 (2H, m), 1.22 (3H, d), 1.52−1.59 (2H, m), 1.62−1.64 (2H, m), 2.95−3.02 (1H, m), 3.14−3.22 (1H, m), 3.45−3.51 (1H, m), 3.61−3.65 (1H, m), 3.76 (1H, d), 3.95−3.98 (1H, m), 4.14−4.17 (1H, m), 4.48−4.51 (1H, m), 5.53 (2H, d), 6.60 (2H, d), 6.75 (1H, s), 8.03−8.06 (2H, m). ESI (m/z): [M + H]+ = 415. 4-[4-[1-(4-Fluorophenyl)sulfonylcyclopropyl]-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-2-yl]aniline (Intermediate AI). Yield 78%. 1 H NMR (DMSO-d6) δ 1.17 (3H, d), 1.57−1.59 (2H, m), 1.86− 1.88 (2H, m), 3.12 (1H, dt), 3.45 (1H, dt), 3.60 (1H, dd), 3.73 (1H, d), 3.95 (1H, dd), 4.10 (1H, d), 4.38 (1H, s), 5.52 (2H, s), 6.49 (2H, d), 6.55 (1H, s), 7.41 (2H, t), 7.64 (2H, d), 7.82−7.85 (2H, m). ESI (m/z): [M + H]+ = 469. 4-[4-[(3S)-3-Methylmorpholin-4-yl]-6-(1-pyridin-4ylsulfonylcyclopropyl)pyrimidin-2-yl]aniline (Intermediate AJ). Yield 73%. 1H NMR (DMSO-d6) δ 1.17 (3H, d), 1.66 (2H, q), 1.95 (2H, q), 3.13 (1H, td), 3.18 (1H, d), 3.45 (1H, td), 3.60 (1H, dd), 3.73 (1H, d), 3.94 (1H, dd), 4.15 (1H, d), 4.43 (1H, s), 5.51 (1H, d), 6.47 (2H, d), 6.56 (1H, s), 7.50 (2H, d), 7.76 (2H, dd), 8.85 (2H, dd). ESI (m/z): [M + H]+ = 452. 4-[4-[1-(Benzenesulfonyl)cyclopropyl]-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-2-yl]aniline (Intermediate AK). Yield 97%. 1H NMR (DMSO-d6) δ 1.15−1.16 (3H, d), 1.58−1.66 (2H, m), 1.85− 1.91 (2H, m), 3.07−3.14 (1H, td), 3.41−3.48 (1H, td), 3.58−3.61 (1H, dd), 3.72−3.75 (1H, d), 3.93−3.96 (1H, dd), 4.05−4.08 (1H, U

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry

were dried over magnesium sulfate, filtered, and concentrated in vacuo. The crude material was chromatographed on silica, eluting with 5% methanol in DCM, to give the desired material as a brown solid (1.25 g, yield 32%). 1H NMR (DMSO-d6) δ 1.23 (3H, d), 2.66 (3H, d), 3.18−3.23 (1H, m), 3.46−3.52 (1H, m), 3.62−3.66 (1H, m), 3.76 (1H, d), 3.96−3.99 (1H, m), 4.16 (1H, d), 4.45 (2H, d), 4.49 (1H, d), 5.38 (1H, t), 6.05 (1H, q), 6.66 (1H, s), 7.46−7.48 (2H, m), 8.18−8.21 (2H, m), 8.69 (1H, s). ESI (m/z): [M + H]+ = 358.

d), 4.33 (1H, bs), 5.50 (2H, s), 6.49−6.53 (3H, t), 7.57−7.61 (2H, t), 7.68−7.71 (3H, m), 7.78−7.81 (2H, d). ESI (m/z): [M + H]+ = 451. Intermediates G, I, and K were prepared from intermediates D, D, and B, respectively, in a method analogous to general procedure G but using (4-amino-3-methylphenyl)boronic acid, (N-methyl-4aminophenyl)boronic acid pinacol ester, and 4-(3cyclopropylureido)phenylboronic acid, respectively. 2-Methoxy-4-[4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidin-2-yl]aniline (Intermediate G). Yield 71%. 1H NMR (DMSO-d6) δ1.24 (d, 3H), 3.17−3.21 (m, 1H), 3.23 (s, 3H), 3.47−3.54 (m, 1H), 3.64−3.67 (m, 1H), 3.78 (d, 1H), 3.83 (s, 3H), 3.97−4.01 (m, 1H), 4.16 (d, 1H), 4.46 (s, 3H), 5.23 (s, 2H), 6.68−6.70 (m, 1H), 6.69 (s, 1H), 7.77 (m, 1H), 7.97 (s, 1H). ESI (m/z): [M + H]+ = 393. N-Methyl-4-[4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidin-2-yl]aniline (Intermediate I). Yield 90%. 1H NMR (DMSO-d6) δ1.23 (3H, d), 2.74 (3H, d), 3.06−3.17 (1H, m), 3.21 (3H, s), 3.46−3.52 (1H, m), 3.62−3.66 (1H, m), 3.77 (1H, d), 3.96−4.00 (1H, m), 4.14 (1H, d), 4.44 (2H, s), 4.46 (1H, s), 6.14 (1H, q), 6.57−6.61 (2H, m), 6.67 (1H, s), 8.10−8.13 (2H, m). ESI (m/z): [M + H]+ = 377. 3-Cyclopropyl-1-[4-[4-(hydroxymethyl)-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-2-yl]phenyl]urea (Intermediate K). Yield 83%. 1H NMR (DMSO-d6) δ 0.40−0.44 (2H, m), 0.62−0.67 (2H, m), 1.23 (3H, d), 2.54−2.58 (1H, m), 3.17−3.22 (1H, m), 3.46−3.52 (1H, m), 3.62−3.66 (1H, m), 3.77 (1H, d), 3.96−3.99 (1H, m), 4.15− 4.18 (1H, m), 4.45 (2H, d), 4.49 (1H, d), 5.38 (1H, t), 6.41 (1H, d), 6.66 (1H, s), 7.46−7.50 (2H, m), 8.18−8.22 (2H, m), 8.49 (1H, s). ESI (m/z): [M + H]+ = 384. 2-Methyl-4-[4-[(3S)-3-methylmorpholin-4-yl]-6(methylsulfonylmethyl)pyrimidin-2-yl]aniline (Intermediate H). 4Bromo-2-methylaniline (1.00 g, 5.37 mmol), potassium acetate (1.59 g, 16.1 mmol), and bis(pinacolato)diboron (1.64 g, 6.45 mmol) were dissolved in 1,4-dioxane (20 mL), and the solution was degassed for 5 min. Dichlorobis(triphenylphosphine)palladium (264 mg, 0.32 mmol) was added and the reaction stirred at 90 °C for 4 h. Intermediate D (1.65 g, 5.37 mmol), ethanol (1.5 mL), 2 M sodium carbonate aqueous solution (3 mL), and dichlorobis(triphenylphosphine)palladium (264 mg) were added. The reaction was maintained at 90 °C for 18 h and then allowed to cool and the mixture partitioned between ethyl acetate and water with insoluble material removed by filtration. The phases were separated, and the aqueous layer was extracted with a second portion of ethyl acetate (15 mL). The combined organics were dried (MgSO4) and concentrated in vacuo. The residue was chromatographed on silica, eluting with 0−3% methanol in DCM, to give the desired compound as a beige foam (290 mg, yield 15%). 1H NMR (DMSO-d6) δ1.22− 1.24 (m, 3H), 3.16−3.19 (m, 1H), 3.20 (s, 3H), 3.46−3.52 (m, 1H), 3.62−3.66 (m, 1H), 3.77 (d, 1H), 3.90 (s, 1H), 3.96−4.00 (m, 1H), 4.14−4.18 (m, 1H), 4.43 (s, 2H), 4.45 (s, 1H), 5.32 (s, 2H), 6.63 (s, 1H), 6.66 (s, 1H), 7.91−7.94 (m, 2H). ESI (m/z): [M + H]+ = 377. 1-[4-[4-(Hydroxymethyl)-6-[(3S)-3-methylmorpholin-4-yl]pyrimidin-2-yl]phenyl]-3-methylurea (Intermediate J). 1-(4-Bromophenyl)-3-methylurea (2.50 g), potassium acetate (3.21 g), and bis(pinacolato)diboron (3.33 g) were dissolved in 1,4 dioxane (120 mL). The solution was degassed for 5 min. Then 1,1′-bis(diphenylphosphino)ferrocenedichloropalladium(II) dichloromethane adduct was added (535 mg) and the reaction was heated to 90 oC for 3 hours. Further 1,1′-bis(diphenylphosphino)ferrocenedichloropalladium(II) dichloromethane adduct (250 mg) was added, and heating continued for 1 hour. Further 1,1′-bis(diphenylphosphino)ferrocenedichloropalladium(II) dichloromethane adduct (250 mg) was added, and heating continued for a further 1 hour. Intermediate B (2.66 g), ethanol (9.5 mL), 2 M sodium carbonate solution (27.3 mL), and 1,1′-bis(diphenylphosphino)ferrocenedichloropalladium(II) dichloromethane adduct (535 mg) were added, and the heating was continued for 16 h. The reaction was cooled and evaporated to dryness and then the residue partitioned between ethyl acetate (250 mL) and water (100 mL). The organics



AUTHOR INFORMATION

Corresponding Author

*Phone: +44 7469 032383. E-mail: kurt.pike@astrazeneca. com. Notes

The authors declare no competing financial interest.



ACKNOWLEDGMENTS The authors thank Dr. Paul Johnson, John Bailey, Jennifer Peed, Matthew Box, Greg Carr, George Hill, Gillian Lamont, Steve Glossop, and Ciorsdaidh Watts for their help synthesizing compounds that underpinned the Free−Wilson models described, Tom Harding and Rebecca Ellston for the generation of the potency data used in the analyses described, Dr. Christine Cresta for helpful discussions regarding mTOR biology, and Anne Ertan for her help generating the small molecule crystal structure of 15 (CCDC 1019570).



ABBREVIATIONS USED mTOR, mammalian target of rapamycin; mTORC1, mTOR complex 1; mTORC2, mTOR complex 2; PIKK, phosphatidylinositol 3-kinase related kinase; ATP, adenosine triphosphate; DMF, N,N-dimethylformamide; DMA, N,N-dimethylacetamide; DME, 1,2-dimethoxyethane; DCM, dichloromethane; DBU, 1,8-diazabicyclo[5.4.0]undec-7-ene; mCPBA, 3chloroperbenzoic acid; DMSO, dimethyl sulfoxide; LCMS, liquid chromatography−mass spectrometry; HPLC, high performance liquid chromatography; TLC, thin layer chromatography; SCX, strong cation exchange



REFERENCES

(1) Huang, S.; Houghton, P. J. Targeting mTOR signaling for cancer therapy. Curr. Opin. Pharmacol. 2003, 3, 371−377. (2) Yap, T. A.; Garrett, M. D.; Walton, M. I.; Raynaud, F.; de Bono, J. S.; Workman, P. Targeting the PI3K-AKT-mTOR pathway:progress, pitfalls, and promises. Curr. Opin. Pharmacol. 2008, 8, 393−412. (3) Brown, E. J.; Albers, M. W.; Shin, T. B.; Ichikawa, K.; Keith, C. T.; Lane, W. S.; Schreiber, S. L. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature 1994, 369, 756− 758. (4) Sabatini, D. M.; Erdjument-Bromage, H.; Lui, M.; Tempst, P.; Snyder, S. H. RAFT1: a mammalian protein that binds to FKBP12 in a rapamycin-dependent fashion and is homologous to yeast TORs. Cell 1994, 78, 35−43. (5) Abraham, R. T. Phosphatidylinositol 3-kinase related kinases. Curr. Opin. Immunol. 1996, 8, 412−418. (6) Fingar, D. C.; Salama, S.; Tsou, C.; Harlow, E.; Blenis, J. Mammalian cell size is controlled by mTOR and its downstream targets S6K1 and 4EBP1/eIF4E. Genes Dev. 2002, 16, 1472−1487. (7) Fingar, D. C.; Richardson, C. J.; Tee, A. R.; Cheatham, L.; Tsou, C.; Blenis, J. mTOR controls cell cycle progression through its cell growth effectors S6K1 and 4E-BP1/eukaryotic translation initiation factor 4E. Mol. Cell. Biol. 2004, 24, 200−216.

V

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry (8) Brunn, G. J.; Williams, J.; Sabers, C.; Wiederrecht, G.; Lawrence, J. C., Jr.; Abraham, R. T. Direct inhibition of the signaling functions of the mammalian target of rapamycin by the phosphoinositide 3-kinase inhibitors, wortmannin and LY294002. EMBO J. 1996, 15, 5256−5267. (9) Sarbassov, D. D.; Ali, S. M.; Sabatini, D. M. Growing roles for the mTOR pathway. Curr. Opin. Cell Biol. 2005, 17, 596−603. (10) Edinger, A. L.; Linardic, C. M.; Chiang, G. G.; Thompson, C. B.; Abraham, R. T. Differential effects of rapamycin on mammalian target of rapamycin signaling functions in mammalian cells. Cancer Res. 2003, 63, 8451−8460. (11) Sun, S. Y.; Rosenberg, L. M.; Wang, X.; Fu, H.; Khuri, F. R.Activation of Akt and eIF4E survival pathways by rapamycinmediated mammalian target of rapamycin inhibition. Cancer Res. 2005, 65, 7052−7058. (12) Cloughesy, T. F.; Yoshimoto, K.; Nghiemphu, P.; Brown, K.; Dang, J.; Zhu, S.; Hsueh, T.; Chen, Y.; Wang, W.; Youngkin, D.; Liau, L.; Martin, N.; Becker, D.; Bergsneider, M.; Lai, A.; Green, R.; Oglesby, T.; Koleto, M.; Trent, J.; Horvath, S.; Mischel, P. S.; Mellinghoff, I. K.; Sawyers, C. L. Antitumor activity of rapamycin in a phase I trial for patient with recurrent PTEN-deficient glioblastoma. PLoS Med. 2008, 5 (1), e8. (13) (a) Garcia-Echeverria, C. Allosteric and ATP-competitive kinase inhibitors of mTOR for cancer treatment. Bioorg. Med. Chem. Lett. 2010, 20, 4308−4312. (b) Faivre, S.; Kroemer, G.; Raymond, E. Current development of mTOR inhibitors as anticancer agents. Nat. Rev. Drug Discovery 2006, 5, 671−688. (c) Finlay, M. R. V.; Griffin, R. J. Modulation of DNA repair by pharmacological inhibitors of the PIKK protein kinase family. Bioorg. Med. Chem. Lett. 2012, 22, 5352−5359. (d) Cano, C.; Saravanan, K.; Bailey, C.; Bardos, J.; Curtin, N. J.; Frigerio, M.; Golding, B. T.; Hardcastle, I. R.; Hummersone, M. G.; Menear, K. A.; Newell, D. R.; Richardson, C. J.; Shea, K.; Smith, G. C. M.; Thommes, P.; Ting, A.; Griffin, R. J. 1Substituted (dibenzo[b,d]thiophen-4-yl)-2-morpholino-4H-chromen4-ones endowed with dual DNA-PK/PI3-K inhibitory activity. J. Med. Chem. 2013, 56, 6386−6401. (14) Walker, E. H.; Pacold, M. E.; Perisic, O.; Stephens, L.; Hawkins, P. T.; Wymann, M. P.; Williams, R. L. Structural determinants of phosphoinositide 3-kinase inhibition by wortmannin, LY294002, quercetin, myricetin, and staurosporine. Mol. Cell 2000, 6, 909−919. (15) Yang, H.; Rudge, D. G.; Koos, J. D.; Vaidialingam, B.; Yang, H. J.; Pavletich, N. P. mTOR kinase structure, mechanism and regulation. Nature 2013, 497, 217−223. (16) Finlay, M. R. V.; Buttar, D.; Critchlow, S. E.; Dishington, A. P.; Fillery, S. M.; Fisher, E.; Glossop, S. C.; Graham, M. A.; Johnson, T.; Lamont, G. M.; Mutton, S.; Perkins, P.; Pike, K. G.; Slater, A. M. Sulfonyl-morpholino-pyrimidines: SAR and development of a novel class of selective mTOR kinase inhibitor. Bioorg. Med. Chem. Lett. 2012, 22, 4163−4168. (17) Inhibition of mTOR kinase was evaluated in a high-throughput assay using α screen capture complex technology (Perkin-Elmer) with a recombinant truncated FLAG-tagged mTOR (amino acids 1362−2549, expressed in HEK293 cells). The activity of the lipid kinases, class I PI3Ks α, β, δ, γ was measured with the lipid PIP2 as substrate. More detailed description of the assay conditions are described in ref 27. (18) Walker, E. H.; Pacold, M. E.; Perisic, O.; Ried, C.; Stephens, L.; Williams, R. L. Structural insights into phosphoinositide 3-kinase catalysis and signalling. Nature 1999, 402, 313−320. (19) Figure 2b was generated using PyMOL: DeLano, W. L. The PyMOL Molecular Graphics System; DeLano Scientific: San Carlos, CA, U.S., 2002; http://www.pymol.org. (20) MDA-MB-468 cells were exposed for 2 h to increasing concentrations of compound. At the end of the incubation period cells were fixed, washed, and probed with antibodies against S473 pAKT or against S235/236 phosphorylated S6 (pS6). Levels of phosphorylation were assessed using an Acumen laser scanning cytometer (TTP Labtech).

(21) (a) Assessments of aqueous solubility were made after an incubation of 24 h in pH 7.4 phosphate buffer. After centrifugation, analysis of the supernatant liquid was performed by LC−UV to quantify the amount of compound in solution. Further details are contained in ref 21b. (b) Buttar, D.; Colclough, N.; Gerhardt, S.; MacFaul, P. A.; Phillips, S. D.; Plowright, A.; Whittamore, P.; Tam, K.; Maskos, K.; Steinbacher, S.; Steuber, H. A combined spectroscopic and crystallographic approach to probing drug− human serum albumin interactions. Bioorg. Med. Chem. 2010, 18, 7486−7496. (22) Finlay, M. R. V; Morris, J.; Pike, K. G. Morpholinopyrimdine derivatives, processes for preparing them, pharmaceutical compositions containing them, and their use for treating proliferative disorders. PCT Int. Appl. WO2008023159, 2008. (23) Pike, K. G. Morpholinopyrimidine derivatives that are useful in the treatment of diseases mediated by mTOR and/or PI3K enzyme and their preparation. PCT Int. Appl. WO2009007750, 2009. (24) Blades, K.; Demeritt, J.; Fillery, S.; Foote, K. M.; Greenwood, R.; Gregson, C.; Hassall, L. A.; McGuire, T. M.; Pike, K. G.; Williams, E. Expedient synthesis of biologically important sulfonylmethyl pyrimidines. Tetrahedron Lett. 2014, 55 (29), 3851−3855. (25) Pike, K. G. Pyrimidine derivatives that are useful in the treatment of diseases mediated by mTOR and/or PI3K enzyme and their preparation. PCT Int. Appl. WO2009007748, 2009. (26) Pike, K. G.; Malagu, K.; Hummersone, M. G.; Menear, K. A.; Duggan, H. M. E.; Gomez, S.; Martin, N. M. B.; Ruston, L.; Pass, S. L.; Pass, M. Optimization of potent and selective dual mTORC1 and mTORC2 inhibitors: The discovery of AZD8055 and AZD2014. Bioorg. Med. Chem. Lett. 2013, 23, 1212−1216. (27) Chresta, C. M.; Davies, B. R.; Hickson, I.; Harding, T.; Cosulich, S.; Critchlow, S. E.; Vincent, J. P.; Ellston, R.; Jones, D.; Sini, P.; James, D.; Howard, Z.; Dudley, P.; Hughes, G.; Smith, L.; Maguire, S.; Hummersone, M.; Malagu, K.; Menear, K.; Jenkins, R.; Jacobsen, M.; Smith, G. C. M.; Guichard, S.; Pass, M. AZD8055 is a potent, selective, and orally bioavailable ATP-competitive mammalian target of rapamycin kinase inhibitor with in vitro and in vivo antitumor activity. Cancer Res. 2010, 70, 288−298. (28) (a) Hopkins, A. L.; Groom, C. R.; Alex, A. Ligand efficiency: a useful metric for lead selection. Drug. Discovery Today 2004, 9, 430− 431. (b) Albert, J. S.; Blomger, N.; Breeze, A. L.; Brown, A. J. H.; Burrows, J. N.; Edwards, P. D.; Folmer, R. H. A.; Geschwinder, S.; Griffen, E. J.; Kenny, P. W.; Nowak, T.; Olssom, L.-L.; Sanganee, H.; Shapiro, A. B. An integrated approach to fragment-based lead generation: philosophy, strategy and case studies from AstraZeneca’s drug discovery programmes. Curr. Top. Med. Chem. 2007, 7, 1600− 1629. (c) Edwards, M. P.; Price, D. A. Role of physicochemical properties and ligand lipophilicity efficiency in addressing drug safety risks. Annu. Rep. Med. Chem. 2010, 45, 381−391. (d) Tarcsay, A.; Nyiri, K.; Keseru, G. M. Impact of lipophilic efficency on compound quality. J. Med. Chem. 2012, 55, 1252−1260. (e) Chiou, W. L.; Robbiel, G.; Chung, S. M.; Wul, T.-C.; Mal, C. Pharm. Res. 1998, 15, 1474. (f) Pike, K. G.; Allen, J. V.; Caulkett, P. W. R.; Clarke, D. S.; Donald, C. S.; Fenwick, M. L.; Johnson, K. M.; Johnstone, C.; McKerrecher, D.; Rayner, J. W.; Walker, R. P.; Wilson, I. Design of a potent, soluble glucokinase activator with increased pharmacokinetic half-life. Bioorg. Med. Chem. Lett. 2011, 21, 3467−3470. (g) Murray, C. W.; Erlanson, D. A.; Hopkins, A. L.; Keseru, G. M.; Leeson, P. D.; Rees, D. C.; Reynolds, C. H.; Richmond, N. J. Validity of ligand efficiency metrics. Med. Chem. Lett. 2014, 5, 616−618. (29) Zask, A.; Kaplan, J.; Verheijen, J. C.; Richard, D. J.; Curran, K.; Brooijmans, N.; Bennett, E. M.; Toral-Barza, L.; Hollander, I.; Ayral-Kaloustian, S.; Yu, K. Morpholine derivatives greatly enhance the selectivity of mammalian target of rapamycin (mTOR) inhibitors. J. Med. Chem. 2009, 52, 7942−7945. (30) Foote, K. M.; Blades, K.; Cronin, A.; Filery, S.; Guichard, S. S.; Hassall, L.; Hickson, I.; Jacq, X.; Jewsbury, P. J.; McGuire, T. M.; Nissink, J. W. M.; Odedra, R.; Page, K.; Perkins, P.; Suleman, A.; Tam, K.; Thommes, P.; Broadhurst, R.; Wood, C. Discovery of 4-{4[(3R)-3-methylmorpholin-4-yl]-6-[1-(methylsulfonyl)cyclopropyl]W

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX

Article

Journal of Medicinal Chemistry pyrimidin-2-yl}-1H-indole (AZ20): a potent and selective inhibitor of ATR protein kinase with monotherapy in vivo antitumor activity. J. Med. Chem. 2013, 56, 2125−2138. (31) Ganellin, R. 1980 Award in medicinal chemistry. Medicinal chemistry and dynamic structure−activity analysis in the discovery of drugs acting at histamine H2 receptors. J. Med. Chem. 1981, 24, 913−920. (32) (a) Free, S. M., Jr.; Wilson, J. W. A mathematical contribution to structure−activity studies. J. Med. Chem. 1964, 7, 395−399. (b) Freeman-Cook, K. D.; Amor, P.; Bader, S.; Buzon, L. M.; Coffey, S. B.; Corbett, J. W.; Dirico, K. J.; Dorn, S. D.; Elliott, R. L.; Esler, W.; Guzman-Perez, A.; Henegar, K. E.; Houser, J. A.; Jones, C. S.; Limberakis, C.; Loomis, K.; McPherson, K.; Murdande, S.; Nelson, K. L.; Phillion, D.; Pierce, B. S.; Song, W.; Sugarman, E.; Tapley, S.; Tu, M.; Zhao, Z. Maximising lipophilic efficiency: the use of Free− Wilson analysis in the design of inhibitors of acetyl-CoA carboxylase. J. Med. Chem. 2012, 55, 935−942. (c) Goldberg, F. W.; Leach, A. G.; Scott, J. S.; Snelson, W. L.; Groombridge, S. D.; Donald, C. S.; Bennett, S. N. L.; Bodin, C.; Morentin Gutierrez, P.; Gyte, A. C. Free−Wilson and structural approaches to co-optimizing human and rodent isoform potency for 11β-hydroxysteriod dehydrogenase type 1 (11β-HSD1) inhibitors. J. Med. Chem. 2012, 55, 10652−10661. (33) Waring, M. J.; Johnstone, C.; McKerrecher, D.; Pike, K. G.; Robb, G. Matrix-based multiparameter optimisation of glucokinase activators: the discovery of AZD1092. Med. Chem. Commun. 2011, 2, 775−779. (34) (a) Monolayers of MDCKII (Madin−Darby canine kidney cells) cells transfected with the human MDR1 (multidrug resistance 1) transporter protein were cultured in Transwells and used to study the permeability and efflux potential of compounds at a concentration of 10 μM.. (b) Keogh, J. P.; Kunta, J. R. Development, validation and utility of an in vitro technique for assessment of potential clinical drug−drug interactions involving P-glycoprotein. Eur. J. Pharm. Sci. 2006, 27, 543−554. (35) Mouse clearance values were obtained following an iv dose of 2.5−10 μmol/kg to CD-1 outbred mice, and dog clearance values were obtained following an iv dose of 1−3 μmol/kg to Beagle dogs. The compounds were dosed as a solution, and the clearance values are quoted in mL min−1 kg−1. Bioavailability values were calculated following an oral dose of 5−20 μmol/kg in mouse and 3−10 μmol/ kg in dog. (36) Redfern, W. S.; Carlsson, L.; Davis, A. S.; Lynch, W. G.; MacKenzie, I.; Palethorpe, S.; Siegl, P. K. S.; Strang, I.; Sullivan, A. T.; Wallis, R.; Camm, A. J.; Hammond, T. G. Relationships between preclinical cardiac electrophysiology, clinical QT interval prolongation and torsade de pointes for a broad range of drugs: evidence for a provisional safety margin in drug development. Cardiovasc. Res. 2003, I, 32−45. (37) The plasma protein binding of compounds was measured using equilibrium dialysis technique and is quoted as the fraction unbound ( f u). A more detailed description of the equilibrium dialysis assay conditions are described in Chiou, W. L.; Robbiel, G.; Chung, S. M.; Wul, T.-C.; Mal, C. Correlation of plasma clearance of 54 extensively metabolized drugs between humans and rats: mean allometric coefficient of 0.66. Pharm. Res. 1998, 15, 1474−1479.

X

DOI: 10.1021/jm501778s J. Med. Chem. XXXX, XXX, XXX−XXX