2,279
Views
0
CrossRef citations to date
0
Altmetric
Research Papers

Identification of highly selective type II kinase inhibitors with chiral peptidomimetic tails

ORCID Icon, , , , , , , , , , ORCID Icon, , ORCID Icon, , , & show all
Pages 1257-1277 | Received 14 Jan 2022, Accepted 12 Apr 2022, Published online: 28 Apr 2022

Abstract

Identification of highly selective type II kinase inhibitors is described. Two different chiral peptidomimetic scaffolds were introduced on the tail region of non-selective type II kinase inhibitor GNF-7 to enhance the selectivity. Kinome-wide selectivity profiling analysis showed that type II kinase inhibitor 7a potently inhibited Lck kinase with great selectivity (IC50 of 23.0 nM). It was found that 7a and its derivatives possessed high selectivity for Lck over even structurally conserved all Src family kinases. We also observed that 7a inhibited Lck activation in Jurkat T cells. Moreover, 7a was found to alleviate clinical symptoms in DSS-induced colitis mice. This study provides a novel insight into the design of selective type II kinase inhibitors by adopting chiral peptidomimetic moieties on the tail region.

1. Introduction

Most signal transduction processes are mediated through a phosphotransfer reactions catalysed by kinases. However, overexpression or mutation of kinases causes tumour cell proliferation and survival. Therefore, kinases are pursued as invaluable targets and a tremendous amount of effort has been devoted towards the discovery of small molecular kinase inhibitors for the treatment of cancer for decadesCitation1. Small molecule kinase inhibitors have been classified by binding modes with protein kinases. Type I inhibitors are the most commonly encountered and occupy the ATP-binding site of the active conformation of kinases (i.e. DFG-in conformation). In contrast to type I inhibitors, type II kinase inhibitors recognise the ATP-binding pocket of inactive DFG-out conformation of kinase proteins. Besides ATP competitive inhibitors (e.g. type I, type II), allosteric and covalent inhibitors have also been investigatedCitation1–4. Although a significant number of small molecular kinase inhibitors have been developed, discovery of selective kinase inhibitors remains challenging. The rationale behind this is that the structure of ATP-binding site in all of the kinase proteins is highly conservedCitation1. In addition, designing an allosteric kinase inhibitor, which is the most selective inhibitor, is difficult since it highly relies on an empirical exerciseCitation2a. Although selective kinases have been uncovered using subtle 3-dimensional structural differences among the kinasesCitation5, discovery of highly selective kinase inhibitors still remains a largely unmet challenge. Type II kinase inhibitors were anticipated to be more selective compared to type I kinase inhibitors in the early phase since the hydrophobic pocket generated by the DFG-out conformation is not quite conserved in contrast to ATP binding pocket and DFG-out conformations are more dynamic. However, type II kinase inhibitors, developed to date have been proven to be largely less selective than type I kinase inhibitors.Citation2a

We noticed that a number of the less selective type II kinase inhibitors possess achiral and limited chemotype tails, which interact with allosteric site of the inactive conformation of kinases. We envisioned that interactions of flat and achiral tail fragments with 3-dimensional structural kinases would be highly limited and resulted in less selective profileCitation6. Thus, we were curious whether type II kinase inhibitors containing chiral tails would be more selective among all of the kinases by affording various binding modesCitation7,Citation8. We were particularly interested in peptidomimetic structure as tail scaffolds. To our best knowledge, development of selective type II kinase inhibitors that consists of peptidomimetic tails is unprecedented. To investigate how peptidomimetic tail structure affects kinase selectivities, we chose the core structure of GNF-7 ()Citation9. GNF-7 has been discovered as a type II T315I Bcr-Abl kinase inhibitor and possesses remarkable potencies against many kinases with highly low kinase selectivity. Among the peptidomimetic structure, we were attracted to synthetically easily accessible solution phase turn mimetic librariesCitation10.

Figure 1. (a) Structure of GNF-7. (b) Designing selective type II kinase inhibitors with peptidomimetic scaffolds.

Figure 1. (a) Structure of GNF-7. (b) Designing selective type II kinase inhibitors with peptidomimetic scaffolds.

2. Results and discussion

2.1. Chemistry

We initially prepared kinase inhibitors, containing small molecular β-turn mimetic scaffolds developed by the Miller group (Scheme 1(a))Citation10b,f. Alkylation of chloromethyl pyrimidine 2 with aniline 1 provided tert-butyl ester 3 under basic conditions in 81% yield. Nucleophilic aromatic substitution of 2,4-dichloropyrimidine 3 with methylamine, followed by cyclic urea formation using triphosgene afforded urea 4. Buchwald coupling of chloropyrimidine 4 with 5-aminopicoline and subsequent removal of tert-butyl group under acidic conditions smoothly generated acid 5. Amide coupling of acid 5 with various β-turn mimetic scaffolds 6 produced amides 7 in 10–20% yields. We also prepared kinase inhibitors containing benzodiazepines as turn mimetic scaffolds (Scheme 1(b)). Amide coupling of benzodiazepines 9 and 10Citation10d,e with aniline 8Citation9a,c afforded amides 11 in 10–20% yields.

Scheme 1. (a) Synthesis of kinase inhibitors containing turn mimetic amide scaffolds. (b) Synthesis of kinase inhibitors possessing benzodiazepines.

Scheme 1. (a) Synthesis of kinase inhibitors containing turn mimetic amide scaffolds. (b) Synthesis of kinase inhibitors possessing benzodiazepines.

2.2. In vitro kinase inhibitory activities

Kinase-inhibitory activities of both 7 and 11 against four selected kinases were assessed by in vitro kinase assay (). To our delight, both 7a and 11b showed high degree of selectivities among selected kinases even between structurally similar Lck and c-Src (over 10-fold selectivities). Replacement of L-Pro with D-Pro led to lower selectivities between Lck and c-Src (7a and 7b). This is not surprising since stereochemical alteration of Pro at i + 1 might potentially change 3-dimensional conformation of the structureCitation10b. The substituents at N-1, C-3, and C-5 positions on the 1,4-benzodiazepin-2-one ring significantly affected selectivities. In all cases, the activities on Lck kinases were found to be superior to those on other kinases. Gratifyingly, 11b possessed over 10-fold selectivity for Lck over c-Src. The iso-butyl group at C-3 position in the 1,4-benzodiazepin-2-one surpassed the methyl group in respect of the selectivity (11b and 11d). Also, the 2-butyl group at the C-3 position causes almost no selectivity (11e).

Table 1. In vitro potency profiling on selected kinases.

With the exciting initial data in hand, selectivities between Lck and c-Src were investigated on derivatives 7 containing the β-turn mimetic scaffolds (). Kinase-inhibitory activities of derivatives 7 against Lck and c-Src were assessed by in vitro kinase assay. Interestingly, 7c possessing alternative stereochemistry compared to 7a showed significantly (>10-fold) diminished selectivity. Changing i + 2 functional groups to cyclopropane (7d and 7e), cyclobutane (7f), and even glycine (7g and 7h) exhibited high selectivities. However, we observed diminished selectivity with benzyl substitution at i + 2 position (7i). High selectivity was kept with piperidine scaffold at i + 1 (7j). In contrast to the pyrrolidine and the piperidine groups, addition of the azetidine at i + 1 (7k) resulted in over 10-fold lower selectivity. The selectivities were not decreased by replacement of iso-butyl with valine at i + 3 (7l, 7m, 7n, and 7o). Substituents at i significantly affected selectivities (7p, 7q, 7r, 7s, and 7t). High selectivities were observed with iso-butyl and cyclohexyl substitution at i (over 10-fold selectivities, 7q and 7s). Compared to 7a, The selectivities of 7v and 7w on Lck and c-Src were highly diminishedCitation10f.

Table 2. In vitro potency profiling on Lck and c-Src.

Additionally, we investigated the selectivities of derivatives containing benzodiazepine scaffolds on Lck and c-Src kinases (). Kinase-inhibitory activities of derivatives 11 against Lck and c-Src were assessed by in vitro kinase assay. The selectivities of 11f was similar to those of its enantiomer 11a and p-fluorobenzyl 11g. It is noteworthy that the benzyl group at N-1 position of the 1,4-benzodiazepin-2-one moiety (11b) was superior to the methyl, allyl, iso-butyl, methoxyethyl, and N-benzylacetamide groups at the corresponding position in terms of selectivities (11h, 11i, 11j, 11n, and 11o). The methyl group in 11k and 11l at C-5 position is slightly more favourable than the phenyl group (11a and 11f) at the corresponding position as regards the selectivity. We observed that introduction of γ-turn mimicryCitation10e at the tail position resulted in almost no selectivity for Lck over c-Src (11p and 11q).

Table 3. In vitro potency profiling on Lck and c-Src.

We explored in vitro potencies of 7a and 11b at both 14 µΜ (Km) and 1 mM ATP concentrations to investigate whether these inhibitors are ATP competitive inhibitors (). We observed that the IC50 values were increased dramatically at 1 mM ATP concentration compared to those at Km ATP concentrations. Thus, we concluded that both 7a and 11b are ATP competitive kinase inhibitors.

Table 4. Biochemical IC50 values of 7a and 11b on Lck at Km and 1 mM ATP concentration.

2.3. Kinome-wide selectivities of 7a described on a kinome phylogenetic tree

We were pleased to confirm that 7a possesses higher selectivity for Lck over other 373 kinases at 1 µM concentration compared to GNF 7 (, Supplementary Table S1–S4). We also obtained IC50 values of 7a against five kinases (Lck, DDR1, Fgr, Bmx, and Blk), which were inhibited greater than 70% in the kinome-wide profiling analysis (, Supplementary Table S1). As shown in has an IC50 value of 23 nM against Lck and possesses 5 to 13-fold selectivity over these four kinases. Furthermore, 7a showed more than 10-fold selectivities over other structurally similar Src family kinases ().

Figure 2. This illustration was reproduced courtesy of Cell Signalling Technology, Inc. (www.cellsignal.com). (a) Kinome phylogenetic tree description of the GNF 7 selectivity profile. (b) Kinome-wide selectivities of 7a described on a kinome phylogenetic tree. (c) In vitro IC50 values of 7a against kinases, which were inhibited greater than 70%. (d) In vitro IC50 values of 7a against Src family kinases.

Figure 2. This illustration was reproduced courtesy of Cell Signalling Technology, Inc. (www.cellsignal.com). (a) Kinome phylogenetic tree description of the GNF 7 selectivity profile. (b) Kinome-wide selectivities of 7a described on a kinome phylogenetic tree. (c) In vitro IC50 values of 7a against kinases, which were inhibited greater than 70%. (d) In vitro IC50 values of 7a against Src family kinases.

2.4. Docking into Lck binding site

Docking of 7a and 11b with Lck kinase by long time (3 μs) molecular dynamics (MD) simulation revealed that turn peptidomimetic scaffolds were located at an allosteric binding site. Also, hydrophobic groups of the peptidomimetic scaffolds interacted with the allosteric helix of the binding site (C-helix in ). The results reveal that the amide moiety of 7a participates in hydrogen-bonding networks with Glu288 in Lck. Additionally, the phenyl group of 11b interacts with Phe354 by π–π stacking interaction. The core scaffolds of 7a and 11b were interacted with kinases hinge region through similar binding poses of the crystal structure of imatinib with kinases (Supplementary Figure S1). The Asp from the “DFG-motif” interacts with the amide functional group of 7a and 11b. Moreover, phenyl moieties on the linker region of 7a and 11b interacts with Lys273 by cation-π interaction.

Figure 3. (a) Predicted structure of Lck and 7a complex. (b) Predicted structure of Lck and 11b complex. (red dashed line: π–π interaction/cation-π interaction, yellow dashed line: hydrogen-bonding).

Figure 3. (a) Predicted structure of Lck and 7a complex. (b) Predicted structure of Lck and 11b complex. (red dashed line: π–π interaction/cation-π interaction, yellow dashed line: hydrogen-bonding).

2.5. Watermap application

Next, we further analysed binding sites using WaterMap applicationCitation11,Citation12. With WaterMap results, many hydration sites with unstable energy (0 > kcal/mol) were found in the allosteric binding site (Supplementary Figure S2). Therefore, when 7a was located at the unstable hydration sites, high binding free energy compensation could be obtained. Five hydration sites with a high energy of over 3.00 kcal/mol were found in Lck, and the peptidomimetic scaffold of 7a occupied these sites (). We envisioned that the energetically unstable regions at the allosteric site of Lck could be compensated with interaction of terminal turn peptidomimetic tail of 7a, which would contribute to high selectivity of 7a for Lck kinase.

Figure 4. Analysis results of Lck and superimpose of 7a at the allosteric site by using WaterMap application. Coloured sphere: hydration sites occupied by the inhibitor. The number on the sphere: ΔG energy (kcal/mol). Green sphere: low energy. Red sphere: high energy over 5 kcal/mol.

Figure 4. Analysis results of Lck and superimpose of 7a at the allosteric site by using WaterMap application. Coloured sphere: hydration sites occupied by the inhibitor. The number on the sphere: ΔG energy (kcal/mol). Green sphere: low energy. Red sphere: high energy over 5 kcal/mol.

With the interesting WaterMap calculation result, we attempted to explain the IC50 difference of 7a and its enantiomer 7c on Lck by the WaterMap application method (). The U-shaped tail of 7a is located in the allosteric binding site on Lck. Also, amino acids at i and i + 3 of 7a tail interact with allosteric helix. However, turn structure of 7c is not fully located at the allosteric binding site of Lck, which is expected to be highly important for kinase selectivities. Although the amino acid at i of 7c tail interacts with allosteric helix, the amino acid at i + 3 is extruded outside and exposed to solvent. Thus, the tail structure of 7c could have high fluctuation, which results in lower selectivities ().

Figure 5. (a) Analysis results of Lck and superimpose of turn scaffold of 7a at the allosteric site by using WaterMap application. (b) Analysis results of Lck and superimpose of turn scaffold of 7c, which is enantiomer of 7a at the allosteric site by using WaterMap application. (green dashed line: van der Waals interaction).

Figure 5. (a) Analysis results of Lck and superimpose of turn scaffold of 7a at the allosteric site by using WaterMap application. (b) Analysis results of Lck and superimpose of turn scaffold of 7c, which is enantiomer of 7a at the allosteric site by using WaterMap application. (green dashed line: van der Waals interaction).

2.6. Inhibitory effect of 7a on Lck activation

We investigated whether Lck activation was affected by 7a in Jurkat cell line (). Western blot analysis revealed that 7a markedly reduced the phosphorylated Lck levels at tyrosine 394 residue in a concentration-dependent manner in anti-CD3-treated Jurkat T cells. These results suggested that 7a inhibited the anti-CD3-activated Lck, similar to positive control A770041.

Figure 6. Effect of 7a on the Lck (Y394) activation in anti-CD3-treated Jurkat cells. After treatment with CD3 antibody for coating in plates, cells were seeded and then treated with various concentrations of 7a (10, 50, or 100 μM) or A770041 (1 μM) used as a positive control. The phosphorylation of Lck tyrosine in Jurkat cells was activated by anti-CD3 mAb. Total cellular protein was resolved by SDS-PAGE, transferred to PVDF membranes, and detected with specific p-Lck (Y394) antibody. β-Actin was used as an internal control.

Figure 6. Effect of 7a on the Lck (Y394) activation in anti-CD3-treated Jurkat cells. After treatment with CD3 antibody for coating in plates, cells were seeded and then treated with various concentrations of 7a (10, 50, or 100 μM) or A770041 (1 μM) used as a positive control. The phosphorylation of Lck tyrosine in Jurkat cells was activated by anti-CD3 mAb. Total cellular protein was resolved by SDS-PAGE, transferred to PVDF membranes, and detected with specific p-Lck (Y394) antibody. β-Actin was used as an internal control.

2.7. In vivo experiment with dextran sulphate sodium (DSS)-induced colitis model

Inflammatory bowel disease (IBD), which is a chronic and immune-mediated disorder of the gastrointestinal tract encompasses Crohn’s disease (CD) and ulcerative colitis (UC)Citation13. Although the exact cause of IBD is unclear, it is widely accepted that an excessive immune response against normal components of microflora results in IBD. Especially, excessive T cell activation plays a pivotal role in mucosal damage in both CD and UCCitation14. Lck plays a crucial role in activation of TCR-linked signal transduction pathways, leading to T cell activation and proliferationCitation15. Additionally, it is reported that overexpression of Lck leads to IBDCitation15. Hence, we evaluated the potential of our selective Lck kinase inhibitor 7a for IBD treatment with dextran sulphate sodium (DSS)-induced colitis model. DSS administration induces acute colonic damage, and changes in clinical parameters can be monitoredCitation16. To determine the recovery effect of 7a in DSS-induced colitis, we assessed the clinical symptom including disease activity index (DAI) and colon length. The DAI scores were evaluated by body weight loss, stool consistency, and occult/gross bleeding (). During the administration of DSS (4%) for 7 days, DAI values were significantly increased (). We discovered that 7a treatment (5 mg/kg, i.p.) improved the symptom changes at the end of experiments (). In addition, the colon length of DSS-treated group was significantly shorter than that of the vehicle-administered control group (8.17 ± 0.32 cm vs. 4.25 ± 0.38 cm, p < 0.001), while 7a treatment (5 mg/kg, i.p.) recovered the DSS-induced colon shortening (4.25 ± 0.38 cm vs. 5.47 ± 0.60 cm, p < 0.05, ).

Figure 7. Effect of 7a on the progression of DSS-induced colitis. (a) Disease activity index (DAI) levels during total experiments periods and (b) at the end of the experiment (day 7). (c, d) Colon length was measured at the end of the experiment (day 7). Data are presented as mean ± SE (n = 6). #p < 0.05, ##p < 0.01, ###p < 0.001 vs. the vehicle-treated control group; *p < 0.05, **p < 0.01 vs. the DSS-treated group. The significance between groups was determined by ANOVA and Dunnett's post-hoc test.

Figure 7. Effect of 7a on the progression of DSS-induced colitis. (a) Disease activity index (DAI) levels during total experiments periods and (b) at the end of the experiment (day 7). (c, d) Colon length was measured at the end of the experiment (day 7). Data are presented as mean ± SE (n = 6). #p < 0.05, ##p < 0.01, ###p < 0.001 vs. the vehicle-treated control group; *p < 0.05, **p < 0.01 vs. the DSS-treated group. The significance between groups was determined by ANOVA and Dunnett's post-hoc test.

Table 5. Assessment of the disease activity index (DAI)

3. Conclusion

In conclusion, we discovered highly selective type II kinase inhibitors by introducing chiral turn peptidomimetic moieties on the tail region for the first time. It turned out that 7a, a novel type II kinase inhibitor, is a potent and exceptionally selective Lck inhibitor. Based on kinome-wide selectivity profiling data, it was confirmed that 7a possesses high selectivity. Kinases selectivities were highly affected by subtle changes of the substituents of peptidomimetic scaffolds. To the best of our knowledge, it has never been reported that low selectivity of a type II kinase inhibitor is dramatically enhanced by adopting chiral peptidomimetic tail. The western blot analysis revealed that 7a is capable of inhibiting Lck activation in anti-CD3-treated Jurkat T cells. Finally, we discovered that 7a could alleviate of clinical symptoms in DSS-induced colitis mice. This study may shed a bright light on the design of selective type II kinase inhibitors by adopting chiral peptidomimetic tails.

4. Experimental

4.1. Chemistry

4.1.1. General procedures

Unless otherwise stated, reactions were performed in flame-dried glassware under a nitrogen atmosphere using dry solvents. Reaction progress was monitored by thin-layer chromatography (TLC). Purified water was obtained using a Barnstead NANOpure Infinity UV/UF system. Brine solutions are saturated aqueous solutions of sodium chloride. Commercially available reagents were purchased from Sigma-Aldrich, Acros Organics, Combi-Blocks, TCI or Alfa Aesar and used as received unless otherwise stated. Reaction temperatures were controlled by an IKAmag temperature modulator unless otherwise indicated. TLC was performed using E. Merck silica gel 60 F254 precoated glass plates (0.25 mm) and visualised by UV fluorescence quenching, KMnO4, or Ninhydrin staining. Silicycle SiliaFlash P60 Academic Silica gel (particle size 0.040–0.064 mm) was used for flash column chromatography. 1H NMR spectra were recorded on Bruker 400 MHz and 600 MHz spectrometer and are reported relative to residual CDCl3 (δ 7.26 ppm), CD3OD (δ 3.31 ppm) or (CD3)2SO (δ 2.50 ppm). 13C NMR spectra are recorded on Bruker 400 MHz and 600 MHz spectrometer (101 MHz & 151 MHz) and are reported relative to CDCl3 (δ 7.26 ppm), CD3OD (δ 3.31 ppm) or (CD3)2SO (δ 2.50 ppm). 19F NMR spectrum is recorded on Bruker 600 MHz spectrometer (377 MHz) and is reported relative to (CD3)2SO (δ 2.50 ppm). Data for 1H NMR are reported as follows: s = singlet, d = doublet, t = triplet, q = quartette, p = pentet, sept = septuplet, m = multiplet, br s = broad singlet, br d = broad doublet, app = apparent. Data for 13C are reported in terms of chemical shifts (δ ppm). Data for 19F is reported in terms of chemical shifts (δ ppm). The purity of final compounds was determined to be ≥95% using HPLC analyses performed on an Agilent 1100 series with a Poroshell C18 column (pore size: 120 Å; particle size: 4 µm, dimensions: 4.6 × 150 mm). Some final compounds are purified using PREP HPLC performed on an Agilent 1260 Infinity II with Agilent Prep-C18 column (particle size: 10 µm, dimensions: 250 × 21.2 mm). IR spectra were obtained using a Nicolet Avatar 330 FT-IR spectrometer and Bruker Alpha Platinum-ATR using thin films deposited on NaCl plates and reported in frequency of absorption (cm−1). Optical rotations were measured with a Rudolph AUTOPOL I automatic polarimeter operating on the sodium D-line (589 nm), using a 100 nm path-length cell and are reported as: [α]DT (concentration in g/100 ml, solvent). High resolution mass spectra (HRMS) were obtained from Waters SYNAPT G2 TOF with a Waters Multimode source in electrospray ionisation (ESI+), atmospheric pressure chemical ionisation (APCI+), or mixed ionisation mode (MM: ESI-APCI+).

4.1.2. Tert-butyl 3-amino-4-methylbenzoate (1)

To a solution of tert-butyl 4-methyl-3-nitrobenzoate (113 mg, 0.475 mmol, 1.00 equiv) in EtOH (1.18 ml) was added Pd/C (24.0 mg, 0.574 mmol), cyclohexene (1.18 ml). The reaction mixture was stirred for 16 h at 80 °C. Solids were removed via a filtration through a celite plug and the resulting solution was concentrated under reduced pressure. The filtrate was purified by flash column chromatography (4:1 hexanes:EtOAc) on silica gel to give aminobenzoate 1 (91.0 mg, 92% yield) as a yellow liquid. Rf: 0.55 (4:1 hexanes:EtOAc); 1H NMR (400 MHz, DMSO) δ 7.17 (d, J = 1.4 Hz, 1H), 7.00 (t, J = 1.2 Hz, 2H), 5.07 (s, 2H), 2.08 (s, 3H), 1.51 (s, 9H); 13 C NMR (101 MHz, DMSO) δ 165.60, 146.64, 129.79, 129.70, 126.17, 116.80, 114.09, 79.75, 30.69, 27.87, 17.57; IR (Neat) 3470, 3377, 2975, 2927, 2857, 1697, 1625, 1577, 1508, 1477, 1456, 1423, 1392, 1367, 1300, 1247, 1164, 1144, 1108, 1071, 1032, 997, 948, 886, 851, 825, 760, 642, 562, 527, 440 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C12H18NO2 [M + H]+: 208.1338; found: 208.1334.

4.1.3. Tert-butyl 3-(((2,4-dichloropyrimidin-5-yl)methyl)amino)-4-methylbenzoate (3)

To a solution of aminobenzoate 1 (1.20 g, 5.97 mmol, 1.00 equiv), 2,4-dichloro-5-(chloromethyl)pyrimidine 2 (1.40 g, 7.16 mmol, 1.20 equiv) in acetone (7.46 ml) was added NaI (1.34 g, 8.95 mmol, 1.50 equiv), K2CO3 (1.34 g, 11.3 mmol, 1.90 equiv). The reaction mixture was stirred for 10 h at 50 °C. The resulting suspension was filtered and washed with CH2Cl2. The filtrate was concentrated in vacuo and purified by flash column chromatography (4:1 hexanes:EtOAc) on silica gel to give dichloropyrimidine 3 (2.10 g, 81% yield) as an orange liquid. Rf: 0.5 (4:1 hexanes:EtOAc); 1H NMR (400 MHz, CDCl3) δ 8.51 (s, 1H), 7.36 (dd, J = 7.7, 1.6 Hz, 1H), 7.13 (dd, J = 7.7, 0.8 Hz, 1H), 7.07 (d, J = 1.6 Hz, 1H), 4.55 (s, 2H), 2.25 (s, 3H), 1.55 (s, 9H); 13 C NMR (101 MHz, CDCl3) δ 165.97, 161.40, 159.49, 159.38, 144.22, 131.17, 130.41, 129.61, 127.51, 119.87, 110.29, 80.90, 42.65, 28.23, 17.72; IR (Neat) 3411, 2977, 2931, 1699, 1610, 1580, 1560, 1519, 1474, 1450, 1422, 1384, 1367, 1349, 1299, 1247, 1163, 1115, 1093, 1065, 1032, 993, 951, 916, 856, 819, 798, 760, 732, 703, 687, 648, 468, 439, 419 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C17H19Cl2N3O2Na [M + Na]+: 390.0754; found: 390.0745.

4.1.4. Tert-butyl 3-(7-chloro-1-methyl-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)-4-methylbenzoate (4)

To a solution of dichloropyrimidine 3 (1.50 g, 4.00 mmol, 1.00 equiv) in 1,4-dioxane (13.6 ml) was added MeNH2 (8.14 ml, 8.14 mmol, 1.50 equiv), i-Pr2NEt (2.12 ml, 12.2 mmol, 3.00 equiv). The reaction mixture was stirred for 1.5 h at 60 °C and then diluted with CH2Cl2 and water. The aqueous phase was extracted with CH2Cl2. The combined organic phases were washed with brine, dried over MgSO4 and concentrated in vacuo. The residue was purified by flash column chromatography (1:1 hexane:EtOAc) on silica gel to give tert-butyl 3-(((2-chloro-4-(methylamino)pyrimidin-5-yl)methyl)amino)-4-methylbenzoate (0.920 g, 62% yield) as a pale-yellow liquid. Rf: 0.18 (4:1 hexanes:EtOAc); 1H NMR (400 MHz, CDCl3) δ 7.94 (s, 1H), 7.45 (dd, J = 7.7, 1.6 Hz, 1H), 7.39 (d, J = 1.6 Hz, 1H), 7.15 (d, J = 7.8 Hz, 1H), 4.20 (s, 2H), 3.04 (d, J = 4.9 Hz, 3H), 2.19 (s, 3H), 1.59 (s, 9H); 13C NMR (101 MHz, CDCl3) δ 166.08, 163.32, 160.61, 154.74, 145.11, 131.32, 130.36, 128.86, 120.96, 112.60, 112.05, 81.11, 43.76, 28.36, 28.11, 17.92; IR (Neat) 3361, 2955, 2923, 2853, 1705, 1597, 1580, 1514, 1455, 1423, 1397, 1367, 1342, 1300, 1270, 1247, 1166, 1115, 1069, 1031, 992, 934, 873, 851, 779, 761, 737, 700, 517, 463 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C18H24ClN4O2 [M + H]+: 363.1588; found: 363.1585.

To a solution of tert-butyl 3-(((2-chloro-4-(methylamino)pyrimidin-5-yl)methyl)amino)-4-methylbenzoate (0.500 g, 1.40 mmol, 1.00 equiv) in THF (4.60 ml) was added triphosgene (0.200 g, 0.700 mmol, 0.500 equiv), Et3N (0.950 ml, 7.00 mmol, 5.00 equiv) at 0 °C under N2 atmosphere. The reaction mixture was stirred for 1 h at 70 °C and then diluted with CH2Cl2 and water. The aqueous phase was extracted with CH2Cl2. The combined organic phases were washed with brine, dried over MgSO4 and concentrated in vacuo and washed with isopropyl ether to give methyl urea 4 (0.920 g, 62% yield) as a pale-yellow solid. Rf: 0.66 (1:1 hexane:EtOAc); 1H NMR (400 MHz, CDCl3) δ 8.12 (s, 1H), 7.90 (dd, J = 7.9, 1.7 Hz, 1H), 7.84 (d, J = 1.7 Hz, 1H), 7.36 (d, J = 8.0 Hz, 1H), 4.83 (d, J = 14.8 Hz, 1H), 4.55 (d, J = 14.8 Hz, 1H), 3.47 (s, 3H), 2.28 (s, 3H), 1.58 (s, 9H); 13 C NMR (101 MHz, CDCl3) δ 164.89, 160.45, 158.98, 153.47, 151.88, 140.88, 140.32, 131.86, 131.48, 129.55, 128.06, 110.34, 81.54, 47.15, 29.05, 28.33, 18.01; IR (Neat) 2978, 1688, 1584, 1471, 1432, 1395, 1360, 1336, 1288, 1253, 1212, 1157, 1143, 1126, 1107, 1069, 1035, 976, 932, 874, 849, 825, 791, 764, 749, 734, 694, 637, 531, 475, 454, 418 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C19H22ClN4O3 [M + H]+: 389.1380; found: 389.1378.

4.1.5. 4-Methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoic acid (5)

To a solution of methyl urea 4 (1.70 g, 4.50 mmol, 1.00 equiv) in butan-2-ol (22.5 ml) was added 5-aminopicoline (0.500 g, 4.54 mmol, 1.01 equiv), K2CO3 (3.10 g, 22.5 mmol, 5.00 equiv), Xphos (0.400 g, 0.800 mmol, 0.200 equiv), Pd2(dba)3 (0.800 g, 0.800 mmol, 0.200 equiv). The reaction mixture was stirred for 2 h at 100 °C. Solids were removed via a filtration through a celite plug and the resulting solution was concentrated under reduced pressure. The filtrate was purified by flash column chromatography (1:20 to 1:10 MeOH:CH2Cl2) on silica gel to give tert-butyl 4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoate (1.65 g, 80% yield) as a white solid. Rf: 0.53 (1:1 hexane:THF); 1H NMR (400 MHz, CDCl3) δ 8.74 (d, J = 2.6 Hz, 1H), 8.01 (dd, J = 8.4, 2.7 Hz, 1H), 7.99 (s, 1H), 7.88 (dd, J = 7.9, 1.8 Hz, 1H), 7.85 (d, J = 1.7 Hz, 1H), 7.34 (d, J = 7.9 Hz, 1H), 7.29 (s, 1H), 7.16 (d, J = 8.4 Hz, 1H), 4.75 (dd, J = 13.8, 1.1 Hz, 1H), 4.52 − 4.41 (m, 1H), 3.45 (s, 3H), 2.56 (s, 3H), 2.28 (s, 3H), 1.58 (s, 9H); 13 C NMR (101 MHz, CDCl3) δ 164.89, 159.20, 157.76, 152.73, 152.61, 151.96, 140.87, 140.73, 140.03, 133.83, 131.51, 131.17, 129.10, 128.02, 127.45, 123.15, 103.08, 81.25, 47.38, 28.72, 28.19, 23.43, 17.90; IR (Neat) 3277, 2956, 2925, 2854, 1709, 1681, 1605, 1576, 1532, 1491, 1413, 1369, 1331, 1290, 1255, 1235, 1167, 1126, 1071, 1032, 951, 848, 786, 752, 682, 643, 520, 454 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C25H29N6O3 [M + H]+: 461.2301; found: 461.2301.

To a solution of tert-butyl 4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoate (0.460 g, 1.00 mmol, 1.00 equiv) in CH2Cl2 (10.0 ml) was added trifluoroacetic acid (1.50 ml, 20.0 mmol, 20.0 equiv). The reaction mixture was stirred for 12 h at 23 °C. The resulting solution was concentrated under reduced pressure and washed with ether to give benzoic acid 5 (0.400 g, 78% yield) as a white solid. Rf: 0.13 (1:10 MeOH:CH2Cl2); 1H NMR (400 MHz, MeOD) δ 9.49 (d, J = 2.5 Hz, 1H), 8.47 (dd, J = 8.9, 2.6 Hz, 1H), 8.22 (d, J = 1.1 Hz, 1H), 7.98 (d, J = 1.8 Hz, 1H), 7.94 (dd, J = 7.9, 1.8 Hz, 1H), 7.80 (d, J = 8.9 Hz, 1H), 7.45 (d, J = 8.0 Hz, 1H), 4.83 (d, J = 1.1 Hz, 1H), 4.61 (dd, J = 14.2, 0.9 Hz, 1H), 3.47 (s, 3H), 2.72 (s, 3H), 2.30 (s, 3H); 13C NMR (101 MHz, MeOD) δ 167.51, 158.20, 157.69, 153.21, 152.77, 145.38, 141.51, 140.91, 138.76, 134.47, 131.06, 130.09, 129.12, 129.05, 128.40, 127.49, 105.25, 46.79, 27.77, 17.36, 16.47.IR (Neat) 3041, 2923, 1674, 1602, 1566, 1503, 1467, 1421, 1340, 1286, 1265, 1234, 1182, 1128, 1107, 1068, 1024, 872, 840, 795, 767, 745, 720, 704, 683, 640, 618, 564, 517, 447 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C21H21N6O3 [M + H]+: 405.1675; found: 405.1674.

4.1.6. General Procedure a for synthesis of amides 7

To a solution of benzoic acid 5 (1.20 equiv) in DMF (0.100 M) was added amine 6 (1.00 equiv), i-Pr2NEt (5.00 equiv), HATU (2.00 equiv). The reaction mixture was stirred for 12 h at 23 °C and then diluted with CH2Cl2 and sat. NaHCO3. The aqueous phase was extracted with CH2Cl2. The combined organic phases were washed with brine, dried over MgSO4 and concentrated in vacuo. The residue was purified by flash column chromatography (1:10 MeOH:CH2Cl2) on silica gel to give amide 7 (10–20% yield) as a white solid.

4.1.7. General Procedure B for synthesis of amides 11

To a solution of aniline 8 (1.20 equiv) and carboxylic acid 9 or 10 (1.00 equv) in DMF (0.100 M) was added HATU (2.00 equiv) and i-Pr2NEt (5.00 equiv) at 23 °C. The reaction mixture was stirred for 16 h at 23 °C. The reaction was diluted with CH2Cl2 and quenched by addition of sat. NaHCO3. The phases were separated and the aqueous phase was extracted with EtOAc. The combined organic layer was washed with brine, dried over MgSO4, and concentrated in vacuo. The residue was purified by column chromatography (1:20 MeOH:CH2Cl2) on silica gel to afford amide 11 (10–20% yield).

Note: Spectra of compounds 7 were also acquired in methanol-d or acetone-d. The integrations of the major and minor peaks changed, providing evidence that species are indeed conformers/rotamers.

4.1.8. (S)-N-(1-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)amino)-2-methyl-1-oxopropan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-valyl)pyrrolidine-2-carboxamide (7a)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 99%; HPLC); Rf: 0.35 (1:10 = MeOH:CH2Cl2); [α]D28 = −102 (c 0.0590, MeOH);) 1H NMR (400 MHz, DMSO) δ 9.64 (s, 1H), 8.79 (d, J = 2.7 Hz, 1H), 8.42 (d, J = 8.0 Hz, 1H), 8.14 (s, 1H), 8.05 (dd, J = 8.5, 2.7 Hz, 1H), 8.00 (s, 1H), 7.93 (dd, J = 22.4, 1.8 Hz, 1H), 7.87 − 7.77 (m, 1H), 7.40 (d, J = 8.0 Hz, 1H), 7.27 (d, J = 8.5 Hz, 1H), 7.17 (d, J = 8.5 Hz, 1H), 4.86 − 4.66 (m, 2H), 4.56 − 4.44 (m, 2H), 4.28 (dd, J = 7.8, 4.8 Hz, 1H), 3.89 (d, J = 9.6 Hz, 1H), 3.63 (t, J = 8.2 Hz, 1H), 3.32 (s, 3H), 2.98 (s, 3H), 2.78 (s, 3H), 2.40 (s, 3H), 2.20 (s, 3H), 2.15 (d, J = 23.1 Hz, 1H), 2.07 − 1.91 (m, 2H), 1.86 (q, J = 5.6 Hz, 2H), 1.50 (dq, J = 11.4, 5.7, 5.1 Hz, 1H), 1.46 − 1.40 (m, 1H), 1.40 − 1.37 (m, 1H), 1.35 (s, 3H), 1.30 (s, 3H), 1.02 − 0.92 (m, 6H), 0.85 (dd, J = 11.2, 6.3 Hz, 6H); 13C NMR (101 MHz, DMSO) δ 173.25, 171.11, 170.96, 170.19, 170.14, 165.56, 165.39, 158.93, 156.93, 153.29, 153.22, 152.17, 152.12, 150.23, 141.10, 141.05, 140.04, 139.26, 134.68, 132.84, 132.74, 130.62, 126.84, 126.78, 126.56, 126.50, 126.24, 122.46, 102.91, 59.92, 59.88, 56.76, 55.99, 47.32, 46.69, 46.57, 40.91, 36.41, 35.13, 29.87, 28.88, 28.21, 25.41, 25.38, 24.70, 24.68, 24.59, 24.07, 23.23, 22.99, 22.97, 21.94, 21.92, 19.14, 19.06, 19.01, 17.28, 17.26; IR (Neat) 3300, 2959, 1632, 1607, 1530, 1496, 1411, 1332, 1240, 1174, 733 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C43H60N11O6 [M + H]+: 826.4728; found: 826.4734.

4.1.9. (R)-N-(1-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)amino)-2-methyl-1-oxopropan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-valyl)pyrrolidine-2-carboxamide (7b)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13 C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 96%; HPLC); Rf: 0.34 (1:10 = MeOH:CH2Cl2); [α]D30 = −176.0 (c 0.00063, MeOH); 1H NMR (400 MHz, DMSO) δ 9.64 (d, J = 7.0 Hz, 2H), 8.79 (t, J = 2.2 Hz, 2H), 8.71 − 8.46 (m, 2H), 8.18 − 8.11 (m, 2H), 8.05 (dd, J = 8.5, 2.7 Hz, 2H), 8.00 − 7.87 (m, 3H), 7.86 − 7.70 (m, 3H), 7.41 (dt, J = 21.0, 8.1 Hz, 4H), 7.18 (d, J = 8.5 Hz, 2H), 4.84 − 4.59 (m, 4H), 4.56 − 4.36 (m, 4H), 4.30 (d, J = 10.6 Hz, 2H), 3.85 − 3.52 (m, 5H), 3.05 − 2.79 (m, 6H), 2.66 (d, J = 23.7 Hz, 6H), 2.40 (s, 6H), 2.21 − 2.16 (m, 6H), 2.06 − 1.93 (m, 3H), 1.87 (s, 3H), 1.61 (t, J = 8.8 Hz, 3H), 1.39 (d, J = 34.3 Hz, 3H), 1.32 (d, J = 5.0 Hz, 2H), 1.28 (d, J = 5.6 Hz, 6H), 1.23 (s, 6H), 1.03 (d, J = 6.1 Hz, 7H), 0.95 (t, J = 6.7 Hz, 9H), 0.90 − 0.86 (m, 3H), 0.84 (dt, J = 7.7, 3.8 Hz, 12H).; 13C NMR (151 MHz, DMSO) δ 173.91, 173.79, 173.78, 173.76, 173.53, 173.50, 171.99, 171.95, 171.93, 171.82, 171.62, 171.60, 171.37, 170.53, 166.74, 159.71, 159.65, 159.29, 157.28, 157.24, 152.78, 152.49, 150.91, 150.56, 141.41, 140.38, 139.70, 135.05, 134.96, 133.53, 130.78, 126.58, 123.11, 122.83, 103.26, 60.39, 57.43, 56.37, 47.65, 47.62, 47.28, 47.13, 47.11, 47.08, 46.92, 46.79, 36.90, 35.53, 29.90, 28.90, 28.58, 26.68, 26.45, 26.23, 25.97, 25.34, 25.09, 24.79, 24.73, 24.32, 24.09, 23.75, 23.60, 23.58, 23.54, 23.52, 22.04, 21.85, 21.79, 21.73, 19.84, 19.53, 19.49, 19.27, 19.22, 17.71, 17.34; IR (Neat) 3306, 2922, 2852, 1720, 1670, 1631, 1600, 1572, 1531, 1404, 1333, 1294, 1259, 1152, 1025, 1006, 817, 756, 732, 699, 668 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C43H60N11O6 [M + H]+: 826.4728; found: 826.4738.

4.1.10. (S)-3-benzyl-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11a)

(Purity: 96%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D25 = +90.7 (c 0.150, MeOH); 1H NMR (400 MHz, DMSO) δ 10.83 (s, 1H), 10.50 (s, 1H), 9.64 (s, 1H), 8.80 (d, J = 2.6 Hz, 1H), 8.16 (d, J = 1.6 Hz, 1H), 8.05 (dd, J = 8.4, 2.6 Hz, 1H), 7.83 (dd, J = 4.0, 2.1 Hz, 1H), 7.76 (s, 1H), 7.69 (dd, J = 8.2, 1.7 Hz, 1H), 7.59 (dd, J = 8.4, 2.2 Hz, 1H), 7.51 (dt, J = 8.6, 4.2 Hz, 1H), 7.44 (d, J = 4.4 Hz, 4H), 7.40 − 7.24 (m, 6H), 7.18 (dd, J = 8.0, 5.8 Hz, 2H), 4.71 (d, J = 14.0 Hz, 1H), 4.52 (d, J = 14.0 Hz, 1H), 3.73 (dd, J = 8.1, 5.5 Hz, 1H), 3.42 (td, J = 18.2, 16.0, 6.9 Hz, 2H), 3.32 (s, 3H), 2.40 (s, 3H), 2.13 (s, 3H); 13C NMR (101 MHz, DMSO) δ 170.58, 167.78, 164.89, 159.42, 157.49, 153.73, 152.56, 150.69, 141.65, 140.51, 139.76, 139.67, 139.01, 138.21, 135.18, 131.35, 131.25, 131.20, 130.93, 130.19, 129.78, 129.14, 128.81, 128.55, 126.71, 126.51, 122.93, 121.84, 121.21, 120.08, 119.52, 119.49, 103.42, 65.58, 47.12, 37.61, 28.71, 23.71, 17.28; IR (Neat) 3357, 2923, 2853, 1667, 1461, 1376, 1256, 1079, 746, 698, 668 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C43H38N9O3 [M + H]+: 728.3098; found: 728.3102.

4.1.11. (S)-1-benzyl-3-isobutyl-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11b)

(Purity: 98%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D26 = −74.4 (c 0.0941, MeOH); 1H NMR (400 MHz, DMSO) δ 10.49 (s, 1H), 9.62 (s, 1H), 8.78 (d, J = 2.6 Hz, 1H), 8.20 (d, J = 1.6 Hz, 1H), 8.15 (s, 1H), 8.05 (dd, J = 8.4, 2.7 Hz, 1H), 7.78 (d, J = 2.8 Hz, 1H), 7.74 (dd, J = 8.2, 1.6 Hz, 1H), 7.62 − 7.57 (m, 1H), 7.53 − 7.48 (m, 1H), 7.42 (dd, J = 8.3, 6.7 Hz, 2H), 7.33 − 7.26 (m, 4H), 7.18 (d, J = 8.5 Hz, 1H), 7.13 − 7.06 (m, 3H), 6.97 − 6.93 (m, 2H), 5.57 (d, J = 15.6 Hz, 1H), 5.01 (d, J = 15.6 Hz, 1H), 4.69 (d, J = 14.1 Hz, 1H), 4.51 (d, J = 14.1 Hz, 1H), 3.74 − 3.64 (m, 2H), 3.33 (s, 3H), 2.40 (s, 3H), 2.13 (s, 3H), 1.93 − 1.83 (m, 2H), 0.94 (d, J = 6.2 Hz, 3H), 0.77 (d, J = 6.0 Hz, 3H); 13 C NMR (101 MHz, DMSO) δ 169.16, 167.47, 164.31, 159.06, 157.14, 153.40, 152.29, 150.40, 141.62, 141.29, 140.10, 138.01, 137.67, 137.14, 134.83, 132.43, 131.18, 130.95, 130.65, 129.73, 129.12, 128.53, 128.43, 127.28, 127.16, 126.52, 123.83, 122.69, 122.42, 119.98, 119.45, 103.08, 61.40, 49.11, 46.76, 28.36, 24.42, 23.41, 23.28, 22.08, 16.89; IR (Neat) 3285, 3031, 2952, 2867, 1661, 1597, 1576, 1530, 1507, 1491, 1466, 1408, 1319, 1293, 1231, 1185, 1143, 1119, 1078, 1029, 991, 908, 824, 784, 737, 696, 553, 518, 461, 411 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C47H46N9O3 [M + H]+: 784.3724; found: 784.3715.

4.1.12. (S)-3-methyl-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11c)

(Purity: 97%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D26 = +40.5 (c 0.124, MeOH); 1H NMR (400 MHz, DMSO) δ 10.76 (s, 1H), 10.53 (s, 1H), 9.63 (s, 1H), 8.79 (d, J = 2.6 Hz, 1H), 8.16 (d, J = 1.4 Hz, 1H), 8.05 (dd, J = 8.4, 2.7 Hz, 1H), 7.84 (t, J = 2.1 Hz, 1H), 7.78 (d, J = 1.7 Hz, 1H), 7.72 (dd, J = 8.2, 1.7 Hz, 1H), 7.62 (dd, J = 8.3, 2.2 Hz, 1H), 7.49 (dt, J = 7.0, 2.4 Hz, 3H), 7.48 − 7.38 (m, 4H), 7.31 (d, J = 8.5 Hz, 1H), 7.17 (d, J = 8.5 Hz, 1H), 4.71 (d, J = 14.0 Hz, 1H), 4.53 (d, J = 14.0 Hz, 1H), 3.70 (q, J = 6.3 Hz, 1H), 3.34 (s, 3H), 2.40 (s, 3H), 2.13 (s, 3H), 1.55 (d, J = 6.4 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 171.34, 167.14, 164.49, 158.97, 157.04, 153.28, 152.13, 150.25, 141.19, 140.04, 139.39, 138.62, 137.76, 137.65, 134.73, 130.90, 130.80, 130.66, 130.35, 129.31, 128.93, 128.33, 126.29, 122.50, 121.26, 120.73, 119.68, 119.11, 102.97, 58.67, 46.67, 28.26, 23.23, 17.25, 16.82; IR (Neat) 3268, 3055, 2930, 2854, 1381, 1600, 1576, 1534, 1508, 1446, 1412, 1321, 1294, 1234, 1216, 1187, 1145, 1120, 1031, 949, 844, 784, 735, 698, 657, 557, 457 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C37H33N9O3Na [M + Na]+: 674.2604; found: 674.2606.

4.1.13. (S)-1-benzyl-3-methyl-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11d)

(Purity: 97%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D30 = −3.70 (c 0.172, MeOH); 1H NMR (600 MHz, DMSO) δ 10.47 (s, 1H), 9.64 (s, 1H), 8.80 (d, J = 2.7 Hz, 1H), 8.21 (d, J = 1.7 Hz, 1H), 8.16 (s, 1H), 8.06 (dd, J = 8.4, 2.7 Hz, 1H), 7.81 (dd, J = 4.7, 2.2 Hz, 1H), 7.75 (dd, J = 8.1, 1.7 Hz, 1H), 7.61 (ddd, J = 7.3, 4.6, 2.1 Hz, 1H), 7.51 (t, J = 7.4 Hz, 1H), 7.42 (t, J = 7.6 Hz, 2H), 7.31 (dq, J = 6.7, 2.4, 1.6 Hz, 4H), 7.18 (d, J = 8.5 Hz, 1H), 7.15 − 7.08 (m, 3H), 7.00 − 6.95 (m, 2H), 5.58 (d, J = 15.6 Hz, 1H), 5.04 (d, J = 15.6 Hz, 1H), 4.70 (d, J = 14.0 Hz, 1H), 4.53 (d, J = 14.0 Hz, 1H), 3.86 (q, J = 6.2 Hz, 1H), 3.34 (s, 3H), 2.41 (s, 3H), 2.14 (s, 3H), 1.61 (d, J = 6.3 Hz, 3H); 13C NMR (151 MHz, DMSO) δ 169.71, 167.06, 164.08, 158.93, 157.02, 153.24, 152.11, 150.17, 141.48, 141.18, 139.94, 137.86, 137.59, 137.45, 137.08, 134.72, 132.49, 130.96, 130.78, 130.44, 129.61, 129.00, 128.38, 128.25, 127.10, 127.04, 126.29, 123.54, 122.49, 122.28, 119.79, 119.29, 102.95, 58.37, 53.60, 48.93, 46.63, 41.84, 28.23, 23.18, 18.08, 17.48, 16.79, 16.72; IR (Neat) 3301, 2918, 2850, 1725, 1670, 1598, 1532, 1507, 1494, 1446, 1411, 1377, 1290, 1263, 1187, 1144, 1120, 1028, 963, 895, 822, 805, 785, 733, 698, 660, 555, 504, 460 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C44H39N9O3Na [M + Na]+: 764.3074; found: 764.3049.

4.1.14. (R)-3-((R)-s-butyl)-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11e)

(Purity: 96%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D28 = −26.2 (c 0.0765, MeOH); 1H NMR (400 MHz, DMSO) δ 10.78 (s, 1H), 10.53 (s, 1H), 9.64 (s, 1H), 8.80 (d, J = 2.7 Hz, 1H), 8.16 (d, J = 1.7 Hz, 1H), 8.05 (dd, J = 8.5, 2.7 Hz, 1H), 7.85 (dd, J = 4.5, 2.2 Hz, 1H), 7.79 (d, J = 1.7 Hz, 1H), 7.72 (dd, J = 8.2, 1.7 Hz, 1H), 7.62 (dd, J = 8.3, 2.2 Hz, 1H), 7.54 − 7.41 (m, 6H), 7.31 (d, J = 8.4 Hz, 1H), 7.18 (d, J = 8.5 Hz, 1H), 4.72 (d, J = 13.9 Hz, 1H), 4.53 (d, J = 14.0 Hz, 1H), 3.34 (s, 3H), 3.13 (d, J = 9.5 Hz, 1H), 2.41 (s, 3H), 2.14 (s, 3H), 2.00 − 1.90 (m, 1H), 1.31 − 1.21 (m, 2H), 0.97 (d, J = 6.6 Hz, 3H), 0.91 (t, J = 7.4 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 169.69, 167.45, 164.96, 159.43, 157.49, 154.35, 153.76, 152.57, 150.69, 141.65, 140.52, 139.77, 139.17, 138.24, 135.18, 131.33, 131.25, 131.13, 130.86, 129.77, 129.12, 128.84, 126.71, 122.92, 121.91, 121.09, 119.53, 103.43, 47.12, 40.63, 40.43, 40.22, 40.01, 39.80, 39.59, 39.38, 35.04, 28.71, 24.97, 23.70, 17.28, 16.53, 11.24; IR (Neat) 3239, 2957, 2924, 2854, 1688, 1658, 1600, 1577, 1534, 1508, 1494, 1465, 1412, 1321, 1296, 1231, 1188, 1145, 1120, 1033, 842, 785, 738, 698, 658, 558, 411 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C40H39N9O3Na [M + Na]+: 716.3074; found: 716.3068.

4.1.15. (R)-N-(1-(((R)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)amino)-2-methyl-1-oxopropan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-D-valyl)pyrrolidine-2-carboxamide (7c)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13 C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 95%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D27 = +48.6 (c 0.0410, MeOH); 1H NMR (400 MHz, DMSO) δ 9.64 (s, 1H), 8.79 (d, J = 2.7 Hz, 1H), 8.42 (d, J = 8.0 Hz, 1H), 8.14 (s, 1H), 8.05 (dd, J = 8.5, 2.7 Hz, 1H), 8.00 (s, 1H), 7.93 (dd, J = 22.4, 1.8 Hz, 1H), 7.87 − 7.77 (m, 1H), 7.40 (d, J = 8.0 Hz, 1H), 7.27 (d, J = 8.5 Hz, 1H), 7.17 (d, J = 8.5 Hz, 1H), 4.86 − 4.66 (m, 2H), 4.56 − 4.44 (m, 2H), 4.28 (dd, J = 7.8, 4.8 Hz, 1H), 3.89 (d, J = 9.6 Hz, 1H), 3.63 (t, J = 8.2 Hz, 1H), 3.32 (s, 3H), 2.98 (s, 3H), 2.78 (s, 3H), 2.40 (s, 3H), 2.20 (s, 3H), 2.15 (d, J = 23.1 Hz, 1H), 2.07 − 1.91 (m, 2H), 1.86 (q, J = 5.6 Hz, 2H), 1.50 (dq, J = 11.4, 5.7, 5.1 Hz, 1H), 1.46 − 1.40 (m, 1H), 1.40 − 1.37 (m, 1H), 1.35 (s, 3H), 1.30 (s, 3H), 1.27 − 1.23 (m, 1H), 1.02 − 0.92 (m, 6H), 0.85 (dd, J = 11.2, 6.3 Hz, 6H); 13C NMR (101 MHz, DMSO) δ 173.25, 171.11, 170.96, 170.19, 170.14, 165.56, 165.39, 158.93, 156.93, 153.29, 153.22, 152.17, 152.12, 150.23, 141.10, 141.05, 140.04, 139.26, 134.68, 132.84, 132.74, 130.62, 126.84, 126.78, 126.56, 126.50, 126.24, 122.46, 102.91, 59.92, 59.88, 56.76, 55.99, 47.32, 46.69, 46.57, 40.91, 36.41, 35.13, 29.87, 28.88, 28.21, 25.41, 25.38, 24.70, 24.68, 24.59, 24.07, 23.23, 22.99, 22.97, 21.94, 21.92, 19.14, 19.06, 19.01, 17.28, 17.26; IR (Neat) 3300, 2959, 1632, 1607, 1530, 1496, 1411, 1332, 1240, 1174, 733 cm−1; IR (Neat) 3293, 2958, 1630, 1607, 1529, 1494, 1413, 1333, 1290, 1234, 1142, 1114, 1033, 845, 787, 735 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C43H60N11O6 [M + H]+: 826.4728; found: 826.4733.

4.1.16. (S)-N-(1-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)carbamoyl)cyclopropyl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-valyl)pyrrolidine-2-carboxamide (7d)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 98%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D27 = −18.9 (c 0.0520, MeOH); 1H NMR (400 MHz, DMSO) δ 9.64 (s, 1H), 8.79 (d, J = 2.6 Hz, 1H), 8.74 (d, J = 2.1 Hz, 1H), 8.42 − 8.36 (m, 1H), 8.14 (d, J = 2.9 Hz, 1H), 8.05 (dd, J = 8.4, 2.7 Hz, 1H), 7.93 (dd, J = 24.4, 1.8 Hz, 1H), 7.84 (ddd, J = 8.1, 3.9, 1.8 Hz, 1H), 7.41 (dd, J = 8.2, 3.5 Hz, 2H), 7.18 (d, J = 8.5 Hz, 1H), 4.84 − 4.71 (m, 2H), 4.58 − 4.47 (m, 2H), 4.13 (t, J = 7.1 Hz, 1H), 3.97 − 3.87 (m, 1H), 3.68 − 3.57 (m, 1H), 3.33 (s, 3H), 3.00 (s, 3H), 2.77 (d, J = 3.0 Hz, 3H), 2.40 (s, 3H), 2.20 (s, 3H), 2.07 − 1.95 (m, 2H), 1.89 − 1.71 (m, 2H), 1.57 − 1.45 (m, 2H), 1.43 − 1.34 (m, 1H), 1.33 − 1.12 (m, 3H), 0.96 (dd, J = 6.7 Hz, 6H), 0.85 − 0.80 (m, 8H).; 13C NMR (101 MHz, DMSO) δ 173.14, 171.52, 171.02, 170.59, 170.53, 166.05, 165.89, 159.41, 157.42, 153.76, 153.69, 152.62, 152.59, 150.71, 141.61, 141.54, 140.51, 139.78, 135.16, 133.32, 133.21, 131.11, 127.31, 127.06, 126.91, 126.72, 122.94, 103.40, 60.61, 60.56, 57.15, 57.09, 47.89, 47.53, 47.04, 41.32, 36.98, 35.64, 34.10, 30.30, 29.36, 28.68, 25.28, 24.56, 24.54, 23.70, 23.33, 23.29, 22.50, 22.46, 19.74, 19.69, 19.42, 19.29, 17.76, 16.86, 16.21; IR (Neat) 3296, 2957, 2871, 1627, 1606, 1575, 1525, 1492, 1410, 1332, 1290, 1233, 1195, 1140, 1112, 1071, 1034, 938, 831, 786, 731, 700, 621, 559, 516, 457 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C43H57N11O6Na [M + Na]+: 846.4391; found: 846.4385.

4.1.17. (R)-N-(1-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)carbamoyl)cyclopropyl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-valyl)pyrrolidine-2-carboxamide (7e)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 98%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D31 = −150.0 (c 0.0530, MeOH); 1H NMR (600 MHz, DMSO) δ 9.63 (d, J = 8.3 Hz, 1H), 8.79 (t, J = 2.8 Hz, 1H), 8.65 − 8.42 (m, 2H), 8.10 (d, J = 21.1 Hz, 1H), 8.05 (dd, J = 8.5, 2.8 Hz, 1H), 7.90 − 7.81 (m, 2H), 7.80 − 7.73 (m, 1H), 7.36 (dd, J = 15.5, 8.0 Hz, 1H), 7.18 (d, J = 8.5 Hz, 1H), 4.75 (td, J = 19.3, 17.8, 10.8 Hz, 2H), 4.63 (dt, J = 25.8, 9.0 Hz, 1H), 4.40 (dd, J = 87.9, 14.0 Hz, 1H), 4.23 − 4.17 (m, 1H), 3.68 (d, J = 6.9 Hz, 2H), 3.33 (s, 3H), 3.00 (d, J = 34.1 Hz, 3H), 2.60 (d, J = 42.9 Hz, 3H), 2.40 (s, 3H), 2.17 (d, J = 14.0 Hz, 4H), 2.11 − 2.03 (m, 1H), 1.97 (m, 1H), 1.86 (m, 1H), 1.79 (m, 1H), 1.72 (m, 1H), 1.65 (m, 1H), 1.39 (m, 1H), 1.25 − 1.05 (m, 3H), 0.94 (d, J = 7.4 Hz, 6H), 0.86 (m, 6H), 0.81 − 0.74 (m, 2H); 13C NMR (151 MHz, DMSO) δ 173.34, 173.14, 172.13, 172.08, 171.12, 170.99, 170.53, 170.49, 167.06, 166.18, 159.29, 159.27, 157.29, 157.26, 153.66, 152.47, 152.38, 150.59, 141.43, 141.32, 140.41, 139.60, 139.14, 135.06, 134.04, 133.30, 130.83, 130.65, 127.00, 126.61, 122.82, 103.37, 60.79, 60.69, 57.03, 56.85, 47.64, 47.32, 47.23, 46.89, 46.69, 40.96, 40.69, 36.93, 35.53, 35.34, 34.05, 30.21, 28.87, 28.77, 28.58, 24.98, 24.95, 24.32, 23.61, 23.59, 23.54, 23.49, 21.82, 19.64, 19.57, 19.00, 18.92, 17.72, 17.70, 16.27, 16.20, 16.00, 15.95; IR (Neat) 3294, 2957, 2923, 2853, 2360, 2340, 2628, 1574, 1533, 1496, 1414, 1333, 1294, 1236, 1149, 1115, 1026, 1007, 821, 753 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C43H58N11O6 [M + H]+: 824.4572; found: 824.4574.

4.1.18. (S)-N-(1-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)carbamoyl)cyclobutyl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-valyl)pyrrolidine-2-carboxamide (7f)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 96%; HPLC); Rf: 0.35 (1:10 = MeOH:CH2Cl2); [α]D31 = −56.7 (c 0.0530, MeOH); 1H NMR (400 MHz, DMSO) δ 9.68 (s, 1H), 8.81 (d, J = 2.7 Hz, 1H), 8.49 (s, 1H), 8.44 (d, J = 8.1 Hz, 1H), 8.15 (s, 1H), 8.07 (dd, J = 8.5, 2.7 Hz, 1H), 7.94 (dd, J = 23.1, 1.8 Hz, 1H), 7.88 − 7.78 (m, 1H), 7.40 (d, J = 8.1 Hz, 1H), 7.19 (dd, J = 8.7, 5.2 Hz, 2H), 4.84 − 4.71 (m, 2H), 4.56 − 4.48 (m, 2H), 4.26 (dd, J = 7.9, 5.3 Hz, 1H), 3.89 (ddt, J = 13.0, 8.8, 4.2 Hz, 1H), 3.64 (dq, J = 10.9, 5.4, 3.9 Hz, 1H), 3.33 (d, J = 2.4 Hz, 3H), 2.99 (s, 3H), 2.78 (d, J = 3.2 Hz, 3H), 2.41 (s, 3H), 2.40 − 2.33 (m, 1H), 2.20 (s, 3H), 2.17 (dd, J = 8.1, 5.4 Hz, 1H), 2.12 − 1.92 (m, 4H), 1.84 (ddt, J = 20.9, 14.2, 7.0 Hz, 4H), 1.53 − 1.40 (m, 2H), 1.38 − 1.31 (m, 1H), 1.26 (t, J = 6.1 Hz, 1H), 0.99 − 0.93 (m, 6H), 0.86 (dd, J = 16.6, 6.2 Hz, 6H); 13C NMR (101 MHz, DMSO) δ 172.72, 171.72, 171.59, 170.64, 170.59, 166.04, 165.87, 159.37, 157.42, 153.75, 153.69, 152.63, 152.58, 150.52, 141.58, 141.53, 140.14, 139.74, 135.27, 133.32, 133.22, 131.09, 127.32, 127.26, 127.04, 126.99, 123.10, 103.45, 60.26, 60.22, 58.87, 57.20, 54.08, 47.81, 47.04, 41.63, 36.89, 35.59, 31.13, 30.79, 30.33, 29.44, 28.68, 25.11, 25.09, 24.52, 23.52, 23.48, 23.46, 22.35, 22.33, 19.67, 19.65, 19.45, 19.40, 18.55, 17.74, 17.20, 15.82; IR (Neat) 3288, 2955, 2360, 2339, 1606, 1527, 1493, 1411, 1331, 1288, 1233, 1196, 1140, 1113, 1033, 953, 843, 787, 732 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C44H60N11O6 [M + H]+: 838.4728; found: 838.4726.

4.1.19. (S)-N-(2-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)amino)-2-oxoethyl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-valyl)pyrrolidine-2-carboxamide (7g)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 95%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D30 = −12.1 (c 0.0820, MeOH); 1H NMR (600 MHz, DMSO) δ 9.75 (s, 1H), 8.85 (s, 1H), 8.45 (dd, J = 8.2, 3.5 Hz, 1H), 8.19 (t, J = 5.1 Hz, 1H), 8.16 (s, 1H), 8.13 − 8.07 (m, 1H), 7.93 (d, J = 28.2 Hz, 1H), 7.86 (d, J = 8.5 Hz, 1H), 7.83 (t, J = 8.5 Hz, 1H), 7.40 (d, J = 7.9 Hz, 1H), 7.26 (d, J = 8.5 Hz, 1H), 4.81 − 4.75 (m, 2H), 4.52 (dt, J = 17.3, 6.5 Hz, 2H), 4.32 (dd, J = 8.0, 4.6 Hz, 1H), 3.90 (dt, J = 10.1, 5.8 Hz, 1H), 3.71 − 3.65 (m, 3H), 3.34 (d, J = 3.2 Hz, 3H), 3.02 (s, 3H), 2.80 (s, 3H), 2.43 (s, 3H), 2.20 (s, 3H), 2.17 (t, J = 7.3 Hz, 1H), 2.07 − 2.03 (m, 1H), 1.98 − 1.94 (m, 1H), 1.85 (t, J = 6.0 Hz, 1H), 1.55 − 1.51 (m, 1H), 1.41 (s, 1H), 1.27 − 1.25 (m, 2H), 0.99 − 0.93 (m, 6H), 0.86 (d, J = 6.6 Hz, 6H); 13C NMR (151 MHz, DMSO) δ 172.22, 171.61, 170.48, 170.43, 168.86, 168.62, 165.86, 165.71, 161.57, 159.16, 157.34, 153.60, 152.50, 152.44, 149.97, 141.44, 141.39, 139.62, 139.10, 135.44, 133.18, 133.08, 130.98, 127.56, 127.21, 126.89, 123.46, 103.52, 63.15, 61.03, 60.28, 59.96, 59.92, 57.12, 57.09, 53.93, 47.81, 47.61, 46.92, 46.86, 42.16, 41.07, 36.88, 35.53, 30.84, 30.21, 30.18, 29.65, 29.40, 28.60, 25.11, 24.88, 24.86, 24.48, 23.46, 23.44, 23.38, 23.37, 23.03, 22.04, 22.02, 22.00, 19.53, 19.49, 19.44, 19.40, 19.34, 19.10, 18.43, 17.82, 17.65, 17.63, 17.09, 12.83;); IR (Neat) 3291, 2958, 1739, 1630, 1574, 1533, 1496, 1415, 1336, 1291, 1232, 1142, 1115, 1033, 844, 787, 736 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C41H56N11O6 [M + H]+: 798.4415; found: 798.4420.

4.1.20. (R)-N-(2-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)amino)-2-oxoethyl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-valyl)pyrrolidine-2-carboxamide (7h)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 98%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D32 = −51.0 (c 0.0590, MeOH); 1H NMR (600 MHz, DMSO) δ 9.65 (s, 1H), 8.79 (s, 1H), 8.48 (dd, J = 16.2, 8.2 Hz, 1H), 8.14 (d, J = 7.4 Hz, 1H), 8.05 (dd, J = 8.5, 2.5 Hz, 2H), 7.92 − 7.88 (m, 1H), 7.86(dd, J = 8.5, 2.0 Hz, 1H), 7.82 − 7.80 (m, 1H), 7.39 − 7.37 (m, 1H), 7.18 (s, 1H), 4.78 − 4.75 (m, 2H), 4.57 − 4.55 (m, 1H), 4.52 − 4.48 (m, 1H), 4.45 (d, J = 14.0 Hz, 1H), 4.31 (dt, J = 8.2, 4.0 Hz, 1H), 3.85 − 3.77 (m, 2H), 3.67 − 3.64 (m, 2H), 3.33 (s, 3H), 3.00 (d, J = 11.9 Hz, 3H), 2.73 (d, J = 11.7 Hz, 3H), 2.40 (s, 3H), 2.20 (d, J = 3.5 Hz, 3H), 1.99 (t, J = 6.3 Hz, 2H), 1.88 (dt, J = 8.6, 5.1 Hz, 2H), 1.61 − 1.55 (m, 2H), 1.35 (ddd, J = 8.9, 4.4, 2.1 Hz, 1H), 0.95 (d, J = 6.7 Hz, 3H), 0.93 (d, J = 6.6 Hz, 3H), 0.85 (s, 6H); 13C NMR (151 MHz, DMSO) δ 172.21, 172.14, 171.89, 171.73, 170.52, 168.66, 168.61, 168.46, 166.39, 166.16, 159.29, 157.29, 153.63, 152.48, 152.41, 150.60, 150.58, 141.44, 141.40, 140.39, 139.67, 139.53, 135.06, 133.33, 133.12, 130.92, 130.86, 127.19, 126.96, 126.79, 126.59, 122.83, 103.32, 60.36, 57.17, 57.14, 47.50, 46.93, 46.88, 46.82, 42.32, 42.28, 41.20, 40.90, 40.79, 40.42, 36.88, 35.54, 35.52, 35.48, 31.06, 30.08, 30.06, 29.49, 29.43, 28.58, 24.55, 24.51, 24.42, 24.39, 23.60, 23.50, 21.95, 21.79, 19.54, 19.52, 19.12, 17.71, 17.68; IR (Neat) 3290, 2957, 2361, 2340, 1629, 1608, 1574, 1533, 1494, 1412, 1333, 1289, 1235, 1142, 1115, 1028, 840, 787, 736 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C41H56N11O6 [M + H]+: 798.4415; found: 798.4425.

4.1.21. (S)-N-((S)-1-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)amino)-1-oxo-3-phenylpropan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-valyl)pyrrolidine-2-carboxamide (7i)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 95%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D29 = −15.5 (c 0.130, MeOH); 1H NMR (400 MHz, DMSO) δ 9.64 (d, J = 4.8 Hz, 1H), 8.79 (d, J = 2.6 Hz, 1H), 8.51 (d, J = 8.1 Hz, 1H), 8.15 (d, J = 4.8 Hz, 1H), 8.09 − 7.97 (m, 2H), 7.97 − 7.87 (m, 2H), 7.81 (td, J = 7.7, 1.9 Hz, 1H), 7.46 − 7.33 (m, 1H), 7.27 − 7.09 (m, 6H), 4.76 (ddd, J = 17.7, 14.0, 9.9 Hz, 2H), 4.56 − 4.31 (m, 4H), 3.85 (dq, J = 11.9, 6.9, 5.9 Hz, 1H), 3.62 (dt, J = 11.8, 6.1 Hz, 1H), 3.32 (s, 3H), 3.02 − 2.95 (m, 1H), 2.93 (s, 3H), 2.91 − 2.82 (m, 1H), 2.80 (s, 3H), 2.40 (s, 3H), 2.20 (s, 3H), 2.15 − 2.08 (m, 1H), 1.97 (dt, J = 11.1, 8.0 Hz, 1H), 1.83 (tt, J = 13.3, 6.6 Hz, 2H), 1.54 (dq, J = 12.8, 6.6 Hz, 1H), 1.39 (tdd, J = 13.7, 9.0, 5.0 Hz, 2H), 1.18 − 1.09 (m, 1H), 1.03 − 0.90 (m, 6H), 0.91 − 0.72 (m, 6H); 13C NMR (101 MHz, DMSO) δ 171.76, 171.56, 170.66, 170.61, 165.96, 165.82, 159.41, 157.42, 153.73, 152.65, 152.57, 150.70, 141.56, 141.51, 140.52, 139.71, 138.01, 135.16, 133.26, 133.18, 131.07, 129.74, 129.63, 128.42, 127.32, 127.04, 126.97, 126.72, 126.65, 122.93, 103.39, 59.83, 57.32, 54.09, 47.66, 47.05, 46.95, 37.68, 36.91, 35.63, 30.33, 30.29, 29.59, 28.69, 24.76, 24.47, 23.71, 23.57, 22.15, 19.63, 19.59, 17.77, 17.74, 17.61; IR (Neat) 3388, 3300, 2955, 2924, 2854, 1728, 1672, 1460, 1415, 1377, 1260, 1018, 952, 799, 705, 667, 609, 554, 533, 498, 469, 447, 411 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C48H62N11O6 [M + H]+: 888.4885; found: 888.4888.

4.1.22. (S)-N-(1-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)amino)-2-methyl-1-oxopropan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-valyl)piperidine-2-carboxamide (7j)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 96%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D31 = −14.2 (c 0.0710, MeOH); 1H NMR (600 MHz, DMSO) δ 9.65 (d, J = 3.2 Hz, 2H), 8.79 (d, J = 2.7 Hz, 2H), 8.75 (dd, J = 10.0, 7.0 Hz, 1H), 8.47 (dd, J = 15.8, 8.6 Hz, 1H), 8.18 − 8.13 (m, 2H), 8.08 − 8.03 (m, 3H), 7.99 (dd, J = 12.1, 1.9 Hz, 1H), 7.94 (dd, J = 32.2, 1.9 Hz, 1H), 7.88 − 7.83 (m, 1H), 7.81 (ddd, J = 7.2, 4.8, 2.0 Hz, 1H), 7.65 (d, J = 4.2 Hz, 1H), 7.46 (t, J = 9.0 Hz, 1H), 7.40 (dq, J = 8.9, 4.9, 3.7 Hz, 3H), 7.18 (d, J = 8.5 Hz, 2H), 5.04 (q, J = 4.3, 3.8 Hz, 1H), 4.93 (d, J = 3.2 Hz, 1H), 4.87 − 4.76 (m, 4H), 4.73 (td, J = 9.6, 5.3 Hz, 2H), 4.70 − 4.62 (m, 1H), 4.56 − 4.42 (m, 3H), 4.07 (t, J = 13.9 Hz, 1H), 3.33 (d, J = 3.7 Hz, 6H), 2.99 − 2.96 (m, 6H), 2.81 − 2.76 (m, 6H), 2.70 (q, J = 11.7 Hz, 1H), 2.40 (s, 6H), 2.20 (d, J = 2.3 Hz, 6H), 2.18 − 2.15 (m, 1H), 2.10 (d, J = 12.2 Hz, 1H), 1.63 − 1.49 (m, 8H), 1.45 (p, J = 6.0, 5.5 Hz, 3H), 1.42 − 1.40 (m, 6H), 1.39 (d, J = 5.3 Hz, 1H), 1.37 (s, 3H), 1.34 (s, 3H), 1.31 − 1.17 (m, 4H), 0.98 (d, J = 6.8 Hz, 3H), 0.93 (d, J = 6.7 Hz, 3H), 0.90 (d, J = 6.6 Hz, 3H), 0.86 (d, J = 6.5 Hz, 3H), 0.83 (dd, J = 10.0, 6.4 Hz, 6H), 0.70 (t, J = 6.3 Hz, 3H);13C NMR (151 MHz, DMSO) δ 206.85, 173.70, 171.94, 171.89, 171.52, 171.44, 171.40, 171.32, 170.25, 169.31, 166.40, 166.22, 165.66, 165.49, 159.30, 159.29, 157.30, 153.59, 152.54, 152.47, 152.44, 152.39, 150.60, 150.59, 141.49, 141.47, 141.38, 140.39, 140.01, 139.63, 135.05, 133.26, 133.13, 132.45, 130.99, 127.06, 126.82, 126.61, 122.83, 103.30, 56.59, 56.54, 56.34, 56.15, 56.05, 55.26, 54.55, 52.46, 47.09, 47.06, 46.94, 43.56, 41.33, 36.77, 36.68, 35.49, 35.38, 31.06, 30.31, 30.24, 30.17, 28.58, 27.06, 26.91, 25.42, 25.31, 25.24, 25.11, 25.07, 25.04, 24.99, 24.35, 24.25, 23.59, 23.52, 22.08, 21.77, 21.73, 20.62, 20.33, 19.90, 19.87, 19.85, 19.82, 19.66, 18.94, 17.72, 17.67, 17.65, 17.63; IR (Neat) 3289, 2932, 1606, 1575, 1530, 1493, 1411, 1333, 1265, 1231, 1193, 1139, 1114, 1019, 952, 842, 787, 733 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C44H62N11O6 [M + H]+: 840.4885; found: 840.4897.

4.1.23. (S)-N-(1-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)amino)-2-methyl-1-oxopropan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-valyl)azetidine-2-carboxamide (7k)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 99%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D28 = −40.5 (c 0.0220, MeOH); 1H NMR (600 MHz, DMSO) δ 9.65 (s, 1H), 8.79 (d, J = 2.6 Hz, 1H), 8.54 (d, J = 8.0 Hz, 1H), 8.15 (d, J = 4.2 Hz, 1H), 8.10 − 8.03 (m, 2H), 7.97 − 7.89 (m, 1H), 7.82 (t, J = 8.5 Hz, 1H), 7.41 (d, J = 7.9 Hz, 1H), 7.34 (d, J = 8.5 Hz, 1H), 7.18 (d, J = 8.5 Hz, 1H), 4.78 (t, J = 14.8 Hz, 1H), 4.73 (td, J = 9.1, 4.8 Hz, 1H), 4.65 (dd, J = 9.0, 5.9 Hz, 1H), 4.52 (dd, J = 14.0, 5.7 Hz, 1H), 4.37 − 4.32 (m, 1H), 4.20 − 4.13 (m, 2H), 3.34 (d, J = 3.9 Hz, 3H), 2.99 (s, 3H), 2.80 (s, 3H), 2.41 (d, J = 8.5 Hz, 3H), 2.38 (d, J = 8.5 Hz, 1H), 2.20 (s, 3H), 2.17 − 2.07 (m, 2H), 1.53 (d, J = 13.5 Hz, 1H), 1.46 − 1.39 (m, 2H), 1.36 (s, 3H), 1.35 (s, 3H), 0.96 (dd, J = 15.1, 6.5 Hz, 6H), 0.86 (dd, J = 16.2, 6.5 Hz, 6H); 13C NMR (151 MHz, DMSO) δ 173.39, 171.78, 171.63, 169.45, 165.93, 165.78, 159.30, 157.31, 153.66, 152.55, 150.60, 141.46, 140.41, 139.66, 135.04, 133.02, 130.99, 127.21, 126.89, 126.61, 122.83, 103.29, 63.15, 60.81, 56.35, 54.98, 54.94, 49.19, 47.12, 46.94, 41.18, 36.78, 35.52, 29.56, 28.59, 25.40, 25.39, 25.13, 25.12, 24.39, 23.60, 23.51, 22.04, 19.61, 19.38, 17.67, 17.64; IR (Neat) 3293, 2926, 1635, 1607, 1531, 1495, 1467, 1412, 1333, 1290, 1241, 1142, 1071, 845, 787, 734 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C42H58N11O6 [M + H]+:812.4572; found: 812.4561.

4.1.24. (S)-N-(1-(((S)-1-(dimethylamino)-3-methyl-1-oxobutan-2-yl)amino)-2-methyl-1-oxopropan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-phenylalanyl)pyrrolidine-2-carboxamide (7l)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 99%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D29 = −46.5 (c 0.0667, MeOH); 1H NMR (400 MHz, DMSO) δ 9.64 (s, 1H), 8.79 (d, J = 2.6 Hz, 1H), 8.42 (d, J = 7.3 Hz, 1H), 8.13 (s, 1H), 8.10 (s, 1H), 8.05 (dd, J = 8.5, 2.7 Hz, 1H), 7.96 − 7.88 (m, 1H), 7.85 − 7.78 (m, 1H), 7.41 (dd, J = 10.6, 8.0 Hz, 2H), 7.23 (dd, J = 8.0, 6.4 Hz, 2H), 7.20 − 7.14 (m, 4H), 4.88 − 4.71 (m, 2H), 4.49 (dd, J = 14.0, 8.9 Hz, 2H), 4.28 (t, J = 6.4 Hz, 1H), 3.91 (m, 1H), 3.67 − 3.56 (m, 1H), 3.33 (s, 3H), 2.96 (dd, J = 13.4, 7.9 Hz, 1H), 2.80 (m, 1H), 2.72 (s, 3H), 2.70 (d, J = 2.1 Hz, 3H), 2.40 (s, 3H), 2.19 (s, 3H), 2.17 − 2.08 (m, 1H), 2.08 − 1.90 (m, 2H), 1.83 (d, J = 6.2 Hz, 2H), 1.28 (d, J = 17.4 Hz, 6H), 0.93 (dd, J = 11.5, 6.6 Hz, 6H); 13C NMR (101 MHz, DMSO) δ 173.58, 171.55, 170.73, 166.06, 159.41, 152.59, 150.72, 140.52, 139.71, 138.04, 135.15, 131.09, 129.75, 128.47, 126.77, 122.93, 103.40, 60.34, 57.27, 56.40, 50.48, 47.82, 47.03, 38.23, 36.77, 35.56, 30.27, 29.33, 28.67, 25.88, 25.11, 23.70, 19.53, 19.48, 17.74; IR (Neat) 3298, 2924, 1628, 1605, 1576, 1528, 1493, 1448, 1411, 1332, 1288, 1294, 1235, 1194, 1142, 1112, 1075, 1031, 943, 830, 787, 732, 700, 623, 548, 514, 480, 458 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C46H58N11O6 [M + H]+: 860.4572; found: 860.4589.

4.1.25. (S)-N-(1-(((S)-1-(dimethylamino)-3-methyl-1-oxobutan-2-yl)amino)-2-methyl-1-oxopropan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-alanyl)pyrrolidine-2-carboxamide (7m)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 96%; HPLC); Rf: 0.35 (1:10 = MeOH:CH2Cl2); [α]D30 = −42.0 (c 0.0667, MeOH); 1H NMR (400 MHz, DMSO) δ 9.62 (s, 1H), 8.79 (d, J = 2.6 Hz, 1H), 8.41 (d, J = 8.0 Hz, 1H), 8.15 (d, J = 4.6 Hz, 2H), 8.05 (dd, J = 8.5, 2.7 Hz, 1H), 7.95 − 7.88 (m, 1H), 7.85 − 7.78 (m, 1H), 7.40 (d, J = 7.9 Hz, 1H), 7.37 (d, J = 4.8 Hz, 1H), 7.24 (d, J = 7.6 Hz, 1H), 7.18 (d, J = 8.4 Hz, 1H), 4.78 (dd, J = 14.0, 6.5 Hz, 1H), 4.63 (t, J = 7.0 Hz, 1H), 4.54 − 4.48 (m, 1H), 4.46 (d, J = 7.6 Hz, 1H), 4.28 (dd, J = 7.6, 5.6 Hz, 1H), 3.96 (d, J = 9.9 Hz, 1H), 3.62 (d, J = 8.8 Hz, 1H), 3.51 (d, J = 24.5 Hz, 2H), 3.33 (s, 3H), 2.97 (d, J = 1.7 Hz, 3H), 2.78 (s, 3H), 2.41 (s, 3H), 2.20 (s, 3H), 2.17 − 2.10 (m, 1H), 2.06 − 1.91 (m, 2H), 1.82 (td, J = 11.9, 11.4, 6.3 Hz, 2H), 1.34 (s, 3H), 1.28 (s, 3H), 1.14 (d, J = 6.8 Hz, 3H), 0.95 (t, J = 6.5 Hz, 6H); 13C NMR (101 MHz, DMSO) δ 173.38, 171.98, 171.96, 171.61, 170.61, 166.06, 159.41, 157.43, 152.65, 150.72, 141.53, 140.52, 139.72, 135.15, 133.30, 133.04, 132.85, 131.09, 129.33, 129.03, 127.23, 127.00, 126.75, 122.93, 103.41, 60.30, 57.38, 56.33, 51.77, 47.87, 47.05, 45.10, 36.84, 35.65, 31.15, 30.20, 29.40, 28.68, 26.48, 25.15, 24.61, 23.69, 19.63, 19.42, 17.74; IR (Neat) 3294, 2962, 2965, 1735, 1632, 1609, 1575, 1531, 1495, 1414, 1333, 1291, 1236, 1196, 1143, 1114, 1034, 947, 832, 787, 740, 699, 625, 598, 513, 452, 418 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C40H54N11O6 [M + H]+:784.4259; found: 784.4248.

4.1.26. (S)-N-(1-(((S)-1-(dimethylamino)-3-methyl-1-oxobutan-2-yl)amino)-2-methyl-1-oxopropan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-valyl)pyrrolidine-2-carboxamide (7n)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 96%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D29 = −90.7 (c 0.0882, MeOH); 1H NMR (400 MHz, DMSO) δ 9.64 (s, 1H), 8.79 (d, J = 2.7 Hz, 1H), 8.48 − 8.41 (m, 1H), 8.15 (d, J = 1.7 Hz, 1H), 8.08 − 8.02 (m, 2H), 7.93 (dd, J = 20.3, 1.9 Hz, 1H), 7.82 (ddd, J = 8.0, 3.9, 1.8 Hz, 1H), 7.40 (d, J = 8.0 Hz, 1H), 7.18 (d, J = 8.5 Hz, 1H), 7.08 (dd, J = 9.0, 5.7 Hz, 1H), 4.79 (dd, J = 14.1, 8.7 Hz, 1H), 4.54 − 4.49 (m, 2H), 4.29 (td, J = 8.9, 8.2, 3.6 Hz, 1H), 3.90 (s, 1H), 3.65 (d, J = 7.5 Hz, 1H), 3.33 (s, 3H), 3.00 (t, J = 1.3 Hz, 3H), 2.80 (d, J = 0.9 Hz, 3H), 2.40 (s, 3H), 2.20 (s, 3H), 2.14 (d, J = 13.7 Hz, 1H), 1.97 (dt, J = 13.7, 7.2 Hz, 3H), 1.85 (d, J = 6.5 Hz, 3H), 1.36 (s, 3H), 1.29 (d, J = 2.2 Hz, 3H), 0.94 (dd, J = 6.7, 3.0 Hz, 6H), 0.81 (d, J = 6.6 Hz, 3H), 0.78 − 0.75 (m, 3H); 13C NMR (101 MHz, DMSO) δ 173.92, 171.63, 171.13, 170.61, 153.71, 150.72, 141.54, 140.52, 135.16, 131.09, 127.32, 127.00, 126.73, 122.94, 60.20, 57.32, 56.54, 53.68, 47.74, 47.05, 37.18, 35.46, 30.81, 30.30, 29.45, 28.68, 25.91, 25.27, 24.99, 23.70, 19.95, 19.59, 18.26, 17.75; IR (Neat) 3239, 2957, 2924, 2854, 1688, 1658, 1600, 1577, 1534, 1508, 1494, 1465, 1412, 1321, 1296, 1231, 1188, 1145, 1120, 1033, 842, 785, 738, 698, 659, 558, 464, 411 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C42H58N11O6 [M + H]+: 812.4572; found: 812.4582.

4.1.27. (R)-N-(1-(((S)-1-(dimethylamino)-3-methyl-1-oxobutan-2-yl)amino)-2-methyl-1-oxopropan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-valyl)pyrrolidine-2-carboxamide (7o)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 99%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D29 = +37.8 (c 0.0530, MeOH); 1H NMR (400 MHz, DMSO) δ 9.65 (s, 1H), 8.79 (d, J = 2.6 Hz, 1H), 8.48 − 8.41 (m, 1H), 8.15 (d, J = 1.7 Hz, 1H), 8.07 (s, 1H), 8.05 (dd, J = 8.4, 2.7 Hz, 1H), 7.93 (dd, J = 20.4, 1.8 Hz, 1H), 7.82 (ddd, J = 8.0, 4.0, 1.8 Hz, 1H), 7.40 (d, J = 8.0 Hz, 1H), 7.18 (d, J = 8.5 Hz, 1H), 7.09 (d, J = 9.0 Hz, 1H), 4.79 (dd, J = 14.1, 8.8 Hz, 1H), 4.50 (ddd, J = 19.3, 10.1, 5.6 Hz, 3H), 4.31 (dd, J = 7.9, 4.7 Hz, 1H), 3.94 − 3.83 (m, 1H), 3.68 − 3.55 (m, 1H), 3.33 (s, 3H), 3.00 (d, J = 1.4 Hz, 3H), 2.80 (s, 3H), 2.40 (s, 3H), 2.20 (s, 3H), 2.17 − 2.08 (m, 1H), 1.98 (m, 3H), 1.87 (dd, J = 11.4, 5.7 Hz, 2H), 1.36 (s, 3H), 1.29 (s, 3H), 0.94 (dd, J = 6.7, 2.9 Hz, 6H), 0.79 (dd, J = 16.8, 6.7 Hz, 6H); 13C NMR (101 MHz, DMSO) δ 173.91, 171.63, 171.12, 159.41, 157.41, 150.71, 140.52, 139.70, 135.15, 131.08, 127.25, 126.99, 126.73, 122.93, 103.38, 60.20, 57.31, 56.54, 53.68, 47.73, 47.04, 37.17, 35.45, 30.81, 30.30, 29.44, 28.68, 25.90, 25.27, 24.99, 23.70, 19.95, 19.59, 18.25, 17.74; IR (Neat) 3296, 2957, 2871, 1627, 1606, 1575, 1525, 1492, 1410, 1332, 1290, 1233, 1195, 1140, 1112, 1071, 1034, 938, 831, 786, 731, 700, 621, 559, 516, 457, 408 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C42H58N11O6 [M + H]+: 812.4572; found: 812.4584.

4.1.28. (S)-N-(1-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)amino)-2-methyl-1-oxopropan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-isoleucyl)pyrrolidine-2-carboxamide (7p)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 96%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D28 = −54.7 (c 0.0500, MeOH); 1H NMR (600 MHz, DMSO) δ 9.64 (s, 1H), 8.79 (d, J = 2.6 Hz, 1H), 8.46 (d, J = 8.1 Hz, 1H), 8.15 (s, 1H), 8.05 (dd, J = 8.4, 2.6 Hz, 1H), 8.00 (s, 1H), 7.92 (dd, J = 27.4, 1.9 Hz, 1H), 7.82 (ddd, J = 8.2, 4.5, 1.9 Hz, 1H), 7.40 (d, J = 8.1 Hz, 1H), 7.25 (d, J = 8.6 Hz, 1H), 7.18 (d, J = 8.5 Hz, 1H), 4.78 (t, J = 13.6 Hz, 1H), 4.72 (td, J = 8.6, 5.2 Hz, 1H), 4.60 − 4.48 (m, 2H), 4.28 (dd, J = 7.8, 5.3 Hz, 1H), 3.93 (d, J = 10.4 Hz, 1H), 3.62 (d, J = 8.4 Hz, 1H), 3.33 (s, 3H), 3.01 − 2.94 (m, 3H), 2.78 (s, 3H), 2.40 (s, 3H), 2.20 (s, 3H), 2.06 − 1.95 (m, 3H), 1.86 (d, J = 11.8 Hz, 2H), 1.61 − 1.40 (m, 4H), 1.35 (s, 3H), 1.30 (s, 3H), 1.20 − 1.13 (m, 1H), 0.94 (d, J = 6.8 Hz, 3H), 0.90 − 0.79 (m, 9H); 13C NMR (151 MHz, DMSO) δ 173.61, 171.48, 171.30, 170.67, 170.62, 165.80, 165.67, 159.30, 157.30, 153.60, 152.55, 152.47, 150.60, 141.45, 141.42, 140.40, 139.64, 139.62, 135.05, 133.13, 133.05, 130.98, 127.22, 126.91, 126.61, 122.83, 103.27, 60.26, 60.24, 56.33, 55.72, 47.72, 47.04, 46.93, 41.24, 36.78, 36.01, 35.98, 35.50, 29.27, 28.58, 25.84, 25.82, 25.07, 24.96, 24.93, 24.91, 24.46, 23.60, 23.36, 22.33, 17.66, 17.63, 15.29, 10.91; IR (Neat) 3300, 2959, 2360, 2340, 1629, 1609, 1531, 1496, 1414, 1332, 1290, 1240, 1143, 1115, 845, 737 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C44H62N11O6 [M + H]+: 840.4885; found: 840.4885.

4.1.29. (S)-N-(1-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)amino)-2-methyl-1-oxopropan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-leucyl)pyrrolidine-2-carboxamide (7q)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 95%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D26 = −34.0 (c 0.171, MeOH); 1H NMR (400 MHz, DMSO) δ 9.63 (s, 1H), 8.78 (d, J = 2.6 Hz, 1H), 8.38 (dd, J = 8.2, 2.3 Hz, 1H), 8.14 (s, 1H), 8.07 − 8.00 (m, 2H), 7.91 (dd, J = 27.5, 1.9 Hz, 1H), 7.84 (dd, J = 8.0, 1.9 Hz, 1H), 7.41 (dd, J = 8.7, 2.6 Hz, 2H), 7.18 (d, J = 8.5 Hz, 1H), 4.84 − 4.63 (m, 3H), 4.51 (dd, J = 14.2, 5.3 Hz, 1H), 4.26 (dd, J = 7.6, 4.9 Hz, 1H), 3.73 (q, J = 6.5 Hz, 1H), 3.55 (dd, J = 15.4, 6.4 Hz, 2H), 3.33 (d, J = 2.0 Hz, 3H), 2.95 (d, J = 2.1 Hz, 3H), 2.77 (s, 3H), 2.40 (s, 3H), 2.20 (s, 3H), 2.06 − 1.94 (m, 2H), 1.87 (td, J = 10.4, 5.7 Hz, 2H), 1.73 (t, J = 11.4 Hz, 2H), 1.51 (tt, J = 17.5, 9.3 Hz, 2H), 1.40 (d, J = 8.9 Hz, 1H), 1.36 (s, 3H), 1.30 (s, 3H), 0.96 − 0.89 (m, 6H), 0.85 (dd, J = 9.8, 6.5 Hz, 6H); 13C NMR (101 MHz, DMSO) δ 173.79, 171.50, 171.47, 171.26, 159.40, 157.40, 150.74, 140.49, 139.82, 135.14, 131.16, 126.79, 122.98, 103.38, 60.49, 56.50, 49.92, 47.30, 47.04, 41.39, 36.81, 35.63, 29.12, 28.68, 26.01, 25.18, 24.99, 24.85, 24.49, 23.76, 23.66, 23.49, 22.43, 21.68, 21.65, 17.74.IR (Neat) 3298, 2924, 1628, 1605, 1576, 1528, 1493, 1448, 1411, 1332, 1288, 1264, 1235, 1194, 1142, 1112, 1075, 1031, 943, 830, 787, 732, 700, 623, 548, 514, 480, 458 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C44H62N11O6 [M + H]+: 840.4806; found: 840.4885.

4.1.30. (S)-N-(1-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)amino)-2-methyl-1-oxopropan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)glycyl)pyrrolidine-2-carboxamide (7r)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 97%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D30 = −27.0 (c 0.100, MeOH); 1H NMR (600 MHz, DMSO) δ 9.64 (d, J = 2.4 Hz, 1H), 8.79 (d, J = 2.7 Hz, 1H), 8.17 − 8.09 (m, 2H), 8.05 (d, J = 8.6, 2.5 Hz, 1H), 7.96 (d, J = 22.1, 1.8 Hz, 1H), 7.86 (dd, J = 13.2, 5.9 Hz, 1H), 7.42 (d, J = 8.0 Hz, 1H), 7.33 (dd, J = 18.9, 8.8 Hz, 1H), 7.17 (d, J = 8.4 Hz, 1H), 4.80 − 4.71 (m, 1H), 4.68 (m, J = 8.9, 5.2 Hz, 1H), 4.52 (d, J = 14.0, 4.2 Hz, 1H), 4.29 (m, J = 5.8, 5.2 Hz, 1H), 3.97 (m, J = 21.2, 17.0, 4.6 Hz, 1H), 3.58 (m, J = 6.2 Hz, 1H), 3.32 (s, 3H), 3.00 − 2.91 (m, 3H), 2.79 (d, 3H), 2.40 (s, 3H), 2.21 (s, 3H), 2.11 − 1.95 (m, 2H), 1.93 − 1.74 (m, 2H), 1.50 (qd, J = 13.7, 6.5 Hz, 1H), 1.39 (m, 2H), 1.34 (s, 3H), 1.29 (s, 3H), 0.79 (m, 6H).13C NMR (151 MHz, DMSO) δ 173.59, 173.45, 171.68, 171.45, 171.44, 171.41, 171.20, 167.88, 167.51, 167.48, 165.76, 165.66, 159.30, 157.34, 157.31, 153.58, 152.50, 152.48, 152.46, 150.59, 141.58, 141.56, 141.53, 140.41, 140.39, 139.74, 139.72, 139.60, 139.58, 135.05, 133.30, 133.15, 133.12, 131.10, 131.07, 126.95, 126.89, 126.82, 126.70, 126.61, 126.59, 122.82, 103.29, 60.23, 59.40, 56.44, 56.40, 47.15, 46.99, 46.98, 46.94, 46.50, 42.40, 41.88, 41.32, 36.81, 36.65, 36.62, 35.49, 35.40, 35.37, 32.05, 29.02, 28.99, 28.59, 28.57, 28.55, 26.33, 26.27, 25.65, 24.88, 24.85, 24.84, 24.59, 24.42, 24.08, 24.05, 23.59, 23.49, 23.42, 23.36, 22.33, 22.20, 22.18, 22.07, 22.05, 17.64; IR (Neat) 3402, 2926, 1610, 1533, 1499, 1463, 1417, 1276, 1120, 846, 750 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C40H54N11O6 [M + H]+: 784.4259; found: 784.4257.

4.1.31. (S)-1-((S)-2-cyclohexyl-2-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzamido)acetyl)-N-(1-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)amino)-2-methyl-1-oxopropan-2-yl)pyrrolidine-2-carboxamide (7s)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 97%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D29 = −51.4 (c 0.0390, MeOH); 1H NMR (400 MHz, DMSO) δ 9.66 (s, 1H), 8.81 (d, J = 2.6 Hz, 1H), 8.38 (d, J = 8.3 Hz, 1H), 8.16 (s, 1H), 8.07 (dd, J = 8.5, 2.7 Hz, 1H), 7.94 (dd, J = 17.8, 1.8 Hz, 2H), 7.84 (d, J = 8.0 Hz, 1H), 7.45 − 7.37 (m, 2H), 7.19 (d, J = 8.5 Hz, 1H), 4.80 (dd, J = 14.0, 7.5 Hz, 1H), 4.77 − 4.65 (m, 1H), 4.63 − 4.48 (m, 2H), 4.29 (t, J = 6.2 Hz, 1H), 3.90 (s, 1H), 3.64 (s, 1H), 3.35 (s, 3H), 3.00 (s, 3H), 2.80 (s, 3H), 2.42 (s, 3H), 2.22 (s, 3H), 2.14 − 1.77 (m, 8H), 1.65 (d, J = 19.5 Hz, 2H), 1.57 (m, 1H), 1.50 − 1.32 (m, 8H), 1.21 (d, J = 32.8 Hz, 3H), 1.05 (d, J = 15.2 Hz, 2H), 0.92 − 0.86 (m, 6H); 13C NMR (101 MHz, DMSO) δ 177.65, 173.79, 171.55, 171.21, 170.71, 169.57, 159.41, 153.76, 150.71, 141.57, 140.51, 139.73, 138.69, 135.16, 131.10, 127.38, 126.97, 126.72, 122.94, 117.93, 103.38, 84.38, 56.45, 47.79, 47.25, 41.35, 36.87, 35.62, 29.13, 28.68, 25.51, 25.28, 25.06, 24.64, 23.70, 22.46, 17.76; IR (Neat) 3300, 2927, 2854, 2360, 2340, 1629, 1609, 1530, 1496, 1414, 1334, 1289, 1236, 1192, 1143, 1117, 845, 788, 737 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C46H64N11O6 [M + H]+: 866.5041; found: 866.5021.

4.1.32. (S)-N-(1-(((S)-1-(dimethylamino)-4-methyl-1-oxopentan-2-yl)amino)-2-methyl-1-oxopropan-2-yl)-1-((S)-2-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzamido)pentanoyl)pyrrolidine-2-carboxamide (7t)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 99%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D30 = +24.8 (c 0.0556, MeOH); 1H NMR (400 MHz, DMSO) δ 9.65 (s, 1H), 8.80 (d, J = 2.6 Hz, 1H), 8.41 − 8.34 (m, 1H), 8.15 (s, 1H), 8.08 − 7.99 (m, 2H), 7.98 − 7.89 (m, 1H), 7.87 − 7.82 (m, 1H), 7.41 (t, J = 7.2 Hz, 2H), 7.18 (d, J = 8.5 Hz, 1H), 4.83 − 4.66 (m, 3H), 4.53 (dd, J = 14.0, 5.1 Hz, 1H), 4.27 (t, J = 6.2 Hz, 1H), 3.75 (d, J = 7.8 Hz, 1H), 3.58 (s, 1H), 3.33 (s, 3H), 2.97 (d, J = 2.4 Hz, 3H), 2.79 (s, 3H), 2.41 (s, 3H), 2.21 (s, 3H), 1.99 (dd, J = 14.4, 7.9 Hz, 2H), 1.88 (s, 2H), 1.76 − 1.69 (m, 2H), 1.53 (dt, J = 15.1, 7.7 Hz, 2H), 1.42 (d, J = 18.8 Hz, 3H), 1.37 (s, 3H), 1.31 (s, 3H), 0.92 (d, J = 7.2 Hz, 3H), 0.85 (dd, J = 9.6, 6.4 Hz, 6H); 13C NMR (101 MHz, DMSO) δ 173.76, 171.48, 171.02, 165.80, 165.68, 159.41, 157.42, 152.61, 150.72, 141.62, 140.52, 139.79, 135.16, 131.15, 126.73, 122.94, 103.38, 60.50, 56.50, 51.45, 47.41, 47.26, 47.06, 41.39, 36.82, 35.59, 33.53, 29.15, 28.68, 26.04, 25.18, 25.04, 24.50, 23.71, 23.49, 22.45, 19.21, 17.76, 14.21; IR (Neat) 3288, 2955, 2918, 2849, 1727, 1633, 1607, 1577, 1532, 1497, 1462, 1411, 1378, 1294, 1118, 1099, 1019, 800, 739 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C43H59N11O6Na [M + Na]+: 848.4547; found: 848.4550.

4.1.33. (S)-N-(1-((2-(dimethylamino)-2-oxoethyl)amino)-2-methyl-1-oxopropan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-valyl)pyrrolidine-2-carboxamide (7u)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 96%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D27 = −59.5 (c 0.0240, MeOH); 1H NMR (600 MHz, DMSO) δ 9.65 (s, 1H), 8.79 (d, J = 2.6 Hz, 1H), 8.49 (dd, J = 8.2, 4.1 Hz, 1H), 8.26 (s, 1H), 8.15 (d, J = 3.7 Hz, 1H), 8.05 (dd, J = 8.4, 2.7 Hz, 1H), 7.93 (dd, J = 29.6, 1.9 Hz, 1H), 7.82 (ddd, J = 7.8, 5.8, 1.9 Hz, 1H), 7.47 (t, J = 5.3 Hz, 1H), 7.40 (dd, J = 8.2, 1.9 Hz, 1H), 7.18 (d, J = 8.5 Hz, 1H), 4.78 (dd, J = 16.0, 14.1 Hz, 1H), 4.53 − 4.45 (m, 2H), 4.30 (t, J = 6.8 Hz, 1H), 4.00 (dd, J = 16.6, 5.8 Hz, 1H), 3.96 (q, J = 7.7 Hz, 1H), 3.61 (ddd, J = 16.8, 12.0, 6.1 Hz, 2H), 3.33 (s, 3H), 2.93 (s, 3H), 2.80 (s, 3H), 2.40 (s, 3H), 2.20 (d, J = 1.5 Hz, 3H), 2.14 (dd, J = 13.5, 6.4 Hz, 1H), 2.08 − 1.97 (m, 2H), 1.86 (qd, J = 12.1, 11.4, 5.3 Hz, 2H), 1.34 (d, J = 12.4 Hz, 6H), 0.97 − 0.92 (m, 6H); 13C NMR (151 MHz, DMSO) δ 174.17, 171.68, 170.83, 170.79, 168.40, 165.92, 165.77, 159.30, 157.30, 153.60, 152.55, 152.48, 150.60, 141.45, 141.42, 140.40, 139.63, 135.04, 133.13, 133.04, 131.00, 127.21, 127.15, 126.91, 126.61, 122.83, 103.28, 63.15, 60.24, 60.21, 57.21, 56.39, 47.80, 46.93, 35.98, 35.38, 30.42, 30.37, 29.12, 28.58, 25.96, 25.94, 25.16, 25.13, 25.12, 23.60, 19.40, 19.38, 19.17, 17.66, 17.63; IR (Neat) 3289, 2927, 1605, 1575, 1528, 1493, 1411, 1332, 1289, 1234, 1195, 1142, 1113, 1033, 947, 842, 786, 731 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C39H52N11O6 [M + H]+: 770.4102; found: 770.4101.

4.1.34. (R)-N-((S)-3-methyl-1-oxo-1-(((R)-1-phenylethyl)amino)butan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-valyl)pyrrolidine-2-carboxamide (7v)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) (Purity: 95%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D29 = +4.25 (c 0.230, MeOH); 1H NMR (400 MHz, DMSO) δ 10.60 (s, 1H), 8.93 − 8.89 (m, 1H), 8.84 (s, 1H), 8.53 (dd, J = 7.3, 3.1 Hz, 1H), 8.20 (d, J = 8.0 Hz, 1H), 8.07 (dd, J = 8.4, 2.7 Hz, 1H), 7.94 − 7.87 (m, 1H), 7.83 (s, 1H), 7.45 (d, J = 8.0 Hz, 1H), 7.40 − 7.32 (m, 2H), 7.31 − 7.24 (m, 5H), 4.92 − 4.79 (m, 1H), 4.45 − 4.34 (m, 2H), 4.09 − 3.99 (m, 1H), 3.96 (s, 1H), 3.66 − 3.58 (m, 1H), 3.53 (d, J = 4.0 Hz, 3H), 3.46 − 3.37 (m, 1H), 3.67 − 3.56 (m, 1H), 2.45 (s, 3H), 2.13 (s, 3H), 2.08 − 1.98 (m, 2H), 1.97 − 1.75 (m, 5H), 1.27 (d, J = 7.0 Hz, 3H), 0.98 (d, J = 6.6 Hz, 6H), 0.76 (t, J = 6.6 Hz, 3H), 0.62 (dd, J = 6.9, 4.3 Hz, 3H); 13C NMR (101 MHz, DMSO) δ 172.56, 171.70, 170.91, 170.32, 170.23, 166.19, 165.84, 161.86, 160.08, 159.76, 159.68, 158.24, 152.92, 151.03, 150.97, 144.74, 144.63, 141.79, 140.38, 134.97, 133.63, 132.87, 132.75, 130.85, 130.71, 129.02, 128.58, 128.50, 128.40, 127.02, 126.94, 126.62, 126.46, 123.20, 101.11, 60.34, 59.75, 58.39, 57.91, 57.12, 48.21, 48.05, 47.67, 47.12, 32.92, 31.30, 30.37, 30.14, 29.87, 29.57, 29.42, 24.48, 23.86, 22.77, 22.73, 22.42, 19.79, 19.66, 19.58, 19.47, 19.41, 18.84, 18.49, 17.45; IR (Neat) 3294, 2956, 2922, 2853, 1725, 1664, 1595, 1571, 1529, 1494, 1462, 1398, 1377, 1333, 1300, 1267, 1236, 1207, 1174, 1115, 1070, 1030, 958, 916, 885, 829, 804, 756, 732, 699, 670, 629, 596, 560, 501, 460, 447, 426 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C44H55N10O5 [M + H]+: 803.4357; found: 803.4371.

4.1.35. (R)-N-((S)-3-methyl-1-oxo-1-(((R)-1-phenylethyl)amino)butan-2-yl)-1-((4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)benzoyl)-L-phenylalanyl)pyrrolidine-2-carboxamide (7w)

(Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum) (Purity: 99%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D27 = +104 (c 0.0560, MeOH); (Due to the distinct presence of rotameric isomers, the 1H NMR and 13C NMR contained extra peaks. See the attached spectrum in the supporting information) 1H NMR (400 MHz, DMSO) δ 9.66 (d, J = 3.8 Hz, 1H), 8.83 (t, J = 5.6 Hz, 1H), 8.80 − 8.75 (m, 1H), 8.21 − 8.10 (m, 2H), 8.05 (dt, J = 8.4, 2.2 Hz, 1H), 7.89 (dd, J = 3.7, 1.8 Hz, 1H), 7.80 − 7.73 (m, 1H), 7.49 − 7.32 (m, 2H), 7.32 − 7.26 (m, 5H), 7.25 − 7.09 (m, 5H), 6.98 − 6.89 (m, 1H), 4.91 − 4.83 (m, 1H), 4.80 (dd, J = 8.9, 6.9 Hz, 1H), 4.67 (d, J = 14.0 Hz, 1H), 4.55 − 4.50 (m, 1H), 4.34 − 4.21 (m, 1H), 3.69 (s, 1H), 3.17 (d, J = 5.2 Hz, 1H), 3.06 (d, J = 7.6 Hz, 1H), 3.02 − 2.84 (m, 1H), 2.41 (s, 3H), 2.19 (d, J = 14.1 Hz, 3H), 2.08 (s, 3H), 2.06 − 1.92 (m, 1H), 1.84 − 1.65 (m, 3H), 1.29 − 1.21 (m, 3H), 0.92 − 0.64 (m, 6H);13C NMR (101 MHz, DMSO) δ 171.65, 171.62, 170.78, 170.73, 170.30, 170.27, 170.14, 165.98, 165.82, 159.45, 157.42, 153.73, 152.53, 152.51, 150.73, 144.80, 144.76, 141.53, 140.54, 139.93, 139.79, 138.78, 137.81, 137.77, 135.15, 132.83, 132.78, 131.01, 129.77, 129.59, 128.66, 128.58, 128.34, 127.13, 126.99, 126.81, 126.78, 126.74, 126.42, 122.93, 103.29, 60.34, 58.51, 58.42, 53.96, 53.85, 53.62, 49.07, 48.46, 48.17, 47.16, 47.04, 31.15, 30.26, 29.78, 28.68, 23.70, 22.75, 22.70, 19.73, 19.60, 19.46, 18.54, 18.47, 18.14, 17.76); IR (Neat) 3291, 2966, 1640, 1607, 1574, 1531, 1496, 1451, 1414, 1332, 1292, 1235, 1193, 1145, 1118, 1029, 843, 739, 701 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C48H55N10O5 [M + H]+: 851.4357; found: 851.4377.

4.1.36. (R)-3-benzyl-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11f)

(Purity: 96%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D21 = −62.7 (c 0.0390, MeOH); 1H NMR (400 MHz, DMSO) δ 10.85 (s, 1H), 10.57 (s, 1H), 9.65 (s, 1H), 8.80 (d, J = 2.6 Hz, 1H), 8.16 (d, J = 1.7 Hz, 1H), 8.05 (dd, J = 8.4, 2.7 Hz, 1H), 7.85 (dd, J = 4.2, 2.2 Hz, 1H), 7.79 (d, J = 1.7 Hz, 1H), 7.72 (dd, J = 8.2, 1.7 Hz, 1H), 7.62 (dd, J = 8.3, 2.2 Hz, 1H), 7.53 − 7.48 (m, 1H), 7.46 − 7.42 (m, 4H), 7.37 − 7.33 (m, 3H), 7.28 (td, J = 8.1, 6.2 Hz, 3H), 7.18 (dd, J = 8.0, 5.3 Hz, 2H), 4.71 (d, J = 14.0 Hz, 1H), 4.52 (d, J = 14.1 Hz, 1H), 3.73 (dd, J = 8.1, 5.4 Hz, 1H), 3.47 − 3.35 (m, 2H), 2.40 (s, 3H), 2.13 (s, 3H); 13C NMR (101 MHz, DMSO) δ 170.54, 167.78, 159.42, 157.48, 152.55, 150.67, 141.62, 140.52, 139.76, 139.66, 139.01, 138.18, 135.18, 131.30, 131.14, 130.91, 130.18, 129.76, 129.12, 128.80, 128.54, 126.72, 126.49, 122.92, 121.27, 47.12, 40.63, 40.43, 40.22, 40.01, 39.80, 39.59, 39.38, 28.70, 23.70, 17.27; IR (Neat) 3298, 2925, 1671, 1601, 1535, 1413, 1321, 1119, 1032, 747, 698 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C43H37N9O3Na [M + Na]+: 750.2917; found: 750.2933.

4.1.37. (S)-3-(4-fluorobenzyl)-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11g)

(Purity: 95%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D30 = +0.706 (c 0.0940, MeOH);); 1H NMR (600 MHz, DMSO) δ 10.83 (s, 1H), 10.51 (s, 1H), 9.64 (s, 1H), 8.80 (s, 1H), 8.16 (s, 1H), 8.05 (d, J = 7.2 Hz, 1H), 7.84 (d, J = 5.8 Hz, 1H), 7.76 (s, 1H), 7.70 (d, J = 8.3 Hz, 1H), 7.60 (d, J = 8.3 Hz, 1H), 7.51 (m, 2H), 7.47 − 7.35 (m, 8H), 7.30 (d, J = 8.4 Hz, 1H), 7.18 (d, J = 8.4 Hz, 1H), 7.10 (t, J = 8.7 Hz, 3H), 4.71 (d, J = 14.0 Hz, 1H), 4.53 (d, J = 14.0 Hz, 1H), 3.73 (t, J = 6.8 Hz, 1H), 3.41 (dd, J = 13.2, 5.3 Hz, 2H), 2.40 (s, 3H), 2.13 (s, 3H); 13C NMR (151 MHz, DMSO) δ 170.10, 167.41, 164.40, 161.62, 160.02, 158.94, 157.01, 153.26, 152.09, 150.22, 141.17, 140.03, 139.28, 138.53, 137.73, 135.25 (d, J = 2.9 Hz), 134.71, 131.51 (d, J = 7.6 Hz), 130.87, 130.77, 130.75, 129.57 (d, J = 271.9 Hz), 129.30, 129.25, 128.35, 126.23, 122.47, 121.37, 120.76, 119.62, 119.06, 119.03, 114.72 (d, J = 20.9 Hz), 102.95, 65.00, 46.64, 38.25, 36.26, 28.24, 23.24, 16.81; 19F NMR (377 MHz, DMSO) δ −117.19; IR (Neat) 3324, 2925, 2854, 1674, 1607, 1511, 1463, 1408, 1261, 1100, 748, 669 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C43H37FN9O3 [M + H]+: 746.3003; found: 746.3005.

4.1.38. (S)-3-isobutyl-1-methyl-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11h)

(Purity: 99%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D26 = +55.8 (c 0.110, MeOH); 1H NMR (600 MHz, DMSO) δ 10.53 (s, 1H), 9.76 (s, 1H), 8.87 (t, J = 2.5 Hz, 1H), 8.18 (s, 1H), 8.12 (d, J = 8.5 Hz, 1H), 8.06 (s, 1H), 7.85 − 7.81 (m, 1H), 7.80 (d, J = 8.2 Hz, 1H), 7.65 − 7.61 (m, 1H), 7.55 (d, J = 7.6 Hz, 2H), 7.52 (d, J = 7.3 Hz, 1H), 7.46 (dd, J = 11.7, 7.8 Hz, 3H), 7.33 (d, J = 8.4 Hz, 1H), 7.27 (d, J = 8.5 Hz, 1H), 4.72 (d, J = 14.0 Hz, 1H), 4.55 (d, J = 14.0 Hz, 1H), 3.62 (ddq, J = 9.6, 6.8, 3.4, 2.8 Hz, 1H), 3.57 (dt, J = 8.9, 5.4 Hz, 1H), 3.35 (s, 3H), 3.14 (qd, J = 7.4, 4.1 Hz, 2H), 2.44 (s, 3H), 2.15 (s, 4H), 1.92 − 1.78 (m, 2H), 0.94 (d, J = 6.4 Hz, 3H), 0.76 (d, J = 6.3 Hz, 3H); 13C NMR (151 MHz, DMSO) δ 170.28, 167.36, 164.71, 159.15, 157.42, 153.61, 152.45, 149.85, 143.69, 141.53, 138.36, 138.03, 137.98, 135.53, 131.31, 131.17, 130.91, 130.80, 130.16, 129.57, 128.76, 123.56, 123.31, 121.38, 120.16, 119.61, 103.59, 61.52, 53.96, 46.99, 42.21, 35.10, 28.64, 24.53, 23.74, 22.93, 22.14, 18.45, 17.17, 17.09, 12.86; IR (Neat) 2954, 2360, 2340, 1659, 1599, 1533, 1492, 1411, 1323, 1266, 1188, 1143, 1030, 837, 784, 733, 698 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C41H42N9O3 [M + H]+: 708.3411; found: 708.3407.

4.1.39. (S)-1-allyl-3-isobutyl-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11i)

(Purity: 96%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D28 = +44.6 (c 0.180, MeOH); 1H NMR (600 MHz, DMSO) δ 10.51 (s, 1H), 9.65 (s, 1H), 8.89 − 8.64 (m, 1H), 8.14 (d, J = 31.3 Hz, 2H), 8.08 − 8.01 (m, 1H), 7.81 (m, 2H), 7.62 (d, J = 8.3 Hz, 1H), 7.57 − 7.39 (m, 6H), 7.32 (d, J = 8.3 Hz, 1H), 7.18 (d, J = 8.3 Hz, 1H), 5.76 (ddt, J = 16.1, 10.3, 5.0 Hz, 1H), 5.04 (d, J = 10.4 Hz, 1H), 4.98 (d, J = 17.3 Hz, 1H), 4.73 (dd, J = 18.1, 7.8 Hz, 2H), 4.60 − 4.50 (m, 2H), 3.64 (dd, J = 8.8, 4.5 Hz, 1H), 3.34 (s, 3H), 2.40 (s, 3H), 2.14 (s, 3H), 2.11 (m, 1H), 1.88 (dd, J = 12.9, 7.1 Hz, 2H), 0.94 (d, J = 5.7 Hz, 3H), 0.77 (d, J = 5.6 Hz, 3H); 13C NMR (151 MHz, DMSO) δ 169.22, 167.53, 164.63, 159.31, 157.38, 153.62, 152.48, 150.58, 142.44, 141.56, 140.39, 138.37, 138.02, 138.00, 135.06, 133.77, 131.85, 131.33, 131.16, 130.93, 130.15, 129.45, 128.84, 126.59, 123.70, 122.83, 122.08, 120.15, 120.13, 119.63, 119.60, 116.64, 103.31, 61.69, 49.16, 47.00, 40.42, 28.61, 24.63, 23.66, 23.60, 22.30, 17.17; IR (Neat) 3294, 2953, 2360, 2340, 1663, 1598, 1532, 1491, 1410, 1293, 1231, 1187, 1142, 1119, 1030, 989, 914, 841, 784, 739, 697 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C43H44N9O3 [M + H]+: 734.3567; found: 734.3565.

4.1.40. (S)-1,3-diisobutyl-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11j)

(Purity: 97%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D31 = +21.4 (c 0.0889, MeOH); 1H NMR (600 MHz, DMSO) δ 10.48 (s, 1H), 9.65 (s, 1H), 8.80 (d, J = 2.6 Hz, 1H), 8.17 (d, J = 3.0 Hz, 2H), 8.05 (dd, J = 8.6, 2.7 Hz, 1H), 7.86 − 7.77 (m, 2H), 7.64 (dt, J = 5.9, 2.9 Hz, 1H), 7.57 − 7.51 (m, 3H), 7.48 (t, J = 7.4 Hz, 2H), 7.44 (d, J = 8.1 Hz, 1H), 7.33 (d, J = 8.4 Hz, 1H), 7.18 (d, J = 8.5 Hz, 1H), 4.71 (d, J = 13.9 Hz, 1H), 4.54 (d, J = 14.0 Hz, 1H), 4.21 (dd, J = 13.8, 8.9 Hz, 1H), 3.66 (dd, J = 13.8, 5.9 Hz, 1H), 3.57 (dd, J = 8.9, 4.4 Hz, 1H), 3.34 (s, 3H), 2.41 (s, 3H), 2.15 (s, 3H), 2.14 − 2.09 (m, 1H), 1.83 (tt, J = 13.0, 7.4 Hz, 2H), 1.66 (p, J = 6.7 Hz, 1H), 0.93 (d, J = 6.1 Hz, 3H), 0.75 (d, J = 6.1 Hz, 3H), 0.71 (d, J = 6.6 Hz, 3H), 0.53 (d, J = 6.6 Hz, 3H); 13C NMR (151 MHz, DMSO) δ 170.12, 167.31, 164.54, 159.31, 157.39, 153.63, 152.48, 150.58, 142.48, 141.55, 140.39, 138.16, 137.97, 135.06, 132.44, 131.34, 131.15, 130.93, 130.09, 129.35, 128.88, 126.59, 123.91, 122.83, 122.41, 120.25, 119.77, 119.73, 103.31, 61.72, 52.75, 47.01, 31.32, 28.61, 27.11, 24.59, 23.71, 23.60, 22.43, 22.24, 20.23, 19.50, 17.18, 14.33; IR (Neat) 3239, 2957, 2924, 2854, 1688, 1658, 1600, 1577, 1534, 1508, 1494, 1465, 1412, 1321, 1296, 1231, 1188, 1145, 1120, 1033, 842, 785, 738, 698, 659, 558, 464, 411 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C44H48N9O3 [M + H]+: 750.3880; found: 750.3883.

4.1.41. (S)-3-benzyl-5-methyl-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11k)

(Purity: 99%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D33 = +60.3 (c 0.110, MeOH); 1H NMR (600 MHz, DMSO) δ 10.66 (s, 1H), 10.44 (s, 1H), 9.66 (s, 1H), 8.81 (d, J = 2.7 Hz, 1H), 8.15 (d, J = 3.0 Hz, 1H), 8.06 (dd, J = 8.5, 2.6 Hz, 1H), 7.88 − 7.79 (m, 2H), 7.75 − 7.71 (m, 1H), 7.64 (s, 1H), 7.59 (d, J = 8.2 Hz, 1H), 7.29 (d, J = 8.4 Hz, 1H), 7.27 − 7.16 (m, 5H), 7.14 (t, J = 7.3 Hz, 1H), 4.70 (d, J = 13.7 Hz, 1H), 4.55 − 4.45 (m, 1H), 3.55 (d, J = 14.0 Hz, 1H), 3.44 − 3.40 (m, 1H), 3.34 (s, 3H), 3.16 (dt, J = 15.0, 7.5 Hz, 1H), 2.41 (s, 6H), 2.13 (s, 3H); 13C NMR (151 MHz, DMSO) δ 169.93, 167.27, 164.76, 159.28, 157.37, 153.59, 152.45, 150.45, 141.52, 140.14, 139.58, 138.08, 137.61, 137.59, 135.14, 131.19, 131.12, 130.88, 129.90, 129.87, 129.14, 128.40, 126.75, 126.30, 122.93, 122.23, 120.87, 119.95, 119.42, 119.38, 103.34, 64.85, 46.99, 37.34, 28.61, 25.86, 23.49, 17.16; IR (Neat) 3278, 2923, 2853, 2360, 2340, 1669, 1600, 1534, 1496, 1411, 1296, 1236, 1189, 1145, 1113, 1072, 1029, 834, 786, 737, 700 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C38H36N9O3 [M + H]+: 666.2941; found: 666.2943.

4.1.42. (R)-3-benzyl-5-methyl-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11l)

(Purity: 98%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D25 = −73.5 (c 0.0890, MeOH); 1H NMR (600 MHz, DMSO) δ 10.66 (s, 1H), 10.44 (s, 1H), 9.66 (s, 1H), 8.81 (d, J = 2.7 Hz, 1H), 8.15 (d, J = 3.0 Hz, 1H), 8.06 (dd, J = 8.5, 2.6 Hz, 1H), 7.88 − 7.79 (m, 2H), 7.75 − 7.71 (m, 1H), 7.64 (s, 1H), 7.59 (d, J = 8.2 Hz, 1H), 7.29 (d, J = 8.4 Hz, 1H), 7.27 − 7.16 (m, 5H), 7.14 (t, J = 7.3 Hz, 1H), 4.70 (d, J = 13.7 Hz, 1H), 4.55 − 4.45 (m, 1H), 3.55 (d, J = 14.0 Hz, 1H), 3.44 − 3.40 (m, 1H), 3.34 (s, 3H), 3.16 (dt, J = 15.0, 7.5 Hz, 1H), 2.41 (s, 6H), 2.13 (s, 3H); 13C NMR (151 MHz, DMSO) δ 169.93, 167.27, 164.76, 159.28, 157.37, 153.59, 152.45, 150.45, 141.52, 140.14, 139.58, 138.08, 137.61, 137.59, 135.14, 131.19, 131.12, 130.88, 129.90, 129.87, 129.14, 128.40, 126.75, 126.30, 122.93, 122.23, 120.87, 119.95, 119.42, 119.38, 103.34, 64.85, 46.99, 37.34, 28.61, 25.86, 23.49, 17.16; IR (Neat) 3279, 2921, 1668, 1598, 1531, 1494, 1408, 1239, 1186, 1143, 1112, 1071, 1026, 826, 785, 732, 698 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C38H36N9O3 [M + H]+: 666.2941; found: 666.2947.

4.1.43. (S)-3-isobutyl-1,5-dimethyl-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11m)

(Purity: 98%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D30 = +36.0 (c 0.0500, MeOH); 1H NMR (600 MHz, DMSO) δ 10.45 (s, 1H), 9.64 (s, 1H), 8.80 (d, J = 2.7 Hz, 1H), 8.16 (s, 1H), 8.05 (dd, J = 8.5, 2.7 Hz, 1H), 7.95 (s, 1H), 7.88 (d, J = 8.1 Hz, 1H), 7.85 − 7.77 (m, 2H), 7.68 − 7.61 (m, 1H), 7.32 (d, J = 8.4 Hz, 1H), 7.17 (d, J = 8.4 Hz, 1H), 4.70 (d, J = 13.9 Hz, 1H), 4.53 (d, J = 14.0 Hz, 1H), 3.40 (m, 1H), 3.36 (s, 3H), 3.34 (s, 3H), 2.43 (s, 3H), 2.40 (s, 3H), 2.14 (s, 3H), 1.93 (ddd, J = 13.5, 8.4, 5.2 Hz, 1H), 1.72 (ddp, J = 33.0, 13.6, 6.6, 6.1 Hz, 2H), 0.85 (d, J = 6.4 Hz, 3H), 0.70 (d, J = 6.3 Hz, 3H); 13C NMR (151 MHz, DMSO) δ 170.07, 167.18, 164.65, 159.31, 157.39, 153.61, 152.48, 150.57, 141.77, 141.54, 140.39, 138.00, 137.50, 135.06, 132.63, 131.27, 131.14, 128.14, 126.59, 123.59, 122.82, 121.23, 121.20, 120.14, 119.66, 103.31, 60.66, 47.00, 35.06, 28.61, 25.44, 24.23, 23.60, 23.58, 22.11, 17.17; IR (Neat) 3304, 2954, 1666, 1602, 1535, 1508, 1413, 1319, 1187, 1144, 1120, 1023, 827, 786, 746 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C36H40N9O3 [M + H]+: 646.3254; found: 646.3265.

4.1.44. (S)-3-isobutyl-1-(2-methoxyethyl)-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11n)

(Purity: 98%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D23 = +20.3 (c 0.0720, MeOH); 1H NMR (600 MHz, DMSO) δ 10.51 (s, 1H), 9.65 (s, 1H), 8.80 (d, J = 2.7 Hz, 1H), 8.16 (d, J = 2.8 Hz, 2H), 8.05 (dd, J = 8.5, 2.7 Hz, 1H), 7.85 − 7.76 (m, 2H), 7.64 (dd, J = 8.4, 2.2 Hz, 1H), 7.55 − 7.50 (m, 3H), 7.49 − 7.44 (m, 2H), 7.39 (d, J = 8.1 Hz, 1H), 7.33 (d, J = 8.4 Hz, 1H), 7.18 (d, J = 8.5 Hz, 1H), 4.71 (d, J = 14.0 Hz, 1H), 4.54 (d, J = 14.0 Hz, 1H), 4.40 (dt, J = 14.5, 5.6 Hz, 1H), 3.99 (ddd, J = 14.4, 6.5, 4.2 Hz, 1H), 3.58 (dd, J = 8.7, 4.5 Hz, 1H), 3.42 − 3.35 (m, 2H), 3.34 (s, 3H), 2.95 (s, 3H), 2.41 (s, 3H), 2.14 (s, 3H), 2.11 (q, J = 8.8 Hz, 1H), 1.86 − 1.80 (m, 2H), 0.93 (d, J = 5.9 Hz, 3H), 0.76 (d, J = 5.9 Hz, 3H); 13C NMR (151 MHz, DMSO) δ 169.32, 167.58, 164.67, 159.31, 157.39, 153.62, 152.48, 150.58, 142.78, 141.56, 140.39, 138.48, 138.04, 138.03, 137.81, 135.06, 132.37, 131.29, 131.16, 131.14, 130.78, 129.81, 129.42, 128.69, 126.59, 123.91, 122.86, 122.83, 120.16, 120.14, 119.64, 119.60, 103.31, 69.70, 61.57, 58.27, 47.01, 28.61, 24.63, 23.67, 23.60, 22.29, 17.18; IR (Neat) 3292, 2953, 2360, 1662, 1598, 1533, 1492, 1411, 1293, 1238, 1187, 1144, 1119, 1022, 909, 825, 784, 737, 697 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C43H46N9O4 [M + H]+: 752.3673; found: 752.3667.

4.1.45. (S)-1-(2-(benzylamino)-2-oxoethyl)-3-isobutyl-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)-2-oxo-5-phenyl-2,3-dihydro-1H-benzo[e][1,4]diazepine-8-carboxamide (11o)

(Purity: 96%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D30 = +39.0 (c 0.0670, MeOH); 1H NMR (600 MHz, DMSO) δ 10.54 (s, 1H), 9.65 (s, 1H), 8.81 (d, J = 2.8 Hz, 1H), 8.67 (t, J = 6.0 Hz, 1H), 8.17 (s, 1H), 8.09 − 7.99 (m, 2H), 7.82 (q, J = 2.4 Hz, 1H), 7.80 − 7.77 (m, 1H), 7.63 (dd, J = 8.1, 2.4 Hz, 1H), 7.56 − 7.53 (m, 2H), 7.51 (t, J = 7.3 Hz, 1H), 7.45 (t, J = 7.5 Hz, 2H), 7.40 (d, J = 8.1 Hz, 1H), 7.34 (d, J = 8.5 Hz, 1H), 7.25 (dt, J = 15.7, 7.6 Hz, 4H), 7.18 (dd, J = 8.1, 4.6 Hz, 2H), 4.66 (dtd, J = 46.8, 16.2, 15.1, 3.8 Hz, 3H), 4.55 (d, J = 14.0 Hz, 1H), 4.34 (dd, J = 15.4, 6.0 Hz, 1H), 4.26 (dd, J = 15.4, 5.7 Hz, 1H), 3.66 (qd, J = 4.5, 1.9 Hz, 1H), 3.35 (s, 3H), 2.41 (s, 3H), 2.15 (s, 3H), 2.13 (d, J = 8.8 Hz, 1H), 1.90 − 1.82 (m, 2H), 0.95 (d, J = 6.1 Hz, 3H), 0.78 (d, J = 5.9 Hz, 3H); 13C NMR (151 MHz, DMSO) δ 169.56, 167.99, 167.90, 164.84, 159.32, 157.39, 153.62, 152.49, 150.55, 143.08, 141.58, 140.35, 139.43, 138.73, 138.07, 138.05, 137.94, 135.08, 131.62, 131.32, 131.19, 130.70, 130.04, 129.66, 128.61, 128.59, 127.44, 127.08, 126.63, 123.43, 122.84, 122.13, 122.12, 120.09, 119.60, 119.56, 103.30, 63.16, 61.40, 50.68, 47.02, 42.48, 40.22, 28.61, 24.61, 23.73, 23.57, 22.25, 17.18; IR (Neat) 3294, 2953, 2360, 1661, 1597, 1531, 1409, 1292, 1233, 1187, 1144, 1119, 1028, 908, 826, 784, 736, 697 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C49H49N10O4 [M + H]+: 841.3938; found: 841.3947.

4.1.46. (S)-2-(5-(but-3-en-1-yl)-1-methyl-2-oxo-2,3-dihydro-1H-benzo[e][1,4]diazepin-3-yl)-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)acetamide (11p)

(Purity: 97%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D30 = −17.0 (c 0.0780, MeOH); 1H NMR (600 MHz, DMSO) δ 10.18 (s, 1H), 9.69 (s, 1H), 8.83 (s, 1H), 8.13 (s, 1H), 8.08 (d, J = 8.5 Hz, 1H), 7.71 (d, J = 7.8 Hz, 1H), 7.67 (d, J = 14.5 Hz, 1H), 7.60 (t, J = 7.9 Hz, 1H), 7.50 (d, J = 8.4 Hz, 1H), 7.32 (t, J = 8.2 Hz, 2H), 7.22 (dd, J = 14.1, 8.4 Hz, 2H), 5.67 (dt, J = 16.6, 8.4 Hz, 1H), 4.84 (d, J = 13.6 Hz, 2H), 4.63 (d, J = 14.0 Hz, 1H), 4.47 (d, J = 14.1 Hz, 1H), 3.90 (d, J = 7.2 Hz, 1H), 3.31 (s, 3H), 3.25 (s, 3H), 3.12 (td, J = 15.8, 6.9 Hz, 1H), 2.97 (ddt, J = 30.1, 14.6, 7.4 Hz, 2H), 2.75 (dt, J = 15.4, 7.9 Hz, 1H), 2.42 (s, 3H), 2.21 (m, 1H), 2.14 (m, 1H), 2.08 (s, 3H); 13C NMR (151 MHz, DMSO) δ 170.61, 169.82, 169.67, 159.18, 157.39, 153.53, 152.39, 150.08, 142.17, 141.47, 138.45, 137.66, 131.59, 131.07, 130.00, 129.83, 127.62, 127.32, 124.88, 123.29, 122.25, 118.45, 118.04, 115.54, 103.48, 59.82, 46.92, 39.00, 36.78, 34.93, 31.22, 31.20, 28.58, 23.16, 18.45, 17.04; IR (Neat) 3305, 2924, 2361, 1666, 1601, 1535, 1493, 1413, 1293, 1235, 1194, 1144, 1117, 1025, 845, 750 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C36H38N9O3 [M + H]+: 644.3098; found: 644.3099.

4.1.47. (S)-2-(1-benzyl-5-(but-3-en-1-yl)-2-oxo-2,3-dihydro-1H-benzo[e][1,4]diazepin-3-yl)-N-(4-methyl-3-(1-methyl-7-((6-methylpyridin-3-yl)amino)-2-oxo-1,4-dihydropyrimido[4,5-d]pyrimidin-3(2H)-yl)phenyl)acetamide (11q)

(Purity: 98%; HPLC); Rf: 0.35 (1:10 MeOH:CH2Cl2); [α]D25 = +45.8 (c 0.0610, MeOH); 1H NMR (400 MHz, DMSO) δ 10.20 (s, 1H), 9.63 (s, 1H), 8.79 (d, J = 2.6 Hz, 1H), 8.13 (d, J = 1.7 Hz, 1H), 8.05 (dd, J = 8.4, 2.7 Hz, 1H), 7.72 − 7.62 (m, 2H), 7.62 − 7.48 (m, 2H), 7.34 (td, J = 8.3, 2.2 Hz, 1H), 7.27 (t, J = 7.4 Hz, 1H), 7.24 − 7.13 (m, 5H), 7.03 (d, J = 7.3 Hz, 2H), 5.79 − 5.66 (m, 1H), 5.33 (dd, J = 15.7, 3.8 Hz, 1H), 4.97 − 4.85 (m, 3H), 4.63 (d, J = 14.1 Hz, 1H), 4.48 (d, J = 14.1 Hz, 1H), 4.02 (t, J = 7.1 Hz, 1H), 3.34 (s, 3H), 3.22 (ddd, J = 16.8, 9.9, 7.4 Hz, 1H), 3.10 − 2.99 (m, 1H), 2.79 − 2.70 (m, 2H), 2.40 (s, 3H), 2.09 (s, 3H), 1.98 (q, J = 7.5 Hz, 2H); 13C NMR (101 MHz, DMSO) δ 170.79, 169.79, 169.35, 169.33, 159.41, 157.50, 153.66, 152.53, 150.67, 141.62, 140.53, 140.51, 138.54, 138.04, 137.62, 135.18, 131.66, 131.18, 131.14, 130.15, 128.79, 127.72, 127.61, 127.56, 126.69, 125.49, 122.98, 122.91, 118.63, 118.27, 115.55, 103.42, 59.96, 49.63, 47.06, 39.01, 36.96, 31.09, 31.07, 28.67, 23.70, 17.16; IR (Neat) 3300, 2923, 1667, 1599, 1534, 1491, 1449, 1409, 1330, 1290, 1236, 1186, 1143, 1116, 1079, 1024, 914, 826, 734, 699 cm−1; HRMS (MM: ESI-APCI+) m/z calc’d for C42H42N9O3 [M + H]+: 720.3411; found: 720.3410.

4.2. Computational methods

For molecular modelling studies, crystal structures of Lck (PDB: 2PL0), c-Src (PDB: 3OEZ), p38a (PDB: 3HEC) and Abl1 (PDB: 2HYY) were downloaded from the RCSB protein data bank homepage (https://www.rcsb.org). Only the crystal structures bound with Imatinib were selected to obtain the DFG-out state of kinases. The chicken c-Src crystal structure was converted into a human c-Src structure using a homology modelling method. The missing activation loop in p38a crystal structure was restored based on the crystal structure of Lck using Prime loop prediction applicationCitation17. For protein-ligand complex prediction, Glide docking applicationCitation18 in Schrödinger suite was employed. OPLS3e force field and SP mode of Glide were used. Flexible ligand sampling was allowed. Molecular dynamics simulation was performed using Desmond molecular dynamics packageCitation19 in Schrödinger suite. The kinase-inhibitor complexes were solvated using TIP3P water model and the solvated MD systems were described using OPLS3e force field. The Nose-Hoover chain thermostat and the Martyna-Tobias-Klein barostat methods were used to maintain the system temperature at 300 K and system pressure at 1 bar, respectively. A periodic boundary condition was employed. MD simulations were performed for 3 μs simulation time. Binding free energies of inhibitors were calculated by Prime molecular mechanics/generalized Born surface area (MM/GBSA) application with VSGB2.0 implicit solvation model. WaterMapCitation20 application in Schrödinger suites was used.

4.3. Biology

4.3.1. In vitro kinase assay

Full panel kinase profiling and kinase IC50 measurement were performed by using Reaction Biology Corp. (San Diego, USA).

4.3.2. Cell culture and sample treatment

Jurkat cells were purchased from the Korea Cell Line Bank (Seoul, Republic of Korea) and maintained in RPMI-1640 medium containing 10% FBS, streptomycin sulphate, penicillin, HEPES, and sodium bicarbonate in a 5% CO2 atmosphere at 37 °C. Jurkat cells were stimulated with 1 μg/ml of plate-bound anti-CD3 mAb (clone HIT3a, BD Bioscience) and then incubated with KITS 1–001 (10, 50, or 100 μM) for 30 min. KIST 1–001 were dissolved in DMSO and added to the culture media in serial dilution (the final concentration of DMSO in all experiments did not exceed 0.1%).

4.3.3. Western blot analysis

The protein of Jurkat cells was extracted using a PRO-PREP (Intron Biotechnology, Seoul, Republic of Korea). The protein concentration was determined using Bio-Rad protein assay reagent according to the manufacturer’s instruction and BSA (Bio-Rad, Hercules, CA, USA) was used as a standard for quantification. Equal protein amounts were separated by 10% sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to PVDF membranes. The membranes were incubated for 1 h with blocking solution (5% skim milk) at room temperature and followed by incubation for primary antibodies overnight at 4 °C. p-Lck (Y394) antibody (1:500, MBS128234, MyBioSource) was used as a primary antibody, and β-actin (1:1000, sc-81178, Santa Cruz Biotechnology) was used as an internal control. And then membranes were incubated with a 1:2000 dilution of horseradish peroxidase-conjugated secondary antibody for 2 h at room temperature. The membranes were analysed using an enhanced chemiluminescence (ECL) substrate and imaged by LAS-4000 luminescent image analyser (FUJIFILM, Tokyo, Japan).

4.3.4. Animals

Male C57BL/6 mice (21 ± 2 g; 6 weeks) were obtained from Oriental Bio Inc. (Seongnam-si, Korea). All mice were bred under constant conditions (temperature: 22 ± 2 °C, humidity: 40–60%, light/dark cycle: 12 h). All animal experiments were conducted under the university guidelines of the ethical committee for Animal Care and Use of the Kyung Hee University (KHSASP-22–002).

4.3.5. Induction of colitis by dextran sulphate sodium (DSS) and treatment

Colitis in mice was induced by providing water containing 4% (w/v) DSS for 7 days. Mice were randomly divided into 5 groups (n = 6/group, ) as follows: control group treated with vehicle; DSS plus vehicle group exposed to 4% DSS and treated with vehicle; the other 3 groups consist of mice receiving 4% DSS was treated with 5-ASA (75 mg/kg/day, p.o.) as a positive control or 7a (1, 5 mg/kg/day, i.p.) daily for 7 days.

Figure 8. Schematic diagram of DSS-induced colitis mouse model.

Figure 8. Schematic diagram of DSS-induced colitis mouse model.

4.3.6. Assessment of the disease activity index (DAI)

To calculate the severity of colitis, body weight, stool consistency, and occult/gross bleeding of all mice were assessed. DAI score was measured every day according to the following table (). The colon length was measured at end of the experiment.

4.3.7. Statistical analysis

Results are expressed as the mean ± SE of triplicate experiments with similar patterns. Statistically significant values were compared using ANOVA and Dunnett’s post hoc test, and p values of less than 0.05 were considered statistically significant.

Supplemental material

Supplemental Material

Download PDF (20 MB)

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

This work was supported by Korea Institute of Science and Technology (KIST), the National Research Foundation of Korea (NRF) funded by the Korean government (MEST) [2017R1A2B3006704, 2019R1A6A1A03032869, NRF2016M3A9B5940991, NRF-2021R1A2C3011992], the Brain Korea 21 FOUR Project for Medical Science in Yonsei University College of Medicine, and the Technology Development Program to Solve Climate Change of the National Research Foundation (NRF) of Korea funded by the Ministry of Science and ICT (NRF-2020M1A2A2079798). This research was supported by the National Research Council of Science & Technology (NST) granted by the Korea government (MSIT) (No. CPS21061-100).

References

  • For selected reviews, see: (a) Wu P, Nielsen TE, Clausen MH. Small-molecule kinase inhibitors: an analysis of FDA-approved drugs. Drug Discovery Today 2016;21:5–10. (b) Wu P, Nielsen TE, Clausen MH. FDA-approved small-molecule kinase inhibitors. Trends Pharmaco Sci 2015;36:422–39. (c) Zhang J, Yang PL, Gray NS. Targeting cancer with small molecule kinase inhibitors. Nat Rev Cancer 2009;9:28–39. (d) Ferguson FM, Gray NS. Kinase inhibitors: the road ahead. Nat Rev Drug Discov 2018;17:353–77. (e) Fedorov O, Müller S, Knapp S. The (un)targeted cancer kinome. Nat Chem Biol 2010;6:166–9. (f) Schwartz PA, Murray BW. Protein kinase biochemistry and drug discovery. Bioorg Chem 2011;39:192–210. (g) Cohen P. Protein kinases-the major drug targets of the twenty-first century? Nat Rev Drug Discov 2002;1:309–15. (h) Grant SK. Therapeutic protein kinase inhibitors. Cell Mol Life Sci 2009;66:1163–77. (i) Pottier C, Fresnais M, Gilon M, et al. Tyrosine kinase inhibitors in cancer: breakthrough and challenges of targeted therapy. Cancers 2020;12:731. (j) Gagic Z, Ruzic D, Djokovic N, et al. In silico methods for design of kinase inhibitors as anticancer drugs. Frontiers in Chemistry 2019;7:873.
  • (a) Zhao Z, Wu H, Wang L, et al. Exploration of type II binding mode: a privileged approach for kinase inhibitor focused drug discovery? ACS Chem Biol 2014;9:1230–41. (b) Roskoski R., Jr. Classification of small molecule protein kinase inhibitors based upon the structures of their drug-enzyme complexes. Pharmacol Res 2016;103:26–48.
  • (a) Sun J, Niu Y, Wang C, et al. Discovery of 3-benzyl-1,3-benzoxazine-2,4-dione analogues as allosteric mitogen-activated kinase kinase (MEK) inhibitors and anti-enterovirus 71 (EV71) agents. Bioorg Med Chem 2016;24:3472–82. (b) Comess KM, Sun C, Abad-Zapatero C, et al. Discovery and characterization of non-ATP site inhibitors of the mitogen activated protein (MAP) kinases. ACS Chem Biol 2011;6:234–44. (c) Wu P, Clausen MH, Nielsen TE. Allosteric small-molecule kinase inhibitors. Pharmacol Therapeut 2015;156:59–68. (d) Converso A, Hartingh T, Garbaccio RM, Tasber E, et al. Development of thioquinazolinones, allosteric Chk1 kinase inhibitors. Bioorg Med Chem Lett 2009;19:1240–4. (e) Lu X, Smaill JB, Ding K. New promise and opportunities for allosteric kinase inhibitors. Angew Chem Int Ed Engl 2020;59:13764–76.
  • (a) Zhao Z, Bourne PE. Progress with covalent small-molecule kinase inhibitors. Drug Discovery Today 2018;23:727–35. (b) Liu Q, Sabnis Y, Zhao Z, et al. Developing irreversible inhibitors of the protein kinase cysteinome. Chem Biol 2013;20:146–59. (c) Weisner J, Gontla R, van der Westhuizen L, et al. Covalent-allosteric kinase inhibitors. Angew Chem Int Ed Engl 2015;54:10313–6. (d) Abdeldayem A, Raouf YS, Constantinescu SN, et al. Advances in covalent kinase inhibitors. Chem Soc Rev 2020;49:2617–87.
  • (a) Norman RA, Toader D, Ferguson AD. Structural approaches to obtain kinase selectivity. Trends Pharmacol Sci 2012;33:273–8. (b) Davies SP, Reddy H, Caivano M, Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J 2000;351:95–105. (c) Müller S, Chaikuad A, Gray NS, Knapp S. The ins and outs of selective kinase inhibitor development. Nat Chem Biol 2015;11:818–21.
  • Liu Y, Han SJ, Liu WB, Stoltz BM. Catalytic enantioselective construction of quaternary stereocenters: assembly of key building blocks for the synthesis of biologically active molecules. Acc Chem Res 2015;48:740–51.
  • (a) Zask A, Murphy J, Ellestad GA. Biological stereoselectivity of atropisomeric natural products and drugs. Chirality 2013;25:265–74. (b) Porter J, Payne A, Whitcombe I, et al. Atropisomeric small molecule Bcl-2 ligands: Determination of bioactive conformation. Bioorg Med Chem Lett 2009;19:1767–72. (c) Xing L, Devadas B, Devraj RV, et al. Discovery and characterization of atropisomer PH-797804, a p38 MAP kinase inhibitor, as a clinical drug candidate. ChemMedChem 2012;7:273–80. (d) Smith DE, Marquez I, Lokensgard ME, et al. Exploiting atropisomerism to increase the target selectivity of kinase inhibitors. Angew Chem Int Ed Engl 2015;54:11754–9.
  • (a) Jiang J, Shen M, Thomas CJ, Boxer MB. Chiral kinase inhibitors. Curr Top Med Chem 2011;11:800–9. (b) Norman P. Evaluation of WO2011134971, chiral 1,6-napthyridine Syk kinase inhibitors. Expert Opin Ther Patents 2012;22:335–9. (c) Kitano Y, Suzuki T, Kawahara E, Yamazaki T. Synthesis and inhibitory activity of 4-alkynyl and 4-alkenylquinazolines: Identification of new scaffolds for potent EGFR tyrosine kinase inhibitors. Bioorg Med Chem Lett 2007;17:5863–7. (d) Bühler S, Goettert M, Schollmeyer D, et al. Chiral sulfoxides as metabolites of 2-thioimidazole-based p38α mitogen-activated protein kinase inhibitors: enantioselective synthesis and biological evaluation. J Med Chem 2011;54:3283–97. (e) Wu CH, Coumar MS, Chu CY, et al. Design and synthesis of tetrahydropyridothieno[2,3-d]pyrimidine scaffold based epidermal growth factor receptor (EGFR) kinase inhibitors: The role of side chain chirality and michael acceptor group for maximal potency. J Med Chem 2010;53:7316–26.
  • (a) Choi HG, Ren P, Adrian F, et al. A type-II kinase inhibitor capable of inhibiting the T315I “gatekeeper” mutant of Bcr-Abl. J Med Chem 2010;53:5439–48. (b) Nonami A, Sattler M, Weisberg E, et al. Identification of novel therapeutic targets in acute leukemias with NRAS mutations using a pharmacologic approach. Blood 2015;125:3133–43. (c) Cho H, Shin I, Ju E, et al. First SAR study for overriding NRAS mutant driven acute myeloid leukemia. J Med Chem 2018;61:8353–73.
  • (a) Whitby LR, Ando Y, Setola V, et al. Design, Synthesis, and validation of a β-turn mimetic library targeting protein-protein and peptide-receptor interactions. J Am Chem Soc 2011;133:10184–94. (b) Metrano AJ, Abascal NC, Mercado BQ, et al. Diversity of secondary structure in catalytic peptides with β-turn-biased sequences. J Am Chem Soc 2017;139:492–516. (c) Hata M, Marshall GR. Do benzodiazepines mimic reverse-turn structures? J Comput Aided Mol Des 2006;20:321–31. (d) Chauhan J, Chen SE, Fenstermacher KJ, et al. Synthetic, structural mimetics of the β-hairpin flap of HIV-1 protease inhibit enzyme function. Bioorg Med Chem 2015;23:7095–109. (e) Dörr AA, Lubell W. γTurn γ-Turn Mimicry with Benzodiazepinones and pyrrolobenzodiazepinones synthesized from a Common Amino ketone intermediate. Org Lett 2015;17:3592–5. (f) Peris G, Jakobsche CE, Miller SJ. Aspartate-catalyzed asymmetric epoxidation reactions. J Am Chem Soc 2007; 129:8710–1. (g) Lenci E, Trabocchi A. Peptidomimetic toolbox for drug discovery. Chem Soc Rev 2020;49:3262–77.
  • WaterMap finds the hydration sites in the binding site using MD simulation and calculates the enthalpy and entropy energies of each the hydration site.
  • (a) Spyrakis F, Ahmed MH, Bayden AS, et al. The roles of water in the protein matrix: a largely untapped resource for drug discovery. J Med Chem 2017;60:6781–827. (b) Robinson DD, Sherman W, Farid R. Understanding kinase selectivity through energetic analysis of binding site waters. ChemMedChem 2010;5:618–27. (c) Wang Y, Fu Q, Zhou Y, et al. Replacement of protein binding-site waters contributes to favorable halogen bond interactions. J Chem Inf Model 2019;59:3136–43. (d) Cappel D, Sherman W, Beuming T. Calculating water thermodynamics in the binding site of Proteins - Applications of WaterMap to Drug Discovery. Curr Top Med Chem 2017;17:2586–98. (e) Breiten B, Lockett MR, Sherman W, et al. Water networks contribute to enthalpy/entropy compensation in protein-ligand binding. J Am Chem Soc 2013;135:15579–84.
  • Ananthakrishnan AN, Kaplan GG, Ng SC. Changing global epidemiology of inflammatory bowel diseases: sustaining health care delivery into the 21st Century. Clin Gastroenterol Hepatol 2020;18:1252–60.
  • Giuffrida P, Sabatino AD. Targeting T cells in inflammatory bowel disease. Pharmacol Res 2020;159:105040.
  • Singh PK, Kashyap A, Silakari O. Exploration of the therapeutic aspects of Lck: a kinase target in inflammatory mediated pathological conditions. Biomed Pharmacother 2018;108:1565–71.
  • Rose WA, Sakamoto K, Leifer CA. Multifunctional role of dextran sulfate sodium for in vivo modeling of intestinal diseases. BMC Immunology 2012;13:41.
  • Schrödinger Release 2019-4: Prime. Schrödinger. New York, NY: LLC; 2019.
  • Schrödinger Release 2019-4: Glide. Schrödinger. New York, NY: LLC; 2019.
  • Schrödinger Release 2019-4: Desmond, Desmond Molecular Dynamics System, D. E. Shaw Research, New York, NY, 2020. Maestro-Desmond Interoperability Tools, Schrödinger, LLC, New York, NY, 2019.
  • Schrödinger Release 2019-4: WaterMap. Schrödinger. New York, NY: LLC; 2019.