Abstract
Receptor interacting protein kinase-2 (RIPK2) is an enzyme involved in the transduction of pro-inflammatory nucleotide-binding oligomerization domain (NOD) cell signaling, a pathway implicated in numerous chronic inflammatory conditions. Herein, a pyrido[2,3-d]pyrimidin-7-one based class of RIPK2 kinase and NOD2 cell signaling inhibitors is described. For example, 33 (e.g. UH15-15) inhibited RIPK2 kinase (IC50 = 8 ± 4 nM) and displayed > 300-fold selectivity versus structurally related activin receptor-like kinase 2 (ALK2). This molecule blocked NOD2-dependent HEKBlue NF-κB activation (IC50 = 20 ± 5 nM) and CXCL8 production (at concentrations > 10 nM). Molecular docking suggests that engagement of Ser25 in the glycine-rich loop may provide increased selectivity versus ALK2 and optimal occupancy of the region between the gatekeeper and the αC-helix may contribute to potent NOD2 cell signaling inhibition. Finally, this compound also demonstrated favorable in vitro ADME and pharmacokinetic properties (e.g. Cmax = 5.7 μM, Tmax = 15 min, t1/2 = 3.4 h and Cl = 45 mL/min/kg following single 10 mg/kg intraperitoneal administration) further supporting the use of pyrido[2,3-d]pyrimidin-7-ones as a new structure class of RIPK2 kinase and NOD cell signaling inhibitors.
Keywords: Inhibitor; Kinase; NOD; Nucleotide-binding oligomerization domain; Pyrido[2,3-d]pyrimidin-7-one; Receptor-interacting protein kinase 2; RIPK2
Graphical Abstract

1. Introduction
Receptor interacting protein kinase-2 (RIPK2), also known as RICK/CARDIAK/CARD3, is a dual serine/threonine and tyrosine kinase that plays a pivotal role in signal transduction from nucleotide-binding oligomerization containing proteins 1/2 (NOD1/2) [1, 2]. These two proteins are cytosolic receptors and part of the pattern recognition receptor family along with Toll-like receptors that cause activation of nuclear factor κ-light chain enhancer of activated B cells (NF-κB) in immune cells after bacterial invasion. These cytosolic proteins detect bacterial peptidoglycan fragments, namely diaminopimelic acid (DAP) and muramyl dipeptide (MDP), present in Gram-positive and Gram-negative bacteria [3]. Upon activation, NOD1/2 binds to RIPK2 through its caspase activation and recruitment domain (CARD) propagating pro-inflammatory signaling [4]. RIPK2 undergoes autophosphorylation of its kinase domain and binds to X-linked inhibitor of apoptosis proteins (XIAP) and other E3 ubiquitin ligases for non-degradative polyubiquitination [5]. Polyubiquitinated RIPK2 then activates transforming growth factor beta-activated kinase 1 (TAK1) and IκB kinase complex (IKK) causing upregulation of mitogen-activated protein kinases (MAPK) and NF-κB. Activation of NF-κB results in transcription of multiple pro-inflammatory cytokine genes [6].
Normal regulation of the NOD1/2-RIPK2 signaling cascade is beneficial in providing defense against invading bacteria. But dysregulation, including a genetic mutations in NOD1/2, may cause excessive RIPK2 activation and thereby contribute to inflammation in multiple conditions, such as inflammatory rheumatoid arthritis and Blau syndrome and in neuroinflammatory conditions (e.g. multiple sclerosis) [7, 8]. Small molecule pharmacological probes for RIPK2/NOD signaling would be useful for furthering the understanding of this pathway and may provide a foundation for therapeutic development.
Several RIPK2 kinase inhibitors have been reported in the literature, including SB203580 (1) [9] , Gefitinib (2) [10], OD36 (3) and OD38 (4) [11], WEHI-345 (5) [12], Ponatinib (6) [13], GSK583 (7) [14], 8a and its pro-drug 8b [15], 9 and 10 [16], and 11 [17] (Figure 1). Recently, our laboratory identified another class of potent RIPK2/NOD signaling inhibitors based on a 3,5-diaryl-2-aminopyridine scaffold, exemplified by CSLP37 (12) [18]. This latter class of compounds was shown to disrupt XIAP-RIPK2 interactions mitigating NOD2 inflammatory signaling [19]. Furthermore, these studies demonstrated that impairing RIPK2 ubiquitination was critical for functional activity, as opposed to blocking catalysis per se. Herein, we describe a structurally distinct class of pyrido[2,3-d]pyrimidin-7-one RIPK2 inhibitors. Importantly, optimization of the solvent exposed region and appropriate occupancy of the region between the gatekeeper and the αC-helix were necessary to achieve potent NOD2 cell signaling inhibition as assessed in NF-κB activation HEKBlue and pro-inflammatory CXCL8 production assays. These compounds will contribute to the collective molecular toolset of RIPK2/NOD signaling inhibitors for further elucidation of this emerging therapeutic paradigm.
Figure 1.

Previously reported RIPK2 kinase inhibitors.
2. Compound design and synthesis
During an activin receptor-like kinase 2 (ALK2) screening campaign, the pyrido[2,3-d]pyrimidin-7-one PD180970 (13) was identified as a modest inhibitor (ALK2 IC50 ~ 1 μM). This compound has previously been reported to bind Abl kinase in a conformational state intermediate between DFG-in and DFG-out [20]. Addition of a solubilizing group as in PD166285 (14) [21] provided potent ALK2 inhibition (IC50 = 0.021 ± 0.016 μM) (Figure 2A). We and others have found that various ALK2 inhibitors oftentimes demonstrate inhibitory activity against RIPK2 [22, 23]. In the case of 14, RIPK2 was indeed potently inhibited (IC50 = 0.013 ± 0.004 μM). In addition, the compound blocked NOD2 cell signaling (IC50 = 0.037 ± 0.004 μM) in the HEKBlue assay of NOD2-dependent NF-κB activation in response to L18-MDP stimulation. Encouraged by these results, a structure-activity relationship (SAR) analysis was conducted examining several regions of the pyrido[2,3-d]pyrimidin-7-one scaffold (Figure 2B) assessing RIPK2 and ALK2 kinase inhibitory activities, as well inhibition of NOD2 cell signaling via the HEKBlue assay. One specific direction of the SAR study was to introduce a sulfone as a hydrogen bond acceptor to engage Ser25 in the glycine-rich loop of RIPK2 that is only present in three other kinases [24], as was previously done with 7 – 10 [14 - 16].
Figure 2.
(A) Structures of 13 and 14, as well as a summary of ALK2, RIPK2 kinase and NOD cell signaling inhibitory data. (B) Regions of the pyrido[2,3-d]pyrimidin-7-one scaffold explored for SAR analysis.
Compounds used for the SAR analysis were synthesized using 2-(methylthio)-6-phenylpyrido[2,3-d]pyrimidin-7-one intermediates that were generated via two different routes. For the first method, the synthesis of the 6-(2,4-dichlorophenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9a) started with the commercially available ethyl-4-chloro-2-(methylthio)pyrimidine-5-carboxylate (A1) as shown in Scheme 1. This material was either treated with ammonium hydroxide in the presence of triethylamine (TEA) to furnish A2 in 98% yield or with aqueous methylamine to generate A3 in 85% yield. The ethyl esters were reduced in the presence of lithium aluminum hydride (LAH) to provide the corresponding alcohols A4 and A5 in 61% and 95% yield, respectively. The alcohols were then treated with manganese dioxide to obtain the aldehydes A6 and A7 in 83% and 95% yield, respectively. Oxidation of alcohol A5 to A7 was also performed using Dess-Martin periodinane, but the yield was lower (i.e. 56%). Condensation of A7 with different phenylacetonitriles in the presence of potassium carbonate generated pyrido[2,3-d]pyrimidine-7-imines A8a-c in 54-63% yield. The imines were then converted to the corresponding pyrido[2,3-d]pyrimidin-7-ones A9a-c in 55-65% yields through sequential N-acetylation with acetic anhydride followed by hydrolysis with concentrated hydrochloric acid (HCl) [25, 26]. Although this method produced several of the desired pyrido[2,3-d]pyrimidine-7-ones, it was not particularly efficient.
Scheme 1. Synthesis of 2-(methylthio)-6-phenylpyrido[2,3-d]pyrimidin-7-one - intermediates A9a-c.

* Reagents and conditions: (a) A2: NH4OH, TEA, THF, rt, 2 h (98%), A3: CH3NH2 (aq), THF, rt, 2 h (85%); (b) LAH, THF, rt, 1.5 h (60-95%); (c) MnO2, DCM, rt, overnight (83-95%); (d) PhCH2CN, K2CO3, DMF, 105 °C, 18 h (54-63%); (e) i) Ac2O, 139 °C, 30 min, ii) conc. HCl, 100 °C, 5 min (55-65% for two steps).
Alternatively, pyrido[2,3-d]pyrimidine-7-ones were synthesized by condensing A6 or A7 with substituted phenylacetate esters in the presence of KF/Al2O3 to furnish A9d-j in 30-60% yield, as shown in Scheme 2 [27]. Furthermore, intermediate A10 was deprotonated with sodium hydride (NaH) and then exposed to different alkyl iodides to furnish A9m-t in 28-74% yield. The 4-hydroxyphenyl derivative A9j was treated with t-butyldimethylsilyl chloride in the presence of dimethylaminopyridine (DMAP) and imidazole or methylated with methyl iodide in the presence of potassium carbonate to afford A9k and A9l in 88% and 95% yield, respectively.
Scheme 2. Synthesis of 2-(methylthio)-6-phenylpyrido[2,3-d]pyrimidin-7-one intermediates A9d-t.

* Reagents and conditions: (a) KF/Al2O3, DMA, rt, 3-4 h (23-60%); (b) R2I, NaH, DMF, 50 °C, 1 h (28-74%); (c) TBSCl, DMAP, imidazole, DMF, 0 °C to rt, overnight (88%); (d) MeI, K2CO3, acetone, reflux, 8 h (95%).
The various 2-(methylthio)-6-phenylpyrido[2,3-d]pyrimidin-7-ones A9a-i and A9k-t were oxidized using m-chloroperbenzoic acid (m-CPBA) to afford methyl sulfones A11a-i and A11k-t in 40-93% yield (Scheme 3). Initial attempts to displace the methyl sulfones with anilines did not furnish the desired products. Alternatively, the anilines A12a-c were converted to their corresponding formamides A13a-c by heating in formic acid, then deprotonated with sodium hydride followed by treatment with methyl sulfones to generate the desired compounds in 33-95% yield [28].
Scheme 3. Synthesis of 6-phenyl-2-(phenylamino)pyrido[2,3-d]pyrimidin-7-ones 15 – 37 and 39-40 *.

* Reagents and conditions: (a) Pd/C (10%), H2 (1 atm), CH3OH, rt, overnight (96%); (b) HCO2H, rt or 60 °C, overnight (79-80%); (c) m-CPBA, DCM, rt, 3-5 h (40-93%); (d) A13a, A13b or A13c, NaH, THF:DMF (1:1), 0 °C to rt, 2.5 h (33-95%). Numbering of the phenylpyrido[2,3-d]pyrimidin-7-one ring system present in compounds 15 – 40 is indicated.
In a case where requisite phenylacetate esters were not readily available, an alternative synthetic method was pursued as illustrated in Scheme 4. Intermediate A7 was treated with ethyl 2-chloro-2-(diethoxyphosphoryl)acetate to give A14 in 42% yield [29]. This was followed by coupling A14 with the commercially available 2-chloro-4-methylphenylboronic acid under Suzuki conditions to afford A15 in a 57% yield. Furthermore, oxidation of A15 with m-CPBA gave A16 in 68% yield, which upon treatment with the anion of A13c generated in the presence of NaH gave 38 in 75% yield.
Scheme 4. Synthesis of 38 *.

* Reagents and conditions: (a) A7, NaH, THF, reflux, 2.5 h (42%); (b) 2-Cl-4-MePhB(OH)2, Pd(PPh3)4, Na2CO3, DMF, ACN, 90 °C, 5 h (57%); (c) m-CPBA, DCM, rt, 5 h (68%); (d) A13c, NaH, THF, DMF, 0 °C to rt, 2.5 h (75%).
3. Results and discussion
To increase the selectivity for RIPK2 versus ALK2 while maintaining cellular potency, the hydrophobic substitutions (R1) at different positions on the phenyl ring, which projects towards the glutamate residue of the α-C helix in the docking model, was initially explored. In addition, this region of the kinase appears to be more flexible for RIPK2 since it has been demonstrated to adopt a Glu-out conformation [30], whereas ALK2 seems to be less accommodating of this conformation as evidenced by only DFG-in/Glu-in inhibitors having been reported to date [23, 31]. Gratifyingly, 15, having 2,4-di-Cl substitution, showed improved selectivity towards RIPK2 (Table 1). The absence of the 6-Cl substituent perhaps allow for optimal torsional rotation of the phenyl group favoring RIPK2. However, this compound showed a 4-fold decrease in RIPK2 cellular potency compared to 14. Removal of the 4-Cl (16) resulted in potent RIPK2 enzyme and NOD cell signaling inhibition, but poor ALK2 selectivity. Having only a substituent in the 4-position, including Cl (17), t-Bu (18), OH (19) or OMe (20), was well tolerated for RIPK2 enzyme inhibition, but resulted in loss of selectivity, cell signaling inhibition or both.
Table 1.
RIPK2 and ALK2 kinases, and NOD2 cell (HEKBlue) signaling inhibitory activities of 14 and 15 – 20.
![]() | ||||
|---|---|---|---|---|
| Compound | R1 | Kinase IC50 (μM) | RIPK2/NOD2 Cell Assay IC50 (μM) |
|
| RIPK2 | ALK2 | |||
| 14 | 2,6-di-Cl | 0.013 ± 0.004 | 0.021 ± 0.016 | 0.037 ± 0.004 |
| 15 | 2,4-di-Cl | 0.016 ± 0.0036 | NI | 0.16 ± 0.024 |
| 16 | 2-Cl | 0.017 ± 0.008 | 0.062 ± 0.080 | 0.004 ± 0.0005 |
| 17 | 4-Cl | 0.013 ± 0.0006 | 2.940 ± 1.633 | 0.95 ± 0.35 |
| 18 | 4-t-Bu | 0.105 ± 0.047 | NI | 1.98 ± 1.73 |
| 19 | 4-OH | 0.025 ± 0.014 | 0.059 ± 0.065 | 0.005 ± 0.002 |
| 20 | 4-OMe | 0.021 ± 0.0016 | NI | 0.51 ± 0.12 |
NI: No inhibition observed at 100 μM; IC50 values are shown as the mean of two or more determinations ± standard deviation.
In order to better balance enzyme and cell signaling inhibition with selectivity versus ALK2, the R2 position on the central heterocycle was explored. Although this group projects towards the glycine-rich loop, the region appears accommodating for larger and more polar substituents. Consequently, an array of groups were introduced (21 – 28) into this region of the pyrido[2,3-d]pyrimidine-7-one scaffold. In general, they were well tolerated with regard to maintaining potent RIPK2 and NOD cell signaling inhibition, but offered no selectivity improvement versus ALK2 (Table 2).
Table 2.
RIPK2 and ALK2 kinases, and NOD2 cell (HEKBlue) signaling inhibitory activities of 21- 28.
![]() | |||||
|---|---|---|---|---|---|
| Compound | R1 | R2 | Kinase IC50 (μM) | RIPK2/NOD2 Cell Assay IC50 (μM) |
|
| RIPK2 | ALK2 | ||||
| 21 | H | Me | 0.0105 ± 0.002 | 0.284 ± 0.356 | 0.014 ± 0.004 |
| 22 | H | Et | ~ 30 nM | 0.004 ± 0.005 | 0.015 ± 0.005 |
| 23 | H | CH2CH2OH | ~ 30 nM | 0.018 ± 0.024 | 0.27 ± 0.22 |
| 24 | 2,6-di-Cl | CH2CH2OMe | 0.128 ± 0.001 | 0.018 ± 0.019 | 0.013 ± 0.007 |
| 25 | 2,6-di-Cl | CH2Ph | 0.021 ± 0.003 | 0.008 ± 0.004 | 0.009 ± 0.003 |
| 26 | 2,6-di-Cl | i-Bu | 0.005 ± 0.002 | 0.009 ± 0.005 | 0.003 ± 0.0005 |
| 27 | 2,6-di-Cl | CH2Ph-3-SO2Me | 0.025 ± 0.008 | 0.092 ± 0.005 | 0.005 ± 0.005 |
| 28 | 2,6-di-Cl | CH2Ph-4-SO2Me | 0.021 ± 0.007 | ND | 0.094 ± 0.004 |
ND: Not determined; IC50 values are shown as the mean of two or more determinations ± standard deviation, unless otherwise indicated.
Next, the effect of R3 substituents on RIPK2 selectivity was investigated. This region of the inhibitor is expected to project towards solvent, but also in the vicinity of Ser25. This residue, which is rather unique to RIPK2, has previously been targeted for engagement as a means to improve selectivity [14]. Incorporation of a hydrogen bond acceptor, such as a sulfone, at either the 3- or 4-positions of the phenyl might provide such an interaction. Indeed, this strategy proved to be effective (Table 3) as it provided 10-fold and 160-fold increase in RIPK2 selectivity versus ALK2 for the 4- (29) and 3-methyl sulfone (30), respectively. Furthermore, the 3-methyl sulfone 30 retained excellent cellular inhibition. Similar to previous observations, introduction of a substituent in the 4-position for the R1 group projecting towards the α-C helix, as in 31 and 32, resulted in loss of cell activity, but not RIPK2 enzyme inhibition. Surprisingly, 33, which is a mono-substituted phenyl version of 30, showed further improvement (310-fold) in RIPK2 selectivity versus ALK2 and excellent cellular potency in the HEKBlue assay. Several other substitution arrangements on the phenyl ring projecting towards the α-C helix, including the 3-, 4- and 5-positions (34 – 40), were detrimental to cellular activity, despite maintaining potent RIPK2 enzyme inhibition.
Table 3.
RIPK2 and ALK2 kinases, and NOD2 cell (HEKBlue) signaling inhibitory activities of methylsulfones 29 – 40.
![]() | ||||||
|---|---|---|---|---|---|---|
| Compound | R1 | R2 | R3 | Kinase IC50 (μM) | RIPK2 Cell Assay IC50 (μM) |
|
| RIPK2 | ALK2 | |||||
| 29 | 2,6-di-Cl | Me | 4-SO2Me | 0.014 ± 0.0001 | 0.136 ± 0.118 | 0.136 ± 0.016 |
| 30 | 2,6-di-Cl | Me | 3-SO2Me | 0.006 ± 0.0001 | 0.972 ± 0.510 | 0.022 ± 0.018 |
| 31 | 4-Cl | Me | 3-SO2Me | 0.0146 ± 0.005 | NIa | NIb |
| 32 | 4-Cl | i-Bu | 3-SO2Me | 0.0647 ± 0.024 | NIa | NIb |
| 33 | 2-Cl | Me | 3-SO2Me | 0.008 ± 0.004 | 2.516 ± 2.168 | 0.020 ± 0.005 |
| 34 | 2,4-diCl | Me | 3-SO2Me | 0.027 ± 0.013 | NIa | 0.878 ± 0.23 |
| 35 | 2,3-di-Cl | Me | 3-SO2Me | 0.034 ± 0.011 | NIa | 1.23 ± 0.156 |
| 36 | 2-Cl, 4-F | Me | 3-SO2Me | 0.017 ± 0.001 | NIa | 0.218 ± 0.049 |
| 37 | 2,5-di-Cl | Me | 3-SO2Me | 0.022 ± 0.0007 | ND | 0.73 ± 0.32 |
| 38 | 2-Cl, 4-Me | Me | 3-SO2Me | 0.015 ± 0.002 | ND | 0.48 ± 0.04 |
| 39 | 4-OH | Me | 3-SO2Me | 0.016 ± 0.001 | 24.80 | NIb |
| 40 | 4-OMe | Me | 3-SO2Me | 0.011 ± 0.004 | NIa | NIb |
NI: No inhibition observed at 100 μM
NI: No inhibition observed at 10 μM; ND: Not determined; IC50 values are shown as the mean of two or more determinations ± standard deviation.
In order to further evaluate cellular activities of the pyrido[2,3-d]pyrimidin-7-one series of RIPK2 kinase inhibitors, several representative compounds were assessed in an assay of chemokine CXCL8 positive U2OS/NOD2 cells in response to L18-MDP stimulation [19]. Four potent inhibitors of RIPK2 kinase activity with variable potency in the HEKBlue assay were selected for testing. Compounds 16 and 33 showed potent inhibition of CXCL8 positive cells compared to 17 and 31 (Figure 3). However, these compounds were inactive in RIPK2 T95W gatekeeper mutant U2OS/NOD2 cells confirming that blocking NF-κB mediated CXCL8 production was mediated through RIPK2 engagement (see supporting information, Figure S1). Compound 33 also demonstrated only marginal activity (IC50 > 4 μM) blocking RIPK1- and RIPK3-dependent necroptosis in human U937 monocytes (Figure S2, [32]) suggesting ~3-orders of magnitude difference in potency in inhibiting cellular RIPK2 signaling versus its two most closely related homologues [33].
Figure 3:
Intracellular flow cytometry analysis of CXCL8 in U2OS/NOD2 cells treated with L18-MDP (200 ng/mL, 4 h). Percentage of CXCL8 positive cells in the presence of inhibitors 16, 17, 31 and 33 at various concentrations versus DMSO control shown. Data points are the average of three experiments.
Molecular docking studies were performed using AutoDock 4.2 (with tools version 1.5.6) software. Inhibitor 14 was docked into the ATP binding site of the kinase domain of RIPK2 using the apo structure (PDB: 5AR2) [30]. The protein was kept rigid, but the ligand was allowed partial flexibility by setting the number of rotatable bonds. The top five poses showed a clustered root mean squared deviation (RMSD) < 2 Å and the binding energy for the top pose was −8.73 kcal/mol (Figure 4A). Importantly, two hydrogen bonds (both 1.8 Å) involving the ligand NH and N-3 with the backbone of M98 in the hinge were observed. The 2,6-dichlorophenyl projected towards a region between the gatekeeper and the αC-helix, while the aminoether on the aniline was directed towards the solvent exposed region and the glycine-rich loop
Figure 4.
(A) Molecular docking model of 14 (cyan) with RIPK2 (PDB:5AR2). Hydrogen bonding interactions (1.8 Å) between the inhibitor and hinge residue M98, as well as the salt-bridge between K47 and E66 of the αC-helix are shown. Also highlighted is the DFG motif. (B) Molecular docking model of 33 (green) with RIPK2 (PDB:5AR2). Hydrogen bonding interaction (3.2 Å) between the inhibitor and S25 is indicated.
Based on excellent selectivity versus ALK2, lack of necroptosis inhibition, and potent NOD cell signaling inhibition, 33 was similarly docked with RIPK2 (PDB: 5AR2). The molecular docking model (Figure 4B, binding energy of −9.07 kcal/mol) resembles the pose of 14 with respect to the hinge interaction and projection of the 2-chlorophenyl towards a region between the gatekeeper and the αC-helix. In addition, the sulfone forms a H-bonding interaction with Ser25 (3.2 Å) in the glycine-rich loop. This model also resembles the RIPK2•7 co-crystal structure (PDB: 5J7B) [14], except interaction with the backbone of Asp164 in the DFG motif was not observed.
For a previous class of RIPK2 inhibitors (e.g. 12), we proposed that appropriate occupancy of the region between the gatekeeper and the αC-helix was necessary to achieve NOD cell signaling inhibition, but not required for disruption of RIPK2 kinase activity [18, 19]. The pyrido[2,3-d]pyrimidin-7-ones series displayed similar discordance between NOD2 signaling and RIPK2 inhibitory activity with the determinant again being the phenyl directed towards the region between the gatekeeper and the αC-helix. To further explore this observation, three derivatives with different substituents on the phenyl (e.g. 31, 33 and 34) demonstrating potent RIPK2 kinase inhibition but an array of potencies for NOD2 signaling inhibition were docked to RIPK2. In all three cases, similar docking poses showed interactions with hinge residue M98 and Ser25, which was consistent with the observed selectivity versus ALK2 (Figure S3A). The phenyl directed towards the region between the gatekeeper and αC-helix was slightly shifted for 31 and 34 possibility due to unfavorable interaction of the 4-Cl with E66 of the αC-helix. In poses where H-bonding to Ser25 was disrupted due to rotation of the phenylsulfone, this shift was even greater (Figure S3B). Overall, only 33 maintains the positioning of the 2-chlorophenyl in the sub-pocket between the αC-helix and T95 gatekeeper, which is critical for the inhibition of NOD2 signaling [19] independent of H-bonding to Ser25 (Figure S3C). The robustness of the binding provides a possible explanation for the improved inhibition of NOD2 cell signaling by 33 compared to the other two compounds.
Finally, several in vitro ADME as well as in vivo pharmacokinetic and brain permeability properties of 33 were assessed. The inhibitor demonstrated relatively good stability in mouse liver microsomes (t1/2 = 19.7 min; Clint= = 35 μL/min/mg), aqueous kinetic solubility at pH 7.4 of 0.7 μM, and good permeability (PAMPA Papp = 23.6 × 10−6 cm/s). A pharmacokinetic study in female ICR mice (N=18) following a 10 mg/kg single intraperitoneal administration [formulated with DMSO (5%), Solutol® (10%) and water (85%)] achieved maximum plasma concentration (Cmax) of 5.7 μM, time to maximum plasma concentration (Tmax) of 15 min, plasma elimination half-life (t1/2) of 3.4 h and clearance (Cl) of 45 mL/min/kg. Brain concentrations of 2.45 μM and 0.32 μM were also observed at 30 min and 2 h post-administration.
4. Conclusions
In conclusion, a new class of pyrido[2,3-d]pyrimidin-7-one based RIPK2 kinase inhibitors was discovered. In addition, optimal substitution of the phenyl group directed towards the region between the gatekeeper and the αC-helix as well as the 3-methyl sulfone on the phenyl ring extending towards the solvent-exposed region with possible engagement of Ser25 in the glycine-rich loop provides increased selectivity versus ALK2 and potent NOD2 cell signaling inhibition. For example, a representative compound 33 demonstrated both potent inhibition of RIPK2 kinase (IC50 = 8 ± 4 nM), > 300-fold selectivity versus ALK2, as well as blocked HEKBlue NF-κB activation (IC50 = 20 ± 5 nM) and L18-MDP stimulated CXCL8 production (at concentrations > 10 nM and was eliminated in RIPK2 T95W mutant U2OS/NOD2 cells). In addition, this compound has in vitro ADME and pharmacokinetic characteristics (e.g. Cmax = 5.7 μM, Tmax = 15 min, t1/2 = 3.4 h and Cl = 45 mL/min/kg following single 10 mg/kg intraperitoneal administration) further supporting use of pyrido[2,3-d]pyrimidin-7-ones (e.g. 33, a.k.a. UH15-15) as a new structure class of RIPK2 kinase and NOD cell signaling inhibitors.
5. Experimental section
All reactions were carried out under an argon atmosphere with dry solvents unless otherwise stated. All commercially available chemicals and reagent grade solvents were used directly without further purification unless otherwise specified. Reactions were monitored by thin-layer chromatography (TLC) on Baker-flex® silica gel plates (IB2-F) using UV-light (254 and 365 nm) as a visualizing agent and either an ethanolic solution of phosphomolybdic acid or ninhydrin solution and heat as developing agents. Flash chromatography was conducted on silica gel (230–400 mesh) using Teledyne ISCO CombiFlash® Rf. Melting points were measured using a Thomas Hoover Uni-Melt capillary melting point apparatus. NMR spectra were recorded at room temperature using a JEOL ECA-500 (1H NMR at 400, 500 and 600 MHz and 13C NMR at 100, 125 and 150 MHz) with tetramethylsilane (TMS) as an internal standard. Chemical shifts (δ) are given in parts per million (ppm) with reference to solvent signals [1H-NMR: CDCl3 (7.26 ppm), CD3OD (3.30 ppm), DMSO-d6 (2.50 ppm); 13C-NMR: CDCl3 (77.0 ppm), CD3OD (49.0 ppm), DMSO-d6 (39.5 ppm)]. Signal patterns are reported as s (singlet), d (doublet), t (triplet), q (quartet), dd (doublet of doublets), td (triplet of doublets), m (multiplet) and brs (broad singlet). Coupling constants (J) are given in Hz. High-resolution mass spectra (HRMS) were carried out using Agilent 6530 Q-TOF instrument by the mass spectrometry facility at the Department of Chemistry, University of Texas at Austin. Electrospray ionization (ESI) were used as ionization source and the spectra were reported as m/z (relative intensity) for the molecular [M] or [M + H]+ ion species. The purity of the final compounds was determined to be ≥ 95% by analytical high-performance liquid chromatography (HPLC) using binary HPLC pump (Waters) and Kinetex 5 μm C18 100A column (250 × 4.6 mm). UV absorption was monitored at λ = 254 nm. The injection volume was 15 μL. The HPLC gradient of acetonitrile/water (both containing 0.1% trifluoroacetic acid) went from 2:98 to 90:10 with a total run time of 30 min and a flow rate of 1 mL/min.
Ethyl 4-amino-2-(methylthio)pyrimidine-5-carboxylate (A2).
To a solution of ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate (A1) (100 mg, 0.43 mmol) in dry THF (2 mL) was added triethylamine (0.2 mL, 1.29 mmol) and ammonium hydroxide (0.5 mL). The resulting mixture was stirred at room temperature (rt) for 2 h. After evaporation in vacuo to remove THF, the crude mixture was partitioned between water and EtOAc. The organic layer was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (15 % EtOAc/hexane) to afford A2 (90 mg, 98%) as a white solid. 1H NMR (400 MHz, CDCl3) δ 8.65 (s, 1H), 7.81 (s, 1H), 6.05 (s, 1H), 4.29 (q, J = 7.2 Hz, 2H), 2.46 (s, 3H), 1.32 (t, J = 7.1 Hz, 3H). 13C NMR (100 MHz, CDCl3) δ 176.1, 166.4, 161.9, 158.9, 101.1, 61.0, 14.3, 14.1.
Ethyl 4-(methylamino)-2-(methylthio)pyrimidine-5-carboxylate (A3).
Aqueous methyl amine (6 mL) was added to the solution of ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate (A1) (3.0 g, 12.93 mmol) in dry THF (20 mL) and the mixture was stirred at rt for 2 h. After evaporation in vacuo to remove THF, the crude mixture was then partitioned between H2O and EtOAc. The organic layer was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (10 % EtOAc/hexane) to afford A3 (2.5 g, 85%) as a white solid. 1H NMR (400 MHz, CDCl3) δ 8.59 (s, 1H), 8.16 (s, 1H), 4.29 (q, J = 7.0 Hz, 2H), 3.06 (d, J = 5.0 Hz, 3H), 2.53 (s, 3H), 1.35 (t, J = 7.1 Hz, 3H). 13C NMR (100 MHz, CDCl3) δ 176.1, 167.1, 160.8, 158.2, 101.0, 60.9, 27.4, 14.3, 14.3.
(4-Amino-2-(methylthio)pyrimidin-5-yl)methanol (A4).
A solution of A2 (450 mg, 2.11 mmol) in THF (2 mL) was added dropwise to the suspension of LiAlH4 (120 mg, 3.16 mmol) in THF (4 mL) at 0 °C and the resulting mixture was then allowed to stir at rt for 30 min. The reaction mixture was cooled at 0 °C and 15% NaOH (0.5 mL) and water (1 mL) was added dropwise. The reaction mixture was then stirred for 1 h, filtered and washed with EtOAc. Evaporation to remove EtOAc in vacuo afforded A4 (220 mg, 61%) as a light-yellow solid, which was used in the next step without purification. 1H NMR (400 MHz, CD3OD) δ 7.84 (s, 1H), 4.45 (s, 2H), 2.47 (s, 3H). 13C NMR (100 MHz, CD3OD) δ 170.4, 162.3, 152.6, 111.9, 58.5, 12.6.
(4-(Methylamino)-2-(methylthio)pyrimidin-5-yl)methanol (A5).
This compound was synthesized in a similar manner as A4 to give a pale yellow solid (95% yield). 1H NMR (400 MHz, CD3OD) δ 7.72 (s, 1H), 4.42 (s, 2H), 3.01 (s, 3H), 2.51 (s, 3H).
4-Amino-2-(methylthio)pyrimidine-5-carbaldehyde (A6).
MnO2 (670 mg, 7.71 mmol) was added to the solution of A4 (220 mg, 1.28 mmol) in DCM (5 mL) and the resulting mixture was stirred overnight at rt under argon. The reaction mixture was then filtered, concentrated to remove DCM and purified by column chromatography on silica gel (30 % EtOAc/hexane) to afford A6 (180 mg, 83%) as a light yellow solid. 1H NMR (400 MHz, CDCl3) δ 9.77 (s, 1H), 8.41 (s, 1H), 8.19 (s, 1H), 5.81 (s, 1H), 2.54 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 190.7, 177.6, 162.9, 160.4, 109.4, 14.3.
4-(Methylamino)-2-(methylthio)pyrimidine-5-carbaldehyde (A7).
This compound was synthesized in a similar manner as A6 to give a pale yellow solid (77% yield). 1H NMR (400 MHz, CDCl3) δ (ppm) 9.69 (s, 1H), 8.55 (s, 1H), 8.29 (s, 1H), 3.11 (d, J = 5.0 Hz, 3H), 2.56 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 190.2, 177.6, 162.8, 159.5, 109.5, 27.2, 14.4.
General procedure for the preparation of pyrimidin-7(8H)-imines (A8a-c). Exemplified for 6-(2,4-dichlorophenyl)-8-methyl-2-(methylthio)pyrido[2,3-d] pyrimidin-7(8H)-imine (A8a).
To a mixture of 4-(methylamino)-2-(methylthio)pyrimidine-5-carbaldehyde (A7) (250 mg, 1.37 mmol), 2-(2,4-dichlorophenyl)acetonitrile (381 mg, 2.05 mmol) and K2CO3 (944 mg, 6.82 mmol) in a round-bottom flask was added DMF (4 mL) under argon and then the solution was refluxed for 18 h. The reaction mixture was partitioned between water and EtOAc. The organic layer was washed with a brine solution. Evaporation of the organic layer in vacuo gave a residue that was purified by column chromatography on silica gel (5% MeOH/DCM) to afford A8a as a pale red solid (63% yield). 1H NMR (400 MHz, CDCl3) δ 8.34 (s, 1H), 7.57 (d, J = 2 Hz, 1H), 7.41-7.37 (dd, J = 2.4, 2 Hz, 1H), 7.25-7.23 (m, 1H), 7.05 (s, 1H), 3.79 (s, 3H), 2.63 (s, 3H).
6-(2-chlorophenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-imine (A8b).
This compound was prepared in the similar manner as A8a and used in the next step without purification.
8-Methyl-2-(methylthio)-6-phenylpyrido[2,3-d]pyrimidin-7(8H)-imine (A8c).
Light red solid (Yield 54%): 1H NMR (400 MHz, CDCl3) δ 8.32 (s, 1H), 7.50-7.45 (m, 3H), 7.41-7.37 (m, 2H), 7.04 (s, 1H), 3.79 (s, 3H), 2.63 (s, 3H).
General procedure for the preparation of pyrido[2,3-d]pyrimidin-7(8H)-ones A9ac). Exemplified for 6-(2,4-dichlorophenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9a).
A suspension of A8a (200 mg, 0.57 mmol) in acetic anhydride (3 mL) was refluxed for 30 min. Evaporation of solvent in vacuo gave a residue that was treated with concentrated HCl (2 mL) and refluxed for 5 min. The reaction mixture was then neutralized with saturated solution of NaHCO3 and partitioned between water and EtOAc. The organic layer was washed with brine solution, filtered and concentrated to get a residue that was purified by column chromatography on silica gel (30 % EtOAc/Hexane) to afford A9a as a pale yellow solid (65% yield). 1H NMR (400 MHz, CDCl3) δ 8.64 (s, 1H), 7.65-7.62 (m, 1H), 7.51 (s, 1H), 7.34-7.29 (m, 2H), 3.82 (s, 3H), 2.66 (s, 3H).
6-(2-Chlorophenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9b).
Pale yellow solid (61% yield): 1H NMR (400 MHz, CDCl3) δ 8.64 (s, 1H), 7.64 (s, 1H), 7.48-7.46 (m, 1H), 7.36-7.33 (m, 3H), 3.81 (s, 3H), 2.65 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 173.5, 161.8, 156.5, 154.3, 135.0, 134.7, 133.7, 131.6, 131.5, 129.9, 129.9, 126.9, 109.4, 28.5, 14.6.
8-Methyl-2-(methylthio)-6-phenylpyrido[2,3-d]pyrimidin-7(8H)-one (A9c).
Yellow solid (55% yield): 1H NMR (500 MHz, CDCl3) δ (ppm) 8.63 (s, 1H), 7.69 (s, 1H), 7.67-7.63 (m, 2H), 7.45-7.36 (m, 3H), 3.81 (s, 3H), 2.64 (s, 3H). 13C NMR (125 MHz, CDCl3) δ 172.8, 162.4, 156.3, 153.9, 135.6, 132.8, 132.6, 128.9, 128.7, 128.4, 109.9, 28.5, 14.6.
General procedure for the preparation of pyrido[2,3-d]pyrimidin-7(8H)-ones (A9d-j) and (A10a-c). Exemplified for 6-(4-chlorophenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9d).
KF/Al2O3 (70 mg, 40 wt %) was added to a stirring solution of A7 (15 mg, 0.08 mmol) and methyl 2-(4-chlorophenyl)acetate (23 mg, 0.12 mmol) in dry DMA (1.5 mL). The mixture was stirred at rt for 3 h under argon. The reaction mixture was then filtered through Celite and the residual solid was washed with DCM. The filtrate was concentrated and the residue was purified by column chromatography over silica gel (15% EtOAc/hexane) to afford A9d as a light-yellow solid (58% yield). 1H NMR (500 MHz, CDCl3) δ (s, 1H), 7.67 (s, 1H), 7.61 (d, J = 8.5 Hz, 2H), 7.40 (d, J = 8.5 Hz, 2H), 3.81 (s, 3H), 2.65 (s, 3H). 13C NMR (125 MHz, CDCl3) δ 173.2, 162.2, 156.4, 153.9, 134.7, 134.0, 132.7, 131.5, 130.2, 128.6, 109.7, 28.5, 14.6.
6-(4-(tert-butyl)phenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9e).
Yellow solid (41% yield): 1H NMR (400 MHz, CDCl3) δ 8.64 (s, 1H), 7.68 (s, 1H), 7.61 (d, J = 6 Hz, 2H), 7.46 (d, J = 7.6 Hz, 2H), 3.81 (s, 3H), 2.64 (s, 3H), 1.35 (s, 9H). 13C NMR (100 MHz, CDCl3) δ 172.6, 162.5, 156.1, 153.9, 151.8, 132.7, 132.7, 132.1, 128.6, 125.4, 110.0, 34.8, 31.4, 28.5, 14.6.
6-(2,6-Dichlorophenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9f).
White solid (30% yield): 1H NMR (400 MHz, CDCl3) δ (ppm) 8.67 (s, 1H), 7.61 (s, 1H), 7.42 (d, J = 7.8 Hz, 2H), 7.33-7.26 (m, 1H), 3.84 (s, 3H), 2.67 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 173.8, 161.0, 156.6, 154.5, 136.0, 135.5, 133.7, 130.3, 129.5, 128.2, 109.2, 28.5, 14.6.
6-(2,3-Dichlorophenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9g).
White solid (38% yield): 1H NMR (500 MHz, CDCl3) δ 8.65 (s, 1H), 7.64 (s, 1H), 7.52 (dd, J = 7.5, 2 Hz, 1H), 7.30-7.24 (m, 2H), 3.82 (s, 3H), 2.67 (s, 3H).
6-(2-Chloro-4-fluorophenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9h).
White solid (35% yield): 1H NMR (400 MHz, CDCl3) δ 8.63 (s, 1H), 7.64 (s, 1H), 7.35-7.30 (m, 1H), 7.24-7.15 (m, 1H), 7.04 (td, J = 8.4, 2.8 Hz, 1H), 3.80 (s, 3H), 2.64 (s, 3H). 13C NMR (150 MHz, CDCl3) δ 173.6, 163.3, 161.8, 161.7, 156.5, 154.3, 135.3, 134.7, 134.6, 132.6, 132.6, 130.8, 130.5, 117.4, 117.2, 114.3, 114.1, 109.3, 28.5, 14.6.
6-(2,5-Dichlorophenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9i).
White solid (23% yield): 1H NMR (400 MHz, CDCl3) δ 8.65 (s, 1H), 7.66 (s, 1H), 7.43-7.31 (m, 3H), 3.82 (s, 3H), 2.66 (s, 3H).
6-(4-Hydroxyphenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9j).
Light yellow solid (55% yield): 1H NMR (400 MHz, DMSO-d6) δ 9.68 (s, 1H), 8.87 (s, 1H), 7.99 (s, 1H), 7.54 (d, J = 8.8 Hz, 2H), 6.81 (d, J = 8 Hz, 2H), 3.64 (s, 3H), 2.59 (s, 3H).
6-(4-((Tert-butyldimethylsilyl)oxy)phenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9k).
To a solution of A9j (160 mg, 0.53 mmol) in dry DCM (3 mL) were added a catalytic amount of DMAP (3 mg) and imidazole (109 mg, 1.60 mmol). TBDMSCl (121 mg, 0.80 mmol) was then added to the mixture at 0 °C. The resulting mixture was stirred at rt for 4 h. Evaporation of DCM in vacuo gave a crude mixture that was portioned between water and DCM. The organic layer was then washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (5 % EtOAc/DCM) to afford A9k as light yellow solid (88% yield). 1H NMR (600 MHz, CDCl3) δ 8.64 (s, 1H), 7.66 (s, 1H), 7.57 (d, J = 8.4 Hz, 2H), 6.89 (d, J = 7.8 Hz, 2H), 3.81 (s, 3H), 2.65 (s, 3H), 0.99 (s, 9H), 0.22 (s, 6H).
6-(4-Methoxyphenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9l).
Anhydrous K2CO3 was added to the solution of A9j (20 mg, 0.06 mmol) in dry acetone (2 mL). Iodomethane (10 μL, 0.10 mmol) was added to the mixture and the contents were refluxed for 8 h. The reaction mixture was allowed to cool to rt and then partitioned between water and EtOAc. The organic layer was washed with brine solution. Evaporation to remove EtOAc in vacuo gave a residue that was purified by column chromatography on silica gel (50% EtOAc/Hex) to afford A9l (20 mg, 95%) as light yellow solid. 1H NMR (400 MHz, CDCl3) δ 8.64 (s, 1H), 7.65-7.62 (m, 3H), 6.96 (d, J = 8.8 Hz, 2H), 3.85 (s, 3H), 3.81 (s, 3H), 2.65 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 172.4, 162.6, 160.0, 156.0, 153.8, 132.4, 131.4, 130.2, 128.0, 113.8, 110.1, 55.5, 28.5, 14.6.
2-(methylthio)-6-phenylpyrido[2,3-d]pyrimidin-7(8H)-one (A10a).
White solid (51% yield): 1H NMR (500 MHz, CDCl3) δ 9.66 (s, 1H), 8.70 (s, 1H), 7.77 (s, 1H), 7.70 (d, J = 7.5 Hz, 2H), 7.47-7.39 (m, 3H), 2.62 (s, 3H). 13C NMR (125 MHz, CDCl3) δ 173.6, 162.3, 156.0, 152.3, 134.7, 134.0, 133.6, 129.0, 128.8, 128.5, 109.6, 14.5.
6-(2,6-Dichlorophenyl)-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A10b).
Light yellow solid (28% yield): 1H NMR (400 MHz, CDCl3) δ (ppm) 9.81 (s, 1H), 8.70 (s, 1H), 7.65 (s, 1H), 7.41 (d, J = 8.4 Hz, 2H), 7.30 (dd, J = 8.7, 7.8 Hz, 1H), 2.60 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 174.5, 160.9, 156.4, 153.9, 137.7, 135.6, 132.9, 130.5, 130.3, 128.2, 108.8, 14.5.
6-(4-Chlorophenyl)-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A10c).
White solid (40% yield): 1H NMR (400 MHz, CDCl3) δ 9.54 (s, 1H), 8.71 (s, 1H), 7.77 (s, 1H), 7.66 (d, J = 8.8 Hz, 2H), 7.42 (d, J = 8.8 Hz, 2H), 2.62 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 173.9, 161.9, 156.1, 153.3, 135.0, 134.1, 133.1, 132.3, 130.1, 128.7, 109.4, 14.5.
General procedure for the preparation of pyrido[2,3-d]pyrimidin-7(8H)-ones (A9m-t). Exemplified for 8-ethyl-2-(methylthio)-6-phenylpyrido[2,3-d]pyrimidin-7(8H)-one (A9m).
Sodium hydride (2.67 mg, 0.11 mmol) was added to the solution of A10a (20 mg, 0.07 mmol) in dry DMF (1.5 mL). Ethyl iodide (50 μL, 0.11 mmol) was added to the mixture and then the reaction vessel was heated at 50 °C for 1h. The reaction mixture was partitioned between water and EtOAc and the organic layer was washed with brine solution. Evaporation to remove EtOAc in vacuo gave a residue that was purified by column chromatography on silica gel (50% EtOAc/Hex) to afford A9m (25 mg, 74%) as a colorless liquid. 1H NMR (400 MHz, CDCl3) δ 8.66 (s, 1H), 7.71-7.65 (m, 3H), 7.47-7.36 (m, 3H), 4.56 (q, J = 7.2 Hz, 2H), 2.65 (s, 3H), 1.37 (t, J = 7.1 Hz, 3H). 13C NMR (100 MHz, CDCl3) δ 172.8, 161.8, 156.4, 153.4, 135.6, 133.0, 132.6, 129.0, 128.7, 128.4, 109.9, 29.8, 14.5, 13.1.
8-(2-Hydroxyethyl)-2-(methylthio)-6-phenylpyrido[2,3-d]pyrimidin-7(8H)-one (A9n).
Pale yellow viscous liquid (48% yield): 1H NMR (400 MHz, CDCl3) δ 8.61 (s, 1H), 7.67 (s, 1H), 7.54 (d, J = 8 Hz, 2H), 7.39-7.31 (m, 3H), 4.63 (t, J = 6 Hz, 2H), 3.88 (t, J = 6 Hz, 2H), 2.56 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 172.9, 163.0, 156.5, 153.7, 135.3, 133.4, 132.9, 128.8, 128.8, 128.4, 110.0, 59.9, 43.6, 14.4.
6-(2,6-Dichlorophenyl)-8-(2-methoxyethyl)-2-(methylthio)pyrido[2,3-d] pyrimidin-7(8H)-one (A9o).
Colorless liquid (44% yield): 1H NMR (400 MHz, CDCl3) δ 8.67 (s, 1H), 7.62 (s, 1H), 7.41 (d, J = 7.8 Hz, 2H), 7.34-7.25 (m, 1H), 4.74 (t, J = 6.2 Hz, 2H), 3.78 (t, J = 6.2 Hz, 2H), 3.39 (s, 3H), 2.65 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 173.7, 160.7, 156.7, 154.4, 136.2, 135.5, 133.7, 130.2, 129.6, 128.2, 109.3, 69.0, 58.9, 40.3, 14.6.
8-Benzyl-6-(2,6-dichlorophenyl)-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9p).
White semisolid (29% yield): 1H NMR (400 MHz, CDCl3) δ 8.65 (s, 1H), 7.61 (s, 1H), 7.47 (d, J = 6.9 Hz, 2H), 7.41 (d, J = 8.2 Hz, 2H), 7.32-7.22 (m, 4H), 5.70 (s, 2H), 2.61 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 173.9, 160.9, 156.7, 154.2, 136.8, 136.3, 135.5, 133.7, 130.3, 129.8, 128.9, 128.6, 128.6, 128.2, 127.6, 109.4, 44.7, 14.7.
6-(2,6-Dichlorophenyl)-8-isobutyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9q).
White solid (28% yield): 1H NMR (400 MHz, CDCl3) δ (ppm) 8.66 (s, 1H), 7.60 (s, 1H), 7.41 (d, J = 8 Hz, 2H), 7.29 (t, J = 8, 7.2 Hz, 1H), 4.36 (d, J = 7.6 Hz, 2H), 2.65 (s, 3H), 2.40-2.30 (sep, J = 6.8, 7.2 Hz, 1H), 0.98 (d, J = 6.9 Hz, 6H). 13C NMR (100 MHz, CDCl3) δ 173.5, 161.0, 156.7, 154.5, 135.9, 135.5, 133.9, 130.2, 129.8, 128.1, 109.1, 48.4, 27.6, 20.4, 14.6.
6-(4-Chlorophenyl)-8-isobutyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9r).
Yellow solid (73% yield): 1H NMR (400 MHz, CDCl3) δ 8.66 (s, 1H), 7.70 (s, 1H), 7.63 (d, J = 8.8 Hz, 2H), 7.41 (d, J = 8.8 Hz, 2H), 4.36 (d, J = 7.3 Hz, 2H), 2.64 (s, 3H), 2.33 (sep, J = 6.8 Hz, 1H), 0.98 (d, J = 6.8 Hz, 6H).
6-(2,6-Dichlorophenyl)-8-(3-(methylsulfonyl)benzyl)-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9s).
Light yellow solid (36% yield): 1H NMR (400 MHz, CDCl3) δ (s, 1H), 8.08 (s, 1H), 7.84 (d, J = 8 Hz, 1H), 7.75 (d, J = 8 Hz, 1H), 7.65 (s, 1H), 7.51 (t, J = 7.8 Hz, 1H), 7.43 (d, J = 8 Hz, 2H), 7.32-7.28 (m, 1H), 5.76 (s, 2H), 3.03 (s, 3H), 2.63 (s, 3H).
6-(2,6-Dichlorophenyl)-8-(4-(methylsulfonyl)benzyl)-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A9t).
Pale yellow solid (53% yield): 1H NMR (400 MHz, CDCl3) δ 8.70 (s, 1H), 7.87 (d, J = 8.4 Hz, 2H), 7.67-7.61 (m, 3H), 7.42 (d, J = 8 Hz, 2H), 7.32-7.28 (m, 1H), 5.76 (s, 2H), 3.02 (s, 3H), 2.58 (s, 3H).
4-(2-(Diethylamino)ethoxy)aniline (A12a).
To a solution of N,N-diethyl-2-(4-nitrophenoxy)ethanamine (490 mg, 2.06 mmol) in methanol (10 mL) was added 10% Pd/C (125 mg). The reaction was stirred in the presence of H2 gas (1 atm) for 4 h. The reaction mixture was then filtered through Celite and concentrated to afford A12a as a brown viscous liquid (96% yield). 1H NMR (400 MHz, CDCl3) δ 6.75-6.73 (m, 2H), 6.64-6.62 (m, 2H), 3.98 (t, J = 6.4 Hz, 2H), 2.84 (t, J = 6.4 Hz, 2H), 2.63 (q, J = 7.2 Hz, 4H), 1.06 (t, J = 7.2 Hz, 6H). 13C NMR (100 MHz, CDCl3) δ 152.1, 140.0, 116.5, 115.7, 67.0, 51.9, 47.8, 11.8.
General procedure for the preparation of N-phenylformamides A13a-c. Exemplified for N-(4-(2-(diethylamino)ethoxy)phenyl)formamide (A13a).
Formic acid (1 mL) was added to A12a (100 mg, 0.48 mmol) in a round-bottom flask containing molecular sieves (4Å, 8-12 mesh)). The reaction mixture was heated at 60 °C for 6 h and then partitioned between a saturated solution of NaHCO3 and EtOAc. The organic layer was washed with brine solution and concentrated to give A13a (79% yield) as a brown viscous liquid which was used without purification in the next step.
N-(4-(methylsulfonyl)phenyl)formamide (A13b).
This compound was prepared in a similar manner as A13a and used in the next step without purification.
N-(3-(methylsulfonyl)phenyl)formamide (A13c).
White solid (80% yield): 1H NMR (400 MHz, DMSO-d6) δ 10.64 (s, 1H), 8.40 (s, 1H), 8.28 (s, 1H), 7.89-7.86 (m, 1H), 7.67-7.63 (m, 2H), 3.24 (s, 3H). 13C NMR (100 MHz, DMSO-d6) δ 160.7, 141.9, 139.4, 130.8, 124.1, 122.5, 117.5, 44.1.
General Procedure for the preparation of 2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-ones (A11a-i) and (A11k-t). Exemplified for 6-(2,4-dichlorophenyl)-8-methyl-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A11a).
m-CPBA (85 mg, 55%) was added to the solution of A9a (38 mg, 0.108 mmol) in DCM (2 mL). The solution was stirred for 3 h. The reaction mixture was then partitioned between water and DCM and the organic layer was washed with brine, filtered and concentrated. The residue was purified by column chromatography using silica gel (40% EtOAc/Hexane) to get A11a as a white solid (63% yield). 1H NMR (400 MHz, CDCl3) δ 9.01 (s, 1H), 7.82 (s, 1H), 7.53 (d, J = 1.8 Hz, 1H), 7.36 (dd, J = 8.2, 2.3 Hz, 1H), 7.31 (d, J = 8.2 Hz, 1H), 3.89 (s, 3H), 3.43 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 164.7, 161.0, 157.4, 155.2, 136.0, 135.4, 134.3, 134.0, 132.1, 132.0, 131.0, 127.4, 114.9, 39.3, 29.4.
6-(2-Chlorophenyl)-8-methyl-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A11b).
White solid (45% yield): 1H NMR (400 MHz, CDCl3) δ 9.00 (s, 1H), 7.82 (s, 1H), 7.48 (d, J = 7.2 Hz, 1H), 7.42-7.32 (m, 3H), 3.87 (s, 3H), 3.42 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 164.5, 161.2, 157.3, 155.1, 136.5, 133.8, 133.7, 133.4, 131.2, 130.6, 130.0, 127.0, 115.0, 39.4, 29.3.
8-Methyl-2-(methylsulfonyl)-6-phenylpyrido[2,3-d]pyrimidin-7(8H)-one (A11c).
Light yellow solid (54% yield): 1H NMR (400 MHz, CDCl3) δ 9.00 (s, 1H), 7.85 (s, 1H), 7.70-7.66 (m, 2H), 7.49-7.44 (m, 3H), 3.88 (s, 3H), 3.42 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 164.0, 161.9, 156.9, 154.6, 137.6, 134.5, 131.15, 129.7, 129.0, 128.6, 115.6, 39.3, 29.3.
6-(4-Chlorophenyl)-8-methyl-2-(methylsulfonyl)pyrido[2,3-d] pyrimidin-7(8H)-one (A11d).
White solid (72% yield): 1H NMR (400 MHz, CDCl3) δ 9.01 (s, 1H), 7.86 (s, 1H), 7.652 (d, J = 8.8 Hz, 2H), 7.445 (d, J = 8.8 Hz, 2H), 3.89 (s, 3H), 3.43 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 164.2, 161.6, 157.0, 154.7, 136.4, 135.9, 132.9, 131.2, 130.3, 128.9, 115.4, 39.3, 29.4.
6-(4-(Tert-butyl)phenyl)-8-methyl-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A11e).
White solid (81% yield): 1H NMR (400 MHz, CDCl3) δ 8.98 (s, 1H), 7.84 (s, 1H), 7.63 (d, J = 8.8 Hz, 2H), 7.47 (d, J = 8 Hz, 2H), 3.86 (s, 3H), 3.40 (s, 3H), 1.34 (s, 9H). 13C NMR (100 MHz, CDCl3) δ 163.8, 162.0, 156.8, 154.5, 153.0, 137.4, 131.7, 130.6, 128.7, 125.6, 115.7, 39.3, 34.9, 31.3, 29.3.
6-(2,6-Dichlorophenyl)-8-methyl-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A11f).
White solid (93% yield): 1H NMR (600 MHz, CDCl3) δ 9.03 (s, 1H), 7.77 (s, 1H), 7.45 (d, J = 7.8 Hz, 2H), 7.35 (t, J = 7.8, 9 Hz, 1H), 3.91 (s, 3H), 3.44 (s, 3H).
6-(2,3-Dichlorophenyl)-8-methyl-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A11g).
Light yellow solid (65% yield): 1H NMR (400 MHz, CDCl3) δ 9.02 (s, 1H), 7.82 (s, 1H), 7.57 (dd, J = 8, 2 Hz, 1H), 7.34-7.25 (m, 2H), 3.90 (s, 3H), 3.44 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 164.7, 160.9, 157.4, 155.2, 136.4, 135.8, 133.9, 133.7, 132.1, 131.4, 129.2, 127.6, 114.9, 39.3, 29.4.
6-(2-Chloro-4-fluorophenyl)-8-methyl-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A11h).
White solid (40% yield): 1H NMR (400 MHz, CDCl3) δ 9.02 (s, 1H), 7.83 (s, 1H), 7.37 (dd, J = 14, 6.6 Hz, 1H), 7.29-7.26 (m, 1H), 7.11 (td, J = 8, 2.4 Hz, 1H), 3.90 (s, 3H), 3.44 (s, 3H). 13C NMR (150 MHz, CDCl3) δ 164.7, 163.7, 162.0, 161.2, 157.3, 155.1, 135.5, 134.6, 134.5, 134.0, 132.5, 132.4, 129.7, 117.7, 117.5, 114.9, 114.5, 114.4, 39.3, 29.4.
6-(2,5-Dichlorophenyl)-8-methyl-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A11i).
White solid (75% yield): 1H NMR (400 MHz, CDCl3) δ 9.02 (s, 1H), 7.83 (s, 1H), 7.46-7.47 (m, 1H), 7.39-7.36 (m, 2H), 3.89 (s, 3H), 3.44 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 164.8, 160.8, 157.4, 155.2, 135.3, 135.0, 134.1, 132.9, 131.9, 131.2, 131.1, 130.6, 114.8, 39.3, 29.4.
6-(4-((Tert-butyldimethylsilyl)oxy)phenyl)-8-methyl-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A11k).
Light yellow solid (64 % yield): 1H NMR (600 MHz, CDCl3) δ 8.96 (s, 1H), 7.80 (s, 1H), 7.60 (d, J = 8.4 Hz, 2H), 6.88 (d, J = 8.4 Hz, 2H), 3.85 (s, 3H), 3.39 (s, 3H), 0.98 (s, 9H), 0.21 (s, 6H). 13C NMR (150 MHz, CDCl3) δ 163.6, 162.0, 157.2, 156.6, 154.3, 136.9, 130.4, 129.9, 127.5, 120.2, 115.7, 39.3, 29.2, 25.7, 18.3.
6-(4-Methoxyphenyl)-8-methyl-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A11l).
Yellow solid (97 % yield): 1H NMR (400 MHz, CDCl3) δ 8.98 (s, 1H), 7.80 (s, 1H), 7.68 (d, J = 8.8 Hz, 2H), 6.98 (d, J = 8.8 Hz, 2H), 3.88 (s, 3H), 3.86 (s, 3H), 3.42 (s, 3H).
8-Ethyl-2-(methylsulfonyl)-6-phenylpyrido[2,3-d]pyrimidin-7(8H)-one (A11m).
White solid (55% yield): 1H NMR (400 MHz, CDCl3) δ 9.01 (s, 1H), 7.85 (s, 1H), 7.72-7.67 (m, 2H), 7.52-7.44 (m, 3H), 4.60 (q, J = 7.0 Hz, 2H), 3.42 (s, 3H), 1.40 (t, J = 7.1 Hz, 3H).
8-(2-Hydroxyethyl)-2-(methylsulfonyl)-6-phenylpyrido[2,3-d]pyrimidin-7(8H)-one (A11n).
Light yellow solid (81% yield), which was used without purification in next step.
6-(2,6-Dichlorophenyl)-8-(2-methoxyethyl)-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A11o).
White solid (58% yield): 1H NMR (400 MHz, CDCl3) δ 9.02 (s, 1H), 7.78 (s, 1H), 7.43 (d, J = 8.2 Hz, 2H), 7.33 (dd, J = 8.8, 7.2 Hz, 1H), 4.79 (t, J = 5.6 Hz, 2H), 3.80 (t, J = 5.6 Hz, 2H), 3.41 (s, 3H), 3.36 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 164.6, 160.2, 157.6, 155.4, 135.1, 134.6, 132.7, 130.9, 128.3, 114.9, 69.1, 59.0, 41.2, 39.4.
8-Benzyl-6-(2,6-dichlorophenyl)-2-(methylsulfonyl)pyrido[2,3-d] pyrimidin-7(8H)-one (A11p).
White solid (61% yield): 1H NMR (400 MHz, CDCl3) δ 9.01 (s, 1H), 7.78 (s, 1H), 7.59 (d, J = 7.6 Hz, 2H), 7.45 (d, J = 8 Hz, 2H), 7.39-7.24 (m, 4H), 5.73 (s, 2H), 3.33 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 164.6, 160.4, 157.6, 154.9, 135.8, 135.1, 134.9, 132.7, 130.9, 129.3, 128.7, 128.3, 128.2, 115.0, 45.5, 39.5.
6-(2,6-Dichlorophenyl)-8-isobutyl-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A11q).
White solid (87% yield): 1H NMR (400 MHz, CDCl3) δ 9.03 (s, 1H), 7.77 (s, 1H), 7.43 (d, J = 7.6 Hz, 2H), 7.33 (dd, J = 9.0, 7.4 Hz, 1H), 4.40 (d, J = 7.3 Hz, 2H), 3.41 (s, 3H), 2.37-2.27 (sep, J = 6.8, 7.2 Hz, 1H), 0.98 (d, J = 6.9 Hz, 6H). 13C NMR (100 MHz, CDCl3) δ 164.7, 160.4, 157.7, 155.2, 135.0, 134.8, 134.7, 132.9, 130.8, 128.3, 114.8, 49.1, 39.3, 27.6, 20.3.
6-(4-Chlorophenyl)-8-isobutyl-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A11r).
Light brown solid (66% yield), which was used without purification in next step.
6-(2,6-Dichlorophenyl)-2-(methylsulfonyl)-8-(3-(methylsulfonyl)benzyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A11s).
White powder (79% yield): 1H NMR (500 MHz, CDCl3) δ 9.03 (s, 1H), 8.25 (s, 1H), 7.91 (d, J = 8 Hz, 1H), 7.86 (d, J = 8 Hz, 1H), 7.81 (s, 1H), 7.52 (t, J = 7.5 Hz, 1H), 7.46 (d, J = 7.5 Hz, 2H), 7.38-7.33 (m, 1H), 5.79 (s, 2H), 3.39 (s, 3H), 3.09 (s, 3H).
6-(2,6-Dichlorophenyl)-2-(methylsulfonyl)-8-(4-(methylsulfonyl)benzyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A11t).
White powder (72% yield): 1H NMR (500 MHz, CDCl3) δ 9.03 (s, 1H), 7.89-7.81 (m, 5H), 7.46 (d, J = 8 Hz, 2H), 7.38-7.35 (m, 1H), 3.37 (s, 3H), 3.01 (s, 3H).
General procedure for the preparation of 2,6-diphenyl-pyrido[2,3-d]pyrimidin-7(8H)-ones (15-40). Exemplified for 6-(2,4-dichlorophenyl)-2-((4-(2-(diethylamino)ethoxy)phenyl)amino)-8-methylpyrido[2,3-d]pyrimidin-7(8H)-one (15).
To a solution of A13a (30 mg, 0.13 mmol) in THF (0.5 mL) and DMF (0.5 mL) was added 60% NaH (8 mg, 0.33 mmol) at 0 °C. The mixture was stirred for 30 min at rt under argon. The mixture was cooled to 0 °C and then A11a (25 mg, 0.06 mmol) was added. The reaction mixture was stirred at rt for 2 h. The reaction mixture was quenched by addition of ice and NaOH (0.5 mL, 2N) solution and then partitioned between water and EtOAc. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated to give a residue that was purified by column chromatography using silica gel (5% MeOH/DCM) to afford 15 as a yellow solid (74% yield). 1H NMR (600 MHz, CDCl3) δ 8.54 (s, 1H), 7.55 (d, J = 7.8 Hz, 4H), 7.49 (s, 1H), 7.30 (s, 2H), 6.94 (d, J = 9.2 Hz, 2H), 4.07 (t, J = 6.2 Hz, 2H), 3.75 (s, 3H), 2.89 (t, J = 6.2 Hz, 2H), 2.65 (q, J = 7.2 Hz, 4H), 1.08 (t, J = 7.1 Hz, 6H). 13C NMR (150 MHz, CDCl3) δ 162.1, 159.4, 158.6, 155.9, 155.6, 135.7, 134.7, 134.6, 133.7, 132.6, 131.5, 129.7, 127.1, 126.5, 122.0, 115.0, 106.4, 66.9, 51.8, 47.9, 28.6, 11.9. HRMS m/z calculated for C26H27Cl2N5O2 [M + H]+: 512.1615; found 512.1626. Purity 95.6% (tR 21.56 min), mp 245-247 °C.
6-(2-Chlorophenyl)-2-((4-(2-(diethylamino)ethoxy)phenyl)amino)-8-methylpyrido[2,3-d]pyrimidin-7(8H)-one (16).
Synthesized from A11b and A13a by using the general procedure for 15 to give 16 (62%) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ 8.54 (s, 1H), 7.55 (d, J = 6.9 Hz, 3H), 7.48-7.45 (m, 1H), 7.37-7.30 (m, 3H), 6.94 (d, J = 8.7 Hz, 2H), 4.07 (t, J = 6.4 Hz, 2H), 3.76 (s, 3H), 2.89 (t, J = 6.2 Hz, 2H), 2.65 (q, J = 7.2 Hz, 4H), 1.08 (t, J = 7.3 Hz, 6H). 13C NMR (100 MHz, CDCl3) δ 162.3, 159.3, 158.5, 155.9, 155.5, 135.5, 135.2, 133.9, 131.7, 131.6, 129.8, 129.5, 127.8, 126.7, 122.0, 115.0, 106.5, 66.9, 51.8, 47.9, 28.6, 11.9. HRMS m/z calculated for C26H28ClN5O2 [M + H]+: 478.2004; found 478.2007. Purity 99.15% (tR 19.93 min), mp 193-195 °C.
6-(4-Chlorophenyl)-2-((4-(2-(diethylamino)ethoxy)phenyl)amino)-8-methylpyrido[2,3-d]pyrimidin-7(8H)-one (17).
Synthesized from A11d and A13a by using the general procedure for 15 to give 17 (45%) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ 8.56 (s, 1H), 7.62-7.60 (m, 3H), 7.54 (d, J = 8.8 Hz, 2H), 7.38 (d, J = 8.8 Hz, 2H), 6.94 (d, J = 8.8 Hz, 2H), 4.07 (t, J = 6.2 Hz, 2H), 3.75 (s, 3H), 2.89 (t, J = 6.2 Hz, 2H), 2.66 (q, J = 7.2 Hz, 4H), 1.08 (t, J = 7.3 Hz, 6H). 13C NMR (150 MHz, CDCl3) δ 162.7, 159.1, 158.4, 155.4, 155.3, 134.6, 134.0, 133.3, 131.7, 130.1, 128.5, 127.8, 122.0, 115.0, 106.9, 66.6, 51.7, 47.8, 28.6, 11.6. HRMS m/z calculated for C26H28ClN5O2 [M + H]+: 478.2004; found 478.2010. Purity 96.5% (tR 21.33 min), mp 216-218 °C.
6-(4-(Tert-butyl)phenyl)-2-((4-(2-(diethylamino)ethoxy)phenyl)amino)-8-methylpyrido[2,3-d]pyrimidin-7(8H)-one (18).
Synthesized from A11e and A13a by using the general procedure for 15 to give 18 (58%) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ 8.54 (s, 1H), 7.63-7.59 (m, 3H), 7.56 (d, J = 8.8 Hz, 2H), 7.45 (d, J = 8.4 Hz, 2H), 6.94 (d, J = 8.8 Hz, 2H), 4.07 (t, J = 6.4 Hz, 2H), 3.76 (s, 3H), 2.89 (t, J = 6.4 Hz, 2H), 2.65 (q, J = 7.2 Hz, 4H), 1.35 (s, 9H), 1.08 (t, J = 7.1 Hz, 6H). 13C NMR (100 MHz, CDCl3) δ 163.1, 159.0, 158.1, 155.4, 155.3, 151.2, 133.3, 132.8, 131.8, 129.1, 128.5, 125.3, 121.9, 114.9, 107.2, 66.9, 51.8, 47.9, 34.7, 31.4, 28.6, 11.9. HRMS m/z calculated for C30H37N5O2 [M + H]+: 500.3020; found 500.3028. Purity 99.8% (tR 23.44 min), mp 208-210 °C.
2-((4-(2-(Diethylamino)ethoxy)phenyl)amino)-6-(4-hydroxyphenyl)-8-methylpyrido[2,3-d]pyrimidin-7(8H)-one (19).
Synthesized from A11k and A13a by using the general procedure for 15 to give 19 (64%) as pale yellow solid. Note the TBS-protecting group from A11k was removed during the reaction. 1H NMR (400 MHz, CD3OD) δ 8.66 (s, 1H), 7.79 (s, 1H), 7.68 (d, J = 9.2 Hz, 2H), 7.50 (d, J = 8.8 Hz, 2H), 6.99 (d, J = 9.2 Hz, 2H), 6.83 (d, J = 8.8 Hz, 2H), 4.25 (t, J = 5.2 Hz, 2H), 3.74 (s, 3H), 3.36-3.34 (m, 2H), 3.11 (q, J = 7.2 Hz, 4H), 1.28 (t, J = 7.4 Hz, 6H). HRMS m/z calculated for C26H29N5O3 [M + H]+: 460.2343; found 460.2348. Purity 99.82% (tR 17.44 min), mp 215-217 °C.
2-((4-(2-(Diethylamino)ethoxy)phenyl)amino)-6-(4-methoxyphenyl)-8-methylpyrido[2,3-d]pyrimidin-7(8H)-one (20).
Synthesized from A11l and A13a by using the general procedure for 15 to give 20 (66%) as yellow solid. 1H NMR (400 MHz, CD3OD) δ 8.59 (s, 1H), 7.73 (s, 1H), 7.61 (d, J = 8.4 Hz, 2H), 7.55 (d, J = 8.8 Hz, 2H), 6.94-6.90 (m, 4H), 4.11 (t, J = 5.4 Hz, 2H), 3.80 (s, 3H), 3.68 (s, 3H), 3.04 (t, J = 5.2 Hz, 2H), 2.82 (q, J = 7.2 Hz, 4H), 1.16 (t, J = 7 Hz, 6H). HRMS m/z calculated for C27H31N5O3 [M + H]+: 474.2500; found 474.2505. Purity 98.89% (tR 19.86 min), mp 188-190 °C.
2-((4-(2-(Diethylamino)ethoxy)phenyl)amino)-8-methyl-6-phenylpyrido[2,3-d]pyrimidin-7(8H)-one (21).
Synthesized from A11c and A13a by using the general procedure for 15 to give 21 (64%) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ 8.55 (s, 1H), 7.68-7.63 (m, 3H), 7.56 (d, J = 8.7 Hz, 2H), 7.45-7.33 (m, 3H), 6.94 (td, J = 6.2, 4.1 Hz, 2H), 4.12 (t, J = 6.2 Hz, 2H), 3.77 (s, 3H), 2.96 (t, J = 6.2 Hz, 2H), 2.73 (q, J = 7.2 Hz, 4H), 1.13 (t, J = 7.1 Hz, 6H). 13C NMR (100 MHz, CDCl3) δ 162.9, 159.0, 158.3, 155.4, 155.2, 136.2, 133.3, 131.9, 129.2, 128.9, 128.3, 128.2, 121.9, 115.0, 107.1, 66.4, 51.7, 47.8, 28.6, 11.5. HRMS m/z calculated for C26H29N5O2 [M + H]+: 444.2394; found 444.2397. Purity 99.4% (tR 19.75 min), mp 172-174 °C.
2-((4-(2-(Diethylamino)ethoxy)phenyl)amino)-8-ethyl-6-phenylpyrido[2,3-d]pyrimidin-7(8H)-one (22).
Synthesized from A11m and A13a by using the general procedure for 15 to give 22 (54%) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ 8.56 (s, 1H), 7.67 (d, J = 3.6 Hz, 2H), 7.64 (s, 1H), 7.56 (d, J = 9.2 Hz, 2H), 7.46-7.30 (m, 3H), 6.94 (d, J = 8.8 Hz, 2H), 4.50 (q, J = 7.0 Hz, 2H), 4.07 (t, J = 6.2 Hz, 2H), 2.90 (t, J = 6.2 Hz, 2H), 2.66 (q, J = 7.0 Hz, 4H), 1.38 (t, J = 7.1 Hz, 3H), 1.09 (t, J = 7.1 Hz, 6H). HRMS m/z calculated for C27H31N5O2 [M + H]+: 458.2551; found 458.2556. Purity 95.8% (tR 20.50 min), mp 133-135 °C.
2-((4-(2-(Diethylamino)ethoxy)phenyl)amino)-8-(2-hydroxyethyl)-6-phenylpyrido[2,3-d]pyrimidin-7(8H)-one (23).
Synthesized from A11n and A13a by using the general procedure for 15 to give 23 (33%) as a yellow solid. 1H NMR (600 MHz, CD3OD) δ 8.70 (s, 1H), 7.88 (s, 1H), 7.68 (d, J = 7.8 Hz, 2H), 7.64 (d, J = 7.8 Hz, 2H), 7.41 (t, J = 7.5 Hz, 2H), 7.35 (d, J = 7.5 Hz, 1H), 7.01 (d, J = 9Hz, 2H), 4.63 (t, J = 6.3 Hz, 2H), 4.27 (t, J = 4.8 Hz, 2H), 3.89 (t, J = 6.6 Hz, 2H), 3.37-3.34 (m, 2H), 3.13-3.12 (m, 4H), 1.28 (t, J = 7.2 Hz, 6H). HRMS m/z calculated for C27H31N5O3 [M + H]+: 474.2500; found 474.2504. Purity 95.0% (tR 18.48 min), mp 157-159 °C.
6-(2,6-Dichlorophenyl)-2-((4-(2-(diethylamino)ethoxy)phenyl)amino)-8-(2-methoxyethyl)pyrido[2,3-d]pyrimidin-7(8H)-one (24).
Synthesized from A11o and A13a by using the general procedure for 15 to give 24 (61%) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ 8.55 (s, 1H), 7.56-7.51 (m, 3H), 7.41 (d, J = 8 Hz, 3H), 7.28-7.24 (m, 1H), 6.94 (d, J = 9.2 Hz, 2H), 4.66 (t, J = 6 Hz, 2H), 4.06 (t, J = 6.4 Hz, 2H), 3.76 (t, J = 6.2 Hz, 2H), 3.36 (s, 3H), 2.89 (t, J = 6.2 Hz, 2H), 2.66 (q, J = 7.2 Hz, 4H), 1.08 (t, J = 7.2 Hz, 6H). 13C NMR (100 MHz, CDCl3) δ 161.2, 159.5, 158.8, 155.9, 155.6, 136.6, 135.8, 134.2, 131.5, 129.9, 128.1, 125.7, 122.2, 114.9, 106.4, 69.1, 66.9, 58.9, 51.8, 47.9, 40.4, 11.9. HRMS m/z calculated for C28H31Cl2N5O3 [M + H]+: 556.1877; found 556.1879. Purity 97.4% (tR 20.76 min), mp 133-135 °C.
8-Benzyl-6-(2,6-dichlorophenyl)-2-((4-(2-(diethylamino)ethoxy)phenyl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (25).
Synthesized from A11p and A13a by using the general procedure for 15 to give 25 (68%) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ 8.54 (s, 1H), 7.53 (s, 1H), 7.41-7.19 (m, 11H), 6.91 (d, J = 9.2 Hz, 2H), 5.60 (s, 2H), 4.08 (t, J = 6.4 Hz, 2H), 2.90 (t, J = 6.2 Hz, 2H), 2.67 (q, J = 7.2 Hz, 4H), 1.09 (t, J = 7.3 Hz, 6H). 13C NMR (100 MHz, CDCl3) δ 161.4, 159.7, 158.8, 155.9, 155.7, 137.0, 136.7, 135.8, 134.3, 131.3, 129.9, 128.4, 128.2, 128.1, 127.3, 125.9, 123.1, 114.9, 106.4, 66.8, 51.8, 47.9, 44.5, 11.9. HRMS m/z calculated for C32H31Cl2N5O2 [M + H]+: 588.1928; found 588.1932. Purity 98.4% (tR 22.59 min), mp 163-165 °C.
6-(2,6-Dichlorophenyl)-2-((4-(2-(diethylamino)ethoxy)phenyl)amino)-8-isobutylpyrido[2,3-d]pyrimidin-7(8H)-one (26).
Synthesized from A11q and A13a by using the general procedure for 15 to give 26 (36%) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ 8.54 (s, 1H), 7.83 (brs, 1H), 7.57 (d, J = 9.2 Hz, 2H), 7.49 (s, 1H), 7.38 (d, J = 7.8 Hz, 2H), 7.23 (d, J = 8.2 Hz, 1H), 6.93 (d, J = 9.2 Hz, 2H), 4.28 (d, J = 7.3 Hz, 2H), 4.07 (t, J = 6.2 Hz, 2H), 2.90 (t, J = 6.2 Hz, 2H), 2.66 (q, J = 7.0 Hz, 4H), 2.42-2.31 (sep, J = 6.8, 7.2 Hz, 1H), 1.09 (t, J = 7.1 Hz, 6H), 0.97 (d, J = 6.4 Hz, 6H). 13C NMR (100 MHz, CDCl3) δ 161.6, 159.3, 158.6, 155.9, 155.4, 136.3, 135.8, 134.4, 131.8, 129.8, 128.1, 125.9, 122.0, 114.8, 106.3, 66.8, 51.8, 48.3, 47.9, 27.4, 20.3, 11.8. HRMS m/z calculated for C29H33Cl2N5O2 [M + H]+: 554.2084; found 554.2085. Purity 97.7% (tR 22.55 min), mp 196-198 °C.
6-(2,6-Dichlorophenyl)-2-((4-(2-(diethylamino)ethoxy)phenyl)amino)-8-(3-(methylsulfonyl)benzyl)pyrido[2,3-d]pyrimidin-7(8H)-one (27).
Synthesized from A11s and A13a by using the general procedure for 15 to give 27 (84%) as pale yellow solid. 1H NMR (600 MHz, CDCl3) δ 8.58 (s, 1H), 7.99 (s, 1H), 7.81 (d, J = 7.8 Hz, 1H), 7.60-7.39 (m, 7H), 7.32-7.26 (m, 2H), 6.94 (d, J = 9 Hz, 2H), 5.68 (s, 2H), 4.10 (t, J = 5.7 Hz, 2H), 2.93-2.91 (m, 5H), 2.68 (q, J = 7.2 Hz, 4H), 1.09 (t, J = 7.2 Hz, 6H). HRMS m/z calculated for C33H33Cl2N5O4S [M + H]+: 666.1703; found 666.1703. Purity 95.67% (tR 21.35 min), mp 203-205 °C.
6-(2,6-Dichlorophenyl)-2-((4-(2-(diethylamino)ethoxy)phenyl)amino)-8-(4-(methylsulfonyl)benzyl)pyrido[2,3-d]pyrimidin-7(8H)-one (28).
Synthesized from A11t and A13a by using the general procedure for 15 to give 28 (70%) as pale yellow solid. 1H NMR (600 MHz, CDCl3) δ 8.58 (s, 1H), 7.81 (d, J = 8.4 Hz, 2H), 7.57 (s, 1H), 7.49-7.36 (m, 6H), 7.29-7.26 (m, 2H), 6.92 (d, J = 8.4 Hz, 2H), 5.64 (s, 2H), 4.08 (t, J = 6.3 Hz, 2H), 3.00 (s, 3H), 2.91 (t, J = 6 Hz, 2H), 2.67 (t, J = 7.2 Hz, 4H), 1.09 (t, J = 6.9 Hz, 6H). HRMS m/z calculated for C33H33Cl2N5O4S [M + H]+: 666.1703; found 666.1716. Purity 95.61% (tR 21.24 min), mp 208-210 °C.
6-(2,6-Dichlorophenyl)-8-methyl-2-((4-(methylsulfonyl)phenyl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (29).
Synthesized from A11f and A13b by using the general procedure for 15 to give 29 (75%) as a white solid. 1H NMR (400 MHz, CDCl3) δ 8.67 (s, 1H), 7.99-7.91 (m, 4H), 7.78 (s, 1H), 7.79 (s, 1H), 7.42 (d, J = 7.8 Hz, 2H), 7.32-7.28 (m, 1H), 3.83 (s, 3H), 3.09 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 161.3, 158.5, 158.4, 155.9, 143.8, 136.1, 135.6, 134.0, 133.9, 130.2, 129.0, 128.2, 127.5, 119.0, 107.7, 44.9, 28.9. HRMS m/z calculated for C21H16Cl2N4O3S [M + H]+: 475.0393; found 475.0397. Purity 97.6% (tR 22.92 min), mp 298-300 °C.
6-(2,6-Dichlorophenyl)-8-methyl-2-((3-(methylsulfonyl)phenyl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (30).
Synthesized from A11f and A13c by using the general procedure for 15 to give 30 (92%) as a white solid. 1H NMR (400 MHz, CDCl3) δ 8.69 (d, J = 14.2 Hz, 2H), 8.22 (s, 1H), 7.82 (d, J = 8 Hz, 1H), 7.65 (d, J = 7.2 Hz, 1H), 7.59 (t, J = 4.2 Hz, 2H), 7.41 (d, J = 8 Hz, 2H), 7.31-7.25 (m, 1H), 3.86 (s, 3H), 3.11 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 161.4, 158.7, 158.6, 155.9, 141.4, 140.0, 136.2, 135.7, 134.0, 130.2, 130.1, 128.1, 127.1, 124.1, 121.6, 118.0, 107.3, 44.6, 29.0. HRMS m/z calculated for C21H16Cl2N4O3S [M + H]+: 475.0393; found 475.0393. Purity 98.2% (tR 22.98 min), mp 238-240 °C.
6-(4-Chlorophenyl)-8-methyl-2-((3-(methylsulfonyl)phenyl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (31).
Synthesized from A11d and A13c by using the general procedure for 15 to give 31 (61%) as a pale yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 10.59 (s, 1H), 8.90 (s, 1H), 8.78 (s, 1H), 8.11 (s, 1H), 7.95 (d, J = 8 Hz, 1H), 7.73 (d, J = 8.8 Hz, 2H), 7.63 (t, J = 7.8 Hz, 1H), 7.58 (d, J = 8.4 Hz, 1H), 7.51 (d, J = 8.8 Hz, 2H), 3.71(s, 3H), 3.22 (s, 3H). HRMS m/z calculated for C21H17ClN4O3S [M + H]+: 441.0783; found 441.0781. Purity 98.15% (tR 23.99 min), mp 296-298 °C.
6-(4-Chlorophenyl)-8-isobutyl-2-((3-(methylsulfonyl)phenyl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (32).
Synthesized from A11r and A13c by using the general procedure for 15 to give 32 (33%) as a pale yellow solid. 1H NMR (400 MHz, CDCl3) δ 8.66 (s, 1H), 8.32 (t, J = 1.8 Hz, 1H), 8.05 (d, J = 8.2 Hz, 1H), 7.74 (s, 1H), 7.71-7.56 (m, 5H), 7.41 (d, J = 8.8 Hz, 2H), 4.38 (d, J = 7.2 Hz, 2H), 3.11 (s, 3H), 2.40-2.30 (sep, J = 6.8, 8.4 Hz, 1H), 0.99 (d, J = 6.4 Hz, 6H). HRMS m/z calculated for C24H23ClN4O3S [M + H]+: 483.1252; found 483.1261. Purity 97.49% (tR 26.82 min), mp 225-227 °C.
6-(2-Chlorophenyl)-8-methyl-2-((3-(methylsulfonyl)phenyl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (33).
Synthesized from A11b and A13c by using the general procedure for 15 to give 33 (72%) as a white solid. 1H NMR (500 MHz, CDCl3) δ 8.70 (s, 1H), 8.64 (s, 1H), 7.84 (brs, 1H), 7.79 (d, J = 7.2 Hz, 1H), 7.66 (d, J = 9 Hz, 1H), 7.63 (s, 1H), 7.58 (t, J = 7.8 Hz, 1H), 7.49-7.47 (m, 1H), 7.38-7.32 (m, 3H), 3.85 (s, 3H), 3.10 (s, 3H). 13C NMR (150 MHz, CDCl3) δ 162.1, 158.5, 158.4, 155.7, 141.5, 140.0, 135.1, 134.9, 133.8, 131.6, 130.2, 129.8, 129.7, 129.3, 126.8, 123.9, 121.5, 117.9, 107.5, 44.6, 28.9. HRMS m/z calculated for C21H17ClN4O3S [M + H]+: 441.0783; found 441.0784. Purity 98.1% (tR 22.36 min), mp 230-232 °C.
6-(2,4-Dichlorophenyl)-8-methyl-2-((3-(methylsulfonyl)phenyl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (34).
Synthesized from A11a and A13c by using the general procedure for 15 to give 34 (63%) as a pale yellow solid. 1H NMR (500 MHz, CDCl3) δ 8.70 (s, 1H), 8.65 (s, 1H), 7.78 (d, J = 6.5 Hz, 2H), 7.67-7.56 (m, 3H), 7.50 (s, 1H), 7.32 (s, 2H), 3.84 (s, 3H), 3.10 (s, 3H). 13C NMR (125 MHz, CDCl3) δ 162.0, 158.6, 158.5, 155.7, 141.5, 139.9, 135.4, 134.9, 134.6, 133.4, 132.5, 130.2, 129.7, 128.0, 127.2, 123.9, 121.6, 118.0, 107.4, 44.6, 29.0. HRMS m/z calculated for C21H16Cl2N4O3S [M + H]+: 475.0393; found 475.0400. Purity 99.5% (tR 24.04 min), mp 231-233 °C.
6-(2,3-Dichlorophenyl)-8-methyl-2-((3-(methylsulfonyl)phenyl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (35).
Synthesized from A11g and A13c by using the general procedure for 15 to give 35 (68%) as a pale yellow solid. 1H NMR (600 MHz, DMSO-d6) δ 10.64 (s, 1H), 8.90 (s, 1H), 8.77 (s, 1H), 7.96 (m, 2H), 7.70 (d, J = 7.8 Hz, 1H), 7.63 (t, J = 8.1 Hz, 1H), 7.58 (d, J = 8.4 Hz, 1H), 7.45 (t, J = 7.8 Hz, 1H), 7.39 (d, J = 7.8 Hz, 1H), 3.69 (s, 3H), 3.23 (s, 3H). 13C NMR (150 MHz, DMSO-d6) δ 161.4, 159.9, 159.2, 155.6, 141.9, 141.0, 138.5, 136.3, 132.3, 131.9, 131.0, 130.7, 130.4, 128.7, 127.6, 124.4, 120.9, 117.6, 106.9, 44.2, 28.7. HRMS m/z calculated for C21H16Cl2N4O3S [M + H]+: 475.0393; found 475.0397. Purity 98.5% (tR 23.50 min), mp 270-272 °C.
6-(2-Chloro-4-fluorophenyl)-8-methyl-2-((3-(methylsulfonyl)phenyl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (36).
Synthesized from A11h and A13c by using the general procedure for 15 to give 36 (60%) as a white solid. 1H NMR (600 MHz, DMSO-d6) δ 10.63 (s, 1H), 8.90 (s, 1H), 8.77 (brs, 1H), 7.96 (d, J = 7.8 Hz, 1H), 7.93 (s, 1H), 7.63 (t, J = 8.1 Hz, 1H), 7.59-7.56 (m, 2H), 7.49-7.46 (m, 1H), 7.36 (td, J = 8.5, 3 Hz, 1H), 3.69 (s, 3H), 3.22 (s, 3H). 13C NMR (150 MHz, DMSO-d6) δ 162.9, 161.7, 161.3, 159.8, 159.1, 155.5, 141.9, 141.0, 136.6, 134.6, 134.5, 133.9, 132.5, 130.4, 126.8, 124.4, 120.9, 117.6, 117.2, 117.0, 114.9, 114.7, 106.9, 44.2, 28.7. HRMS m/z calculated for C21H16ClFN4O3S [M + H]+: 459.0688; found 459.0688. Purity 97.6% (tR 22.72 min), mp 248-250 °C.
6-(2,5-Dichlorophenyl)-8-methyl-2-((3-(methylsulfonyl)phenyl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (37).
Synthesized from A11i and A13c by using the general procedure for 15 to give 37 (92%) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.66 (s, 1H), 8.90 (s, 1H), 8.77 (s, 1H), 7.99-7.96 (m, 2H), 7.66-7.57 (m, 3H), 7.54-7.51 (m, 2H), 3.69 (s, 3H), 3.23 (s, 3H). HRMS m/z calculated for C21H16Cl2N4O3S [M + H]+: 475.0393; found 475.0390. Purity 97.4% (tR 23.76 min), mp 263-265 °C.
Preparation of 6-chloro-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A14).
The mixture of A7 (20 mg, 0.11 mmol) and 60% NaH (8 mg, 0.33 mmol) in THF (1.5 mL) was stirred at rt for 10 min under argon. A solution of ethyl 2-chloro-2-(diethoxyphosphoryl)acetate (42 mg, 0.16 mmol) in THF (0.5 mL) was added dropwise to and the resulting mixture was heated at reflux for 2.5 h. The reaction mixture was cooled to rt, concentrated and extracted using EtOAc and water. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated to give a residue that was purified by column chromatography using silica gel (30% EtOAc/Hexane) to afford A14 as a white solid (42% yield). 1H NMR (600 MHz, CDCl3) δ 8.59 (s, 1H), 7.82 (s, 1H), 3.80 (s, 3H), 2.62 (s, 3H). 13C NMR (150 MHz, CDCl3) δ (ppm) 173.6, 159.3, 155.7, 153.3, 132.6, 126.7, 109.0, 29.2, 14.6.
6-(2-Chloro-4-methylphenyl)-8-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (A15).
A14 (24 mg, 0.11 mmol), (2-chloro-4-methylphenyl)boronic acid (28 mg, 0.16 mmol) and Pd(PPh3)4 (13 mg, 0.011 mmol) were placed in a round-bottom flask and purged with argon for 10 min. DMF (1 mL) and CH3CN (2 mL) were added and the flask was again purged with argon for 10 min. A 1M solution of Na2CO3 (23 mg, 0.21 mmol) (220 μL) was added dropwise and the reaction mixture was heated at 90 °C for 5 h. The reaction mixture was allowed to cool to rt, the solvent was evaporated, and the residue was partitioned between water and EtOAc. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated to give a residue that was purified by column chromatography using silica gel (10% EtOAc/DCM) to afford A15 as a white solid (57%). 1H NMR (400 MHz, CDCl3) δ 8.64 (s, 1H), 7.64 (s, 1H), 7.31-7.23 (m, 2H), 7.14 (d, J = 7.6 Hz, 1H), 3.82 (s, 3H), 2.66 (s, 3H), 2.38 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 173.3, 161.9, 156.3, 154.3, 140.3, 134.8, 133.3, 131.6, 131.2, 130.4, 127.7, 109.4, 28.5, 21.1, 14.6.
6-(2-Chloro-4-methylphenyl)-8-methyl-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (A16).
This compound was synthesized by using the general procedure for the preparation of A11a. White solid (68% yield): 1H NMR (400 MHz, CDCl3) δ 8.99 (s, 1H), 7.80 (s, 1H), 7.34 (s, 1H), 7.27-7.24 (m, 1H), 7.17 (d, J = 8.4 Hz, 1H), 3.89 (s, 3H), 3.44 (s, 3H), 2.41 (s, 3H). 13C NMR (100 MHz, CDCl3) δ 164.4, 161.3, 157.1, 155.1, 141.2, 136.6, 133.6, 133.1, 130.9, 130.6, 130.5, 127.8, 115.1, 39.3, 29.3, 21.2.
6-(2-Chloro-4-methylphenyl)-8-methyl-2-((3-(methylsulfonyl)phenyl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (38).
Synthesized from A16 and A13c by using the general procedure for 15 to give 38 (75%) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 10.61 (s, 1H), 8.89 (s, 1H), 8.77 (s, 1H), 7.97 (d, J = 8.8 Hz, 1H), 7.88 (s, 1H), 7.65-7.57 (m, 2H), 7.39 (s, 1H), 7.29 (d, J = 8 Hz, 1H), 7.22 (d, J = 7.7 Hz, 1H), 3.69 (s, 3H), 3.23 (s, 3H), 2.36 (s, 3H). HRMS m/z calculated for C22H19ClN4O3S [M + H]+: 455.0939; found 455.0944. Purity 99.6% (tR 23.51 min), mp 182-184 °C.
6-(4-Hydroxyphenyl)-8-methyl-2-((3-(methylsulfonyl)phenyl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (39).
Synthesized from A11k and A13c by using the general procedure for 15 to give 39 (79%) as pale yellow solid. Note the TBS-protecting group from A11k was removed during the reaction. 1H NMR (400 MHz, DMSO-d6) δ 10.53 (s, 1H), 9.63 (s, 1H), 8.87 (s, 1H), 8.79 (s, 1H), 7.95-7.94 (m, 2H), 7.62 (t, J = 7.8 Hz, 1H), 7.57-7.53 (m, 3H), 6.82 (d, J = 8.8 Hz, 2H), 3.70 (s, 3H), 3.22 (s, 3H). HRMS m/z calculated for C21H18N4O4S [M + H]+: 423.1122; found 423.1120. Purity 95.0% (tR 19.63 min), mp 268-270 °C.
6-(4-Methoxyphenyl)-8-methyl-2-((3-(methylsulfonyl)phenyl)amino)pyrido[2,3-d]pyrimidin-7(8H)-one (40).
Synthesized from A11l and A13c by using the general procedure for 15 to give 40 (95%) as yellow solid. 1H NMR (400 MHz, DMSO-d6) δ 10.52 (s, 1H), 8.87-8.77 (m, 2H), 7.99-7.94 (m, 2H), 7.66-7.54 (m, 4H), 6.99 (d, J = 8.4 Hz, 2H), 3.79 (s, 3H), 3.70 (s, 3H), 3.21 (s, 3H). HRMS m/z calculated for C22H20N4O4S [M + H]+: 437.1278; found 437.1282. Purity 97.52% (tR 22.42 min), mp 291-293 °C.
Molecular docking studies.
AutoDock 4.2 (AutoDock Tools 1.5.6) was used to perform molecular docking studies. Ligands were docked into the ATP binding site of the protein using the ligand docking protocol in AutoDock 4.2. The structure of the protein was kept rigid, while the ligand structures were set partially flexible by setting the number of rotatable bonds. In brief, ligands were built using ChemBio3D and subjected to MM2 (force field) energy minimization. Using AutoGrid, the grid box was set to 70.00 Å, 60 Å and 50 Å along the x-, y- and z-axis, with 0.375 Å spacing. Docking calculations were carried out using the Lamarckian genetic algorithm with default parameters. Docking poses were selected based on the hydrogen bond interaction between the pyrimidine nitrogen (N-3) and amino −NH of the pyrido[2,3-d]pyrimidin-7-one with RIPK2 hinge residue Met98 and the highest binding energy (kcal/mol). Docking pose analyses were performed using the PyMOL Molecular Graphics System, Version 2.0 Schrodinger LLC software.
ADPGlo kinase assay.
Recombinant RIPK2 protein (20 ng per reaction) was diluted in the reaction buffer consisting of 40 mM Tris (pH 7.5); 20 mM MgCl2; 0.1 mg/ml BSA; 50 μM DTT. Diluted protein was added to low volume white 384 well plates (2 μL/well). Inhibitors were diluted 1:3 in reaction buffer, and then 1 μL was added to each well and incubated 5 min at room temperature. Reactions were initiated by the addition of 2 μL of 100 μM ATP and 1 mg/ml RS repeat peptide (SignalChem) in the reaction buffer. The final RIPK2 and substrate concentrations were 105 nM and 207 μM, respectively. Plates were sealed with plastic coverslips and incubated at room temperature for 2 h. Reactions were stopped by the addition of 5 μL of ADP-Glo reagent (Promega) and the ADP generation reaction was performed for 40 min at room temperature. Luminescence signal was generated by the addition of 10 μL of Kinase detection reagent (Promega) for 30 min at room temperature. Luminescence signals were determined using appropriate luminescence plate-reader (typical integration time 0.3-1 sec). To calculate percent inhibition, the average background signal was subtracted from test well and maximal signal wells. Inhibition, % = (1- (test signal/maximal signal))*100. The percent inhibition at a specified concentration was determined or IC50 values were calculated based on a dose range of inhibitor concentrations using non-linear regression in GraphPad Prism software. Compound 12 [18] and DMSO were used as a positive and negative controls, respectively.
Radioisotope filter binding kinase assay.
Enzyme inhibitory activity was evaluated by Reaction Biology Corp using a radiometric HotSpot™ assay system [34] by incubating human ALK2 with the protein substrate casein (1 mg/mL) and [ϒ-33P] ATP (10 μM) in the presence of various concentrations of test compounds (10 nM – 100 μM). After 30 min the amount of 33P-casein was determined. A plot of inhibitor concentration versus % activity was constructed and from this plot, an IC50 value was determined.
NOD2-RIPK2 signaling assay.
HEK-Blue cells expressing human NOD2 and NFkB-SEAP reporter (Invivogen) were seeded into 96 well clear plates at 7.5x103 cells per well in 100 μL of DMEM media supplemented with 10% FBS and 1% antibiotic-antimycotic mix. Cells were allowed to attach for 48 h in 5% CO2 tissue culture incubator at 37 °C. On the morning of the experiment, media in the wells was replaced with 100 μL of HEK-Blue detection media (Invivogen). Cells were treated with the inhibitors, diluted in DMSO (0.5 μL per well) for 15 min in 5% CO2 tissue culture incubator at 37 °C. After that, cells were stimulated by the addition of 1 ng/well L18-MDP (Invivogen). Cells were incubated in 5% CO2 tissue culture incubator at 37 °C for 8 h and absorbance, corresponding to the SEAP in the media, was determined in Wallac3V plate reader (Perkin Elmer). Inhibition, %= (1-((sample signal-unstimulated and DMSO treated cells)/(L18-MDP stimulated and DMSO treated cells - unstimulated and DMSO treated cells)))*100. IC50 values were calculated based on a dose range of inhibitor concentrations using non-linear regression in GraphPad Prism software.
Intracellular flow cytometry of CXCL8.
U2OS/NOD2 cells including RIPK2 KO cells reconstituted with WT RIPK2 or T95W mutant are described in Hrdinka et al. [19]. Cells were treated with inhibitors and stimulated with L18-MDP (InvivoGen) as indicated in figure legends. The intracellular staining of CXCL8 was performed as previously described [35]. The results were analyzed by FACS Canto Flow Cytometer (BD Biosciences, Franklin Lakes, NJ) and data processed using FlowJo software (FlowJo, LLC, Ashland, OR).
In vitro ADME studies of 33.
In vitro pharmacokinetic parameters such as metabolic stability (t1/2 in hepatic microsomes and intrinsic clearance), solubility and permeability were performed for 33. Hepatic microsomal stability was determined by incubating 1μM compound with 1 mg/mL mouse hepatic microsomes in 100 mM potassium phosphate buffer, pH 7.4 at 37 °C with continuous shaking. NADPH was added with 1 mM final concentration to initiate the reaction and 40 μL aliquots were removed from 300 μL of the final incubation volume at different time points (0, 5, 10, 20, 40 and 60 mins). These aliquots were then added to 160 μL acetonitrile to stop the reaction. NADPH dependence of the reaction was evaluated in parallel incubations without NADPH. The samples were centrifuged through a 0.45-micron filter plate at the end of the assay and analyzed by LC-MS/MS to determine the half-life of the compound. The kinetic aqueous solubility of 33 was determined by diluting 1 μL of a 10 mM DMSO stock with 99 μL PBS solution (pH 7.4), stored at room temperature for 24 h, filtered and analyzed by LC-MS/MS. These studies were performed by Cameron, et al. at the Scripps Research Institute, Florida.
Pharmacokinetic study of 33.
The pharmacokinetic study (Medicilon Preclinical Research LLC, Shanghai, China) was conducted in female ICR mice (N=18) following a 10 mg/kg single intraperitoneal administration [formulated with DMSO (5%), solutol (10%) and water (85%)]. Blood samples were collected at 0.25, 0.5, 1, 2, 6, and 24 h. Brain samples were collected after 0.5 and 2 h. The plasma and brain concentrations of 33 were determined by liquid chromatography–tandem mass spectrometry. The lower limit of detection in plasma samples was 5 ng/mL, while in brain samples were 50 ng/mL.
Supplementary Material
Highlights.
Demonstration of pyrido[2,3-d]pyrimidin-7-ones as potent inhibitors of RIPK2.
Installation of a sulfone enhanced selectivity versus structurally related ALK2.
SAR insights into achieving RIPK2 dependent NOD cell signaling inhibition.
In vitro ADME and in vivo PK characterization of inhibitor 33 illustrated.
Acknowledgments
This work was supported in whole or in part with funds from the National Institutes of Health, Department of Health and Human Services (CA190542, AI124049, AG058642, AI144400 and NS111395). Work in the M.G-H lab was supported by the Ludwig Institute for Cancer Research Ltd. and a Wellcome Trust Senior Research Fellowship (102894/Z/13/Z).
Abbreviations used
- ADME
adsorption, distribution, metabolism and excretion
- ALK2
activin receptor-like kinase 2
- ADP
adenosine diphosphate
- ATP
adenosine triphosphate
- AUC
area under the curve
- BRET
bioluminescence resonance energy transfer
- BSA
bovine serum albumin
- CARD
caspase-activated recruitment domain
- CL
clearance
- Cmax
concentration maximum
- CXCL8
C-X-C motif chemokine ligand 8
- DAP
diaminopimelic acid
- DFG
aspartic acid-phenyl alanine-glycine
- DMEM
Dulbecco’s modified eagle medium
- DTT
dithiothreitol
- FBS
fetal bovine serum
- HEPES
(4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid)
- HPLC
high-performance liquid chromatography
- HRMS
high resolution mass spectra
- IC50
half maximal inhibitory concentration
- IKK
IκB kinase
- IL
interleukin
- ip
intraperitoneal
- L18-MDP
lipidated form of muramyl dipeptide
- LUBAC
linear ubiquitin chain assembly complex
- MAPK
mitogen-activated protein kinase
- NF-κB
nuclear factor kappa-light-chain-enhancer of activated B cells
- NLR
NOD-like receptor
- NOD
nucleotide-binding oligomerization domain
- PAMPA
parallel artificial membrane permeability assay
- PBS
phosphate-buffered saline
- PDB
protein data bank
- PG
peptidoglycan
- PPI
protein-protein interaction
- RIPK2
receptor-interacting protein kinase 2
- SAR
structure-activity relationship
- SEAP
secreted embryonic alkaline phosphatase
- TAK1
transforming growth factor beta-activated kinase 1
- TNF
tumor necrosis factor
- Tris
tris(hydroxymethyl)aminomethane
- Ub
ubiquitin
- XIAP
X-linked inhibitor of apoptosis
Footnotes
Accession codes
PDB 5AR2 and 5J7B are available from the RCSB Protein Data Bank (http://www.rscb.org/).
Declaration of competing interest
S.N., A.D. and G.D.C. declare the following financial interests/ personal relationships which may be considered as potential competing interests: PCT patent application (Publication Number: US 2020/0172536 A1) assigned to the University of Houston System and Trustees of Tufts College. All other authors declare no conflict of interest about this article. All other authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Appendix A. Supplementary data
Supplementary data to this article can be found online at https://doi.org/10.1016/j.ejmech.2021.113252
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Declaration of interests
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
References
- (1).McCarthy JV; Ni J; Dixit VM RIP2 is a novel NF-κB-activating and cell death-inducing kinase. J. Biol. Chem 273 (1998) 16968–16975, 10.1074/jbc.273.27.16968. [DOI] [PubMed] [Google Scholar]
- (2).Dorsch M; Wang A; Cheng H; Lu C; Bielecki A; Charron K; Clauser K; Ren H; Polakiewicz RD; Parsons T; Li P; Ocain T; Xu Y Identification of a regulatory autophosphorylation site in the serine-threonine kinase RIP2. Cell Signal. 18 (12) (2006) 2223–2229, 10.1016/j.cellsig.2006.05.005. [DOI] [PubMed] [Google Scholar]
- (3).Girardin SE; Boneca IG; Viala J; Chamaillard M; Labigne A; Thomas G; Philpott DJ; Sansonetti PJ NOD2 is a general sensor of peptidoglycan through muramyl dipeptide (MDP) detection. J. Biol. Chem 278 (2003) 8869–8872, 10.1074/jbc.c200651200 [DOI] [PubMed] [Google Scholar]
- (4).Fridh V; Rittinger K The tandem CARDs of NOD2: Intramolecular interactions and recognition of RIP2. PLoS One. 7 (3) (2012) e34375, 10.1371/journal.pone.0034375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (5).Damgaard RB; Nachbur U; Yabal M; Wong WW; Fiil BK; Kastirr M; Rieser E; Rickard JA; Bankovacki A; Peschel C; Ruland J; Bekker-Jensen S; Mailand N; Kaufmann T; Strasser A; Walczak H; Silke J; Jost PJ; Gyrd-Hansen M The ubiquitin ligase XIAP recruits LUBAC for NOD2 signaling in inflammation and innate immunity. Mol Cell. 46 (6) (2012) 746–758, 10.1016/j.molcel.2012.04.014. [DOI] [PubMed] [Google Scholar]
- (6).Hrdinka M; Gyrd-Hansen M The Met1-Linked Ubiquitin Machinery: Emerging Themes of (De)regulation. Mol Cell. 68 (2) (2017) 265–280, 10.1016/j.molcel.2017.09.001. [DOI] [PubMed] [Google Scholar]
- (7).Foley KP; Desai B; Vossenkämper A; Reilly MA; Biancheri P; Wang L; Lipshutz DB; Connor J; Miller M; Haile PA; Casillas LN; Votta BJ; Gough PJ; MacDonald TT; Wouters CH; Rose CD; Bertin J OR3–001–RIP2 kinase is activated in Blau syndrome and IBD. Pediatr. Rheumatol 11 A3 (2013), 10.1186/1546-0096-11-S1-A3. [DOI] [Google Scholar]
- (8).Shaw PJ; Barr MJ; Lukens JR; McGargill MA; Chi H; Mak TW; Kanneganti TD Signaling via the RIP2 adaptor protein in central nervous system-infiltrating dendritic cells promotes inflammation and autoimmunity. Immunity. 34 (1) (2011) 75–84, 10.1016/j.immuni.2010.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (9).Argast GM; Fausto N; Campbell JS Inhibition of RIP2/RICK/CARDIAK activity by pyridinyl imidazole inhibitors of p38 MAPK. Mol. Cell. Biochem 268 (2005) 129–140, 10.1007/s11010-005-3701-0. [DOI] [PubMed] [Google Scholar]
- (10).Tigno-Aranjuez JT; Asara JM; Abbott DW Inhibition of RIP2’s tyrosine kinase activity limits NOD2-driven cytokine responses. Genes Dev. 24 (2010) 2666–2677, 10.1101/gad.1964410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (11).Tigno-Aranjuez JT; Benderitter P; Rombouts F; Deroose F; Bai X; Mattioli B; Cominelli F; Pizarro TT; Hoflack J; Abbott DW In vivo inhibition of RIPK2 kinase alleviates inflammatory disease. J. Biol. Chem 289 (43) (2014) 29651–29664, 10.1074/jbc.M114.591388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (12).Nachbur U; Stafford CA; Bankovacki A; Zhan Y; Lindqvist LM; Fiil BK; Khakham Y; Ko HJ; Sandow JJ; Falk H; Holien JK; Chau D; Hildebrand J; Vince JE; Sharp PP; Webb AI; Jackman KA; Muhlen S; Kennedy CL; Lowes KN; Murphy JM; Gyrd-Hansen M; Parker MW; Hartland EL; Lew AM; Huang DC; Lessene G; Silke J A RIPK2 inhibitor delays NOD signaling events yet prevents inflammatory cytokine production. Nat Commun. 6 (2015) 6442, 10.1038/ncomms7442. [DOI] [PubMed] [Google Scholar]
- (13).Canning P; Ruan Q; Schwerd T; Hrdinka M; Maki JL; Saleh D; Suebsuwong C; Ray S; Brennan PE; Cuny GD; Uhlig HH; Gyrd-Hansen M; Degterev A; Bullock AN Inflammatory signaling by NOD-RIPK2 is inhibited by clinically relevant type II kinase inhibitors. Chem Biol. 22 (9) (2015) 1174–1184, 10.1016/j.chembiol.2015.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (14).Haile PA; Votta BJ; Marquis RW; Bury MJ; Mehlmann JF; Singhaus R Jr.; Charnley AK; Lakdawala AS; Convery MA; Lipshutz DB; Desai BM; Swift B; Capriotti CA; Berger SB; Mahajan MK; Reilly MA; Rivera EJ; Sun HH; Nagilla R; Beal AM; Finger JN; Cook MN; King BW; Ouellette MT; Totoritis RD; Pierdomenico M; Negroni A; Stronati L; Cucchiara S; Ziolkowski B; Vossenkamper A; MacDonald TT; Gough PJ; Bertin J; Casillas LN The identification and pharmacological characterization of 6-(tert-Butylsulfonyl)-N-(5-fluoto-1H-indazil-3-yl)quinoline-4-amine (GSK583), a highly potent and selective inhibitor of RIP2 kinase. J. Med. Chem 59 (10) (2016) 4867–4880, 10.1021/acs.jmedchem.6b00211. [DOI] [PubMed] [Google Scholar]
- (15).Haile PA; Casillas LN; Votta BJ; Wang GZ; Charnley AK; Dong X; Bury MJ; Romano JJ; Mahlmann JF; King BW; Erhard KF; Hanning CR; Lipshutz DB; Desai BM; Capriotti CA; Schaeffer MC; Berger SB; Mahajan MK; Reilly MA; Nagilla R; Rivera EJ; Sun HH; Kenna JK; Beal AM; Ouellette MT; Kelly M; Stemp G; Convery MA; Vossenkamper A; MacDonald TT; Gough PJ; Bertin J; Marquis RW Discovery of first-in-class receptor interacting protein 2 (RIP2) kinase specific clinical candidate, 2-((4-(Benzo[d]thiazol-5-ylamino)-6-(tert-butylsulfonyl)quinazolin-7-yl)oxy)ethyl dihydrogen phosphate, for the treatment of inflammatory diseases. J. Med. Chem 62 (14) (2019) 6482–6494, 10.1021/acs.jmedchem.9b00575. [DOI] [PubMed] [Google Scholar]
- (16).He X; Da Ros S; Nelson J; Zhu X; Jiang T; Okram B; Jiang S; Michellys PY; Iskandar M; Espinola S; Jia Y; Bursulaya B; Kreusch A; Gao MY; Spraggon G; Baaten J; Clemmer L; Meeusen S; Huang D; Hill R; Nguyen-Tran V; Fathman J; Liu B; Tuntland T; Gordon P; Hollenbeck T; Ng K; Shi J; Bordone L; Liu H Identification of potent and selective RIPK2 inhibitors for the treatment of inflammatory diseases. ACS Med. Chem Lett. 8 (10) (2017) 1048–1053, 10.1021/acsmedchemlett.7b00258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (17).Shairaz B; Carlos VM; Rodrigo AO; Ratmir D Preparation of substituted benzamides as RIPK2 inhibitors as antiinflammatory and antitumor agents. Patent WO 2019161495 A1, 2019.
- (18).Suebsuwong C; Dai B; Pinkas DM; Duddupudi AL; Li L; Bufton JC; Schlicher L; Gyrd-Hansen M; Hu M; Bullock AN; Degterev A; Cuny GD Receptor-interacting protein kinase 2 (RIPK2) and nucleotide-binding oligomerization domain (NOD) cell signaling inhibitors based on a 3,5-diphenyl-2-aminopyridine scaffold. Eur. J. Med. Chem 200 (2020) 112417, 10.1016/j.ejmech.2020.112417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (19).Hrdinka M; Schlicher L; Dai B; Pinkas DM; Bufton JC; Picaud S; Ward JA; Rogers C; Suebsuwong C; Nikhar S; Cuny GD; Huber KVM; Filippakopoulos P; Bullock AN; Degterev A; Gyrd-Hansen M Small molecule inhibitors reveal an indispensable scaffolding role of RIPK2 in NOD2 signaling. EMBO J, 37 (17) 2018. e99372, 10.15252/embj.201899372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (20).Cowan-Jacob SW; Fendrich G; Floersheimer A; Furet P; Liebetanz J; Rummel G; Rheinberger P; Centeleghe M; Fabbro D; Manley PW Structural biology contributions to the discovery of drugs to treat chronic myelogenous leukaemia. Acta Cryst. D 63 (2007) 80–93, 10.1107/S0907444906047287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (21).Panek RL; Lu GH; Klutchko SR; Batley BL; Dahring TK; Hamby JM; Hallak H; Doherty AM; Keiser JA In vitro pharmacological characterization of PD 166285, a new nanomolar potent and broadly active protein tyrosine kinase inhibitor. J Pharmacol Exp Ther. 283 (3) (1997) 1433–1444, https://jpet.aspetjournals.org/content/283/3/1433. [PubMed] [Google Scholar]
- (22).Mohedas AH; Xing X; Armstrong KA; Bullock AN; Cuny GD; Yu PB Development of an ALK2-biased BMP type I receptor kinase inhibitor. ACS Chem Biol. 8 (6) (2013) 1291–1302, 10.1021/cb300655w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (23).Mohedas AH; Wang Y; Sanvitale CE; Canning P; Choi S; Xing X; Bullock AN; Cuny GD; Yu PB Structure-activity relationship of 3,5-diaryl-2-aminopyridine ALK2 inhibitors reveals unaltered binding affinity for Fibrodysplasia Ossificans Progressiva causing mutants. J. Med. Chem 57 (19) (2014) 7900–7915, 10.1021/jm501177w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (24).Buzko O; Shokat KM A kinase sequence database: sequence alignments and family assignment. Bioinformatics. 18 (9) (2002) 1274–1275, 10.1093/bioinformatics/18.9.1274. [DOI] [PubMed] [Google Scholar]
- (25).Boschelli DH; Wu Z; Klutchko SR; Hollis Showalter HD; Hamby JM; Lu GH; Major TC; Dahring TK; Batley B; Panek RL; Keiser J; Hartl BG; Kraker AJ; Klohs WD; Roberts BJ; Patmore S; Elliot WL; Steinkampf R; Bradford LA; Hallak H; Doherty AM Synthesis and tyrosine kinase inhibitory activity of a series of 2-Amino-8H-pyrido[2,3-d]pyrimidines: Identification of potent, selective platelet-derived growth factor receptor tyrosine kinase inhibitors. J. Med. Chem 41 (22) (1998) 4365–4377, 10.1021/jm980398y. [DOI] [PubMed] [Google Scholar]
- (26).Klutchko SR; Hamby JM; Boschelli DH; Wu Z; Kraker AJ; Amar AM; Hartl BG; Shen C; Klohs WD; Steinkampf RW; Driscoll DL; Nelson JM; Elliot WL; Roberts BJ; Stoner CL; Vincent PW; Dykes DJ; Panek RL; Lu GH; Major TC; Dahring TK; Hallak H; Bradford LA; Hollis Showalter HD; Doherty AM 2-Substituted Aminopyrido[2,3-d]pyrimidin-7(8H)-ones. Structure-activity relationships against selected tyrosine kinases and in vitro and in vivo anticancer activity. J. Med. Chem 41 (17) (1998) 3276–3292, 10.1021/jm9802259. [DOI] [PubMed] [Google Scholar]
- (27).Kraus GA; Gupta V; Mokhtarian M; Mehanovic S; Nilsen-Hamilton M New effective inhibitors of the Abelson kinase. Bioorg. Med. Chem 18 (17) (2010) 6316–6321, 10.1016/j.bmc.2010.07.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (28).Jaekyoo L; Jang-Sik J; Hae-Jun H; Ho-Juhn S; Jung-Ho K; Se-Won K; Jong Sung K; Jaesang L; Tae-Im L; Yung-Geun C; Sung-Ho P; In Yong L; Byung-Chul S; Paresh Devidas S; Dong-Sik J Substituted pyrimidine compounds and their use as SYK inhibitors. Patent WO2015061369 A1, 2015.
- (29).VanderWel SN; Harvey PJ; McNamara DJ; Repine JT; Keller PR; Quin J 3rd.; Booth RJ; Elliott WL; Dobrusin EM; Fry DW; Toogood PL Pyrido[2,3-d]pyrimidin-7-ones as specific inhibitors of cyclin-dependent kinase 4. J. Med. Chem 48 (7) (2005) 2371 – 2387, 10.1021/jm049355. [DOI] [PubMed] [Google Scholar]
- (30).Charnley AK; Convery MA; Lakdawala Shah A; Jones E; Hardwicke P; Bridges A; Ouellette M; Totoritis R; Schwartz B; King BW; Wisnoski DD; Kang J; Eidam PM; Votta BJ; Gough PJ; Marquis RW; Bertin J; Casillas L Crystal structures of human RIP2 kinase catalytic domain complexed with ATP-competitive inhibitors: Foundations for understanding inhibitor selectivity. Bioorg. Med. Chem 23 (21) (2015) 7000–7006, 10.1016/j.bmc.2015.09.038. [DOI] [PubMed] [Google Scholar]
- (31).Sanvitale CE; Kerr G; Chaikuad A; Ramel M-C; Mohedas AH; Reichert S; Wang Y; Triffitt JT; Cuny GD; Yu PB; Hill CS; Bullock AN A new class of small molecule inhibitor of BMP signaling. PLoS One, 8 (4) (2013) e62721, 10.1371/journal.pone.0062721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (32).Degterev A; Huang Z; Boyce M; Li Y; Jagtap P; Mizushima N; Cuny GD; Mitchison TJ; Moskowitz MA; Yuan J Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol. 1 (2) (2005) 112–119, 10.1038/nchembio711. [DOI] [PubMed] [Google Scholar]
- (33).Cuny GD; Degterev A RIPK protein kinase family: Atypical lives of typical kinases. Semin Cell Dev Biol. (2020) S1084–9521, 10.1016/j.semcdb.2020.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (34).Anastassiadis T; Deacon SW; Devarajan K; Ma H; Peterson JR Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity. Nat Biotechnol. 29 (11) (2011) 1039–1045, 10.1038/nbt.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (35).Hrdinka M; Fiil BK; Zucca M; Leske D; Bagola K; Yabal M; Elliott PR; Damgaard RB; Komander D; Jost PJ; Gyrd-Hansen M CYLD limits Lys63- and Met1-linked ubiquitin at receptor complexes to regulate innate immune signaling. Cell Rep, 14 (12) (2016) 2846–2858, 10.1016/j.celrep.2016.02.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.






