EGFR dimer disruptors and uses thereof

文档序号:975788 发布日期:2020-11-03 浏览:34次 中文

阅读说明:本技术 Egfr二聚体干扰物及其用途 (EGFR dimer disruptors and uses thereof ) 是由 穆克什·K·尼亚蒂 西奥多·S·劳伦斯 克里斯托弗·怀特黑德 詹森·克里斯托弗·雷希 布雷南 于 2019-02-25 设计创作,主要内容包括:本文提供了调节EGFR的化合物以及使用所述化合物例如治疗癌症的方法。(Provided herein are compounds that modulate EGFR and methods of using the compounds, for example, to treat cancer.)

1.a compound having the structure of formula I:

Figure FDA0002688847210000011

wherein

X is O-C0-6Alkylene, S-C0-6Alkylene or NR3-C0-6Alkylene, and said alkylene is optionally substituted with 1 to 3 substituents independently selected from halo, N (R)3)2And OR3Substituted with a group of (1);

y is C0-6Alkylene, and said alkylene is optionally substituted with 1 to 3 substituents independently selected from halo, N (R)3)2And OR3Substituted with a group of (1);

a is C6-10Aryl or 5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S, and said aryl or heteroaryl is optionally substituted with 1 to 3R4Substitution;

b is C6-10Aryl, 5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S, a 3-8 membered cycloalkyl ring, or 3-12 membered heterocycloalkyl having 1-3 ring heteroatoms selected from O, S and N, and said aryl, heteroaryl, cycloalkyl or heterocycloalkyl optionally substituted with 1 to 3R5Substitution;

R1and R2Each independently is C1-6Alkyl, or R1And R2Together with the carbon atom to which they are attached form a 4-8 membered cycloalkyl or heterocycloalkyl ring, wherein the heterocycloalkyl ring has 1 or 2 ring heteroatoms selected from O, S and N, and wherein the cycloalkyl or heterocycloalkyl ring is optionally interrupted by 1-2R6Substitution;

each R3Independently is H or C1-6An alkyl group;

each R4And R5Independently is C1-6Alkyl radical, C1-6Haloalkyl, halo or C1-6An alkoxy group; and is

R6Is C1-6Alkyl radical, C1-6Haloalkyl, (C ═ O) R3、(C=O)OR3、CON(R3)2、C0-3alkylene-C3-8Cycloalkyl radical, C0-3alkylene-C6-10Aryl or C0-3Alkylene- (5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S), wherein the aryl or heteroaryl is optionally substituted with 1 to 3R5And (4) substitution.

2.A compound or salt according to claim 1, wherein R1And R2Each independently is C1-6An alkyl group.

3.A compound or salt according to claim 2, wherein R1And R2Each is methyl.

4.A compound or salt according to claim 1, wherein R1And R2Together with the carbon atom to which they are attached form a 4-8 membered cycloalkyl or heterocycloalkyl ring.

5.A compound or salt according to claim 4, wherein R1And R2Together with the carbon atom to which they are attached form a 5-or 6-membered cycloalkyl or heterocycloalkyl ring.

6. A compound or salt according to claim 5, wherein R1And R2Together with the carbon atom to which they are attached form a cyclohexyl ring.

7. A compound or salt according to claim 5, wherein R1And R2Together with the carbon atom to which they are attached form a heterocycloalkyl ring having the structure:wherein denotes the point of attachment to the remainder of the compound of formula I.

8. A compound or salt according to any one of claims 1-7, wherein each R4And R5Independently is C1-6Alkyl, halo or C1-6An alkoxy group.

9. The compound or salt of any one of claims 1-8, wherein R6Is C1-6Alkyl, (C)=O)R3、(C=O)OR3Or CON (R)3)2

10. The compound or salt of any one of claims 4-9, wherein R6Is C1-6An alkyl group.

11.A compound or salt according to claim 10, wherein R6Is methyl.

12. The compound or salt of any one of claims 1-11 wherein a is C6-10And (4) an aryl group.

13. A compound or salt according to claim 12, wherein a is phenyl.

14. The compound or salt of any one of claims 1-13 wherein B is C6-10And (4) an aryl group.

15.A compound or salt according to claim 14, wherein B is phenyl.

16. The compound or salt of any one of claims 1 to 13 wherein B is a 5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S.

17. A compound or salt of claim 16 wherein B is pyridyl.

18. A compound or salt according to claim 16, wherein B is quinolinyl.

19. The compound or salt of any one of claims 1-13 wherein B is 3-8 membered cycloalkyl.

20. The compound or salt of claim 18 wherein B is a5 or 6 membered cycloalkyl.

21. The compound or salt of any one of claims 1 to 13 wherein B is a 3-12 membered heterocycloalkyl having 1-3 ring heteroatoms selected from O, S and N.

22. The compound or salt according to any one of claims 1-21, wherein a is substituted with one R4And (4) substitution.

23. A compound or salt according to claim 22, wherein a has the structure:

24. the compound or salt of any one of claims 1-21, wherein a is substituted with two R4And (4) substitution.

25. The compound or salt of any one of claims 1-24, wherein at least one R4Is C1-6An alkyl group.

26. A compound or salt according to claim 25, wherein at least one R4Is methyl.

27. The compound or salt of any one of claims 1-24, wherein at least one R4Is a halo group.

28. A compound or salt according to claim 27, wherein R4Is bromine.

29. The compound or salt of any one of claims 1-24, wherein at least one R4Is C1-6An alkoxy group.

30. A compound or salt according to claim 29, wherein at least one R4Is methoxy.

31. The compound or salt according to any one of claims 1-30, wherein B is substituted with one R5And (4) substitution.

32. The compound or salt of any one of claims 1-30 wherein B is substituted with two R5And (4) substitution.

33. The compound of claim 32, wherein B has the structure

Figure FDA0002688847210000031

34. The compound or salt of any one of claims 1 to 33, wherein at least one R5Is a halo group.

35. A compound or salt according to claim 34, wherein at least one R5Is fluorine or chlorine.

36. The compound or salt of claim 30 or 33, wherein one R5Is fluorine and the other R5Is chlorine.

37. The compound or salt of any one of claims 1 to 33, wherein at least one R5Is C1-6An alkoxy group.

38. A compound or salt according to claim 37, wherein at least one R5Is methoxy.

39. The compound or salt of claim 30 or 33, wherein one R5Is halo and the other R is5Is C1-6An alkoxy group.

40. A compound or salt according to claim 39, wherein one R5Is chlorine and the other isR5Is methoxy.

41. The compound or salt of any one of claims 1 to 40 wherein X is O-C0-6Alkylene or S-C0-6An alkylene group.

42. A compound or salt according to claim 41, wherein X is S-C0-6An alkylene group.

43. A compound or salt of claim 41 wherein X is O, S, O-CH2-or S-CH2-。

44. The compound or salt of any one of claims 1-43, wherein Y is C0-2An alkylene group.

45. The compound or salt of claim 44 wherein Y is null or CH2

46. The compound of claim 45, wherein X is NR3-CH2、O-CH2-or S-CH2-, and Y is null.

47. The compound or salt of claim 45 wherein X is NR3-CH2、O-CH2-or S-CH2-, and Y is CH2

48. The compound or salt of any one of claims 1-47, wherein R3Is H.

49. A compound listed in table 1 or a pharmaceutically acceptable salt thereof.

50. A compound listed in table 2 or a pharmaceutically acceptable salt thereof.

51. A pharmaceutical composition comprising a compound or salt according to any one of claims 1-50, and a pharmaceutically acceptable carrier or excipient.

52. A method of modulating EGFR comprising administering to a subject in need thereof a therapeutically effective amount of a compound or salt of any one of claims 1 to 50.

53. The method of claim 52, wherein said modulating comprises inhibiting EGFR dimerization.

54. The method of claim 52 or 53, wherein said modulating comprises inducing EGFR degradation.

55. A method of treating or preventing a disease or disorder associated with aberrant EGFR activity, comprising administering to a subject in need thereof a therapeutically effective amount of a compound or salt according to any one of claims 1 to 50.

56. The method of claim 55, wherein the disease or disorder is cancer.

57. The method of claim 56, wherein the cancer is selected from lung cancer, colorectal cancer, glioblastoma, and head and neck cancer.

58. The method of claim 56, wherein the cancer is melanoma, colon cancer, renal cancer, leukemia, or breast cancer.

59. The method of any one of claims 56-58, wherein the cancer is an Onchitinib-resistant cancer.

60. The method of any one of claims 52-59, wherein the subject is a human.

Background

EGFR small molecule Tyrosine Kinase Inhibitors (TKI) erlotinib, gefitinib and afatinib were most successful as single drugs for treating lung adenocarcinomas with somatic mutations (e.g., L858R or exon 19 deletion, i.e., E746-a750 deletion) that confer sensitivity to such drugs, which occur in 7-20% of patients depending on race and sex (19). Unfortunately, the response rarely lasts for more than a year, as almost all patients develop resistance to treatment (20). The third generation irreversible inhibitor, oxitinib (AZD9291), is effective in treating untreated patients and patients who develop resistance to first or second generation TKIs (7). However, within one year of treatment with oxitinib, most patients develop another mutation in the EGFR kinase domain (C797S), which is the drug binding site (12, 21, 22). Although several approaches to targeting axitinib resistant EGFR have been reported (12, 13, 23), until now, these patients with the C797S mutation have no TKI treatment option. Chemotherapy is the only option.

In view of the foregoing, there is a need for cancer therapeutics that target EGFR in ways other than inhibiting EGFR tyrosine kinase activity. There is also a need for a therapeutic agent for the treatment of cancer that does not develop resistance after initial use.

Disclosure of Invention

Provided herein are compounds and methods for modulating EGFR. More specifically, EGFR modulators and the use of such modulators in the treatment or prevention of diseases or disorders associated with aberrant EGFR activity (e.g., cancer) are provided.

In one aspect, the present disclosure provides a compound of formula I:

Figure BDA0002688847220000011

wherein X is O-C0-6Alkylene, S-C0-6Alkylene or NR3-C0-6Alkylene and the alkylene is X is O-C0-6Alkylene, S-C0-6Alkylene or NR3-C0-6Alkylene, and said alkylene is optionally substituted with 1 to 3 substituents independently selected from halo, N (R)3)2And OR3Substituted with a group of (1); y is C0-6Alkylene, and said alkylene is optionally substituted with 1 to 3 substituents independently selected from halo, N (R)3)2And OR3Substituted with a group of (1); a is C6-10Aryl or 5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S, and said aryl or heteroaryl is optionally substituted with 1 to 3R4Substitution; b is C6-10Aryl, 5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S, a 3-8 membered cycloalkyl ring, or 3-12 membered heterocycloalkyl having 1-3 ring heteroatoms selected from O, S and N, and said aryl, heteroaryl, cycloalkyl or heterocycloalkyl optionally substituted with 1 to 3R5Substitution; r1And R2Each independently is C1-6Alkyl, or R1And R2Together with the carbon atom to which they are attached form a 4-8 membered cycloalkyl or heterocycloalkyl ring, wherein the heterocycloalkyl ring has 1 or 2 ring heteroatoms selected from O, S and N, and wherein the cycloalkyl or heterocycloalkyl ring is optionally interrupted by 1-2R6Substitution; each R3Independently is H or C1-6An alkyl group; each R4And R5Independently is C1-6Alkyl radical, C1-6Haloalkyl, halo or C1-6An alkoxy group; and R is6Is C1-6Alkyl radical, C1-6Haloalkyl, (C ═ O) R3、(C=O)OR3、CON(R3)2、C0-3alkylene-C3-8Cycloalkyl radical, C0-3alkylene-C6-10Aryl or C0-3Alkylene- (5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S), wherein the aryl or heteroaryl is optionally substituted with 1 to 3R5And (4) substitution.

Further provided herein are methods of modulating EGFR using the disclosed compounds. Other aspects of the disclosure include methods of inhibiting EGFR dimerization using the disclosed compounds, and methods of inducing EGFR degradation using the disclosed compounds.

Other aspects of the disclosure include a compound as disclosed herein for use in the preparation of a medicament for treating or preventing a disease or disorder associated with aberrant EGFR activity in a subject, and the use of a compound as disclosed herein for a method of treating or preventing a disease or disorder associated with aberrant EGFR activity in a subject.

Drawings

Figure 1 shows (left) treatment with small peptides that inhibit tumor (marked by x) -specific EGFR for EGFR dimerization more than adjacent normal tissues; and (right) EGFR degradation schematic. EGFR is shown as an extracellular domain (small spot ═ n-leaf, large spot ═ C-leaf) and a flexible C-terminal tail linked to a biplate kinase domain via a transmembrane portion. EGF binding promotes active (asymmetric) dimer formation between the extracellular domains and/or between the n-lobe and c-lobe of the two monomers. EGFR in this conformation remains stable and induces phosphorylation of the C-terminal tail, thereby promoting tumor progression tyrosine kinase inhibitors (e.g., ocitinib), inhibiting ATP recruitment and promoting inactive (symmetric) dimerization between the n-lobes of the two monomers. EGFR in this conformation retains the kinase inactive conformation but maintains protein stability. Inactivation of EGFR activity is associated with tumor growth inhibition. The α C-helix and β 4-sheet of the C-lobe of EGFR and the loop between the h-helix of the n-lobe are involved in EGF-induced active dimer formation. This model postulates that interferon (irterptin) or compound 8C binds in the capsule and interferes with EGF-induced active dimer formation. EGFR monomers bound to ligand and compound 8C degraded rapidly. Loss of EGFR protein is associated with cell death. The thickness of the arrow shows the effect of compound 8C in tumor cells versus normal cells.

FIG. 2 shows (A) a schematic of an asymmetrically active dimer of EGFR. One monomer of EGFR that binds EGF is sufficient to induce dimerization. The insert shows a model of interferon binding to the c-leaf of EGFR (PDB code: 2RFD), and (B) the putative interaction between the purified EGFR kinase domain and interferon.

FIG. 3 shows (A) the effect of interferon treatment on EGF-induced dimerization in NCI-H1975 cells; (B) the effect of interferon on its target EGFR in NCI-H1975 xenografts; (C) the efficacy of interferon in vivo; and (D) long-term effects of treatment on tumor histology, EGFR expression, and mitotic index (measured by Ki-67 scoring).

Figure 4 shows (a) the procedure for pre-screening lead compounds, and the structures of two pre-lead compounds C95 and C67; (B) the resulting SAR and microsomal stability (shown in blue boxes), and the structure of the most selective potent molecule (compound 8C); (C) the effect of two pre-lead compounds 8C on EGF-stimulated EGFR dimerization in erlotinib-resistant NCI-H1975 lung cancer cell lines, as shown in fig. 3A (lysates prepared and immunoblotted with anti-EGFR antibodies); and (D) the effect of the selected three lead molecules (1 μ M) on the steady state EGFR protein assessed 24 hours after treatment.

Figure 5 shows (a) competitive EGFR binding of compound 8C to interferon; (B) the effect of compound 8C on the thermal stability of purified EGFR as demonstrated by the thermal stability assay; and (C) the effect of compound 8C concentration on EGFR thermostability at 44 ℃ in the presence of 0 to 10 μ M compound 8C.

FIG. 6 shows (A) the effect of Compound 8C in whole cell lysates from Ba/F3-AZD cells on thermal stability of EGFR; (B) the potency of compound 8C measured against oxitinib-resistant Ba/F3 cells expressing specific EGFR mutations; (C) effect of compound 8C on EGF-induced EGFR dimerization, as shown in figure 3A; (C-D) Effect of Compound 8C treatment on EGFR-induced dimer and EGFR protein levels; (E) IC's in response to erlotinib, oxitinib and Compound 8C tabulated from two oxitinib-resistant Ba/F3 cell lines50The value is obtained.

Figure 7 shows (a) the specificity and potency of compound 8C determined against a panel of ocitinib-resistant cells and compared to normal lung fibroblasts (MRC5) using a clonogenic survival assay; (B) effect of compound 8C concentration on EGF-induced EGFR dimerization in axitinib-resistant PC9 cells, as shown in figure 6; (C) effect of compound 8C treatment on EGFR-induced dimer and EGFR protein levels in PC9-AZR cells.

FIG. 8 shows (A) a human NCI-H1975 tumor xenograft (A)>150mm3) The pharmacokinetics of a single 100mg/kg dose of compound 8C administered intraperitoneally in nude mice; and (B) in carrying a human NCI-H1975 tumor xenograft (>150mm3) The pharmacokinetics of compound 8C in nude mice given by oral gavage at a single 100mg/kg dose.

Figure 9 shows (a) the effect of basal bioluminescence and pre-lead compound 95 on different time points in mice bearing NCI-H1975 xenografts; and (B) quantification and mapping changes in bioluminescence.

FIG. 10 shows the mean tumor volume as a function of time for nude mice bearing UMSCC74B, head and neck tumor model (about 100 mm)2) And treated with compound 8C (30mg/kg daily for one week) or vehicle (5% DMSO in PBS). There were at least 5 mice per group. Tumor volumes and body weights were recorded 3-4 times weekly and plotted. The average loss of body weight during treatment was less than 10%. Error bars represent standard error of the mean.

Figure 11 shows the effect of compound 8C in an axitinib-resistant tumor model. Nude mice with oxitinib-resistant Ba/F3 ascites tumor were treated with vehicle, oxitinib or compound 8C. The effect of treatment on EGFR, pEGFR and other molecules was determined by immunoblotting.

Fig. 12 shows the pathological assessment of FFPE-lung PDX in primary tumors, 1 st and 2 nd xenografts of UMLCA7, and analysis of EGFR protein expression using IHC in two additional PDX. Note that there was a high amount of EGFR expression in squamous cell carcinoma (UMLT16), but not in large cell carcinoma (UMLT14) PDX sample (20X).

Figure 13 shows that treatment of the mouse model of pancreatic cancer with compound 8C showed a significantly reduced propensity to develop PanIn, a pancreatic ductal lesion. Histopathological examination of the pancreas tissue showed a significant reduction in lesion involvement in the treated mice compared to the control group (quantified as B).

Figure 14 shows that mouse xenografts carrying the UMSCC74B head and neck tumor model treated with compound 8C showed significantly smaller tumors compared to control mice receiving treatment vehicle or cetuximab.

Detailed Description

Although inhibition of kinase activity of oncogenic proteins using small molecules and antibodies has been the primary means of anticancer drug development efforts, leading to several FDA-approved cancer therapies, the clinical efficacy of kinase-targeted agents has been inconsistent (22, 24). EGFR has shown scaffold function in addition to tyrosine kinase activity (24-36). This can be demonstrated by Kinase Death (KD) mutants expressing EGFR (e.g., K745A, V741G and Y740F) or by expression of ErbB3 (no kinase activity) in Ba/F3 cells that do not express these receptors (37-39). Expression of these kinase-deficient mutants promotes cell survival, suggesting that these receptors may still transmit survival signals by forming dimers, suggesting that EGFR functions beyond kinase activity (39).

EGFR dimers are known to be relatively stable when compared to monomers (40). The dimer is capable of producing a downstream mitogenic signal (41). Without being bound by theory, it is hypothesized that blocking EGFR dimerization will accelerate EGFR degradation, and this approach will be effective against tumors driven by TKI resistant EGFR (14, 24, 27). Briefly, it was demonstrated that EGFR binding to EGF (phosphorylated EGFR prevalent in most tumors) protein stability is modulated by dimer formation by fragments in the EGFR kinase domain located between the α C helix and β 4 sheet of the C-lobe and the h-helix of the n-lobe of the EGFR kinase domain (15, 42). EGFR protein stability in normal cells is largely unregulated by this dimer interface, as EGFR does not form asymmetric dimers in the absence of EGF (43). This difference between tumor cells and normal cells provides a novel targetable protein-protein interaction.

To test this idea, a dozen peptides were generated that mimic the binding surface. The most potent peptide contains six amino acids from the α C- β 4 loop of EGFR, which is called interferon (17). Interferons are capable of inhibiting EGF-induced EGFR dimerization. This peptide binds directly to EGFR and repeated HEPES washes did not significantly affect this binding compared to the control peptide (scrambled peptide). Although interferons are effective in a Tyrosine Kinase Inhibitor (TKI) resistant lung xenograft model (14), delivery of peptides in humans remains challenging (44).

Provided herein are compounds that modulate EGFR, e.g., block EGFR dimerization, induce EGFR degradation, and kill EGFR-driven cells. These compounds are useful in the prevention or treatment of a variety of diseases and disorders, for example, in the treatment of cancer.

Accordingly, provided herein is a compound having the structure of formula I:

wherein

X is O-C0-6Alkylene, S-C0-6Alkylene or NR3-C0-6Alkylene, and said alkylene is optionally substituted with 1 to 3 substituents independently selected from halo, N (R)3)2And OR3Substituted with a group of (1);

y is C0-6Alkylene, and said alkylene is optionally substituted with 1 to 3 substituents independently selected from halo, N (R)3)2And OR3Substituted with a group of (1);

a is C6-10Aryl or 5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S, and said aryl or heteroaryl is optionally substituted with 1 to 3R4Substitution;

b is C6-10Aryl, 5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S, a 3-8 membered cycloalkyl ring, or 3-12 membered heterocycloalkyl having 1-3 ring heteroatoms selected from O, S and N, and said aryl, heteroaryl, cycloalkyl or heterocycloalkyl optionally substituted with 1 to 3R5Substitution;

R1and R2Each of which isIndependently is C1-6Alkyl, or R1And R2Together with the carbon atom to which they are attached form a 4-8 membered cycloalkyl or heterocycloalkyl ring, wherein the heterocycloalkyl ring has 1 or 2 ring heteroatoms selected from O, S and N, and wherein the cycloalkyl or heterocycloalkyl ring is optionally interrupted by 1-2R6Substitution;

each R3Independently is H or C1-6An alkyl group;

each R4And R5Independently is C1-6Alkyl radical, C1-6Haloalkyl, halo or C1-6An alkoxy group; and is

R6Is C1-6Alkyl radical, C1-6Haloalkyl, (C ═ O) R3、(C=O)OR3、CON(R3)2、C0-3alkylene-C3-8Cycloalkyl radical, C0-3alkylene-C6-10Aryl or C0-3Alkylene- (5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S), wherein aryl or heteroaryl is optionally substituted with 1 to 3R5And (4) substitution.

In various embodiments, R1And R2Each independently is C1-6An alkyl group. In some embodiments, R1And R2Each is methyl.

In various embodiments, R1And R2Together with the carbon atom to which they are attached form a 4-8 membered cycloalkyl or heterocycloalkyl ring. In some embodiments, R1And R2Together with the carbon atom to which they are attached form a 5-or 6-membered cycloalkyl or heterocycloalkyl ring. In some embodiments, R1And R2Together with the carbon atom to which they are attached form a cyclohexyl ring.

In various embodiments, R1And R2Together with the carbon atom to which they are attached form a heterocycloalkyl ring having the structure:

Figure BDA0002688847220000061

wherein denotes the point of attachment to the remainder of the compound of formula I. In some embodiments, R6Is C1-6Alkyl radical, C1-6Haloalkyl, (C ═ O) R3、(C=O)OR3、CON(R3)2、C0-3alkylene-C3-8Cycloalkyl radical, C0-3alkylene-C6-10Aryl or C0-3Alkylene- (5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S), wherein aryl or heteroaryl is optionally substituted with 1 to 3R5And (4) substitution. In some embodiments, R6Is C1-6Alkyl, (C ═ O) R3、(C=O)OR3Or CON (R)3)2. In some embodiments, R6Is C1-6An alkyl group. In some embodiments, R6Is methyl, ethyl, propyl, isopropyl, isobutyl or isoamyl. In some embodiments, R6Is methyl. In some embodiments, R6Is deuterated. In some embodiments, R6Is C1-6A haloalkyl group. In some embodiments, R6Is 3,3, 3-trifluoropropyl. In some embodiments, R6Is C0-3alkylene-C3-8A cycloalkyl group. In some embodiments, R6Is cyclobutyl, cyclopentyl or cyclohexyl. In some embodiments, R6Is cyclobutyl or cyclopentyl. In some embodiments, R6Is C0-3alkylene-C6-10And (4) an aryl group. In some embodiments, R6Is benzyl. In some embodiments, R6Is C0-3Alkylene- (5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S), wherein the heteroaryl is optionally substituted with 1 to 3R5And (4) substitution. In some embodiments, R6Is C1Alkylene- (5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S), wherein the heteroaryl is optionally substituted with 1 to 3R5And (4) substitution. In some embodiments, R6Is C0-3Alkylene- (5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S), wherein the heteroaryl is substituted with 1 to 3R5And (4) substitution. In some embodiments, R6Is C0-3Alkylene- (5-to 10-membered heteroaryl having 1-4 heteroatoms selected from N, O and S), wherein the heteroaryl is unsubstituted. In some embodiments, R6Is composed of

Figure BDA0002688847220000062

In various embodiments, A is C6-10And (4) an aryl group. In some embodiments, a is phenyl.

In various embodiments, B is C6-10And (4) an aryl group. In some embodiments, B is phenyl. In various embodiments, B is a 5-10 membered heteroaryl having 1-4 heteroatoms selected from N, O and S. In some embodiments, B is pyridyl. In some embodiments, B is quinolinyl. In various embodiments, B is a 3-8 membered cycloalkyl group. In some embodiments, B is a 5-or 6-membered cycloalkyl. In various embodiments, B is a 3-12 membered heterocycloalkyl having 1-3 ring heteroatoms selected from O, S and N.

In some embodiments, A is substituted with one R4And (4) substitution. In some embodiments, a has the structure:

Figure BDA0002688847220000071

in some embodiments, A is substituted with two R4And (4) substitution. In some embodiments, at least one R4Is C1-6An alkyl group. In some embodiments, at least one R4Is methyl. In some embodiments, at least one R4Is a halo group. In some embodiments, R4Is bromine. In some embodiments, at least one R4Is C1-6An alkoxy group. In some embodiments, at least one R4Is methoxy.

In some embodiments, B is substituted with one R5And (4) substitution. In some embodiments, B is substituted with two R5And (4) substitution. In some embodiments, B has the structure

Figure BDA0002688847220000072

In some embodiments, at least one R5Is a halo group. In some embodiments, at least one R5Is fluorine or chlorine. In some embodiments, one R5Is fluorine and the other R5Is chlorine. In some embodiments, at least one R5Is C1-6An alkoxy group. In some embodiments, at least one R5Is methoxy. In some embodiments, one R5Is halo and the other R is5Is C1-6An alkoxy group. In some embodiments, one R5Is chlorine and the other R is5Is methoxy.

In some embodiments, each R is4And R5Independently is C1-6Alkyl, halo or C1-6An alkoxy group. In some embodiments, R6Is C1-6Alkyl, (C ═ O) R3、(C=O)OR3Or CON (R)3)2

In various embodiments, X is O-C0-6Alkylene or S-C0-6An alkylene group. In some embodiments, X is S-C0-6An alkylene group. In some embodiments, X is O, S, O-CH2-or S-CH2-. In various embodiments, Y is C0-2An alkylene group. In some embodiments, Y is null or CH2. In some embodiments, X is NR3-CH2、O-CH2-or S-CH2-, and Y is null. In some embodiments, X is NR3-CH2、O-CH2-or S-CH2-, and Y is CH2. In some embodiments, R3Is H.

Particular compounds contemplated include those listed in tables 1,2 or pharmaceutically acceptable salts thereof:

TABLE 1

Figure BDA0002688847220000091

TABLE 2

In some cases, the compound is a compound listed in table 1 or a salt thereof.

Compound 8C showed significant improvements in pharmacological properties as well as biological activity (microsomal half-life within 46 minutes, submicromolar IC in clonogenic cell assay50). Compound 8C inhibits EGF-induced EGFR dimerization, binds directly to purified EGFR, and is selectively active in EGFR-driven axitinib-resistant cell lines and xenograft models.

Definition of

As used herein, the term "alkyl" refers to straight and branched chain saturated hydrocarbon groups containing from one to thirty carbon atoms (e.g., from one to twenty carbon atoms or from one to ten carbon atoms). Term CnMeaning that the alkyl group has "n" carbon atoms. E.g. C4Alkyl refers to an alkyl group having 4 carbon atoms. C1-C7Alkyl refers to alkyl groups having a number of carbon atoms that encompasses the entire range (e.g., 1 to 7 carbon atoms) as well as all subranges (e.g., 1-6, 2-7, 1-5, 3-6, 1,2, 3,4, 5,6, and 7 carbon atoms). Non-limiting examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl (2-methylpropyl), tert-butyl (1, 1-dimethylethyl), 3-dimethylpentyl, and 2-ethylhexyl. Unless otherwise specified, an alkyl group may be an unsubstituted alkyl group or a substituted alkyl group.

The term "alkylene" as used herein refers to an alkyl group having a substituent. For example: the term "alkylene halo" refers to an alkyl group substituted with a halo group. The alkylene group may be-CH2CH2-or-CH2-. Operation of the artLanguage CnDenotes an alkylene group having "n" carbon atoms. E.g. C1-6Alkylene refers to alkylene groups having a number of carbon atoms that spans the entire range as well as all subranges, as previously described for "alkyl" groups. Unless otherwise specified, an alkylene group may be an unsubstituted alkylene group or a substituted alkylene group.

As used herein, the term "cycloalkyl" refers to an aliphatic cyclic hydrocarbon group containing three to eight carbon atoms (e.g., 3,4, 5,6, 7, or 8 carbon atoms). Term CnRepresents a cycloalkyl group having "n" carbon atoms. E.g. C5Cycloalkyl refers to cycloalkyl having 5 carbon atoms in the ring. C6-C8Cycloalkyl refers to cycloalkyl groups having a number of carbon atoms that encompasses the entire range (e.g., 6 to 8 carbon atoms) as well as all subranges (e.g., 6-7, 7-8, 6, 7, and 8 carbon atoms). Non-limiting examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Unless otherwise specified, cycloalkyl groups may be unsubstituted cycloalkyl groups or substituted cycloalkyl groups. The cycloalkyl groups described herein may be alone or fused with another cycloalkyl, heterocycloalkyl, aryl, and/or heteroaryl group. When a cycloalkyl group is fused to another cycloalkyl group, each cycloalkyl group can contain three to eight carbon atoms, unless otherwise specified. Unless otherwise specified, cycloalkyl groups may be unsubstituted or substituted.

As used herein, the term "heterocycloalkyl" is defined similarly to cycloalkyl, except that the ring contains one to three heteroatoms independently selected from oxygen, nitrogen, and sulfur. Specifically, the term "heterocycloalkyl" refers to a ring containing a total of three to twelve atoms (e.g., 3-8, 5-8, 3-6, 3,4, 5,6, 7, 8, 9, 10, 11, or 12) wherein 1,2, or 3 of the ring atoms are heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, and the remaining atoms in the ring are carbon atoms. Non-limiting examples of heterocycloalkyl groups include piperidine, pyrazolidine, tetrahydrofuran, tetrahydropyran, dihydrofuran, morpholine, and the like.

Cycloalkyl and heterocycloalkyl groups may be saturated or partially unsaturated ring systems, optionally substituted, for example, by one to three groups independently selected from alkyl, alkylene OH, C (O) NH2、NH2Oxy (═ O), aryl, alkylene halide, and OH. The heterocycloalkyl group can be optionally further N-substituted with alkyl (e.g., methyl or ethyl), alkylene-OH, alkylenearyl, and alkyleneheteroaryl. The heterocycloalkyl groups described herein may be alone or fused with another heterocycloalkyl, cycloalkyl, aryl, and/or heteroaryl group. When a heterocycloalkyl group is fused to another heterocycloalkyl group, each heterocycloalkyl group can contain three to twelve total ring atoms, and one to three heteroatoms, unless otherwise specified. Unless otherwise specified, heterocycloalkyl groups may be unsubstituted or substituted.

As used herein, the term "aryl" refers to a monocyclic or bicyclic aromatic group having 6 to 10 ring atoms. Unless otherwise specified, an aryl group may be unsubstituted or substituted with one or more (and specifically, one to four) groups selected from, for example, halo, alkyl, alkenyl, OCF3、NO2CN, NC, OH, alkoxy, amino, alkylamino, CO2H、CO2Alkyl, aryl and heteroaryl. The aryl group can be alone (e.g., phenyl), or fused with another aryl group (e.g., naphthyl, anthracenyl), cycloalkyl (e.g., tetrahydronaphthyl), heterocycloalkyl, and/or heteroaryl

As used herein, the term "heteroaryl" refers to a monocyclic or bicyclic aromatic ring having 5 to 10 total ring atoms and containing one to four heteroatoms selected from nitrogen, oxygen, and sulfur atoms in the aromatic ring. Unless otherwise specified, heteroaryl groups may be unsubstituted or substituted with one or more (and specifically, one to four) substituents selected from, for example, halo, alkyl, alkenyl, OCF3、NO2CN, NC, OH, alkoxy, amino, CO2H、CO2Alkyl, aryl and heteroaryl. In some cases, the heteroaryl group is substituted with one or more of alkyl and alkoxyAnd (4) substitution. Examples of heteroaryl groups include, but are not limited to, thienyl, furyl, pyridyl, pyrrolyl, oxazolyl, triazinyl, triazolyl, isothiazolyl, isoxazolyl, imidazolyl, pyrazinyl, pyrimidinyl, thiazolyl, and thiadiazolyl.

As used herein, the term "alkoxy (alkoxy or alkxyl)" as used herein refers to an "-O-alkyl" group. The alkoxy group (alkoxy or alkoxyl) may be unsubstituted or substituted.

As used herein, the term "therapeutically effective amount" refers to an amount of a compound or combination of therapeutically active compounds that ameliorates, reduces, or eliminates one or more symptoms of a particular disease or condition (e.g., cancer) or prevents or delays the onset of one or more symptoms of a particular disease or condition.

As used herein, the terms "patient" and "subject" are used interchangeably and refer to animals, such as dogs, cats, cows, horses, and sheep (e.g., non-human animals), and humans. A particular patient or subject is a mammal (e.g., a human).

As used herein, the term "pharmaceutically acceptable" means that the reference substance (e.g., a compound of the present disclosure, or a formulation or specific excipient containing the compound) is safe and suitable for administration to a patient or subject. The term "pharmaceutically acceptable excipient" refers to a medium that does not interfere with the efficacy of the biological activity of the active ingredient or ingredients and is non-toxic to the host to which it is administered.

The compounds disclosed herein can be pharmaceutically acceptable salts. As used herein, the term "pharmaceutically acceptable salt" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without excessive toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in detail in journal of pharmaceutical Sciences (j. pharmaceutical Sciences), 1977,66,1-19, by s.m. berge et al, which is incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of the present invention compriseSalts derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable non-toxic acid addition salts are amino salts formed with inorganic acids (such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid) or organic acids (, such as acetic acid, trifluoroacetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid) or by using other methods used in the art (such as ion exchange). Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartates, benzenesulfonates, benzoates, bisulfates, borates, butyrates, camphorates, camphorsulfonates, citrates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, formates, fumarates, glucoheptonates, glycerophosphates, gluconates, glutamates, hemisulfates, heptanoates, hexanoates, hydroiodides, 2-hydroxy-ethanesulfonates, lactobionates, lactates, laurates, malates, maleates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, oxalates, palmitates, pamoates, pectinates, persulfates, 3-phenylpropionates, persulfates, salts of alginic acid, salts of, Phosphates, picrates, pivalates, propionates, stearates, succinates, sulfates, tartrates, thiocyanates, p-toluenesulfonates, undecanoates, pentanoates, and the like. Salts of compounds containing carboxylic acid or other acidic functional groups may be prepared by reaction with a suitable base. Such salts include, but are not limited to, alkali metals, alkaline earth metals, aluminum salts, ammonium, N+(C1-4Alkyl radical)4Salts and salts with organic bases such as trimethylamine, triethylamine, morpholine, pyridine, piperidine, picoline, dicyclohexylamine, N '-dibenzylethylenediamine, 2-hydroxyethylamine, bis- (2-hydroxyethyl) amine, tris- (2-hydroxyethyl) amine, procaine, dibenzylpiperidine, dehydroabietylamine, N' -didehydroabietylamine, glucosamine, N-methylglucamine, collidine, quinine, quinoline, and basic amino acids such as lysine and arginine. The present invention also contemplates the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water-or oil-soluble or dispersible products can be obtained by this quaternization.Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Additional pharmaceutically acceptable salts include non-toxic ammonium, quaternary ammonium, and amine cations formed using counterions (e.g., halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, lower alkyl sulfonates, and aryl sulfonates) as appropriate.

As used herein, the term "treating" encompasses both prophylactic (e.g., prophylactic) treatment and palliative treatment.

As used herein, the term "excipient" refers to any pharmaceutically acceptable additive, carrier, diluent, adjuvant, or other ingredient other than an Active Pharmaceutical Ingredient (API).

Synthesis of Compounds of the disclosure

The compounds disclosed herein can be prepared in a variety of ways using commercially available starting materials, compounds known in the literature or from readily prepared intermediates, by employing standard synthetic methods and procedures known to those skilled in the art or in view of the teachings herein. The compounds disclosed herein can generally be synthesized by following the synthetic schemes described in the examples section and modifying the particular desired substituents.

Standard synthetic methods and procedures for the preparation of organic molecules and the transformation and manipulation of functional groups can be obtained from the relevant scientific literature or from standard textbooks in the field. Although not limited to any one or several sources, classical text (e.g., Smith, M.B., March, J., "March's advanced Organic Chemistry: Reactions, Mechanisms, and structures)," 5 th edition, John Wiley & Sons, New York, 2001, and Greene, T.W., Wuts, P.G.M., "Protective Groups in Organic Synthesis (Protective Groups in Organic Synthesis), 3 rd edition, John Wiley's parent, New York, 1999) are useful and recognized Organic synthetic reference textbooks known to those skilled in the art. The following description of the synthetic methods is intended to illustrate, but not limit, the general procedures for preparing the compounds of the present disclosure.

The synthetic methods disclosed herein may tolerate a wide variety of functional groups; various substituted starting materials may therefore be used. The methods provide substantially the desired final compound at or near the end of the overall process, but in some cases may require further conversion of the compound to its pharmaceutically acceptable salt.

Generally, compounds of formula (I) can be synthesized according to scheme 1.

Scheme 1

Figure BDA0002688847220000171

Compounds having structure c can be synthesized using the procedure shown in scheme 1. The substituted 2, 5-dihydroimidazole derivative a is reacted with an amide compound b in a suitable solvent (e.g., acetonitrile) to produce the compounds described herein, i.e., compounds of formula (I) having structure c. The amide compound b includes a suitable leaving group LG selected based on the nature of the group Q, for example, a halogen or tosylate. Can be based on substituents R1、R2Nature of A, Y and B the appropriate additional derivatization reaction of the compound having structure c is selected.

The coupling of compounds a and b can be catalyzed by appropriate reagents selected based on the precise nature of compounds a and b. For example, when LG of compound b is halogen (e.g., when LG is chlorine), coupling of compounds a and b can be catalyzed by a base (e.g., sodium carbonate or potassium carbonate). Sometimes the coupling reaction may not require a catalyst.

In some cases, compounds having a structure selected from O, S and NR can be prepared by treatment with an appropriate reagent prior to coupling with a compound having structure b3Compound a of Q is converted to a compound having a structure selected from the group consisting of O, S and NR3Compounds of Q of different members of the group. For example, a compound having structure a of Q ═ O can be converted to a compound having structure a of Q ═ S by treatment with a sulfur hybridizing reagent (e.g., lawson' S reagent or diphosphorus pentasulfide). This compound can then be coupled with a compound having structure b to produce a compound described herein, i.e., havingA compound of formula (I) having structure c.

Compounds a and b can be commercially available or prepared by a variety of methods from commercially available starting materials. For example, amide compounds having structure b can be prepared by, for example, the reaction of an acid chloride with an amine.

May be based on the substituent R in the compound c1、R2The nature of A, Y and B, and the desired functionality in the derivative compound, select additional derivatization reactions that convert compounds having structure c to other compounds disclosed herein. For example, R1And R2Together with the carbon atom to which they are attached, can form a heterocyclic ring, such as a piperidine ring, which can be further derivatized (e.g., methylated, addition of protecting groups, etc.) by methods known in the art to form a variety of other compounds of formula (I) described herein.

Pharmaceutical formulations, dosages and routes of administration

Further provided are pharmaceutical formulations comprising a compound described herein (e.g., a compound of formula I or a pharmaceutically acceptable salt thereof) and a pharmaceutically acceptable excipient.

The compounds described herein can be administered to a subject in a therapeutically effective amount (e.g., an amount sufficient to prevent or alleviate symptoms of a disease or disorder associated with aberrant EGFR). The compounds may be administered alone or as part of a pharmaceutically acceptable composition or formulation. In addition, the compound may be administered all at once, multiple times, or delivered substantially uniformly over a period of time. It should also be noted that the dosage of the compound may vary over time.

The particular administration regimen for a particular subject will depend, in part, on the compound, the amount of compound administered, the route of administration, and the cause and extent of any side effects. The amount of a compound administered to a subject (e.g., a mammal, such as a human) in accordance with the present disclosure should be sufficient to affect the desired response within a reasonable time frame. The dosage will generally depend on the route, time and frequency of administration. Thus, clinicians titrate dosages and vary routes of administration to achieve optimal therapeutic effects, and conventional ranging techniques are known to those of ordinary skill in the art.

Purely by way of example, the method comprises administering, in accordance with the factors described above, for example from about 0.1mg/kg to about 100mg/kg of a compound disclosed herein. In other embodiments, the dosage range is from 1mg/kg to about 100 mg/kg; or 5mg/kg to about 100 mg/kg; or from 10mg/kg to about 100 mg/kg. Some conditions require prolonged treatment, which may or may not result in administration of lower doses of the compound after multiple administrations. If desired, doses of the compound may be administered in two, three, four, five, six or more sub-doses, respectively, at appropriate intervals throughout the day, optionally in unit dosage forms. The treatment period will be as the case may be, and may last from one day to several months.

Suitable methods of administering physiologically acceptable compositions, such as pharmaceutical compositions comprising a compound disclosed herein (e.g., a compound of formula (I)), are well known in the art. Although more than one route may be used to administer a compound, a particular route may provide a more direct and more effective response than another route. The pharmaceutical composition comprising the compound is administered or instilled into a body cavity, absorbed through the skin or mucosa, ingested, inhaled and/or introduced into the circulation as the case may be. For example, in certain instances, it is desirable to deliver a pharmaceutical composition comprising a pharmaceutical agent orally, by injection or by one of the following: intravenous, intraperitoneal, intracerebral (intraparenchymal), intracerebroventricular, intramuscular, intraocular, intraarterial, intraportal, intralesional, intramedullary, intrathecal, intraventricular, transdermal, subcutaneous, intraperitoneal, intranasal, enteral, topical, sublingual, urethral, vaginal, or rectal. The compounds may be administered by a sustained release system or an implanted device.

To facilitate administration, in various aspects, the compounds are formulated in a physiologically acceptable composition that includes a carrier (e.g., vehicle, adjuvant, or diluent). The particular carrier employed is limited by chemical-physical considerations such as inadequate solubility and reactivity with the compound, as well as the route of administration. Physiologically acceptable carriers are well known in the art. Illustrative pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (see, e.g., U.S. Pat. No. 5,466,468). Injectable formulations are further described in the following documents: for example, Pharmacy and pharmaceutical Practice (Pharmacy and pharmaceutical Practice), Leipingtech publishing Co., J.B. Lippincott Co., edited by Bank and Chalmers, pp.238-. In one aspect, a pharmaceutical composition comprising the compound is placed within a container along with packaging material that provides instructions regarding the use of such pharmaceutical composition. Typically, such instructions contain tangible expressions describing the concentration of the agent, and in certain embodiments, the relative amounts of excipient ingredients or diluents (e.g., water, saline, or PBS) that may be required for the recombinant pharmaceutical composition.

Compositions suitable for parenteral injection may include pharmaceutically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, as well as sterile powders for reconstitution in sterile injectable solutions or dispersions. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propylene glycol, polyethylene glycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil), and injectable organic esters, such as ethyl oleate. For example, proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.

These compositions may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. Microbial contamination can be prevented by adding various antibacterial and antifungal agents (e.g., parabens, chlorobutanol, phenol, sorbic acid, and the like). It may also be desirable to include isotonic agents, for example, sugars, sodium chloride and the like. Prolonged absorption of the injectable pharmaceutical composition can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.

Solid dosage forms for oral administration include capsules, tablets, powders and granules. In such solid dosage forms, the active compound is mixed with: at least one inert customary excipient (or carrier), such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, for example starches, lactose, sucrose, mannitol, and silicic acid; (b) binders such as carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, such as glycerol; (d) disintegrating agents, such as agar-agar, calcium carbonate, potato tapioca starch, alginic acid, certain complex silicates and sodium carbonate; (a) solution retarders, such as paraffin; (f) absorption accelerators, such as quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glyceryl monostearate; (h) adsorbents such as kaolin and bentonite; and (i) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate or mixtures thereof. In the case of capsules and tablets, the dosage form may also include buffering agents. Solid compositions of a similar type may also be employed as fillers in soft-filled gelatin capsules and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.

Solid dosage forms (e.g., tablets, dragees, capsules, pills, and granules) can be prepared with coatings and shells such as enteric coatings and other coatings well known in the art. The solid dosage form may also contain opacifying agents. Furthermore, the solid dosage form may be an embedded composition, such that it releases one or more active compounds in certain parts of the intestinal tract in a delayed manner. Examples of embedding compositions which can be used are polymeric substances and waxes. The active compound may also be in microencapsulated form, optionally together with one or more excipients.

Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn germ, olive, castor and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, or mixtures of these substances and the like.

In addition to such inert diluents, the compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.

After formulation, the solution will be administered in a manner compatible with the dosage formulation and in a therapeutically effective amount. The formulations can be readily administered in a variety of dosage forms, such as injectable solutions, drug-releasing capsules, and the like. For example, for parenteral administration in aqueous solution, the solution should be suitably buffered if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are particularly suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.

In the jurisdictions where patenting methods for practicing on the human body is prohibited, the meaning of "administering" a composition to a human subject should be limited to specifying a controlled substance that the human subject can self-administer by any technique (e.g., oral, inhalation, topical administration, injection, insertion, etc.). Is intended to be the broadest reasonable interpretation consistent with the law or law defining the subject matter of the patent. In jurisdictions that do not prohibit patenting of methods practiced on humans, the "administration" of a composition encompasses both the methods practiced on humans and the aforementioned activities.

Application method

The compounds described herein can modulate EGFR. In some embodiments, the compound inhibits EGFR dimerization. In various embodiments, the compound induces EGFR degradation.

Although EGFR has been clearly identified as an important molecular target in oncogenes and cancers, there remains a great need and opportunity for improved methods of modulating the activity of this oncogene. EGFR degradation has also been shown to have a profound effect on cell survival even in TKI resistant cells by using cell penetrating peptides that block dimerization (interferon) or siRNA (14, 23, 25, 26). Without being bound by theory, it is speculated that small molecules that inhibit dimerization are selective for the degradation of active EGFR (abundant in tumors), as most normal cells do not express high levels of EGF/EGFR and thus form symmetric dimers that are not expected to be affected by compound 8C. This approach works even in TKI-resistant tumor cells because the dimeric interface in TKI-resistant tumors remains intact (see fig. 1A and 1B). The methods described herein are unique compared to currently approved therapies. Extensive activity has been demonstrated in preclinical models by degrading the EGFR protein rather than simply inhibiting its kinase activity, while the ability to target tumor tissue is improved since the agent only affects EGFR binding to EGF (which is abundant in tumor cells compared to normal tissue), thereby improving safety and therapeutic range.

Methods that degrade EGFR, rather than simply inhibit its kinase activity, overcome the ongoing resistance to oxitinib in non-small cell lung cancer patients. Although lung cancer is the focus of the present application, there is also an additional significant clinical opportunity in other EGFR-driven cancers (e.g., head and neck, colorectal, and glioblastoma). Thus, targeted selective degradation of oncoproteins in tumors represents a new mechanism that may not only inhibit kinase activity, but that may be applicable to other oncogenic proteins (22, 25, 26, 46).

The compounds disclosed herein are particularly advantageous for treating or preventing diseases or disorders caused by aberrant EGFR activity.

As used herein, "aberrant EGFR activity" refers to activity associated with mutations and overexpression of the Epidermal Growth Factor Receptor (EGFR). Such mutations and overexpression have been implicated in the development of a variety of cancers (Shan et al, Cell (Cell) 2012,149(4) 860-870).

Given the importance of the biological role of EGFR, the compounds of the present disclosure may be useful in a number of applications in a variety of contexts. For example, and most simply, the active agents of the present disclosure can be used to inhibit dimerization of EGFR in a cell. In this regard, the present disclosure provides a method of inhibiting EGFR dimerization in a cell. The method comprises contacting the cell with an amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, effective to inhibit dimerization. In some aspects, the cell is part of an in vitro or ex vivo cell culture or an in vitro or ex vivo tissue sample. In some aspects, the cell is an in vivo cell. In certain embodiments, the methods are intended for research purposes, while in other embodiments, the methods are intended for therapeutic purposes.

Inhibition of EGFR dimerization results in increased EGFR degradation. Accordingly, the present disclosure further provides a method of increasing EGFR degradation in a cell. The method comprises contacting the cell with an amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, effective to increase degradation. In some aspects, the cell is part of an in vitro or ex vivo cell culture or an in vitro or ex vivo tissue sample. In some aspects, the cell is an in vivo cell. In certain embodiments, the methods are intended for research purposes, while in other embodiments, the methods are intended for therapeutic purposes.

As shown herein, compounds that inhibit EGFR dimerization increase tumor cell death. Accordingly, the present disclosure provides a method of increasing tumor cell death in a subject. The method comprises administering to the subject a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, in an amount effective to increase tumor cell death.

In accordance with the foregoing, the present disclosure further provides a method of treating cancer in a subject. The method comprises administering to a subject a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, in an amount effective to treat cancer in the subject.

As used herein, the term "treatment" and words related thereto do not necessarily imply 100% or complete treatment. Rather, there are varying degrees of treatment, one of which is considered by those of ordinary skill in the art to have a potential benefit or therapeutic effect. In this regard, the methods of treating cancer of the present disclosure can provide any number or any level of cancer treatment. Furthermore, the treatment provided by the methods of the present disclosure may comprise treating one or more conditions or symptoms of the cancer being treated. Moreover, the treatment provided by the methods of the present disclosure may encompass slowing the progression of the cancer. For example, the methods can treat cancer by reducing the growth of a tumor or cancer, reducing metastasis of tumor cells, increasing cell death of a tumor or cancer cells, and the like.

The cancer treatable by the methods disclosed herein can be any cancer, for example, any malignant growth or tumor caused by abnormal and uncontrolled cell division, which can spread to other parts of the body through the lymphatic system or the bloodstream. In some embodiments, the cancer is a cancer in which EGFR is expressed by cancer cells. In some aspects, the cancer is a cancer in which EGFR protein is overexpressed, a gene encoding EGFR is amplified, and/or an EGFR mutein (e.g., truncated EGFR, point-mutated EGFR) is expressed.

In some aspects, the cancer is a cancer selected from the group consisting of: acute lymphocytic cancer; acute myeloid leukemia; alveolar rhabdomyosarcoma; bone cancer; brain cancer; breast cancer; anus, anal canal, or anorectal cancer; eye cancer; intrahepatic bile duct cancer; joint cancer; neck, gall bladder or pleural cancer; nasal, nasal or middle ear cancer; oral cancer; vulvar cancer; leukemia (e.g., chronic lymphocytic leukemia); chronic medullary carcinoma; colon cancer; esophageal cancer; cervical cancer; gastrointestinal carcinoids; hodgkin lymphoma; hypopharyngeal carcinoma; kidney cancer; laryngeal cancer; liver cancer; lung cancer; malignant mesothelioma; melanoma; multiple myeloma; nasopharyngeal carcinoma; non-hodgkin lymphoma; ovarian cancer; pancreatic cancer; peritoneal, omental and mesenteric cancers; pharyngeal cancer; prostate cancer; rectal cancer; kidney cancer (e.g., Renal Cell Carcinoma (RCC)); small bowel cancer; soft tissue cancer; gastric cancer; testicular cancer; thyroid cancer; ureteral cancer and bladder cancer. In particular aspects, the cancer is selected from the group consisting of: head and neck cancer, ovarian cancer, cervical cancer, bladder and esophageal cancer, pancreatic cancer, gastrointestinal cancer, gastric cancer, breast cancer, endometrial and colorectal cancer, hepatocellular cancer, glioblastoma, bladder cancer, lung cancer (e.g., non-small cell lung cancer (NSCLC)), bronchoalveolar carcinoma. In a particular aspect, the cancer is an ocitinib-resistant cancer. In some cases, the cancer is pancreatic cancer, head and neck cancer, melanoma, colon cancer, renal cancer, leukemia, or breast cancer. In some cases, the cancer is melanoma, colon cancer, renal cancer, leukemia, or breast cancer.

Also provided herein is the use of a compound disclosed herein in the manufacture of a medicament for modulating EGFR or for treating or preventing a disease or disorder associated with aberrant EGFR activity.

The disclosure herein will be more readily understood by reference to the following examples.

In view of the many possible embodiments to which the principles of this disclosure may be applied, it should be recognized that the illustrated embodiments are only examples and should not be taken as limiting the scope of the invention.

Examples of the invention

Example 1: general procedure a:

Figure BDA0002688847220000241

to 3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5]]To a solution of dec-3-en-2-thione 6 in anhydrous acetonitrile was added acetamides 7A-G (1 equivalent effect). The reaction mixture was warmed to 40 ℃. Next, 2M aqueous potassium carbonate (1 eq) was added to the reaction mixture. The reaction was maintained at 40 ℃ until TLC showed loss of starting material and new RfSpots (typically 2-6 hours). Once the reaction was completed by TLC, it was subjected to post-treatment. The crude reaction mixture was poured into a separatory funnel and ethyl acetate and water were added. The organic layer was separated and then washed with brine (1 ×). The organic layer was then dried over anhydrous MgSO4Dried, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by flash chromatography. The crude product was loaded onto a silica column using a minimum amount of ethyl acetate. The column was placed on top of another pre-equilibrated silica column. The product is usually eluted using a gradient of ethyl acetate/heptane (1-100%) or methanol/dichloromethane (0-10%). The product fractions were concentrated under reduced pressure to give the desired product.

EXAMPLE 2 Synthesis of the Compound 8A (2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) thio) -N- (6-methoxypyridin-3-yl) acetamide)

Following general procedure a, acetamide 7A was used. The crude product was purified by flash chromatography eluting in 10% methanol in dichloromethane to afford the title compound.1H NMR(400MHz,DMSO-d6)10.30(s,1H),8.31(d,J=2.29Hz,1H),7.74-7.86(m,5H),6.78(d,J=8.87Hz,1H),4.14(s,2H),3.79(s,3H),2.50-2.67(m,4H),2.24(s,3H),1.55-1.75(m,4H);MS(ESI+m/z 503.10,ESI-m/z 501.10);TLC:(90:10:0.5,DCM:MeOH:NH4OH)Rf=0.56。

EXAMPLE 3 Synthesis of Compound 8B (2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) thio) -N- (6-chloropyridin-3-yl) acetamide)

Acetamide 7B was used following general procedure a. The crude product was purified by flash chromatography eluting in 10% methanol in dichloromethane to afford the title compound.1H NMR(400MHz,DMSO-d6)10.72(s,1H),8.63(s,1H),8.10(br d,J=9.15Hz,1H),7.85(q,J=8.23Hz,4H),7.53(d,J=8.60Hz,1H),4.24(s,2H),2.56-2.81(m,4H),2.27(br s,3H),1.55-1.85(m,4H);MS(ESI+m/z507.95,ESI-m/z505.95);TLC:(95:5:0.5,DCM:MeOH:NH4OH)Rf=0.17。

EXAMPLE 4 Synthesis of the Compound 8C (2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide)

Acetamide 7C was used following general procedure a. The crude product was purified by flash chromatography eluting in 10% methanol in dichloromethane to afford the title compound.1H NMR(400MHz,DMSO-d6)10.89(br s,1H),8.95(s,1H),8.67(s,1H),7.96(br d,J=8.33Hz,1H),7.92(br d,J=8.33Hz,1H),7.78-7.87(m,4H),7.52-7.72(m,2H),4.29(s,2H),2.43-2.52(m,4H),2.18(br s,3H),1.55-1.85(m,4H);MS(ESI+m/z 523.05,ESI-m/z 521.00);TLC:(90:10:0.5,DCM:MeOH:NH4OH)Rf=0.47。

EXAMPLE 5 Synthesis of the Compound 8D (2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) thio) -N- (pyridin-4-yl) acetamide)

Acetamide 7D was used following general procedure a. The crude product was purified by flash chromatography eluting in 13% methanol in dichloromethane to afford the title compound.1H NMR(400MHz,DMSO-d6)10.66(s,1H),8.41(brd,J=6.13Hz,2H),7.78(q,J=8.51Hz,4H),7.52(d,J=6.04Hz,2H),4.18(s,2H),2.50-2.73(m,4H),2.19(br s,3H),1.28-1.91(m,4H);MS(ESI+m/z 473.05,ESI-m/z 471.05);TLC:(90:10:0.5,DCM:MeOH:NH4OH)Rf=0.24。

EXAMPLE 6 Synthesis of the Compound 8E (2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) thio) -N-cyclohexylacetamide)

Acetamide 7E was used following general procedure a. Following the work-up procedure in general procedure a, the title compound was obtained without column purification.1H NMR(400MHz,DMSO-d6)8.04(br d,J=7.78Hz,1H),7.78(q,J=8.63Hz,4H),3.89(s,2H),3.45-3.55(m,1H),2.50-2.71(m,4H),2.29(s,3H),1.58-1.80(m,7H),1.51(br d,J=12.44Hz,1H),1.05-1.28(m,6H);MS(ESI+m/z 478.90,ESI-m/z476.90);TLC:(90:10:0.5,DCM:MeOH:NH4OH)Rf=0.60。

EXAMPLE 7 Synthesis of the Compound 8F (2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) thio) -N-cyclopentylacetamide)

Acetamide 7F was used following general procedure a. The crude product was purified by flash chromatography eluting in 10% methanol in dichloromethane to afford the title compound.1H NMR(400MHz,DMSO-d6)8.15(br d,J=6.77Hz,1H),7.79(q,J=8.42Hz,4H),3.92-4.01(m,1H),3.91(s,2H),2.50-2.71(m,4H),2.32(s,3H),1.58-1.82(m,8H),1.45-1.54(m,2H),1.38(td,J=6.27,12.35Hz,2H);MS(ESI+m/z 464.10,ESI-m/z 462.15);TLC:(90:10:0.5,DCM:MeOH:NH4OH)Rf=0.62。

EXAMPLE 8 Synthesis of the Compound 8G (N-benzyl-2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) thio) acetamide)

Acetamide 7G was used following general procedure a. The crude product was purified by flash chromatography eluting in 10% methanol in dichloromethane to afford the title compound.1H NMR(400MHz,DMSO-d6)8.65(br t,J=5.81Hz,1H),7.73-7.82(m,4H),7.17-7.30(m,5H),4.27(d,J=5.95Hz,2H),3.99(s,2H),2.50-2.73(m,5H),2.28(s,3H),1.66(br s,3H);MS(ESI+m/z 486.00,ESI-m/z 484.10);TLC:(90:10:0.5,DCM:MeOH:NH4OH)Rf=0.59。

EXAMPLE 9-11 Synthesis of Compound 3-5

Example 9 Synthesis of the Compound 3(4- (2- ((3-chloro-4-methoxyphenyl) amino) -2-oxoethyl) -2- (4-methoxyphenyl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylic acid tert-butyl ester)

To 2- (4-methoxyphenyl) -3-oxo-1, 4, 8-triazaspiro [4.5]]To a solution of tert-butyl dec-1-ene-8-carboxylate, 1 in anhydrous DMF was added sodium hydride (2 equivalents). The reaction mixture was warmed to 40 ℃ for thirty minutes. Next, 2-chloro-N- (3-chloro-4-methoxyphenyl) acetamide hydrochloride, 2(1.76 eq) was added to the reaction mixture. The reaction mixture was maintained at 40 ℃ and under N2Stirring under flowing down. After five hours, the reaction was purified, although TLC indicated that starting material remained. The crude reaction mixture was cooled to room temperature and then loaded onto silica. The dry loaded material was placed in a dry loading column and packed. The dry support column was placed on top of a pre-equilibrated (1% ethyl acetate/heptane) silica gel column. The crude product was purified using flash chromatography, eluting with a gradient of 1-100% ethyl acetate in heptane. The desired fractions were collected (eluted under 50% ethyl acetate) and reduced pressureAnd concentrated to give the title compound.1H NMR(400MHz,CDCl3)8.67(s,1H),8.46(d,J=8.97Hz,2H),7.55(d,J=2.56Hz,1H),7.27-7.39(m,1H),6.99(d,J=8.97Hz,2H),6.84(d,J=8.87Hz,1H),4.01-4.22(m,2H),3.88(s,3H),3.86(s,3H),3.39(m,2H),2.21(dt,J=4.80,12.69Hz,2H),1.50(s,9H),1.19-1.40(m,4H);MS(ESI+m/z 501,ESI-m/z 556.15);TLC:(50:50EA:HEP)Rf=0.21。

EXAMPLE 10 Synthesis of the Compound 4(N- (3-chloro-4-methoxyphenyl) -2- (3- (4-methoxyphenyl) -2-oxo-1, 4, 8-triazaspiro [4.5] dec-3-en-1-yl) acetamide hydrochloride)

To 3,4- (2- ((3-chloro-4-methoxyphenyl) amino) -2-oxoethyl) -2- (4-methoxyphenyl) -3-oxo-1, 4, 8-triazaspiro [4.5]]To a solution of tert-butyl dec-1-ene-8-carboxylate in anhydrous dioxane was added 4M HCl in dioxane (3.3 equivalents). After one hour, no reaction was observed by TLC, so another portion of 4M HCl in dioxane (3.3 eq) was added. After one hour, a new spot (baseline) was observed, but starting material remained. Another portion of 4M HCl in dioxane (3.3 eq) was added and the reaction mixture was stirred at 40 ℃ overnight. Between one night, a yellow precipitate formed in the reaction mixture. The solid was filtered through a sintered funnel (fritted tunnel) and rinsed with excess dioxane. The solid was transferred to a vial and dried under reduced pressure to give the title compound.1H NMR(400MHz,DMSO-d6)10.48(s,1H),9.22(br d,J=9.70Hz,1H),8.93(br d,J=10.70Hz,1H),8.39(d,J=8.97Hz,2H),7.78(d,J=2.47Hz,1H),7.45(dd,J=2.52,9.01Hz,1H),7.07-7.14(m,3H),4.98(br s,2H),4.25(s,2H),3.84(s,3H),3.81(s,3H),3.39-3.48(m,2H),3.25-3.39(m,2H),1.55(br d,J=13.45Hz,2H);MS(ESI+m/z 457.10,ESI-m/z455.10);TLC:(95:5:0.5,DCM:MeOH:NH4OH)Rf=0.03。

EXAMPLE 11 Synthesis of the Compound 5(N- (3-chloro-4-methoxyphenyl) -2- (3- (4-methoxyphenyl) -8-methyl-2-oxo-1, 4, 8-triazaspiro [4.5] dec-3-en-1-yl) acetamide)

To 4, N- (3-chloro-4-methoxyphenyl) -2- (3- (4-methoxyphenyl) -2-oxo-1, 4, 8-triazaspiro [4.5]]Dec-3-en-1-yl) acetamideTo a solution of the hydrochloride in methanol was added aqueous formaldehyde (37 wt% solution, 7 equivalents). The reaction mixture was stirred for 5 minutes, then sodium triacetoxyborohydride (3 equivalents) was added. The reaction mixture was stirred at room temperature overnight. The next morning, TLC (95:5:0.5, DCM: MeOH: NH)4OH) shows reaction completion with a new higher RfAnd (4) speckle. The crude reaction was poured into a separatory funnel. Dichloromethane was added to the separatory funnel. The organic layer was successively saturated NaHCO3Aqueous (1x) and brine (1 x). Subjecting the organic layer to anhydrous Na2SO4Dried, filtered and concentrated under reduced pressure to give the title compound.1H NMR(400MHz,DMSO-d6)10.15(s,1H),8.34(d,J=8.97Hz,2H),7.74(d,J=2.47Hz,1H),7.39(dd,J=2.56,8.97Hz,1H),7.03-7.12(m,3H),4.24(s,2H),3.81(s,3H),3.80(s,3H),2.78(br s,2H),2.29(br s,3H),2.15-2.26(m,2H)1.13-1.33(m,3H);MS(ESI+m/z 471.10);TLC:(95:5:0.5,DCM:MeOH:NH4OH)Rf=0.23

Examples 12-14-Synthesis of Compounds 10, 11 and 12

EXAMPLE 12 Synthesis of the Compound 10(2- (4-methoxyphenyl) -3- ((2- ((6-methoxypyridin-3-yl) amino) -2-oxoethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylic acid tert-butyl ester)

To 2- (4-methoxyphenyl) -3-oxo-1, 4, 8-triazaspiro [4.5]]To a solution of tert-butyl dec-1-ene-8-carboxylate, 1 in anhydrous THF was added lawson's reagent (1 eq). The reaction mixture was stirred at room temperature for 48 hours, then the crude product was purified by flash chromatography. The crude reaction mixture was dry loaded onto silica. The dry loaded material was placed in a dry loading column and packed. The dry support column was placed on top of a pre-equilibrated (2% ethyl acetate/heptane) silica gel column. The crude product was purified using flash chromatography, eluting with a gradient of 2-100% ethyl acetate in heptane. The desired fraction was collected (eluted under 50% ethyl acetate) and concentrated under reduced pressure to give a crude solid. The solid was triturated with a mixture of heptane, dichloromethane and ethyl acetate (3:2:1v/v) and filtered in a sinter funnel to give 92- (4-methoxyphenyl) -3-thio-1, 4, 8-triazaspiro [4.5]Dec-1-ene-8-carboxylic acid tert-butyl ester. MS (ESI + m/z 376.9, ESI-m/z 374.9); TLC (95:0.5:0.5, DCM: MeOH: NH)4OH)Rf=0.68。

Next, to 2- (4-methoxyphenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5]]To a solution of tert-butyl dec-1-ene-8-carboxylate, 9 in anhydrous acetonitrile was added 2-chloro-N- (3-chloro-4-methoxyphenyl) acetamide hydrochloride, 2(1 eq). The reaction mixture was warmed to 40 ℃. Next, 2M aqueous potassium carbonate (2 eq) was added to the reaction mixture. The reaction was continued overnight at 40 ℃ until TLC showed loss of starting material and two new lower R's were observedfAnd (4) speckle. The crude reaction mixture was poured into a separatory funnel. Ethyl acetate and water were added to the funnel. The organic layer was separated and then washed with brine (1 ×). The organic layer was then dried over anhydrous MgSO4Dried, filtered and concentrated under reduced pressure to give the crude product. The crude product was loaded onto a silica column using a minimum amount of dichloromethane. The column was placed on top of another pre-equilibrated silica column. The product was eluted by flash chromatography using a gradient of ethyl acetate/heptane (1-35%). The product fractions were concentrated under reduced pressure to give the title compound.1H NMR(400MHz,DMSO-d6)10.30(s,1H),8.32(d,J=2.38Hz,1H),7.84-7.91(m,3H),7.12(d,J=8.97Hz,2H),6.80(d,J=8.97Hz,2H),4.15(s,2H),3.84(s,3H),3.82(s,3H),3.63-3.72(m,2H),3.47-3.56(m,2H),1.72(br t,J=8.78Hz,2H),1.42(br s,11H);MS(ESI+m/z540.20,ESI-m/z 538.15);TLC:(50:50,EA:Hep)Rf=0.29。

EXAMPLE 13 Synthesis of the Compound 11(2- ((3- (4-methoxyphenyl) -1,4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (6-methoxypyridin-3-yl) acetamide hydrochloride)

To a solution of 10, 2- (4-methoxyphenyl) -3- ((2- ((6-methoxypyridin-3-yl) amino) -2-oxoethyl) thio) -1,4, 8-triazaspiro [4.5]Addition of 4 MHC-containing solution of tert-butyl deca-1, 3-diene-8-carboxylate in anhydrous dioxanel dioxane (3 equivalents). After two hours, no starting material was observed by TLC and a new spot (baseline) was formed. A yellow precipitate formed in the reaction mixture. The solid was filtered through a sintered funnel and rinsed with excess dioxane. The solid was not pure and was therefore loaded onto a silica column using a minimum amount of dichloromethane. The product was eluted by flash chromatography using a gradient of methanol/dichloromethane (0-15%). The product fractions were concentrated under reduced pressure to give the title compound.1H NMR(400MHz,DMSO-d6)10.40(s,1H),8.45-8.60(br s,1H),8.25-8.43(m,1H),7.85-7.93(m,3H),7.12(d,J=8.87Hz,2H),6.80(d,J=8.78Hz,2H),4.18(s,2H),3.84(s,3H),3.81(s,3H),3.22-3.56(m,4H),1.84-2.01(m,2H),1.60-1.75(m,2H);MS(ESI+m/z 440.10,ESI-m/z438.10);TLC:(50:50,EA:Hep)Rf=0.25

EXAMPLE 14 Synthesis of Compound 12(2- ((3- (4-methoxyphenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (6-methoxypyridin-3-yl) acetamide)

To 11, 2- ((3- (4-methoxyphenyl) -1,4, 8-triazaspiro [4.5]]To a solution of deca-1, 3-dien-2-yl) thio) -N- (6-methoxypyridin-3-yl) acetamide hydrochloride in 95:5 dichloromethane in methanol was added aqueous formaldehyde (37 wt% solution, 7 equivalents). The reaction mixture was stirred for one hour, then sodium triacetoxyborohydride (3 equivalents) was added. The reaction mixture was stirred at room temperature overnight. The next morning, TLC (95:5:0.5, DCM: MeOH: NH)4OH) shows reaction completion with a new higher RfAnd (4) speckle. The crude reaction was poured into a separatory funnel. Dichloromethane and water were added to the separatory funnel. The organic layer was separated and washed with brine (1 ×). Subjecting the organic layer to anhydrous Na2SO4Dried, filtered and concentrated under reduced pressure to give the crude product. The crude product is not pure and is therefore loaded onto a silica column using a minimum amount of dichloromethane. The product was eluted by flash chromatography using a gradient of methanol/dichloromethane (0-10%). The product fractions were concentrated under reduced pressure to give the title compound.1H NMR(400MHz,DMSO-d6)10.33(s,1H),8.35(d,J=2.38Hz,1H),7.85-7.91(m,3H),7.12(d,J=8.02Hz,2H),6.81(d,J=9.06Hz,1H),4.16(s,2H),3.85(s,3H),3.82(s,3H),2.53-2.81(m,4H),2.15-2.40(m,2H),1.92(s,3H),1.50-1.80(m,2H);MS(ESI+m/z 454.10,ESI-m/z452.10);TLC:(95:5:0.5,DCM:MeOH:NH4OH)Rf=0.22。

Examples 15 to 28: synthesis of Compounds 33 a-33 l

Figure BDA0002688847220000301

(i) General reaction conditions of (a): to a solution of tert-butyl 2- (4-bromophenyl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) was added THF (0.1M) containing lawson's reagent (0.6 eq) and the reaction mixture was heated to 60 ℃ until the reaction was observed to be complete by TLC. The reaction mixture was concentrated onto silica gel and purified by flash column chromatography (0-100% EtOAc/hexanes) to give 2- (4-bromophenyl) -3-thio-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylic acid tert-butyl ester.

(ii) General reaction conditions of (a): to a solution of tert-butyl 2- (4-bromophenyl) -3-thio-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-3-yl) acetamide (1 eq) and triethylamine (3 eq). The reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organic layer was dried, concentrated and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (4-bromophenyl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate.

(iii) General reaction conditions of (a): to a solution of tert-butyl 2- (4-bromophenyl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate (1 eq) in DCM (0.1M) was added 4M HCl in dioxane (2 eq) and the reaction mixture was stirred at room temperature for 5 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF (0.1M) and triethylamine (5 equivalents) and the appropriate electrophile (3 equivalents) were added followed by nabh (oac)3(3 equivalents), and the reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give the desired product.

Examples 15 to 28

The compound 33a 2- ((3- (4-bromophenyl) -8-isopropyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide. 1H NMR (400MHz, DMSO-d6)10.81(s,1H), 8.97-8.90 (m,1H), 8.00-7.95 (m,1H), 7.95-7.90 (m,1H), 7.89-7.83 (m,2H), 7.83-7.77 (m,2H), 7.71-7.63 (m,1H), 7.62-7.55 (m,1H),4.26(s,2H), 2.73-2.60 (m,4H), 1.88-1.71 (m,2H), 1.62-1.40 (m,2H),0.90(s,3H),0.88(s, 3H).

The compound 33b 2- ((3- (4-bromophenyl) -8-isopropyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide. 1H NMR (400MHz, DMSO-d6)10.80(s,1H),8.94(d, J ═ 2.5Hz,1H),8.67(d, J ═ 2.5Hz,1H), 7.99-7.94 (m,1H), 7.94-7.90 (m,1H), 7.88-7.83 (m,2H), 7.83-7.78 (m,2H), 7.69-7.62 (m,1H), 7.62-7.54 (m,1H),4.26(s,2H), 3.48-3.36 (m,4H), 2.67-2.56 (m,2H), 2.31-2.12 (m,2H), 1.62-1.38 (m,2H), 1.28-1.15 (m,2H),0.81(d, 6.5 Hz), 0.81(d, 6H).

The compound 33c 2- ((8-phenyl-3- (4-bromophenyl) -1,4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) thio) -N- (quinolin-3-yl) acetamide 1H NMR (400MHz, Methanol-d4)8.17(d, J ═ 2.5Hz,1H),7.95(d, J ═ 2.5Hz,1H), 7.25-7.19 (m,1H), 7.12-7.02 (m,3H), 6.96-6.87 (m,3H), 6.85-6.78 (m,1H), 6.43-6.30 (m,3H), 6.30-6.17 (m,2H), 3.42-3.34 (m,2H), 2.76-2.60 (m,4H), 2.10-1.95 (m,2H), 1.95-2H (m,1H), 2H-78H).

Compound 33d 2- ((3- (4-bromophenyl) -8-ethyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) thio) -N- (quinolin-3-yl) acetamide 1H NMR (400MHz, DMSO-d6)10.82(s,1H),8.94(d, J ═ 2.5Hz,1H),8.67(d, J ═ 2.4Hz,1H),7.97(d, J ═ 8.4Hz,1H),7.93(d, J ═ 8.1Hz,1H),7.85(d, J ═ 8.5Hz,2H),7.81(d, J ═ 8.5Hz,2H),7.69 to 7.63(m,1H),7.62 to 7.53(m,1H),4.27(s,2H), 2.81 (d, J ═ 8.5Hz,2H),7.69 to 7.63(m,1H),7.62 to 7.53(m,1H),4.27(s,2H), 3(s,3H), 3(m,3H), 0.50, 3(m, 3-, 3H) in that respect

The compound 33e 2- ((3- (4-bromophenyl) -8-propyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) thio) -N- (quinolin-3-yl) acetamide 1H NMR (400MHz, DMSO-d6)10.80(s,1H),8.94(d, J ═ 2.5Hz,1H),8.67(d, J ═ 2.4Hz,1H), 7.99-7.95 (m,1H),7.93(dd, J ═ 8.1,1.5Hz,1H), 7.88-7.83 (m,2H), 7.83-7.78 (m,2H),7.66(ddd, J ═ 8.4,6.8,1.5Hz,1H),7.58(dd, 8.2, 6.8,1.5Hz,1H), 1.9, 3.9, 2H, 1.6, 2H, 6.5Hz, 1.5Hz,1H, 7.55H, 6.6, 2H (m,2H, 6, 6.6.6, 2H, 6H, 2H, 6, 2H, 6, 2H) 1.63-1.41 (m,2H), 1.40-1.29 (m,2H),0.79(t, J ═ 7.3Hz, 3H).

Compound 33f 2- ((3- (4-bromophenyl) -8-isobutyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) thio) -N- (quinolin-3-yl) acetamide 1H NMR (400MHz, DMSO-d6)10.80(s,1H),8.95(d, J ═ 2.6Hz,1H),8.67(d, J ═ 2.5Hz,1H), 8.00-7.94 (m,1H), 7.94-7.90 (m,1H), 7.87-7.83 (m,2H), 7.83-7.78 (m,2H), 7.69-7.62 (m,1H), 7.62-7.54 (m,1H),4.26(s,2H), 2.65-2.52 (m,4H), 2.06-1.93 (m,2H), 2.93 (m,1H), 1.80 (m,1H), 0.80 (m,1H), j ═ 6.5Hz, 6H).

Compound 33g 2- ((3- (4-bromophenyl) -8- (3,3, 3-trifluoropropyl) -1,4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide 1H NMR (400MHz, DMSO-d6)10.80(s,1H),8.94(d, J ═ 2.5Hz,1H),8.67(d, J ═ 2.4Hz,1H), 8.00-7.95 (m,1H), 7.95-7.91 (m,1H), 7.88-7.83 (m,2H), 7.83-7.78 (m,2H),7.66(ddd, J ═ 8.4,6.9,1.5Hz,1H),7.58(ddd, J ═ 8.1,6.9, 1.5H), 1.68 (ddd, J ═ 8.1,6.9,1, 1.68, 6.6H), 6.96H, 6.54 (m, 1H).

The compound 33H 2- ((3- (4-bromophenyl) -8-cyclopentyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide 1H NMR (400MHz, DMSO-d6)10.82(s,1H),8.96(s,1H),8.68(s,1H), 8.03-7.90 (m,2H), 7.90-7.77 (m,4H), 7.73-7.63 (m,1H), 7.62-7.54 (m,1H),4.25(s,2H), 2.77-2.23 (m,5H), 2.03-0.83 (m, 12H).

Compound 33i 2- ((3- (4-bromophenyl) -8-cyclobutyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) thio) -N- (quinolin-3-yl) acetamide 1H NMR (400MHz, DMSO-d6)10.81(s,1H),8.96(d, J ═ 2.5Hz,1H),8.67(d, J ═ 2.5Hz,1H), 8.00-7.96 (m,1H), 7.95-7.90 (m,1H), 7.88-7.83 (m,2H), 7.83-7.77 (m,2H),7.66(ddd, J ═ 8.4,6.9,1.5Hz,1H),7.58(ddd, J ═ 8.2,6.9,1.4, 1H), 2.25.91 (s,2H), 2.49-7.5H, 1H), 1.49 (m,1H), 1.63-1.10 (m, 4H).

The compound 33J 2- ((3- (4-bromophenyl) -8- ((1-methyl-1H-pyrazol-4-yl) methyl) -1,4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide 1H NMR (400MHz, Chloroform-d)9.88(s,1H), 8.85-8.76 (m,1H),8.65(d, J ═ 2.6Hz,1H), 8.11-8.02 (m,1H), 7.87-7.77 (m,3H), 7.70-7.61 (m,3H),7.56(ddd, J ═ 8.1,6.9,1.2Hz,1H),7.47(d, J ═ 0.7Hz,1H),7.37(s,1H),4.00(s, 3H), 3.91, 2H, 62(s, 3H), 2H) 3.04-2.72 (m,4H), 2.40-2.07 (m,2H),1.78(s, 2H).

The compounds 33k 2- ((3- (4-bromophenyl) -8- ((1-isopropyl-1H-pyrazol-4-yl) methyl) -1,4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide 1H NMR (400MHz, Chloroform-d)9.89(s,1H),8.80(d, J ═ 2.5Hz,1H),8.66(d, J ═ 2.6Hz,1H), 8.08-7.98 (m,1H), 7.87-7.76 (m,3H), 7.69-7.63 (m,3H),7.56(ddd, J ═ 8.2,6.9,1.2Hz,1H),7.50(d, J ═ 0.9Hz,1H),7.44(s, 4H), 4.01(s, 4H), 4.4H, 4H, 2H) 3.62(s,2H), 3.09-2.76 (m,4H), 2.40-2.12 (m,2H),1.74(d, J ═ 35.2Hz,2H),1.53(d, J ═ 6.7Hz, 5H).

The compounds 33l 2- ((3- (4-bromophenyl) -8- (methyl-d 2) -1,4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) thio) -N- (quinolin-3-yl) acetamide 1H NMR (400MHz, DMSO-d6)10.80(s,1H),8.94(d, J ═ 2.5Hz,1H),8.67(d, J ═ 2.4Hz,1H),7.97(dd, J ═ 8.3,1.1Hz,1H),7.93(dd, J ═ 8.1,1.4Hz,1H), 7.89-7.83 (m,2H), 7.83-7.78 (m,2H),7.66(dd, J ═ 8.4,6.9,1.5, 1H),7.58 (m, 9, 9.54, 9.9, 1H), 1.54, 28.9, 9, 9.7.7.7.7.7.7.7.9, 8 ═ 8.9, 8.7.7.7.7.7.7.7.7.7.7.7.7.7., 4H) in that respect

Examples 29 to 34: synthesis of Compounds 34a-34f

Synthesis of compound 34a 2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-4-yl) acetamide

To a solution of tert-butyl 2- (4-bromophenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-4-yl) acetamide (1 eq) and triethylamine (3 eq) and the reaction mixture was stirred for 10 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organics were combined, dried, concentrated and purified by flash column chromatography (0-100% EtOAc in hexanes) to give tert-butyl 2- (4-bromophenyl) -3- ((2-oxo-2- (quinolin-6-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate 1H NMR (400MHz, DMSO-d6)10.65(s,1H), 8.80(dd, J ═ 4.2,1.7Hz,1H), 8.34(d, J ═ 2.4Hz,1H), 8.33-8.27 (m,1H), 8.04-7.92 (m,1H), 7.92-7.76 (m,5H),7.49(dd, J ═ 8.3,4.2Hz,1H), 4.27(s,2H), 3.70-3.70 (m,5H), 3.46(m, 3-5H), 2H) 1.74(d, J ═ 12.6, 8.4, 3.9Hz, 2H), 1.56-1.44(m, 2H),1.37(s, 9H).

To a solution of tert-butyl 2- (4-bromophenyl) -3- ((2-oxo-2- (quinolin-6-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate (1 eq) in DCM was added dioxane (2 eq) containing 4M HCl and the reaction mixture was stirred for 4 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF and formaldehyde (5 eq) and triethylamine (5 eq) were added followed by nabh (oac)3(3 eq) and the reaction mixture was stirred for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give 2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] dec-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-6-yl) acetamide. 1H NMR (400MHz, DMSO-d6)10.67(s,1H),8.80(dd, J ═ 4.2,1.7Hz,1H),8.35(d, J ═ 2.4Hz,1H),8.29(d, J ═ 8.1Hz,1H),7.99(d, J ═ 9.1Hz,1H), 7.89-7.77 (m,5H),7.49(dd, J ═ 8.3,4.2Hz,1H),4.26(s,2H), 2.67-2.54 (m,4H),2.22(s,3H), 1.85-1.40 (m, 4H).

Synthesis of compound 34b 2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-6-yl) acetamide

To a solution of tert-butyl 2- (4-bromophenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-6-yl) acetamide (1 eq) and triethylamine (3 eq) and the reaction mixture was stirred for 10 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organics were combined, dried, concentrated and purified by flash column chromatography (0-100% EtOAc in hexanes) to give tert-butyl 2- (4-bromophenyl) -3- ((2-oxo-2- (quinolin-6-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate 1H NMR (400MHz, DMSO-d6)10.65(s,1H), 8.80(dd, J ═ 4.2,1.7Hz,1H), 8.34(d, J ═ 2.4Hz,1H), 8.33-8.27 (m,1H), 8.04-7.92 (m,1H), 7.92-7.76 (m,5H),7.49(dd, J ═ 8.3,4.2Hz,1H), 4.27(s,2H), 3.70-3.70 (m,5H), 3.46(m, 3-5H), 2H) 1.74(d, J ═ 12.6, 8.4, 3.9Hz, 2H), 1.56-1.44(m, 2H),1.37(s, 9H).

To a solution of tert-butyl 2- (4-bromophenyl) -3- ((2-oxo-2- (quinolin-6-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate (1 eq) in DCM was added dioxane (2 eq) containing 4M HCl and the reaction mixture was stirred for 4 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF and formaldehyde (5 eq) and triethylamine (5 eq) were added followed by nabh (oac)3(3 eq) and the reaction mixture was stirred for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give 2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] dec-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-6-yl) acetamide. 1H NMR (400MHz, DMSO-d6)10.67(s,1H),8.80(dd, J ═ 4.2,1.7Hz,1H),8.35(d, J ═ 2.4Hz,1H),8.29(d, J ═ 8.1Hz,1H),7.99(d, J ═ 9.1Hz,1H), 7.89-7.77 (m,5H),7.49(dd, J ═ 8.3,4.2Hz,1H),4.26(s,2H), 2.67-2.54 (m,4H),2.22(s,3H), 1.85-1.40 (m, 4H).

Synthesis of compound 34c 2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-5-yl) acetamide

To a solution of tert-butyl 2- (4-bromophenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-5-yl) acetamide (1 eq) and triethylamine (3 eq) and the reaction mixture was stirred for 10 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organics were combined, dried, concentrated and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (4-bromophenyl) -3- ((2-oxo-2- (quinolin-5-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate.

To a solution of tert-butyl 2- (4-bromophenyl) -3- ((2-oxo-2- (quinolin-5-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate (1 eq) in DCM was added dioxane (2 eq) containing 4M HCl and the reaction mixture was stirred for 4 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF and formaldehyde (5 eq) and triethylamine (5 eq) were added followed by nabh (oac)3(3 eq) and the reaction mixture was stirred for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give 2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] dec-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-5-yl) acetamide. 1H NMR (400MHz, DMSO-d6)10.41(s,1H),8.93(dd, J ═ 4.2,1.6Hz,1H),8.52(ddd, J ═ 8.6,1.7,0.9Hz,1H), 7.96-7.71 (m,7H),7.57(dd, J ═ 8.6,4.2Hz,1H),4.36(s,2H), 2.75-2.56 (m,4H),2.25(s,3H), 1.89-1.59 (m, 4H).

Synthesis of compound 34d 2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-7-yl) acetamide

To a solution of tert-butyl 2- (4-bromophenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-7-yl) acetamide (1 eq) and triethylamine (3 eq) and the reaction mixture was stirred for 10 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organics were combined, dried, concentrated and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (4-bromophenyl) -3- ((2-oxo-2- (quinolin-7-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate.

To a solution of tert-butyl 2- (4-bromophenyl) -3- ((2-oxo-2- (quinolin-7-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate (1 eq) in DCM was added dioxane (2 eq) containing 4M HCl and the reaction mixture was stirred for 4 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF and formaldehyde (5 eq) and triethylamine (5 eq) were added followed by nabh (oac)3(3 eq) and the reaction mixture was stirred for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give 2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] dec-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-7-yl) acetamide. 1H NMR (400MHz, DMSO-d6)10.69(s,1H),8.85(dd, J ═ 4.2,1.8Hz,1H),8.40(d, J ═ 2.0Hz,1H),8.28(d, J ═ 8.1Hz,1H),7.94(d, J ═ 8.9Hz,1H), 7.91-7.84 (m,2H), 7.83-7.77 (m,2H),7.73(dd, J ═ 8.8,2.1Hz,1H),7.43(dd, J ═ 8.2,4.2Hz,1H),4.28(s,2H), 2.67-2.55 (m,4H),2.20(s,3H), 1.91-1.38 (m, 4H).

Synthesis of compound 34e 2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-8-yl) acetamide

To a solution of tert-butyl 2- (4-bromophenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-8-yl) acetamide (1 eq) and triethylamine (3 eq) and the reaction mixture was stirred for 10 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organics were combined, dried, concentrated and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (4-bromophenyl) -3- ((2-oxo-2- (quinolin-8-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate.

To a solution of tert-butyl 2- (4-bromophenyl) -3- ((2-oxo-2- (quinolin-8-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate (1 eq) in DCM was added dioxane (2 eq) containing 4M HCl and the reaction mixture was stirred for 4 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF and formaldehyde (5 eq) and triethylamine (5 eq) were added followed by nabh (oac)3(3 eq) and the reaction mixture was stirred for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give 2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] dec-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-8-yl) acetamide. 1H NMR (400MHz, DMSO-d6)10.65(s,1H),8.85(dd, J ═ 4.2,1.7Hz,1H),8.60(dd, J ═ 7.6,1.3Hz,1H),8.42(dd, J ═ 8.3,1.7Hz,1H), 7.90-7.79 (m,4H),7.69(dd, J ═ 8.3,1.4Hz,1H),7.64(dd, J ═ 8.3,4.2Hz,1H), 7.62-7.56 (m,1H),4.34(s,2H), 2.50-2.37 (m,4H),2.07(s,3H), 1.86-1.17 (m, 4H).

Synthesis of compound 34f 2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N-methyl-N- (quinolin-3-yl) acetamide

To a solution of tert-butyl 2- (4-bromophenyl) -3-thio-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in DCM (0.1M) was added 2-bromo-N-methyl-N- (quinolin-3-yl) acetamide (1 eq) and triethylamine (3 eq) and the reaction mixture was stirred for 10 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organics were combined, dried, concentrated and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (4-bromophenyl) -3- ((2- (methyl (quinolin-3-yl) amino) -2-oxoethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate.

To a solution of tert-butyl 2- (4-bromophenyl) -3- ((2- (methyl (quinolin-3-yl) amino) -2-oxoethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate (1 eq) in DCM was added 4M HCl in dioxane (2 eq) and the reaction mixture was stirred for 4 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF and formaldehyde (5 eq) and triethylamine (5 eq) were added followed by nabh (oac)3(3 eq) and the reaction mixture was stirred for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give 2- ((3- (4-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] dec-1, 3-dien-2-yl) thio) N-methyl-N- (quinolin-3-yl) acetamide. 1H NMR (400MHz, DMSO-d6)9.03(s,1H),8.51(s,1H),8.10(d, J ═ 8.5Hz,1H),8.00(dd, J ═ 8.4,1.4Hz,1H), 7.89-7.63 (m,6H),4.01(s,2H),3.37(s,3H),3.30(s,3H), 2.65-2.16 (m,7H), 1.89-1.18 (m, 4H).

Examples 35 to 43: synthesis of Compounds 35a-35i

Figure BDA0002688847220000371

Synthesis of the compound 35a 2- ((3- (4-fluorophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide

Methyl amino (4-fluorophenyl) acetate HCl was stirred in 28% ammonium hydroxide (5mL/g) solution for 96 hours, resulting in the formation of 2-amino-2- (4-fluorophenyl) acetamide in the form of a white precipitate, which was collected by filtration and used without further purification. To a solution of 2-amino-2- (4-fluorophenyl) acetamide (1 eq) in ethanol (0.1M) was added tert-butyl 4-oxo-1-piperidinecarboxylate (1 eq) and the reaction mixture was heated to reflux for 12 hours. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The resulting residue was dissolved in DCM (0.1M) and N-bromosuccinimide (1 eq) was added. The reaction mixture was stirred for 8 hours and saturated sodium bicarbonate was added. The resulting mixture was extracted with DCM. The organics were dried, concentrated and purified by FCC (0-100% EtOAc/hexanes) to give tert-butyl 2- (4-fluorophenyl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)10.28(s,1H), 8.48-8.37 (m,2H), 7.41-7.31 (m,2H), 3.71-3.50 (m,4H), 1.79-1.69 (m,2H), 1.68-1.56 (m,2H),1.44(s, 9H).

To a solution of tert-butyl 2- (4-fluorophenyl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in THF (0.1M) was added lawson's reagent (0.6 eq) and the reaction mixture was heated to 60 ℃ until the reaction was observed to be complete by TLC. The reaction mixture was concentrated onto silica gel and purified by flash column chromatography (0-100% EtOAc/hexanes) to give 2- (4-fluorophenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylic acid tert-butyl ester. 1H NMR (400MHz, DMSO-d6)12.60(s,1H),8.38(dd, J ═ 8.5,5.7Hz,2H),7.34(t, J ═ 8.7Hz,2H), 3.80-3.70 (m,2H), 3.60-3.45 (m,2H), 1.86-1.67 (m,4H),1.45(s, 9H).

To a solution of tert-butyl 2- (4-fluorophenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-3-yl) acetamide (1 eq) and triethylamine (3 eq). The reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organic layer was dried, concentrated and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (4-fluorophenyl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate.

To a solution of tert-butyl 2- (4-fluorophenyl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decan-1, 3-diene-8-carboxylate (1 eq) in DCM (0.1M) was added 4M HCl in dioxane (2 eq) and the reaction mixture was stirred at room temperature for 5 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF and triethylamine (5 equivalents) and formaldehyde (3 equivalents) were added followed by nabh (oac)3(3 equivalents) and the reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give 2- ((3- (4-fluorophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide. 1H NMR (400MHz, DMSO-d6)10.80(s,1H),8.94(d, J ═ 2.5Hz,1H), 8.74-8.64 (m,1H), 7.99-7.95 (m,1H), 7.95-7.92 (m,1H), 7.91-7.88 (m,2H), 7.70-7.51 (m,4H),4.27(s,2H),3.34(s,3H), 2.64-2.55 (m,4H), 2.26-2.11 (m,2H), 1.91-1.42 (m, 4H).

Synthesis of compound 35b 2- ((8-methyl-3-phenyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide

Methyl 2-amino-2-phenylacetate hydrochloride was stirred in 28% ammonium hydroxide (5mL/g) solution for 96 hours, resulting in the formation of 2-amino-2-phenylacetamide as a white precipitate, which was collected by filtration and used without further purification. To a solution of 2-amino-2-phenylacetamide (1 eq) in ethanol (0.1M) was added tert-butyl 4-oxo-1-piperidinecarboxylate (1 eq) and the reaction mixture was heated to reflux for 12 hours. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The resulting residue was dissolved in DCM (0.1M) and N-bromosuccinimide (1 eq) was added. The reaction mixture was stirred for 8 hours and saturated sodium bicarbonate was added. The resulting mixture was extracted with DCM. The organics were dried, concentrated and purified by FCC (0-100% EtOAc/hexanes) to give 3-oxo-2-phenyl-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylic acid tert-butyl ester. 1H NMR (400MHz, DMSO-d6)10.26(s,1H), 8.47-8.23 (m,2H), 7.61-7.48 (m,3H), 3.70-3.50 (m,4H), 1.80-1.68 (m,2H), 1.68-1.58 (m,2H),1.44(s, 9H).

To a solution of 3-oxo-2-phenyl-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylic acid tert-butyl ester (1 eq) in THF (0.1M) was added lawson's reagent (0.6 eq) and the reaction mixture was heated to 60 ℃ until the reaction was observed to be complete by TLC. The reaction mixture was concentrated onto silica gel and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2-phenyl-3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)12.58(s,1H), 8.30-8.23 (m,2H), 7.60-7.46 (m,3H), 3.81-3.70 (m,2H), 3.60-3.44 (m,2H), 1.86-1.62 (m,4H),1.45(s, 9H).

To a solution of 2-phenyl-3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylic acid tert-butyl ester (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-3-yl) acetamide (1 eq) and triethylamine (3 eq). The reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organic layer was dried, concentrated and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -3-phenyl-1, 4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate.

To a solution of tert-butyl 2- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -3-phenyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-diene-8-carboxylate (1 eq) in DCM (0.1M) was added 4M HCl in dioxane (2 eq) and the reaction mixture was stirred at room temperature for 5 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF and triethylamine (5 equivalents) and formaldehyde (3 equivalents) were added followed by nabh (oac)3(3 equivalents) and the reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give 2- ((8-methyl-3-phenyl-1, 4, 8-triazaspiro [4.5] dec-1, 3-dien-2-yl) thio) -N- (quinolin-3-yl) acetamide. 1HNMR (400MHz, DMSO-d6)10.81(s,1H),8.94(d, J ═ 2.5Hz,1H),8.67(d, J ═ 2.5Hz,1H), 8.02-7.90 (m,4H),7.66(ddd, J ═ 8.4,6.9,1.5Hz,1H),7.58(ddd, J ═ 8.1,6.8,1.3Hz,1H), 7.49-7.39 (m,2H),4.28(s,2H), 2.75-2.56 (m,4H),2.24(s,3H), 1.99-1.40 (m, 4H).

Synthesis of the compound 35c 2- ((3- (4-chloro-3-fluorophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide

Methyl 2-amino-2- (4-chloro-3-fluorophenyl) acetate hydrochloride was stirred in 28% ammonium hydroxide (5mL/g) solution for 96 hours, resulting in the formation of 2-amino-2- (4-chloro-3-fluorophenyl) acetamide as a white precipitate, which was collected by filtration and used without further purification. To a solution of 2-amino-2- (4-chloro-3-fluorophenyl) acetamide (1 eq) in ethanol (0.1M) was added tert-butyl 4-oxo-1-piperidinecarboxylate (1 eq) and the reaction mixture was heated to reflux for 12 hours. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The resulting residue was dissolved in DCM (0.1M) and N-bromosuccinimide (1 eq) was added. The reaction mixture was stirred for 8 hours and saturated sodium bicarbonate was added. The resulting mixture was extracted with DCM. The organics were dried, concentrated and purified by FCC (0-100% EtOAc/hexanes) to give 2- (4-chloro-3-fluorophenyl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylic acid tert-butyl ester. 1HNMR (400MHz, DMSO-d6)10.37(s,1H),8.29(dd, J ═ 10.4,1.8Hz,1H),8.20(ddd, J ═ 8.4,1.8,0.8Hz,1H),7.78(dd, J ═ 8.4,7.7Hz,1H), 3.69-3.52 (m,4H),1.73(d, J ═ 6.8Hz,4H),1.44(s, 9H).

To a solution of tert-butyl 2- (4-chloro-3-fluorophenyl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in THF (0.1M) was added lawson's reagent (0.6 eq) and the reaction mixture was heated to 60 ℃ until the reaction was complete by TLC. The reaction mixture was concentrated onto silica gel and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (4-chloro-3-fluorophenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)12.58(s,1H), 8.30-8.23 (m,2H), 7.60-7.46 (m,3H), 3.81-3.70 (m,2H), 3.60-3.44 (m,2H), 1.86-1.62 (m,4H),1.45(s, 9H).

To a solution of tert-butyl 2- (4-chloro-3-fluorophenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-3-yl) acetamide (1 eq) and triethylamine (3 eq). The reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organic layer was dried, concentrated and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (4-chloro-3-fluorophenyl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)10.78(s,1H),8.91(d, J ═ 2.5Hz,1H), 8.71-8.61 (m,1H), 8.00-7.95 (m,1H), 7.95-7.91 (m,1H), 7.90-7.83 (m,2H), 7.81-7.76 (m,1H),7.67(ddd, J ═ 8.4,6.9,1.5Hz,1H),7.58(ddd, J ═ 8.2,6.9,1.3Hz,1H),4.29(s,2H), 3.70-3.58 (m,2H), 3.58-3.45 (m,2H), 1.82-1.65 (m,2H), 1.60-1.43 (m,2H), 1.38H (m, 9H).

To 2- (4-chloro-3-fluorophenyl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5]]To a solution of tert-butyl deca-1, 3-diene-8-carboxylate (1 eq) in DCM (0.1M) was added 4M HCl in dioxane (2 eq) and the reaction mixture was stirred at room temperature for 5 hours. The reaction mixture was concentrated. The residue obtained is taken up in THF and triethyl amine is addedAmine (5 equiv.) and Formaldehyde (3 equiv.), followed by addition of NaBH (OAc)3(3 equivalents) and the reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give 2- ((3- (4-chloro-3-fluorophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5]]Dec-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide. 1H NMR (400MHz, DMSO-d6)10.80(s,1H),8.94(d, J ═ 2.5Hz,1H),8.67(d, J ═ 2.5Hz,1H), 7.99-7.95 (m,1H),7.93(d, J ═ 7.7Hz,1H), 7.89-7.82 (m,2H), 7.80-7.75 (m,1H),7.66(ddd, J ═ 8.5,6.9,1.5Hz,1H),7.58(ddd, J ═ 8.1,6.8,1.3Hz,1H),4.29(s,2H), 2.63-2.55 (m,4H),2.20(s,3H), 1.93-1.38 (m, 4H).

Synthesis of compound 35d 2- ((3- (3-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide

Methyl 2-amino-2- (3-bromophenyl) acetate was stirred in 28% ammonium hydroxide (5mL/g) solution for 96 hours, resulting in the formation of 2-amino-2- (3-bromophenyl) acetamide as a white precipitate, which was collected by filtration and used without further purification. To a solution of 2-amino-2- (3-bromophenyl) acetamide (1 eq) in ethanol (0.1M) was added tert-butyl 4-oxo-1-piperidinecarboxylate (1 eq) and the reaction mixture was heated to reflux for 12 hours. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The resulting residue was dissolved in DCM (0.1M) and N-bromosuccinimide (1 eq) was added. The reaction mixture was stirred for 8 hours and saturated sodium bicarbonate was added. The resulting mixture was extracted with DCM. The organics were dried, concentrated and purified by FCC (0-100% EtOAc/hexanes) to give tert-butyl 2- (3-bromophenyl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)10.33(s,1H), 8.54-8.48 (m,1H), 8.39-8.25 (m,1H),7.80(d, J ═ 8.0,2.1,1.1Hz,1H), 7.57-7.44 (m,1H),3.60(d, J ═ 19.5Hz,4H),1.73(d, J ═ 7.4Hz,2H), 1.69-1.55 (m,2H),1.44(s, 9H).

To a solution of tert-butyl 2- (3-bromophenyl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in THF (0.1M) was added lawson's reagent (0.6 eq) and the reaction mixture was heated to 60 ℃ until the reaction was observed to be complete by TLC. The reaction mixture was concentrated onto silica gel and purified by flash column chromatography (0-100% EtOAc/hexanes) to give 2- (3-bromophenyl) -3-thio-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylic acid tert-butyl ester. 1H NMR (400MHz, DMSO-d6)12.63(s,1H), 8.51-8.42 (m,1H),8.27(dt, J ═ 7.8,1.3Hz,1H),7.77(ddd, J ═ 8.0,2.1,1.1Hz,1H), 7.51-7.44 (m,1H), 3.82-3.68 (m,2H), 3.61-3.43 (m,2H), 1.86-1.66 (m,4H),1.45(s, 9H).

To a solution of tert-butyl 2- (3-bromophenyl) -3-thio-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-3-yl) acetamide (1 eq) and triethylamine (3 eq). The reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organic layer was dried, concentrated and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (3-bromophenyl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)10.78(s,1H),8.91(d, J ═ 2.6Hz,1H), 8.70-8.59 (m,1H),8.05(t, J ═ 1.8Hz,1H), 8.00-7.95 (m,1H), 7.95-7.90 (m,2H),7.84(ddd, J ═ 8.1,2.1,1.0Hz,1H),7.67(ddd, J ═ 8.4,6.9,1.5Hz,1H), 7.61-7.54 (m,2H),4.28(s,2H), 3.71-3.58 (m,2H), 3.57-3.46 (m,2H), 1.80-1.68 (m,2H), 1.60-1.42 (m,2H),1.38 (m, 2H).

To a solution of tert-butyl 2- (3-bromophenyl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate (1 eq) in DCM (0.1M) was added 4M HCl in dioxane (2 eq) and the reaction mixture was stirred at room temperature for 5 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF and triethylamine (5 equivalents) and formaldehyde (3 equivalents) were added followed by nabh (oac)3(3 equivalents) and the reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give 2- ((3- (3-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) thio) -N- (quinolin-3-yl) acetamide. 1H NMR (400MHz, DMSO-d6)10.79(s,1H),8.94(d, J ═ 2.5Hz,1H),8.67(d, J ═ 2.5Hz,1H),8.03(t, J ═ 1.8Hz,1H), 7.99-7.93 (m,1H), 7.93-7.88 (m,2H),7.83(ddd, J ═ 8.1,2.1,1.0Hz,1H),7.66(ddd, J ═ 8.4,6.9,1.5Hz,1H), 7.62-7.54 (m,2H),4.28(s,2H),2.59(s,4H),2.21(s,3H), 1.94-1.48 (m, 4H).

Synthesis of the compound 35e 2- ((8-methyl-3- (naphthalen-2-yl) -1,4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide

Methyl methylamino (2-naphthyl) acetate hydrochloride was stirred in 28% ammonium hydroxide (5mL/g) solution for 96 hours resulting in the formation of 2-amino-2- (naphthalen-2-yl) acetamide in the form of a white precipitate, which was collected by filtration and used without further purification. To a solution of 2-amino-2- (naphthalen-2-yl) acetamide (1 eq) in ethanol (0.1M) was added tert-butyl 4-oxo-1-piperidinecarboxylate (1 eq) and the reaction mixture was heated to reflux for 12 hours. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The resulting residue was dissolved in DCM (0.1M) and N-bromosuccinimide (1 eq) was added. The reaction mixture was stirred for 8 hours and saturated sodium bicarbonate was added. The resulting mixture was extracted with DCM. The organics were dried, concentrated and purified by FCC (0-100% EtOAc/hexanes) to give tert-butyl 2- (naphthalen-2-yl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)10.35(s,1H), 9.18-9.11 (m,1H),8.28(dd, J ═ 8.6,1.6Hz,1H), 8.12-7.96 (m,3H), 7.68-7.57 (m,2H), 3.77-3.64 (m,2H), 3.64-3.51 (m,1H), 1.86-1.73 (m,2H), 1.73-1.62 (m,2H),1.45(s, 9H).

To a solution of tert-butyl 2- (naphthalen-2-yl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in THF (0.1M) was added lawson's reagent (0.6 eq) and the reaction mixture was heated to 60 ℃ until the reaction was complete by TLC observation. The reaction mixture was concentrated onto silica gel and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (naphthalen-2-yl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate.

To a solution of tert-butyl 2- (naphthalen-2-yl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-3-yl) acetamide (1 eq) and triethylamine (3 eq). The reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organic layer was dried, concentrated and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (naphthalen-2-yl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)10.80(s,1H),8.93(d, J ═ 2.5Hz,1H), 8.73-8.64 (m,1H), 8.59-8.52 (m,1H), 8.17-8.09 (m,2H), 8.09-7.88 (m,4H), 7.74-7.63 (m,3H), 7.62-7.55 (m,1H),4.32(s,2H),3.68(s,2H), 3.61-3.46 (m,2H), 1.88-1.71 (m,2H), 1.57-1.43 (m,1H),1.39(s, 9H).

To a solution of tert-butyl 2- (naphthalen-2-yl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decan-1, 3-diene-8-carboxylate (1 eq) in DCM (0.1M) was added 4M HCl in dioxane (2 eq) and the reaction mixture was stirred at room temperature for 5 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF and triethylamine (5 equivalents) and formaldehyde (3 equivalents) were added followed by nabh (oac)3(3 equivalents) and the reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give 2- ((8-methyl-3- (naphthalen-2-yl) -1,4, 8-triazaspiro [4.5] dec-1, 3-dien-2-yl) thio) -N- (quinolin-3-yl) acetamide. 1H NMR (400MHz, DMSO-d6)10.79(s,1H),8.94(d, J ═ 2.5Hz,1H),8.67(d, J ═ 2.5Hz,1H), 7.99-7.95 (m,1H), 7.95-7.91 (m,1H), 7.91-7.85 (m,2H),7.66(ddd, J ═ 8.4,6.9,1.5Hz,1H),7.58(ddd, J ═ 8.1,6.8,1.3Hz,1H), 7.16-7.10 (m,2H),4.26(s,2H),3.86(s,3H),3.32(s,3H), 2.65-2.53 (m,4H), 2.24-2.15 (m,2H), 1.29.89-1H (m, 1H).

Synthesis of the compound 35f 2- ((3- (4-methoxyphenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide

Methyl amino (4-methoxyphenyl) acetate hydrochloride was stirred in 28% ammonium hydroxide (5mL/g) solution for 96 hours, resulting in the formation of 2-amino-2- (4-methoxyphenyl) acetamide in the form of a white precipitate, which was collected by filtration and used without further purification. To a solution of 2-amino-2- (4-methoxyphenyl) acetamide (1 eq) in ethanol (0.1M) was added tert-butyl 4-oxo-1-piperidinecarboxylate (1 eq) and the reaction mixture was heated to reflux for 12 hours. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The resulting residue was dissolved in DCM (0.1M) and N-bromosuccinimide (1 eq) was added. The reaction mixture was stirred for 8 hours and saturated sodium bicarbonate was added. The resulting mixture was extracted with DCM. The organics were dried, concentrated and purified by FCC (0-100% EtOAc/hexanes) to give tert-butyl 2- (4-methoxyphenyl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)10.20(s,1H), 8.39-8.29 (m,2H), 7.11-7.01 (m,2H),3.83(s,3H), 3.71-3.48 (m,4H), 1.78-1.67 (m,2H), 1.67-1.54 (m,2H),1.44(s, 9H).

To a solution of tert-butyl 2- (4-methoxyphenyl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in THF (0.1M) was added lawson's reagent (0.6 eq) and the reaction mixture was heated to 60 ℃ until the reaction was observed to be complete by TLC. The reaction mixture was concentrated onto silica gel and purified by flash column chromatography (0-100% EtOAc/hexanes) to give 2- (4-methoxyphenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylic acid tert-butyl ester.

To a solution of tert-butyl 2- (4-methoxyphenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-3-yl) acetamide (1 eq) and triethylamine (3 eq). The reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organic layer was dried, concentrated and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (4-methoxyphenyl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)10.77(s,1H),8.92(d, J ═ 2.5Hz,1H), 8.70-8.61 (m,1H), 7.99-7.93 (m,1H), 7.93-7.87 (m,2H),7.66(ddd, J ═ 8.4,6.9,1.5Hz,1H),7.58(ddd, J ═ 8.1,6.8,1.3Hz,1H), 7.20-7.08 (m,2H),4.27(s,2H),3.86(s,3H), 3.73-3.57 (m,2H), 3.57-3.42 (m,2H), 1.81-1.67 (m,2H), 1.50-1.40 (m,2H),1.37(s, 9H).

To a solution of tert-butyl 2- (4-methoxyphenyl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate (1 eq) in DCM (0.1M) was added 4M HCl in dioxane (2 eq) and the reaction mixture was stirred at room temperature for 5 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF and triethylamine (5 equivalents) and formaldehyde (3 equivalents) were added followed by nabh (oac)3(3 equivalents) and the reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give 2- ((3- (4-methoxyphenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide. 1H NMR (400MHz, DMSO-d6)10.79(s,1H),8.94(d, J ═ 2.5Hz,1H),8.67(d, J ═ 2.5Hz,1H), 7.99-7.95 (m,1H), 7.95-7.91 (m,1H), 7.91-7.85 (m,2H),7.66(ddd, J ═ 8.4,6.9,1.5Hz,1H),7.58(ddd, J ═ 8.1,6.8,1.3Hz,1H), 7.16-7.10 (m,2H),4.26(s,2H),3.86(s,3H), 2.65-2.53 (m,4H),2.20(s,3H), 1.89-1.29 (m, 4H).

Synthesis of the compound 35g 2- ((3- (2-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide

Methyl 2-amino-2- (2-bromophenyl) acetate was stirred in 28% ammonium hydroxide (5mL/g) solution for 96 hours, resulting in the formation of 2-amino-2- (2-bromophenyl) acetamide as a white precipitate, which was collected by filtration and used without further purification. To a solution of 2-amino-2- (2-bromophenyl) acetamide (1 eq) in ethanol (0.1M) was added tert-butyl 4-oxo-1-piperidinecarboxylate (1 eq) and the reaction mixture was heated to reflux for 12 hours. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The resulting residue was dissolved in DCM (0.1M) and N-bromosuccinimide (1 eq) was added. The reaction mixture was stirred for 8 hours and saturated sodium bicarbonate was added. The resulting mixture was extracted with DCM. The organics were dried, concentrated and purified by FCC (0-100% EtOAc/hexanes) to give tert-butyl 2- (2-bromophenyl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)10.23(s,1H), 7.82-7.74 (m,1H), 7.64-7.40 (m,3H), 3.73-3.49 (m,4H), 1.78-1.68 (m,4H),1.44(s, 9H).

To a solution of tert-butyl 2- (2-bromophenyl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in THF (0.1M) was added lawson's reagent (0.6 eq) and the reaction mixture was heated to 60 ℃ until the reaction was observed to be complete by TLC. The reaction mixture was concentrated onto silica gel and purified by flash column chromatography (0-100% EtOAc/hexanes) to give 2- (2-bromophenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylic acid tert-butyl ester. 1H NMR (400MHz, DMSO-d6)12.55(s,1H),7.73(dt, J ═ 7.8,0.9Hz,1H), 7.53-7.40 (m,3H), 3.85-3.72 (m,2H), 3.55-3.43 (m,2H), 1.90-1.70 (m,4H),1.44(s, 9H).

To a solution of tert-butyl 2- (2-bromophenyl) -3-thio-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-3-yl) acetamide (1 eq) and triethylamine (3 eq). The reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organic layer was dried, concentrated and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (2-bromophenyl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)10.78(s,1H),8.90(d, J ═ 2.5Hz,1H),8.65(d, J ═ 2.4Hz,1H), 8.01-7.91 (m,2H),7.83(dd, J ═ 8.2,1.2Hz,1H),7.66(ddd, J ═ 8.4,6.9,1.5Hz,1H), 7.63-7.47 (m,4H),4.23(s,2H), 3.68-3.53 (m,4H), 1.83-1.68 (m,2H), 1.68-1.54 (m,2H),1.40(s, 9H).

To a solution of tert-butyl 2- (2-bromophenyl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate (1 eq) in DCM (0.1M) was added 4M HCl in dioxane (2 eq) and the reaction mixture was stirred at room temperature for 5 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF and triethylamine (5 equivalents) and formaldehyde (3 equivalents) were added followed by nabh (oac)3(3 equivalents) and the reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give 2- ((3- (2-bromophenyl) -8-methyl-1, 4, 8-triazaspiro [4.5] dec-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide. 1H NMR (400MHz, DMSO-d6)10.79(s,1H),8.91(d, J ═ 2.5Hz,1H),8.66(d, J ═ 2.5Hz,1H),7.95(ddd, J ═ 12.8,8.2,1.3Hz,2H), 7.88-7.76 (m,1H),7.66(ddd, J ═ 8.4,6.9,1.5Hz,1H), 7.62-7.46 (m,4H),4.22(s,2H), 2.72-2.53 (m,4H),2.24(s,3H), 1.94-1.64 (m, 4H).

Synthesis of the compound 35h 2- ((8-methyl-3- (4- (trifluoromethyl) phenyl) -1,4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) sulfanyl) -N- (quinolin-3-yl) acetamide

Methyl 2-amino-2- [4- (trifluoromethyl) phenyl ] acetate hydrochloride was stirred in 28% ammonium hydroxide (5mL/g) solution for 96 hours, resulting in the formation of 2-amino-2- (4- (trifluoromethyl) phenyl) acetamide in the form of a white precipitate, which was collected by filtration and used without further purification. To a solution of 2-amino-2- (4- (trifluoromethyl) phenyl) acetamide (1 eq) in ethanol (0.1M) was added tert-butyl 4-oxo-1-piperidinecarboxylate (1 eq) and the reaction mixture was heated to reflux for 12 hours. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The resulting residue was dissolved in DCM (0.1M) and N-bromosuccinimide (1 eq) was added. The reaction mixture was stirred for 8 hours and saturated sodium bicarbonate was added. The resulting mixture was extracted with DCM. The organics were dried, concentrated and purified by FCC (0-100% EtOAc/hexanes) to give 3-oxo-2- (4- (trifluoromethyl) phenyl) -1,4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylic acid tert-butyl ester. 1H NMR (400MHz, DMSO-d6)10.36(s,1H),8.53(d, J ═ 8.1Hz,2H),7.91(d, J ═ 8.1Hz,2H), 3.72-3.52 (m,4H), 1.85-1.60 (m,4H),1.45(s, 9H).

To a solution of 3-oxo-2- (4- (trifluoromethyl) phenyl) -1,4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylic acid tert-butyl ester (1 eq) in THF (0.1M) was added lawson's reagent (0.6 eq) and the reaction mixture was heated to 60 ℃ until the reaction was complete by TLC observation. The reaction mixture was concentrated onto silica gel and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -3- (4- (trifluoromethyl) phenyl) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)12.66(s,1H),8.58(d, J ═ 2.0Hz,1H),8.27(dd, J ═ 8.4,2.0Hz,1H),7.80(d, J ═ 8.4Hz,1H), 3.83-3.71 (m,2H), 3.61-3.45 (m,2H), 1.89-1.66 (m,4H),1.45(s, 9H).

To a solution of 3-thioxo-2- (4- (trifluoromethyl) phenyl) -1,4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylic acid tert-butyl ester (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-3-yl) acetamide (1 eq) and triethylamine (3 eq). The reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organic layer was dried, concentrated and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -3- (4- (trifluoromethyl) phenyl) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)10.80(s,1H), 8.97-8.88 (m,1H), 8.79-8.58 (m,1H), 8.19-8.08 (m,1H), 8.08-7.86 (m,4H), 7.77-7.52 (m,2H),4.29(s,2H), 3.72-3.46 (m,4H), 1.82-1.67 (m,2H), 1.61-1.43 (m,2H),1.38(s, 9H).

To a solution of tert-butyl 2- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -3- (4- (trifluoromethyl) phenyl) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate (1 eq) in DCM (0.1M) was added 4M HCl in dioxane (2 eq) and the reaction mixture was stirred at room temperature for 5 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF and triethylamine (5 equivalents) and formaldehyde (3 equivalents) were added followed by nabh (oac)3(3 equivalents) and the reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give 2- ((8-methyl-3- (4- (trifluoromethyl) phenyl) -1,4, 8-triazaspiro [4.5] decan-1, 3-dien-2-yl) thio) -N- (quinolin-3-yl) acetamide. 1H NMR (400MHz, DMSO-d6)10.80(s,1H),8.94(d, J ═ 2.5Hz,1H),8.67(d, J ═ 2.5Hz,1H),8.09(t, J ═ 1.2Hz,1H), 8.01-7.91 (m,2H), 7.91-7.83 (m,2H),7.67(ddd, J ═ 8.4,6.9,1.5Hz,1H),7.59(ddd, J ═ 8.1,6.8,1.3Hz,1H),4.29(s,2H), 2.70-2.57 (m,4H),2.23(s,3H), 1.98-1.46 (m, 4H).

Synthesis of the compound tert-butyl 35i 2- (3, 4-dichlorophenyl) -3- ((quinolin-3-ylcarbamoyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate

Methyl 2-amino-2- (3, 4-dichlorophenyl) acetate hydrochloride was stirred in 28% ammonium hydroxide (5mL/g) solution for 96 hours, resulting in the formation of 2-amino-2- (3, 4-dichlorophenyl) acetamide as a white precipitate, which was collected by filtration and used without further purification. To a solution of 2-amino-2- (3, 4-dichlorophenyl) acetamide (1 eq) in ethanol (0.1M) was added tert-butyl 4-oxo-1-piperidinecarboxylate (1 eq) and the reaction mixture was heated to reflux for 12 hours. The reaction mixture was cooled to room temperature and concentrated under reduced pressure. The resulting residue was dissolved in DCM (0.1M) and N-bromosuccinimide (1 eq) was added. The reaction mixture was stirred for 8 hours and saturated sodium bicarbonate was added. The resulting mixture was extracted with DCM. The organics were dried, concentrated and purified by FCC (0-100% EtOAc/hexanes) to give tert-butyl 2- (3, 4-dichlorophenyl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate. 1HNMR (400MHz, DMSO-d6)10.37(s,1H),8.55(d, J ═ 1.9Hz,1H),8.29(dd, J ═ 8.4,1.9Hz,1H),7.82(d, J ═ 8.4Hz,1H), 3.70-3.51 (m,4H), 1.81-1.58 (m,4H),1.44(s, 9H).

To a solution of tert-butyl 2- (3, 4-dichlorophenyl) -3-oxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in THF (0.1M) was added lawson's reagent (0.6 eq) and the reaction mixture was heated to 60 ℃ until the reaction was complete by TLC observation. The reaction mixture was concentrated onto silica gel and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (3, 4-dichlorophenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)12.66(s,1H),8.58(d, J ═ 2.0Hz,1H),8.27(dd, J ═ 8.4,2.0Hz,1H),7.80(d, J ═ 8.4Hz,1H), 3.83-3.71 (m,2H), 3.61-3.45 (m,2H), 1.89-1.66 (m,4H),1.45(s, 9H).

To a solution of tert-butyl 2- (3, 4-dichlorophenyl) -3-thioxo-1, 4, 8-triazaspiro [4.5] dec-1-ene-8-carboxylate (1 eq) in DCM (0.1M) was added 2-bromo-N- (quinolin-3-yl) acetamide (1 eq) and triethylamine (3 eq). The reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was diluted with DCM and washed with saturated aqueous sodium bicarbonate. The organic layer was dried, concentrated and purified by flash column chromatography (0-100% EtOAc/hexanes) to give tert-butyl 2- (3, 4-dichlorophenyl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)10.80(s,1H), 8.97-8.88 (m,1H), 8.79-8.58 (m,1H), 8.19-8.08 (m,1H), 8.08-7.86 (m,4H), 7.77-7.52 (m,2H),4.29(s,2H), 3.72-3.46 (m,4H), 1.82-1.67 (m,2H), 1.61-1.43 (m,2H),1.38(s, 9H).

To a solution of tert-butyl 2- (3, 4-dichlorophenyl) -3- ((2-oxo-2- (quinolin-3-ylamino) ethyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate (1 eq) in DCM (0.1M) was added 4M HCl in dioxane (2 eq) and the reaction mixture was stirred at room temperature for 5 hours. The reaction mixture was concentrated. The resulting residue was taken up in THF and triethylamine (5 equivalents) and formaldehyde (3 equivalents) were added followed by nabh (oac)3(3 equivalents) and the reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was concentrated, taken up in EtOAc and washed with saturated aqueous sodium bicarbonate. The organics were dried, concentrated and purified by flash column chromatography (0-30% MeOH/DCM) to give tert-butyl 2- (3, 4-dichlorophenyl) -3- ((quinolin-3-ylcarbamoyl) thio) -1,4, 8-triazaspiro [4.5] decane-1, 3-diene-8-carboxylate. 1H NMR (400MHz, DMSO-d6)10.80(s,1H),8.94(d, J ═ 2.5Hz,1H),8.67(d, J ═ 2.5Hz,1H),8.09(t, J ═ 1.2Hz,1H), 8.01-7.91 (m,2H), 7.91-7.83 (m,2H),7.67(ddd, J ═ 8.4,6.9,1.5Hz,1H),7.59(ddd, J ═ 8.1,6.8,1.3Hz,1H),4.29(s,2H), 2.70-2.57 (m,4H),2.23(s,3H), 1.98-1.46 (m, 4H).

Pharmacokinetic Studies

Example 44 interaction between purified EGFR kinase Domain and Interferon

The WT-EGFR kinase domain (aa696-1022, active kinase) was expressed and purified from SF9 insect cells. 100ng of pure EGFR was incubated with biotin-bound interferon and, after washing with citrate buffer, bound EGFR was captured using CaptAvidin beads. The specificity of this interaction can be confirmed by co-incubating the reaction with more and more non-biotinylated interferons. Bound EGFR protein was released in Laemmli buffer and resolved by SDS pages. Interferon-bound EGFR was quantified using ImageJ software and shown below the blot. Competitive inhibition of interferon-EGFR binding by cold interferon indicates that interferon binds directly to EGFR (see fig. 2B).

Example 45 Effect of Interferon treatment on EGF-induced dimerization in NCI-H1975 cells

NCI-H1975 cells were treated with 10. mu.M interferon for 1 hour, EGF (30ng/ml) for 30 minutes, and disuccinimidyl suberate (DSS, 150. mu.M) for 30 minutes to crosslink interacting proteins. Lysates were prepared and immunoblotted with anti-EGFR antibody. (see FIG. 3A).

Example 46-Effect of Interferon on its target EGFR in NCI-H1975 xenografts

On days 1 and 2, mice carrying NCI-H1975 xenografts were injected with interferon (10mg/kg, i.p.). On day 3, tumors were removed and prepared for immunoblotting (see fig. 3B). The effect of interferon on NCI-H1975 xenografts was evaluated after two consecutive days (monday to tuesday) of treatment with interferon (10mg/kg, i.p.). Tumor volumes were plotted for each treatment condition. Scrambled peptides were used as controls (see fig. 3C). The long-term effects of treatment on tumor histology, EGFR expression and mitotic index (measured by Ki-67 score) were assessed by immunostaining the tumors 2 weeks after treatment (see figure 3D).

Example 47-development of Compound 8C and confirmation of biological Activity

Interferons have been shown to be effective in the TKI (erlotinib) resistant NCI-H1975 xenograft model (see FIGS. 3A-D) (14). However, due to other challenges in poor PK and peptide drug development, a drug discovery program was developed to develop a small molecule with a similar mechanism of action but better pharmacokinetics (see figures 4A-D). Based on the active EGFR dimer interface, using virtual screens, commercial library acquisition and custom synthesis, a series of new molecules were developed, comprising lead compounds 95(C95) and 67(C67) (see fig. 4A-D). Both lead molecules were active in vivo against the erlotinib resistant NCI-H1975 xenograft model. To test the in vivo activity of EGFR dimer inhibition, the TKI resistance NCI-H1975 EGFR reporter was used. These tumor xenografts show induction of bioluminescence after loss of EGFR activity (47). A single injection of 100mg/kg of C95 resulted in a 2.5-fold induction of the EGFR reporter, which lasted 48 hours. (see fig. 9). The effect of C95 treatment on the reduction of EGFR levels was demonstrated to correlate with changes in bioluminescence. These data indicate that drugs that induce EGFR degradation may be effective against TKI resistant tumors.

Compound 8C was approximately 20-fold more stable than its lead (C95) in mouse and human liver microsomes. Efficacy of compound 8C in UMSCC74B (an EGFR-driven head and neck aggressive tumor model). Tumor-bearing mice were treated daily (Monday through Friday) at a dose of 30mg/kg for one week. The dose was selected based on a single dose PK profile (see figure 8A). This treatment was safe and resulted in significant tumor growth delay (see fig. 10).

Ascitic fluid mediated by oxitinib-resistant Ba/F3 cells in nude mice (by intraperitoneal injection of cells) and solid tumor models (by subcutaneous injection of cells) were also used for efficacy studies. An initial study was performed in the ascites model and the effect of compound 8C was compared to the effect of an equivalent dose of oxitinib (30 mg/kg). Although treatment was initiated only after tumor burden became high, a single 30mg/kg dose of compound 8C extended the lifespan of these mice compared to the ocitinib-treated group (p ═ 0.072, data not shown). The effect of treatment on EGFR was confirmed in cells collected from the same mouse before, 18 hours after, and 24 hours after treatment (see fig. 11).

Example 48 direct interaction between EGFR kinase Domain and Compound 8C

To determine whether compound 8C competes for the same binding site on EGFR, biotin-interferon-avidin coupled beads were incubated with purified EGFR at 37 ℃ for 15 minutes, with or without compound 8C. The agarose beads were spun and then non-specific proteins were removed by washing in citrate buffer. Bound proteins were released in Laemmli buffer and resolved by SDS pages. Competitive inhibition of interferon-EGFR binding by compound 8C indicates that the molecule has a similar mechanism of action (see fig. 5A).

The effect of compound 8C on the thermal stability of purified EGFR was confirmed by thermal stability assay. 100ng of purified EGFR was incubated with DMSO or Compound 8C (10. mu.M) for 30 min at 4 ℃. The samples were subjected to heat inactivation (20 to 44 ℃) for 3 minutes. The soluble fraction was separated from the aggregate by centrifugation at 13,000RPM for 10 minutes at 4 ℃. Approximately 20ng of soluble protein was isolated on a 4-12% bis-tris gel and blotted with anti-EGFR antibody. EGFR band intensity quantified and plotted by ImageJ software (see figure 5B). The effect of compound 8C concentration on thermal stability of EGFR at 44 ℃ was determined in the presence of 0 to 10 μ M compound 8C. Soluble fractions of EGFR were quantified and plotted as described above (see figure 5C).

Preliminary data show that compound 8C may bind to the same site on the EGFR molecule as interferon (see fig. 5A) and may thermally stabilize wild-type and axitinib-resistant (C797S) EGFR. As shown in fig. 5B and 5C, incubation of compound 8C with purified wild-type EGFR kinase domain changed the melting curve, indicating that there was an interaction between EGFR and compound 8C. To determine the effect of compound 8C on the axitinib-resistant EGFR mutants, a cell-based assay was used. Oxitinib or Compound 8C was incubated with whole cell lysates prepared from Ba/F3(C797S-EGFR) cells. Aliquots were heated to different temperatures in the presence of DMSO, 10 μ M axitinib, or compound 8C. After cooling, the samples were centrifuged, the soluble protein fraction was collected and immunoblotted with anti-EGFR antibody (see fig. 6A). Axitinib was effective in altering the melting temperature of purified WT-EGFR (see fig. 5B), but had minimal effect on the C797S mutant EGFR protein (see fig. 6A). As expected, compound 8C effectively altered the melting temperature of the axitinib-resistant EGFR. These data indicate that there may be interactions between compound 8C and EGFR even in EGFR-C797S.

Based on these data, it was hypothesized that compound 8C would remain potent in cells resistant to ocitinib due to the C797S mutation. By using these isogenic 6-Ba/F3 cell types (containing TKI-sensitive and drug-resistant cells, Table I), it was demonstrated that Compound 8C remains active in the nanomolar range of each cell line. This is expected from this class of drugs, since the effect of the drug is not associated with EGFR kinase mutations. Inhibition of EGF-induced EGFR dimerization and protein expression by compound 8C was also demonstrated in all of these cells. Data shown are from ocitinib-resistant Ba/F3 cells with the C797S mutation (see fig. 6B-6E).

Example 49-validation of Selective Activity of Compound 8C in Lung cancer cell lines

To test the selectivity of compound 8C, the ocitinib-resistant lung cancer cell lines (PC9-AZR and HCC827-AZR) and normal lung fibroblasts (MRC5) were treated with different concentrations and cell survival was determined using the clonogenic survival assay (see fig. 7A and table 3 below). These preliminary data indicate that compound 8C selectively kills cancer cells driven by EGFR. It was determined whether the response to compound 8C correlated with inhibition of EGF-induced dimerization and EGFR degradation. For this purpose, PC9-AZR cells were treated with 1. mu.M Compound 8C for 1 hour, EGF (30ng/ml) for 30 minutes and disuccinimidyl suberate (DSS, 150. mu.M) for 30 minutes to crosslink the interacting proteins. Lysates were prepared and immunoblotted with anti-EGFR antibody. Figure 7B shows that 1 μ M compound 8C can inhibit EGFR dimerization and induce EGFR degradation. Overall, these results confirm the discovery of Ba/F3 cells.

Example 50 viability assay

In RKO, UM10B, UM1, MCR5 and UMCC92 cells according to the manufacturer's protocol

Figure BDA0002688847220000511

The reagents assess the viability of the cells after treatment. Briefly, 10,000 cells were plated tetraploid into 96-well plates. One day after inoculation, cells were treated with a range of concentrations (0.1 to 30 micromolar). 3 days after treatment, the cells were incubated withThe reagents were incubated together for 4 hours. Only living cells convert redox dyes (reszurin) into fluorescent products (resofurin). The fluorescence emission (excitation 560nM) was measured at 590 nM. IC (integrated circuit)50Values were calculated as the average concentration of compound required to inhibit cell proliferation, as measured by fluorescence at 590nM, which is 50% compared to vehicle-treated controls. The results are presented in tables 3 and 4 below.

TABLE 3

TABLE 4

Example 51-in vivo pharmacokinetics of Compound 8C in tumors and plasma

Short-term PK studies with i.p. injections of the agent showed that compound 8C selectively accumulated in the tumor despite the fact that it had been rapidly cleared from plasma (see fig. 8A and 8B). The peak concentration of compound 8C reached in tumors (. gtoreq.7 hours 69.7. + -. 15.68. mu.M) was much greater than 99% killing of TKI resistantThe concentration required for the tumor cells to be drug-resistant. IC of PC9-AZR and HCC827-AZR50And IC90In the range ofTo

Figure BDA0002688847220000535

(see FIG. 7A). Interestingly, the peak plasma concentration at 30 min was 5.4. + -. 1.5. mu.M. Compound 8C had a plasma half-life of 7.96 hours, but the half-life in tumors was much greater than 24 hours (see figure 8A). Despite rapid systemic clearance, prolonged drug accumulation in tumors is ideal for therapeutic use. Data at the 24 hour time point indicate that the agent may be suitable for daily administration. After observing selective accumulation of compound 8C in tumors injected intraperitoneally, it was determined whether compound 8C was orally available. For oral administration, compound 8C was formulated in PBS (v/v) containing 20% tween80 after brief sonication. Mice received 100mg/kg by oral gavage. The concentration of compound 8C in tumor and plasma samples at 7 hours, 15 hours and 24 hours was determined and plotted in figure 8B.

For intraperitoneal injection, compound 8C was formulated at 10mg/mL in PBS with 5% DMSO by adjusting the pH to 5.5. For oral administration, a homogeneous suspension was prepared in 20% tween80 with brief sonication. Initially, a single dose of 100mg/kg of Compound 8C was administered intraperitoneally (FIG. 8A) or gavage into nude mice carrying human NCI-H1975 tumor xenografts (>150mm3) (FIG. 8B). Mice were euthanized at 0, 30 minutes, 1 hour, 3 hours, 7 hours, 15 hours, and 24 hours. Tumor and plasma samples were collected. In the case of mice with oral gavage, plasma and tumor samples were collected at only 7 hours, 15 hours and 24 hours. The concentration of compound 8C in plasma and tumors was determined and the data obtained was plotted as molar concentration.

Example 52 in vivo validation of EGFR reporter

This method was verified with compound C95. Briefly, once the tumor reaches about 100mm3The mice were imaged to obtain the basal bioluminescence and the effect of the lead compound 95 at different time points (see figure 9A). The change in bioluminescence was quantified and plotted (see fig. 9B). Finally, the effect of treatment on EGFR protein levels was confirmed by immunoblotting 48 hours after treatment.

Example 53 in vivo Activity of Compound 8C

Treatment of UMSCC 74-carrying 74B with (30mg/kg daily for one week) or vehicle (5% DMSO in PBS) () The nude mouse of (1). There were at least 5 mice per group. Tumor volumes and body weights were recorded 3-4 times weekly and the mean tumor volume was plotted against time. The average loss of body weight during treatment was less than 10%. Error bars represent standard error of the mean.

For the compound 8C treatment group, day 0 was defined as the first day of treatment. In vehicle-controlled mice, day 0 was defined as the day in the compound 8C-treated group where the tumor volume was closest to the mean tumor volume on the day of treatment initiation. To assess whether tumor volume growth rate varied from treatment to treatment, a mixed-response model was fitted to a random intercept term at the mouse level to account for the correlation between intratumoral and 2 tumors in mice over time. After one week of single administration, single drug treatment significantly reduced the growth rate of the tumor (see fig. 10). The tumor growth rate was significantly lower in the compound 8C-treated group compared to the time-matched control group (p < 0.013) and the global control group (p < 0.001).

Example 54-effect of compound 8C in an ocitinib-resistant tumor model.

To test the activity of compound 8C against the axitinib-resistant EGFR-driven tumors, an ascites tumor model was developed using Ba/F3-AZR cells (L858R + T790M + C797S-EGFR) as previously reported (65). 500 ten thousand BA/F3-AZR cells were injected by intraperitoneal injection into 6-week-old female nude mice. Mice developed ascites tumors and an average survival time of 20 days was observed. To test the efficacy of compound 8C compared to oxitinib, 15 mice injected with Ba/F3-AZR cells were injected. Mice were randomized into three groups 18 days after tumor cell injection. Mice were treated by intraperitoneal injection with vehicle, single oral dose of either 30mg/kg of ocitinib or 30mg/kg of compound 8C. The health of the mice was monitored and euthanized according to the ULAM final guidelines. Although treatment was initiated after tumor burden became high, a single 30mg/kg dose of compound 8C extended the lifespan of these mice compared to the axitinib-treated group. There was no difference between vehicle control and oxitinib treated groups. The difference between ocitinib and compound 8C was calculated using the log rank test (p ═ 0.072). To test the effect of treatment on the target, tumor cells were collected from one mouse before, 18 hours after, and 24 hours after treatment. Cells were washed with PBS and treated as described in figure 3, and immunoblot results showing the effect of treatment on EGFR, pEGFR and other molecules are given in figure 11.

Example 55-preliminary safety testing of compound 8C in a mouse model.

A preliminary safety test was performed using C57BL6 mice for a 30mg/kg daily dose for one week. The overall health and body weight of a group of 6 mice was monitored during the treatment period. A modest decrease in body weight of 3.5 ± 2.4% was observed after one week of treatment, but mice fully recovered body weight 2 days after cessation of treatment.

Example 56: NCI 60 cell line selection

Compound 8C was tested for activity against 60 different human tumor cell lines at the national cancer institute using standard NCI 60 screening protocols. The percentage growth inhibition of the cell lines performing best is given in table 5 below.

TABLE 5

Group of Cell lines Percentage of growth
Melanoma (MEA) SK-MEL-5 -96.2
Cancer of colon HCT-116 -90.7
Melanoma (MEA) M14 -83.8
Renal cancer 786-0 -81.7
Melanoma (MEA) UACC-62 -78.9
Melanoma (MEA) LOXIMVI -78.5
Cancer of colon COLO205 -76.2
Melanoma (MEA) MALME-3M -75.5
Melanoma (MEA) SK-MEL-28 -70.1
Cancer of colon HT29 -67.1
Leukemia (leukemia) K-562 -61.5
Melanoma (MEA) UACC-257 -61.2
Cancer of colon HCC-2998 -51.3
Breast cancer MDA-MB-468 -43.8
Breast cancer MCF7 -42.7
Leukemia (leukemia) HL-60(TB) -40.7
Breast cancer MDA-MB-231/ATCC -40.5

Example 57: effect of Compound 8C in pancreatic tumor model

KC mice (30mg/kg body weight per day) at 6 weeks of age were treated with Compound 8C by oral gavage. The final effect on PanIn levels was observed compared to control mice that did not receive compound 8C. Pancreas of mice treated with compound 8C showed a significantly reduced tendency to develop PanIn, a pancreatic ductal lesion, as shown in fig. 13.

Example 58: effect of Compound 8C in head and neck tumor models

Mouse xenografts (30mg/kg body weight, twice weekly) of UMSCC74B (head and neck tumor cell line) were treated with compound 8C by oral gavage. The final effect on tumor volume was observed compared to control mice that did not receive compound 8C and control mice that received cetuximab. Mice treated with compound 8C showed significantly reduced tumor volume compared to both controls, as shown in figure 14.

Reference to

Midha et al, J.S. J.cancer Res.) -2015; 5: 2892-911.

Dahabreh et al clinical Cancer research (Clin Cancer Res.) 2010; 16: 291-303.

Rudin et al clinical cancer research 2009; 15: 5646-61.

Balak et al, clinical cancer research 2006; 12: 6494-501.

Yun et al, Proc Natl Acad Sci U S.A. 2008; 105: 2070-5.

Book 2010, Bublil et al, Association of American society for laboratory and biology (Faeb J.); 24: 4744-55.

Soria et al, N Engl J Med 2018; 378: 113-25.

Niederst et al "clinical cancer research" 2015; 21:3924-33.

Mineral et al, Nature medicine (Nat Med.) 2015; 21:560-2.

Book of Oncology of the American medical Association (JAMA Oncol.) 2015, Yu et al; 1:981-3.

2017 in Arula nanda et al Journal of Thoracic Oncology (Journal of clinical Oncology); 12:1728-32.

Jia Y et al, Nature (Nature.) 2016; 534:129-32.

2017, Kong et al, "communication of biochemistry and biophysical research (Biochem Biophys Res Commun.); 488:266-72.

2014 in Ahsan et al, Neoplasia (Neoplasia); 16:105-14.

2013, Ahsan et al, J Biol Chem., or J Biol Chem.; 288:26879-86.

Lichtner et al Cancer Research 2001; 61:5790-5.

17.WO2012087943-A2;US2012190622;WO2012087943;EP2655401;US9029502;US2015218277。

18.WO2014176475

Amador et al cancer research 2004; 64:9139-43.

Wheeler et al, Nature Reviews Clinical Oncology (Nature Reviews Clinical Oncology.) 2010; 7:493-507.

Pao et al, "public science library medicine (PLoS Med.)" 2005; 2: e 73.

Cuneo et al, "pharmacology and therapeutics (Pharmacol Ther.)" 2015; 154:67-77.

2018 in Burslem et al, Cell Chem Biol.; 25:67- +.

Ray et al, "tumor target (Oncotarget.)" 2016.

Raina et al, journal of the national academy of sciences, usa 2016; 113:7124-9.

Book 2016 in Crews et al, J Med Chem; 59:5129-30.

Ray et al tumor formation 2015; 17:697-703.

2015 in Nature Chemical Biology (Nature Chemical Biology), Bondeson et al; 11: 611-U120.

Shukla et al, tumorigenesis 2014; 16:115-28.

Hines et al, journal of the national academy of sciences of the United states, 2013; 110:8942-7.

Ray et al tumor formation 2011; 13:570-8.

Arigiris et al, clinical cancer research 2011; 17:5755-64.

Ahsan et al, cancer research 2010; 70:2862-69.

34, Feng et al, tumor Gene (Oncogene) 2007; 26:3431-9.

Piao et al Cancer Gene therapy (Cancer Gene Ther 2009) 2009; 16:256-65.

2013 in Wang et al, J Gene medicine journal (J Gene Med.); 15:42-50.

Spivakkroizman et al journal of biochemistry 1992; 267:8056-63.

Ewald et al, Experimental cytology research (Exp Cell Res.) 2003; 282:121-31.

Walker et al, molecular Cell Biol 1998; 18:7192-204.

2015 in Coban O et al journal of biophysics (Biophys J.); 108:1013-26.

Book et al, Nature (Nature.))) 2010; 464: 783-U163.

Zhang et al Nature 2007; 450:741-4.

Zhang et al, cytology (Cell.) 2006; 125:1137-49.

Torchilin et al, Drug discovery Today, 2003; 8:259-66.

45.Ripphausen et al, drug development today 2011; 16:372-6.

2012, Corcoran et al Cancer Discovery (Cancer Discovery); 2:227-35.

2011 in Khan et al Analytical Biochemistry (Analytical Biochemistry); 417:57-64.

In the journal of science transformation medicine (Sci Transl Med.) 2011, Chmielecki et al; and 3:90ra 59.

Lovly et al Nature medicine 2014; 20:1027-34.

Ichihara et al cancer research 2017; 77:2990-3000.

Journal of The Association of American pharmaceutical scientists (The AAPS journal), 2011; 13:417-26.

Sun et al journal of pharmaceutical chemistry 2011; 54:3306-18.

53.Cai et al journal of pharmaceutical chemistry 2011; 54:2714-26.

Zheng et al drug metabolism and disposition: chemical biological fate (Drug metabolism and position of chemicals) 2011; 39:627-35.

Fang et al drug metabolism and disposition: biological fate of chemicals 2008; 36:1153-65.

56.Fang et al journal of clinical pharmacology (JClin Pharmacol.) 2007; 47:227-37.

Zou et al Expert opinion of Drug metabolism toxicology (Expert Opin Drug Metab Toxicol.) 2012; 8:855-72.

58.Han et al Cancer communication (Cancer Lett.) 2012; 318:124-34.

Weihua et al Cancer Cell 2008; 13:385-93.

Wei et al cytology 2013; 154:1269-84.

61.Li et al Autophagy (Autophagy) 2010; 6:1066-77.

2017, Leighl et al, "clinical Lung Cancer" (Clin Lung Cancer.); 18:34-42.

63.Pan et al, "public library of sciences in the United states (PLoS One.)" 2015; 10.

2012 in Sangodkar et al J.Clin Invest.); 122:2637-51.

65.Ma et al, J Blood Cancer (Blood Cancer J.) 2013; 3.

66.Morgan et al, "Front of oncology" (Front oncol.) "2017; 7.

61页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:用于治疗与ETBR激活相关的癌症的氘化化合物、组合物和方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!