1,447
Views
0
CrossRef citations to date
0
Altmetric
Research Article

Design and synthesis of doublecortin-like kinase 1 inhibitors and their bioactivity evaluation

, , &
Article: 2287990 | Received 11 Sep 2023, Accepted 21 Nov 2023, Published online: 07 Dec 2023

Abstract

Doublecortin-like kinase 1 (DCLK) is a microtubule-associated serine/threonine kinase that is upregulated in a wide range of cancers and is believed to be related to tumour growth and development. Upregulated DCLK1 has been used to identify patients at high risk of cancer progression and tumours with chemotherapy-resistance. Moreover, DCLK1 has been identified as a cancer stem cell (CSC) biomarker in various cancers, which has received considerable attention recently. Herein, a series of DCLK1 inhibitors were prepared based on the previously reported XMD8-92 structure. Among all the synthesised compounds, D1, D2, D6, D7, D8, D12, D14, and D15 showed higher DCLK1 inhibitory activities (IC50 40–74 nM) than XMD8-92 (IC50 161 nM). Compounds D1 and D2 were selective DCLK1 inhibitors as they showed a rather weak inhibitory effect on LRRK2. The antiproliferative activities of these compounds were also preliminarily evaluated. The structure-activity relationship revealed by our compounds provides useful guidance for the further development of DCLK1 inhibitors.

Introduction

Doublecortin-like kinase 1 (DCLK1) was first identified in the brain as a microtubule-associated protein (MAP) and it was found to play a crucial role in early neurogenesisCitation1. Accumulating studies indicated that DCLK1 is related to tumour development. DCLK1 isoforms are overexpressed in many types of cancer cells, especially in colorectal cancer, which worsens the survival outcomes of colorectal cancer (CRC) patientsCitation2,Citation3. Epithelial-mesenchymal transition (EMT) plays a key role in cancer invasion and metastasis, especially in pancreatic cancer. It was shown that small-interfering RNA (siRNA)-mediated knockdown of DCLK-1 in human pancreatic cancer cells induced the downregulation of EMT-associated transcription factorsCitation4. Moreover, pharmacological inhibition of DCLK1 prevented the development of DCLK1+ pancreatic ductal adenocarcinoma (PDAC) in clinically relevant patient-derived PDAC organoid modelsCitation5.

Additionally, DCLK1 is recognised as a regulator of type II immune response and linked with functional regulation of the tumour microenvironment. DCLK1 is strongly linked to the infiltration of multiple immune cell types, especially tumour-associated macrophages (TAMs) and regulatory T-cells (Treg). And DCLK1 might contribute to TAM-mediated inhibition of CD8+ T-cells to enhance tumour growth in the tumour microenvironmentCitation6. In addition, it was reported that the overexpression of miR-539 enhanced the sensitivity of cisplatin (DDP)-resistant NSCLC cells to DDP by directly targeting DCLK1Citation7. Considering DCLK1 played key roles in tumour growth, EMT, tumour immunity, and drug resistance, targeting DCLK1 is a potential strategy for treating cancers.

Cancer stem cells (CSCs) exhibit unique self-renewal properties and are regarded as the key initiator in tumour formation, development, metastasis, and recurrenceCitation8,Citation9. Because CSCs have strong resistance to traditional drug treatment, new strategies targeting CSCs have been soughtCitation10,Citation11. DCLK1 is regarded as a marker of various cancer stem cells and it regulates pro-survival signalling and self-renewal of tumour cellsCitation12–14 (). In the adenomatous polyposis coli (APC) mouse model, the expression of DCLK1 is positively correlated with other stem cell markers, and the enteroids formed from the intestinal DCLK+ cells display higher pluripotency and pro-survival signalling. DCLK1 knockdown in APCMin/+ mice reduced the ability of colon cancer cells to self-renew and surviveCitation14. Furthermore, DCLK1 marks a population of tumour stem-like cells in renal cell carcinoma (RCC). A novel monoclonal antibody targeting DCLK1 alternative splice variants-positive cells blocked RCC tumorigenesis in vivo and siRNA mediated knockdown of DCLK1 resulted in decreased expression of EMT and pluripotency factors in RCCCitation15,Citation16. In addition, small extracellular vesicle (sEV/exosome) biogenesis and EMT are inhibited in gastric cancer (GC)Citation17 by a specific small molecule inhibitor of DCLK1.

Figure 1. The role of DCLK1 in tumours.

Figure 1. The role of DCLK1 in tumours.

The crystal structure of DCLK1 has been reported, which provides the necessary structure information for the design of DCLK1 inhibitorsCitation18. However, DCLK1 and the Leucine-rich repeat kinase 2 (LRRK2) share a similar inhibitor profile. And XMD8-92 and LRRK-IN-1, which are widely used DCLK1 inhibitors, do not have good selectivityCitation19,Citation20. Therefore, we conducted chemical modifications of the structures of XMD8-92 and LRRK2-IN-1 (), and various effective DCLK1 inhibitors with higher selectivity profiles were synthesised. Their structure–activity relationship was also investigated.

Figure 2. Crystal structure of DCLK1-KD in complex with DCLK1-IN-1 (PDB: 7kxw) and molecular design.

Figure 2. Crystal structure of DCLK1-KD in complex with DCLK1-IN-1 (PDB: 7kxw) and molecular design.

Results and discussion

The molecular design

DCLK1-IN-1 was an efficient DCLK1 selective inhibitor developed by Gray et alCitation5. XMD8-92, LRRK-IN-1, and DCLK1-IN-1 share similar scaffold structures, namely the benzopyrimido diazepine moiety. As shown in , we plan to introduce substitution groups on the A-ring, such as electron withdrawing or electron donating groups, to enhance the interactions with surrounding amino acid residues. On the B-ring, we plan to replace the R2 and R3 with more bulky groups because there is still space in the kinase pocket adjacent to them. The C-ring has a strong interaction with VAL468, and we intend to keep it. G1 and G2 are located at the interface between protein and solvent, and we hope to introduce hydrophilic groups to improve the physicochemical properties of the resulting compounds and their selectivity to DCLK1.

Chemistry

The synthesis method for XMD8-92 analogues reported by Gray’s et al.Citation20,Citation21 was adopted and modified in this study. As shown in Scheme 1, substituted anthranilates 1-1–1-10 were transformed into N-methylated products 2-1–2-10 in the presence of dimethyl carbonate and NaY molecular sievesCitation22. Compounds 3-1–3-10 were synthesised through the nucleophilic substitution of 2,4-dichloro-5-nitropyrimidine by the secondary amine of compounds 2-1–2-10. The benzopyrimido diazepines 4-1–4-10 were achieved by reduction of the nitro group of compounds 3-1–3-10 followed by the spontaneous closure of the seven membered ring. Next, compounds 5-1–5-10 were obtained via methylation of compounds 4-1–4-10 by methyl iodide and NaH in anhydrous THF. Finally, A1–10 was obtained via Pd-catalysed Buchwald-Hartwig cross coupling reaction in low to moderate yields. As shown in Scheme 2, the synthesis of the B series is similar to that of the A series. The series C and D compounds were modified at the G1 and G2 positions ().

Scheme 1. Synthesis of compounds A1A10. Reagents and conditions: (a) NaY, 150 °C, dimethyl carbonate, 23–55%; (b) DIEA, 1,4-dioxane, 50 °C, 33–89%; (c) SnCl2·2H2O, EtOAc, 70 °C, 41–89%; (d) CH3I, NaH, THF, 0 °C, 48–100%; (e) Pd(OAc)2, XPhos, Cs2CO3, t-BuOH, 110 °C, 15–41%.

Scheme 1. Synthesis of compounds A1–A10. Reagents and conditions: (a) NaY, 150 °C, dimethyl carbonate, 23–55%; (b) DIEA, 1,4-dioxane, 50 °C, 33–89%; (c) SnCl2·2H2O, EtOAc, 70 °C, 41–89%; (d) CH3I, NaH, THF, 0 °C, 48–100%; (e) Pd(OAc)2, XPhos, Cs2CO3, t-BuOH, 110 °C, 15–41%.

Scheme 2. Synthesis of compounds B1B3. (a) DIEA, 1,4-dioxane, 50 °C, 70–85%; (c) SnCl2·2H2O, EtOAc, 70 °C, 43–95%; (d) R2I, NaH, THF, 0 °C, 57–100%; (e) Pd(OAc)2, XPhos, Cs2CO3, t-BuOH, 110 °C, 15–41%.

Scheme 2. Synthesis of compounds B1–B3. (a) DIEA, 1,4-dioxane, 50 °C, 70–85%; (c) SnCl2·2H2O, EtOAc, 70 °C, 43–95%; (d) R2I, NaH, THF, 0 °C, 57–100%; (e) Pd(OAc)2, XPhos, Cs2CO3, t-BuOH, 110 °C, 15–41%.

The synthesis of series C compounds proceeded from the synthesis of G1 and G2 groups before carrying out coupling reaction with compound 13 (Scheme 3). Most of the compounds of series D were also prepared via similar procedure. Alternatively, compounds D1, D2, D5, D7, and D8 were obtained by coupling reaction of G1/G2 moiety, followed by further modification (Scheme 4). It is worth mentioning that compound D8 was synthesised with a longer linker, which could be used as a probe for DCLK1. Furthermore, some compounds were introduced with hydrophilic groups, such as compounds D5 and D6 with hydrophilic sulfoxide or sulphone groups, respectively, and compound D7 with a fructose group via the Maillard reactionCitation23. In addition, compounds with lipophilic G2 groups were also synthesised, such as D10D14. In total, more than 30 compounds were synthesised to evaluate their DCLK1 inhibitory activity.

Scheme 3. Synthesis of compounds C1, C2. (a) K2CO3, DMF, 50 °C, 80–96%; (b) SnCl2·2H2O, EtOAc, 70 °C, 52–78%; (c) Pd(OAc)2, XPhos, Cs2CO3, t-BuOH, 110 °C, 35–46%.

Scheme 3. Synthesis of compounds C1, C2. (a) K2CO3, DMF, 50 °C, 80–96%; (b) SnCl2·2H2O, EtOAc, 70 °C, 52–78%; (c) Pd(OAc)2, XPhos, Cs2CO3, t-BuOH, 110 °C, 35–46%.

Scheme 4. Synthesis of compounds D1D16. (a) Pd(OAc)2, XPhos, Cs2CO3, t-BuOH, 110 °C, 30–48%; (b)H2, Pd/C, r.t., 100%; (c) Et3N, CH2Cl2, r.t.; (d) LiOH (2 M), THF, 90% over two steps; (e) CH3COSK, DMF, 100 °C, 38%; (f) K2CO3, DMF, 90 °C, 91%; (g) H2O2, AcOH, r.t., 95%; (h) TFA, CH2Cl2, r.t., 100%; (i) AcOH, EtOH, reflux, 82%.

Scheme 4. Synthesis of compounds D1–D16. (a) Pd(OAc)2, XPhos, Cs2CO3, t-BuOH, 110 °C, 30–48%; (b)H2, Pd/C, r.t., 100%; (c) Et3N, CH2Cl2, r.t.; (d) LiOH (2 M), THF, 90% over two steps; (e) CH3COSK, DMF, 100 °C, 38%; (f) K2CO3, DMF, 90 °C, 91%; (g) H2O2, AcOH, r.t., 95%; (h) TFA, CH2Cl2, r.t., 100%; (i) AcOH, EtOH, reflux, 82%.

In vitro evaluation of DCLK1 inhibitory activity

All compounds were tested for their kinase inhibitory activity at the concentration of 1 μM. As shown in , the kinase activity of DCLK1 and LRRK2 after drug treatment was presented as a percentage, with 100% indicating no inhibitory activity and 0% indicating complete inhibition. Compounds D1, D2, D5-D8, D11, D12, D14, and D15 exhibited higher DCLK1 kinase inhibitory activity than XMD8-92. In addition, at the same concentration, compounds A6, A9, B3, D1, D2, and D7 exhibited higher selectivity towards DCLK1 comparing with LRRK2. Compounds with high activity or good selectivity were selected for further IC50 determination (). The IC50 values of XMD8-92 for DCLK1 and LRRK2 kinases are similar, which indicates poor selectivity. Most compounds exhibited good DCLK1 inhibitory activity, and nine compounds exhibited higher DCLK1 inhibitory activity than XDM8-92. It is worth noting that compounds D1, D2, and D8 had significantly increased DCLK1 kinase selectivity with enhanced kinase inhibitory activity.

Table 1. DCLK1 and LRRK2 inhibitory activities of the synthesised compounds at the concentration of 1 uM.

Table 2. DCLK1 and LRRK2 inhibitory IC50 values of the synthesised compounds.

Structure–activity relationships

We analysed the structure–activity relationship based on the kinase inhibitory data of the synthesised compounds (). Firstly, any substituents on the A-ring led to decreased or lost activity, even the smallest F atom. Secondly, when the R2 substituent was ethyl, the DCLK1 selectivity of compound B3 was improved, indicating that this position may accommodate larger groups, which was also mentioned in Gray’s reportsCitation5,Citation24. The DCLK1 inhibitory activity of compound B2 decreased, indicating that the R3 cannot be H atom. Compared with compound C2 with a pyridine ring structure, the DCLK1 inhibitory activity of compound C1 with an aliphatic ring was weakened, indicating the necessity of G1 being an aromatic ring. The introduction of carboxyl groups at G2 increased the DCLK1 selectivity for compounds D1 and D2. The introduction of difluorinated compound D14 also has higher DCLK1 selectivity than monofluorinated compounds D10 and D13. Therefore, we believe that the G2 group is more important for the selectivity of DCLK1. In addition, compared with D10–D14 with hydrophobic groups, D5–D7 carrying hydrophilic G2 groups exhibited little effects on the activity and selectivity of DCLK1. Interestingly, when the G2 moiety was glucose (D7) or long-chain group (D8), enhanced DCLK1 inhibitory activity and selectivity were observed, indicating the potential in developing more potent and selective DCLK1 inhibitors with this class of compounds.

Figure 3. Structure–activity relationship analysis of DCLK1 inhibitors.

Figure 3. Structure–activity relationship analysis of DCLK1 inhibitors.

Anti-proliferation activity assay

To determine whether DCLK1 was required for tumour cell proliferation, the cytotoxicity of compounds was evaluated in colorectal cancer cell line HCT116. HCT116 was treated with compounds for 72 h, and the cell viability was determined using CCK8 assay (). The antiproliferative effects of most compounds on HCT116 were correlated with their DCLK1 inhibitory activities, except for compounds D1 and D2. The poor cytotoxicity of compounds D1 and D2 may be attributed to their poor cell membrane permeability due to the negatively charged carboxylates. Compounds D5–8, D11–12, and D14–15 showed higher anti-proliferate activity than the positive control XMD8-92. Overall, targeting DCLK1 inhibited the proliferation of HCT116, indicating the importance of DCLK1 in tumour growth.

Figure 4. The antiproliferative effects of most compounds on HCT116.

Figure 4. The antiproliferative effects of most compounds on HCT116.

Molecular docking study

Due to the excellent kinase inhibitory activity exhibited by compounds D1 and D8, the molecular docking study was performed to predict the possible binding mode with DCLK1 subunitCitation18 (PDB ID: 5JZN). Through docking software, compounds D1 and D8 were successfully localised in the binding pocket of DCLK1 subunit. Similar to the crystal structures of DCLK1-KD in complex with reported inhibitorsCitation25, the benzodiazepine scaffolds are located on the inner side of the hydrophobic pocket. As shown in , compound D1 exhibited three hydrogen bonds. The carbonyl group on the diazepine ring formed a hydrogen bond with LYS419. Notably, two hydrogen bonds were formed by the carboxyl group with residues LYS469 and ARG394. In addition, the carboxyl group formed a salt bridge with LYS469 which enhanced the interaction between D1 and DCLK1. The docking results of compounds D8 and D1 were similar. Compound D8 formed two hydrogen bonds with residues LYS469 and ARG394. The hydrophobic interaction of benzodiazepine rings matched the surrounding hydrophobic amino acids. And the benzene ring had a pπ conjugation effect with LYS419. These interactions maintained the activity of compound D8 against DCLK1 even with a long “tail”. Compared to the crystal structure of DCLK1-IN-1 in , compounds D1 and D8 gave up hydrogen bonding with residue VAL468 and chose to form more hydrogen bonds with residues LYS469 and ARG394, which may be more advantageous in enhancing the binding force and thus enhancing the inhibitory activity against DCLK1. Interestingly, compound D8 with a long-chain group replacing the carboxylic group did not undermine the binding to DCLK1, which also suggests that compound D8 may be suitable for developing a probe targeting DCLK1, such as proteolysis-targeting chimaeras (PROTACs)Citation26. The docking results somewhat explain the elevation of DCLK1 kinase activity by compounds D1 and D8 as well as validate the discussion in the structure–activity relationship analysis.

Figure 5. Docking of compounds D1 and D8 to the residues of DCLK1 subunit (PDB ID: 5JZN).

Figure 5. Docking of compounds D1 and D8 to the residues of DCLK1 subunit (PDB ID: 5JZN).

Conclusion

DCLK1 is an important marker of cancer stem cell and an important anti-tumour target. In this study, we synthesised a series of derivatives of XMD8-92 which is widely used as a non-selective DCLK1 inhibitor. We found that compounds D1, D2, D5, D6, D7, D8, D12, D14, and D15 exhibited higher activity than XMD8-92, among which compounds D1, D2, and D8 exhibited good DCLK1 selectivity. The cytotoxicity assay demonstrated that this series of compounds are suitable leads for developing new anti-tumour drugs.

Experimental section

Chemistry

All reagents and solvents were obtained from commercial suppliers (Bide Pharmatech, Energy Chemical, Alfa, etc.) without further purification unless noted. Reactions were monitored by TLC. Thin layer chromatography was carried out using TLC silica gel 60 F254 plates. Flash column chromatography was performed with 200–300 mesh silica gel. The NMR spectrum was recorded on a Bruker-400 NMR spectrometer, with TMS as an internal standard and chemical shifts reported in ppm (δ). Coupling constants (J) are reported in Hz. The melting point was measured by an X-5 micro melting point metre. High-resolution mass spectra (HRMS) were obtained on a Shimadzu LCMS-IT-TOF mass spectrometer.

Preparation of intermediate 2-1–2-10

A sealed tube (100 ml of internal volume) was charged with a solution of compound 1-1–1-10 (0.12 M) and NaY (1:1 w/w) in dimethyl carbonate (30 ml). Air in the reaction mixture was replaced by N2 at room temperature. The sealed tube was then heated at 150 °C for 15 h. After the reaction was completed, the sealed tube was cooled to room temperature and opened slowly. The NaY was filtered out and the filtrate was concentrated in vacuo. Purification by silica gel column chromatography (petroleum ether/EtOAc, 20:1 v/v) gave intermediate 2-1–2-10 (23–55%).

Methyl 4-methyl-2-(methylamino)benzoate (2–4)

White solid; yield 51%; m.p. 49–50 °C; 1H NMR (400 MHz, CDCl3) δ 7.80 (d, J = 8.1 Hz, 1H), 7.62 (s, 1H), 6.49 (s, 1H), 6.44 (d, J = 8.3 Hz, 1H), 3.86 (s, 3H), 2.93 (d, J = 5.1 Hz, 3H), 2.35 (s, 3H).13C NMR (101 MHz, CDCl3) δ 169.06, 152.04, 145.41, 131.49, 115.76, 110.95, 107.45, 51.28, 29.54, 22.19.

Preparation of intermediate 3-1–3-10 or 7-1–7-3

2 or 6 (1 eq), 2,4-dichloro-5-nitropyrimidine (1.5 eq), and DIEA (1.5 eq) were dissolved in 1,4-dioxane and heated at 50 °C for 6–10 h. The solvent was concentrated, and the residue was diluted with water (50 ml) and extracted with DCM (50 ml) × 3. The organics were combined and washed with brine (20 ml), dried over Na2SO4, filtered, and concentrated in vacuo. Purification by silica gel column chromatography (eluant: petroleum ether/EtOAc, 10:1 v/v) gave intermediate 3-1–3-10 or 7-1–7-3 (33–89%).

Methyl 3-chloro-2-((2-chloro-5-nitropyrimidin-4-yl)amino)benzoate (3-1)

Yellow solid; yield 33%; m.p. 115–116 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.71 (s, 1H), 9.24 (s, 1H), 7.91 (t, J = 8.2 Hz, 2H), 7.56 (t, J = 8.0 Hz, 1H), 3.73 (s, 3H). 13C NMR (101 MHz, DMSO) δ 165.27, 162.65, 158.52, 154.90, 134.15, 134.04, 133.60, 130.23, 129.88, 129.58, 127.83, 53.00. HRMS-ESI (m/z): [M + H]+ calcd for C12H8Cl2N4O4: 342.9995, found: 342.9993.

Methyl 2-((2-chloro-5-nitropyrimidin-4-yl)amino)-3-methylbenzoate (3-2)

Yellow solid; yield 62%; m.p. 167–168 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.57 (s, 1H), 9.19 (s, 1H), 7.79 (d, J = 7.7 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.43 (t, J = 7.7 Hz, 1H), 3.70 (s, 3H), 2.24 (s, 3H). 13C NMR (101 MHz, DMSO) δ 166.32, 162.67, 158.19, 154.98, 137.50, 135.15, 135.07, 128.60, 128.02, 127.99, 52.63, 18.45. HRMS-ESI (m/z): [M + H]+ calcd for C13H11ClN4O4: 323.0542, found: 323.0543.

Methyl 4-chloro-2-((2-chloro-5-nitropyrimidin-4-yl)(methyl)amino)benzoate (3-3)

White solid; yield 51%; m.p. 135–136 °C; 1H NMR (400 MHz, CDCl3) δ 8.55 (s, 1H), 8.00 (d, J = 8.5 Hz, 1H), 7.43 (dd, J = 8.5, 2.1 Hz, 1H), 7.24 (d, J = 2.1 Hz, 1H), 3.81 (s, 3H), 3.57 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 163.99, 161.00, 155.55, 154.64, 144.11, 140.10, 133.67, 131.71, 129.11, 127.99, 52.68, 41.97. HRMS-ESI (m/z): [M + H]+ calcd for C13H10Cl2N4O4: 357.0152, found: 357.0151.

Methyl 2-((2-chloro-5-nitropyrimidin-4-yl)(methyl)amino)-4-methylbenzoate (3-4)

Yellow solid; yield 85%; m.p. 143–144 °C; 1H NMR (400 MHz, CDCl3) δ 8.45 (s, 1H), 7.96 (d, J = 8.0 Hz, 1H), 7.24 (d, J = 8.1 Hz, 1H), 6.98 (s, 1H), 3.81 (s, 3H), 3.57 (s, 3H), 2.40 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 164.69, 160.80, 155.06, 154.54, 145.54, 142.95, 132.70, 131.94, 129.60, 127.78, 122.77, 52.35, 41.91, 21.39. HRMS-ESI (m/z): [M + H]+ calcd for C14H13ClN4O4: 337.0698, found: 337.0696.

Methyl 5-chloro-2-((2-chloro-5-nitropyrimidin-4-yl)(methyl)amino)benzoate (3-5)

Brown solid; yield 73%; m.p. 198–199 °C; 1H NMR (400 MHz, CDCl3) δ 8.49 (s, 1H), 7.99 (d, J = 2.5 Hz, 1H), 7.54 (dd, J = 8.5, 2.5 Hz, 1H), 7.16 (d, J = 8.5 Hz, 1H), 3.80 (s, 3H), 3.52 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 163.60, 160.89, 155.49, 154.63, 141.47, 134.59, 134.23, 132.51, 131.69, 129.07, 127.36, 52.81, 42.02. HRMS-ESI (m/z): [M + H]+ calcd for C13H10Cl2N4O4: 357.0152, found: 357.0161.

Methyl 2-((2-chloro-5-nitropyrimidin-4-yl)(methyl)amino)-5-fluorobenzoate (3-6)

White solid; yield 72%; m.p. 118–119 °C; 1H NMR (400 MHz, CDCl3) δ 8.48 (s, 1H), 7.75 (dd, J = 8.7, 2.9 Hz, 1H), 7.36–7.29 (m, 1H), 7.23 (dd, J = 8.8, 4.9 Hz, 1H), 3.84 (d, J = 1.0 Hz, 3H), 3.56 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 163.60, 162.68, 160.86, 160.18, 155.28, 154.76, 138.96, 129.90, 129.82, 121.44, 119.61, 52.82, 42.10. HRMS-ESI (m/z): [M + H]+ calcd for C13H10ClFN4O4: 341.0447, found: 341.0446.

Methyl 2-((2-chloro-5-nitropyrimidin-4-yl)amino)-5-(trifluoromethyl)benzoate (3-7)

Yellow solid; yield 46%; m.p. = 149–150 °C; 1H NMR (400 MHz, CDCl3) δ 12.89 (s, 1H), 9.28 (s, 1H), 8.82 (d, J = 8.8 Hz, 1H), 8.41 (d, J = 2.3 Hz, 1H), 7.91 (dd, J = 8.9, 2.3 Hz, 1H), 4.07 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 166.49, 163.63, 157.80, 153.13, 141.08, 130.45, 128.64, 127.94, 126.87, 124.68, 123.51, 119.06, 53.21. HRMS-ESI (m/z): [M + H]+ calcd for C13H8ClF3N4O4: 377.0259, found: 377.0258.

Methyl 2-((2-chloro-5-nitropyrimidin-4-yl)(methyl)amino)-5-methylbenzoate (3-8)

White solid; yield 89%; m.p. 96–97 °C; 1H NMR (400 MHz, CDCl3) δ 8.46 (s, 1H), 7.86 (s, 1H), 7.40 (d, J = 8.1 Hz, 1H), 7.09 (d, J = 8.1 Hz, 1H), 3.82 (s, 3H), 3.57 (s, 3H), 2.42 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 164.90, 160.79, 155.11, 154.73, 140.45, 139.16, 134.83, 133.07, 131.90, 127.32, 125.55, 52.44, 42.03, 21.07. HRMS-ESI (m/z): [M + H]+ calcd for C14H13ClN4O4: 337.0698, found: 337.0695.

Methyl 2-((2-chloro-5-nitropyrimidin-4-yl)(methyl)amino)-6-fluorobenzoate (3-9)

Yellow solid; yield 73%; m.p. 91–92 °C; 1H NMR (400 MHz, CDCl3) δ 8.60 (s, 1H), 7.52 (q, J = 7.8 Hz, 1H), 7.17 (t, J = 8.9 Hz, 1H), 7.03 (d, J = 8.0 Hz, 1H), 3.81 (s, 3H), 3.56 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 163.21, 162.26, 161.07, 159.70, 155.82, 154.72, 143.11, 133.48, 131.71, 122.47, 116.95, 52.89, 42.31. HRMS-ESI (m/z): [M + H]+ calcd for C13H10ClFN4O4: 341.0447, found: 341.0445.

Methyl 2-((2-chloro-5-nitropyrimidin-4-yl)(methyl)amino)-6-methylbenzoate (3-10)

Yellow solid; yield 85%; m.p. 122–123 °C; 1H NMR (400 MHz, CDCl3) δ 8.55 (s, 1H), 7.41 (t, J = 7.8 Hz, 1H), 7.24 (d, J = 7.7 Hz, 1H), 7.08 (d, J = 7.9 Hz, 1H), 3.74 (s, 3H), 3.55 (s, 3H), 2.37 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 167.13, 160.79, 155.50, 154.74, 141.01, 138.72, 131.97, 131.50, 130.87, 129.77, 124.26, 52.30, 42.25, 20.17. HRMS-ESI (m/z): [M + H]+ calcd for C14H13ClN4O4: 337.0698, found: 337.0695.

Preparation of intermediate 4-1–4-10 or 8-1–8-3

3-1–3-10 or 7-1–7-10 (1 eq) and SnCl2·2H2O (2 eq) in EtOAc (10 ml) was heated at 70 °C for 5–8 h. The solvent was concentrated, and the residue diluted with water (50 ml) and extracted with EtOAc (50 ml) × 3. The organics were combined and washed with brine (20 ml), dried over Na2SO4, filtered, and concentrated in vacuo to give the white or yellow powder, washed with MeOH and air dried to give intermediate 4-1–4-10 or 8-1–8-3 (43–95%).

2,10-Dichloro-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (4-1)

White solid; yield 52%; m.p. 246–247 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.40 (s, 1H), 8.24 (s, 1H), 8.12 (s, 1H), 7.78 (dd, J = 8.0, 1.6 Hz, 1H), 7.68 (dd, J = 7.9, 1.6 Hz, 1H), 7.09 (t, J = 7.9 Hz, 1H). 13C NMR (101 MHz, DMSO) δ 165.54, 157.00, 153.20, 149.80, 139.13, 134.75, 132.36, 124.51, 124.12, 123.16, 122.61. HRMS-ESI (m/z): [M + H]+ calcd for C11H6Cl2N4O: 280.9991, found: 280.9992.

2-Chloro-10-methyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (4-2)

White solid; yield 58%; m.p. 210–211 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.23 (s, 1H), 8.30 (s, 1H), 8.06 (s, 1H), 7.62 (d, J = 7.8 Hz, 1H), 7.36 (d, J = 7.3 Hz, 1H), 6.98 (t, J = 7.6 Hz, 1H), 2.34 (s, 3H). 13C NMR (101 MHz, DMSO) δ 167.15, 158.41, 153.17, 149.23, 141.40, 136.08, 130.99, 128.54, 123.70, 123.29, 122.77, 18.45. HRMS-ESI (m/z): [M + H]+ calcd for C12H9ClN4O: 261.0532, found: 261.0537.

2,9-Dichloro-11-methyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (4-3)

White solid; yield 71%; m.p. 241–242 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.52 (s, 1H), 8.18 (s, 1H), 7.74 (d, J = 8.4 Hz, 1H), 7.34 (s, 1H), 7.28 (d, J = 8.4 Hz, 1H), 3.35 (s, 3H). 13C NMR (101 MHz, DMSO) δ 166.66, 160.88, 153.48, 150.13, 148.97, 138.89, 133.65, 124.62, 124.56, 124.31, 120.13, 37.35. HRMS-ESI (m/z): [M + H]+ calcd for C12H8Cl2N4O: 295.0148, found: 295.0150.

2-Chloro-9,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (4-4)

White solid; yield 82%; m.p. 238–239 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.36 (s, 1H), 8.13 (s, 1H), 7.62 (d, J = 7.9 Hz, 1H), 7.09 (s, 1H), 7.02 (d, J = 8.0 Hz, 1H), 3.33 (s, 3H), 2.34 (s, 3H). 13C NMR (101 MHz, DMSO) δ 167.52, 161.41, 153.35, 149.61, 147.69, 144.76, 131.96, 125.27, 124.42, 123.01, 120.46, 37.25, 21.51. HRMS-ESI (m/z): [M + H]+ calcd for C13H11ClN4O: 275.0694, found: 275.0692.

2,8-Dichloro-11-methyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (4-5)

White solid; yield 68%; m.p. 230–231 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.60 (s, 1H), 8.16 (s, 1H), 7.66 (s, 1H), 7.63 (s, 1H), 7.30 (s, 1H), 3.32 (s, 3H). 13C NMR (101 MHz, DMSO) δ 166.27, 161.11, 153.57, 150.12, 146.76, 133.70, 131.10, 128.58, 127.49, 124.16, 122.28, 37.35. HRMS-ESI (m/z): [M + H]+ calcd for C12H8Cl2N4O: 295.0148, found: 295.0148.

2-Chloro-8-fluoro-11-methyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (4-6)

White solid; yield 70%; m.p. 216–217 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.60 (s, 1H), 8.16 (s, 1H), 7.50–7.43 (m, 2H), 7.33 (dd, J = 9.9, 4.6 Hz, 1H), 2.09 (s, 3H). 13C NMR (101 MHz, DMSO) δ 166.32, 161.40, 159.74, 153.58, 149.93, 144.15, 127.71, 124.19, 122.41, 121.02, 117.88, 37.42. HRMS-ESI (m/z): [M + H]+ calcd for C12H8ClFN4O: 279.0443, found: 279.0442.

2-Chloro-8-(trifluoromethyl)-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (4-7)

Yellow solid; yield 41%; m.p. = 241–242 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.26 (s, 1H), 10.25 (s, 1H), 8.09 (d, J = 2.4 Hz, 1H), 7.98 (s, 1H), 7.73 (dd, J = 8.7, 2.4 Hz, 1H), 7.26 (d, J = 8.5 Hz, 1H). 13C NMR (101 MHz, DMSO) δ 164.36, 155.50, 153.01, 148.59, 146.06, 131.27, 130.53, 125.68, 122.64, 121.53, 121.06, 120.18. HRMS-ESI (m/z): [M + H]+ calcd for C12H6ClF3N4O: 315.0225, found: 315.0225.

2-Chloro-8,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (4-8)

White solid; yield 89%; m.p. 218–219 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.40 (s, 1H), 8.12 (s, 1H), 7.53 (s, 1H), 7.39 (d, J = 8.3 Hz, 1H), 7.16 (d, J = 8.5 Hz, 1H), 3.33 (s, 3H), 2.28 (s, 3H). 13C NMR (101 MHz, DMSO) δ 167.64, 161.56, 153.39, 149.56, 145.42, 134.71, 133.87, 132.02, 125.61, 124.30, 120.03, 37.18, 20.33. HRMS-ESI (m/z): [M + H]+ calcd for C13H11ClN4O: 257.0694, found: 257.0693.

2-Chloro-7-fluoro-11-methyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (4-9)

White solid; yield 43%; m.p. 215–216 °C; 1H NMR (400 MHz, DMSO-d6) δ 10.58 (s, 1H), 8.20 (s, 1H), 7.55 (q, J = 7.8, 7.4 Hz, 1H), 7.15 (d, J = 8.4 Hz, 1H), 7.08 (t, J = 9.4 Hz, 1H), 3.33 (s, 3H). 13C NMR (101 MHz, DMSO) δ 163.77, 162.34, 153.59, 150.77, 150.24, 133.89, 133.78, 124.31, 115.99, 113.10, 112.88, 37.13. HRMS-ESI (m/z): [M + H]+ calcd for C12H8ClFN4O: 279.0443, found: 279.0444.

2-Chloro-7,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (4-10)

Yellow solid; yield 49%; m.p. 145–146 °C; 1H NMR (400 MHz, CDCl3) δ 7.63 (s, 1H), 7.32 (t, J = 7.8 Hz, 1H), 7.21 (d, J = 7.6 Hz, 1H), 6.93 (d, J = 7.8 Hz, 1H), 3.89 (s, 3H), 2.38 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 167.91, 165.01, 154.59, 151.69, 149.62, 140.86, 130.81, 128.96, 127.81, 126.28, 115.68, 36.00, 21.08, 0.01. HRMS-ESI (m/z): [M + H]+ calcd for C13H11ClN4O: 275.0694, found: 275.0699.

Preparation of intermediate 5-1–5-10 or 9-1–9-3

4-1–4-10 or 8-1–8-3 (1 eq), MeI (6 eq) and NaH (2 eq, 60% suspension in mineral oil) was added in THF (10 ml) at −5 °C. After the reaction was complete as monitored by TLC, the solvent was diluted with water (30 ml) and extracted with EtOAc (30 ml) × 3. The organics were combined, dried over Na2SO4, filtered, and concentrated in vacuo. Purification by silica gel column chromatography (eluant: petroleum ether/EtOAc, 4:1 v/v) gave intermediate 5-1–5-10 or 9-1–9-3 (48–100%).

2,10-Dichloro-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (5-1)

Yellow solid; yield 48%; m.p. 178–179 °C; 1H NMR (400 MHz, CDCl3) δ 7.94 (d, J = 8.0 Hz, 1H), 7.53 (d, J = 7.8 Hz, 1H), 7.13 (s, 1H), 6.99 (t, J = 8.2 Hz, 1H), 3.86 (s, 3H), 3.38 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 166.93, 151.53, 145.55, 142.63, 136.51, 134.35, 132.95, 132.03, 128.90, 127.22, 124.22, 39.63, 36.79. HRMS-ESI (m/z): [M + H]+ calcd for C13H10Cl2N4O: 309.0304, found: 309.0303.

2-Chloro-5,10,11-trimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (5-2)

Yellow solid; yield 68%; m.p. 188–189 °C; 1H NMR (400 MHz, CDCl3) δ 7.88 (d, J = 8.0 Hz, 1H), 7.32 (d, J = 7.4 Hz, 1H), 7.04 (s, 1H), 6.99 (t, J = 7.6 Hz, 1H), 3.74 (s, 3H), 3.38 (s, 3H), 2.34 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 168.07, 149.81, 145.19, 143.70, 136.49, 135.37, 134.93, 131.26, 128.80, 125.75, 124.16, 39.56, 36.36, 19.77, 0.03. HRMS-ESI (m/z): [M + H]+ calcd for C14H13ClN4O: 289.0851, found: 289.0847.

2,9-Dichloro-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (5-3)

White solid; yield 100%; m.p. 165–166 °C; 1H NMR (400 MHz, CDCl3) δ 8.28 (s, 1H), 7.81 (d, J = 8.4 Hz, 1H), 7.17 (dd, J = 8.4, 1.9 Hz, 1H), 7.11 (d, J = 1.9 Hz, 1H), 3.52 (s, 3H), 3.43 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 166.74, 163.33, 154.90, 151.60, 149.27, 139.30, 133.89, 128.32, 124.62, 124.13, 118.35, 38.29, 36.35. HRMS-ESI (m/z): [M + H]+ calcd for C13H10Cl2N4O: 309.0304, found: 309.0302.

2-Chloro-5,9,11-trimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (5-4)

White solid; yield 100%; m.p. 162–163 °C; 1H NMR (400 MHz, CDCl3) δ 8.23 (s, 1H), 7.76 (d, J = 7.9 Hz, 1H), 7.00 (d, J = 8.0 Hz, 0H), 6.90 (s, 1H), 3.51 (s, 3H), 3.43 (s, 3H), 2.39 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 167.69, 163.94, 154.60, 151.17, 148.26, 143.94, 132.57, 128.55, 125.25, 123.00, 118.47, 38.27, 36.26, 21.67. HRMS-ESI (m/z): [M + H]+ calcd for C14H13ClN4O: 289.0851, found: 289.0849.

2,8-Dichloro-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (5-5)

White solid; yield 96%; m.p. 179–180 °C; 1H NMR (400 MHz, CDCl3) δ 8.27 (s, 1H), 7.82 (d, J = 2.6 Hz, 1H), 7.42 (dd, J = 8.9, 2.6 Hz, 1H), 7.05 (d, J = 8.8 Hz, 1H), 3.52 (s, 3H), 3.41 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 166.37, 163.54, 154.88, 151.62, 146.88, 132.81, 132.18, 129.96, 128.16, 127.17, 119.40, 38.47, 36.36. HRMS-ESI (m/z): [M + H]+ calcd for C13H10Cl2N4O: 309.0304, found: 309.0305.

2-Chloro-8-fluoro-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (5-6)

White solid; yield 92%; m.p. 168–169 °C; 1H NMR (400 MHz, CDCl3) δ 8.25 (s, 1H), 7.53 (dd, J = 8.7, 3.1 Hz, 1H), 7.17 (m, 1H), 7.07 (dd, J = 9.1, 4.4 Hz, 1H), 3.51 (s, 3H), 3.39 (s, 3H).13C NMR (101 MHz, CDCl3) δ 166.41, 163.84, 160.19, 157.76, 154.84, 151.53, 144.44, 128.23, 127.48, 120.03, 118.86, 38.50, 36.44. HRMS-ESI (m/z): [M + H]+ calcd for C13H10ClFN4O: 293.0600, found: 293.0596.

2-Chloro-5,11-dimethyl-8-(trifluoromethyl)-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(5-7)

White solid; yield 90%; m.p. = 167–168 °C; 1H NMR (400 MHz, CDCl3) δ 8.31 (s, 1H), 8.15 (d, J = 2.3 Hz, 1H), 7.72 (dd, J = 8.7, 2.3 Hz, 1H), 7.23 (d, J = 8.7 Hz, 1H), 3.55 (s, 3H), 3.48 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 166.39, 163.10, 155.01, 151.87, 151.14, 130.25, 129.71, 128.12, 126.80, 126.08, 124.78, 118.47, 38.36, 36.47. HRMS-ESI (m/z): [M + H]+ calcd for C14H10ClF3N4O: 343.0568, found: 343.0565.

2-Chloro-5,8,11-trimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (5-8)

White solid; yield 100%; m.p. 173–174 °C; 1H NMR (400 MHz, CDCl3) δ 8.22 (s, 1H), 7.65 (d, J = 2.2 Hz, 1H), 7.27 (dd, J = 8.3, 2.8 Hz, 1H), 6.99 (d, J = 8.4 Hz, 1H), 3.51 (s, 3H), 3.40 (s, 3H), 2.33 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 167.85, 164.09, 154.62, 151.20, 145.98, 134.15, 133.72, 132.65, 128.42, 125.54, 117.87, 38.45, 36.17, 20.38. HRMS-ESI (m/z): [M + H]+ calcd for C14H13ClN4O: 289.0851, found: 289.0848.

2-Chloro-7-fluoro-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (5-9)

White solid; yield 97%; m.p. 172–173 °C; 1H NMR (400 MHz, CDCl3) δ 8.33 (s, 1H), 7.40 (m, 1H), 6.98–6.88 (m, 2H), 3.53 (s, 3H), 3.41 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 164.28, 163.50, 160.78, 154.81, 152.27, 150.56, 132.60, 128.49, 115.63, 113.67, 112.81, 37.96, 36.32. HRMS-ESI (m/z): [M + H]+ calcd for C13H10ClFN4O: 293.0600, found: 293.0597.

2-Chloro-5,7,11-trimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one (5-10)

White solid; yield 81%; m.p. 132–133 °C; 1H NMR (400 MHz, CDCl3) δ 8.29 (s, 1H), 7.29 (t, J = 8.1 Hz, 1H), 7.05 (d, J = 7.5 Hz, 1H), 6.99 (d, J = 8.3 Hz, 1H), 3.53 (s, 3H), 3.39 (s, 3H), 2.45 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 167.91, 165.01, 154.59, 151.69, 149.62, 140.86, 130.81, 128.96, 127.81, 126.28, 115.68, 38.29, 36.00, 21.08. HRMS-ESI (m/z): [M + H]+ calcd for C14H13ClN4O: 289.0851, found: 289.0848.

General procedure for synthesis of compound A1–A10 and B1–B3

A sealed tube with a mixture of 5-1–5-10 or 9-1–9-3 (1 eq), 1–(4-amino-3-ethoxyphenyl)piperidin-4-ol (1 eq), Pd(OAc)2 (0.05 eq), XPhos (0.1 eq), and Cs2CO3 (2 eq) in t-BuOH (5.0 ml) was heated at 110 °C for 8–16 h. The reaction was then filtered through celite and eluted with dichloromethane. The dichloromethane was removed in vacuo and the resulting crude product was purified by silica gel column chromatography (eluant: DCM/MeOH, 40:1 v/v) to afford the target product (15–41%).

10-Chloro-2-((2-ethoxy-4-(4-hydroxypiperidin-1-yl)phenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(A1)

Yellow solid; yield 21%; m.p. 130–131 °C; 1H NMR (400 MHz, CDCl3) δ 8.32 (d, J = 8.6 Hz, 1H), 8.15 (s, 1H), 7.68 (dd, J = 7.7, 1.6 Hz, 1H), 7.52 (s, 1H), 7.45 (dd, J = 8.0, 1.7 Hz, 1H), 7.15 (t, J = 7.8 Hz, 1H), 6.65–6.56 (m, 2H), 4.13 (q, J = 6.9 Hz, 2H), 3.90–3.83 (m, 1H), 3.65 (s, 3H), 3.54–3.47 (m, 5H), 2.95–2.87 (m, 2H), 2.10–2.00 (m, 2H), 1.81–1.72 (m, 2H), 1.49 (t, J = 7.0 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 167.62, 163.36, 156.20, 151.67, 148.04, 147.11, 144.56, 134.46, 132.83, 129.92, 128.70, 126.04, 122.38, 121.63, 119.04, 108.72, 102.09, 67.85, 64.23, 48.46, 38.56, 38.45, 34.43, 14.99. HRMS-ESI (m/z): [M + H]+ calcd for C26H29ClN6O3: 509.2062, found: 509.2059.

2-((2-Ethoxy-4-(4-hydroxypiperidin-1-yl)phenyl)amino)-5,10,11-trimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(A2)

Brown solid; yield 28%; m.p. 177–178 °C; 1H NMR (400 MHz, CDCl3) δ 7.98 (d, J = 8.5 Hz, 1H), 7.86 (d, J = 7.9 Hz, 1H), 7.28 (s, 1H), 7.22 (s, 1H), 6.95–6.87 (m, 2H), 6.60–6.55 (m, 2H), 4.13 (q, J = 7.0 Hz, 2H), 3.89–3.82 (m, 1H), 3.73 (s, 3H), 3.53–3.47 (s, 2H), 3.37 (s, 3H), 2.96–2.85 (m, 2H), 2.36 (s, 3H), 2.08–1.99 (m, 2H), 1.77–1.67 (m, 2H), 1.49 (t, J = 7.0 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 168.97, 152.28, 149.03, 148.59, 148.50, 146.33, 139.66, 135.64, 134.21, 130.99, 126.07, 122.61, 121.94, 121.77, 120.19, 108.75, 101.62, 67.74, 64.22, 48.11, 39.93, 34.25, 29.70, 20.05, 15.00. HRMS-ESI (m/z): [M + H]+ calcd for C27H32N6O3: 489.2609, found: 489.2608.

9-Chloro-2-((2-ethoxy-4-(4-hydroxypiperidin-1-yl)phenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(A3)

Brown solid; yield 31%; m.p. 162–163 °C; 1H NMR (400 MHz, CDCl3) δ 8.22 (d, J = 8.5 Hz, 1H), 8.14 (s, 1H), 7.80 (d, J = 8.4 Hz, 1H), 7.50 (s, 1H), 7.14–7.07 (m, 2H), 6.63–6.58 (m, 2H), 4.12 (q, J = 6.9 Hz, 2H), 3.91–3.82 (m, 1H), 3.54–3.47 (m, 5H), 3.41 (s, 3H), 2.95–2.87 (m, 2H), 2.06–2.01 (m, 2H), 1.81–1.70 (m, 2H), 1.48 (t, J = 6.9 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 167.42, 163.13, 156.15, 151.75, 150.41, 148.18, 147.20, 138.41, 133.53, 124.95, 123.81, 122.19, 120.51, 119.20, 117.75, 108.59, 102.15, 67.75, 64.25, 48.43, 38.11, 35.97, 34.36, 14.97. HRMS-ESI (m/z): [M + H]+ calcd for C26H29ClN6O3: 509.2062, found: 509.2063.

2-((2-Ethoxy-4-(4-hydroxypiperidin-1-yl)phenyl)amino)-5,9,11-trimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(A4)

White solid; yield 40%; m.p. 171–172 °C; 1H NMR (400 MHz, CDCl3) δ 8.26 (d, J = 8.6 Hz, 1H), 8.11 (s, 1H), 7.75 (d, J = 7.9 Hz, 1H), 7.48 (s, 1H), 6.95 (d, J = 7.9 Hz, 1H), 6.89 (s, 1H), 6.64–6.55 (m, 2H), 4.12 (q, J = 7.0 Hz, 2H), 3.90–3.83 (m, 1H), 3.55–3.44 (m, 5H), 3.42 (s, 3H), 2.94–2.86 (m, 2H), 2.36 (s, 3H), 2.08–2.01 (m, 2H), 1.80–1.70 (m, 2H), 1.48 (t, J = 7.0 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 168.38, 163.75, 156.04, 151.33, 149.39, 148.07, 147.09, 142.98, 132.21, 124.50, 123.75, 122.44, 120.87, 119.05, 117.91, 108.61, 102.17, 67.78, 64.23, 48.48, 38.11, 35.91, 34.40, 21.64, 14.98. HRMS-ESI (m/z): [M + H]+ calcd for C27H32N6O3: 489.2609, found: 489.2602.

8-Chloro-2-((2-ethoxy-4-(4-hydroxypiperidin-1-yl)phenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(A5)

Yellow solid; yield 28%; m.p. 173–174 °C; 1H NMR (400 MHz, CDCl3) δ 8.21 (d, J = 8.6 Hz, 1H), 8.12 (s, 1H), 7.81 (s, 1H), 7.50 (s, 1H), 7.35 (d, J = 8.9 Hz, 1H), 7.00 (d, J = 9.1 Hz, 1H), 6.60 (s, 1H), 6.58 (s, 1H), 4.11 (q, J = 7.0 Hz, 2H), 3.89–3.80 (m, 1H), 3.52–3.44 (m, 5H), 3.38 (s, 3H), 2.93–2.83 (m, 2H), 2.07–2.00 (m, 2H), 1.79–1.68 (m, 2H), 1.47 (t, J = 7.0 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 167.05, 163.40, 156.12, 151.75, 148.17, 148.05, 147.28, 132.05, 131.82, 129.06, 127.92, 122.10, 120.31, 119.19, 118.82, 108.55, 102.09, 67.67, 64.24, 48.41, 38.31, 35.98, 34.35, 14.97. HRMS-ESI (m/z): [M + H]+ calcd for C26H29ClN6O3: 509.2062, found: 509.2061.

2-((2-Ethoxy-4-(4-hydroxypiperidin-1-yl)phenyl)amino)-8-fluoro-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(A6)

Yellow solid; yield 23%; m.p. 166–167 °C; 1H NMR (400 MHz, CDCl3) δ 8.21 (d, J = 8.5 Hz, 1H), 8.11 (s, 1H), 7.57–7.49 (m, 2H), 7.14–7.06 (m, 1H), 7.02 (dd, J = 9.2, 4.4 Hz, 1H), 6.61–6.56 (m, 2H), 4.10 (q, J = 6.9 Hz, 2H), 3.87–3.78 (m, 1H), 3.53–3.42 (m, 5H), 3.37 (s, 3H), 2.92–2.83 (m, 2H), 2.06–1.98 (m, 2H), 1.78–1.68 (m, 2H), 1.46 (t, J = 7.0 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 167.13, 163.75, 159.87, 157.45, 156.13, 151.63, 148.16, 147.27, 145.62, 145.59, 128.06, 122.12, 120.39, 119.20, 118.21, 108.55, 102.09, 67.58, 64.24, 48.44, 38.35, 36.06, 34.34, 14.97. HRMS-ESI (m/z): [M + H]+ calcd for C26H29FN6O3: 493.2358, found: 493.2353.

2-((2-Ethoxy-4-(4-hydroxypiperidin-1-yl)phenyl)amino)-5,11-dimethyl-8-(trifluoromethyl)-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(A7)

Yellow solid; yield 15%; m.p. 119–120 °C; 1H NMR (400 MHz, CDCl3) δ 8.27–8.12 (m, 3H), 7.66 (d, J = 8.7 Hz, 1H), 7.52 (s, 1H), 7.19 (d, J = 8.7 Hz, 1H), 6.61 (s, 1H), 6.59 (s, 1H), 4.12 (q, J = 6.9 Hz, 2H), 3.90–3.83 (m, 1H), 3.60–3.44 (m, 8H), 2.96–2.87 (m, 2H), 2.10–1.99 (m, 2H), 1.79–1.74 (m, 2H), 1.48 (t, J = 6.9 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 167.08, 162.91, 156.18, 152.26, 152.00, 148.22, 147.34, 129.88, 129.56, 128.95, 126.79, 125.95, 122.02, 120.21, 119.22, 117.82, 108.53, 102.09, 67.76, 64.25, 48.37, 38.21, 36.09, 34.35, 14.96. HRMS-ESI (m/z): [M + H]+ calcd for C27H29F3N6O3: 543.2326, found: 543.2322.

2-((2-Ethoxy-4-(4-hydroxypiperidin-1-yl)phenyl)amino)-5,8,11-trimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(A8)

Yellow solid; yield 41%; m.p. 170–171 °C; 1H NMR (400 MHz, CDCl3) δ 8.23 (d, J = 8.5 Hz, 1H), 8.08 (s, 1H), 7.62 (d, J = 2.2 Hz, 1H), 7.48 (s, 1H), 7.20 (dd, J = 8.5, 2.2 Hz, 1H), 6.95 (d, J = 8.4 Hz, 1H), 6.60–6.56 (m, 2H), 4.09 (q, J = 7.0 Hz, 2H), 3.86–3.77 (m, 1H), 3.49–3.44 (m, 5H), 3.37 (s, 3H), 2.90–2.82 (m, 2H), 2.30 (s, 3H), 2.04–1.97 (m, 2H), 1.78–1.67 (m, 2H), 1.45 (t, J = 7.0 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 168.52, 163.95, 156.01, 151.33, 148.08, 147.15, 147.13, 133.21, 133.01, 132.29, 126.22, 122.34, 120.68, 119.09, 117.28, 108.60, 102.14, 67.57, 64.23, 48.49, 38.29, 35.82, 34.36, 20.38, 14.97. HRMS-ESI (m/z): [M + H]+ calcd for C27H32N6O3: 489.2609, found: 489.2602.

2-((2-Ethoxy-4-(4-hydroxypiperidin-1-yl)phenyl)amino)-7-fluoro-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(A9)

Yellow solid; yield 23%; m.p. 174–175 °C; 1H NMR (400 MHz, CDCl3) δ 8.21 (d, J = 8.9 Hz, 1H), 8.18 (s, 1H), 7.51 (s, 1H), 7.37–7.29 (m, 1H), 6.91–6.83 (m, 2H), 6.59 (s, 1H), 6.57 (s, 1H), 4.11 (q, J = 6.9 Hz, 2H), 3.89–3.80 (m, 1H), 3.54–3.44 (m, 5H), 3.38 (s, 3H), 2.93–2.85 (m, 2H), 2.10–2.00 (m, 2H), 1.79–1.68 (m, 2H), 1.47 (t, J = 7.0 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 164.20, 163.31, 160.77, 156.05, 152.42, 151.85, 148.17, 147.30, 131.93, 122.05, 120.52, 119.21, 116.01, 113.10, 111.99, 108.53, 102.05, 67.67, 64.23, 48.39, 37.80, 35.98, 34.34, 14.96. HRMS-ESI (m/z): [M + H]+ calcd for C26H29FN6O3: 493.2358, found: 493.2354.

2-((2-Ethoxy-4-(4-hydroxypiperidin-1-yl)phenyl)amino)-5,7,11-trimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(A10)

Yellow solid; yield 25%; m.p. = 184–185 °C; 1H NMR (400 MHz, CDCl3) δ 8.25 (d, J = 8.6 Hz, 1H), 8.16 (s, 1H), 7.48 (s, 1H), 7.22 (t, J = 7.9 Hz, 1H), 6.98 (dd, J = 12.0, 7.9 Hz, 2H), 6.64–6.55 (m, 2H), 4.12 (q, J = 7.0 Hz, 2H), 3.89–3.81 (m, 1H), 3.53–3.46 (m, 5H), 3.37 (s, 3H), 2.94–2.86 (m, 2H), 2.47 (s, 3H), 2.10–1.99 (m, 2H), 1.80–1.69 (m, 2H), 1.48 (t, J = 7.0 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 168.46, 164.94, 155.99, 151.90, 150.75, 148.09, 147.13, 140.40, 130.12, 127.03, 126.90, 122.38, 121.12, 119.11, 115.04, 108.63, 102.15, 67.77, 64.24, 48.45, 38.12, 35.67, 34.40, 21.06, 14.97. HRMS-ESI (m/z): [M + H]+ calcd for C27H32N6O3: 489.2609, found: 489.2606.

2-((2-Ethoxy-4-(4-hydroxypiperidin-1-yl)phenyl)amino)-11-methyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(B1)

Yellow solid; yield 29%; m.p. 165–166 °C; 1H NMR (400 MHz, CDCl3) δ 8.83 (s, 1H), 8.26 (d, J = 8.6 Hz, 1H), 8.01 (s, 1H), 7.91 (dd, J = 7.8, 1.7 Hz, 1H), 7.54–7.48 (m, 1H), 7.47 (s, 1H), 7.20–7.09 (m, 2H), 6.63–6.57 (m, 2H), 4.12 (q, J = 7.0 Hz, 2H), 3.90–3.83 (m, 1H), 3.53–3.47 (m, 2H), 3.46 (s, 3H), 2.94–2.85 (m, 2H), 2.10–2.01 (m, 2H), 1.79–1.73 (m, 2H), 1.48 (t, J = 7.0 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 169.51, 161.75, 156.61, 149.25, 149.08, 148.02, 146.96, 133.33, 132.05, 125.53, 123.65, 122.60, 118.93, 118.71, 115.15, 108.64, 102.22, 67.84, 64.22, 48.56, 36.67, 34.41, 14.99. HRMS-ESI (m/z): [M + H]+ calcd for C25H28N6O3: 461.2296, found: 461.2293.

2-((2-Ethoxy-4-(4-hydroxypiperidin-1-yl)phenyl)amino)-5-methyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(B2)

Yellow solid; yield 20%; m.p. 194–195 °C; 1H NMR (400 MHz, CDCl3) δ 8.16–8.09 (m, 2H), 7.95 (dd, J = 7.9, 1.6 Hz, 1H), 7.39–7.30 (m, 2H), 7.07 (t, J = 7.6 Hz, 1H), 6.81 (d, J = 8.0 Hz, 1H), 6.57 (s, 1H), 6.55 (s, 1H), 6.39 (s, 1H), 4.08 (q, J = 7.0 Hz, 2H), 3.88–3.81 (m, 1H), 3.49–3.45 (m, 5H), 2.91–2.83 (m, 2H), 2.06–1.99 (m, 2H), 1.74–1.70 (m, 2H), 1.44 (t, J = 7.0 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 167.61, 160.63, 156.30, 151.81, 148.40, 147.45, 145.07, 133.27, 132.86, 123.46, 123.18, 121.96, 119.63, 119.10, 118.89, 108.57, 102.15, 67.79, 64.19, 48.41, 38.51, 34.37, 14.97. HRMS-ESI (m/z): [M + H]+ calcd for C25H28N6O3: 461.2296, found: 461.2292.

2-((2-Ethoxy-4-(4-hydroxypiperidin-1-yl)phenyl)amino)-5-ethyl-11-methyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(B3)

Brown solid; yield 25%; m.p. 162–163 °C; 1H NMR (400 MHz, CDCl3) δ 8.25 (d, J = 8.7 Hz, 1H), 8.19 (s, 1H), 7.80 (dd, J = 7.8, 1.7 Hz, 1H), 7.50 (s, 1H), 7.40 (t, J = 7.9 Hz, 1H), 7.13 (t, J = 7.7 Hz, 1H), 7.07 (d, J = 8.4 Hz, 1H), 6.62–6.57 (m, 2H), 4.12 (q, J = 6.9 Hz, 2H), 3.99 (q, J = 6.9 Hz, 2H), 3.87–3.81 (m, 1H), 3.52–3.46 (m, 2H), 3.41 (s, 3H), 2.93–2.84 (m, 2H), 2.06–2.01 (m, 2H), 1.78–1.70 (m, 2H), 1.47 (t, J = 7.0 Hz, 3H), 1.30 (t, J = 6.9 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 168.26, 164.80, 156.05, 151.89, 149.62, 148.08, 147.15, 132.02, 131.89, 127.29, 123.72, 122.30, 119.27, 119.05, 117.27, 108.59, 102.12, 67.73, 64.20, 48.52, 46.19, 35.79, 34.37, 14.99, 13.87. HRMS-ESI (m/z): [M + H]+ calcd for C27H32N6O3: 489.2609, found: 489.2603.

General procedure for synthesis of compounds C1 and C2

The compounds C1 and C2 are synthesised by a similar method using the above-described procedure for the reaction of compound A1.

2-((4-(4-Hydroxypiperidin-1-yl)cyclohexyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(C1)

Yellow solid; yield 45%; m.p. 203–204 °C; 1H NMR (400 MHz, CDCl3) δ 7.99 (s, 1H), 7.82 (dd, J = 7.8, 1.6 Hz, 1H), 7.40 (td, J = 7.9, 1.7 Hz, 1H), 7.11 (t, J = 7.5 Hz, 1H), 7.04 (d, J = 8.3 Hz, 1H), 4.08–4.03 (m, 1H), 3.82–3.62 (m, 3H), 3.45 (s, 3H), 3.29 (s, 3H), 2.96–2.87 (m, 2H), 2.46–2.37 (m, 2H), 2.00–1.93 (m, 4H), 1.78–1.73 (m, 2H), 1.64–1.58 (m, 4H). 13C NMR (101 MHz, CDCl3) δ 168.40, 164.19, 158.13, 151.46, 149.54, 132.20, 132.17, 126.59, 123.53, 117.20, 67.28, 62.30, 46.54, 46.22, 38.22, 35.42, 34.19, 29.18, 23.62. HRMS-ESI (m/z): [M + H]+ calcd for C24H32N6O2: 437.2660, found: 437.2657.

2-((6-(4-Hydroxypiperidin-1-yl)pyridin-3-yl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(C2)

Yellow solid; yield 62%; m.p. 189–190 °C; 1H NMR (400 MHz, Methanol-d4) δ 8.55 (d, J = 2.3 Hz, 1H), 8.32 (s, 1H), 8.18 (dd, J = 9.9, 2.4 Hz, 1H), 7.83 (dd, J = 7.9, 1.5 Hz, 1H), 7.65–7.57 (m, 1H), 7.54 (d, J = 9.8 Hz, 1H), 7.34 (d, J = 8.3 Hz, 1H), 7.30 (t, J = 7.5 Hz, 1H), 4.09–4.01 (m, 3H), 3.68–3.60 (m, 2H), 3.56 (s, 3H), 3.52 (s, 3H), 2.12–2.04 (m, 2H), 1.79–1.68 (m, 2H). 13C NMR (101 MHz, CD3OD) δ 167.73, 166.34, 150.90, 149.47, 146.33, 142.22, 139.36, 133.09, 131.78, 127.68, 125.59, 125.04, 124.80, 121.19, 118.90, 113.44, 64.77, 43.93, 38.04, 36.46, 32.72. HRMS-ESI (m/z): [M + H]+ calcd for C23H25N7O2: 432.2142, found: 432.2137.

Preparation of intermediate 14

A sealed tube with a mixture of 13 (1 eq), 2-Methoxy-4-nitroaniline (1 eq), Pd(OAc)2 (0.05 eq), XPhos (0.1 eq),and Cs2CO3 (2 eq) in t-BuOH was heated at 110 °C for 10 h. The reaction was then filtered through celite and eluted with dichloromethane. The dichloromethane was removed in vacuo and the resulting crude product was purified by silica gel column chromatography (eluant: DCM/MeOH, 40:1 v/v) to afford the title intermediate 14 (36%).

2-((2-Methoxy-4-nitrophenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(14)

Yellow solid; yield 36%; m.p. 215–216 °C; 1H NMR (400 MHz, CDCl3) δ 8.71 (d, J = 9.1 Hz, 1H), 8.23 (s, 1H), 8.01 (dd, J = 9.0, 2.3 Hz, 2H), 7.88 (dd, J = 7.8, 1.8 Hz, 1H), 7.78 (d, J = 2.5 Hz, 1H), 7.51–7.44 (m, 1H), 7.18 (td, J = 7.5, 1.0 Hz, 1H), 7.13 (dd, J = 8.4, 1.0 Hz, 1H), 4.05 (s, 3H), 3.54 (s, 3H), 3.48 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 168.07, 163.65, 154.71, 151.14, 149.03, 146.73, 141.17, 135.67, 132.60, 132.44, 126.37, 124.01, 123.04, 118.07, 117.50, 115.56, 105.09, 56.33, 38.30, 36.07. HRMS-ESI (m/z): [M + H]+ calcd for C20H18N6O4: 407.1462, found: 407.1461.

Preparation of intermediate 15

A mixture of 14 (1 eq), Pd/C (0.05 eq), and methanol was stirred at room temperature for 12 h under a hydrogen atmosphere. The reaction mixture was filtered and the residue obtained by evaporation of the solvent of the filtrate under reduced pressure. This residue is prepared as the hydrochloride salt to afford the intermediate 15 (100%).

2-((4-Amino-2-methoxyphenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(15)

Brown solid; yield 100%; m.p. 199–200 °C; 1H NMR (400 MHz, Methanol-d4) δ 8.28 (s, 1H), 8.14–8.07 (m, 1H), 7.85 (dd, J = 7.8, 1.6 Hz, 1H), 7.66–7.60 (m, 1H), 7.37 (d, J = 8.3 Hz, 1H), 7.33 (t, J = 7.7 Hz, 1H), 7.21 (s, 1H), 7.16 (dd, J = 8.5, 2.3 Hz, 1H), 4.02 (s, 3H), 3.58 (s, 3H), 3.51 (s, 3H). 13C NMR (101 MHz, CD3OD) δ 167.49, 167.19, 152.44, 149.74, 145.43, 138.96, 133.23, 131.85, 128.83, 125.51, 125.43, 125.08, 120.65, 119.19, 114.98, 106.56, 55.73, 38.19, 36.51. HRMS-ESI (m/z): [M + H]+ calcd for C20H20N6O2: 377.1721, found: 377.1715.

General procedure for synthesis of compounds D1 and D2

To a solution of 18 (1 eq) and triethylamine (2 eq) in DCM ethyl 3-chloro-3-oxopropanoate/ethyl 4-chloro-4-oxobutanoate (1.1 eq) in DCM was added dropwise. The reaction mixture was stirred at RT for 1.5 h. After the reaction was complete as monitored by TLC, water was added to the solution. The organic phase is washed with saturated NaHCO3 and dried over anhydrous Na2SO4. After filtration, the combined organic solution is concentrated to dryness under reduced pressure. The crude product was dissolved in THF and treated with 2 N LiOH. The reaction was stirred for 1 h. The solution was added with 1 N HCl to make the pH = 6. This mixture was then washed with EtOAc (three times), and the combined extracts were washed with brine, dried with Na2SO4, and evaporated under reduced pressure to afford the crude product. The resulting crude product was purified by silica gel column chromatography (eluant: DCM/MeOH, 40:1 v/v) to afford the compound D1, D2 (70%).

3-((4-((5,11-Dimethyl-6-oxo-6,11-dihydro-5H-benzo[e]pyrimido[5,4-b][1,4]diazepin-2-yl)amino)-3-methoxyphenyl)amino)-3-oxopropanoic acid(D1)

White solid; yield 70%; m.p. 187–188 °C; 1H NMR (400 MHz, DMSO-d6) δ 12.59 (br, 1H), 10.11 (s, 1H), 8.35 (s, 1H), 8.05 (s, 1H), 7.94 (d, J = 8.7 Hz, 1H), 7.68 (dd, J = 7.7, 1.7 Hz, 1H), 7.52–7.45 (m, 1H), 7.40 (d, J = 2.2 Hz, 1H), 7.23 (d, J = 8.4, 1.0 Hz, 1H), 7.19–7.12 (m, 2H), 3.81 (s, 3H), 3.39 (s, 3H), 3.35 (s, 2H), 3.29 (s, 3H). 13C NMR (101 MHz, DMSO) δ 169.75, 167.52, 164.73, 163.50, 152.76, 150.03, 149.70, 135.15, 132.90, 132.11, 126.65, 124.43, 123.89, 121.50, 120.89, 118.21, 111.17, 103.05, 56.05, 44.45, 37.90, 35.95. HRMS-ESI (m/z): [M + H]+ calcd for C23H22N6O5: 463.1724, found: 463.1726.

4-((4-((5,11-Dimethyl-6-oxo-6,11-dihydro-5H-benzo[e]pyrimido[5,4-b][1,4]diazepin-2-yl)amino)-3-methoxyphenyl)amino)-4-oxobutanoic acid(D2)

White solid; yield 70%; m.p. 237–238 °C; 1H NMR (400 MHz, DMSO-d6) δ 12.12 (s, 1H), 9.94 (s, 1H), 8.34 (s, 1H), 8.03 (s, 1H), 7.90 (d, J = 8.6 Hz, 1H), 7.68 (dd, J = 7.8, 1.7 Hz, 1H), 7.53–7.45 (m, 1H), 7.45 (d, J = 2.2 Hz, 1H), 7.23 (dd, J = 8.4, 1.0 Hz, 1H), 7.19–7.14 (m, 1H), 7.12 (dd, J = 8.7, 2.2 Hz, 1H), 3.80 (s, 3H), 3.39 (s, 3H), 3.28 (s, 3H), 2.60–2.52 (m, 4H). 13C NMR (101 MHz, DMSO) δ 174.30, 170.25, 167.52, 163.50, 156.53, 152.76, 150.08, 149.71, 135.63, 132.90, 132.10, 126.66, 123.95, 123.89, 121.58, 120.82, 118.20, 110.96, 102.97, 56.01, 37.89, 35.93, 31.48, 29.28. HRMS-ESI (m/z): [M + H]+ calcd for C24H24N6O5: 477.1881, found: 477.1875.

Preparation of intermediate 16

The intermediate 16 are synthesised by a similar method using the above-described procedure for the reaction of compounds D1 and D2.

6-Bromo-N-(4-((5,11-dimethyl-6-oxo-6,11-dihydro-5H-benzo[e]pyrimido[5,4-b][1,4]diazepin-2-yl)amino)-3-methoxyphenyl)hexanamide(16)

White solid; yield 87%; m.p. 137–138 °C; 1H NMR (400 MHz, CDCl3) δ 8.30 (d, J = 8.4 Hz, 2H), 8.11 (s, 1H), 7.82 (d, J = 7.8 Hz, 1H), 7.59 (s, 1H), 7.51 (s, 1H), 7.40 (t, J = 7.8 Hz, 1H), 7.10 (t, J = 7.5 Hz, 1H), 7.05 (d, J = 8.3 Hz, 1H), 6.97 (d, J = 8.7 Hz, 1H), 3.84 (s, 3H), 3.47 (s, 3H), 3.40–3.34 (m, 5H), 2.35 (t, J = 7.5 Hz, 2H), 1.90–1.80 (m, 2H), 1.73 (m, 2H), 1.52–1.43 (m, 2H). 13C NMR (101 MHz, CDCl3) δ 171.37, 168.45, 163.71, 155.85, 151.39, 149.43, 147.94, 132.95, 132.46, 132.17, 126.41, 125.29, 123.69, 121.11, 118.12, 117.44, 111.71, 103.32, 55.78, 38.28, 37.14, 35.87, 33.68, 32.46, 27.77, 24.71. HRMS-ESI (m/z): [M + H]+ calcd for C26H29BrN6O3: 553.1557, found: 553.1553.

General procedure for the synthesis of compound D8

A mixture of 16 (1 eq) and potassium ethanethioate (6 eq) in DMF was heated to 100 °C for 6 h. After the reaction was complete as monitored by TLC, the solvent was diluted with water (20 ml) and extracted with EtOAc (20 ml) × 3. The organics were combined, dried over Na2SO4, filtered, and concentrated in vacuo. Purification by silica gel column chromatography (eluant: DCM/MeOH, 50:1 v/v) gave compound D8 (72%).

S-(6-((4-((5,11-dimethyl-6-oxo-6,11-dihydro-5H-benzo[e]pyrimido[5,4-b][1,4]diazepin-2-yl)amino)-3-methoxyphenyl)amino)-6-oxohexyl) ethanethioate(D8)

Brown solid; yield 72%; m.p. 159–160 °C; 1H NMR (400 MHz, CDCl3) δ 8.35 (d, J = 8.7 Hz, 1H), 8.14 (s, 1H), 7.86 (dd, J = 7.8, 1.7 Hz, 1H), 7.60 (s, 1H), 7.56 (d, J = 2.3 Hz, 1H), 7.46–7.40 (m, 1H), 7.38 (s, 1H), 7.15 (t, J = 7.5 Hz, 1H), 7.09 (d, J = 8.3 Hz, 1H), 6.87 (dd, J = 8.7, 2.3 Hz, 1H), 4.10 (t, J = 6.7 Hz, 2H), 3.92 (s, 3H), 3.51 (s, 3H), 3.42 (s, 3H), 2.39 (t, J = 7.4 Hz, 2H), 2.06 (s, 3H), 1.84–1.76 (m, 2H), 1.75–1.66 (m, 2H), 1.53–1.40 (m, 2H). 13C NMR (101 MHz, CDCl3) δ 171.23, 170.91, 168.34, 163.75, 155.82, 151.35, 149.40, 148.01, 132.53, 132.37, 132.26, 126.53, 125.51, 123.71, 121.26, 118.08, 117.38, 111.36, 103.18, 64.26, 55.84, 38.25, 37.46, 35.88, 28.42, 25.61, 25.14, 21.02. HRMS-ESI (m/z): [M + H]+ calcd for C28H32N6O4S: 549.2279, found: 549.2286.

Preparation of intermediate 18

A mixture of 17 (1.5 eq), 5-fluoro-2-nitrophenyl ethyl ether (1 eq), and K2CO3 in DMF was heated to 90 °C for 8–10 h. After the reaction was complete as monitored by TLC, the solution was diluted with water (50 ml) and extracted with EtOAc (50 ml) × 3. The organics were combined, dried over Na2SO4, filtered, and concentrated in vacuo. Purification by silica gel column chromatography (eluant: petroleum ether/EtOAc, 5:1 v/v) gave intermediate 18 (62–91%).

4-(3-Ethoxy-4-nitrophenyl)thiomorpholine (18-4)

Yellow solid; yield 90%; m.p. 118–119 °C; 1H NMR (400 MHz, CDCl3) δ 7.98 (d, J = 9.3 Hz, 1H), 6.38 (dd, J = 9.4, 2.5 Hz, 1H), 6.29 (d, J = 2.5 Hz, 1H), 4.15 (q, J = 6.9 Hz, 2H), 3.85–3.78 (m, 4H), 2.76–2.68 (m, 4H), 1.51 (t, J = 6.9 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 155.84, 154.33, 129.86, 128.89, 105.77, 98.80, 65.35, 50.44, 25.93, 14.65. HRMS-ESI (m/z): [M + H]+ calcd for C12H16N2O3S: 269.0954, found: 269.0953.

Preparation of intermediate 19

The intermediate 19 are synthesised by similar method using the above-described procedure for the reaction of intermediate 15.

1-(4-Amino-3-ethoxyphenyl)piperidine-4-carboxylic acid (19-3)

White solid; yield 100%; m.p. 146–147 °C; 1H NMR (400 MHz, DMSO-d6) δ 7.64 (d, J = 14.6 Hz, 1H), 7.51–7.46 (m, 1H), 7.41–7.33 (m, 1H), 4.18 (q, J = 7.0 Hz, 2H), 3.66 (s, 2H), 3.56 (s, 4H), 2.24–2.08 (m, 4H), 1.38 (t, J = 6.9 Hz, 3H). 13C NMR (101 MHz, DMSO) δ 175.25, 174.12, 151.96, 123.95, 113.07, 109.98, 106.59, 65.40, 52.27, 37.99, 26.13, 14.79. HRMS-ESI (m/z): [M + H]+ calcd for C14H20N2O3: 265.1547, found: 265.1544.

General procedure for synthesis of compounds D3, D4, D6, D9–D16

The compounds D3, D4, D6, D9D16 are synthesised by a similar method using the above-described procedure for the reaction of compound A1.

1-(4-((5,11-Dimethyl-6-oxo-6,11-dihydro-5H-benzo[e]pyrimido[5,4-b][1,4]diazepin-2-yl)amino)-3-ethoxyphenyl)piperidine-4-carboxylic acid(D3)

White solid; yield 48%; m.p. 159–160 °C; 1H NMR (400 MHz, Methanol-d4) δ 8.38 (d, J = 8.8 Hz, 1H), 8.32 (s, 1H), 7.80 (dd, J = 7.8, 1.7 Hz, 1H), 7.63–7.57 (m, 1H), 7.46 (s, 1H), 7.41 (dd, J = 8.8, 2.5 Hz, 1H), 7.32 (d, J = 8.5 Hz, 1H), 7.28 (t, J = 7.5 Hz, 1H), 4.30 (q, J = 6.9 Hz, 2H), 3.88–3.72 (m, 5H), 3.53 (s, 3H), 3.50 (s, 3H), 2.45–2.38 (m, 2H), 2.33–2.26 (m, 2H), 1.51 (t, J = 7.0 Hz, 3H). 13C NMR (101 MHz, CD3OD) δ 175.52, 174.12, 168.40, 165.84, 152.07, 149.96, 147.16, 144.57, 137.80, 133.26, 131.70, 128.29, 125.47, 124.65, 121.05, 118.59, 112.76, 104.89, 65.34, 55.50, 55.38, 38.07, 37.14, 36.06, 25.84, 25.74, 13.61. HRMS-ESI (m/z): [M + H]+ calcd for C27H30N6O4: 503.2401, found: 503.2397.

2-((2-Ethoxy-4-thiomorpholinophenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(D4)

Yellow solid; yield 38%; m.p. 190–191 °C; 1H NMR (400 MHz, CDCl3) δ 8.28 (d, J = 8.8 Hz, 1H), 8.13 (s, 1H), 7.86 (dd, J = 7.8, 1.7 Hz, 1H), 7.50 (s, 1H), 7.46–7.40 (m, 1H), 7.15 (t, J = 7.5 Hz, 1H), 7.10 (d, J = 8.3 Hz, 1H), 6.58 (dd, J = 8.8, 2.6 Hz, 1H), 6.54 (d, J = 2.5 Hz, 1H), 4.12 (q, J = 7.0 Hz, 2H), 3.51 (s, 3H), 3.49–3.44 (m, 4H), 3.43 (s, 3H), 2.85–2.77 (m, 4H), 1.49 (t, J = 7.0 Hz, 3H). 13 C NMR (101 MHz, CDCl3) δ 168.35, 163.77, 156.01, 151.47, 149.48, 148.15, 147.49, 132.29, 132.25, 126.60, 123.66, 122.97, 120.86, 119.06, 117.36, 109.55, 102.99, 64.29, 53.41, 38.23, 35.90, 27.45, 14.96. HRMS-ESI (m/z): [M + H]+ calcd for C25H28N6O2S: 477.2067, found: 477.2065.

2-((4-(1,1-Dioxidothiomorpholino)-2-ethoxyphenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(D6)

White solid; yield 32%; m.p. 157–158 °C; 1H NMR (400 MHz, Methanol-d4) δ 8.12 (s, 1H), 7.83 (dd, J = 7.8, 1.7 Hz, 1H), 7.69–7.62 (m, 1H), 7.52 (s, 1H), 7.40–7.31 (m, 2H), 6.80–6.70 (m, 2H), 4.16 (q, J = 7.0 Hz, 2H), 3.99–3.89 (m, 4H), 3.55 (s, 3H), 3.49 (s, 3H), 3.25–3.20 (m, 4H), 1.35 (t, J = 6.9 Hz, 3H). 13C NMR (101 MHz, CD3OD) δ 168.01, 167.57, 145.24, 133.56, 131.79, 125.25, 125.07, 119.95, 119.23, 107.95, 101.15, 64.33, 49.99, 47.17, 38.65, 36.48, 13.81. HRMS-ESI (m/z): [M + H]+ calcd for C25H28N6O4S: 509.1966, found: 509.1962.

2-((2-Ethoxy-4-(4-hydroxy-4-methylpiperidin-1-yl)phenyl)amino)-5-ethyl-11-methyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(D9)

White solid; yield 30%; m.p. 178–179 °C; 1H NMR (400 MHz, CDCl3) δ 8.25 (d, J = 8.7 Hz, 1H), 8.18 (s, 1H), 7.80 (d, J = 7.8 Hz, 1H), 7.50 (s, 1H), 7.40 (t, J = 7.8 Hz, 1H), 7.12 (t, J = 7.5 Hz, 1H), 7.06 (d, J = 8.3 Hz, 1H), 6.66–6.56 (m, 2H), 4.26–3.71 (m, 4H), 3.40 (s, 3H), 3.32–3.22 (m, 2H), 3.21–3.09 (m, 2H), 1.85–1.70 (m, 4H), 1.46 (t, J = 7.0 Hz, 3H), 1.38–1.22 (m, 7H). 13C NMR (101 MHz, CDCl3) δ 168.24, 164.80, 156.10, 151.89, 149.63, 148.14, 147.36, 132.00, 131.89, 127.30, 123.69, 122.17, 119.23, 119.17, 117.26, 108.66, 102.06, 77.45, 77.13, 76.81, 67.65, 64.23, 47.16, 46.16, 38.59, 35.76, 29.87, 14.98, 13.85. HRMS-ESI (m/z): [M + H]+ calcd for C28H34N6O3: 503.2771, found: 503.2767.

2-((2-Ethoxy-4-(4-fluoro-4-methylpiperidin-1-yl)phenyl)amino)-5-ethyl-11-methyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(D10)

White solid; yield 33%; m.p. 189–190 °C; 1H NMR (400 MHz, CDCl3) δ 8.28 (d, J = 8.7 Hz, 1H), 8.20 (s, 1H), 7.82 (d, J = 7.7 Hz, 1H), 7.51 (s, 1H), 7.40 (t, J = 7.8 Hz, 1H), 7.14 (t, J = 7.6 Hz, 1H), 7.07 (d, J = 8.3 Hz, 1H), 6.67–6.57 (m, 2H), 4.21–3.75 (m, 4H), 3.47–3.33 (m, 5H), 3.11 (t, J = 11.9 Hz, 2H), 2.01–1.77 (m, 4H), 1.53–1.37 (m, 6H), 1.31 (t, J = 7.1 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 168.20, 164.78, 156.09, 151.90, 149.63, 148.14, 147.10, 131.98, 131.90, 127.35, 123.69, 122.44, 119.34, 119.15, 117.25, 108.83, 102.18, 92.92, 91.25, 77.41, 77.09, 76.78, 64.24, 46.97, 46.94, 46.14, 36.45, 36.24, 35.76, 26.88, 14.97, 13.85. HRMS-ESI (m/z): [M + H]+ calcd for C28H33FN6O2: 505.2702, found: 505.2719.

2-((2-Methoxy-4-(piperidin-1-yl)phenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(D11)

White solid; yield 45%; m.p. 166–167 °C; 1H NMR (400 MHz, CDCl3) δ 8.22 (d, J = 9.3 Hz, 1H), 8.13 (s, 1H), 7.86 (dd, J = 7.8, 1.7 Hz, 1H), 7.48–7.38 (m, 2H), 7.19–7.05 (m, 2H), 6.60 (d, J = 7.4 Hz, 2H), 3.90 (s, 3H), 3.51 (s, 3H), 3.42 (s, 3H), 3.17–3.08 (m, 4H), 1.81–1.72 (m, 4H), 1.65–1.55 (m, 2H). 13C NMR (101 MHz, CDCl3) δ 168.36, 163.77, 156.19, 151.47, 149.52, 148.92, 148.34, 132.23, 126.64, 123.60, 121.95, 120.76, 119.32, 117.33, 108.56, 101.21, 77.33, 77.02, 76.70, 55.65, 51.83, 38.19, 35.84, 26.06, 24.28. HRMS-ESI (m/z): [M + H]+ calcd for C25H28N6O2: 445.2352, found: 445.2346.

2-((2-Methoxy-4-(4-methoxypiperidin-1-yl)phenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(D12)

White solid; yield 47%; m.p. 179–180 °C; 1H NMR (400 MHz, CDCl3) δ 8.22 (d, J = 8.5 Hz, 1H), 8.12 (s, 1H), 7.86 (dd, J = 7.8, 1.8 Hz, 1H), 7.47–7.38 (m, 2H), 7.18–7.05 (m, 2H), 6.64–6.56 (m, 2H), 3.90 (s, 3H), 3.54–3.44 (m, 5H), 3.43–3.34 (m, 7H), 2.97–2.86 (m, 2H), 2.12–2.00 (m, 2H), 1.81–1.70 (m, 2H). 13C NMR (101 MHz, CDCl3) δ 168.34, 163.76, 156.15, 151.46, 149.50, 148.91, 147.42, 132.25, 132.23, 126.62, 123.60, 122.17, 120.81, 119.32, 117.33, 108.64, 101.19, 77.36, 77.04, 76.73, 76.06, 55.67, 55.59, 48.40, 38.19, 35.84, 30.84. HRMS-ESI (m/z): [M + H]+ calcd for C26H30N6O3: 475.2458, found: 475.2450.

2-((4-(4-Fluoropiperidin-1-yl)-2-methoxyphenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(D13)

White solid; yield 33%; m.p. 160–161 °C; 1H NMR (400 MHz, CDCl3) δ 8.17 (s, 1H), 8.15 (s, 1H), 7.87 (dd, J = 7.8, 1.7 Hz, 1H), 7.49–7.39 (m, 2H), 7.20–7.06 (m, 2H), 6.56 (s, 1H), 5.86–5.71 (m, 1H), 5.08–4.97 (m, 2H), 4.40 (s, 2H), 3.90 (s, 3H), 3.52 (s, 3H), 3.45 (s, 3H), 3.24 (t, J = 7.4 Hz, 2H), 2.29 (q, J = 7.2 Hz, 2H). 13C NMR (101 MHz, CDCl3) δ 168.34, 163.76, 156.20, 151.56, 149.47, 148.14, 136.49, 132.27, 132.26, 126.66, 123.67, 122.44, 120.71, 120.55, 117.27, 115.93, 101.15, 77.35, 77.03, 76.71, 55.76, 51.76, 38.22, 35.85, 32.64. HRMS-ESI (m/z): [M + H]+ calcd for C25H27FN6O2: 463.2252, found: 463.2261.

2-((4-(4,4-Difluoropiperidin-1-yl)-2-methoxyphenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(D14)

White solid; yield 32%; m.p. 171–172 °C; 1H NMR (400 MHz, CDCl3) δ 8.27 (d, J = 8.5 Hz, 1H), 8.14 (s, 1H), 7.87 (d, J = 7.5 Hz, 1H), 7.51–7.39 (m, 2H), 7.20–7.06 (m, 2H), 6.67–6.56 (m, 2H), 3.91 (s, 3H), 3.55–3.36 (m, 6H), 3.35–3.23 (m, 4H), 2.23–2.05 (m, 4H). 13C NMR (101 MHz, CDCl3) δ 168.30, 163.77, 156.07, 151.44, 149.46, 148.93, 146.07, 132.27, 126.62, 123.65, 122.96, 121.01, 119.26, 117.34, 109.13, 101.51, 77.37, 77.05, 76.74, 55.73, 47.99, 47.94, 47.89, 38.19, 35.85, 34.12, 33.89, 33.67. HRMS-ESI (m/z): [M + H]+ calcd for C25H26F2N6O2: 481.2164, found: 481.2155.

2-((4-(4-Hydroxy-4-methylpiperidin-1-yl)-2-methoxyphenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(D15)

White solid; yield 37%; m.p. 162–163 °C; 1H NMR (400 MHz, CDCl3) δ 8.22 (d, J = 8.6 Hz, 1H), 8.12 (s, 1H), 7.89–7.82 (m, 1H), 7.47–7.38 (m, 2H), 7.19–7.05 (m, 2H), 6.67–6.58 (m, 2H), 3.90 (s, 3H), 3.50 (s, 3H), 3.42 (s, 3H), 3.35–3.25 (m, 2H), 3.23–3.11 (m, 2H), 1.91–1.80 (m, 2H), 1.76–1.71 (m, 2H), 1.33 (s, 3H). 13C NMR (101 MHz, CDCl3) δ 168.40, 163.80, 156.16, 151.46, 149.51, 148.95, 147.48, 132.28, 132.23, 126.59, 123.63, 122.12, 120.79, 119.38, 117.36, 108.72, 101.15, 77.35, 77.03, 76.72, 67.82, 55.69, 47.18, 38.66, 38.23, 35.85, 29.88. HRMS-ESI (m/z): [M + H]+ calcd for C26H30N6O3: 475.2458, found: 475.2451.

2-((4-(4,4-Dimethylpiperidin-1-yl)-2-methoxyphenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(D16)

White solid; yield 40%; m.p. 181–182 °C; 1H NMR (400 MHz, CDCl3) δ 8.22 (d, J = 8.5 Hz, 1H), 8.12 (s, 1H), 7.90–7.82 (m, 1H), 7.47–7.39 (m, 2H), 7.18–7.04 (m, 2H), 6.66–6.57 (m, 2H), 3.90 (s, 3H), 3.50 (s, 3H), 3.41 (s, 3H), 3.19–3.11 (m, 4H), 1.61–1.53 (m, 4H), 1.02 (s, 6H). 13C NMR (101 MHz, CDCl3) δ 168.35, 163.76, 156.20, 151.48, 149.52, 148.96, 147.94, 132.25, 132.22, 126.63, 123.59, 121.75, 120.73, 119.40, 117.33, 108.22, 100.77, 77.39, 77.07, 76.75, 55.65, 47.03, 38.67, 38.19, 35.84, 28.43, 27.94. HRMS-ESI (m/z): [M + H]+ calcd for C27H32N6O2: 473.2665, found: 473.2658.

General procedure for the synthesis of compound D5

D4 (1 eq) was dissolved in acetic acid, and hydrogen peroxide (1 eq) was added dropwise. After the reaction was complete as monitored by TLC, the residue was obtained by evaporation of solvent under reduced pressure. Purification by silica gel column chromatography (eluant: DCM/MeOH, 60:1 v/v) gave compound D5 (95%).

2-((2-ethoxy-4-(1-Oxidothiomorpholino)phenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(D5)

White solid; yield 95%; m.p. 163–164 °C; 1H NMR (400 MHz, Methanol-d4) δ 8.19 (s, 1H), 7.83 (dd, J = 7.8, 1.7 Hz, 1H), 7.76 (s, 1H), 7.68–7.61 (m, 1H), 7.40–7.29 (m, 2H), 7.08–6.94 (m, 2H), 4.22 (q, J = 7.0 Hz, 2H), 4.16–4.05 (m, 2H), 3.83–3.76 (m, 2H), 3.56 (s, 3H), 3.50 (s, 3H), 3.31–3.24 (m, 2H), 3.14–3.04 (m, 2H), 1.40 (t, J = 6.9 Hz, 3H). 13C NMR (101 MHz, CD3OD) δ 167.89, 167.31, 152.63, 150.25, 145.43, 138.51, 133.48, 131.80, 125.17, 120.20, 119.19, 109.55, 102.47, 64.61, 43.36, 42.38, 38.53, 36.51, 13.75. HRMS-ESI (m/z): [M + H]+ calcd for C25H28N6O3S: 493.2016, found: 493.2011.

Preparation of intermediate 20

The intermediate 20 are synthesised by a similar method using the above-described procedure for the reaction of compound A1.

Tert-butyl 4-(4-((5,11-dimethyl-6-oxo-6,11-dihydro-5H-benzo[e]pyrimido[5,4-b][1,4] diazepin-2-yl)amino)-3-ethoxyphenyl)piperazine-1-carboxylate(20)

White solid; yield 29%; m.p. 237–238 °C; 1H NMR (400 MHz, CDCl3) δ 8.29 (d, J = 8.5 Hz, 1H), 8.13 (s, 1H), 7.86 (dd, J = 7.8, 1.7 Hz, 1H), 7.49 (s, 1H), 7.46–7.40 (m, 1H), 7.14 (t, J = 7.5 Hz, 1H), 7.10 (d, J = 8.4 Hz, 1H), 6.63–6.54 (m, 2H), 4.12 (q, J = 6.9 Hz, 2H), 3.61 (t, J = 5.1 Hz, 4H), 3.51 (s, 3H), 3.43 (s, 3H), 3.09 (t, J = 5.1 Hz, 4H), 1.52–1.46 (m, 12H). 13C NMR (101 MHz, CDCl3) δ 168.32, 163.75, 156.01, 154.72, 151.46, 149.48, 148.10, 146.92, 132.27, 132.25, 126.61, 123.65, 123.06, 120.87, 119.02, 117.34, 108.68, 102.15, 79.89, 64.27, 50.59, 38.21, 35.89, 28.45, 14.95. HRMS-ESI (m/z): [M + H]+ calcd for C30H37N7O4: 560.2980, found: 560.2978.

Preparation of intermediate 21

A mixture of 21 (1 eq), trifluoroacetic acid (10 eq), and DCM was stirred at room temperature for 1.5 h. After the reaction was complete as monitored by TLC, the residue was obtained by evaporation of solvent under reduced pressure. This residue is prepared as the hydrochloride salt to afford the title intermediate 21 (100%).

2-((2-Ethoxy-4-(piperazin-1-yl)phenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(21)

Yellow solid; yield 100%; m.p. 185–186 °C; 1H NMR (400 MHz, Methanol-d4) δ 8.15 (s, 1H), 7.85 (dd, J = 7.8, 1.7 Hz, 1H), 7.67–7.56 (m, 1.7 Hz, 2H), 7.38 (d, J = 8.0 Hz, 1H), 7.34 (t, J = 7.6 Hz, 1H), 6.92 (d, J = 2.5 Hz, 1H), 6.84 (dd, J = 8.7, 2.5 Hz, 1H), 4.20 (q, J = 7.0 Hz, 2H), 3.64–3.59 (m, 4H), 3.57 (s, 3H), 3.50 (s, 3H), 3.49–4.46 (m, 4H), 1.38 (t, J = 7.0 Hz, 3H). 13C NMR (101 MHz, CD3OD) δ 167.59, 167.46, 145.13, 137.19, 133.26, 131.81, 125.34, 125.09, 120.16, 119.20, 108.85, 102.33, 64.31, 46.87, 43.07, 38.30, 36.44, 13.68. HRMS-ESI (m/z): [M + H]+ calcd for C25H29N7O2: 460.2455, found: 460.2449.

Preparation of compound D7

A mixture of 21 (1 eq), glucose (1.5 eq), and acetic acid (1 eq) in ethanol was heated to reflux for 8 h. After the reaction was complete as monitored by TLC, the residue was obtained by evaporation of solvent under reduced pressure. Purification by silica gel column chromatography (eluant: DCM/MeOH, 30:1 v/v) gave compound D7 (82%).

2-((2-Ethoxy-4-(4-(((2R,3S,4R,5R)-2,3,4,5-tetrahydroxytetrahydro-2H-pyran-2-yl)methyl)piperazin-1-yl)phenyl)amino)-5,11-dimethyl-5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one(D7)

Yellow solid; yield 82%; m.p. 126–127 °C; 1H NMR (400 MHz, CDCl3) δ 8.25 (d, J = 8.5 Hz, 1H), 8.12 (s, 1H), 7.85 (dd, J = 7.8, 1.7 Hz, 1H), 7.54 (s, 1H), 7.46–7.40 (m, 1H), 7.14 (t, J = 7.3 Hz, 1H), 7.09 (d, J = 8.3 Hz, 1H), 6.59–5.64 (m, 2H), 4.11 (q, J = 7.0 Hz, 2H), 4.03 (d, J = 12.4 Hz, 2H), 3.92–3.69 (m, 7H), 3.57 (d, J = 9.4 Hz, 1H), 3.50 (s, 3H), 3.42 (s, 3H), 3.19–3.10 (m, 5H), 2.97 (d, J = 13.3 Hz, 1H), 2.76–2.66 (m, 2H), 2.08 (s, 1H), 1.47 (t, J = 7.0 Hz, 3H). 13C NMR (101 MHz, CDCl3) δ 168.42, 163.82, 155.99, 151.37, 149.45, 148.21, 146.76, 132.34, 132.24, 126.52, 123.69, 122.70, 120.76, 119.22, 117.38, 108.17, 101.68, 96.71, 71.56, 70.89, 69.27, 64.28, 62.79, 61.16, 54.32, 50.42, 38.29, 35.90, 14.95. HRMS-ESI (m/z): [M + H]+ calcd for C31H39N7O7: 622.2984, found: 622.2996.

Biological evaluation

In vitro kinase inhibition assay

Eurofins (Belgium) conducted in vitro DCLK1 and LRRK2 kinase activity assays. Compounds were sent as dry powders to Eurofins.

In vitro cytotoxicity assay

A 96-well plate (3599, Corning, USA) was inoculated with 100 μL of complete medium containing 10 000 cells, and the experimental, control, and blank groups were established. Three parallel wells were used per concentration. After 10 h, media from the experimental and control groups were replaced with a complete medium containing the indicated concentration of the compound. Cells were incubated for 72 h before 10 μL of CCK8 reagent (CA1210, Solarbio, China) was added to each well and incubated for 1 h. The absorbance at 450 nm was measured with a microplate reader (Tecan, Switzerland). The IC50 was determined using GraphPad Prism 8 and nonlinear regression curve fitting.

Molecular modelling

Docking results were carried out by Schrödinger Maestro and Pymol. All parameters follow the default values. The DCLK1 crystal structure was acquired from RCSB Protein Data Bank (http://www.rcsb.org, PDB ID: 5JZN). The compound was prepared with ChemDraw and Schrödinger Maestro.

Consent form

All authors agree with the submission of the final version for publication in the journal.

Supplemental material

Supplemental Material

Download PDF (6.3 MB)

Disclosure statement

The authors report no conflicts of interest.

Additional information

Funding

This work was supported by the National Natural Science Foundation of China (Grant Nos. 81872738 and 81772565) and the National Key R&D Program of China (2022YFF1203005 and 2022YFC2303700).

References

  • Dijkmans TF, van Hooijdonk LW, Fitzsimons CP, Vreugdenhil E. The doublecortin gene family and disorders of neuronal structure. Cent Nerv Syst Agents Med Chem. 2010;10(1):32–46.
  • Gzil A, Szylberg Ł, Jaworski D, Dominiak J, Zarębska I, Grzanka D. The essential role of DCLK1 in pathogenesis, diagnostic procedures and prognostic stratification of colorectal cancer. Anticancer Res. 2019;39(6):2689–2697.
  • Kalantari E, Razmi M, Tajik F, Asadi-Lari M, Ghods R, Madjd Z. Oncogenic functions and clinical significances of DCLK1 isoforms in colorectal cancer: a systematic review and meta-analysis. Cancer Cell Int. 2022;22(1):217.
  • Sureban SM, May R, Lightfoot SA, Hoskins AB, Lerner M, Brackett DJ, Postier RG, Ramanujam R, Mohammed A, Rao CV, et al. Dcamkl-1 regulates epithelial-mesenchymal transition in human pancreatic cells through a mir-200a-dependent mechanism. Cancer Res. 2011;71(6):2328–2338.
  • Ferguson FM, Nabet B, Raghavan S, Liu Y, Leggett AL, Kuljanin M, Kalekar RL, Yang A, He S, Wang J, et al. Discovery of a selective inhibitor of doublecortin like kinase 1. Nat Chem Biol. 2020;16(6):635–643.
  • Wu X, Qu D, Weygant N, Peng J, Houchen CW. Cancer stem cell marker DCLK1 correlates with tumorigenic immune infiltrates in the colon and gastric adenocarcinoma microenvironments. Cancers. 2020;12(2):274.
  • Deng H, Qianqian G, Ting J, Aimin Y. Mir-539 enhances chemosensitivity to cisplatin in non-small cell lung cancer by targeting DCLK1. Biomed Pharmacother. 2018;106:1072–1081.
  • Polyak K, Hahn WC. Roots and stems: stem cells in cancer. Nat Med. 2006;12(3):296–300.
  • Sarkar B, Dosch J, Simeone DM. Cancer stem cells: a new theory regarding a timeless disease. Chem Rev. 2009;109(7):3200–3208.
  • Khan IN, Al-Karim S, Bora RS, Chaudhary AG, Saini KS. Cancer stem cells: a challenging paradigm for designing targeted drug therapies. Drug Discov Today. 2015;20(10):1205–1216.
  • Park JH, Chung S, Matsuo Y, Nakamura Y. Development of small molecular compounds targeting cancer stem cells. Med Chem Commun. 2017;8(1):73–80.
  • Cao Z, Weygant N, Chandrakesan P, Houchen CW, Peng J, Qu D. Tuft and cancer stem cell marker DCLK1: a new target to enhance anti-tumor immunity in the tumor microenvironment. Cancers. 2020;12(12):3801.
  • Chhetri D, Vengadassalapathy S, Venkadassalapathy S, Balachandran V, Umapathy VR, Veeraraghavan VP, Jayaraman S, Patil S, Iyaswamy A, Palaniyandi K, et al. Pleiotropic effects of DCLK1 in cancer and cancer stem cells. Front Mol Biosci. 2022;9:965730.
  • Chandrakesan P, Yao J, Qu D, May R, Weygant N, Ge Y, Ali N, Sureban SM, Gude M, Vega K, et al. DCLK1, a tumor stem cell marker, regulates pro-survival signaling and self-renewal of intestinal tumor cells. Mol Cancer. 2017;16(1):30.
  • Ge Y, Weygant N, Qu D, May R, Berry WL, Yao J, Chandrakesan P, Zheng W, Zhao L, Zhao KL, et al. Alternative splice variants of DCLK1 mark cancer stem cells, promote self-renewal and drug-resistance, and can be targeted to inhibit tumorigenesis in kidney cancer. Int Journal of Cancer. 2018;143(5):1162–1175.
  • Weygant N, Qu D, May R, Tierney RM, Berry WL, Zhao L, Agarwal S, Chandrakesan P, Chinthalapally HR, Murphy NT, et al. DCLK1 is a broadly dysregulated target against epithelial-mesenchymal transition, focal adhesion, and stemness in clear cell renal carcinoma. Oncotarget. 2015;6(4):2193–2205.
  • Carli ALE, Afshar-Sterle S, Rai A, Fang H, O'Keefe R, Tse J, Ferguson FM, Gray NS, Ernst M, Greening DW, et al. Cancer stem cell marker DCLK1 reprograms small extracellular vesicles toward migratory phenotype in gastric cancer cells. Proteomics. 2021;21(13–14):e2000098.
  • Patel O, Dai W, Mentzel M, Griffin MD, Serindoux J, Gay Y, Fischer S, Sterle S, Kropp A, Burns CJ, et al. Biochemical and structural insights into doublecortin-like kinase domain 1. Structure. 2016;24(9):1550–1561.
  • Sureban SM, May R, Weygant N, Qu D, Chandrakesan P, Bannerman-Menson E, Ali N, Pantazis P, Westphalen CB, Wang TC, et al. Xmd8-92 inhibits pancreatic tumor xenograft growth via a DCLK1-dependent mechanism. Cancer Lett. 2014;351(1):151–161.
  • Deng X, Dzamko N, Prescott A, Davies P, Liu Q, Yang Q, Lee JD, Patricelli MP, Nomanbhoy TK, Alessi DR, et al. Characterization of a selective inhibitor of the Parkinson’s disease kinase LRRK2. Nat Chem Biol. 2011;7(4):203–205.
  • Ferguson FM, Ni J, Zhang T, Tesar B, Sim T, Kim ND, Deng X, Brown JR, Zhao JJ, Gray NS. Discovery of a series of 5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-ones as selective pi3k-delta/gamma inhibitors. ACS Med Chem Lett. 2016;7(10):908–912.
  • Selva M, Tundo P, Perosa A. Reaction of functionalized anilines with dimethyl carbonate over nay faujasite. 3. Chemoselectivity toward mono-n-methylation. J Org Chem. 2003;68(19):7374–7378.
  • Xia X, Zhai Y, Cui H, Zhang H, Hayat K, Zhang X, Ho CT. Glycine, diglycine, and triglycine exhibit different reactivities in the formation and degradation of amadori compounds. J Agric Food Chem. 2022;70(47):14907–14918.
  • Ferguson FM, Liu Y, Harshbarger W, Huang L, Wang J, Deng X, Capuzzi SJ, Muratov EN, Tropsha A, Muthuswamy S, et al. Synthesis and structure-activity relationships of DCLK1 kinase inhibitors based on a 5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one scaffold. J Med Chem. 2020;63(14):7817–7826.
  • Patel O, Roy MJ, Kropp A, Hardy JM, Dai W, Lucet IS. Structural basis for small molecule targeting of doublecortin like kinase 1 with DCLK1-in-1. Commun Biol. 2021;4(1):1105.
  • Bekes M, Langley DR, Crews CM. Protac targeted protein degraders: the past is prologue. Nat Rev Drug Discov. 2022;21(3):181–200.