Isoxazole compounds for the treatment of diseases associated with HBV infection

文档序号:1301527 发布日期:2020-08-07 浏览:12次 中文

阅读说明:本技术 用于治疗与hbv感染相关的疾病的异噁唑化合物 (Isoxazole compounds for the treatment of diseases associated with HBV infection ) 是由 S.库杜克 S.M.H.文德维尔 于 2018-12-21 设计创作,主要内容包括:本文提供了异噁唑化合物,其药物组合物,制备此类化合物和组合物的方法,以及抑制、遏制或预防受试者中HBV感染的方法。<Image he="430" wi="587" file="DDA0002547303920000011.GIF" imgContent="drawing" imgFormat="GIF" orientation="portrait" inline="no"></Image>(Provided herein are isoxazole compounds, pharmaceutical compositions thereof, methods of making such compounds and compositions, and methods of inhibiting, suppressing, or preventing HBV infection in a subject.)

1. A compound, and pharmaceutically acceptable salts, solvates, stereoisomers, isotopic variations or N-oxides thereof, said compound having the structure of formula (I):

wherein

X1And X2Each independently is O or N, wherein when X is1When is O, X2Is N, and wherein when X1When is N, X2Is O;

het is a 5 or 6 membered heteroaryl ring selected from the group consisting of: thiazoles, pyrimidines and pyrazoles, wherein each thiazole, pyrimidine and pyrazole is optionally substituted with at least one R7Substitution; wherein R is7Selected from the group consisting of: halo and C1-4Alkyl radical, wherein said C1-4The alkyl group is optionally substituted with one or more groups selected fromSubstituted with a substituent of the group (b): OH and C1-4A haloalkyl group;

R1、R2、R3and R4Each independently is H or C1-4An alkyl group;

X3and X4Each independently is C-R6Or N, wherein when X3Is C-R6Or N is, X4Is C-R6And wherein when X4Is C-R6Or N is, X3Is C-R6

R5And R6Each independently selected from the group consisting of: H. halo, C1-4Haloalkyl and CN; and is

n is 0, 1 or 2.

2. The compound of claim 1, wherein X1Is O and X2Is N.

3. The compound of claim 1, wherein X1Is N and X2Is O.

4. The compound of claim 1, wherein Het is

5. The compound of claim 1, wherein Het is

6. The compound of claim 1, wherein Het is

7. The compound of claim 1, wherein R1Is H.

8. The compound of claim 1, wherein R1Is CH3

9. The compound of claim 1, wherein R2Is H.

10. The compound of claim 1, wherein R2Is CH3

11. The compound of claim 1, wherein R1And R2Each is CH3

12. The compound of claim 1, wherein R3Is H.

13. The compound of claim 1, wherein R3Is CH3

14. The compound of claim 1, wherein R4Is H.

15. The compound of claim 1, wherein R4Is CH3

16. The compound of claim 1, wherein X3Is C-R6And R is6Is H.

17. The compound of claim 1, wherein X3Is N.

18. The compound of claim 1, wherein X4Is C-R6And R is6Is halogenated.

19. The compound of claim 1, wherein X4Is C-R6And R is6Is F.

20. The compound of claim 1, wherein X4Is N.

21. The compound of claim 1, wherein R5Selected from halo, C1-4Haloalkyl and CN.

22. The compound of claim 1, wherein n is 0.

23. The compound of claim 1, wherein n is 1.

24. The compound of claim 1, wherein n is 2.

25. The compound of claim 1, wherein R7Is H, F or CF2H。

26. The compound of claim 1, wherein

X1Is N;

X2is O; and is

Het isWherein R is7Is H.

27. The compound of claim 26, wherein

R1、R2And R3Each is H; and is

R4Is CH3

28. The compound of claim 27, wherein

X3Is C-R6Wherein R is6Selected from the group consisting of: H. CF (compact flash)3And CN;

X4is C-R6Wherein R is6Is F;

n is 0 or 1; and is

R5Is CF3Or CN.

29. The compound of claim 27, wherein

X3Is C-R6Wherein R is6Selected from the group consisting of: H. CF (compact flash)2H and Br;

X4is N;

n is 1 or 2; and is

Each R5Independently selected from the group consisting of: F. CF (compact flash)2H and Br.

30. The compound of claim 1, wherein

X1Is O;

X2is N; and is

Het isWherein R is7Is H.

31. The compound of claim 30, wherein

R1、R2And R3Each is H; and is

R4Is CH3

32. The compound of claim 31, wherein

X3Is C-R6Wherein R is6Selected from the group consisting of: H. CF (compact flash)3And CN;

X4is C-R6Wherein R is6Is F;

n is 0 or 1; and is

R5Is CF3Or CN.

33. The compound of claim 31, wherein

X3Is C-R6Wherein R is6Selected from the group consisting of: H. CF (compact flash)2H and Br;

X4is N;

n is 1 or 2; and is

Each R5Independently selected from the group consisting of: F. CF (compact flash)2H and Br.

34. The compound of claim 1, and pharmaceutically acceptable salts, solvates, stereoisomers, isotopic variations or N-oxides thereof, having the structure of formula (IA):

wherein

Het is

R1Is H or CH3

R2Is H or CH3

R3Is H;

R4is H or CH3

X3Is C-R6Wherein R is6Is H;

X4is N or C-R6Wherein R is6Is F;

each R5Independently selected from the group consisting of: br, F, CF2H、CF3And CN;

n is 1 or 2; and is

R7Is H.

35. The compound of claim 1, and pharmaceutically acceptable salts, solvates, stereoisomers, isotopic variations or N-oxides thereof, having the structure of formula (IB):

wherein the content of the first and second substances,

het is

R1Is H or CH3

R2Is H or CH3

R3Is H;

R4is H or CH3

X3Is C-R6Wherein R is6Is H;

X4is N or C-R6Wherein R is6Is F;

each R5Independently selected from the group consisting of: br, F, CF2H、CF3And CN;

n is 1 or 2; and is

R7Is H.

36. A compound selected from the group consisting of:

(S) -N- (3-cyano-4-fluorophenyl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 3-c ] pyridine-5 (4H) -carboxamide;

(S) -N- (2- (difluoromethyl) -3-fluoropyridin-4-yl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 3-c ] pyridine-5 (4H) -carboxamide;

(S) -N- (4-fluoro-3- (trifluoromethyl) phenyl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 3-c ] pyridine-5 (4H) -carboxamide;

(S) -N- (2-bromo-3-fluoropyridin-4-yl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 3-c ] pyridine-5 (4H) -carboxamide;

(S) -N- (3-cyano-4-fluorophenyl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 5-c ] pyridine-5 (4H) -carboxamide;

(S) -N- (2- (difluoromethyl) -3-fluoropyridin-4-yl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 5-c ] pyridine-5 (4H) -carboxamide;

(S) -N- (4-fluoro-3- (trifluoromethyl) phenyl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 5-c ] pyridine-5 (4H) -carboxamide;

(S) -N- (2-bromo-3-fluoropyridin-4-yl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 5-c ] pyridine-5 (4H) -carboxamide;

n- (3-cyano-4-fluorophenyl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 3-c ] pyridine-5 (4H) -carboxamide;

n- (4-fluoro-3- (trifluoromethyl) phenyl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 3-c ] pyridine-5 (4H) -carboxamide;

n- (2- (difluoromethyl) -3-fluoropyridin-4-yl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 3-c ] pyridine-5 (4H) -carboxamide;

n- (2-bromo-3-fluoropyridin-4-yl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 3-c ] pyridine-5 (4H) -carboxamide;

n- (3-cyano-4-fluorophenyl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 5-c ] pyridine-5 (4H) -carboxamide;

n- (4-fluoro-3- (trifluoromethyl) phenyl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 5-c ] pyridine-5 (4H) -carboxamide;

n- (2- (difluoromethyl) -3-fluoropyridin-4-yl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 5-c ] pyridine-5 (4H) -carboxamide; and

n- (2-bromo-3-fluoropyridin-4-yl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 5-c ] pyridine-5 (4H) -carboxamide;

and pharmaceutically acceptable salts, solvates or N-oxides thereof.

37. A pharmaceutical composition comprising:

(A) at least one compound selected from compounds having the formula (I) wherein:

wherein

X1And X2Each independently is O or N, wherein when X is1When is O, X2Is N, and wherein when X1When is N, X2Is O;

het is a 5 or 6 membered heteroaryl ring selected from the group consisting of: thiazoles, pyrimidines and pyrazoles, wherein each thiazole, pyrimidine and pyrazole is optionally substituted with at least one R7Substitution; wherein R is7Selected from the group consisting of: halo and C1-4Alkyl radical, wherein said C1-4The alkyl group is optionally substituted with one or more substituents selected from the group consisting of: OH and C1-4A haloalkyl group;

R1、R2、R3and R4Each independently is H or C1-4An alkyl group;

X3and X4Each independently is C-R6Or N, wherein when X3Is C-R6Or N is, X4Is C-R6And wherein when X4Is C-R6Or N is, X3Is C-R6

R5And R6Each independently selected from the group consisting of: H. halo, C1-4Haloalkyl and CN; and is

n is 0, 1 or 2;

and pharmaceutically acceptable salts, solvates, stereoisomers, isotopic variations or N-oxides of the compounds having formula (I); and

(B) at least one pharmaceutically acceptable excipient.

38. A pharmaceutical composition comprising at least one compound of claim 36 and at least one pharmaceutically acceptable excipient.

39. A method of treating an HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of at least one compound of claim 1.

40. A method of inhibiting or reducing the formation or presence of HBV DNA-containing particles or HBV RNA-containing particles in an individual in need thereof, the method comprising administering to the individual a therapeutically effective amount of a compound of claim 1.

41. The method of claim 39 or 40, further comprising administering to the individual at least one additional therapeutic agent.

42. The method according to claim 41, wherein the additional therapeutic agent is selected from at least one of the group consisting of HBV polymerase inhibitors, immunomodulators, interferons, viral entry inhibitors, viral maturation inhibitors, capsid assembly modulators, reverse transcriptase inhibitors, cyclophilin/TNF inhibitors, T L R-agonists and HBV vaccines.

43. The method of claim 41, wherein the therapeutic agent is a reverse transcriptase inhibitor selected from the group consisting of: zidovudine, didanosine, zalcitabine, ddA, stavudine, lamivudine, abacavir, emtricitabine, entecavir, aliscitabine, altiveline, ribavirin, acyclovir, famciclovir, valacyclovir, valganciclovir, tenofovir, adefovir, PMPA, cidofovir, efavirenz, nevirapine, delavirdine, and eltravirine.

44. The method of claim 41, wherein the therapeutic agent is a T L R agonist selected from the group consisting of SM360320 (9-benzyl-8-hydroxy-2- (2-methoxy-ethoxy) adenine) and AZD 8848([3- ({ [3- (6 amino 2-butoxy-8-oxo-7, 8-dihydro-9H-purin-9-yl) propyl ] [3- (4-morpholinyl) propyl ] amino } methyl) phenyl ] acetic acid methyl ester).

45. The method of claim 41, wherein the therapeutic agent is an HBV vaccine selected from the group consisting of RECOMBIVAX HB, ENGERIX-B, E L OVAC B, GENEVAC-B, and SHANTVAC B.

46. The present disclosure relates to compounds having formula (I) for use in the treatment of HBV infection.

47. A compound having the formula (X):

wherein

X1And X2Each independently is O or N, wherein when X is1When is O, X2Is N, and wherein when X1When is N, X2Is O;

het is a 5 or 6 membered heteroaryl ring selected from the group consisting of: thiazoles, pyrimidines and pyrazoles, wherein each thiazole, pyrimidine and pyrazole is optionally substituted with at least one R7Is substituted in which R7Selected from the group consisting of: halo and C1-4Alkyl radical, wherein said C1-4The alkyl group is optionally substituted with one or more substituents selected from the group consisting of: OH and C1-4A haloalkyl group;

R1、R2、R3and R4Each independently is H or C1-4An alkyl group.

48. A process for preparing a compound having formula (I), the process comprising combining a compound having formula (X) with a compound having formula (XI) in the presence of a base and a solvent

Wherein

X1And X2Each independently is O or N, wherein when X is1When is O, X2Is N, and wherein when X1When is N, X2Is O;

het is a 5 or 6 membered heteroaryl ring selected from the group consisting of: thiazoles, pyrimidines and pyrazoles, wherein each thiazole, pyrimidine and pyrazole is optionally substituted with at least one R7Is substituted in which R7Selected from the group consisting of: halo and C1-4Alkyl radical, wherein said C1-4The alkyl group is optionally substituted with one or more substituents selected from the group consisting of: OH and C1-4A haloalkyl group;

R1、R2、R3and R4Each independently is H or C1-4An alkyl group;

X3and X4Each independently is C-R6Or N, wherein when X3Is C-R6Or N is, X4Is C-R6And wherein when X4Is C-R6Or N is, X3Is C-R6

R5And R6Each independently selected from the group consisting of: H. halo, C1-4Haloalkyl and CN; and is

n is 0, 1 or 2.

Technical Field

The present disclosure relates to isoxazole compounds, pharmaceutical compositions comprising these compounds, chemical processes for preparing these compounds, and their use in treating diseases associated with HBV infection in animals, particularly humans.

Background

Chronic Hepatitis B Virus (HBV) infection is a serious global health problem affecting more than 5% of the world population (more than 3.5 million people worldwide, 1.25 million people in the united states).

Despite the availability of prophylactic HBV vaccines, the burden of chronic HBV infection remains a significant unmet global medical problem because treatment options are not ideal in most areas of developing countries and the rate of new infections continues to be constant.

HBV capsid protein plays an important role in the life cycle of the virus. The HBV capsid/core protein forms a metastable viral particle or protein shell that protects the viral genome during intercellular passage and also plays a central role in viral replication, including genome encapsidation, genome replication, and virion morphogenesis and egress. The capsid structure also reacts to environmental cues to allow non-encapsulation after viral entry. Consistently, it has been found that proper timing of capsid assembly and disassembly, proper capsid stability, and function of the core protein are critical for viral infectivity.

The critical functions of HBV capsid proteins impose strict evolutionary constraints on the viral capsid protein sequence, leading to the observation of low sequence variability and high conservation. Consistently, mutations in the HBV capsid that disrupt its assembly are lethal, and mutations that disturb capsid stability can severely attenuate viral replication. The high functional constraints on the multifunctional HBV core/capsid protein are consistent with high sequence conservation, as many mutations are detrimental to function. In fact, the core/capsid protein sequences are > 90% identical in HBV genotype and show only a few polymorphic residues. Therefore, it is difficult to select drug resistance to HBV core/capsid protein binding compounds without having a major impact on virus replication adaptability.

Reports describe compounds that bind to the viral capsid and inhibit HIV, rhinovirus, and HBV replication, providing strong pharmacological evidence for the concept of viral capsid proteins as targets for antiviral drugs.

There is a need in the art for therapeutic agents that can increase the suppression of viral production and can treat, ameliorate and/or prevent HBV infection. Administration of such therapeutic agents to HBV infected patients as monotherapy or in combination with other HBV treatments or adjunctive therapies would increase the likelihood of reduced viral load, improved prognosis, impaired disease progression and enhanced seroconversion.

In view of the clinical impact of HBV on infected patients, there is a need to identify compounds that are capable of increasing viral suppression and compounds that can treat, ameliorate and/or prevent HBV infection, which represents an attractive approach to the development of new therapeutic agents. Such compounds are provided herein.

Disclosure of Invention

The present disclosure relates to general and preferred embodiments as defined in the independent and dependent claims appended hereto, respectively, which are incorporated herein by reference. In particular, the disclosure relates to compounds having formula (I):

and pharmaceutically acceptable salts, stereoisomers, isotopic variations, N-oxides, or solvates of the compounds having formula (I).

In the examplesIn, R1、R2、R3And R4Each of which is independently hydrogen or C1-4An alkyl group.

Het represents a 5-or 6-membered heteroaryl ring. In embodiments, the 5-or 6-membered heteroaryl ring is a thiazole, a pyrimidine, or a pyrazole. Heteroaryl rings may be optionally substituted with at least one R7And (4) substitution. Each R7May be independently halo, C1-4Alkyl, C substituted by hydroxy (OH)1-4Alkyl, or C1-4A haloalkyl group.

In the examples, X1And X2Is independently oxygen (O) or nitrogen (N). In the examples, when X1When is O, X2Is N. In the examples, when X1When is N, X2Is O.

In the examples, X3And X4Each of which is independently C-R6Or nitrogen. In the examples, when X3Is C-R6Or when nitrogen is present, X4Is C-R6. In the examples, when X4Is C-R6Or when nitrogen is present, X3Is C-R6. Each R6May independently be: hydrogen, halo, C1-4Haloalkyl or Cyano (CN).

None, one or two RThe substituent-n is 0, 1 or 2. Each R5May independently be: hydrogen, halo, C1-4Haloalkyl or Cyano (CN).

Additional embodiments include pharmaceutically acceptable salts of compounds having formula (I), pharmaceutically acceptable prodrugs of compounds having formula (I), pharmaceutically active metabolites of compounds having formula (I), and enantiomers and diastereomers of compounds having formula (I), and pharmaceutically acceptable salts thereof.

In the examples, the compounds having formula (I) are compounds selected from those classes described or exemplified in the detailed description below.

The disclosure also relates to pharmaceutical compositions comprising one or more compounds having formula (I), pharmaceutically acceptable salts of compounds having formula (I), pharmaceutically acceptable prodrugs of compounds having formula (I), and pharmaceutically active metabolites having formula (I). The pharmaceutical composition may further comprise one or more pharmaceutically acceptable excipients or one or more other agents or therapies.

The disclosure also relates to methods of use or uses of compounds having formula (I). In embodiments, the compounds having formula (I) are used to treat or ameliorate Hepatitis B Virus (HBV) infection, increase suppression of HBV production, interfere with HBV capsid assembly or other HBV viral replication steps or HBV products. The method comprises administering to a subject in need of such a method an effective amount of at least one compound having formula (I), a pharmaceutically acceptable salt of a compound having formula (I), a pharmaceutically acceptable prodrug of a compound having formula (I), and a pharmaceutically active metabolite of a compound having formula (I). Additional examples of methods of treatment are set forth in the detailed description.

The disclosure also relates to compounds having formula (X):

in the examples, R1、R2、R3And R4Each of which is independently hydrogen or C1-4An alkyl group.

Het represents a 5-or 6-membered heteroaryl ring. In embodiments, the 5-or 6-membered heteroaryl ring is a thiazole, a pyrimidine, or a pyrazole. Heteroaryl rings may be optionally substituted with at least one R7And (4) substitution. Each R7May be independently halo, C1-4Alkyl, C substituted by hydroxy (OH)1-4Alkyl, or C1-4A haloalkyl group.

In the examples, X1And X2Is independently oxygen (O) or nitrogen (N). In the examples, when X1When is O, X2Is N. In the examples, when X1When is N, X2Is O.

The disclosure also relates to a process or method of making a compound having formula (I), comprising combining a compound having formula (X) with a compound having formula (XI) in the presence of a base and a solvent.

In the examples, R1、R2、R3And R4Each of which is independently hydrogen or C1-4An alkyl group.

Het represents a 5-or 6-membered heteroaryl ring. In embodiments, the 5-or 6-membered heteroaryl ring is a thiazole, a pyrimidine, or a pyrazole. Heteroaryl rings may be optionally substituted with at least one R7And (4) substitution. Each R7May be independently halo, C1-4Alkyl, C substituted by hydroxy (OH)1-4Alkyl, or C1-4A haloalkyl group.

In the examples, X1And X2Is independently oxygen (O) or nitrogen (N). In the examples, when X1When is O, X2Is N. In the examples, when X1When is N, X2Is O.

In the examples, X3And X4Each of which is independently C-R6Or nitrogen. In the examples, when X3Is C-R6Or when nitrogen is present, X4Is C-R6. In the examples, when X4Is C-R6Or when nitrogen is present, X3Is C-R6. Each R6May independently be: hydrogen, halo, C1-4Haloalkyl or Cyano (CN).

None, one or two R5The substituent-n is 0, 1 or 2. Each R5May independently be: hydrogen, halo, C1-4Haloalkyl or Cyano (CN).

In embodiments, the base is an alkylamine, such as Triethylamine (TEA), Diisopropylethylamine (DIEA), and the like.

In embodiments, the solvent is a halogenated alkane, such as Dichloromethane (DCM), Dichloroethane (DCE), and the like.

It is an object of the present disclosure to overcome or ameliorate at least one of the disadvantages of the conventional approaches and/or the prior art or to provide a useful alternative thereto. Further embodiments, features, and advantages of the present disclosure will be apparent from the following detailed description and from the practice of the disclosed subject matter.

Detailed Description

Further embodiments, features, and advantages of the presently disclosed subject matter will be apparent from the following detailed description of the presently disclosed subject matter, and from the practice thereof. For the sake of brevity, publications (including patents) cited in this specification are hereby incorporated by reference.

Provided herein are compounds having formula (I), including compounds having formulae (IA) and (IB), and pharmaceutically acceptable salts, pharmaceutically acceptable prodrugs, and pharmaceutically active metabolites of the disclosed compounds thereof.

In one aspect, provided herein are compounds having formula (I), and pharmaceutically acceptable salts, stereoisomers, isotopic variations, N-oxides, or solvates thereof,

wherein

R1、R2、R3And R4Each independently is H or C1-4An alkyl group;

het is a 5 or 6 membered heteroaryl ring selected from the group consisting of: thiazoles, pyrimidines and pyrazoles, wherein each thiazole, pyrimidine and pyrazole is optionally substituted with at least one R7Substitution;

X1and X2Each independently is O or N;

wherein when X is1When is O, X2Is N; wherein when X is1When is N, X2Is O;

X3and X4Each independently is C-R6Or N;

wherein when X is3Is C-R6Or N is, X4Is C-R6(ii) a Wherein when X is4Is C-R6Or N is, X3Is C-R6

R5And R6Each independently selected from the group consisting of: H. halo, C1-4Haloalkyl and CN;

n is 0, 1 or 2; and is

R7Selected from the group consisting of: halo, C1-4Alkyl, C substituted by OH1-4Alkyl, and C1-4A haloalkyl group;

and pharmaceutically acceptable salts, solvates, stereoisomers, isotopic variations or N-oxides of the compounds having formula (I).

In embodiments, the compound having formula (I) is wherein R1A compound which is H.

In embodiments, the compound having formula (I) is wherein R1Is CH3The compound of (1).

In embodiments, the compound having formula (I) is wherein R1And R2Each is CH3The compound of (1).

In embodiments, the compound having formula (I) is wherein R4A compound which is H.

In embodiments, the compound having formula (I) is wherein R4Is CH3The compound of (1).

In embodiments, the compound having formula (I) is wherein Het isThe compound of (1).

In embodiments, the compound having formula (I) is wherein Het isThe compound of (1).

In embodiments, the compound having formula (I) is wherein Het isThe compound of (1).

In embodiments, the compound having formula (I) is wherein X3Is C-R6And R is6A compound which is H.

In embodiments, the compound having formula (I) is wherein X3A compound which is N.

In embodiments, the compound having formula (I) is wherein X4Is C-R6And R is6Are halogenated compounds.

In embodiments, the compound having formula (I) is wherein X4Is C-R6And R is6A compound which is F.

In embodiments, the compound having formula (I) is wherein X4A compound which is N.

In embodiments, the compound having formula (I) is wherein R5Selected from halo, C1-4A haloalkyl and CN.

In embodiments, the compound having formula (I) is wherein R5Is fluorine.

In embodiments, the compound having formula (I) is a compound wherein n is 0.

In embodiments, the compound having formula (I) is a compound wherein n is 1.

In embodiments, the compound having formula (I) is a compound wherein n is 2.

In embodiments, the compound having formula (I) is wherein R7Is H, F, CF2H or (CHF)2The compound of (1).

In embodiments, the compound having formula (I) is wherein X1Is O, X2A compound which is N.

In embodiments, the compound having formula (I) is wherein X1Is N, X2A compound which is O.

Embodiments of the present disclosure are compounds of formula (I) having formula (IA):

wherein

Het is

R1Is H or CH3

R2Is H or CH3

R3Is H;

R4is H or CH3

X3Is C-R6Wherein R is6Is H, Br, F, CF2H (or CHF)2、CF3Or CN;

X4is N or C-R6Wherein R is6Is F;

each RIndependently selected from the group consisting of: br, F, CF2H、CF3And CN;

n is 0, 1 or 2; and is

R7Is H;

and pharmaceutically acceptable salts, N-oxides, or solvates of the compounds having formula (IA).

Embodiments of the present disclosure are compounds having formula (I) having formula (IB):

wherein

Het is

R1Is H or CH3

R2Is H or CH3

R3Is H;

R4is H or CH3

X3Is C-R6Wherein R is6Is H, Br, F, CF2H (or CHF)2)、CF3Or CN;

X4is N or C-R6Wherein R is6Is F;

each R5Independently selected from the group consisting of: br, F, CF2H、CF3And CN;

n is 0, 1 or 2; and is

R7Is H;

and pharmaceutically acceptable salts, N-oxides, or solvates of the compounds having formula (IB).

In embodiments, the compound having formula (I) is a compound wherein X is1Is N; x2Is O; r1And R2Each is H; r4Is CH3(ii) a Het isX3And X4Each is C-R6And each R6Is H; and R is5Selected from halo, C1-4Haloalkyl and CN.

In embodiments, the compound having formula (I) is a compound wherein X is1Is O; x2Is N; r1And R2Each is H; r4Is CH3(ii) a Het isX3And X4Each is C-R6And each R6Is H; and R is5Selected from halo, C1-4Haloalkyl and CN.

In embodiments, the compound having formula (I) is a compound wherein X is1Is N; x2Is O; r1And R2Each is CH3;R4Is H; het isX3And X4Each is C-R6And each R6Is H; and R is5Selected from halo, C1-4Haloalkyl and CN.

In embodiments, the compound having formula (I) is a compound wherein X is1Is O; x2Is N; r1And R2Each is CH3;R4Is H; het isX3And X4Each is C-R6And each R is6Is H; and R is5Selected from halo, C1-4Haloalkyl and CN.

In embodiments, the compound having formula (I) is a compound wherein X is1Is N; x2Is O; r1And R2Each is H; r4Is CH3(ii) a Het isX3And X4Each is C-R6And each R6Is H; and at least one R5Is F.

In embodiments, the compound having formula (I) is a compound wherein X is1Is O; x2Is N; r1And R2Each is CH3;R4Is H; het isX3And X4Each is C-R6And each R is6Is H; and at least one RIs F.

In embodiments, the compound having formula (I) is a compound wherein X is1Is N; x2Is O; r1And R2Each is H; r4Is CH3(ii) a Het isX4Is N; and R isSelected from halo, C1-4Haloalkyl and CN.

In embodiments, the compound having formula (I) is a compound wherein X is1Is O; x2Is N; r1And R2Each is H; r4Is CH3(ii) a Het isX4Is N; and R is5Selected from halo, C1-4Haloalkyl and CN.

In embodiments, the compound having formula (I) is a compound wherein X is1Is N; x2Is O; r1And R2Each is H; r4Is CH3(ii) a Het isX4Is C-R6Wherein R is6Is F; and R is5Selected from halo, C1-4Haloalkyl and CN.

In embodiments, the compound having formula (I) is a compound wherein X is1Is O; x2Is N; r1And R2Each is CH3;R4Is H; het isX4Is C-R6Wherein R is6Is F; and R is5Selected from halo, C1-4Haloalkyl and CN.

In embodiments, the compound having formula (I) is a compound wherein X is1Is N; x2Is O; r1And R2Each is CH3;R4Is H; het isX4Is N; and R isSelected from halo, C1-4Haloalkyl and CN.

In embodiments, the compound having formula (I) is a compound wherein X is1Is O; x2Is N; r1And R2Each is H; r4Is CH3(ii) a Het isX4Is N; and R is5Selected from the group consisting of halo、C1-4Haloalkyl and CN.

In embodiments, the compound having formula (I) is a compound wherein X is1Is N; x2Is O; r1And R2Each is CH3;R4Is H; het isX4Is C-R6Wherein R is6Is F; and R is5Selected from halo, C1-4Haloalkyl and CN.

In embodiments, the compound having formula (I) is a compound wherein X is1Is O; x2Is N; r1And R2Each is CH3;R4Is H; het isX4Is C-R6Wherein R is6Is F; and R is5Selected from halo, C1-4Haloalkyl and CN.

In one embodiment, the compound having formula (I) is a compound wherein

X1Is N; x2Is O; and Het isWherein R is7Is H.

In one embodiment, the compound having formula (I) is a compound wherein

X1Is N; x2Is O; het isWherein R is7Is H; r1、R2And R3Each is H; and R is4Is CH3

In one embodiment, the compound having formula (I) is a compound wherein X is1Is N; x2Is O; het isWherein R is7Is H; r1、R2And R3Each is H; r4Is CH3;X3Is C-R6Wherein R is6Selected from the group consisting of: H. CF (compact flash)3And CN; x4Is C-R6Wherein R is6Is F; n is 0 or 1; and R is5Is CF3Or CN.

In one embodiment, the compound having formula (I) is a compound wherein X is1Is N; x2Is O; het isWherein R is7Is H; r1、R2And R3Each is H; r4Is CH3;X3Is C-R6Wherein R is6Selected from the group consisting of: H. CF (compact flash)2H and Br; x4Is N; n is 1 or 2; and each R5Independently selected from the group consisting of: F. CF (compact flash)2H and Br.

In one embodiment, the compound having formula (I) is a compound wherein X is1Is O; x2Is N; and is

Het isWherein R is7Is H.

In one embodiment, the compound having formula (I) is a compound wherein X is1Is O; x2Is N;

het isWherein R is7Is H; r1、R2And R3Each is H; and R is4Is CH3

In one embodiment, the compound having formula (I) is a compound wherein X is1Is O; x2Is N;

het isWherein R is7Is H; r1、R2And R3Each is H; r4Is CH3;X3Is C-R6Wherein R is6Selected from the group consisting of: H. CF (compact flash)3And CN; x4Is C-R6Wherein R is6Is F; n is 0 or 1; and R is5Is CF3Or CN.

In one embodiment, the compound having formula (I) is a compound wherein X is1Is O; x2Is N;

het isWherein R is7Is H; r1、R2And R3Each is H; r4Is CH3;X3Is C-R6Wherein R is6Selected from the group consisting of: H. CF (compact flash)2H and Br; x4Is N; n is 1 or 2; and each R5Independently selected from the group consisting of: F. CF (compact flash)2H and Br.

Another embodiment of the disclosure is a compound as shown in table 1 below.

Table 1.

And pharmaceutically acceptable salts, N-oxides, or solvates thereof.

Pharmaceutical composition

Also disclosed herein are pharmaceutical compositions comprising

(A) At least one compound having the formula (I):

wherein

R1、R2、R3And R4Each independently is H or C1-4An alkyl group;

het is a 5 or 6 membered heteroaryl ring selected from the group consisting of: thiazoles, pyrimidines and pyrazoles, wherein each thiazole, pyrimidine and pyrazole is optionally substituted with at least one R7Substitution;

X1and X2Each independently is O or N;

wherein when X is1When is O, X2Is N; wherein when X is1When is N, X2Is O;

X3and X4Each independently is C-R6Or N;

wherein when X is3Is C-R6Or N is, X4Is C-R6(ii) a Wherein when X is4Is C-R6Or N is, X3Is C-R6

R5And R6Each independently selected from the group consisting of: H. halo, C1-4Haloalkyl and CN;

n is 0, 1 or 2; and is

R7Selected from the group consisting of: halo, C1-4Alkyl, C substituted by OH1-4Alkyl, and C1-4A haloalkyl group; and

a pharmaceutically acceptable salt, stereoisomer, isotopic variation, N-oxide or solvate of a compound having formula (I); and

(B) at least one pharmaceutically acceptable excipient.

One embodiment of the present disclosure is a pharmaceutical composition comprising at least one pharmaceutically acceptable excipient and at least one of the compounds listed in table 1, as well as any pharmaceutically acceptable salt, N-oxide, or solvate of such a compound, or any pharmaceutically acceptable prodrug of such a compound, or any pharmaceutically active metabolite of such a compound.

Additional active therapeutic agents may include, for example, anti-HBV agents (such as HBV polymerase inhibitors, interferons, viral entry inhibitors, viral maturation inhibitors, capsid assembly modulators, reverse transcriptase inhibitors, immunomodulators (such as T L R-agonists), or any other agent that affects the HBV life cycle and/or the outcome of an HBV infection).

As used herein, the term "composition" or "pharmaceutical composition" refers to a mixture of at least one compound useful in the present disclosure and a pharmaceutically acceptable carrier. The pharmaceutical composition facilitates administration of the compound to a patient or subject. There are a variety of techniques in the art for administering compounds including, but not limited to, intravenous, oral, aerosol, parenteral, ocular, pulmonary, and topical administration.

As used herein, the term "pharmaceutically acceptable carrier" means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, stabilizer, dispersant, suspending agent, diluent, excipient, thickener, solvent or encapsulating material, involved in carrying or transporting or carrying or delivering a compound useful in the present disclosure in a patient such that it may perform its intended function. Typically, such constructs are carried or transported from one organ or part of the body to another organ or part of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation (including the compounds useful in the present disclosure) and not injurious to the patient. Some examples of materials that can serve 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 gum 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; glycols, 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; a surfactant; 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.

As used herein, "pharmaceutically acceptable carrier" also includes any and all coating agents, antibacterial and antifungal agents, and absorption delaying agents, and the like, that are compatible with the activity of compounds useful in the present disclosure and physiologically acceptable to a patient. Supplementary active compounds may also be incorporated into the compositions. The "pharmaceutically acceptable carrier" may further include pharmaceutically acceptable salts of the compounds useful in the present disclosure. Other additional ingredients that may be included in pharmaceutical compositions for practicing the present disclosure are known in the art and are described, for example, in Remington's pharmaceutical sciences [ Remington pharmaceutical science ] (genano editors, Mack Publishing Co. [ mark Publishing company ], 1985, easton, pa), which is incorporated herein by reference.

"pharmaceutically acceptable excipient" refers to a substance that is non-toxic, biologically tolerable, and biologically suitable for administration to a subject (e.g., an inert substance), which is added to a pharmacological composition, or which serves as a vehicle, carrier, or diluent to facilitate and is compatible with administration of a pharmaceutical agent. Examples of excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.

Delivery forms of pharmaceutical compositions containing one or more active agent dosage units can be prepared using suitable pharmaceutical excipients and mixing techniques known or available to those skilled in the art. In the methods of the invention, the compositions may be administered by a suitable delivery route, for example, orally, parenterally, rectally, topically or by ocular route or by inhalation.

The formulations may be in the form of tablets, capsules, sachets, dragees, powders, granules, lozenges, reconstitutable powders, liquid preparations, or suppositories. Preferably, the composition is formulated for intravenous infusion, topical administration, or oral administration.

For oral administration, the compounds of the present disclosure may be provided in the form of tablets or capsules, or in the form of solutions, emulsions, or suspensions. To prepare oral compositions, the compounds can be formulated to produce, for example, a dose of from about 0.05 to about 100 mg/kg/day, or from about 0.05 to about 35 mg/kg/day, or from about 0.1 to about 10 mg/kg/day. For example, a total daily dose of about 5mg to 5g per day may be achieved by once, twice, three or four times daily administration.

Oral tablets may comprise a compound according to the present disclosure admixed with pharmaceutically acceptable excipients such as inert diluents, disintegrants, binders, lubricants, sweeteners, flavoring agents, coloring agents and preservatives. Suitable inert fillers include sodium and calcium carbonate, sodium and calcium phosphate, lactose, starch, sucrose, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol, and the like. Examples of liquid oral vehicles include ethanol, glycerol, water, and the like. Starch, polyvinylpyrrolidone (PVP), sodium starch glycolate, microcrystalline cellulose and alginic acid are suitable disintegrating agents. The binder may include starch and gelatin. The lubricant (if present) may be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate to delay absorption in the gastrointestinal tract, or may be coated with an enteric coating.

Capsules for oral administration include hard and soft gelatin capsules. To prepare hard gelatin capsules, the compounds of the present disclosure may be mixed with a solid, semi-solid, or liquid diluent. Soft capsules can be prepared by mixing a compound of the present disclosure with water, oil (e.g., peanut oil, olive oil), liquid paraffin, a mixture of short chain fatty acid monoglycerides and short chain fatty acid diglycerides, polyethylene glycol 400, or propylene glycol.

Liquids for oral administration may be in the form of suspensions, solutions, emulsions or syrups, or may be presented as a dry product, lyophilized or otherwise reconstituted with water or other suitable vehicle prior to use. Such liquid compositions may optionally contain: pharmaceutically acceptable excipients such as suspending agents (e.g., sorbitol, methyl cellulose, sodium alginate, gelatin, hydroxyethyl cellulose, carboxymethyl cellulose, aluminum stearate gelatin, and the like); non-aqueous vehicles such as oils (e.g., almond oil or fractionated coconut oil), propylene glycol, ethanol, or water; preservatives (e.g., methyl or propyl paraben, or sorbic acid); wetting agents, such as lecithin; and flavoring and coloring agents (if desired).

The active agents of the present disclosure may also be administered by non-oral routes. For example, the compositions may be formulated as suppositories for rectal administration. For parenteral administration, including intravenous, intramuscular, intraperitoneal, or subcutaneous routes, the compounds of the disclosure can be provided in sterile aqueous solutions or suspensions buffered to appropriate pH and isotonicity or in parenterally acceptable oils. Suitable aqueous vehicles include ringer's solution and isotonic sodium chloride. Such forms will be presented in unit dosage form (e.g., ampoules or single use injection devices), in multi-dose form (e.g., vials from which appropriate doses may be withdrawn), or in solid form or preconcentrate useful in the preparation of injectable formulations. Exemplary infusion doses mixed with a pharmaceutically acceptable carrier over a period of time ranging from several minutes to several days can range from about 1 to 1000 μ g/kg/minute of the compound.

For topical administration, the compound may be combined with a pharmaceutically acceptable carrier at a concentration of about 0.1% to about 10% (drug to carrier). Another mode of administration of the compounds of the present disclosure may utilize patch formulations to affect transdermal delivery.

Alternatively, in the methods of the present disclosure, the compounds of the present disclosure may be administered by inhalation via the nasal or oral route (e.g., in spray formulations further containing a suitable carrier).

Application method

The disclosed compounds are useful for treating and preventing HBV infection in a subject (e.g., a human subject).

In non-limiting aspects, these compounds can (i) modulate or disrupt HBV assembly and other HBV core protein functions necessary for HBV replication or infectious particle production, (ii) inhibit production or infection of infectious viral particles, or (iii) interact with HBV capsid to affect defective viral particles with reduced infectivity or replication capacity as a modulator of capsid assembly. In particular, and without being bound by any particular mechanism of action, it is believed that the disclosed compounds are useful in HBV therapy by disrupting, accelerating, reducing, delaying and/or inhibiting the assembly of normal viral capsids and/or disassembly of immature or mature particles thereby inducing abnormal capsid morphology leading to antiviral effects (such as disrupting virion assembly and/or disassembly, virion maturation, viral egress, and/or infection of target cells). The disclosed compounds can interact with mature or immature viral capsids as an interfering agent of capsid assembly to perturb capsid stability, thereby affecting their assembly and/or disassembly. The disclosed compounds can perturb the protein folding and/or salt bridges required for stability, function, and/or normal morphology of the viral capsid, thereby disrupting and/or accelerating the assembly and/or disassembly of the capsid. The disclosed compounds can bind to the capsid and alter the metabolism of cellular polyproteins and precursors, leading to abnormal accumulation of protein monomers and/or oligomers and/or abnormal particles, which causes cytotoxicity and death of infected cells. The disclosed compounds can cause failure of optimal stable capsid formation, affecting effective uncoating and/or disassembly of the virus (e.g., during infection). When the capsid protein is immature, the disclosed compounds can disrupt and/or accelerate capsid assembly and/or disassembly. The disclosed compounds can disrupt and/or accelerate capsid assembly and/or disassembly as the capsid protein matures. The disclosed compounds can disrupt and/or accelerate capsid assembly and/or disassembly during viral infection, which can further attenuate HBV viral infectivity and/or reduce viral load. Disruption, acceleration, inhibition, delay, and/or reduction of capsid assembly and/or disassembly by the disclosed compounds can eradicate the virus from the host organism. Eradication of HBV from a subject by the disclosed compounds advantageously eliminates the need for chronic long-term therapy and/or reduces the duration of long-term therapy.

The present disclosure relates to compounds having formula (I) for use in the treatment of HBV infection.

The present disclosure relates to compounds having formula (I) for use as medicaments for the treatment of HBV infection.

The present disclosure relates to the use of compounds having formula (I) for the treatment of HBV infection.

Another embodiment of the present disclosure is a method of treating a subject having an HBV infection, comprising administering to a subject in need of such treatment an effective amount of at least one compound having formula (I).

The present disclosure relates to compounds having formula (I) for use in reducing the viral load associated with HBV infection.

The present disclosure relates to compounds having formula (I) for use as medicaments for reducing the viral load associated with HBV infection.

The present disclosure relates to the use of compounds having formula (I) for reducing the viral load associated with HBV infection.

In another aspect, provided herein is a method of reducing the viral load associated with HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.

The present disclosure relates to compounds having formula (I) for use in reducing the recurrence of HBV infection.

The present disclosure relates to compounds having formula (I) for use as medicaments for reducing the recurrence of HBV infection.

The present disclosure relates to the use of compounds having formula (I) for reducing the recurrence of HBV infection.

In another aspect, provided herein is a method of reducing the recurrence of HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.

The present disclosure relates to compounds having formula (I) for use in inhibiting or reducing the formation or presence of HBV dna na-containing particles or HBV RNA-containing particles.

The present disclosure relates to compounds having formula (I) for use as medicaments for inhibiting or reducing the formation or presence of HBV dna na-containing particles or HBV RNA-containing particles.

The present disclosure relates to the use of a compound having formula (I) for inhibiting or reducing the formation or presence of hbv dna a-containing particles or hbv rna-containing particles.

In another aspect, provided herein is a method of inhibiting or reducing the formation or presence of hbv dna a-containing particles or hbv rna-containing particles in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound having formula (I), or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method of reducing the adverse physiological effects of HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound having formula (I) or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method of inducing remission of HBV infected liver injury in an individual in need thereof, the method comprising administering to the individual a therapeutically effective amount of a compound having formula (I), or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method of reducing the physiological effects of chronic antiviral therapy of HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method of prophylactically treating an HBV infection in an individual in need thereof, wherein the individual has a latent HBV infection, comprising administering to the individual a therapeutically effective amount of a compound having formula (I), or a pharmaceutically acceptable salt thereof.

In embodiments, the disclosed compounds are suitable for monotherapy. In the examples, the disclosed compounds are effective against natural or natural HBV strains. In the examples, the disclosed compounds are effective against HBV strains resistant to currently known drugs.

In another embodiment, the compounds provided herein may be used in methods of modulating (e.g., inhibiting or disrupting) the activity, stability, function, and viral replication properties of HBVcccDNA.

In yet another embodiment, the compounds of the present disclosure can be used in a method of attenuating or preventing the formation of HBV cccDNA.

In another embodiment, the compounds provided herein may be used in a method of modulating (e.g., inhibiting or disrupting) HBVcccDNA activity.

In yet another embodiment, the compounds of the present disclosure can be used in a method of attenuating the formation of HBV cccDNA.

In another embodiment, the disclosed compounds may be used in methods of modulating, inhibiting, or disrupting the production or release of HBV RNA particles from an infected cell.

In another embodiment, the total burden (or concentration) of HBV RNA particles is modulated. In a preferred embodiment, the overall burden of HBV RNA is attenuated.

In another embodiment, the methods provided herein reduce the viral load in said individual to a greater extent or at a faster rate than administration of a compound selected from the group consisting of: HBV polymerase inhibitors, interferons, viral entry inhibitors, viral maturation inhibitors, different capsid assembly modulators, antiviral compounds of different or unknown mechanisms, and any combination thereof.

In another embodiment, the methods provided herein result in a lower incidence of viral mutation and/or viral resistance as compared to administration of a compound selected from the group consisting of: HBV polymerase inhibitors, interferons, viral entry inhibitors, viral maturation inhibitors, different capsid assembly modulators, antiviral compounds of different or unknown mechanisms, and combinations thereof.

In another embodiment, the methods provided herein further comprise administering to the individual at least one HBV vaccine, nucleoside HBV inhibitor, interferon, or any combination thereof.

In one aspect, provided herein is a method of treating an HBV infection in an individual in need thereof, the method comprising reducing the HBV viral load by: administering to the subject a therapeutically effective amount of a compound of formula (I) (and of formula (IA) or formula (IB)), or a pharmaceutically acceptable salt thereof, alone or in combination with a reverse transcriptase inhibitor; and further administering to the individual a therapeutically effective amount of an HBV vaccine.

Another embodiment of the present disclosure is a method of treating a subject having an HBV infection, comprising administering to a subject in need of such treatment an effective amount of at least one compound having formula (I).

In another aspect, provided herein is a method of reducing the viral load associated with HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method of reducing the recurrence of HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method of inhibiting or reducing the formation or presence of HBV DNA-containing particles or HBV rna-containing particles in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound having formula (I), or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method of reducing the adverse physiological effects of HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound having formula (I) or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method of inducing remission of HBV infected liver injury in an individual in need thereof, the method comprising administering to the individual a therapeutically effective amount of a compound having formula (I), or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method of reducing the physiological effects of chronic antiviral therapy of HBV infection in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method of prophylactically treating an HBV infection in an individual in need thereof, wherein the individual has a latent HBV infection, comprising administering to the individual a therapeutically effective amount of a compound having formula (I), or a pharmaceutically acceptable salt thereof.

In embodiments, the methods provided herein further comprise monitoring the HBV viral load of the subject, wherein the method is performed for a period of time such that the HBV virus is undetectable.

Combination of

Provided herein are combinations of one or more of the disclosed compounds with at least one additional therapeutic agent. In embodiments, the methods provided herein may further comprise administering to the individual at least one additional therapeutic agent. In embodiments, the disclosed compounds are suitable for use in combination therapy. The compounds of the present disclosure may be used in combination with one or more additional compounds for the treatment of HBV infection, or HBV-related or induced diseases, or liver diseases. These additional compounds may comprise a compound of the present disclosure or a compound known to be useful for treating, preventing, or reducing the symptoms or effects of HBV infection, or symptoms or effects of HBV-related or HBV-induced diseases, or symptoms or effects of liver disease.

In one aspect, there is provided a product comprising a first compound and a second compound as a combined preparation for simultaneous, separate or sequential use in the prevention or treatment of HBV infection or HBV-induced disease in a mammal in need thereof, wherein the first compound is different from the second compound, wherein the first compound is a compound or a pharmaceutically acceptable salt of the present application or a pharmaceutical composition of the present application, and wherein the second compound is another HBV inhibitor.

In exemplary embodiments, the additional active ingredients are those ingredients known or found to be effective in treating a condition or disorder involving HBV infection, such as another HBV capsid assembly modulator or an active compound directed against another target associated with a particular disease or disorder involving HBV infection, or HBV infection itself. The combinations can be used to enhance therapeutic efficacy (e.g., by including compounds in the combination that enhance the efficacy or effectiveness of the active agents according to the present disclosure), reduce one or more side effects, or reduce the required dosage of the active agents according to the present disclosure. In another embodiment, the methods provided herein allow for administration of at least one additional therapeutic agent at a lower dose or frequency than the sole administration of the at least one additional therapeutic agent required to achieve a similar result in prophylactically treating an HBV infection in an individual in need thereof.

The HBV inhibitor may be selected from the group consisting of HBV codrug, HBV DNA polymerase inhibitors, reverse transcriptase inhibitors, immunomodulators (such as toll-like (T L R) receptor modulators), interferons (such as polyethylene glycol interferons) and interferon α receptor ligands, hyaluronidase inhibitors, hepatitis b surface antigen (HbsAg) inhibitors, cytotoxic T lymphocyte-associated protein 4(ipi4) inhibitors, cyclophilin inhibitors, TNF inhibitors, HBV viral entry inhibitors, antisense oligonucleotides targeted to viral mRNA, short interfering rna (sirna) and ddRNAi endonuclease modulators, ribonucleotide reductase inhibitors, HBV E antigen inhibitors, covalently closed circular DNA (cccdna) inhibitors, farnesoid X receptor agonists, HBV antibodies, chemokine antagonists, CCR2 agonists, cytokines, nuclear protein modulators, retinoic acid inducible gene stimulators, NOD2 stimulators, phosphatidylkinase 3-kinase (P13) inhibitors, HBV amine 2 inhibitors, thymosin kinase 3-amine 2 inhibitors, HBV kinase inhibitors, and other inhibitors of the effects on replication cycle of HBV infection, or other drugs affecting the outcome of HBV pathway (BTK-cycle) infection.

Additional active ingredients may include inhibitors of HBV reverse transcriptase and inhibitors of DNA and RNA polymerases, including but not limited to lamivudine (3TC, Zeffix, Heptovir, Epivir and Epivir-HBV), entecavir (Baraclude, Entavir), adefovir dipivoxil (Hepsara, Preveon, bis-POM PMEA), tenofovir fumarate (Viread, TDF or PMPA), interferons, including but not limited to interferon α (IFN- α), interferon β (IFN- β), interferon lambda (IFN-lambda), and interferon gamma (IFN-gamma), viral entry inhibitors, viral maturation inhibitors, capsid assembly modulators described in the literature, such as but not limited to BAY41-4109, reverse transcriptase inhibitors, immunomodulators, such as T L R-agonists, and different or unknown mechanisms, such as but not limited to AT-61-E-N- (1-chloro-3-oxo-phenyl-1-phenyl-3-phenyl-propane-1- (1-3-phenyl) -1-phenyl-3-phenyl-propane-1-3-phenyl-propane-1-phenyl-3-phenyl-propane-2-phenyl-propane-1-4-phenyl-propane-amide, and analogs.

In embodiments, the additional therapeutic agent is an interferon, the term "interferon" or "IFN" refers to any member of a family of highly homologous species-specific proteins that inhibit viral replication and cellular proliferation and modulate immune responses human interferons are divided into three classes, type I, which includes interferon- α (IFN- α), interferon- β (IFN- β), and interferon-omega (IFN-omega), type II, which includes interferon-gamma (IFN-gamma), and type III, which includes interferon-lambda (IFN-lambda). the term "interferon" as used herein includes recombinant forms of interferons that have been developed and are commercially available.

Thus, in one embodiment, a compound having formula I, II, III or IV may be administered in combination with an interferon selected from the group consisting of interferon α (IFN- α), interferon β (IFN- β), interferon lambda (IFN-lambda), and interferon gamma (IFN-gamma). in one embodiment, the interferon is interferon- α 0-2a, interferon- α -2b, or interferon- α -n1.. in another embodiment, either interferon- α -2a or interferon- α -2b is pegylated.in a preferred embodiment, interferon- α -2a is pegylated interferon- α -2a (PEGASYS).

In another embodiment, the additional therapeutic agent is selected from an immunomodulatory or immunostimulatory therapy comprising a biological agent belonging to the interferon class.

Furthermore, the additional therapeutic agent may be an agent that disrupts the function of one or more other essential viral proteins or host proteins required for HBV replication or persistence.

In another embodiment, the additional therapeutic agent is an antiviral agent that blocks viral entry or maturation or targets HBV polymerase, such as a nucleoside or nucleotide or non-nucleoside (nucleotide) polymerase inhibitor. In another embodiment of the combination therapy, the reverse transcriptase inhibitor and/or the DNA and/or RNA polymerase inhibitor is zidovudine, didanosine, zalcitabine, ddA, stavudine, lamivudine, abacavir, emtricitabine, entecavir, aliscitabine, ativelapine (Atevirapine), ribavirin, acyclovir, famciclovir, valacyclovir, valganciclovir, tenofovir, adefovir, PMPA, cidofovir, efavirenz, nevirapine, delavirdine, or etravirine.

In other words, the immunomodulator may affect maturation of antigen presenting cells, proliferation of T cells, and cytokine release (e.g., I L-12, I L-18, IFN- α, IFN- β, and IFN-. gamma., TNF- α, and the like).

In another embodiment, the additional therapeutic agent is a T L R modulator or a T L R agonist, such as a T L R-7 agonist or a T L R-9 agonist in an additional embodiment of the combination therapy, the T L R-7 agonist is selected from the group consisting of SM 320 (9-benzyl-8-hydroxy-2- (2-methoxy-ethoxy) adenine) and AZD 8848([ methyl 3- ({ [3- (6-amino-2-butoxy-8-oxo-7, 8-dihydro-9H-purin-9-yl) propyl ] [3- (4-morpholinyl) propyl ] amino } methyl) phenyl ] acetate).

In one embodiment, the HBV vaccine is at least one of RECOMBIVAX HB, ENGERIX-B, E L OVAC B, GENEVAC-B, or SHANTAC B.

In another aspect, provided herein is a method of treating an HBV infection in an individual in need thereof, the method comprising reducing the HBV viral load by: administering to the individual a therapeutically effective amount of a compound of the disclosure, alone or in combination with a reverse transcriptase inhibitor; and further administering to the individual a therapeutically effective amount of an HBV vaccine. The reverse transcriptase inhibitor may be one of zidovudine, didanosine, zalcitabine, ddA, stavudine, lamivudine, abacavir, emtricitabine, entecavir, aliscitabine, altiveline, ribavirin, acyclovir, famciclovir, ganciclovir, valganciclovir, tenofovir, adefovir, PMPA, cidofovir, efavirenz, nevirapine, delavirdine, or etravirine.

Examples of anti-HBV agents suitable for use with the anti-HBV agents of the present application include, but are not limited to, small molecules, antibodies, and/or CAR-T therapies that bind HBV env (S-CAR cells), capsid assembly modulators, T L R agonists (e.g., T L R7 and/or T L R8 agonists), cccDNA inhibitors, HBV polymerase inhibitors (e.g., entecavir and tenofovir), and/or immune checkpoint inhibitors, etc. the at least one anti-HBV agent may be selected, for example, from HBV DNA polymerase inhibitors, immunomodulators, Toll-like receptor 7 modulators, Toll-like receptor 8 modulators, Toll-like receptor 3 modulators, interferon α receptor ligands, hyaluronidase inhibitors, modulators of I L-10, HBsAg inhibitors, Toll-like receptor 9 modulators, cyclophilin inhibitors, HBV receptor 8 modulators, therapeutic antisense vaccines, HBV antisense nucleotide entry inhibitors, oligonucleotide 58hyaluronidase inhibitors, particularly anti-HBV DNA receptor siRNA inhibitors such as CD 961, CD L inhibitors, siRNA inhibitors, CD 6326 inhibitors, siRNA targeting HBV DNA polymerase receptor targeting HBV DNA polymerase sequence, siRNA receptor binding to HBV receptor 12, siRNA receptor binding to HBV epitope receptor binding, HIV receptor binding to HIV receptor binding agents, HIV receptor binding to HIV receptor binding agents, HIV binding to HIV receptor binding to HIV binding agents, epitope receptor binding agents, epitope binding to HIV binding agents, epitope binding to HIV binding agents, epitope binding agents, epitope binding to HIV binding agents, epitope binding to HIV binding agents, epitope binding to HIV binding agents, epitope.

For any combination therapy described herein, a suitable method can be used to calculate the synergistic effect, e.g., Sigmoid-EmaxEquation (Holford and Scheiner, 19981, clin. pharmacokinet. [ clinical pharmacokinetics ]]6: 429-]114: 313-326) and the median effect equation (Chou and Talalay, 1984, adv. enzyme Regul. [ progression of enzyme regulation ]]22: 27-55). Each of the above-mentioned equations can be applied to experimental data to generate a corresponding graph to help assess the effect of the drug combination. The corresponding graphs associated with the above equations are the concentration-effect curve, the isobologram curve, and the joint index curve, respectively.

Definition of

The following sets forth definitions of various terms used to describe the present disclosure. Unless otherwise limited to a specific context, these definitions apply to the terms as used throughout the specification and claims, either individually or as part of a larger group.

Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the applicable arts. Generally, the nomenclature used herein and the laboratory procedures in cell culture, molecular genetics, organic chemistry, and peptide chemistry are those well known and commonly employed in the art.

As used herein, the articles "a" and "an" refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. For example, "an element" means one element or more than one element.

As used herein, the terms "comprising", "containing" and "comprising" are used herein in their open, non-limiting sense.

The term "alkyl" refers to straight or branched chain alkyl groups having 1 to 12 carbon atoms in the chain. Examples of alkyl groups include methyl (Me, which may also be structurally represented by the symbol "/"), ethyl (Et), n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl (tBu), pentyl, isopentyl, tert-pentyl, hexyl, isohexyl, and groups considered equivalent to any of the above examples by one of ordinary skill in the art and from the perspectives provided herein. The term C as used herein1-4Alkyl refers to a straight or branched alkyl group having 1 to 4 carbon atoms in the chain. The term C as used herein1-6Alkyl refers to a straight or branched alkyl group having 1 to 6 carbon atoms in the chain.

The term "heteroaryl" refers to a monocyclic heterocycle or fused bicyclic heterocycle having 3 to 9 ring atoms per heterocycle (a ring structure having ring atoms selected from carbon atoms and up to four heteroatoms selected from nitrogen, oxygen, and sulfur). Illustrative examples of heteroaryl groups include the following entities in the form of suitable bonding moieties:

those skilled in the art will recognize that the above recited or illustrated classes of heteroaryl groups are not exhaustive and that additional classes may be selected within the scope of these defined terms.

The term "cyano" refers to the group-CN.

The term "halogen" denotes chlorine, fluorine, bromine or iodine.

The term "perhaloalkyl" or "haloalkyl" refers to a straight or branched alkyl group having 1 to 6 carbon atoms in the chain, optionally replacing hydrogen with halogen. The term "C" as used herein1-4Haloalkyl "refers to a straight or branched alkyl group having 1 to 4 carbon atoms in the chain, optionally with the substitution of hydrogen with halogen. The term "C" as used herein1-6Haloalkyl "refers to a straight or branched alkyl group having 1 to 6 carbon atoms in the chain, optionally with the substitution of hydrogen with halogen. Examples of "perhaloalkyl", "haloalkyl" groups include trifluoromethyl (CF)3) Difluoromethyl (CF)2H) Monofluoromethyl (CH)2F) Pentafluoroethyl (CF)2CF3) Tetrafluoroethyl (CHFCF)3) Monofluoroethyl (CH)2CH2F) Trifluoroethyl (CH)2CF3) Tetrafluorotrifluoromethylethyl (-CF (CF)3)2) And groups considered equivalent to any of the above examples, by one of ordinary skill in the art and from the viewpoints provided herein.

The term "substituted" means that the specified group or moiety bears one or more substituents. The term "unsubstituted" means that no substituent is present in the indicated group. The term "optionally substituted" means that the specified group is unsubstituted or substituted with one or more substituents. Where the term "substituted" is used to describe a structural system, it is intended that the substitution occur at any valency-allowed position on the system. Where it is not explicitly stated that a specified moiety or group is optionally substituted or substituted with a specified substituent, it is to be understood that such moiety or group is intended to be unsubstituted.

The terms "pair (para)", "meta" and "ortho" have meanings as understood in the art. Thus, for example, a fully substituted phenyl group has substituents at the two "ortho" (o) positions adjacent to the attachment point of the phenyl ring, the two "meta" (m) positions, and one "para" (p) position across the attachment point. To further clarify the position of the substituents on the phenyl ring, two different ortho positions are designated ortho and ortho ', and two different meta positions are designated meta and meta', as set forth below.

When referring to a substituent on a pyridyl group, the terms "para", "meta" and "ortho" refer to the position of the substituent relative to the point of attachment of the pyridyl ring. For example, the following structure is depicted as 3-pyridyl, where X1The substituents being in the ortho position, X2The substituent is in the meta position, and X3The substituent is positioned at the para position:

to provide a more concise description, some of the quantitative expressions given herein are not defined by the term "about". It is understood that each quantity given herein is meant to refer to the actual value given, whether or not the term "about" is used explicitly, and also means approximations based on such given value as can be reasonably inferred by one of ordinary skill in the art, including equivalents and approximations due to the experimental and/or measurement conditions for such given value. Whenever a yield is given in percent, such yield refers to the mass of an entity giving the yield relative to the maximum amount that the same entity can be obtained according to a particular stoichiometric condition. Concentrations given in percentages, unless otherwise indicated, refer to mass ratios.

The terms "buffered" solution or "buffer" solution are used interchangeably herein according to their standard meaning. The buffer solution is used to control the pH of the medium, and its selection, use and function are known to those of ordinary skill in the art. See, for example, the g.d. consistency editors, Van Nostrand's Encyclopedia of Chem, which describe (among others) how the concentration of buffer solutions and buffer components correlate with the pH of the bufferistry [ Van Nuo Stelan chemical encyclopedia]Page 261, 5 th edition (2005). For example, by mixing MgSO4And NaHCO3The buffer solution was obtained by adding to the solution at a ratio of 10: 1w/w to maintain the pH of the solution at about 7.5.

Any formula given herein is intended to represent a compound having the structure depicted by the structural formula, as well as certain variations or forms thereof. In particular, compounds of any of the formulae given herein may have asymmetric centers and thus exist in different enantiomeric forms. All optical isomers of the compounds of the general formula and mixtures thereof are considered to be within the scope of the formula. Thus, any formula given herein is intended to represent the racemate, one or more enantiomeric forms, one or more diastereomeric forms, one or more atropisomeric forms, and mixtures thereof. In addition, certain structures may exist as geometric isomers (i.e., cis and trans isomers), tautomers, or atropisomers.

It is also understood that compounds having the same molecular formula but differing in the nature or order of bonding of their atoms or the arrangement of their atoms in space are referred to as "isomers".

Stereoisomers that are not mirror images of each other are referred to as "diastereomers", and stereoisomers that are not mirror images of each other are referred to as "enantiomers". When a compound has an asymmetric center, for example, the asymmetric center is bonded to four different groups, and there may be a pair of enantiomers. Enantiomers can be characterized by the absolute configuration of their asymmetric centers and described by the R-and S-order rules of Cahn and Prelog, or by the way the molecules rotate the plane of polarized light, and are designated dextrorotatory or levorotatory (i.e., designated as the (+) -or (-) -isomers, respectively). The chiral compounds may exist as individual enantiomers or as mixtures thereof. Mixtures containing the same ratio of enantiomers are referred to as "racemic mixtures".

"tautomer" refers to compounds that are interchangeable forms of a particular compound structure and differ in hydrogen atom and electron displacement. Thus, the two structures can be in equilibrium by the motion of redundant electrons and atoms (usually H). For example, enols and ketones are tautomers because they are rapidly converted to each other by treatment with an acid or a base. Another example of tautomerism is the acid-and nitro-forms of phenylnitromethane, which examples are likewise formed by treatment with an acid or a base.

The tautomeric form may be associated with optimal chemical reactivity and biological activity to obtain the target compound.

The compounds of the present disclosure may have one or more asymmetric centers; thus, such compounds may be produced as the (R) -or (S) -stereoisomer alone or as a mixture thereof.

Unless otherwise indicated, the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers as well as racemic or other mixtures thereof. Methods for the determination of stereochemistry and the separation of stereoisomers are well known in the art.

Certain examples contain chemical structures depicted as absolute enantiomers, but are intended to indicate enantiomerically pure materials of unknown configuration. In these cases, the absolute stereochemistry of the corresponding stereocenter is not known, indicated in the name by (R) or (S). Thus, a compound designated (R) refers to an enantiomerically pure compound having the absolute configuration (R) or (S). In the case where absolute stereochemistry has been proven, the (R) and (S) named structures are used.

SymbolAndused to mean the same spatial arrangement in the chemical structures shown herein. Similarly, symbolsAndforMeaning the same spatial arrangement in the chemical structure shown herein.

In addition, any formula given herein is also intended to refer to hydrates, solvates, and polymorphs of such compounds, and mixtures thereof, even if such forms are not explicitly listed. Certain compounds having formula (I), or pharmaceutically acceptable salts of compounds having formula (I), may be obtained as solvates. Solvates include solvates formed by the interaction or complexation of a compound of the present disclosure with one or more solvents in solution or in solid or crystalline form. In some embodiments, the solvent is water and the solvate is a hydrate. Furthermore, certain crystalline forms of the compound of formula (I) or pharmaceutically acceptable salts of the compound of formula (I) may be obtained as co-crystals. In certain embodiments of the present disclosure, the compound having formula (I) is obtained in crystalline form. In other embodiments, the crystalline form of the compound having formula (I) is cubic in nature. In other embodiments, the pharmaceutically acceptable salt of the compound having formula (I) is obtained in crystalline form. In still other embodiments, the compound having formula (I) is obtained in one of several polymorphic forms, as a mixture of crystalline forms, as a polymorphic form, or as an amorphous form. In other embodiments, the compound having formula (I) is converted in solution between one or more crystalline forms and/or polymorphs.

Representative compounds referred to herein relate to any one of: (a) the actual listed forms of such compounds, and (b) any form of such compounds in media in which they are considered when named. For example, reference herein to a compound such as R-COOH encompasses reference to, for example, any one of: R-COOH(s)、R-COOH(sol)And R-COO- (sol). In this example, R-COOH(s)Refers to a solid compound as it may, for example, be present in a tablet or some other solid pharmaceutical composition or formulation; R-COOH(sol)Refers to the undissociated form of the compound in a solvent; and R-COO- (sol)Refers to the dissociated form of the compound in a solvent, e.g. the compound is aqueousDissociated form in the environment, whether such dissociated form is derived from R-COOH, from a salt thereof, or from R-COO produced upon dissociation in the medium in question-Any other entity of (1). In another example, expressions such as "exposing an entity to a compound having the formula R-COOH" refer to exposing the entity to one or more forms of the compound R-COOH present in a medium in which such exposure is performed. In yet another example, expressions such as "reacting an entity with a compound having the formula R-COOH" refer to reacting (a) one or more chemically-related forms of such entity present in the medium in which such reaction occurs with (b) one or more chemically-related forms of compound R-COOH present in the medium in which such reaction occurs. In this respect, if such an entity is for example in an aqueous environment, it is understood that the compound R-COOH is in such the same medium and thus the entity is exposed to e.g. R-COOH(aq)And/or R-COO- (aq)Of the classes of media, where the subscript "(aq)" represents "aqueous" according to its conventional meaning in chemistry and biochemistry. The carboxylic acid function is chosen among these named examples; however, this choice is not intended to be limiting, but rather is merely illustrative. It is understood that similar examples may be provided in terms of other functional groups, including but not limited to hydroxyl groups, basic nitrogen members (such as those in amines), and any other group that interacts or transforms in a medium containing the compound according to known means. Such interactions and transformations include, but are not limited to, dissociation, association, tautomerism, solvolysis (including hydrolysis), solvation (including hydration), protonation, and deprotonation. No further examples are provided herein in this regard, as these interactions and transformations in a given medium are known to any person of ordinary skill in the art.

In another example, zwitterionic compounds are encompassed herein by reference to compounds known to form zwitterions, even if not explicitly named in their zwitterionic form. The term one or more zwitterionic compounds, such as one or more zwitterions and synonyms thereof, is an IUPAC approved labelQuasi-names, which are well known and are part of a standard set of defined scientific names. In this regard, the name of zwitterion is designated by the molecular entity dictionary of the biologically relevant Chemical entity database (Chemical Entities of Biological Interest (ChEBI)) as the name identification ChEBI: 27369. as is generally well known, zwitterionic or zwitterionic compounds are neutral compounds having formal unit charges of opposite sign. Sometimes, these compounds are referred to by the term "inner salt". Other sources refer to these compounds as "dipolar ions," although the latter terms are considered by other sources as misnomers. As a specific example, the aminoacetic acid (i.e., the aminoglycine) is of the formula H2NCH2COOH, and in some media (in this case neutral media) as zwitterions+H3NCH2COO-Exist in the form of (1). Zwitterions, zwitterionic compounds, internal salts, and dipolar ions are within the scope of the present disclosure in the known and well-defined meaning of these terms, and in any event should be so understood by one of ordinary skill in the art. The structures of the zwitterionic compounds associated with the compounds of the present disclosure are not explicitly set forth herein, as there is no necessity to name every example which would be recognized by one of ordinary skill in the art. But is part of an embodiment of the present disclosure. In this regard, no further examples are provided herein, as the various forms of interactions and transformations that result in a given compound in a given medium are known to any one of ordinary skill in the art.

Any formula given herein is also intended to represent the unlabeled form as well as the isotopically labeled form of the compound. Isotopically-labeled compounds have the structure depicted by the formulae given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or number of atoms. Examples of isotopes that can be incorporated into compounds of the present disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, as respectively2H、3H、11C、13C、14C、15N、18O、17O、31P、32P、35S、18F、36Cl、125I. Such isotopically labeled compounds can be used in metabolic studies (preferably14C) Reaction kinetics studies (with, for example, deuterium (i.e. D or2H) (ii) a Or tritium (i.e. T or3H) Detection or imaging processes including measurement of tissue distribution of drugs or substrates, such as Positron Emission Tomography (PET) or single-photon emission computed tomography (SPECT), or in the radiation treatment of patients. In particular, it is possible to use, for example,18f or11The C-labeled compound may be particularly preferably used for studies of PET or SPECT. In addition, the heavy isotopes such as deuterium (i.e.,2H) substitution may confer certain therapeutic advantages resulting from better metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements). Isotopically labeled compounds of the present disclosure and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples below, and by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent as described below.

The selection of a particular section from a list of possible categories of a given variable, when referring to any formula given herein, is not intended to limit the categories of said variable occurring elsewhere to the same selection. In other words, when a variable occurs more than once, the choice of a category from a given list is independent of the choice of a category of the same variable elsewhere in the formula, unless otherwise specified.

In light of the above explanatory disclosure of assignments and nomenclature, it should be understood that explicit reference to a setting herein (where chemically meaningful and unless otherwise indicated) implies embodiments that independently refer to such a setting, and every possible embodiment that refers to a subset of the explicitly referred settings.

By way of a first example of terminology with respect to substituents, if substituent S1 Examples of the inventionIs S1And S2And a substituent S2 Examples of the inventionIs S3And S4Of the present disclosure, then, these assignments refer to embodiments of the present disclosure given according to the following choices: s1 Examples of the inventionIs S1And S2 Examples of the inventionIs S3;S1 Examples of the inventionIs S1And S2 Examples of the inventionIs S4;S1 Examples of the inventionIs S2And S2 Examples of the inventionIs S3;S1 Examples of the inventionIs S2And S2 Examples of the inventionIs S4(ii) a And equivalents of each of such choices. For the sake of brevity, the shorter term "S" is used herein accordingly1 Examples of the inventionIs S1And S2And S is2 Examples of the inventionIs S3And S4One "of (a), but not in a limiting manner. The first example of the above terminology with respect to substituents described in general terms is intended to illustrate the various substituent assignments described herein. The above rules for substituents given herein extend, where applicable, to as R1、R2、R3、R4、R5、R6、R7、X1、X2、X3、X4N, Het, PG, and any other generic substituent symbol used herein.

Furthermore, when more than one assignment is given to any member or substituent, embodiments of the disclosure encompass various combinations that may be made of the enumerated assignments taken independently and their equivalents. By way of a second example of nomenclature for substituents, if substituent S is described hereinExamples of the inventionIs S1、S2And S3Of the present disclosure, then this list refers to embodiments of the present disclosure, wherein SExamples of the inventionIs S1;SExamples of the inventionIs S2;SExamples of the inventionIs S3;SExamples of the inventionIs S1And S2One of (a); sExamples of the inventionIs S1And S3One of (a); sExamples of the inventionIs S2And S3One of (a); sExamples of the inventionIs S1、S2And S3One of (a); and SExamples of the inventionIs any equivalent of each of these options. For the sake of brevitySee, the shorter term "S" is used herein accordinglyExamples of the inventionIs S1、S2And S3One "of (a), but not in a limiting manner. The second example of the above terminology with respect to substituents, as described in general terms, is intended to illustrate the various substituent assignments described herein. The above rules for substituents given herein extend, where applicable, to as R1、R2、R3、R4、R5、R6、R7、X1、X2、X3、X4N, Het, PG, and any other generic substituent symbol used herein.

Name "Ci-j", wherein j > i, when applied to a class of substituents herein, means embodiments of the disclosure wherein each number of carbon atom members from i to j (including i and j) is independently achieved. For example, the term C1-4Independently means a member having one carbon atom (C)1) Examples of (A) a member having two carbon atoms (C)2) Example of (1), a member having three carbon atoms (C)3) Examples of (A) and members having four carbon atoms (C)4) Examples of (1).

Term Cn-mAlkyl refers to a straight or branched aliphatic chain having a total number N of carbon atoms in the chain, which satisfies n.ltoreq.N.ltoreq.m, with m > N. When more than one such attachment possibility is allowed, any reference herein to a di-substituent is meant to encompass a variety of attachment possibilities. For example, reference to a disubstituent-a-B- (wherein a ≠ B) refers herein to such disubstituent with a attached to a first substitution member and B attached to a second substitution member, and it also refers to such disubstituent with a attached to a second substitution member and B attached to the first substitution member.

The disclosure also includes pharmaceutically acceptable salts of compounds having formula (I) (as well as formula (IA) and formula (IB)), preferably those described above and the specific compounds exemplified herein, and methods of treatment using such salts.

The term "pharmaceutically acceptable" means approved or approvable by a regulatory agency of the federal or a state government or a corresponding agency of a country outside the united states, or listed in the U.S. pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.

"pharmaceutically acceptable salt" is intended to mean a free acid or base salt of a compound represented by formula (I) that is non-toxic, biologically tolerable, or in other forms that are biologically suitable for administration to a subject. It should have the desired pharmacological activity of the parent compound. See generally, g.s.paulekuhn et al, "Trends in Active pharmaceutical ingredient Salt Selection based on Analysis of the Orange Book Database" [ Active drug ingredient Salt screening trend based on Orange Book Database Analysis ], j.med.chem. [ journal of pharmaceutical chemistry ], 2007, 50: 6665-72, s.m. berge et al, "Pharmaceutical Salts" [ drug Salts ], J Pharm Sci. [ journal of Pharmaceutical science ], 1977, 66: 1-19, and Handbook of Pharmaceutical Salts, Properties, Selection, and uses, [ Handbook of Salts: properties, selection and use ] Stahl and Wermuth eds, Wiley-VCH and VHCA, zurich, 2002. Examples of pharmaceutically acceptable salts are those that are pharmacologically effective without undue toxicity, irritation, or allergic response and are suitable for contact with patient tissues. The compounds of formula (I) may have sufficiently acidic groups, sufficiently basic groups, or both functional groups, and thus react with various inorganic or organic bases, and inorganic and organic acids, to form pharmaceutically acceptable salts.

Examples of pharmaceutically acceptable salts include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, propionate, decanoate, octanoate, acrylate, formate, isobutyrate, hexanoate, heptanoate, propiolate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, butyne-1, 4-dioate, hexyne-1, 6-dioate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, phthalate, sulfonate, xylenesulfonate, phenylacetate, phenylpropionate, phenylbutyrate, dihydrogenphosphate, pyrophosphate, chloride, bromide, iodide, propionate, and butyrate, Citrate, lactate, gamma-hydroxybutyrate, glycolate, tartrate, methanesulfonate, propanesulfonate, naphthalene-1-sulfonate, naphthalene-2-sulfonate, and mandelate.

For example, the free base may be treated with an inorganic acid such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, nitric acid, boric acid, phosphoric acid, and the like, or with an organic acid such as acetic acid, phenylacetic acid, propionic acid, stearic acid, lactic acid, ascorbic acid, maleic acid, hydroxymaleic acid, isethionic acid, succinic acid, valeric acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, oleic acid, palmitic acid, lauric acid, pyranosidyl acid (such as glucuronic acid or galacturonic acid), α -hydroxy acid (such as mandelic acid, citric acid or tartaric acid), an amino acid (such as aspartic acid, glutaric acid or glutamic acid), an aromatic acid (such as benzoic acid, 2-acetoxybenzoic acid, naphthoic acid or cinnamic acid), a sulfonic acid (such as lauryl sulfonic acid, p-toluenesulfonic acid, methanesulfonic acid, ethanesulfonic acid), an acid (such as exemplified herein), and any other mixtures thereof, and equivalents thereof, which are considered to be acceptable by a person of ordinary skill in the art.

When the compound of formula (I) is an acid, such as a carboxylic or sulfonic acid, the desired pharmaceutically acceptable salt may be prepared by any suitable method, for example by treating the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide, an alkaline earth metal hydroxide, any compatible mixture of bases (such as those given herein as examples), and any other base and mixtures thereof which are considered equivalents or acceptable substitutes from the standpoint of one of ordinary skill in the art. Illustrative examples of suitable salts include organic salts derived from amino acids (e.g., N-methyl-D-glucamine, lysine, choline, glycine, and arginine), ammonia, carbonates, bicarbonates, primary, secondary, and tertiary amines, and cyclic amines (e.g., tromethamine, benzylamine, pyrrolidine, piperidine, morpholine, and piperazine), and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.

The disclosure also relates to pharmaceutically acceptable prodrugs of compounds having formula (I), and methods of treatment employing such pharmaceutically acceptable prodrugs. The term "prodrug" means a precursor of a specified compound that, upon administration to a subject, produces the compound in vivo via a chemical or physiological process (e.g., solvolysis or enzymatic cleavage) or under physiological conditions (e.g., a prodrug at near physiological pH is converted to a compound of formula (I)). A "pharmaceutically acceptable prodrug" is a prodrug that is non-toxic, biologically tolerable, or in other forms that are biologically suitable for administration to a subject. Exemplary procedures for selecting and preparing suitable prodrug derivatives are described, for example, in "Design of Prodrugs Design ]" h.bundgaard editions, eisweier (Elesevier), 1985.

Exemplary prodrugs include compounds having an amino acid residue or a polypeptide chain of two or more (e.g., two, three, or four) amino acid residues covalently linked through an amide or ester bond to a free amino, hydroxyl, or carboxylic acid group of a compound having formula (I) examples of amino acid residues include the twenty naturally occurring amino acids commonly designated by three letter symbols, as well as 4-hydroxyproline, hydroxylysine, desmosine (demosine), isodesmosine (isodesmosine), 3-methylhistidine, norvaline (norvlin), β -alanine, γ -aminobutyric acid, citrulline homocysteine, homoserine, ornithine, and methionine sulfone.

Additional types of prodrugs can be generated, for example, by derivatizing a free carboxyl group having the structure of formula (I) as an amide or alkyl ester. Examples of amides include those derived from ammonia, C1-6Alkyl primary amines and di (C)1-6Alkyl) secondary amines. Secondary amines include 5-or 6-membered heterocycloalkyl or heteroaryl ring moieties. Examples of amides include those derived from ammonia, C1-3Primary alkyl amines and di (A), (B), (CC1-2Alkyl) amines. Examples of esters of the present disclosure include C1-7Alkyl radical, C5-7Cycloalkyl, phenyl and phenyl (C)1-6Alkyl) esters. Preferred esters include methyl esters. Prodrugs can also be prepared by following the general principles of Fleisher et al, adv. drug Delivery Rev. [ review of advanced drug Delivery]1996, 19, 115-130 using groups including hemisuccinate, phosphate, dimethylaminoacetate and phosphoryloxymethyloxycarbonyl to derivatize the free hydroxyl groups. Carbamate derivatives of hydroxy and amino groups may also produce prodrugs. Carbonate derivatives, sulfonates and sulfates of hydroxyl groups may also provide prodrugs. Derivatization of hydroxyl groups as (acyloxy) methyl and (acyloxy) ethyl ethers, where the acyl group may be an alkyl ester, optionally substituted with one or more ether, amine or carboxylic acid functionalities, or where the acyl group is an amino acid ester as described above, may also be used to produce prodrugs. This type of prodrug can be, for example, Robinson et al, J Med Chem [ J. PharmacoChem]1996, 39(1), 10-18. The free amine may also be derivatized as an amide, sulfonamide, or phosphoramide. All of these prodrug moieties may incorporate groups including ether, amine, and carboxylic acid functional groups.

The disclosure also relates to pharmaceutically active metabolites of compounds having formula (I), which may also be used in the methods of the disclosure, "pharmaceutically active metabolites" means pharmacologically active products of in vivo metabolism of compounds having formula (I) or salts thereof. Prodrugs and active metabolites of compounds may be determined using conventional techniques known or available in the art. see, e.g., Bertolini et al, JMed Chem. [ J. Pharmacochemical ]1997, 40, 2011-2016; Shan et al, pharm Sci. [ J. Pharmacoc. 1997, 86(7), 765 767; Bagshawei, Drug Dev. Res. [ Drug development research ]1995, 34, 220-230; Bodor, Adv Drug Res. [ Drug research evolution ]1984, 13, 224-331; Bundggard, Design of Prodrugs [ Design of prodrug of drugs [ Elsedearner, Design of prodrug ], and Application of Drug discovery publication 3576, Kressar publication 3576, and Application of drugs [ Design of drugs ]1984, 13, 224-Press, Design of drugs [ Design of drugs ] 3576, and Application of Hardrags et al (published edison & edition, publication L, Application of drugs).

The term "modulator" includes both inhibitors and activators, where "inhibitor" refers to a compound that reduces, prevents, inactivates, desensitizes, or down regulates HBV assembly and other HBV core protein functions necessary for HBV replication or infectious particle production.

As used herein, the term "capsid assembly modulator" refers to a compound that disrupts or accelerates or inhibits or hinders or retards or reduces or modifies normal capsid assembly (e.g., during maturation) or normal capsid disassembly (e.g., during infection) or perturbs capsid stability thereby inducing aberrant capsid morphology and function. In one embodiment, the capsid assembly modulator accelerates capsid assembly or disassembly, thereby inducing aberrant capsid morphology. In another embodiment, the capsid assembly modulator interacts with (e.g., binds to at an active site, binds to at an allosteric site, modifies and/or hinders folding, etc.) a major capsid assembly protein (CA), thereby disrupting capsid assembly or disassembly. In yet another embodiment, the capsid assembly modulator causes perturbation of the structure or function of the CA (e.g., the ability of the CA to assemble, disassemble, bind to a substrate, fold into a proper conformation, etc.), which reduces viral infectivity and/or is lethal to the virus.

As used herein, the term "treatment" is defined as the application or administration of a therapeutic agent, i.e., a compound of the present disclosure (alone or in combination with another agent), to a patient suffering from, having symptoms of, or having the potential to suffer from an HBV infection, with the goal of curing, healing, alleviating, ameliorating, altering, remediating, ameliorating, improving, or affecting the HBV infection, symptoms of HBV infection, or the potential to suffer from an HBV infection, or the application or administration of a therapeutic agent to an isolated tissue or cell line from a patient (e.g., for diagnostic or ex vivo applications). Such treatments can be specifically tailored or modified based on knowledge gained from the pharmacogenomics field.

As used herein, the term "prevention" means no disorder or disease development (if no disorder or disease occurs), or no further disorder or disease development (if the disorder or disease has already occurred). The ability to prevent some or all of the symptoms associated with a disorder or disease is also contemplated.

As used herein, the term "patient", "individual" or "subject" refers to a human or non-human mammal. Non-human mammals include, for example, farm animals as well as companion animals such as ovine, bovine, porcine, canine, feline, and murine mammals. Preferably, the patient, subject or individual is a human.

In a method of treatment according to the disclosure, an effective amount of an agent according to the disclosure is administered to a subject suffering from or diagnosed with such a disease, disorder, or condition. By "effective amount" is meant an amount or dose that is generally sufficient to elicit the desired therapeutic or prophylactic benefit for a given disease, disorder or condition in a patient in need of such treatment. An effective amount or dose of a compound of the present disclosure can be determined by conventional methods, such as modeling, dose escalation studies, or clinical trials, and taking into account conventional factors, such as the mode or route of administration or drug delivery, the pharmacokinetics of the compound, the severity and course of the disease, disorder, or condition, previous or ongoing treatment of the subject, the health and response of the subject to the drug, and the judgment of the treating physician. Examples of dosages are in the range of from about 0.001 to about 200mg of compound per kg of subject body weight per day, preferably about 0.05 to 100 mg/kg/day, or about 1 to 35 mg/kg/day, administered in single or divided dosage units (e.g., BID, TID, QID). Exemplary ranges for suitable dosages for a 70-kg human are from about 0.05 to about 7 g/day, or from about 0.2 to about 2.5 g/day.

An example of a dosage of the compound is from about 1mg to about 2,500 mg. In some embodiments, the dose of a compound of the present disclosure for use in the compositions described herein is less than about 10,000mg, or less than about 8,000mg, or less than about 6,000mg, or less than about 5,000mg, or less than about 3,000mg, or less than about 2,000mg, or less than about 1,000mg, or less than about 500mg, or less than about 200mg, or less than about 50 mg. Similarly, in some embodiments, the dose of the second compound (i.e., another drug for HBV treatment) as described herein is less than about 1,000mg, or less than about 800mg, or less than about 600mg, or less than about 500mg, or less than about 400mg, or less than about 300mg, or less than about 200mg, or less than about 100mg, or less than about 50mg, or less than about 40mg, or less than about 30mg, or less than about 25mg, or less than about 20mg, or less than about 15mg, or less than about 10mg, or less than about 5mg, or less than about 2mg, or less than about 1mg, or less than about 0.5mg, and any and all whole or partial increments thereof.

Once the patient's disease, disorder or condition has improved, the dosage can be adjusted for prophylactic or maintenance treatment. For example, as symptoms change, the dose or frequency of administration, or both, can be reduced to a level that maintains the desired therapeutic or prophylactic effect. Of course, if the symptoms have been alleviated to an appropriate level, treatment may be discontinued. However, when any symptoms recur, the patient may require intermittent treatment for a long period of time.

Examples of the invention

Exemplary compounds useful in the methods of the present disclosure will now be described with reference to the following illustrative synthetic schemes for their general preparation and the specific examples that follow. The skilled artisan will recognize that, in order to obtain the various compounds herein, the starting materials may be suitably selected such that the ultimately desired substituent will be carried through the reaction scheme with or without suitable protection to yield the desired product. Alternatively, it may be necessary or desirable to replace the ultimately desired substituent with a suitable group that can be carried through the reaction scheme and appropriately replaced with the desired substituent. Variables are as defined above with reference to formula (I), unless otherwise specified. The reaction may be carried out between the melting point of the solvent and the reflux temperature, preferably between 0 ℃ and the reflux temperature of the solvent. Conventional heating or microwave heating may be employed to heat the reaction. The reaction can also be carried out in a sealed pressure vessel above the normal reflux temperature of the solvent.

Abbreviations and acronyms used herein include those shown in table 2 below:

table 2:

synthesis of

Exemplary compounds useful in the methods of the present disclosure will now be described with reference to the following illustrative synthetic schemes for their general preparation and the specific examples that follow.

Scheme 1

According to scheme 1, β ketoesters having formula (IV) are prepared by reacting a compound having formula (II) (where PG is a suitable nitrogen protecting group, such as Boc and the like, and R is3And R4Is H or C1-4Alkyl) with a compound of formula (III) (wherein Het is an optionally substituted five or six membered heteroaryl ring (e.g. pyrimidine etc.)), a suitable base (e.g. L iHMDS or L DA etc.) in a solvent (e.g. THF or dioxane) at a temperature in the range of 0 ℃ to 15 ℃ for 3-6h to form, for example, a compound of formula (II) (wherein PG is Boc, R is Boc)3Is H, and R4Is CH3) Reaction with a suitable base (such as L iHMDS), a compound of formula (III) wherein Het is a pyrimidine in a solvent (such as THF) to provide a compound of formula (IV) β keto ester of formula (IV) is reacted with hydroxylamine hydrochloride in a suitable solvent (such as EtOH and the like) at a temperature of about 70 ℃ for about 60h to provide isoxazole compounds of formula (Va) and formula (Vb) cyclizing to isoxazole and subsequent deprotection in situ.

Scheme 2

According to scheme 2, compounds having formula (VII) (wherein R1And R2Is C1-4E.g., 3, 3-dimethyl-4-oxo-piperidine-1-carboxylic acid tert-butyl ester at a temperature of about-78 ℃ with a base (e.g., L iHMDS) in a solvent (e.g., THF), followed by addition of a compound of formula (III) (wherein Het is a pyrimidine) at a temperature range of-78 ℃ to 20 ℃ for 2h to provide a compound of formula (VII): the compounds of formula (VIIIa) and (VIIIb) are prepared from the compound of formula (VII) in two steps1And R2Is C1-4Alkyl, and PG is Boc) with hydroxylamine hydrochloride and NaOAc in a suitable solvent (e.g., EtOH, etc.) at a temperature of about 70 ℃ for about 60h to provide an oxime intermediate. In a second step, the oxime intermediate is reacted with MsCl, a base (e.g., TEA, etc.) in a solvent (e.g., DCM, etc.) at a temperature of about 0 ℃ to 25 ℃ to provide the cyclized isoxazole compound having formula (VIIIa) and (VIIIb). Deprotecting a compound having formula (VIIIa) or (VIIIb) with an acid (e.g., TFA, HCl, etc.) in a suitable solvent (e.g., DCM, etc.) to provide a compound having formula (IXa) or (IXb).

Scheme 3

According to scheme 3, compounds of formula (X) [ and Va, Vb, IXa, IXb](wherein R is1、R2、R3And R4Each independently is H or C1-4Alkyl radical, and X1And X2Each independently O or N) with a commercially available or synthetically produced compound of the formula (XI), in which X is3And X4Each independently is C-R6Or N; wherein when X is3Is C-R6Or N is, X4Is C-R6(ii) a Wherein when X is4Is C-R6Or N is, X3Is C-R6;R5And R6Each independently selected from the group consisting of: H. halo, C1-4Haloalkyl and CN; and n is 1 or 2; a base (e.g., TEA or DIEA, etc.) in a suitable solvent (e.g., DCM or DCE) to provide a compound having formula (I).

The compounds of formula (I) may be converted into their corresponding salts using methods known to those of ordinary skill in the art. For example, in Et2O、CH2Cl2The amine of formula (I) is treated with trifluoroacetic acid, HCl or citric acid in a solvent of THF, MeOH, chloroform or isopropanol to provide the corresponding salt form alternatively, the conditions are purified by reverse phase HP L C, thus obtaining trifluoroacetic acid or a formate salt the crystalline form of the pharmaceutically acceptable salt of the compound of formula (I) is obtained in crystalline form by recrystallization from a polar solvent (including mixtures of polar solvents and aqueous mixtures of polar solvents) or from a non-polar solvent (including mixtures of non-polar solvents).

When compounds according to the present disclosure have at least one chiral center, they may accordingly exist as enantiomers. When the compounds have two or more chiral centers, they may additionally exist as diastereomers. It is understood that all such isomers and mixtures thereof are encompassed within the scope of the present disclosure.

The compounds prepared according to the above schemes may be obtained in a single form (e.g. a single enantiomer) by synthesis of the particular form or by resolution. The compounds prepared according to the above schemes may alternatively be obtained as mixtures of various forms, such as racemic (1: 1) or non-racemic (non-1: 1) mixtures. When racemic and non-racemic mixtures of enantiomers are obtained, the individual enantiomers may be separated using conventional separation methods known to those of ordinary skill in the art, such as chiral chromatography, recrystallization, salt formation of the diastereomers, adducts derived as diastereomers, biotransformation, or enzymatic transformations. Where a mixture of regioisomers or diastereomers is obtained, the individual isomers may be separated, where applicable, using conventional methods, such as chromatography or crystallization.

Examples of the invention

The following specific examples are provided to further illustrate the disclosure and various preferred embodiments.

In obtaining the compounds and corresponding analytical data described in the examples below, the following experimental and analytical protocols were followed, unless otherwise indicated.

Unless otherwise stated, the reaction mixture was magnetically stirred at room temperature (rt) under a nitrogen atmosphere. When the solutions are "dried", they are usually passed through a drying agent (e.g., Na)2SO4Or MgSO 24) Drying is carried out. When the mixture, solution and extract are "concentrated", they are typically concentrated under reduced pressure in a rotary evaporator.

Pre-packed column on silica gel (SiO)2) Normal phase silica gel chromatography (FCC) was performed above.

Preparative reverse phase high performance liquid chromatography (RP HP L C) was performed under any of the following methods:

method A.Gilson GX-281 semi-manufactured HP L C, using Synergic C18(10 μm, 150x25mm) or Boston Green ODS C18(5 μm, 150x30mm) from Phinomenex, and flowing at 5% -99% ACN (containing 0.225% FA) over 10min in water and then held for 2min in 100% ACN at a flow rate of 25m L/min.

Or

Gilson GX-281 semi-preparative HP L C, using Synergi C18(10 μm, 150X25mm) or Boston Green ODS C18(5 μm, 150X30mm) from Philomo, and 10min 5% -99% ACN (0.1% TFA) in water as the mobile phase, and then held for 2min in 100% ACN at a flow rate of 25m L/min or

Method C.Gilson GX-281 semi-manufactured HP L C, using Synergi C18(10 μm, 150X25mm) or Boston Green ODS C18(5 μm, 150X30mm) from Philomo and flowing 10min 5% -99% ACN (0.05% HCl) in water and then maintaining for 2min in 100% ACN at a flow rate of 25m L/min.

Or

Method d.gilson GX-281 semi-preparative HP L C, using Gemini C18(10 μm, 150x25mm), AD (10 μm, 250mmx30mm), or Waters XBridge C18 column (5 μm, 150x30mm) from philippine corporation, mobile phase 0% -99% ACN (0.05% ammonium hydroxide v/v) over 10min in water and then held for 2min in 100% ACN at a flow rate of 25m L/min.

Or

Method e.gilson GX-281 semi-preparative HP L C, using Gemini C18(10 μm, 150x25mM) from philips inc or a Waters XBridge C18 column (5 μm, 150x30mM) with a mobile phase of 5% -99% ACN (10mM NH4HCO3) in water over 10min, and then held in 100% ACN for 2min at a flow rate of 25m L/min.

Preparative supercritical fluid high performance liquid chromatography (SFC) is carried out on a Thar 80Prep-SFC system or a Waters 80QPrep-SFC system. ABPR was set to 100 bar for CO2Maintained under SF conditions and flow rates were verified by compound characterization, ranging from 50g/min to 70 g/min. The column temperature is ambient temperature

Unless otherwise indicated, Mass Spectra (MS) were obtained by electrospray ionization (ESI) in positive mode on a SHIMADZU L CMS-2020MSD or Agilent 1200\ G6110A MSD.

Nuclear Magnetic Resonance (NMR) spectra were obtained on a Bruker model avim 400 spectrometer. The multiplicities are defined as follows: s is singlet, d is doublet, t is triplet, q is quartet, m is multiplet, br is broad. It will be appreciated that for compounds containing exchangeable protons, the protons may or may not be visible in the NMR spectrum, depending on the choice of solvent used to run the NMR spectrum and the concentration of the compound in solution.

Chemical names were generated using ChemDraw Ultra 12.0, ChemDraw Ultra 14.0 (cambridge software corporation, harvard university, ma, cambridge Corp.) or ACD/Name Version 10.01 (Advanced Chemistry).

The compounds designated R or S are enantiomerically pure compounds with no defined absolute configuration.

Intermediate 1: (S) -6-methyl-3- (pyrimidine-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 3-c]Pyridine.

Step a. (2S) -2-methyl-4-oxo-5- (pyrimidine-2-carbonyl) piperidine-1-carboxylic acid tert-butyl ester.To a mixture of (S) -tert-butyl 2-methyl-4-oxopiperidine-1-carboxylate (5g, 23.44mmol) in THF (40M L) at 0 deg.C under N2 was added L iHMDS (1M, 46.88M L) in one portion, the reaction mixture was stirred at 0 deg.C for 0.5h, then methyl pyrimidine-2-carboxylate (5.50g, 39.85mmol) in THF (40M L) was added to the solution at 0 deg.C and stirred at 15 deg.C for 4h4Cl (200m L) and stirring for 1min the aqueous phase was extracted with ethyl acetate (200m L× 2) the organic layer was separated, washed with brine (100m L× 2) and dried over anhydrous Na2SO4Dried, filtered and concentrated in vacuo. Purification (FCC, SiO)2Petroleum ether/ethyl acetate 10/1 to 0: 1) gave tert-butyl (2S) -2-methyl-4-oxo-5- (pyrimidine-2-carbonyl) piperidine-1-carboxylate as a yellow oil (5.5g, 17.22mmol, 73.47% yield). Ms (esi): for C16H21N3O4Is 319.2; found M/z 342.1[ M + Na]+

(S) -6-methyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 3-c]Pyridine compound

To a solution of (2S) -2-methyl-4-oxo-5- (pyrimidine-2-carbonyl) piperidine-1-carboxylic acid tert-butyl ester (0.5g, 1.57mmol) in EtOH (15M L) was added hydroxylamine hydrochloride (587.51mg, 8.45mmol), the reaction mixture was stirred at 70 ℃ for 60h, the reaction mixture was washed with EtOAc (15M L× 3), then the pH of the aqueous phase was adjusted to 9 by aqueous NaOH (1M), the aqueous layer was extracted with DCM (20M L× 3), the combined DCM layers were washed with brine (30M L), over Na L2SO4The residue was purified via RP HP L C (Condition D), followed by SFC (column: AD (250mm 30mm, 10 μm); mobile phase: [ 0.1% NH ]3H2O MeOH](ii) a B%: 50% -50%) to give a yellow solidThe title compound (Rt ═ 4.121min, 60mg, 76.80% yield, 96% purity) and (S) -6-methyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 5-c]Pyridine. Ms (esi): for C11H12N4The calculated mass of O is 216.1; found M/z, 217.0[ M +1]+1HNMR(400MHz,CDCl3)8.87(d,J=4.8Hz,2H),7.29-7.26(m,1H),4.53-4.49(m,1H),4.13-4.09(m,1H),3.07-2.99(m,2H),2.48-2.41(m,1H),1.34(d,J=6.4Hz,3H)。

Intermediate 2: (S) -6-methyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 5-c]Pyridine compound

From intermediate 1, step B: (retention time 3.603min, 50mg, 64.00% yield, 96% purity) the title compound was isolated. Ms (esi): for C11H12N4The calculated mass of O is 216.1; found M/z, 217.0[ M +1]+1HNMR(400MHz,CDCl3)8.86(d,J=4.8Hz,2H),7.31(t,J=4.8Hz,1H),4.32-4.28(m,1H),4.12-4.03(m,1H),3.16-3.05(m,1H),2.96-2.86(m,1H),2.49-2.42(m,1H),1.34(d,J=6.4Hz,3H)。

Intermediate 3: 7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 3-c]Pyridine-5 (4H) -formazan Tert-butyl ester

Step A.3, 3-dimethyl-4-oxo-5- (pyrimidine-2-carbonyl) piperidine-1-carboxylic acid tert-butyl esterTo a solution of 3, 3-dimethyl-4-oxo-piperidine-1-carboxylic acid tert-butyl ester (3g, 13.20mmol) in THF (30M L) at-78 ℃ was added L iHMDS (1M, 26.40M L.) the reaction mixture was heated at-78 ℃ under N2Stirring for 0.5 h. At-78 ℃ under N2Next, pyrimidine-2-carboxylic acid methyl ester (3.10g, 22.44mmol) was added to the mixture. Mixing the mixture inAt 20 ℃ under N2Stirred under atmosphere for 2 h. To react with NH4Aqueous Cl (80m L) and then neutralized by dilute HCl (1N, 80m L.) the aqueous layer was extracted with EtOAc (40m L X3.) the combined organic layers were dried over anhydrous Na2SO4Dried, filtered and concentrated in vacuo. Purification (FCC, SiO)2Petroleum ether/ethyl acetate 100/1 to 3: 1) gave the title compound as a yellow solid (3.8g, 11.25mmol, 85.24% yield, 98.7% purity). Ms (esi): for C17H23N3O4The calculated mass of (3) is 333.2; found M/z 356.1[ M + Na]+

Step B.7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 3-c]Pyridine-5 (4H) -carboxylic acid Tert-butyl ester and 7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 5-c]Pyridine-5 (4H) -carboxylic acid tert-butyl ester Esters.3, 3-dimethyl-4-oxo-5- (pyrimidine-2-carbonyl) piperidine-1-carboxylic acid tert-butyl ester (2.6g, 7.80mmol), AcONa (1.28g, 15.60mmol), NH2A mixture of OH & HCl (1.08g, 15.60mmol) in EtOH (30m L) was stirred at 60 ℃ for 16hr, the reaction mixture was cooled and concentrated under reduced pressure, the residue was diluted with ethyl acetate (30m L) and washed with H2O (50m L) the organic layer was separated, dried, filtered and concentrated under reduced pressure to give 3g of crude product as a yellow solid which was used directly in the next step without further purification at 0 ℃ under N2Next, to a cooled solution of the crude product (3g) in DCM (30m L) were added TEA (1.31g, 12.92mmol, 1.80m L) and MsCl (986.39mg, 8.61mmol, 666.48 μ L) the reaction mixture was heated at 25 ℃ under N2Stir 2h under stirring the reaction mixture was poured into ice water (30m L) and stirred for 1min the aqueous phase was extracted with DCM (30m L x2) the combined organic layers were washed with brine (60m L), separated and washed with anhydrous Na2SO4Dried, filtered and concentrated under reduced pressure. Purification (SiO)2Petroleum ether/ethyl acetate 100/1 to 1: 1) gave 1.9g of a mixture of the title compound, purified (SFC (OD-3S _3_5_40_3M L column: Chiralcel OD-3100 × 4.6mm i.d., 3 μ M, mobile phase methanol (0.05% DEA) in CO2From 5 percent to 40 percent, the flow rate is 3m L/min, the wavelength is 220nm)) to give the title compound (7, 7-dimethyl-3-pyrimidin-2-yl-4, 6-dihydroisoxazolo [4, 3-c) as a white solid]Pyridine-5-carboxylic acid tert-butyl ester, 256mg, 16.38% yield, 91% purity); and 7, 7-dimethyl-3-pyrimidin-2-yl-4, 6-dihydroisoxazolo [4, 5-c as a white solid]Pyridine-5-carboxylic acid tert-butyl ester (1.52g, 99% purity). Ms (esi): for C17H22N4O3The calculated mass of (a) is 330.2; found M/z 353.1[ M + Na]+

Step C.7, 7-dimethyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 3-c]Pyridine.To 7, 7-dimethyl-3-pyrimidin-2-yl-4, 6-dihydroisoxazolo [4, 3-c at 30 ℃]To a solution of pyridine-5-carboxylic acid tert-butyl ester (350mg, 1.06mmol, 1 equiv.) in DCM (4m L) was added TFA (4.01g, 35.15mmol, 2.60m L, 33.18 equiv.) with stirring for 1 hour the reaction mixture was concentrated under reduced pressure to give the title compound as a TFA salt as a yellow oil (391mg), which was used without further purification MS (ESI) for C12H14N4The calculated mass of O is 230.1; found M/z, 231.0[ M + H]+

Intermediate 4: 7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4, 5-c]Pyridine-5 (4H) -formazan Tert-butyl ester

The title compound was synthesized in a similar manner to intermediate 3, step C, using 7, 7-dimethyl-3-pyrimidin-2-yl-4, 6-dihydroisoxazolo [4, 5-C]Pyridine-5-carboxylic acid tert-butyl ester (product from intermediate 3, step 2) was prepared. The crude title compound was used without further purification. Ms (esi): for C12H14N4The calculated mass of O is 230.1; found M/z, 231.0[ M + H]+。Example 1: (S) -N- (3-cyano-4-fluorophenyl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazole And [4, 3-c ]]Pyridine-5 (4H) -carboxamide.

To (S) -6-methyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 3-c]To a solution of pyridine (intermediate 1, 50mg, 217.35 μmol) in DCM (5m L) were added TEA (116.99mg, 1.16mmol, 160.92 μ L) and phenyl (3-cyano-4-fluorophenyl) carbamate (56.85mg, 219.66 μmol). the reaction mixture was stirred at 20 ℃ for 3 h. the reaction mixture was concentrated under reduced pressure purification (RP HP L C (condition a)) to give the title compound (55.29mg, 61.97% yield, 98.05% purity) as a white solid ms (esi) for C19H15FN6O2Is 378.1; found M/z 379.1[ M + H]+1H NMR(400MHz,CDCl3)8.93(d,J=4.8Hz,2H),7.74-7.72(m,1H),7.70-7.65(m,1H),7.36(t,J=4.8Hz,1H),7.18(t,J=8.8Hz,1H),6.63(s,1H),5.19-5.15(m,1H),5.13-5.05(m,1H),4.66-4.62(d,1H),3.18-3.10(m,1H),3.02-2.95(m,1H),1.26(d,J=7.2Hz,3H)。

Example 2: (S) -N- (, 2- (difluoromethyl) -3-fluoropyridin-4-yl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-di Hydroisoxazolo [4, 3-c ] s]Pyridine-5 (4H) -carboxamide.

The title compound was prepared in analogy to example 1 by substituting phenyl (3-cyano-4-fluorophenyl) carbamate with phenyl (2- (difluoromethyl) -3-fluoropyridin-4-yl) carbamate. Ms (esi): for C18H15F3N6O2Is 404.1; found M/z, 405.1[ M + H]+1H NMR(400MHz,CDCl3)8.94(d,J=4.8Hz,2H),8.38-8.34(m,2H),7.37(t,J=4.8Hz,1H),7.13(d,J=4.0Hz,1H),6.91-6.61(m,1H),5.27-5.20(m,1H),5.25-5.21(m,1H),5.11-5.08(m,1H),4.73-4.69(m,1H),4.75-4.68(m,1H),3.20-3.11(m,1H),3.06-2.98(m,1H),1.29(d,J=7.2Hz,3H)。

Example 3: (S) -N- (4-fluoro-3- (trifluoromethyl) phenyl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazole Azolo [4, 3-c ] s]Pyridine-5 (4H) -carboxamide.

The title compound was prepared in analogy to example 1 by substituting phenyl (3-cyano-4-fluorophenyl) carbamate with phenyl (4-fluoro-3- (trifluoromethyl) phenyl) carbamate. Ms (esi): for C19H15F4N5O2Is 421.1; found M/z 422.1[ M + H]+1H NMR(400MHz,CDCl3)8.93(d,J=4.8Hz,2H),7.68-7.60(m,2H),7.36(t,J=4.8Hz,1H),7.16(t,J=9.2Hz,1H),6.65(s,1H),5.21-5.17(m,1H),5.12-5.06(m,1H),4.65-4.61(m,1H),3.18-3.10(m,1H),3.02-2.94(m,1H),1.25(d,J=6.8Hz,3H)。

Example 4: (S) -N- (2-bromo-3-fluoropyridin-4-yl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazole And [4, 3-c ]]Pyridine-5 (4H) -carboxamide.

The title compound was prepared in analogy to example 1 by substituting phenyl (3-cyano-4-fluorophenyl) carbamate with phenyl (2-bromo-3-fluoropyridin-4-yl) carbamate. Ms (esi): for C17H14BrFN6O2Is 432.0; found M/z 433.0/435.0[ M + H ]]+1H NMR(400MHz,CDCl3)8.94(d,J=4.8Hz,2H),8.23-8.16(m,1H),8.10(d,J=5.2Hz,1H),7.37(t,J=4.8Hz,1H),7.04(s,1H),5.24-5.20(m,1H),5.13-5.04(m,1H),4.72-4.68(m,1H),3.20-3.12(m,1H),3.05-2.97(m,1H),1.28(d,J=7.2Hz,3H)。

Example 5: (S) -N- (3-cyano-4-fluorophenyl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4,5-c]Pyridine-5 (4H) -carboxamide.

In a similar manner to example 1, with (S) -6-methyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 5-c)]Pyridine (intermediate 2) substituted (S) -6-methyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 3-c)]Pyridine (intermediate 1) the title compound was prepared. Ms (esi): for C19H15FN6O2Is 378.1; found M/z 379.1[ M + H]+1H NMR(400MHz,CDCl3)8.92(d,J=4.8Hz,2H),7.73-7.71(m 1H),7.70-7.65(m,1H),7.40(t,J=4.8Hz,1H),7.17(t,J=8.8Hz,1H),6.74(s,1H),5.20-5.15(m,1H),5.06-5.02(m,1H),4.54-4.50(m,1H),3.22-3.17(m,1H),2.85-2.81(m,1H),1.27(d,J=6.8Hz,3H)。

Example 6: (S) -N- (2- (difluoromethyl) -3-fluoropyridin-4-yl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-di Hydroisoxazolo [4, 5-c ] s]Pyridine-5 (4H) -carboxamides

In a similar manner to example 1, using (S) -6-methyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 5-c)]The title compound was prepared from pyridine (intermediate 2) and phenyl (2- (difluoromethyl) -3-fluoropyridin-4-yl) carbamate. Ms (esi): for C18H15F3N6O2Is 404.1; found M/z, 405.1[ M + H]+1H NMR(400MHz,CDCl3)8.94(d,J=4.8Hz,2H),8.96-8.92(m,1H),8.37-8.32(m,2H),7.41(t,J=4.8Hz,1H),7.14(d,J=4.0Hz,1H),6.90-6.61(m,1H),5.22-5.08(m,2H),4.62-4.58(m,1H),4.64-4.56(m,1H),3.26-3.20(m,1H),2.90-2.86(m,1H),2.93-2.84(m,1H),1.31(d,J=6.8Hz,3H)。

Example 7:(S) -N- (4-fluoro-3- (trifluoromethyl) phenyl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazole Azolo [4, 5-c]Pyridine-5 (4H) -carboxamide.

In a similar manner to example 1, using (S) -6-methyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 5-c)]Pyridine (intermediate 2) was reacted with phenyl (4-fluoro-3- (trifluoromethyl) phenyl) carbamate to prepare the title compound. Ms (esi): for C19H15F4N5O2Is 421.1; found M/z 422.1[ M + H]+1H NMR(400MHz,CDCl3)8.93(d,J=4.8Hz,2H),7.67-7.60(m,2H),7.41(t,J=4.8Hz,1H),7.16(t,J=9.2Hz,1H),6.62(s,1H),5.20-5.14(m,1H),5.07-5.03(m,1H),4.55-4.51(m,1H),3.24-3.18(m,1H),2.86-2.82(m,1H),1.27(d,J=6.8Hz,3H)。

Example 8: (S) -N- (2-bromo-3-fluoropyridin-4-yl) -6-methyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazole And [4, 5-c ]]Pyridine-5 (4H) -carboxamide.

In a similar manner to example 1, using (S) -6-methyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 5-c)]Pyridine (intermediate 2) and phenyl (2-bromo-3-fluoropyridin-4-yl) carbamate the title compound was prepared. Ms (esi): for C17H14BrFN6O2Is 432.0; found M/z 433.0/435.0[ M + H ]]+1H NMR(400MHz,CDCl3)8.94(d,J=4.8Hz,2H),8.21-8.15(m,1H),8.10(d,J=5.4Hz,1H),7.41(t,J=4.8Hz,1H),7.06(br s,1H),5.20-5.14(m,1H),5.12-5.08(m,1H),4.61-4.57(m,1H),3.25-3.20(m,1H),2.89-2.85(m,1H),1.30(d,J=6.8Hz,3H)。

Example 9: n- (3-cyano-4-fluoro)Phenyl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4,3-c]Pyridine-5 (4H) -carboxamide.

In a similar manner to example 1, 7-dimethyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 3-c ] was used]Pyridine (intermediate 3) and phenyl (3-cyano-4-fluorophenyl) carbamate. Ms (esi): for C20H17FN6O2Is 392.1; found M/z 393.1[ M + H]+1H NMR(400MHz,CDCl3)8.94(s,1H),7.81-7.67(m,2H),7.36(t,J=4.9Hz,1H),7.23-7.14(m,1H),6.70(s,1H),4.97(s,2H),3.62(s,2H),1.52(s,6H)。

Example 10: n- (4-fluoro-3- (trifluoromethyl) phenyl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroiso Oxazolo [4, 3-c]Pyridine-5 (4H) -carboxamide.

In a similar manner to example 1, 7-dimethyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 3-c ] was used]Pyridine (intermediate C) and phenyl (4-fluoro-3- (trifluoromethyl) phenyl) carbamate. Ms (esi): for C20H17F4N5O2The calculated mass of (d) is 435.1; found m/z, 436.1.[ M + H ]]+1HNMR(400MHz,CDCl3)8.94(d,J==4.9Hz,2H),7.72-7.62(m,2H),7.36(t,J==4.9Hz,1H),7.18(t,J==9.3Hz,1H),6.66(s,1H),4.97(s,2H),3.62(s,2H),1.52(s,6H)。

Example 11: n- (2- (difluoromethyl) -3-fluoropyridin-4-yl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-di Hydroisoxazolo [4, 3-c ] s]Pyridine-5 (4H) -carboxamide.

In a similar manner to example 1, 7-dimethyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 3-c ] was used]Pyridine (intermediate 3) and phenyl (2- (difluoromethyl) -3-fluoropyridin-4-yl) carbamate the title compound was prepared. Ms (esi): for C19H17F3N6O2The calculated mass of (a) is 418.1; found M/z 419.1[ M + H]+1HNMR(400MHz,CDCl3)8.94(d,J=4.9Hz,2H),8.45-8.34(m,2H),7.37(t,J=4.9Hz,1H),7.17(br d,J=4.1Hz,1H),6.92-6.60(m,1H),5.03(s,2H),3.64(s,2H),1.53(s,6H)。

Example 12: n- (2-bromo-3-fluoropyridin-4-yl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazole And [4, 3-c ]]Pyridine-5 (4H) -carboxamide.

In a similar manner to example 1, 7-dimethyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 3-c ] was used]Pyridine (intermediate 3) and phenyl (2-bromo-3-fluoropyridin-4-yl) carbamate the title compound was prepared. Ms (esi): for C18H16BrFN6O2Calculated mass of (d) is 446.1; found M/z, 447.0/449.0.[ M + H ]]+1HNMR(400MHz,CDCl3)8.94(d,J=4.9Hz,2H),8.26-8.08(m,2H),7.37(t,J=5.0Hz,1H),7.09(brd,J=4.0Hz,1H),5.02(s,2H),3.63(s,2H),1.53(s,6H)。

Example 13: n- (3-cyano-4-fluorophenyl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazolo [4,5-c]Pyridine-5 (4H) -carboxamide.

In a similar manner to example 1, use 77-dimethyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 5-c]The title compound was prepared by reaction of pyridine (intermediate 4) and phenyl (3-cyano-4-fluorophenyl) carbamate. Ms (esi): for C20H17FN6O2Is 392.1; found M/z 393.1.[ M + H ]]+1H NMR(400MHz,CDCl3)8.92(t,J=4.2Hz,2H),7.82-7.65(m,2H),7.41(t,J=4.6Hz,1H),7.18(t,J=8.6Hz,1H),6.89-6.73(m,1H),4.82(s,2H),3.67(s,2H),1.47(s,6H)。

Example 14: n- (4-fluoro-3- (trifluoromethyl) phenyl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroiso Oxazolo [4, 5-c]Pyridine-5 (4H) -carboxamide.

In a similar manner to example 1, 7-dimethyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 5-c ] was used]Pyridine (intermediate 4) and phenyl (4-fluoro-3- (trifluoromethyl) phenyl) carbamate. Ms (esi): for C20H17F4N5O2The calculated mass of (d) is 435.1; found M/z 436.1.[ M + H ]]+1HNMR(400MHz,CDCl3)8.93(d,J=4.9Hz,2H),7.70-7.61(m,2H),7.40(t,J=4.9Hz,1H),7.25-7.08(m,1H),6.69(s,1H),4.83(s,2H),3.67(s,2H),1.47(s,6H)。

Example 15: n- (2- (difluoromethyl) -3-fluoropyridin-4-yl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-di Hydroisoxazolo [4, 5-c ] s]Pyridine-5 (4H) -carboxamide.

In a similar manner to example 1, 7-dimethyl-3- (pyrimidin-2-yl) -4, 5, 6, 7-tetrahydroisoxazolo [4, 5-c ] was used]Preparation of the titled compound from pyridine (intermediate 4) and phenyl (2- (difluoromethyl) -3-fluoropyridin-4-yl) carbamateA compound (I) is provided. Ms (esi): for C19H17F3N6O2The calculated mass of (a) is 418.1; found M/z 419.1[ M + H]+1HNMR(400MHz,CDCl3)8.94(d,J=4.9Hz,2H),8.42-8.35(m,2H),7.42(t,J=4.9Hz,1H),7.28(s,1H),7.18(br d,J=4.3Hz,1H),6.93-6.59(m,1H),4.90(s,2H),3.69(s,2H),1.49(s,6H)。

Example 16: n- (2-bromo-3-fluoropyridin-4-yl) -7, 7-dimethyl-3- (pyrimidin-2-yl) -6, 7-dihydroisoxazole And [4, 5-c ]]Pyridine-5 (4H) -carboxamide.

In a similar manner to example 1, 7-dimethyl-3-pyrimidin-2-yl-5, 6-dihydro-4H-isoxazolo [4, 5-c ] was used]Pyridine (intermediate 4) and phenyl N- (2-bromo-3-fluoro-4-pyridyl) carbamate. Ms (esi): for C18H16BrFN6O2Calculated mass of (d) is 446.1; found M/z 447.0/449.0[ M + H ]]+1H NMR(400MHz CDCl3)8.93(d,J=5.0Hz,2H),8.23-8.18(m,1H),8.13-8.08(m,1H),7.42-7.38(m,1H),7.10-7.05(m,1H),4.93-4.85(m,2H),3.67(s,2H),1.47(s,6H)。

Biological data

HBV replication inhibition assay

The inhibitory effect of the disclosed compounds on HBV replication was determined in cells infected or transfected with HBV or cells with stably integrated HBV (e.g. hepgs 2.2.15 cells) (Sells et al 1987.) in this example hepgs 2.2.15 cells were maintained in cell culture medium containing 10% Fetal Bovine Serum (FBS), geneticin, L-glutamine, penicillin and streptomycin hepgs 2.2.15 cells were seeded in 96 well plates at a density of 40,000 cells/well and treated in checkerboard (checker box) format with serially diluted compounds with a final DMSO concentration of 0.5% or by adding drug combinations, cells were incubated with compounds for 3 days, then the medium was removed and fresh medium containing compounds was added to the cells and further day 3, the supernatant was removed and treated with DNA enzyme at 37 ℃ for 60 minutes, then enzyme inactivation at 75 ℃ for 15 minutes by adding fresh medium containing compounds to the cells and the cells were extracted with a PCR kit containing the DNA polymerase for the following the PCR protocol of the PCR-mediated inactivation of HBV DNA by adding additional DNA polymerase for the cells using the PCR-DNA polymerase enzyme amplification of the PCR-amplified HBV DNA by the PCR-DNA polymerase (pgna) and the PCR assay kit-DNA amplification of the PCR assay kit for the fluorescence of all DNA-DNA extracted from the cells using the PCR-DNA-amplified DNA of the PCR-DNA-amplified HBV-DNA-protein-DNA-enriched cells (pgna-enriched cell culture medium alone was determined by the PCR-enriched recombinant DNA-enriched with the PCR-enriched recombinant DNA-enriched hepga-DNA-enriched HBV-enriched DNA-enriched recombinant DNA-enriched DNA.

E1: % inhibition ═ DMSOave-Xi)/DMSOave x 100%

Where DMSOave is the average signal calculated from wells treated with DMSO control (0% inhibition control), Xi is the signal measured from individual wells. EC was determined by non-linear fitting using Graphpad Prism software (san Diego, Calif.) and equation E250The value, i.e., the effective concentration to achieve 50% inhibition.

E2: Y ═ Ymin + (Ymax-Ymin)/(1+10 (L ogEC50-X) X hill slope) where Y represents the percent inhibition value and X represents the logarithm of the compound concentration.

Selected disclosed compounds were assayed in the HBV replication assay (BDNA assay) as described above, and a representative set of these active compounds is shown in table 3. Table 3 shows the EC obtained by BDNA assay for a selected group of compounds50The value is obtained. In Table 3, "A" means 1nM < EC50≤100nM;"B" means 100nM < EC50Less than or equal to 1,000 nM; and "C" means 1,000nM < EC50≤10,000nM。

TABLE 3 Activity in BDNA assay (EC)50)

The disclosure of each and every patent, patent application, and publication cited herein is hereby incorporated by reference in its entirety.

Although the present disclosure has been disclosed with reference to specific embodiments, it is apparent that other embodiments and variations of the present disclosure may be devised by others skilled in the art without departing from the true spirit and scope of the present disclosure. It is intended that the following claims be interpreted to embrace all such embodiments and equivalents.

50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:Bcl-2抑制剂

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!