Substituted pyrimidines, pharmaceutical compositions and methods of treatment

文档序号:1173441 发布日期:2020-09-18 浏览:12次 中文

阅读说明:本技术 取代嘧啶类化合物及其药物组合物和治疗方法 (Substituted pyrimidines, pharmaceutical compositions and methods of treatment ) 是由 陈志宏 于 2018-08-27 设计创作,主要内容包括:本发明提供了对某些酪氨酸激酶(例如布鲁顿酪氨酸激酶(BTK)和/或黏着斑激酶(FAK),细胞外信号调节激酶(ERK))的活性具有抑制作用的新型嘧啶衍生物和类似物,其药物组合物,以及治疗、减轻或预防由酪氨酸激酶介导的某些疾病或障碍的方法,例如癌症、肿瘤、纤维症、炎症性疾病、自身免疫性疾病、糖尿病或免疫介导性疾病。(The present invention provides novel pyrimidine derivatives and analogues having inhibitory effect on the activity of certain tyrosine kinases, such as Bruton's Tyrosine Kinase (BTK) and/or Focal Adhesion Kinase (FAK), extracellular signal-regulated kinase (ERK), pharmaceutical compositions thereof, and methods of treatment, alleviation or prevention of certain diseases or disorders mediated by tyrosine kinases, such as cancer, tumors, fibrosis, inflammatory diseases, autoimmune diseases, diabetes or immune-mediated diseases.)

1. A compound having the structural formula (I):

Figure FDA0002625886830000011

wherein the content of the first and second substances,

x is NH (CH)2)nO or S, n is 0 or 1;

a is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

b is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R1’may be absent, and R1And R1’Each independently (if present) selected from halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, carboxamide, sulfonamide and R1xWherein R is1xSelected from:

Figure FDA0002625886830000012

optionally R1And R1’Is a group containing an electrophilic moiety, optionally R1And R1’Taken together to form a 4-6 membered aliphatic or aromatic cyclic or heterocyclic moiety;

R2and R3Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, or R2And R3Taken together to form a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R4and R5Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, substituted carboxylic acid amide, substituted sulfonamide, or R4And R5Together form a 6-15 membered aliphatic, cyclic or heterocyclic moiety,

or a pharmaceutically acceptable form thereof.

2. A compound of claim 1, wherein R is2And R3Taken together to form a 5-membered ring having the structural formula (II):

Figure FDA0002625886830000021

wherein the content of the first and second substances,

Y1selected from CH, CH2N, NH, O or S;

Y2selected from CH,CH2N, NH, O or S;

Y3selected from CH, CH2N, NH, O or S; and

Y1and Y2The bond between (a) and (b) may be a double or single bond; y is2And Y3The bond between (a) and (b) may be a double or single bond; provided that Y is1、Y2And Y3Is CH or CH2(ii) a And

Y1and Y2A bond between Y and2and Y3At least one of the bonds in (b) is a single bond.

3. The compound of claim 1, wherein B is a six membered aryl group, and the compound has structural formula (III):

4. a compound of claim 3, wherein R is4And R5Meta and para, respectively, having the structural formula (IV):

Figure FDA0002625886830000031

5. a compound according to claim 3 or 4, wherein R is4And R5Together form a 6-15 membered aliphatic, cyclic or heterocyclic moiety.

6. A compound according to any one of claims 3 to 5, wherein R is4And R5Together form a 12-membered aliphatic heterocyclic moiety.

7. The compound of any one of claims 3-6, having structural formula (V-a):

Figure FDA0002625886830000032

8. a compound according to any one of claims 3 to 7, wherein R is2And R3Taken together to form a 5-membered heterocyclic aryl moiety having the structural formula (VI-a):

Figure FDA0002625886830000033

wherein the content of the first and second substances,

Y1selected from CH, CH2N, NH, O or S;

Y2selected from CH, CH2N, NH, O or S;

Y3selected from CH, CH2N, NH, O or S; and

Y1and Y2The bond between (a) and (b) may be a double or single bond; y is2And Y3The bond between (a) and (b) may be a double or single bond; provided that

Y1、Y2And Y3Is CH or CH2(ii) a And

Y1and Y2At least one of the bonds between Y2 and Y3 is a single bond.

9. The compound of any one of claims 1-8, wherein a is a 6-membered aryl moiety having structural formula (VII):

wherein the content of the first and second substances,

z is CH or N; and

R1' and R2Each independently selected from hydrogen, halogen, C1-C6Alkyl, hydroxy or a group containing an electrophilic moiety, provided that R1' and R2At least one of "is a group comprising an electrophilic moiety.

10. The compound of claim 9, wherein Z is CH.

11. The compound of claim 9, wherein Z is N.

12. The compound of any one of claims 1-8, wherein a is a 6-membered aliphatic, cyclic, or heterocyclic moiety having the structural formula (VIII):

Figure FDA0002625886830000051

wherein the content of the first and second substances,

z is CH or N.

13. The compound of claim 12, wherein Z is CH.

14. The compound of claim 12, wherein Z is N.

15. The compound of any one of claims 1-14, wherein a is a 5-membered aliphatic cyclic moiety having structure (IX):

R1' and R2Each independently selected from hydrogen, halogen, C1-C6Alkyl, hydroxy or a group containing an electrophilic moiety, provided that R1' and R2At least one of "is a group comprising an electrophilic moiety.

16. A compound according to any one of claims 1 to 15, wherein the electrophilic or nucleophilic moiety is selected from the group consisting of acrylamide (CH)2=CH-C(=O)-NH-)、-N-S(=O)2-CH=CH2、-N-C(=O)-CH=CH-CH3and-N-S (═ O)2-CH=CH-CH3

17. The compound according to any one of claims 2 and 8 to 16,

Y1is N;

Y2is CH; and

Y3is NH.

18. The compound according to any one of claims 2 and 8 to 16,

Y1is CH;

Y2is N; and

Y3is NH.

19. The compound according to any one of claims 2 and 8 to 16,

Y1is CH;

Y2is CH; and

Y3is NH.

20. The compound according to any one of claims 2 and 8 to 16,

Y1is CH;

Y2is CH; and

Y3is O or S.

21. A compound according to any one of claims 1 to 20 wherein X is NH.

22. A compound according to any one of claims 1 to 20 wherein X is O.

23. A compound according to any one of claims 1 to 20 wherein X is S.

24. The compound according to any one of claims 1-22, selected from:

Figure FDA0002625886830000061

Figure FDA0002625886830000071

25. the compound of claim 1, having the structural formula:

Figure FDA0002625886830000092

26. the compound of claim 1, having the structural formula:

Figure FDA0002625886830000093

27. the compound of claim 1, having the structural formula:

28. the compound of claim 1, having the structural formula:

Figure FDA0002625886830000102

29. the compound of claim 1, having the structural formula:

30. the compound of claim 1, having the structural formula:

Figure FDA0002625886830000104

Figure FDA0002625886830000111

31. the compound of claim 1, having the structural formula:

32. the compound of claim 1, having the structural formula:

33. the compound of claim 1, having the structural formula:

34. the compound of claim 1, having the structural formula:

35. the compound of claim 1, having the structural formula:

Figure FDA0002625886830000122

36. a compound of claim 1, wherein R is1Is a group comprising an electrophilic moiety.

37. A compound of claim 1, wherein R is1Is a group containing a nucleophilic moiety.

38. A pharmaceutical composition comprising an amount of a compound having the structural formula (I):

wherein the content of the first and second substances,

x is NH (CH)2)nO or S, n is 0 or 1;

a is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

b is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

r1 'may be absent and R1 and R1' (if present) are each independently selected from halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, carboxamide, sulfonamide and R1xWherein R is1xSelected from:

optionally R1And R1’At least one of is composed ofRadical of an electrophilic moiety, optionally R1And R1’Taken together to form a 4-6 membered aliphatic or aromatic cyclic or heterocyclic moiety;

R2and R3Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, or R2And R3Taken together to form a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R4and R5Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, substituted carboxylic acid amide, substituted sulfonamide, or R4And R5Together form a 6-15 membered aliphatic, cyclic or heterocyclic moiety, or a pharmaceutically acceptable form thereof, effective for treating, preventing or alleviating one or more diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

39. The pharmaceutical composition of claim 38, effective for treating, preventing or ameliorating cancer or a related disease or disorder.

40. The pharmaceutical composition of claim 38, effective for treating, preventing or ameliorating an inflammatory disease or related disease or disorder.

41. The pharmaceutical composition of claim 38, effective for treating, preventing or ameliorating fibrosis or a related disease or disorder.

42. The pharmaceutical composition of claim 38, effective to treat, prevent or ameliorate an autoimmune disease or related disease or disorder.

43. The pharmaceutical composition of claim 38, effective for treating, preventing or ameliorating an immune-mediated disease or a related disease or disorder.

44. A pharmaceutical composition comprising a compound of any one of claims 1-37.

45. The pharmaceutical composition of claim 38, wherein R is1Is a group comprising an electrophilic moiety.

46. The pharmaceutical composition of claim 38, wherein R is1Is a group containing a nucleophilic moiety.

47. A unit dosage form comprising the pharmaceutical composition of any one of claims 38-46.

48. A method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound of formula (I):

wherein the content of the first and second substances,

x is NH (CH)2)nO or S, n is 0 or 1;

a is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

b is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R1’may be absent, and R1And R1’Each, if present, is independently selected from halogen, C1-C6 alkyl, C1-C6 alkoxy, cyano, hydroxy, amino, carboxamide, sulfonamide and R1xWherein R is1xSelected from:

optionally R1And R1’At least one of is composed ofRadical of an electrophilic moiety, optionally R1And R1’Taken together to form a 4-6 membered aliphatic or aromatic cyclic or heterocyclic moiety;

R2and R3Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, or R2And R3Taken together to form a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R4and R5Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, substituted carboxylic acid amide, substituted sulfonamide, or R4And R5Together forming a 6-15 membered aliphatic, cyclic or heterocyclic moiety, or a pharmaceutically acceptable form thereof, effective for treating, preventing or alleviating one or more cancers, inflammatory diseases, fibrosis, autoimmune diseases or immune-mediated diseases or related diseases or disorders in a mammal (including a human), and a pharmaceutically acceptable excipient, carrier or diluent.

49. The method of claim 48, effective to treat, prevent or ameliorate cancer or a related disease or disorder.

50. The method of claim 48, effective to treat, prevent or ameliorate an inflammatory disease or related disease or disorder.

51. The method of claim 48, effective to treat, prevent or ameliorate fibrosis or a related disease or disorder.

52. The method of claim 48, effective to treat, prevent or ameliorate an inflammatory disease or related disease or disorder.

53. The method of claim 48, effective to treat, prevent or ameliorate an immune-mediated disease or related disease or disorder.

54. A method for treating, alleviating or preventing a disease or disorder comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound of any one of claims 1-37, wherein the disease or disorder is cancer, an inflammatory disease, fibrosis, an autoimmune disease or an immune-mediated disease or related disease or disorder.

55. A method for treating, alleviating or preventing a disease or disorder comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having an inhibitory effect on the activity of Bruton's Tyrosine Kinase (BTK) and/or Focal Adhesion Kinase (FAK).

56. The method of claim 55, wherein the compound is a BTK inhibitor.

57. The method of claim 55, wherein the compound is a bone marrow tyrosine kinase (BMX) inhibitor.

58. The method of claim 56 or 57, wherein the compound is a BTK and BMX inhibitor.

59. The method of claim 55 or 58, wherein the compound is an interleukin-2 inducible T-cell kinase (ITK) inhibitor.

60. The method of claim 55, wherein said compound is a FAK inhibitor.

61. The method of claim 60, wherein the compound is a BTK and FAK inhibitor.

62. The method of any one of claims 55-61, wherein the compound is a BTK (C481S) mutant inhibitor.

63. The method of any one of claims 55-62, wherein the compound is an extracellular signal-regulated kinase (ERK) inhibitor.

64. The method of any one of claims 55-63, wherein the compound has the structural formula (I):

Figure FDA0002625886830000161

wherein the content of the first and second substances,

x is NH (CH)2)nO or S, n is 0 or 1;

a is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

b is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R1’may be absent, and R1And R1’Each independently (if present) selected from halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, carboxamide, sulfonamide and R1xWherein R is1xSelected from:

optionally R1And R1’Is a group containing an electrophilic moiety, optionally R1And R1’Taken together to form a 4-6 membered aliphatic or aromatic cyclic or heterocyclic moiety;

R2and R3Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, or R2And R3Taken together to form a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R4and R5Each is independentIs selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, substituted carboxylic acid amide, substituted sulfonamide, or R4And R5Together form a 6-15 membered aliphatic, cyclic or heterocyclic moiety, or a pharmaceutically acceptable form thereof.

65. The method of claim 64, wherein R is1Is a group comprising an electrophilic moiety.

66. The method of claim 64, wherein R is1Is a group containing a nucleophilic moiety.

67. A method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

68. A method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

69. A method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure FDA0002625886830000181

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

70. A method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

71. A method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure FDA0002625886830000191

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

72. A method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

73. A method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure FDA0002625886830000193

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

74. A method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

75. A method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

76. A method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure FDA0002625886830000203

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

77. A method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

78. Use of a compound according to any one of claims 1-37 and a pharmaceutically acceptable excipient, carrier or diluent in the manufacture of a medicament for the treatment of a disease or disorder.

79. The use of claim 78, wherein the disease or disorder is cancer, an inflammatory disease, fibrosis, an autoimmune disease, or an immune-mediated disease, or a related disease or disorder.

Technical Field

The present invention relates generally to novel compounds and their therapeutic uses. In particular, the present invention provides novel pyrimidine derivatives and analogues having inhibitory effects on the activity of certain tyrosine kinases, such as Bruton's Tyrosine Kinase (BTK) and/or Focal Adhesion Kinase (FAK), extracellular signal-regulated kinase (ERK), pharmaceutical compositions thereof, and methods of treating, alleviating or preventing certain diseases or disorders mediated by tyrosine kinases, such as cancer, inflammatory diseases, fibrosis, autoimmune diseases or immune-mediated diseases.

Background

Cancer is a group of diseases involving abnormal cell growth, with the potential to invade or spread to other parts of the body. There are over 100 kinds of cancers harmful to human beings. In 2015, over 9000 million people with cancer develop about 1400 million new cases each year, resulting in about 15% of deaths. The most common types of cancer include lung, prostate, colorectal and gastric cancer in men, and breast, colorectal, lung and cervical cancer in women. Despite many therapeutic approaches, such as surgery, chemotherapy, radiation therapy, hormonal therapy, targeted therapy and palliative therapy, cancer remains the biggest health threat to humans.

Inflammatory diseases are a large group of diseases that underlie a variety of human diseases. Inflammation is a protective response of the human body to injury and infection, and is a complex process involving multiple cell types and different blood components. The inflammatory process rapidly destroys and eliminates foreign and damaged cells and isolates infected or injured tissue from the rest of the body. When inflammation becomes uncontrolled, inflammatory diseases occur and healthy tissue is destroyed. When the immune system falsely triggers an inflammatory response in the absence of infection, many inflammatory diseases arise, such as joint inflammation in rheumatoid arthritis. Still other inflammatory diseases are responsive to tissue injury or trauma, but affect the entire body. Overall, the prevalence of immune-mediated inflammatory diseases is estimated to be 5% -7% in western society.

The therapies and means currently available for treating diseases or disorders, such as cancer, tumors, fibrosis, inflammatory diseases, autoimmune diseases or immune-mediated diseases, are inadequate. There remains an urgent need for new and improved therapies to effectively address these diseases and disorders.

Disclosure of Invention

The present invention is based, in part, on the unexpected discovery of novel pyrimidine derivatives and analogs that exhibit inhibitory activity against certain tyrosine kinases, such as Bruton's Tyrosine Kinase (BTK) and/or Focal Adhesion Kinase (FAK), and pharmaceutical compositions thereof. The pyrimidine derivatives disclosed herein have been shown to effectively inhibit BTK and/or other non-receptor tyrosine kinase TEC families, such as bone marrow tyrosine kinase (BMX) or interleukin-2 inducible T cell kinase (ITK). In addition, certain pyrimidine derivatives are effective in inhibiting drug resistance mutations BTK (C481S), FAK, and BTK. In addition, some of the pyrimidine derivatives disclosed in this application are effective in inhibiting drug resistance mutations BTK (C481S), FAK and extracellular signal-regulated kinase (ERK), as well as BTK.

Also disclosed herein are methods of treating, alleviating, or preventing certain diseases or disorders mediated by tyrosine kinases, such as BTK, BTK (C481S), FAK, ERK kinase, and/or other TEC family kinases. Such diseases and disorders include cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes, or immune-mediated diseases.

In one aspect, the invention generally relates to a compound having the structural formula (I):

wherein the content of the first and second substances,

x is NH (CH)2)nO or S, n is 0 or 1;

a is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

b is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R1’may be absent, and R1And R1’Each independently (if present) selected from halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, carboxamide, sulfonamide and R1xWherein R is1xSelected from:

Figure BDA0002467331340000022

optionally R1And R1’Is a group containing an electrophilic moiety, optionally R1And R1’Taken together to form a 4-6 membered aliphatic or aromatic cyclic or heterocyclic moiety;

R2and R3Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, or R2And R3Taken together to form a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R4and R5Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, substituted carboxylic acid amide, substituted sulfonamide, or R4And R5Together form a 6-15 membered aliphatic, cyclic or heterocyclic moiety,

or a pharmaceutically acceptable form thereof.

In another aspect, the invention relates generally to a pharmaceutical composition comprising an amount of a compound having the structural formula (I):

Figure BDA0002467331340000031

wherein the content of the first and second substances,

x is NH (CH)2)nO or S, n is 0 or 1;

a is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

b is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R1’may be absent, and R1And R1’Each independently (if present) selected from halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, carboxamide, sulfonamide and R1xWherein R is1xSelected from:

Figure BDA0002467331340000032

optionally R1And R1’Is a group containing an electrophilic moiety, optionally R1And R1’Taken together to form a 4-6 membered aliphatic or aromatic cyclic or heterocyclic moiety;

R2and R3Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, or R2And R3Taken together to form a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R4and R5Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, substituted carboxylic acid amide, substituted sulfonamide, or R4And R5Together form a 6-15 membered aliphatic, cyclic or heterocyclic moiety,

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention generally relates to a unit dosage form comprising the disclosed pharmaceutical composition.

In another aspect, the invention relates generally to a method for treating, reducing or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the structural formula (I):

wherein the content of the first and second substances,

x is NH (CH)2)nO or S, n is 0 or 1;

a is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

b is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R1’may be absent, and R1And R1’Each independently (if present) selected from halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, carboxamide, sulfonamide and R1xWherein R is1xSelected from:

Figure BDA0002467331340000042

optionally R1And R1’Is a group containing an electrophilic moiety, optionally R1And R1’Taken together to form a 4-6 membered aliphatic or aromatic cyclic or heterocyclic moiety;

R2and R3Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, or R2And R3Taken together to form a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R4and R5Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, substituted carboxylic acid amide, substituted sulfonamide, or R4And R5Together form a 6-15 membered aliphatic, cyclic or heterocyclic moiety,

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In another aspect, the invention relates generally to a method of treating, ameliorating or preventing a disease or disorder. The method comprises administering to a subject in need thereof a pharmaceutical composition disclosed herein, wherein the disease or disorder is selected from cancer, an inflammatory disease, a fibrosis, an autoimmune disease or an immune-mediated disease or a related disease or disorder.

In another aspect, the invention relates generally to a method of treating, ameliorating or preventing a disease or disorder. The method comprises administering to a subject in need thereof a pharmaceutical composition comprising a compound having an inhibitory effect on the activity of Bruton's Tyrosine Kinase (BTK) and/or Focal Adhesion Kinase (FAK). In certain method embodiments, the compound has the structural formula (I):

Figure BDA0002467331340000051

wherein the content of the first and second substances,

x is NH (CH)2)nO or S, n is 0 or 1;

a is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

b is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R1’may be absent, and R1And R1’Each independently (if present) selected from halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, carboxamide, sulfonamide and R1xWherein R is1xSelected from:

Figure BDA0002467331340000052

optionally R1And R1’Is a group containing an electrophilic moiety, optionally R1And R1’Taken together to form a 4-6 membered aliphatic or aromatic cyclic or heterocyclic moiety;

R2and R3Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, or R2And R3Taken together to form a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R4and R5Each independently selected from hydrogen, halogen、C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, substituted carboxylic acid amide, substituted sulfonamide, or R4And R5Together form a 6-15 membered aliphatic, cyclic or heterocyclic moiety,

or a pharmaceutically acceptable form thereof.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000062

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000072

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000081

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000083

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000091

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000092

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the invention relates generally to the use of a compound disclosed herein and a pharmaceutically acceptable excipient, carrier or diluent in the manufacture of a medicament for the treatment of a disease or disorder.

Drawings

The above and other objects and features of the present invention will become apparent from the following description taken in conjunction with the accompanying drawings. The attached drawings are respectively as follows:

FIG. 1 schematically shows Western blot analysis of p-BTK (Y223) in Ramos cells.

FIG. 2 exemplarily shows a BTK IC50And (4) data.

FIG. 3 exemplarily shows a BTK (C481S) IC50And (4) data.

FIG. 4 schematically shows a BMX IC50And (4) data.

FIG. 5 schematically shows an ITK IC50And (4) data.

FIG. 6 exemplarily shows FAK IC50And (4) data.

FIG. 7 exemplarily shows an ERK IC50And (4) data.

Definition of

Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. "Compound" as presented herein means "Compound". General principles of organic chemistry, as well as specific functional motifs and reactions, are described in organic chemistry, thomas sorel, university science book, soxhlet 2006.

Certain compounds of the present invention may exist in specific geometric or stereoisomeric forms. Such compounds, including cis and trans isomers, R-and S-enantiomers, diastereomers, (d) -isomers, (l) -isomers, racemic and other mixtures thereof, contemplated by the present invention are within the scope of the present invention. Additional asymmetric carbon atoms may be present in a substituent, such as an alkyl group. All such isomers and mixtures thereof are included in the present invention.

Mixtures of isomers containing any of a variety of isomer ratios may be used in accordance with the present invention. For example, where only two isomers are combined, the present invention contemplates mixtures comprising ratios of 50: 50, 60: 40, 70: 30, 80: 20, 90: 10, 95: 5, 96: 4, 97: 3, 98: 2, 99: 1, or 100: 0 isomers. One of ordinary skill in the art will readily appreciate that similar ratios can be considered for more complex isomer mixtures.

For example, if a particular enantiomer of a compound of the invention is desired, it may be prepared by asymmetric synthesis or by using chiral auxiliary derivatives, the resulting diastereomeric mixture separated, and the auxiliary group cleaved to provide the pure desired enantiomer. Alternatively, where the molecule contains a basic functionality, such as an amino group, or an acidic functionality, such as a carboxyl group, diastereomeric salts are formed with an appropriate optically active acid or base, followed by resolution of the resulting diastereomers by fractional crystallization or chromatography, as is well known in the art, to recover the pure enantiomers.

Solvates and polymorphs of the compounds of the disclosure are also contemplated by the present invention. Solvates of the compounds of the invention include, for example, hydrates.

The definitions of specific functional groups and chemical terms are described in more detail below. The recitation of range values is intended to include each value and sub-range subsumed within that range. For example, "C1-6 alkyl" is meant to include C1、C2、C3、C4、C5、C6、C1-6、C1-5、C1-4、C1-3、C1-2、C2-6、C2-5、C2-4、C2-3、C3-6、C3-5、C3-4、C4-6、C4-5And C5-6An alkyl group.

As used herein, the term "alkyl" refers to a straight or branched chain group consisting of only carbon and hydrogen atoms, free of unsaturation, having 1 to 10 carbon atoms (e.g., C)1-10Alkyl groups). Herein, numbers such as "1 to 10" refer to each integer within a given range; for example, "1 to 10 carbon atoms" means that the alkyl group can be made up of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, up to and including 10 carbon atoms, and the present definition also encompasses the term "alkyl" where no numerical range is specified. In some embodiments, "alkyl" may be C1-6An alkyl group. In some embodiments, the alkyl group has 1 to 10, 1 to 8, 1 to 6, or 1 to 3 carbon atoms. Representative saturated straight chain alkyl groups include, but are not limited to, -methyl, -ethyl, -n-propyl, -n-butyl, -n-pentyl, and-n-hexyl; and saturated branched alkyl groups include, but are not limited to, -isopropyl, -sec-butyl, -isobutyl, -tert-butyl, -isopentyl, 2-methylbutyl, 3-methylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl, 5-methylhexyl, 2, 3-dimethylbutyl, and the like. The alkyl group is attached to the parent molecule by a single bond. Unless otherwise indicated in the specification, an alkyl group may be optionally substituted with one or more substituents independently including acyl, alkyl, alkenyl, alkynyl, alkoxy, alkaryl, cycloalkyl, aralkyl, aryl, aryloxy, amino, amido, amidino, imino, azide, carbonate, carbamate, carbonyl, heteroalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, hydroxy, cyano, halogen (F, Cl, Br, I), haloalkoxy, haloalkyl, ester, ether, mercapto, thio, alkylthio, arylthio, thiocarbonyl, nitro, oxo, phosphate, phosphonate, phosphinate, silyl, sulfinyl, sulfonyl, sulfonamido, synergistic sulfone, sulfonate, urea, -Si (R, alkyl, aryl, aryloxy, aryl, heteroaryl, amino, nitro, carbonyl, amino, sulfonyl, phosphonite, amino, sulfonyl, urea, or aminoa)3、-ORa、-SRa、-OC(O)-Ra、-N(Ra)2、-C(O)Ra、-C(O)ORa、-OC(O)N(Ra)2、-C(O)N(Ra)2、-N(Ra)C(O)ORa、-N(Ra)C(O)Ra、-N(Ra)C(O)N(Ra)2、-N(Ra)C(NRa)N(Ra)2、-N(Ra)S(O)tN(Ra)2(wherein t is 1 or 2), -P (═ O) (R)a)(Ra) OR-O-P (═ O) (OR)a)2Wherein each R isaIndependently is hydrogen, alkyl, haloalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl, or heteroarylalkyl, and each of these moieties may be optionally substituted as defined herein. In non-limiting examples, substituted alkyl groups may be selected from fluoromethyl, difluoromethyl, trifluoromethyl, 2-fluoroethyl, 3-fluoropropyl, hydroxymethyl, 2-hydroxyethyl, 3-hydroxypropyl, benzyl, and phenethyl.

As used herein, the term "alkoxy" refers to a straight, branched, saturated cyclic configuration of an-O-alkyl group, including from 1 to 10 carbon atoms (C)1-10) And combinations thereof. Examples include methoxy, ethoxy, propoxy, isopropoxy, butoxy, tert-butoxy, pentyloxy, cyclopropyloxy, cyclohexyloxy and the like. "lower alkoxy" refers to alkoxy groups containing 1 to 6 carbons. In some embodiments, C1-3Alkoxy refers to alkoxy groups of straight and branched chain alkyl groups containing 1 to 3 carbon atoms. Unless otherwise indicated in the specification, an alkoxy group may be optionally substituted with one or more substituents independently including acyl, alkyl, alkenyl, alkynyl, alkoxy, alkaryl, cycloalkyl, aralkyl, aryl, aryloxy, amino, amido, amidino, imino, azide, carbonate, carbamate, carbonyl, heteroalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, hydroxy, cyano, halogen, haloalkoxy, haloalkyl, ester, ether, mercapto, thio, alkylthio, arylthio, thiocarbonyl, nitro, oxo, phosphate, phosphonate, phosphinate, silyl, sulfinyl, sulfonyl, sulfonamido, or the likeSynergistic sulfones, sulfonates, ureas, -Si (R)a)3、-ORa、-SRa、-OC(O)-Ra、-N(Ra)2、-C(O)Ra、-C(O)ORa、-OC(O)N(Ra)2、-C(O)N(Ra)2、-N(Ra)C(O)ORa、-N(Ra)C(O)Ra、-N(Ra)C(O)N(Ra)2、-N(Ra)C(NRa)N(Ra)2、-N(Ra)S(O)tN(Ra)2(wherein t is 1 or 2), -P (═ O) (R)a)(Ra) OR-O-P (═ O) (OR)a)2Wherein each R isaIndependently is hydrogen, alkyl, haloalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl, or heteroarylalkyl, and each of these moieties may be optionally substituted as defined herein.

As used herein, the term "aromatic" or "aryl" refers to an atomic group having 6 to 14 ring atoms (e.g., C)6-14Aromatic or C6-14Aryl) having at least one ring with a carbocyclic conjugated pi-electron system (e.g., phenyl, fluorenyl, and naphthyl). In some embodiments, aryl refers to C6-10And (4) an aryl group. For example, a divalent group formed from a substituted benzene derivative and having a free valence at a ring atom is referred to as a substituted phenylene group. In other embodiments, divalent groups derived from monovalent polycyclic hydrocarbon groups (named by removing one hydrogen atom from a carbon atom having a free valence, ending with a "-yl") are named by adding a "-ylidene" to the name of the corresponding monovalent group, e.g., naphthyl having two points of attachment is referred to as dihydronaphthylene. As used herein, numerical ranges such as "6 to 14 aryl" refer to each integer within the given range; for example, "6 to 14 ring atoms" means that the aryl group may consist of 6 ring atoms, 7 ring atoms, etc., up to and including 14 ring atoms. The term includes monocyclic or fused-ring polycyclic (i.e., rings that share adjacent pairs of atoms) groups. Polycyclic aryl groups include bicyclic, tricyclic, tetracyclic, and the like. In polycyclic radicals, only one ring is aromatic, and soAryl definitions include indanyl and like groups. Non-limiting examples of aryl groups include phenyl, propenaphthyl, naphthyl, tetrahydronaphthyl, phenanthryl, anthracenyl, fluorenyl, indolyl, indanyl, and the like. Unless otherwise indicated in the specification, the aryl moiety may be optionally substituted with one or more substituents independently including acyl, alkyl, alkenyl, alkynyl, alkoxy, alkylaryl, cycloalkyl, arylalkyl, aryl, aryloxy, amino, amido, amidino, imino, azide, carbonate, carbamate, carbonyl, heteroalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, hydroxy, cyano, halogen, haloalkoxy, haloalkyl, ester, ether, mercapto, thio, alkylthio, arylthio, thiocarbonyl, nitro, oxo, phosphate, phosphonate, phosphinate, silyl, sulfinyl, sulfonyl, sulfonamido, synergistic sulfone, sulfonate, urea, -Si (R) unless otherwise indicated in the specificationa)3、-ORa、-SRa、-OC(O)-Ra、-N(Ra)2、-C(O)Ra、-C(O)ORa、-OC(O)N(Ra)2、-C(O)N(Ra)2、-N(Ra)C(O)ORa、-N(Ra)C(O)Ra、-N(Ra)C(O)N(Ra)2、-N(Ra)C(NRa)N(Ra)2、-N(Ra)S(O)tN(Ra)2(wherein t is 1 or 2), -P (═ O) (R)a)(Ra) OR-O-P (═ O) (OR)a)2Wherein each R isaIndependently is hydrogen, alkyl, haloalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl, or heteroarylalkyl, and each of these moieties may be optionally substituted as defined herein.

As used herein, the terms "cycloalkyl" and "carbocyclyl" each refer to a monocyclic or polycyclic group containing only carbon and hydrogen, and may be saturated or partially unsaturated. Partially unsaturated cycloalkyl groups may be referred to as "cycloalkenyl" if the carbocyclic ring contains at least one double bond and "cycloalkynyl" if the carbocyclic ring contains at least one triple bond. Ring (C)Alkyl includes groups having 3 to 13 ring atoms (i.e., C)3-13Cycloalkyl groups). As used herein, numerical ranges such as "3 to 10" refer to each integer within the given range; for example, "3 to 13 carbon atoms" means that the cycloalkyl group can consist of 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, and the like, up to and including 13 carbon atoms. The term "cycloalkyl" also includes bridged and spiro-fused ring structures that do not contain heteroatoms. The term also includes monocyclic or fused-ring polycyclic (i.e., rings that share adjacent pairs of atoms) groups. Polycyclic aryl groups include bicyclic, tricyclic, tetracyclic, and the like. In some embodiments, "cycloalkyl" may be C3-8An alkyl group. In some embodiments, "cycloalkyl" may be C3-5An alkyl group. Illustrative examples of cycloalkyl groups include, but are not limited to, the moiety C3-6Carbocyclyl includes, but is not limited to, cyclopropyl (C)3) Cyclobutyl (C)4) Cyclopentyl (C)5) Cyclopentenyl group (C)5) Cyclohexyl (C)6) Cyclohexenyl (C)6) Cyclohexadienyl (C)6) And the like. C3-7Examples of the carbocyclic group include norbornene (C)7)。C3-8Examples of the carbocyclic group include the above-mentioned C3-7Carbocyclyl and cycloheptyl (C)7) Cycloheptadienyl (C)7) Cycloheptatrienyl (C)7) Cyclooctyl (C)8) Bicyclo [2.2.1]Heptylalkyl, bicyclo [2.2.2]An octyl group and the like. C3-13Examples of the carbocyclic group include the above-mentioned C3-8Carbocyclic radicals and octahydro-1H indenyl, decahydronaphthyl, spiro [4.5 ]]Decyl groups, and the like. Unless otherwise indicated in the specification, a cycloalkyl group may be optionally substituted with one or more substituents independently including acyl, alkyl, alkenyl, alkynyl, alkoxy, alkaryl, cycloalkyl, aralkyl, aryl, aryloxy, amino, amido, amidino, imino, azide, carbonate, carbamate, carbonyl, heteroalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, hydroxy, cyano, halogen, haloalkoxy, haloalkyl, ester, ether, mercapto, thio, alkylthio, arylthio, thiocarbonyl, nitro, oxo, phosphate, phosphonate, phosphinate, silyl, sulfinyl, sulfonyl, sulfonamide, synergistic sulfone, sulfolane, and the likeAcid salt, urea, -Si (R)a)3、-ORa、-SRa、-OC(O)-Ra、-N(Ra)2、-C(O)Ra、-C(O)ORa、-OC(O)N(Ra)2、-C(O)N(Ra)2、-N(Ra)C(O)ORa、-N(Ra)C(O)Ra、-N(Ra)C(O)N(Ra)2、-N(Ra)C(NRa)N(Ra)2、-N(Ra)S(O)tN(Ra)2(wherein t is 1 or 2), -P (═ O) (R)a)(Ra) OR-O-P (═ O) (OR)a)2Wherein each R isaIndependently is hydrogen, alkyl, haloalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl, or heteroarylalkyl, and each of these moieties may be optionally substituted as defined herein. The terms "cycloalkenyl" and "cycloalkynyl" correspond to the description above with respect to "cycloalkyl" in which the "group" is substituted with "alkenyl" or "alkynyl", respectively, with the parent term "alkenyl" or "alkynyl" as described herein. For example, cycloalkenyl groups can contain 3 to 13 ring atoms, such as 5 to 8 ring atoms. In some embodiments, cycloalkynyl groups can contain 5 to 13 ring atoms.

As used herein, the term "halogen" refers to fluorine (F), chlorine (Cl), bromine (Br), or iodine (I). As used herein, the term "halide" or "halogen" refers to fluorine, chlorine, bromine or iodine. The terms "haloalkyl", "haloalkenyl", "haloalkynyl" and "haloalkoxy" include alkyl, alkenyl, alkynyl and alkoxy structures substituted with one or more halo groups or combinations thereof. For example, "fluoroalkyl" and "fluoroalkoxy" include haloalkyl and haloalkoxy, respectively, wherein halogen is fluorine, such as, but not limited to, trifluoromethyl, difluoromethyl, 2,2, 2-trifluoroethyl, 1-fluoromethyl-2-fluoroethyl, and the like. Alkyl, alkenyl, alkynyl and alkoxy are all as defined herein and may be optionally further substituted as defined herein.

As used herein, the term "heteroalkyl" refers to an alkyl group having one or moreBackbone chain atoms selected from atoms other than carbon, such as oxygen, nitrogen, sulfur, phosphorus, or combinations thereof. Given value ranges, e.g. C1-4Heteroalkyl refers to the overall chain length, which in this example is 4 atoms long. For example, a-CH2OCH2CH3The radical being referred to as "C4"heteroalkyl, which includes a heteroatom center in the description of the length of the atom chain. The bond to the parent molecular structure may be through a heteroatom or a carbon in a heteroalkyl chain. For example, a nitrogen-containing heteroalkyl moiety refers to a group in which at least one of the backbone atoms is a nitrogen atom. One or more heteroatoms in the heteroalkyl group may be optionally oxidized. If one or more nitrogen atoms are present, they may also optionally be quaternized. For example, heteroalkyl groups also include skeletal chains substituted with one or more nitroxide (-O-) substituents. Exemplary heteroalkyl groups include, but are not limited to, ethers, such as methoxyethyl (-CH)2CH2OCH3) Ethoxymethyl (-CH)2OCH2CH3) (methoxymethoxy) ethyl (-CH)2CH2OCH2OCH3) (methoxymethoxy) methyl (-CH)2OCH2OCH3) And (methoxyethoxy) methyl (-CH)2OCH2CH2OCH3) Etc.; amines such as (-CH)2CH2NHCH3,-CH2CH2N(CH3)2,-CH2NHCH2CH3,-CH2N(CH2CH3)(CH3) Etc.).

As used herein, the term "heteroaryl" or "heteroaromatic" refers to a group of 5-18 membered monocyclic or polycyclic (e.g., bicyclic, tricyclic, tetracyclic, etc.) aromatic ring systems (e.g., sharing 6, 10, or 14 pi electrons in a ring array) having ring carbon atoms and 1-6 ring heteroatoms provided in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, phosphorus, and sulfur ("5-18 membered heteroaryl"). Heteroaryl polycyclic ring systems may contain one or more heteroatoms in one or both rings. As used herein, numerical ranges such as "5 to 18" refer to each integer within the given range; for example, "5 to 18 ring atoms" means that the heteroaryl group may consist of 5 ring atoms, 6 ring atoms, etc., up to and including 18 ring atoms. In some embodiments, heteroaryl groups may contain 5 to 14 ring atoms. In some embodiments, divalent groups derived from monovalent heteroaryl groups (named by removing one hydrogen atom from a carbon atom having a free valence, ending with a "-yl") are named by adding a "pyridine" in the name of the corresponding monovalent group, e.g., a pyridine group having two points of attachment is a pyridyl group.

For example, a nitrogen-containing "heteroaromatic" or "heteroaryl" moiety refers to an aromatic group in which at least one backbone atom of the ring is a nitrogen atom. One or more heteroatoms in the heteroaryl group can be optionally oxidized. If one or more nitrogen atoms are present, they may also optionally be quaternized. Heteroaryl also includes ring systems substituted with one or more nitroxide (-O-) substituents, such as pyridyl nitroxide. The heteroaryl is attached to the parent molecular structure through any atom of the ring.

"heteroaryl" also includes ring systems in which a heteroaryl ring, as defined above, is fused to one or more aryl groups, wherein the point of attachment to the parent molecular structure is on the aryl or heteroaryl ring, or in which a heteroaryl ring, as defined above, is fused to one or more cycloalkyl or heterocyclyl groups, wherein the point of attachment to the parent molecular structure is on the heteroaryl ring. For polycyclic heteroaryl groups in which one ring does not contain a heteroatom (e.g., indolyl, quinolinyl, carbazolyl, and the like), the point of attachment to the parent molecular structure can be on either ring, i.e., the ring bearing the heteroatom (e.g., 2-indolyl) or the ring containing no heteroatom (e.g., 5-indolyl). In some embodiments, a heteroaryl group is a 5-10 membered aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms provided in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, phosphorus, and sulfur ("5-10 membered heteroaryl"). In some embodiments, heteroaryl is a 5-8 membered aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms provided in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, phosphorus, and sulfur ("5-8 membered heteroaryl"). In some embodiments, a heteroaryl group is a 5-6 membered aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms provided in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, phosphorus, and sulfur ("5-6 membered heteroaryl"). In some embodiments, the 5-6 membered heteroaryl has 1-3 ring heteroatoms selected from nitrogen, oxygen, phosphorus, and sulfur. In some embodiments, the 5-6 membered heteroaryl has 1-2 ring heteroatoms selected from nitrogen, oxygen, phosphorus, and sulfur. In some embodiments, the 5-6 membered heteroaryl has 1 ring heteroatom selected from nitrogen, oxygen, phosphorus, and sulfur.

Examples of heteroaryl groups include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzindolyl, 1, 3-benzodiazolyl, benzofuranyl, benzoxazolyl, benzo [ d [ d ] ]]Thiazolyl, benzothiadiazolyl, benzo [ b ]][1,4]Dioxepin, benzo [ b][1,4]Oxazinyl, 1, 4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzoxazolyl, benzopyranyl, benzofuranyl, benzopyranyl, benzofurazan, benzothiazolyl, benzothienyl, methylthiophenyl [3,2-d]Pyridine, benzotriazolyl, benzo [4,6 ]]Imidazole [1,2-a ]]Pyridyl, carbazolyl, sinelinyl, cyclopentane [ d ]]Pyrimidinyl, 6, 7-dihydro-5H-cyclopentane [4, 5 ]]Thieno [2, 3-d ]]Pyrimidinyl, 5, 6-dihydrobenzo [ h ]]Quinazolinyl, 5, 6-dihydrobenzo [ h ]]Sinilinyl, 6, 7-dihydro-5H-benzo [6, 7]]Cycloheptyl [1, 2-c ]]Pyridazinyl, dibenzofuranyl, dibenzothienyl, furanyl, furazanyl, furanonyl, furo [3, 2-c ]]Pyridyl, 5,6,7,8, 9, 10-hexachlorocyclohexane [ d]Pyrimidinyl, 5,6,7,8, 9, 10-hexachlorocyclohexane [ d]Pyridazinyl, 5,6,7,8, 9, 10-hexachlorocyclohexane [ d]Pyridyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolyl, isoindolyl, isoquinolyl, indolizinyl, isoxazolyl, 5, 8-methoxy-5, 6,7, 8-tetrahydroquinazolinyl, naphthyridinyl, 1, 6-naphthyridinyl, oxadiazolyl, 2-oxazapyridyl, oxazolyl, oxiranyl, 5,6, 6a, 7,8, 9, 10, 10 a-octahydrobenzo [ h ] benzo]Quinazolinyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pterin, purinyl, pyranyl, pyrrolyl, quinoxalinyl, phenoxazinyl, purinyl, pyrroniyl, and the like,Pyrazolyl and pyridoxol [3,4-d ]]Pyrimidinyl, pyrido [3,2-d ]]Pyrimidinyl, pyrido [3,4-d ]]Pyrimidinyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyrrolyl, quinazolinyl, quinoxalinyl, quinolinyl, isoquinolinyl, tetrahydroquinoline, 5,6,7, 8-tetrahydroquinazoline, 5,6,7, 8-tetrahydrobenzotriazole [4.5]Thieno [2, 3-d ]]Pyrimidineol, 5,6,7, 8-tetrahydropyrido [4, 5-c]Pyridazinyl, thiazolyl, thiadiazolyl, thiapyranyl, triazolyl, tetrazolyl, triazinyl, thieno [2, 3-d ]]Pyrimidinyl, thieno [3,2-d]Pyrimidinyl, thieno [2, 3-c)]Pyridyl and thiophenyl (i.e., thienyl). Unless otherwise indicated in the specification, a heteroaryl moiety may be optionally substituted with one or more substituents independently including acyl, alkyl, alkenyl, alkynyl, alkoxy, alkylaryl, cycloalkyl, arylalkyl, aryl, aryloxy, amino, amido, amidino, imino, azide, carbonate, carbamate, carbonyl, heteroalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, hydroxy, cyano, halogen, haloalkoxy, haloalkyl, ester, ether, mercapto, thio, alkylthio, arylthio, thiocarbonyl, nitro, oxo, phosphate, phosphonate, phosphinate, silyl, sulfinyl, sulfonyl, sulfonamido, synergistic sulfone, sulfonate, urea, -Si (R) independentlya)3、-ORa、-SRa、-OC(O)-Ra、-N(Ra)2、-C(O)Ra、-C(O)ORa、-OC(O)N(Ra)2、-C(O)N(Ra)2、-N(Ra)C(O)ORa、-N(Ra)C(O)Ra、-N(Ra)C(O)N(Ra)2、-N(Ra)C(NRa)N(Ra)2、-N(Ra)S(O)tN(Ra)2(wherein t is 1 or 2), -P (═ O) (R)a)(Ra) OR-O-P (═ O) (OR)a)2Wherein each R isaIndependently is hydrogen, alkyl, haloalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl, and each of these moieties may optionally be substituted as defined hereinAnd (4) substitution.

As used herein, an "effective amount" of a surgically active agent refers to an amount sufficient to elicit a desired biological response. It will be appreciated by those of ordinary skill in the art that an effective amount of a compound of the invention may vary depending on such factors as the desired biological endpoint, the pharmacokinetics of the compound, the condition being treated, the mode of administration, and the patient.

As used herein, "treatment" or "treating" of a disease or disorder refers to a method of alleviating, delaying or ameliorating the condition before or after the onset of the disease or disorder. The treatment may be one or more effects or symptoms of needle disease and/or underlying pathology. The treatment can be palliation, and can be, but is not limited to, complete regression of the disease or disease symptoms. The degree of such reduction or prevention is at least 5%, 10%, 20%, 40%, 50%, 60%, 80%, 90%, 95% or 100% compared to an equivalent, non-managed control group, regardless of the amount by standard techniques.

As used herein, "preventing" refers to a method for eliminating, delaying, avoiding or preventing the onset, incidence, severity or recurrence of a disease or disorder. For example, a method is "preventing" if the onset, incidence, severity, or recurrence of a disease or disorder or one or more symptoms thereof is reduced or delayed in a subject susceptible to the disease as compared to a subject not receiving the method. A disclosed method is "prevention" if the onset, incidence, severity, or recurrence of osteoporosis or one or more symptoms of the disease or disorder is reduced or delayed in a subject susceptible to the disease after receiving the method as compared to the progression of the condition prior to receiving treatment in the subject. Thus, the degree of reduction or delay in the onset, incidence, severity, or recurrence of osteoporosis may be approximately 5,10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any reduction therebetween.

As used herein, "pharmaceutically acceptable forms" of the disclosed compounds include, but are not limited to, pharmaceutically acceptable salts, hydrates, esters, solvates, isomers, prodrugs, and isotopically labeled derivatives of the disclosed compounds. In one embodiment, "pharmaceutically acceptable forms" include, but are not limited to, pharmaceutically acceptable salts, esters, prodrugs, and isotopically labeled derivatives of the disclosed compounds. In certain embodiments, "pharmaceutically acceptable forms" include, but are not limited to, pharmaceutically acceptable isomers, stereoisomers, prodrugs and isotopically labeled derivatives of the disclosed compounds.

In certain embodiments, the pharmaceutically acceptable form is a pharmaceutically acceptable salt. As used herein, the term "pharmaceutically acceptable salt" refers to a salt that is, within the scope of sound medical judgment, suitable for contact with the tissues of a subject 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, Berge et al, in pharmaceutical sciences (1977)66:1-19 describe in detail pharmaceutically acceptable salts. Pharmaceutically acceptable salts of the compounds provided herein include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable non-toxic acid addition salts are amino salts formed from inorganic acids such as hydrochloric, hydrobromic, phosphoric, sulfuric and perchloric acids or organic acids such as acetic, oxalic, maleic, tartaric, citric, succinic or malonic acids, or by using other methods in the art, such as ion exchange. Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartates, benzenesulfonic acid, benzenesulfonates, benzoates, bisulfates, borates, butyrates, camphorates, camphorsulfonates, citrates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, formates, fumarates, gluconates, glycerophosphates, gluconates, hemisulfates, heptanoates, hexanoates, hydroiodides, 2-hydroxy-ethanesulfonates, lactobionates, laurates, dodecylsulfates, malates, maleates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, oxalates, palmitates, pamoates, pectinates, persulfates, 3-phenylpropionates, phosphates, Picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate and the like. In some embodiments, organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, lactic acid, trifluoroacetic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like.

Salts can be prepared in situ during the isolation and purification of the disclosed compounds or separately, e.g., by reacting the free base or free acid of the parent compound with a suitable base or acid, respectively. Pharmaceutically acceptable salts derived from suitable bases include alkali metals, alkaline earth metals, ammonium and N+(C1-4alkyl)4And (3) salt. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Other pharmaceutically acceptable salts include, non-toxic ammonium, quaternary ammonium and primary amine cations, formed from counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate. Organic bases from which salts can be derived include, for example, primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine. In some embodiments, the pharmaceutically acceptable base addition salt may be selected from the group consisting of ammonium, potassium, sodium, calcium, and magnesium salts.

In certain embodiments, the pharmaceutically acceptable form is a "solvate" (e.g., hydrate). As used herein, the term "solvate" refers to a compound that further includes a stoichiometric or non-stoichiometric amount of solvent bound by non-covalent intermolecular forces. The solvate can be the disclosed compound or a pharmaceutically acceptable salt thereof. When the solvent is water, the solvate is a "hydrate". Pharmaceutically acceptable solvates and hydrates are complexes that may include, for example, about 1 to 100, 1 to 10, 1 to 2, 3, or 4 solvent or water molecules. It is to be understood that the term "compound" as used herein includes compounds and solvates of compounds, as well as mixtures thereof.

In certain embodiments, the pharmaceutically acceptable form is a prodrug. As used herein, the term "prodrug" (or "prodrug") refers to a compound that is converted in vivo to yield a compound disclosed herein or a pharmaceutically acceptable form of the compound. Prodrugs are inactive when administered to a subject, but may be converted to the active compound in vivo by hydrolysis, e.g., in the blood. In some cases, the prodrug has better physical and/or transport properties than the parent compound. Upon administration to a subject, the prodrugs can increase the bioavailability of the compounds (e.g., enhance absorption into the blood by oral administration), which increases the efficiency of delivery to the biological compartment of interest (e.g., the brain or lymphatic system) relative to the parent compound. Exemplary prodrugs include derivatives of the disclosed compounds that have greater aqueous solubility or active transport across the intestinal membrane relative to the parent compound.

Prodrug compounds generally have the advantage of solubility, histocompatibility, or delayed release in mammalian organisms (see Bundgard, H, prodrug design (1985), pages 7-9, 21-24 (elmsevier, amsterdam.) a discussion of prodrugs, Higuchi, T et al, prodrug-novel delivery systems, a.c.s.symposium series, vol.14, and bioreversible vehicles in drug design, Edward b.roche, american society of medicine and pegman press, 1987, are all provided and both are fully incorporated herein.

As used herein, the term "pharmaceutically acceptable" excipient, carrier or diluent refers to a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in transporting or transporting the subject drug from one organ or portion of the body to another organ or portion of the body. Each carrier must be "acceptable", i.e., compatible with the other ingredients of the formulation, and not injurious to the patient. Examples of materials that can be used as pharmaceutically acceptable carriers include sugars such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; ethylene glycol, such as propylene glycol; polyols such as glycerol, sorbitol, mannitol and polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; ringer's solution; ethanol; a phosphate buffer solution; and other non-toxic compatible materials used in pharmaceutical formulations. Wetting, emulsifying and lubricating agents, for example, sodium lauryl sulfate, magnesium stearate and polyethylene oxide-polypropylene oxide copolymers, as well as coloring, release, coating, sweetening, flavoring and perfuming agents, preservatives and antioxidants may also be present in the composition.

As used herein, the term "isolated" or "purified" refers to a material that is substantially or substantially free of components that are normally present in its native state. Purity and homogeneity are typically determined using "analytical chemistry techniques", such as polyacrylamide gel electrophoresis or high performance liquid chromatography.

As used herein, the term "subject" refers to any animal (e.g., a mammal), including but not limited to humans, non-human primates, rodents, and populations awaiting receipt of a particular treatment. Generally, the terms "subject" and "patient" are used interchangeably herein for a human subject.

As used herein, "low dose" refers to a dose that is at least 5% (e.g., at least 10%, 20%, 50%, 80%, 90%, or even 95%) less than the minimum standard recommended dose of a particular compound formulated for a given route of administration, for the treatment of any human disease or disorder. For example, a "low dose" of an agent used to lower glucose levels and administered by inhalation will be different from a "low dose" of the same agent administered orally. For example, a "low dose" of a pharmaceutical agent formulated for administration by inhalation will be different from a "low dose" of the same pharmaceutical agent formulated for administration by oral means.

As used herein, "high dose" refers to a dose that is at least 5% (e.g., at least 10%, 20%, 50%, 100%, 200%, or even 300%) higher than the highest standard recommended dose for a particular compound for the treatment of any human disease or disorder.

As used herein, the term "prodrug" or "prodrug" refers to a pharmacological derivative of the proto-drug molecule that requires biotransformation (spontaneous or enzymatic) in vivo to release the active drug. These prodrugs are pharmaceutically active in vivo when they undergo solvolysis or enzymatic degradation under physiological conditions. The prodrug compounds herein can be mono-, di-, tri-, etc. Depending on the number of biotransformation steps required to release the active drug in the organism, and the number of functional groups present in the precursor type.

Prodrug forms generally have the advantage of solubility, histocompatibility, or delayed release in the mammalian body. (see, Bundgard, prodrug design, pages 7-9, 21-24, Elsevier, Amsterdam, 1985; Silverman, organic chemistry for drug design and drug behavior, pages 352 and 401, academic Press, san Diego, Calif., 1992). Prodrugs generally known in the art include acid derivatives such as esters prepared by reacting the parent acid with a suitable alcohol, amides prepared by reacting the parent acid compound with an amine, basic groups reacted to form acylated base derivatives, and the like. Of course, other prodrug derivatives may be combined with other features disclosed herein to increase bioavailability. Thus, one skilled in the art will appreciate that certain compounds disclosed herein having a free amino, hydroxyl or carboxyl group can be converted into prodrugs. Prodrugs include compounds having an amino acid residue, or a polypeptide chain having two or more (e.g., two, three, or four) amino acid residues covalently linked by peptide bonds to a free amino, hydroxyl, or carboxylic acid group of a compound disclosed herein. Amino acid residues include the 20 natural amino acids commonly represented by three letter symbols, and also include 4-hydroxyproline, hydroxylysine, desmosine, isodesmosine, 3-methylhistidine, norvaline, β -alanine, γ -aminobutyric acid, citrulline, homocysteine, homoserine, ornithine and methionine sulfone. Prodrugs also include compounds having a carbonate, carbamate, amide, or alkyl ester moiety covalently bonded to any of the above substituents disclosed herein.

Detailed Description

The present invention provides novel pyrimidine derivatives and analogues and pharmaceutical compositions thereof which exhibit inhibitory activity against certain tyrosine kinases, such as Bruton's Tyrosine Kinase (BTK) and/or Focal Adhesion Kinase (FAK). Also disclosed herein are methods of treating, alleviating, or preventing certain diseases or disorders mediated by tyrosine kinases, such as BTK, BTK (C481S), FAK, ERK kinase, and/or other TEC family kinases. Such diseases and disorders include cancer, inflammatory diseases, fibrosis, diabetes, autoimmune diseases, or immune-mediated diseases.

Tyrosine kinases play an important role in regulating a variety of cellular processes, such as cell proliferation and cell survival. Certain tyrosine kinases are known to be activated by mutations or to be aberrantly expressed in many human cancers. Selective inhibition of certain tyrosine kinases is useful in the treatment of human diseases such as cancer. For example, Epidermal Growth Factor Receptor (EGFR) is mutated and/or overexpressed in certain human cancers, such as lung cancer. Several EGFR inhibitors have been approved for the treatment of EGFR mutated and/or overexpressed cancers.

Focal Adhesion Kinase (FAK)

Focal Adhesion Kinase (FAK) is a tyrosine kinase that integrates signals from integrin and growth factor receptors and regulates a variety of cellular functions including adhesion, spreading, migration, invasion, polarity, proliferation and survival. Studies have shown that FAK is required for the development and progression of tumors, such as skin and breast tumors. FAK has been reported to be overexpressed in many human tumors (e.g., breast, colon, pancreatic, and prostate cancers). FAK is regulated and activated by phosphorylation, particularly at tyrosine residue Y397. In addition, phosphorylated FAK (Y397) is increased in certain tumors, such as pancreatic cancer, compared to normal tissue. (McLeanet et al 2005Nat Rev Cancer 5(7): 505-15; Frame et al 2010Nat Rev Mol Cell biol.11(11): 802-14; McLean et al 2004genes Dev.18(24):2998-

RNAi inhibition of FAK or dominant negative FAK has been shown to result in loss of adhesion and cell death in human cancer cells (e.g., breast, melanoma, and ovarian cancer cells). Inhibition of FAK by shRNA inhibits lung cancer metastasis and increases survival in mouse models. Furthermore, loss of FAK catalytic activity reduces the growth of v-Src tumors in mice. (Beviglia et al 2003biochem J.373(Pt 1): 201-10; Smith et al 2005Melanoma Res.15(5): 357-62; Haider et al 2005Clin Cancer Res.11(24Pt 1): 8829-36; Mitra et al 2006oncogene.25(32):4429-40.)

In cancer, many functions of FAK are associated with its signaling role downstream of integrin and growth factor receptors on the plasma membrane. FAK can localize in the nucleus, and nuclear FAK regulates transcription of inflammatory cytokines and chemokines, promoting the formation of an immunosuppressive tumor microenvironment. Inhibition of FAK activity by triggering immune-mediated tumor regression may have beneficial therapeutic effects. (Lim et al 2008mol cell.29(1): 9-22; Serrels et al 2015cell 163(1): 160-73; Sulzmaier et al 2014Nature Reviews Cancer 14, 598-

Studies have shown that FAK is the primary mediator of fiber formation. FAK is activated after skin injury. Fibroblast-specific FAK knockout mice had significantly less inflammation and fibrosis than control mice, suggesting that FAK plays an important role in fibrosis. Inhibition of FAK significantly eliminated bleomycin-induced pulmonary fibrosis in mice. FAK is a promising target for the treatment of fibrotic diseases (e.g. pulmonary fibrosis). (Wong et al.2011nat Med.18(1): 148-52; Lagares et al.2012Arthritis Rheum.64(5): 1653-64; Kinoshita et al.2013Am J Respir Cell mol biol.49(4):536-43.)

Thus, there is ample evidence to suggest that FAK is a regulator of adhesion, apoptosis, cell growth inhibition, migration, angiogenesis, fibrosis, and/or immunosuppression. Inhibition of FAK activity is useful for treating FAK-related diseases, such as cancer and fibrosis. Currently, there are no FAK inhibitors approved by the U.S. Food and Drug Administration (FDA) that can be used to treat patients.

Bruton's Tyrosine Kinase (BTK)

BTK is a kinase encoded by the BTK gene. BTK plays a crucial role in B cell development and survival, such as B cell activation and B cell signaling pathways, which link B Cell Receptor (BCR) stimulation to downstream responses in cells. Abnormal B cell proliferation and differentiation may lead to lymphomas, including acute or chronic lymphocytic leukemia. (Rawlings1999Clin Immunol.91(3): 243-53; Khan 2001Immunol Res.23(2-3): 147-56; Hendriks et al.2014nat Rev Cancer 14(4):219-32.)

Inhibition of BTK can be useful as a treatment for B cell mediated diseases such as B cell lymphomas and leukemias.

In addition, BTK-dependent immune cell crosstalk has been reported to drive pancreatic cancer. BTK, a key B cell and macrophage kinase, regulates T cell-dependent immune responses. PDAC mice were treated with the BTK inhibitor ibutinib recombinant macrophage to drive towards the TH1 phenotype and inhibit PDAC growth, indicating that BTK signaling mediates PDAC immunosuppression. Pharmacological inhibition of BTK in PDAC mice reactivates the adaptive immune response, providing a new treatment for this devastating tumor. (Gunderson et al.2016cancer Discov.6(3):270-85.)

Effective BTK inhibitors are useful for the treatment of cancer and rheumatoid arthritis, allergy and asthma. Studies have also shown that BTK signaling is involved in a variety of inflammatory, autoimmune, and immune-mediated diseases. (Corneth et al.2016Curr TopMicrobiol Immunol.393:67-105.)

Other TEC family kinases also play important roles in T cell activation. ITK is one of the family members of the TEC of tyrosine kinases involved in T cell receptor signaling, playing an important role in T cell activation and development. ITK inhibitors are useful for the treatment of diseases such as allergic asthma and atopic dermatitis. (Lucas et al 2003Immunol Rev.191: 119-38; Schwartzberg et al 2005nat Rev Immunol.5(4): 284-95; August et al 2012int Rev Immunol.31(2):155-65.)

In addition, BMK inhibitors may be used as anticancer agents. The research shows that BMX is highly expressed in cancer cells, the cell proliferation and the tumorigenicity are promoted, and the deletion of BMX can inhibit the tumor growth. (Zhuang et al.2014J Exp Clin Cancer Res.33: 25; Li et al.2017Oncotarget.8(30):49238-

Since TEC family kinases (e.g., BTK, ITK, BMX) play a key role in the activation of B and/or T cells that are involved in the pathogenesis of inflammatory, autoimmune and immune-mediated diseases, effective inhibition of these kinases would be promising treatments for such diseases.

Ibrutinib is a small molecule BTK inhibitor that has been approved for the treatment of a variety of B cell lymphomas and leukemias. However, as the data for the application of ibrutinib to cancer therapy matures, concerns about adverse reactions and drug resistance thereof begin to be raised. (Kaur et al.2017Ann Hematol.96(7): 1175-1184; Furman et al.2014N EnglJ Med.370(24):2352-4.)

Studies have shown that ibrutinib also targets the EGFR wild type (wt) present in normal cells, and can cause severe side effects such as skin rash, diarrhea, and weight loss. These side effects limit the clinical use of ibrutinib.

As disclosed herein, it has now been unexpectedly discovered that certain pyrimidine derivatives are effective in inhibiting BTK and/or other TEC family non-receptor tyrosine kinases, such as bone marrow tyrosine kinase (BMX) or interleukin-2 induced T cell kinase (ITK). In addition, certain pyrimidine derivatives are effective in inhibiting drug resistance mutations BTK (C481S), FAK, and BTK. In addition, certain pyrimidine derivatives are effective in inhibiting the drug resistance mutations BTK (C481S), FAK and ERK, and BTK.

In one aspect, the invention generally relates to a compound having the structural formula (I):

Figure BDA0002467331340000221

wherein the content of the first and second substances,

x is NH (CH)2)nO or S, n is 0 or 1;

a is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

b is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R1’may be absent, and R1And R1’Each independently (if present) selected from halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, carboxamide, sulfonamide and R1xWherein R is1xSelected from:

Figure BDA0002467331340000222

optionally R1And R1’Is a group containing an electrophilic moiety, optionally R1And R1’Taken together to form a 4-6 membered aliphatic or aromatic cyclic or heterocyclic moiety;

R2and R3Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, or R2And R3Taken together to form a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R4and R5Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, substituted carboxylic acid amide, substituted sulfonamide, or R4And R5Together form a 6-15 membered aliphatic, cyclic or heterocyclic moiety,

or a pharmaceutically acceptable form thereof.

In certain embodiments, the compound has the following structural formula;

Figure BDA0002467331340000231

in certain embodiments, R in structural formula (I)2And R3Taken together to form a 5-membered ring having the structural formula (II):

Figure BDA0002467331340000232

wherein the content of the first and second substances,

Y1selected from CH, CH2N, NH, O or S;

Y2selected from CH, CH2N, NH, O or S;

Y3selected from CH, CH2N, NH, O or S; and

Y1and Y2The bond between (a) and (b) may be a double or single bond; y is2And Y3The bond between (a) and (b) may be a double or single bond; provided that Y is1、Y2And Y3Is CH or CH2(ii) a And

Y1and Y2A bond between2And Y3At least one of the bonds in (b) is a single bond.

In certain embodiments, B in structural formula (I) is a six membered aryl group, and the compound has structural formula (III):

Figure BDA0002467331340000233

in certain embodiments, R in structural formula (III)4And R5Meta and para, respectively, having the structural formula (IV):

Figure BDA0002467331340000241

in certain embodiments, R in structural formula (III) or (IV)4And R5Co-formA 6 to 15 membered aliphatic, cyclic or heterocyclic moiety.

In certain embodiments, R4And R5Together form a 12-membered aliphatic heterocyclic moiety.

In certain embodiments, the compound has the following structural formula:

Figure BDA0002467331340000242

wherein R is6、R7And R8Each independently selected from hydrogen, halogen, C1-C6Alkyl and C1-C6An alkoxy group.

In certain embodiments, R6、R7And R8Is H, the compound having the structural formula (V-a):

in certain embodiments, R in structural formula (V)2And R3Taken together to form a 5-membered heterocyclic aryl moiety having the structural formula (VI):

Figure BDA0002467331340000251

wherein the content of the first and second substances,

Y1selected from CH, CH2N, NH, O or S;

Y2selected from CH, CH2N, NH, O or S;

Y3selected from CH, CH2N, NH, O or S;

Y1and Y2The bond between (a) and (b) may be a double or single bond; y is2And Y3The bond between (a) and (b) may be a double or single bond; provided that Y is1、Y2And Y3Is CH or CH2(ii) a And

Y1and Y2A bond between Y and2and Y3At least one of the bonds between is a single bond。

In certain embodiments, R6、R7And R8Is H, the compound having the structural formula (VI-a):

in certain embodiments, a is a 6-membered aryl moiety having the structural formula (VII):

Figure BDA0002467331340000261

wherein the content of the first and second substances,

z is CH or N; and

R1' and R1Each independently selected from hydrogen, halogen, C1-C6Alkyl, hydroxy or a group containing an electrophilic moiety, provided that R1' and R1At least one of "is a group comprising an electrophilic moiety.

In certain embodiments, Z in formula (VII) is CH.

In certain embodiments, Z in formula (VII) is N.

In certain embodiments, a is a 6-membered aliphatic cyclic or heterocyclic moiety having the structural formula (VIII):

z is CH or N.

In certain embodiments, Z in structural formula (VIII) is CH.

In certain embodiments, Z in structural formula (VIII) is N.

In certain embodiments, a is a 5-membered aliphatic cyclic moiety having structural formula (IX):

R1' and R2Each independently selected from hydrogen, halogen, C1-C6Alkyl, hydroxy or a group containing an electrophilic moiety, provided that R1' and R2At least one of "is a group comprising an electrophilic moiety.

In certain embodiments of the above compounds, Y1And Y2The bond between is a double bond. In certain embodiments of the above compounds, Y1And Y2The bond in between is a single bond.

In certain embodiments of the above compounds, Y2And Y3The bond between is a double bond. In certain embodiments of the above compounds, Y2And Y3The bond in between is a single bond.

In certain embodiments of the above compounds, Y1Is N; y is2Is CH; y is3Is NH.

In certain embodiments of the above compounds, Y1Is CH; y is2Is N; y is3Is NH.

In certain embodiments of the above compounds, Y1Is CH; y is2Is CH; y is3Is NH.

In certain embodiments of the above compounds, Y1Is CH; y is2Is CH; y is3Is O or S.

In certain embodiments of the above compounds, X is NH.

In certain embodiments of the above compounds, X is O.

In certain embodiments of the above compounds, X is S.

In certain embodiments of the above compounds, the electrophilic or nucleophilic moiety is acrylamide (CH)2CH-C (═ O) -NH-). In certain embodiments of the above compounds, the electrophilic or nucleophilic moiety is-N-S (═ O)2-CH=CH2、-N-C(=O)-CH=CH-CH3or-N-S (═ O)2-CH=CH-CH3

In certain embodiments, the disclosed compounds form irreversible covalent bonds with a target kinase.

In certain embodiments, the disclosed compounds form reversible non-covalent bonds with a target kinase.

Exemplary compounds of the invention include, but are not limited to, those listed in table 1:

table 1.Exemplary Compounds

Figure BDA0002467331340000281

Figure BDA0002467331340000291

In certain embodiments, the compound has the following structural formula:

Figure BDA0002467331340000301

in certain embodiments, the compound has the following structural formula:

Figure BDA0002467331340000302

in certain embodiments, the compound has the following structural formula:

in certain embodiments, the compound has the following structural formula:

Figure BDA0002467331340000311

in certain embodiments, the compound has the following structural formula:

Figure BDA0002467331340000312

in certain embodiments, the compound has the following structural formula:

in certain embodiments, the compound has the following structural formula:

Figure BDA0002467331340000314

Figure BDA0002467331340000321

in certain embodiments, the compound has the following structural formula:

Figure BDA0002467331340000322

in certain embodiments, the compound has the following structural formula:

in certain embodiments, the compound has the following structural formula:

in certain embodiments, the compound has the following structural formula:

in certain embodiments, the compound has the following structural formula:

in certain embodiments, the compound has the following structural formula:

Figure BDA0002467331340000333

in certain embodiments, the compound has the following structural formula:

Figure BDA0002467331340000334

in certain embodiments, the compound has the following structural formula:

in certain embodiments, the compound has the following structural formula:

Figure BDA0002467331340000342

in certain embodiments, the compound has the following structural formula:

Figure BDA0002467331340000343

in certain embodiments, the compound has the following structural formula:

Figure BDA0002467331340000344

in another aspect, the invention relates generally to a pharmaceutical composition comprising an amount of a compound having the structural formula (I):

wherein the content of the first and second substances,

x is NH (CH)2)nO or S, n is 0 or 1;

a is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

b is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R1’may be absent, and R1And R1’Each independently (if present) selected from halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, carboxamide, sulfonamide and R1xWherein R is1xSelected from:

optionally R1And R1’Is a group containing an electrophilic moiety, optionally R1And R1’Taken together to form a 4-6 membered aliphatic or aromatic cyclic or heterocyclic moiety;

R2and R3Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, or R2And R3Taken together to form a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R4and R5Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, substituted carboxylic acid amide, substituted sulfonamide, or R4And R5Together form a 6-15 membered aliphatic, cyclic or heterocyclic moiety,

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In certain embodiments, the pharmaceutical composition is effective to treat, prevent, or ameliorate cancer or a related disease or disorder.

In certain embodiments, the pharmaceutical composition is effective to treat, prevent, or ameliorate an inflammatory disease or related disease or disorder.

In certain embodiments, the pharmaceutical composition is effective to treat, prevent, or ameliorate fibrosis or related diseases or disorders.

In certain embodiments, the pharmaceutical composition is effective to treat, prevent, or ameliorate an autoimmune disease or related disease or disorder.

In certain embodiments, the pharmaceutical composition is effective for treating, preventing, or ameliorating an immune-mediated disease or related disease or disorder.

The pharmaceutical compositions described herein include pharmaceutical compositions containing any of the compounds provided herein.

In certain pharmaceutical composition embodiments, the compound has the following structural formula:

Figure BDA0002467331340000361

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

Figure BDA0002467331340000362

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

Figure BDA0002467331340000371

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

Figure BDA0002467331340000372

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

Figure BDA0002467331340000374

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

Figure BDA0002467331340000383

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

Figure BDA0002467331340000392

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

Figure BDA0002467331340000393

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

Figure BDA0002467331340000395

Figure BDA0002467331340000401

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

Figure BDA0002467331340000403

in certain pharmaceutical composition embodiments, the compound has the following structural formula:

Figure BDA0002467331340000404

in yet another aspect, the present invention generally relates to a unit dosage form comprising the disclosed pharmaceutical composition.

In another aspect, the invention relates generally to a method for treating, reducing or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the structural formula (I):

wherein the content of the first and second substances,

x is NH (CH)2)nO or S, n is 0 or 1;

a is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

b is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R1’may be absent, and R1And R1’Each independently (if present) selected from halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, carboxamide, sulfonamide and R1xWherein R is1xSelected from:

Figure BDA0002467331340000412

optionally R1And R1’Is a group containing an electrophilic moiety, optionally R1And R1’Taken together to form a 4-6 membered aliphatic or aromatic cyclic or heterocyclic moiety;

R2and R3Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, or R2And R3Taken together to form a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R4and R5Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, substituted carboxylic acid amide, substituted sulfonamide, or R4And R5Together form a 6-15 membered aliphatic, cyclic or heterocyclic moiety,

or a pharmaceutically acceptable form thereof, effective to treat, prevent or ameliorate one or more of cancer, inflammatory disease, fibrosis, autoimmune disease or immune-mediated disease or related disease or disorder in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In certain embodiments, the methods are effective to treat, prevent, or ameliorate cancer or a related disease or disorder.

In certain embodiments, the methods are effective to treat, prevent, or ameliorate an inflammatory disease or related disease or disorder.

In certain embodiments, the methods are effective to treat, prevent, or ameliorate fibrosis or a related disease or disorder.

In certain embodiments, the methods are effective to treat, prevent, or ameliorate an autoimmune disease or related disease or disorder.

In certain embodiments, the methods are effective to treat, prevent, or ameliorate an immune-mediated disease or related disease or disorder.

In another aspect, the invention relates generally to a method of treating, ameliorating or preventing a disease or disorder. The method comprises administering to a subject in need thereof a pharmaceutical composition disclosed herein, wherein the disease or disorder is selected from cancer, an inflammatory disease, a fibrosis, an autoimmune disease or an immune-mediated disease or a related disease or disorder.

In another aspect, the invention relates generally to a method of treating, ameliorating or preventing a disease or disorder. The method comprises administering to a subject in need thereof a pharmaceutical composition comprising a compound having an inhibitory effect on the activity of Bruton's Tyrosine Kinase (BTK) and/or Focal Adhesion Kinase (FAK).

In certain method embodiments, the compound is an inhibitor of BTK.

In certain method embodiments, the compound is an inhibitor of bone marrow tyrosine kinase (BMX).

In certain method embodiments, the compound is an inhibitor of BTK and BMX.

In certain method embodiments, the compound is an inhibitor of interleukin-2 induced T cell kinase (ITK).

In certain method embodiments, the compound is an inhibitor of FAK.

In certain method embodiments, the compound is an inhibitor of BTK and FAK.

In certain method embodiments, the compound is an inhibitor of a BTK (C481S) mutant.

In certain method embodiments, the compound is an inhibitor of ERK.

In certain method embodiments, the compound is an inhibitor of BTK, BTK (C481S) mutants, and FAK.

In certain method embodiments, the compound is an inhibitor of BTK, BTK (C481S) mutants, FAK and ERK.

In certain method embodiments, the compound has the structural formula (I):

wherein the content of the first and second substances,

x is NH (CH)2)nO or S, n is 0 or 1;

a is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

b is a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R1’may be absent, and R1And R1’Each independently (if present) selected from halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, carboxamide, sulfonamide and R1xWherein R is1xSelected from:

Figure BDA0002467331340000431

optionally R1And R1’Is a group containing an electrophilic moiety, optionally R1And R1’Taken together to form a 4-6 membered aliphatic or aromatic cyclic or heterocyclic moiety;

R2and R3Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, or R2And R3Taken together to form a 5-7 membered, aliphatic or aromatic, cyclic or heterocyclic moiety;

R4and R5Each independently selected from hydrogen, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, cyano, hydroxy, amino, substituted carboxylic acid amide, substituted sulfonamide, or R4And R5Together form a 6-15 membered aliphatic, cyclic or heterocyclic moiety,

or a pharmaceutically acceptable form thereof.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000441

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000442

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000451

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000461

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000462

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000471

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000472

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000481

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000502

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the present invention relates generally to a method for treating, reducing, or preventing a disease or disorder, comprising administering to a subject in need thereof a pharmaceutical composition comprising a compound having the following structural formula:

Figure BDA0002467331340000503

or a pharmaceutically acceptable form thereof, effective for treating, preventing or ameliorating one or more of cancer, inflammatory diseases, fibrosis, autoimmune diseases, diabetes or immune-mediated diseases or related diseases or disorders in a mammal, including a human, and a pharmaceutically acceptable excipient, carrier or diluent.

In yet another aspect, the invention relates generally to the use of a compound disclosed herein and a pharmaceutically acceptable excipient, carrier or diluent in the manufacture of a medicament for the treatment of a disease or disorder.

In certain embodiments of use, the disease or disorder is cancer or a related disease or condition.

In certain embodiments of use, the disease or disorder is an inflammatory disease or related disease or condition.

In certain embodiments of use, the disease or disorder is fibrosis or a related disease or condition.

In certain embodiments of use, the disease or disorder is an autoimmune disease or related disease or disorder.

In certain embodiments of use, the disease or disorder is an immune-mediated disease or related disease or disorder.

The compounds, pharmaceutical compositions and methods of the invention are useful for treating or preventing proliferative diseases including non-hodgkin's lymphoma (particularly the diffuse large B-cell lymphoma (DLBCL) and Mantle Cell Lymphoma (MCL) subtypes), B-cell chronic lymphocytic leukemia and mature B-cell Acute Lymphocytic Leukemia (ALL), especially Acute Lymphocytic Leukemia (ALL).

The compounds, pharmaceutical compositions and methods of the invention are useful for treating autoimmune and inflammatory diseases, for example rheumatic diseases (such as rheumatoid arthritis, psoriatic arthritis, infectious arthritis, progressive chronic arthritis, osteoarthritis, traumatic arthritis, gouty arthritis, reiter's syndrome, polychondritis, acute synovitis, and spondylitis), glomerulonephritis (with or without nephrotic syndrome), autoimmune hematologic diseases (such as hemolytic anemia, aplastic anemia, idiopathic thrombocytopenia, and neutropenia), autoimmune gastritis, and autoimmune inflammatory bowel diseases (such as ulcerative colitis and crohn's disease), host versus graft disease, allograft rejection, chronic thyroiditis, graves ' disease, scleroderma, diabetes (types 1 and 2), Active hepatitis (acute and chronic), pancreatitis, primary biliary cirrhosis, myasthenia gravis, multiple sclerosis, systemic lupus erythematosus, psoriasis, atopic dermatitis, contact dermatitis, eczema, sunburn of the skin, vasculitis (e.g., behcet's disease), chronic renal insufficiency, stevens-johnson syndrome, inflammatory pain, idiopathic steatorrhea, cachexia, sarcoidosis, guillain-barre syndrome, uveitis, conjunctivitis, keratoconjunctivitis, otitis media, periodontal disease, interstitial pulmonary fibrosis, asthma, bronchitis, rhinitis, sinusitis, pneumoconiosis, pulmonary dysfunction syndrome, emphysema, pulmonary fibrosis, silicosis, chronic inflammatory lung diseases (e.g., chronic obstructive pulmonary disease), and other inflammatory or obstructive diseases of the respiratory tract.

When the compound, the pharmaceutical composition and the method of the present invention are administered to a subject in combination with another therapeutic agent for treating inflammatory diseases, autoimmune diseases or immune-mediated diseases, the anti-inflammatory effect and the therapeutic effect thereof can be enhanced.

Examples of therapeutic agents for treating inflammatory diseases, autoimmune diseases, or immune-mediated diseases include, but are not limited to, steroid drugs (e.g., prednisone, prednisolone, methylprednisolone, cortisone, hydrocortisone, betamethasone, dexamethasone, etc.), methotrexate, leflunomide, anti-TNFa agents (e.g., etanercept, infliximab, adalimumab, etc.), calcineurin inhibitors (e.g., tacrolimus, pimecrolimus, etc.), and antihistamines (e.g., diphenhydramine, hydroxyzine, loratadine, ebastine, ketotifen, cetirizine, levocetirizine, fexofenadine, etc.), and at least one therapeutic agent selected from the above may be included in the pharmaceutical composition of the present invention.

The compounds, pharmaceutical compositions and methods of the invention are useful for treating allergies, such as allergy to food, food additives, pesticides, dust mites, pollen, animal materials and contact allergens, type hypersensitivity asthma, allergic rhinitis, allergic conjunctivitis.

The compounds, pharmaceutical compositions and methods of the invention are useful for treating infectious diseases, such as sepsis, septic shock, endotoxic shock, gram-negative sepsis, shigellosis, meningitis, cerebral malaria, pneumonia, tuberculosis, viral myocarditis, viral hepatitis (hepatitis a, b, and c), aids virus infection, cytomegalovirus-induced retinitis, influenza, herpes, severe burn-induced infection, myalgia due to infection, cachexia secondary infection, and veterinary viral infections, such as lentiviruses, caprine arthritis virus, progressive interstitial pneumonia of sheep, feline immunodeficiency virus, bovine immunodeficiency virus, or canine immunodeficiency virus.

The compounds, pharmaceutical compositions and methods of the invention are useful for treating bone resorption disorders, such as osteoporosis, osteoarthritis, traumatic arthritis, gouty arthritis, and bone disorders associated with multiple myeloma.

The compounds, pharmaceutical compositions and methods of the invention are useful for treating cancers induced by BTK and/or FAK tyrosine kinases or mutants thereof.

The compounds, pharmaceutical compositions and methods of the invention are useful for treating cancer induced by BTK or mutants thereof.

The compounds, pharmaceutical compositions and methods of the invention are useful for treating cancers induced by BTK and/or BMX tyrosine kinase or mutants thereof.

The compounds, pharmaceutical compositions and methods of the invention are useful for treating cancers induced by BMX tyrosine kinase.

The compounds, pharmaceutical compositions and methods of the invention are useful for treating cancers induced by ERK kinases.

Examples of cancers or tumors can include, but are not limited to, liver cancer, hepatocellular cancer, thyroid cancer, colorectal cancer, testicular cancer, bone cancer, oral cancer, basal cell cancer, ovarian cancer, brain cancer, gallbladder cancer, biliary tract cancer, head and neck cancer, colorectal cancer, bladder cancer, tongue cancer, esophageal cancer, glioma, glioblastoma, renal cancer, malignant melanoma, gastric cancer, breast cancer, sarcoma, pharyngeal cancer, uterine cancer, cervical cancer, prostate cancer, rectal cancer, pancreatic cancer, lung cancer, skin cancer, and other solid cancers.

The compound, pharmaceutical composition and method of the present invention can enhance anticancer effects when administered to a subject in addition to another anticancer agent.

Examples of anticancer agents may include, but are not limited to, immunotherapy (e.g., CTLA4 antagonists, cell signaling inhibitors (e.g., imatinib, gefitinib, bortezomib, erlotinib, sorafenib, sunitinib, dasatinib, vorinostat, lapatinib, sirolimus, nilotinib, everolimus, pazopanib, trastuzumab, bevacizumab, cetuximab, ranibizumab, pegaptanib, panitumumab, etc.), mitotic inhibitors (e.g., paclitaxel, vincristine, vinblastine, etc.), alkylating agents (e.g., cisplatin, cyclophosphamide, onconine, carmustine, etc.), antimetabolites (e.g., methotrexate, 5-FU, etc.), intercalating anticancer agents (e.g., actinomycin, anthracycline, bleomycin, mitomycin-C, etc.), topoisomerase inhibitors (e.g., irinotecan, topotecan, teniposide, etc.), immunotherapeutic agents (e.g., interleukins, interferons, etc.) and anti-hormonal agents (e.g., tamoxifen, raloxifene, etc.), at least one anti-cancer agent selected from the above may be included in the pharmaceutical compositions of the invention.

Isotopically labeled compounds are also within the scope of the present disclosure. As used herein, "isotopically-labeled compound" refers to compounds presently disclosed, including pharmaceutically acceptable salts and prodrugs thereof, in which one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into the compounds disclosed herein include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, e.g.2H、3H、13C、14C、15N、18O、17O、31P、32P、35S、18F、36Cl。

By isotopically labeling the compounds disclosed herein, the compounds can be used in drug and/or matrix tissue distribution assays. Tritium (f) is particularly preferred because of its ease of preparation and detection3H) And carbon-14 (14C) To label the compound. In addition, with heavier isotopes such as deuterium (2H) Substitutions may offer therapeutic advantages which benefit from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and are therefore preferred in some circumstances. Isotopically-labeled compounds disclosed herein, as well as pharmaceutically acceptable salts, esters, and prodrugs thereof, can be prepared by methods known in the art.

In addition, the normally abundant hydrogen is replaced by heavier isotopes such as deuterium (1H) Certain therapeutic benefits may be obtained, for example, improved absorption, distribution, metabolism and/or excretion (ADME) properties, and the resulting drug may have improved efficacy, safety and/or tolerability. By using13C substitutions are usually rich12C may also bring benefits. (see, WO 2007/005643, WO 2007/005644, WO 2007/016361 and WO 2007/016431.)

Stereoisomers (e.g., cis and trans isomers) and all optical isomers (e.g., R and S enantiomers) of the compounds disclosed herein, as well as racemic, diastereomeric and other mixtures of such isomers, are within the scope of the disclosure.

After the compounds of the present disclosure are prepared, they are preferably isolated and purified to obtain a composition containing an amount equal to or greater than 95% by weight ("substantially pure"), and then used or formulated as described herein. In certain embodiments, the compounds of the present invention are greater than 99% pure.

Solvates and polymorphs of the compounds of the disclosure are also contemplated by the present invention. Solvates of the compounds of the invention include, for example, hydrates.

Any suitable mode of administration may be employed, for example parenteral, intravenous, subcutaneous, intramuscular, intracerebroventricular, intracorporeal, intraperitoneal, rectal or oral. For some patients, the most suitable mode of administration will depend on the nature and severity of the disease or condition being treated, or the nature of the treatment being used and the nature of the active compound.

Oral solid dosage forms include capsules, tablets, pills, powders and granules. In such solid dosage forms, the compounds described herein or derivatives thereof are mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or (I) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, (ii) binders, such as carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and acacia, (iii) humectants, such as glycerol, (iv) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate, (v) solution retarders, such as paraffin, (vi) absorption accelerators, such as quaternary ammonium compounds, (vii) wetting agents, such as cetyl alcohol and glycerol monostearate, (viii) adsorbents, such as kaolin and bentonite, and (ix) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate or mixtures thereof. When the dosage form is a capsule, tablet or pill, a buffering agent may also be included. Solid compositions of a similar type may also be employed as fillers in soft and hard gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other materials known in the art.

Oral liquid dosage forms 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 oil, groundnut oil, corn germ oil, olive oil, castor oil, sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, or mixtures of these substances, and the like. In addition to these inert diluents, the composition may also include other adjuvants, such as wetting agents, emulsifying agents, suspending agents, sweetening, flavoring, or perfuming agents.

The materials, compositions, and ingredients disclosed herein can be used for, can be combined with, can be used to prepare, or are products of the disclosed methods and compositions. It is to be understood that combinations, subsets, interactions, groups, etc. of these materials are disclosed that while specific reference of each individual, group, and permutation of these compounds may not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a method is disclosed and discussed and modifications to some of the molecules in the method are discussed, each combination and permutation of the method, and the modifications that are possible, are specifically contemplated unless specifically indicated to the contrary. Likewise, any subset or combination of the above is also specifically contemplated and disclosed. This concept applies to all aspects of this disclosure including, but not limited to, steps in methods of using the disclosed compositions. Thus, if there are additional steps that can be performed it is understood that these additional steps can be achieved with any specific step or combination of steps of the methods of the present disclosure, and that such combinations or subsets of combinations are specifically contemplated and should be considered disclosed.

Certain compounds of the present invention may exist in specific geometric or stereoisomeric forms. Such compounds, including cis and trans isomers, R-and S-enantiomers, diastereomers, (d) -isomers, (l) -isomers, racemic and other mixtures thereof, contemplated by the present invention are within the scope of the present invention. Additional asymmetric carbon atoms may be present in a substituent, such as an alkyl group. All such isomers and mixtures thereof are included in the present invention.

Mixtures of isomers containing any of a variety of isomer ratios may be used in accordance with the present invention. For example, where only two isomers are combined, the present invention contemplates mixtures comprising ratios of 50: 50, 60: 40, 70: 30, 80: 20, 90: 10, 95: 5, 96: 4, 97: 3, 98: 2, 99: 1, or 100: 0 isomers. One of ordinary skill in the art will readily appreciate that similar ratios can be considered for more complex isomer mixtures.

For example, if a particular enantiomer of a compound of the invention is desired, it may be prepared by asymmetric synthesis or by using chiral auxiliary derivatives, the resulting diastereomeric mixture separated, and the auxiliary group cleaved to provide the pure desired enantiomer. Alternatively, where the molecule contains a basic functionality, such as an amino group, or an acidic functionality, such as a carboxyl group, diastereomeric salts are formed with an appropriate optically active acid or base, followed by resolution of the resulting diastereomers by fractional crystallization or chromatography, as is well known in the art, to recover the pure enantiomers.

The following examples are intended to illustrate embodiments of the present invention and are not intended to be limiting in any way.

Examples

Synthesis of Compounds

4- [3- [ (5-Ethyl-2, 2-dimethyl-1, 3-dioxan-5-yl) oxo]Propoxy group]Aniline (I-1)

Scheme 1

In a round bottom flask, 2-dimethyl-5-vinyl-1, 3-dioxane (2.9g, 20.4mmol, 2.0eq.), 3- (4-nitrophenoxy) propan-1-ol (2.0g,10.2mmol,1.0eq.), sodium carbonate (206mg,2.0mmol,0.2eq.), RuCl2(PPh3)3(192mg,0.2mmol, 2.0% mol), heated at 115 ℃ for 3 hours under nitrogen. Thin Layer Chromatography (TLC) showed complete reaction (PE/EA ═ 15:1) to give crude 5-ethyl-2, 2-dimethyl-5- [3- (4-nitrophenoxy) propoxy ] propoxy]-1, 3-dioxane (2.78g, yield 80%).

To a solution of the intermediate product (2.78g,8.2mmol) obtained above in ethyl acetate (60mL) was added palladium on carbon (200mg, 7% Wt). The reaction flask was evacuated/refilled with hydrogen several times. The mixture was stirred under hydrogen at room temperature for 16 hours. After filtration and concentration, the residue was purified by silica gel column chromatography using petroleum ether/ethyl acetate (v/v ratio ═ 5:1 → 3:1) to give the desired 4- [3- [ (5-ethyl-2, 2-dimethyl-1, 3-dioxan-5-yl) oxo ] -propoxy ] aniline (I-1) (2.4g, yield 95%).

LC-MS (liquid chromatography-mass spectrometer) (LC-MS) 168.1[ ESI, M +1, 3- (4-aminophenoxy) -1-propanol]6,7, 9, 10, 12, 13-hexahydro-5, 8, 11, 14-tetraoxa-benzocyclododecen-2-ylamine (I-2)

Scheme 2

Step 1: to a mixture (22.02g,199.98mmol) in n-butanol (220mL) under nitrogen was added 46mL of LiOH H2O (18.46g,439.94mmol) in water. The mixture was heated to 95 ℃ and then 1-chloro-2- [2- (2-chloro-ethoxy) -ethoxy ] -ethoxy was added to n-butanol (240mL)]-ethane (37.43g,200.09mmol). The reaction mixture was heated overnight and then filtered. 1M aqueous hydrochloric acid was added to adjust the pH to 8. After removal of most of the n-butanol under reduced pressure, the residue was extracted with dichloromethane (250mL × 2). The combined organic layers were then washed with brine and dried over anhydrous magnesium sulfate. The crude product was purified by silica gel column chromatography to give 6.5g of 6,7, 9, 10, 12, 13-hexahydro-5, 8, 11, 14-tetraoxa-benzocyclododecene as a yellow oil in 14.4% yield.

Step 2: to a refluxing solution of 6,7, 9, 10, 12, 13-hexahydro-5, 8, 11, 14-tetraoxa-benzocyclododecene (6.50g,28.98mmol) was added dropwise nitric acid (56%, 5.4mL) in acetonitrile (65 mL). When TLC showed the reaction was complete, 160mL of water was added. The resulting solution was left at 2 ℃ overnight. The precipitate was collected and washed to neutrality to give the desired 2-nitro-6, 7, 9, 10, 12, 13-hexahydro-5, 8, 11, 14-tetraoxa-benzocyclododecene as a yellow solid (4.61g, 59% yield).

And 3, step 3: to the nitro compound (4.61g,17.12mmol) suspended in 90mL of methanol was added 0.46g of 10% Pd/C. After hydrogen exchange, the mixture is reacted in H2Stirring was continued overnight. TLC showed complete reduction of nitro material. The hydrogen was removed and the reaction flask was refilled with nitrogen several times. The reaction mixture was filtered, concentrated, and purified by silica gel column chromatography to give 3.76g of the desired aniline (I-2) (yield 91.7%).

LC/MS:240.2(ES+,M+1)

1HNMR(CDCl3,400MHz)6.83(d,1H,J=8Hz),6.33(dd,1H,J=4Hz),6.26(dd,1H,J=4,8Hz),(4.11,m 4H),3.89(m,2H),3.80(m,6H),1.75(br s,2H)。

1- [ (3R) -3- [ [2- [4- [3- [ (2-ethyl-5, 5-dimethyl-1, 3-dioxan-2-yl) oxo]Propoxy group]Aniline Base of]-5-fluoro-pyrimidin-4-yl]Amino group]-1-piperidinyl group]Prop-2-en-1-one (Compound-1)

Figure BDA0002467331340000571

Scheme 3

Figure BDA0002467331340000572

Step 1 (Cl-displacement). Tert-butyl (3R) -3-aminopiperidine-1-carboxylate (3.8g,19.2mmol), 2, 4-dichloro-5-F-pyrimidine (3.2g,19.2mmol) and trimethylamine (4.56mL,32mmol) were mixed in 25mL of n-butanol. The reaction mixture was heated at 130 ℃ overnight. Liquid chromatography-mass spectrometry showed the reaction was complete. After concentration, the residue was treated with a conventional aqueous solution and extracted with ethyl acetate. The organic layer was washed with water, brine and dried over anhydrous magnesium sulfate. After concentration by filtration, the crude product was purified by column chromatography on silica gel (petrol ether/ethyl acetate v/v4/1) to give tert-butyl (3R) -3- [ (2-chloro-5-fluoro-pyrimidin-4-yl) amino ] piperidine-1-carboxylate (4.8g, 73%) as a white solid.

Step 2 (De-Boc). The Boc-intermediate (3.3g,10mmol) was subjected to a Boc removal reaction with 6mL of trifluoroacetic acid in 15mL of dichloromethane for 4 h. The solvent was removed under reduced pressure and the residue suspended in ethyl acetate (200mL) and saturated sodium bicarbonate. The organic layer was separated and dried over anhydrous magnesium sulfate. After concentration by filtration, the de-Boc product was obtained (1.4g, 61% yield).

Step 3 (acrylation). Acryloyl chloride (0.4mL,5.0mmol) in 5mL of LDCM was added to a mixture of the de-Boc intermediate (0.8g,3.5mmol) and DIPEA (0.91g,7mmol) in 20mL of DCM/DMA (v/v 3:1) at-7.5 deg.C. The reaction mixture was stirred overnight. After concentration and treatment with ethyl acetate in a conventional aqueous solution, the final product was purified by silica gel column chromatography (petroleum ether/ethyl acetate 3/1) to give 0.62g of acrylamide in 62% yield.

Step 4 (palladium mediated aryl amination). To a mixture of acrylamide (0.31g,1.09mmol) and key intermediate I-1(0.4g,1.3mmol) in 6mL t-butanol was added potassium carbonate (1.4g,9.2mmol), X-phos (0.22g,0.24mmol) and Pd2(dba)3(0.06g,0.12 mmol). After nitrogen exchange, the mixture was heated at 80 ℃ overnight. The obtained product was extracted with ethyl acetate and purified by silica gel column chromatography (petroleum ether/ethyl acetate. cndot.: 3/1 to 2/1) to obtain the desired final product 1- [ (3R) -3- [ [2- [4- [3- [ (2-ethyl-5, 5-dimethyl-1, 3-dioxan-2-yl) oxo]Propoxy group]Anilino radical]-5-fluoro-pyrimidin-4-yl]Amino group]-1-piperidinyl group]Prop-2-en-1-one (0.15g, 25%).

LC-MS M/z (ES +, M-1422, 5, 5-trimethyl-1, 3-dioxane)

1- [ (3R) -3- [ [ 5-fluoro-2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0] tetraoxabicyclo]Hexadecyl-1 (12), 13, 15-triene- 14-ylamino) pyrimidin-4-yl]Amino group]-1-piperidinyl group]Synthesis of prop-2-en-1-one (Compound-2)

Figure BDA0002467331340000581

Scheme 4

Figure BDA0002467331340000582

To a mixture of acrylamide (61mg,0.21mmol,1.07eq.) and key intermediate I-2(48mg,0.2mmol,1eq.) in 4mL of n-butanol was added 2 drops of trifluoroacetic acid. The resulting mixture was refluxed overnight. The reaction mixture was concentrated, and the residue was neutralized with sodium hydrogencarbonate and extracted with ethyl acetate. The crude product was purified by silica gel column chromatography (DCM/MeOHv/v ═ 30/1) to give the desired 1- [3- [ [ 5-fluoro-2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0] hexadecyl-1 (12), 13, 15-trien-14-ylamino) pyrimidin-4-yl ] amino ] -1-piperidinyl ] prop-2-en-1-one as a 46mg white solid (26%).

LC-MS:m/z 488.2(ES+,M+1).

1- [ (3R) -3- [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0] tetraoxabicyclo]Hexadecyl-1 (12), 13, 15-trien-14-yl Amino) -9H-purin-6-yl]Amino group]-1-piperidinyl group]Synthesis of prop-2-en-1-one (Compound-3)

Scheme 5

Step 1 (N-protection). Chloromethyl 2, 2-dimethylpropionate (13.5g,90mmol) was added dropwise to a mixture of 2, 6-dichloro-9H-purine (11g,60mmol) and cesium carbonate (29g,90mmol) in 20mL of LDMSO. After stirring at room temperature for 48 hours, the mixture was poured into ice. The product was extracted with ethyl acetate and purified by silica gel column chromatography (petroleum ether/ethyl acetate 7/1 to 5/1) to give 5g of methyl (2, 6-dichloropurin-9-yl) 2, 2-dimethylpropionate (27%).

Step 2 (Cl-displacement). This step was the same as in step 1 of scheme 3, using methyl (2, 6-dichloropurin-9-yl) 2, 2-dimethylpropionate (2.1g, 6.93mmol) and 2, 4-dichloro-5-F-pyrimidine (1.66g, 8.32mmol) to give tert-butyl (3R) -3- [ [ 2-chloro-9- (2, 2-dimethylpropionyloxymethyl) purin-6-yl ] amino ] piperidine-1-carboxylate as a white foamy solid (2.3g, 71%).

Step 3 (palladium mediated aryl amination). This procedure is identical to scheme 3, step 4, using the product from the last step (1.4g, 3.0mmol), and 2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-trien-14-amine (0.86g, 3.6mmol, X-phos (0.15g,0.3 mmol), Pd2(dba)3(0.2g,0.2mmol) to give (3R) -3- [ [9- (2, 2-dimethylpropionyloxymethyl) -2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-trien-14-ylamino) purin-6-yl]Amino group]Piperidine-1-carboxylic acid tert-butyl ester (0.6g, 30%).

And 4, deprotection. To the diaminopurine obtained in the last step (0.6g, 0.9mol) in a mixture of 10mL of THF and 5mL of methanol at 0 deg.C was added 0.96mL of 3M aqueous sodium hydroxide solution. The mixture was stirred at room temperature for 5 hours, then 0.4mL of ammonium hydroxide (25-28%) was added. Stirring was then continued overnight and the product was extracted with ethyl acetate. After concentration, the crude product (0.35g) was obtained and used directly in the next step.

Step 5 (De-Boc). Boc deprotection was performed with 2ml TFA in 4ml lcm using the crude product obtained above. The reaction mixture was poured into water, sonicated and filtered to give 0.4g of a solid.

Step 6 (acrylation). Using the deprotected intermediate (0.15g,0.33mol), acrylation was performed in 12mL of a mixed solvent of THF/water/DMF (10/1/1) with acryloyl chloride (32. mu.L, 1.1eq.) and trimethylamine (0.1g, 1 mmol). After stirring overnight, the mixture was poured into water. The solid was collected to give 29mL of a yellow solid with a purity of 96.7% (17% yield).

LC-MS:510.2(ES+,M+1).

1- [ (3R) -3- [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0] tetraoxabicyclo]Hexadecyl-1 (12), 13, 15-trien-14-yl Amino) -9H-purin-6-yl]Amino group]-1-piperidinyl group]Prop-2-en-1-one (Compound-4)

Figure BDA0002467331340000601

Compound-4 is prepared by hydrogenating Compound-3 over a Pd/C catalyst in methanol.

LC-MS:512.2(ES+,M+1).

N- [3- [ [2- [4- [3- [ (2-ethyl-5, 5-dimethyl-1, 3-dioxan-2-yl) oxo]Propoxy group]Anilino radical]-5- Fluoro-pyrimidin-4-yl]Amino group]Phenyl radical]Prop-2-enamide (Compound-5)

Synthesis of Compound-5 was performed in the same manner as for Compound-1, using m-N-Boc aniline instead of 3-amino-N-Boc-piperidine as starting material.

LC-MS:m/z=425.1(ES+,M+H-142).

N- [3- [ [ 5-fluoro-2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-trien-14-yl Amino) pyrimidin-4-yl]Amino group]Phenyl radical]Synthesis of prop-2-enamides (Compound-6)

Figure BDA0002467331340000612

Synthesis of Compound-6 was performed in the same manner as for Compound-2, using m-N-Boc aniline instead of 3-amino-N-Boc-piperidine as starting material.

LC-MS:m/z=496.3(ES+,M+H).

1HNMR(DMSO-d6,400MHz)10.15(s,1H),9.41(s,1H),9.04(s,1H),8.08(s,1H),7.93(s,1H),7.48(m,2H),7.37(d,1H,J=4Hz),7.27(dd,1H,J=4,8Hz),7.21(dd,1H,J=4,8Hz),6.83(d,1H,J=8Hz),6.47(dd,1H,J=8,16Hz),6.24(dd,1H,J=4,16Hz),5.74(dd,1H,J=4,8Hz),4.00(br s,2H),3.88(br s,2H),3.67(br s,4H),3.43(br s,4H)。

N- [3- [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-trien-14-ylamino Radical) -9H-purin-6-yl]Amino group]Phenyl radical]Synthesis of prop-2-enamides (Compound-7)

Figure BDA0002467331340000621

Synthesis of Compound-7 was performed in the same manner as for Compound-3, using m-N-Boc aniline instead of 3-amino-N-Boc-piperidine as starting material.

LC-MS:m/z=518.3(ES+,M+H).

1HNMR(DMSO-d6,400MHz)10.27(s,1H),9.07(s,1H),8.10(s,1H),7.72(d,1H,J=8Hz),7.51~7.46(m,3H),6.92(d,1H,J=12Hz),6.50(dd,1H,J=12,20Hz),6.27(d,1H,J=20Hz),5.27(d,1H,J=12Hz),4.00(m,4H),3.68(m,8H)。

N- [3- [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-trien-14-ylamino Radical) -9H-purin-6-yl]Amino group]Phenyl radical]Propionamide (Compound-8)

Figure BDA0002467331340000622

Compound-8 is prepared by hydrogenating Compound-7 over a Pd/C catalyst in methanol.

LC-MS:m/z=520.3(ES+,M+H).

N- [2- [ [2- [4- [3- [ (2-ethyl-5, 5-dimethyl-1, 3-dioxan-2-yl) oxo]Propoxy group]Anilino radical]-5- Fluoro-pyrimidin-4-yl]Amino group]Phenyl radical]Prop-2-enamide (Compound-9)

Compound-9 is prepared by the same method as Compound-1 in scheme 3, except that it starts with tert-butyl N- (2-aminophenyl) carbamate and is not tert-butyl (3R) -3-aminopiperidine-1-carboxylate.

LC-MS: M/z 425.3(ES +, M + H-142 fragment).

1HNMR(CDCl3,400MHz)7.93(d,1H,J=3Hz),7.63(br s,1H),7.53(br s,1H),7.47(br s,1H),7.29(d,2H,J=8Hz),6.75(d,2H,J=8Hz),6.40(d,1H,J=20Hz),6.15(dd,1H,J=12,20Hz),5.73(d,1H,J=12Hz),4.05(t,2H,J=4hz),3.79(d,2H,J=12Hz),3.60(t,2H,J=8Hz),2.10(m,2H),1.75(q,2H,J=8Hz),1.13(s,3H),0.97(t,3H,J=8Hz),0.51(s,3H)。

N- [2- [ [ 5-fluoro-2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-trien-14-yl Amino) pyrimidin-4-yl]Amino group]Phenyl radical]Prop-2-enamide (Compound-10)

Compound-10 is prepared by the same method as Compound-2 in scheme 4, except that it starts with tert-butyl N- (2-aminophenyl) carbamate and is not tert-butyl (3R) -4-aminopiperidine-1-carboxylate.

LC-MS:m/z=496.2(ES+,M+H).

1HNMR(CD3OD,400MHz)7.96(s,1H),7.71(d,1H,J=8Hz),7.56(br d,1H,J=8Hz),7.34(s,1H),7.29(br t,2H,J=8Hz),6.92(d,1H,J=12Hz),6.86(d,1H,J=8Hz),6.33~6.39(m,2H),5.77(d,1H,J=12Hz),4.10(br t,2H,J=4Hz),3.87(br t,2H,J=4Hz),3.78(s,4H),3.75(s,4H)。

N- [2- [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-trien-14-ylamino Radical) -9H-purin-6-yl]Amino group]Phenyl radical]Prop-2-enamide (Compound-11)

Figure BDA0002467331340000632

Compound-11 is prepared by the same method as Compound-3 in scheme 4, except that it starts with tert-butyl N- (2-aminophenyl) carbamate and not tert-butyl (3R) -5-aminopiperidine-1-carboxylate.

LC-MS:m/z=518.2(ES+,M+H).

1HNMR(DMSO-d6,400MHz)12.6(s,1H),10.11(s,1H),8.81(s,1H),8.76(s,1H),7.92(d,1H,J=8Hz),7.50(d,1H,J=8Hz),7.25(m,2H),6.86(d,1H,J=12Hz),6.56(s,1H),6.41(dd,1H,J=12,20Hz),6.25(d,1H,J=20Hz),5.80(dd,1H,J=4,12Hz),4.00(br s,2H),3.89(br s,2H),3.68(m,8H)。

N- [2- [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-trien-14-ylamino Radical) -9H-purin-6-yl]Amino group]Phenyl radical]Propionamide (Compound-12)

Figure BDA0002467331340000641

Compound-12 was prepared by hydrogenation of Compound-11 in methanol at 10% Pd/C. Compound-12 can also be prepared by the method of preparing Compound-11, except that the propane is performed instead of the acryloyl.

LC-MS:m/z=520.2(ES+,M+H).

2-amino-N-methoxy-benzamide (I-3)

Scheme 6

10mL of 2M aqueous sodium hydroxide solution was added to a 25% aqueous solution of 0-methylhydroxylamine (1640mg,19.6mmol,3.2eq.) at 0 ℃. After stirring for 30 minutes, 1H-3, 1-benzoxazine-2, 4-dione (994mg,6.1mmol,1eq.) was added in portions. The reaction mixture was then stirred at room temperature for 3 hours. The desired product was extracted with ethyl acetate and titrated with diethyl ether to give a brown solid (600mg, 60%).

4, 6-dichloro-1-tetrahydropyran-2-yl-pyrazolo [3,4-d]Pyrimidine (I-4)

Scheme 7

Figure BDA0002467331340000652

To 4, 6-dichloro-1H-pyrazolo [3,4-d ] in 3mL THF and 3mL CDM]To a mixture of pyrimidine (200mg,1.06mmol,1.0eq) and p-toluenesulfonic acid (20.1mg,0.106mmol,0.1eq) was added 3, 4-dihydro-2H-pyran (134mg,1.6mmol,1.5 eq). The mixture was stirred at room temperature for 1 hour until TLC showed complete reaction. The reaction mixture was concentrated and then redissolved in 10mL of DCM and saturated NaHCO3Water and brine wash. The organic layer was dried and concentrated to give the desired intermediate I-4(287mg, 100%) as a white solid.

N- [3- (aminomethyl) -2-pyridinyl]-N-methylmethanesulfonamide (I-5)

Figure BDA0002467331340000653

Scheme 8

To 20ml of N-methylmethanesulfonamide at 20ml DMF (2.18g,20mmol,1.0eq) was added potassium tert-butoxide (t-BuOK) (2.24g,20mmol,1.0 eq.). After stirring at room temperature for 30 minutes. 2-fluoro-3-cyanopyridine (2.44g,20mmol,1.0eq) was added in one portion and the resulting mixture was heated to 80 ℃ for 2 hours. (TLC showed the reaction was complete). The reaction mixture was poured onto ice and extracted with ethyl acetate. The dried organic layer was concentrated and purified by silica gel column chromatography using petroleum ether/ethyl acetate (v/v ═ 1/1) as an eluent to give N- (3-cyano-2-pyridyl) -N-methyl-methanesulfonamide (4.0g, 93%).

4.0g of nitrile was hydrogenated in methanol containing 10% Pd/C overnight. TLC showed two products, m/z 216 and 413. The product was purified by column chromatography on silica gel using DCM/MeOH-10/1 as eluent to give the desired I-5(3.0g, 73%).

N- [ (1R, 2R) -2-hydroxycyclohexyl)]-N-methylmethanesulfonamide (I-6)

Figure BDA0002467331340000661

Scheme 9

To a mixture of (1R, 2R) -2-aminocyclohexanol (3.02g,20mmol), TEA (8.5mL,60mmol) and DMAP (0.75g,6.0mmol) in 30mL of DCM was added TBSCl (4.5g,30 mmol). The reaction mixture was stirred overnight. After washing with water, the organic layer was MgSO4And (5) drying. After filtration and concentration, the residue was used directly in the next step.

To the TBS ether obtained above was added triethylamine (4.6mL,34.2mmol) containing 20mL of LPCM followed by MsCl (1.0mL,13.7mmol) in an ice bath. The reaction mixture was stirred at room temperature overnight. After a conventional aqueous workup, the concentrated crude product was purified by silica gel column chromatography (petroleum ether/ethyl acetate, v/v ═ 8/1) to give N- [ (1R, 2R) -2- [ tert-butyl (dimethyl) silyl ] oxycyclohexyl ] methanesulfonamide (3.0g, two-step total yield 49%).

To a mixture of sulfonamide (1.5g,4.9mmol) obtained above and cesium carbonate (3.19g,9.8mmol) in 10mL of DMF was added methyl iodide (0.37mL,5.88 mmol). The reaction mixture was stirred at room temperature overnight. The reaction mixture was poured into water and the product was extracted with ethyl acetate. After drying and concentration, the product was purified by silica gel column chromatography (DCM/MeOH. cndot. 50/130/1) → to give 1.56g (97%) of N-m-sulfonamide.

Deprotection of the TBS group was carried out by reacting N-m-sulfonamide with TBAF (1.5equiv) to give the desired alcohol I-6(660mg, 66%).

N- [ (1R, 2R) -2-hydroxycyclopentyl group]-N-methylmethanesulfonamide (I-7)

Intermediate I-7 was prepared in the same manner as I-6 shown in scheme 8 using (1R, 2R) -2-aminocyclopentanol hydrochloride instead of (1R, 2R) -2-aminocyclohexanol as the starting material.

N- [2- [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-trien-14-ylamino Yl) -7H-pyrrolo [2, 3-d]Pyrimidin-4-yl]Amino group]Phenyl radical]Propionamide (Compound-13)

Figure BDA0002467331340000672

Scheme 1

Figure BDA0002467331340000681

The synthesis of Compound-13 is shown in scheme 10, which is similar to the synthesis of Compound-3 in scheme 5, except that 2, 4-dichloropyrrolo [2, 3-d ] pyrimidin-7-yl) methyl 2, 2-dimethylpropionate is used instead of 2, 6-dichloro-9H-purine as starting material, and the final step is propionylation, rather than acryloylation.

LC-MS:m/z=518.9(ES+,M+H).

1HNMR(DMSO-d6,400MHz)10.17(s,1H),9.74(s,1H),8.64(s,1H),7.72(s,1H),7.56(s,2H),7.18(m,t,J=9~12Hz),6.30(s,1H),4.01(br s,2H),3.92(br s,2H),3.68(m,8H),2.30(q,2H,J=6Hz),1.03(t,3H,J=6Hz)。

N- [2- [ [6- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-trien-14-ylamino Radical) -1H-pyrazolo [3,4-d]Pyrimidin-4-yl]Amino group]Phenyl radical]Propionamide (Compound-14)

Scheme 11

Figure BDA0002467331340000691

The synthesis scheme for Compound-14 is similar to that for Compound-13 in scheme 10, except that 4, 6-dichloro-1-tetrahydropyran-2-yl-pyrazolo [3,4-d ] pyrimidine (I-4) is used as a starting material in place of 2, 4-dichloropyrrolo [2, 3-d ] pyrimidin-7-yl) methyl 2, 2-dimethylpropionate, as shown in scheme 11.

LC-MS:m/z=519.9(ES+,M+H).

1HNMR(DMSO-d6,400MHz)12.9(s,1H),9.58(s,1H),9.18(s,1H),7.71~7.64(m,3H),7.63(s,1H),7.26(m,3H),6.87(s,1H),4.03(br s,2H),3.96(br s,2H),3.64~3.72(m,8H),2.30(q,2H,J=6Hz),1.03(t,3H,J=6Hz)。

N-methoxy-2- [ [6- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (16), 12, 14-triene-14- Alkylamino) -1H-pyrazolo [3,4-d]Pyrimidin-4-yl]Amino group]Benzamide (Compound-15)

Figure BDA0002467331340000701

Compound-15 was prepared in the same manner as Compound-14 in scheme 11, starting from intermediates I-3 and I-4.

LC-MS:m/z=521.9(ES+,M+H).

1HNMR(DMSO-d6,400MHz)13.91(s,1H),11.23(s,1H),10.91(s,1H),8.91(br s,1H),8.78(br s,1H),8.05(d,1H,J=6.0Hz),7.73(t,1H,J=6Hz),7.44(t,1H,J=5.6Hz),7.14(dd,1H,J=1.6,6.8Hz),7.05(d,1H,J=1.6Hz),4.081(br s,2H),4.03(br s,2H),4.01(s,3H),3.70(s,2H),3.61(br s,4H),3.59(s,2H)。

N-methyl-N- [ (1R, 2R) -2- [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (16), 12, 14- Trien-14-ylamino) -7H-pyrrolo [2, 3-d]Pyrimidin-4-yl]Oxo radical]Cyclohexyl radical]Methanesulfonamide (Compound-16)

Compound-16 was prepared in the same manner as Compound-13 in scheme 10, except starting from intermediate I-6 instead of tert-butyl N- (2-aminophenyl) carbamate.

LC-MS:m/z=561.9(ES+,M+H).

1HNMR(DMSO-d6,400MHz)11.41(s,1H),8.93(s,1H),7.57(s,1H),7.32(d,1H,J=6.0Hz),6.95(d,2H,J=5.6Hz),6.25(s,1H),5.37(br s,1H),4.10(br s,2H),4.01(br s,2H),3.93(br s,2H),3.91(br s,2H),3.78(m,6H),2.84(s,3H),2.51(s,3H),2.29(br s,1H),1.75(m 5H),1.07~1.40(m,5H)。

N-methyl-N- [ (1R, 2R) -2- [ [6- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (16), 12, 14- Trien-14-ylamino) -1H-pyrazolo [3,4-d]Pyrimidin-4-yl]Oxo radical]Cyclohexyl radical]Methanesulfonamide (Compound-17)

Compound-17 was prepared in the same manner as Compound-15 in scheme 11, except starting from intermediate I-6 instead of tert-butyl N- (2-aminophenyl) carbamate.

LC-MS:m/z=562.9(ES+,M+H).

1HNMR(DMSO-d6,400MHz)13.20(s,1H),9.36(s,1H),7.82(s,1H),7.55(s,1H),7.35(d,1H,J=5.6Hz),6.98(d,1H,J=6.8Hz),5.41(m,1H),4.16(br s,2H),4.07(br s,2H),3.90(m,1H),3.77(s,2H),3.71(br s,2H),3.65(s,4H).2.86(s,3H),2.68(s,3H),2.28(m,1H),1.78(m,3H),1.25~1.75(m,4H)。

N-methyl-N- [2- [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (16), 12, 14-triene-14- Phenylamino) -7H-pyrrolo [2, 3-d]Pyrimidin-4-yl]Oxo radical]Cyclopentyl group]Methanesulfonamide (Compound-18)

Figure BDA0002467331340000712

The Compound (Compound-18) was prepared in the same manner as Compound-16, except that it was prepared starting from intermediate I-7 instead of I-6.

LC-MS:m/z=547.9(ES+,M+H).

1HNMR(DMSO-d6,400MHz)11.51(s,1H),8.90(s,1H),7.58(d,1H,=3.2Hz),7.32(d,1H,J=6.0Hz),6.95(m,2H),6.21(br s,1H),5.37(br s,1H),4.07(br s,2H),4.01(brs,2H),3.55~3.75(m,6H),2.84(s,3H),2.75(s,3H),2.29(br s,1H),1.96(m,1H),1.72(m,4H)。

N-methyl-N- [ (1R, 2R) -2- [ [6- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (16), 12, 14- Trien-14-ylamino) -1H-pyrazolo [3,4-d]Pyrimidin-4-yl]Oxo radical]Cyclopentyl group]Methanesulfonamide (Compound-19)

Compound-19 was prepared in the same manner as Compound-16, except that it was started from intermediate I-7 instead of I-6.

LC-MS:m/z=548.9(ES+,M+H).

1HNMR(DMSO-d6,400MHz)9.31(s,1H),7.82(s,1H),7.52(s,1H),7.28(d,1H,=8.8Hz),5.54(t,1H,J=3.6Hz),4.36(m,1H),4.06(br s,2H),4.02(br s,2H),3.71(br s,2H),3.65(br s,2H),3.60(s,4H),2.84(s,3H),2.73(s,3H),2.29(m,1H),1.96(m,1H),1.72(m,4H)。

N-methyl-N- [4- [ [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (16), 12, 14-triene- 14-ylamino) -7H-pyrrolo [2, 3-d]Pyrimidin-4-yl]Amino group]Methyl radical]-3-pyridyl]Methanesulfonamide (Compound- 20)

Compound-20 was prepared in the same manner as Compound-16, except that it was started from intermediate I-5 instead of I-6.

LC-MS:m/z=569.9(ES+,M+H).

1HNMR(DMSO-d6,400MHz)11.0(s,1H),8.45(s,1H),8.43(s,1H),7.82(d,1H,=3.2Hz),7.75(br t,1H),7.50(s,1H),7.45(d,1H,J=6.0Hz),7.23(d,1H,J=6.0Hz),6.80(s,1H),6.75(s,1H),6.45(s,1H),4.78(s,2H)4.00(br s,2H),3.95(br s,2H),3.60~3.70(m,8H),3.23(s,3H),3.15(s,3H)。

N-methyl-N- [4- [ [ [6- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (16), 12, 14-triene- 14-ylamino) -1H-pyrazolo [3,4-d]Pyrimidin-4-yl]Amino group]Methyl radical]-3-pyridyl]Methanesulfonamide (Compound- 21) Synthesis of (2)

Figure BDA0002467331340000731

Compound-21 was prepared in the same manner as Compound-15 in scheme 11, except that it was prepared starting from intermediate I-5 instead of tert-butyl N- (2-aminophenyl) carbamate.

LC-MS:m/z=570.9(ES+,M+H).

1HNMR(DMSO-d6,400MHz)10.02(br s,1H),8.49(d,1H,=8Hz),7.86(d,1H,J=8Hz),7.43(dd,1H,J=8,10Hz),7.25(s,1H),6.97(m,2H),4.86(d,2H,J=6Hz),4.05(br s,2H),3.93(br s,2H),3.60~3.70(m,8H),3.17(s,3H),3.13(s,3H)。

N6- (1H-indol-4-yl) -N2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]Hexadecyl-1 (12), 13, 15-tris Synthesis of en-14-yl) -9H-purine-2, 6-diamine (Compound-22)

Figure BDA0002467331340000732

Compound-22 was prepared in the same manner as scheme 5, starting from 1H-indol-4-amine.

LC-MS:m/z=487.9(ES+,M+H).

1HNMR(DMSO-d6,400MHz)12.60(s,1H),11.14(s,1H),8.87(s,1H),8.77(s,1H),7.98(d,1H,=4Hz),7.69(s,1H),7.52(s,1H),7.29(s,1H),7.24(d,1H,J=8Hz),7.20(d,1H,J=8Hz),6.82(d,1H,J=8Hz),6.54(s,1H),6.42(m,1H),4.00(br s,2H),3.81(br s,2H),3.62~3.67(br d,8H)。

N6- (1H-indol-7-yl) -N2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]Hexadecyl-1 (12), 13, 15-tris Synthesis of en-14-yl) -9H-purine-2, 6-diamine (Compound-23)

Figure BDA0002467331340000741

Compound-23 was prepared in the same manner as scheme-5, starting from 1H-indol-7-amine.

LC-MS:m/z=487.9(ES+,M+H).

1HNMR(DMSO-d6,400MHz)11.20(s,1H),9.23(s,1H),8.68(s,1H),7.94(s,1H),7.70(t,1H,=4Hz),7.46(s,1H),7.32(d,1H,J=8Hz),7.28(d,1H,J=4Hz),6.99(d,1H,J=8Hz),6.76(d,1H,J=8Hz),6.45(s,1H),4.00(br s,2H),3.82(br s,2H),3.62~3.67(m,8H)。

N-methyl-N- [ (1R, 2R) -2- [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15- Trien-14-ylamino) thieno [2, 3-d]Pyrimidin-4-yl]Oxocyclopentyl group]Synthesis of methanesulfonamide (Compound-24)

Figure BDA0002467331340000742

The Compound (Compound-24) was prepared following the same procedure as Compound-18, starting from 2, 4-dichlorothieno [2, 3-d ] pyrimidine and the key intermediate I-5.

LC-MS:m/z=564.9(ES+,M+H).

1HNMR(CDCl3,400MHz)7.44(s,1H),7.15(d,1H,=4Hz),7.11(d,1H,J=8Hz),6.96(m,3H),5.51(m,1H),4.56(q,1H,J=8Hz),4.22(br s,2H),4.18(br s,2H),3.90(br s,2H),3.85(br s,2H),3.81(s,4H),2.85(s,3H),2.79(s,3H),2.31(m,1H),2.02(m,1H),1.80(m,4H)。

N- [ 5-methyl-2- [ [6- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-triene-14- Alkylamino) -1H-pyrazolo [3,4-d]Pyrimidin-4-yl]Amino group]Phenyl radical]Prop-2-enamine (Compound-26)

The compounds are prepared according to the same procedure as scheme 11 using N- (2-aminophenyl) propionamide reacted with 4, 6-dichloro-1-tetrahydropyran-2-yl-pyrazolo [3,4-d ] pyrimidine in a chloro-displacement step followed by palladium mediated aryl amination with 2, 5, 8, 11-tetraoxabicyclo [10.4.0] hexadecyl-1 (16), 12, 14-trien-14-amine.

The compound was prepared according to the same procedure as scheme 11 using tert-butyl N- (2-amino-5-methyl-phenyl) carbamate reacted with 4, 6-dichloro-1-tetrahydropyran-2-yl-pyrazolo [3,4-d ] pyrimidine in a chloro-displacement step followed by palladium mediated aryl amination with 2- [2- (4-amino-2-methoxy-phenoxy) ethoxy ] ethanol and final acrylation after Boc-deprotection,

LC-MS:m/z 532.2(ES+,M+H).

HNMR(DMSO-d6,300MHz)9.86(s,1H),9.72(s,1H),9.21(s,1H),8.87(s,1H),7.59(s,1H),7.50(s,1H),7.44(d,1H,J=6Hz),7.21(br s,1H),7.02(t,1H,J=6Hz),6.82(d,1H,J=6Hz),6.44(dd,1H,J=6,12Hz),6.18(d,1H,J=12Hz),5.68(m,1H),3.99(d,2H,J=6Hz),3.90(br s,1H),3.67(s,2H),3.64(s,2H),3.59(s,4H),2.31(s,3H)。

n- [ 5-methyl-2- [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-triene-14- Alkylamino) -9H-purin-6-yl]Amino group]Phenyl radical]Acetamide (Compound-27)

Figure BDA0002467331340000752

The compounds are prepared according to the same procedure as scheme 11 using N- (2-amino-5-methyl-phenyl) acetamide reacted with (2, 6-dichloropurin-9-yl) methyl 2, 2-dimethylpropionate in a chloro-displacement step followed by palladium mediated aryl amination with 2, 5, 8, 11-tetraoxabicyclo [10.4.0] hexadecyl-1 (16), 12, 14-trien-14-amine.

LC-MS:m/z 520.4(ES+,M+H).

HNMR(DMSO-d6,300MHz)9.82(s,1H),8.72(s,1H),8.56(s,1H),7.86(s,1H),7.71(d,1H,J=6Hz),7.48(d,1H,J=6Hz),7.19(m,2H),7.00(d,1H,J=6Hz),6.82(d,1H,J=6Hz),3.98(d,2H,J=6Hz),3.90(br s,1H),3.67(s,2H),3.64(s,2H),3.59(s,4H),2.27(s,3H),2.01(s,3H)。

N- [ 5-fluoro-2- [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-trien-14-yl Amino) -9H-purin-6-yl]Amino group]Phenyl radical]Acetamide (Compound-28)

The compounds are prepared according to the same procedure as scheme 11 using N- (2-amino-5-fluoro-phenyl) acetamide reacted with (2, 6-dichloropurin-9-yl) methyl 2, 2-dimethylpropionate in a chloro-displacement step followed by palladium mediated aryl amination with 2, 5, 8, 11-tetraoxabicyclo [10.4.0] hexadecyl-1 (16), 12, 14-trien-14-amine.

LC-MS:m/z 524.4(ES+,M+H).

HNMR(DMSO-d6,300MHz)9.85(s,1H),8.91(s,1H),8.60(s,1H),7.99(d,1H,J=6Hz),7.91(s,1H),7.44(d,1H,J=3Hz),7.31(m,2H),6.92(m,1H),6.85(d,1H,J=6Hz),3.98(d,2H,J=6Hz),3.90(br s,1H),3.67(s,2H),3.64(s,2H),3.59(s,4H),2.05(s,3H)。

N- [2- [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-trien-14-ylamino Radical) -9H-purin-6-yl]Amino group]-5- (trifluoromethyl) phenyl]Acetamide (Compound29)

Figure BDA0002467331340000762

The compounds are prepared according to the same procedure as in scheme 11, using N- [ 2-amino-5- (trifluoromethyl) phenyl ] acetamide to react with methyl (2, 6-dichloropurin-9-yl) 2, 2-dimethylpropionate in a chloro-displacement step followed by palladium mediated aryl amination with 2, 5, 8, 11-tetraoxabicyclo [10.4.0] hexadecyl-1 (16), 12, 14-trien-14-amine.

LC-MS:m/z 574.4(ES+,M+H).

HNMR(DMSO-d6,300MHz)9.85(s,1H),8.91(s,1H),8.60(s,1H),7.99(d,1H,J=6Hz),7.91(s,1H),7.44(d,1H,J=3Hz),7.31(m,2H),6.92(m,1H),6.85(d,1H,J=6Hz),3.98(d,2H,J=6Hz),3.90(br s,1H),3.67(s,2H),3.64(s,2H),3.59(s,4H),2.05(s,3H)。

N- [3- [ [ 5-fluoro-2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15-trien-14-yl Amino) pyrimidin-4-yl]Phenyl radical]Prop-2-enamine (Compound-30)

The compounds are prepared by the Suzuki reaction of (3-aminophenyl) boronic acid with 2, 4-dichloro-5-F-pyrimidine, followed by acrylation and palladium mediated aryl amination with 2, 5, 8, 11-tetraoxabicyclo [10.4.0] hexadecyl-1 (16), 12, 14-trien-14-amine.

LC-MS:m/z 481.2(ES+,M+H).

HNMR(DMSO-d6,300MHz)10.30(s,1H),9.60(s,1H),8.58(s,1H),8.39(s,1H),7.79(d,1H,J=6Hz),7.71(d,1H,J=6Hz),7.58(s,1H),7.51(t,1H,J=6Hz),7.24(d,1H,J=6Hz),6.94(d,1H,J=6Hz),6.44(dd,1H,J=6,12Hz),6.24(d,1H,J=12Hz),5.75(m,1H),3.99(d,2H,J=6Hz),3.90(br s,1H),3.67(s,2H),3.64(s,2H),3.59(s,4H)。

N-methyl-N- [ (1R, 2R) -2- [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15- Trien-14-ylamino) -9H-purin-6-yl]Oxo radical]Indan-1-yl]Methanesulfonamide (Compound-31)

The compound is prepared according to the same procedure as in scheme 5, using N- [ (1R, 2R) -2-hydroxyindan-1-yl ] -N-methylmethanesulfonamide in a chloro-metathesis step with methyl (2, 6-dichloropurin-9-yl) 2, 2-dimethylpropionate in the presence of cesium carbonate containing dioxane, followed by palladium mediated aryl amination with 2, 5, 8, 11-tetraoxabicyclo [10.4.0] hexadecyl-1 (16), 12, 14-trien-14-amine.

LC-MS:m/z 597.2(ES+,M+H).

HNMR(DMSO-d6,300MHz)12.72(s,1H),9.19(s,1H),7.96(s,1H),7.57(s,1H),7.32(m,3H),7.17(m,2H),6.90(d,1H,J=6Hz),5.83(m,1H),5.61(d,1H,J=6Hz),3.98(d,2H,J=6Hz),3.90(br s,1H),3.67(s,2H),3.64(s,2H),3.59(s,4H),3.03(s,3H),2.62(s,3H)。

N-methyl-N- [ (1R, 2R) -2- [ [2- (2, 5, 8, 11-tetraoxabicyclo [10.4.0]]Hexadecyl-1 (12), 13, 15- Trien-14-ylamino) -5- (trifluoromethyl) -4-pyridinyl]Oxo radical]Indan-1-yl]Methanesulfonamide (Compound-32)

Figure BDA0002467331340000781

The compounds are prepared by iodine displacement of 2-chloro-4-iodo-5- (trifluoromethyl) pyridine with N- [ (1R, 2R) -2-hydroxyindan-1-yl ] -N-methylmethanesulfonamide in the presence of sodium hydride in dioxane, followed by palladium mediated aryl amination with 2, 5, 8, 11-tetraoxabicyclo [10.4.0] hexadecyl-1 (16), 12, 14-trien-14-amine.

LC-MS:m/z 624.4(ES+,M+H).

HNMR(DMSO-d6,300MHz)9.25(s,1H),8.22(s,1H),7.32(m,4H),7.23(m,1H),7.09(m,1H),6.95(d,1H,J=6Hz),6.57(s,1H),5.53(d,1H,J=6Hz),5.30(m,1H),3.98(d,2H,J=6Hz),3.90(br s,1H),3.67(s,2H),3.64(s,2H),3.59(s,4H),2.97(s,3H),2.59(s,3H)。

105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:用于治疗非小细胞肺癌的奧希替尼

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!