Therapeutic compounds and compositions and methods of use thereof

文档序号:1590579 发布日期:2020-01-03 浏览:21次 中文

阅读说明:本技术 治疗化合物和组合物及其使用方法 (Therapeutic compounds and compositions and methods of use thereof ) 是由 M·扎克 F·A·罗梅罗 Y-X·成 于 2018-05-21 设计创作,主要内容包括:本文描述可用作JAK激酶抑制剂的化合物及其盐。还提供包括所述JAK抑制剂和药学上可接受的载体、佐剂或媒介物的药物组合物,和治疗响应于Janus激酶活性抑制的患者疾病或病症或减轻其严重程度的方法。(Described herein are compounds and salts thereof that are useful as JAK kinase inhibitors. Also provided are pharmaceutical compositions comprising the JAK inhibitors and a pharmaceutically acceptable carrier, adjuvant or vehicle, and methods of treating or lessening the severity of a disease or disorder in a patient responsive to inhibition of Janus kinase activity.)

1. a compound of formula (I) or a pharmaceutically acceptable salt or stereoisomer thereof,

Figure FDA0002282880330000011

wherein:

R1is hydrogen or CH3

R2Is halogen, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl OR-ORaWherein R is2Optionally substituted by one or more groups independently selected from halogen, C1-C3Alkyl, cyano, hydroxy and oxo;

Rais C1-C6Alkyl, -phenyl-CORbRc-phenyl- (3-6-membered heterocyclyl) or 3-11-membered heterocyclyl, wherein R isaOptionally substituted by one or more groups independently selected from halogen, C1-C3Alkyl, cyano, hydroxy and oxo;

each RbAnd RcIndependently is hydrogen or CH3

R3Is hydrogen or NH2

R4Is hydrogen or CH3(ii) a And is

R5Is hydrogen or NH2

2. The compound of claim 1, or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R1Is hydrogen.

3. The compound of claim 1, or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R1Is CH3

4. RightsA compound according to any one of claims 1 to 3, or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R3Is hydrogen.

5. A compound according to any one of claims 1 to 4, or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each R4And R5Is hydrogen.

6. The compound of claim 1, or a pharmaceutically acceptable salt or stereoisomer thereof, wherein each R1、R3、R4And R5Is hydrogen.

7. A compound according to any one of claims 1 to 6, or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R2Is C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl OR-ORaWherein R is2Optionally substituted by one or more groups independently selected from halogen, C1-C3Alkyl, cyano, hydroxy and oxo.

8. A compound according to any one of claims 1 to 6, or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R2Selected from halogen, C1-C6Haloalkyl and C1-C6A haloalkoxy group.

9. The compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R2Is selected from

Figure FDA0002282880330000021

10. A compound selected from the following 1-18, or a pharmaceutically acceptable salt or stereoisomer thereof:

Figure FDA0002282880330000022

Figure FDA0002282880330000031

Figure FDA0002282880330000041

11. a compound which is

Figure FDA0002282880330000042

12. A compound which is

Figure FDA0002282880330000043

13. A compound which is

Figure FDA0002282880330000051

14. A compound which is

Figure FDA0002282880330000052

15. A compound which is

Figure FDA0002282880330000053

16. A compound which isOr a pharmaceutically acceptable salt or stereoisomer thereof.

17. A compound which is

Figure FDA0002282880330000055

18. A compound which isOr a pharmaceutically acceptable salt thereof.

19. A compound which isOr a pharmaceutically acceptable salt or stereoisomer thereof.

20. A compound which is

Figure FDA0002282880330000063

21. A compound which isOr a pharmaceutically acceptable salt thereof.

22. A compound which is

Figure FDA0002282880330000071

23. A compound which is

Figure FDA0002282880330000072

24. A compound which is

Figure FDA0002282880330000073

25. A compound which is

Figure FDA0002282880330000074

26. A compound which is

Figure FDA0002282880330000081

27. A pharmaceutical composition comprising a compound of any one of claims 1 to 26, or a pharmaceutically acceptable salt or stereoisomer thereof, and a pharmaceutically acceptable carrier, diluent or excipient.

28. A compound according to any one of claims 1 to 26, or a pharmaceutically acceptable salt or stereoisomer thereof, for use in therapy.

29. Use of a compound of any one of claims 1 to 26, or a pharmaceutically acceptable salt or stereoisomer thereof, in the treatment of an inflammatory disease.

30. Use of a compound of any one of claims 1 to 26, or a pharmaceutically acceptable salt or stereoisomer thereof, in the manufacture of a medicament for the treatment of an inflammatory disease.

31. A compound according to any one of claims 1 to 26, or a pharmaceutically acceptable salt or stereoisomer thereof, for use in the treatment of an inflammatory disease.

32. The compound of any one of claims 29 to 31, or a pharmaceutically acceptable salt or stereoisomer, or a use thereof, wherein the inflammatory disease is asthma.

33. A method of preventing, treating or lessening the severity of a disease or condition in a patient that responds to inhibition of Janus kinase activity, comprising administering to said patient a therapeutically effective amount of a compound of any one of claims 1 to 26, or a stereoisomer or pharmaceutically acceptable salt thereof.

34. The method of claim 33, wherein the disease or disorder is asthma.

35. The method of claim 33, wherein the Janus kinase is JAK 1.

36. The invention as hereinbefore described.

Technical Field

The present invention relates to Janus kinase, e.g., JAK1, inhibitor compounds, as well as compositions and methods of use comprising these compounds, including but not limited to, the diagnosis and treatment of patients suffering from a disorder responsive to JAK kinase inhibition.

Background

Cytokine pathways mediate many biological functions, including many aspects of inflammation and immunity. Janus kinases (JAKs), including JAK1, JAK2, JAK3 and TYK2, are cytoplasmic protein kinases associated with type I and type II cytokine receptors and regulate cytokine signal transduction. Cytokine binding to cognate receptors triggers activation of receptor-associated JAKs, leading to tyrosine phosphorylation of JAK-mediated, Signaling and Transcriptional Activation (STAT) proteins and ultimately transcriptional activation of specific genomes (Schindler et al, 2007, j.biol. chem.282: 20059-63). JAK1, JAK2 and TYK2 showed a broad pattern of gene expression, whereas JAK3 expression was restricted to leukocytes only. Cytokine receptors often act as heterodimers, and thus, there is generally more than one type of JAK kinase associated with the cytokine receptor complex. The specific JAKs associated with different cytokine receptor complexes have been identified in many cases by genetic studies and confirmed by other experimental evidence. Exemplary therapeutic benefits of JAK enzyme inhibition are discussed, for example, in international application No. WO 2013/014567.

JAK1 was originally identified in a novel kinase screen (Wilks a.f.,1989, proc.natl.acad.sci.u.s.a.86: 1603-. Genetic and biochemical studies have shown that JAK1 is functionally and physiologically related to type I interferons (e.g., IFN. alpha.), type II interferons (e.g., IFN. gamma.) and the IL-2 and IL-6 cytokine receptor complexes (Kisseleva et al, 2002, Gene 285: 1-24; Levy et al, 2005, Nat. Rev. mol. Cell biol.3: 651-. JAK1 knockout mice die perinatally due to defects in LIF receptor signaling (Kisseleva et al, 2002, Gene 285: 1-24; O' Shea et al, 2002, Cell,109(suppl.): S121-S131). Characterization of tissues derived from JAK1 knock-out mice demonstrates that this kinase plays an important role in the IFN, IL-10, IL-2/IL-4 and IL-6 pathways. The European Commission approved a humanized monoclonal antibody (Tocilizumab) targeting the IL-6 pathway for the treatment of moderate to severe rheumatoid arthritis (Scheinecker et al, 2009, nat. Rev. drug Discov.8: 273-.

CD 4T cells play an important role in the pathogenesis of asthma by producing TH2 cytokines in the lung, including IL-4, IL-9, and IL-13(Cohn et al, 2004, Annu. Rev. Immunol.22: 789-815). IL-4 and IL-13 induce increased mucus production, recruitment of eosinophils to the lung, and increased IgE production (Kasaian et al, 2008, biochem. Pharmacol.76(2): 147-. IL-9 causes mast cell activation, which exacerbates asthma symptoms (Kearley et al, 2011, am.J.Resp.Crit.Care Med.,183(7): 865-. IL-4R α chain activates JAK1 and binds to IL-4 or IL-13 when linked to the universal γ chain or IL-13R α 1 chain, respectively (Pernis et al, 2002, J.Clin.invest.109(10): 1279-. Universal gamma chains can also be combined with IL-9R α to bind IL-9, and IL-9R α also activates JAK1(Demoulin et al, 1996, mol. cell biol.16(9): 4710-4716). Although the universal gamma chain activates JAK3, JAK1 has been shown to be much stronger than JAK3, and JAK1 inhibition is sufficient to inactivate signaling through the universal gamma chain even in the presence of JAK3 activity (Haan et al, 2011, chem. biol.18(3): 314-. Inhibition of IL-4, IL-13 and IL-9 signaling by blocking the JAK/STAT signaling pathway may alleviate asthma symptoms in preclinical models of pneumonia (Mathew et al, 2001, J.Exp.Med.193(9): 1087-.

Biochemical and genetic studies have shown a relationship between JAK2 and the single-chain (e.g., EPO), IL-3 and interferon gamma cytokine receptor families (Kisseleva et al, 2002, Gene 285: 1-24; Levy et al, 2005, nat. Rev. mol. CellBiol.3: 651-. In agreement, JAK2 knockout mice die of anemia (O' Shea et al, 2002, Cell,109(suppl.): S121-S131). Kinase activating mutations in JAK2 (e.g., JAK 2V 617F) are associated with human myeloproliferative disorders.

JAK3 is only associated with the gamma universal cytokine receptor chain present in the IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21 cytokine receptor complexes. JAK3 is essential for the development and proliferation of lymphoid cells, and mutations in JAK3 lead to Severe Combined Immunodeficiency (SCID) (O' Shea et al, 2002, Cell,109(suppl.): S121-S131). Based on their role in regulating lymphocytes, JAK3 and JAK 3-mediated pathways have been targeted for immunosuppressive indications (e.g., transplant rejection and rheumatoid Arthritis) (Baslund et al, 2005, Arthritis & Rheumatism 52: 2686-.

TYK2 is associated with type I interferons (e.g., IFN. alpha.), IL-6, IL-10, IL-12, and the IL-23 cytokine receptor complex (Kisseleva et al, 2002, Gene 285: 1-24; Watford, W.T. & O' Shea, J.J.,2006, Immunity 25: 695-. In line with this, primary cells derived from TYK2 deficient humans are deficient in type I interferon, IL-6, IL-10, IL-12 and IL-23 signaling. Fully human monoclonal antibodies (Ustekinumab) targeting the p40 subunit common to IL-12 and IL-23 cytokines have recently been approved by the European Commission for use in the treatment of moderate to severe plaque psoriasis (Krueger et al, 2007, N.Engl. J.Med.356: 580-92; Reich et al, 2009, nat. Rev. drug Discov.8: 355-356). In addition, antibodies targeting both the IL-12 and IL-23 pathways have been tested clinically for treatment of Crohn (Crohn) disease (Mannon et al, 2004, N.Engl. J.Med.351: 2069-79).

Certain pyrazolopyrimidine compounds that have been reported to be useful as inhibitors of one or more Janus kinases are discussed in International patent application publication Nos. WO 2010/051549, WO 2011/003065, WO 2015/177326 and WO 2017/089390. It presents data for certain specific compounds showing inhibition of JAK1 as well as JAK2, JAK3 and/or TYK2 kinases.

There remains a need for additional compounds that are Janus kinase inhibitors. For example, there is a need for compounds that have useful potency as inhibitors of one or more Janus kinases (e.g., JAK1) and other pharmacological properties necessary to achieve useful therapeutic benefits. For example, there is a need for an effective compound that generally exhibits selectivity for one Janus kinase over other kinases (e.g., selectivity for JAK1 over other kinases, such as leucine-rich repeat kinase 2(LRRK 2)). There is also a need for effective compounds that exhibit greater selectivity for one Janus kinase over other Janus kinases (e.g., greater selectivity for JAK1 over other Janus kinases). In disorders responsive to JAK1 inhibition, kinases that exhibit selectivity for JAK1 may provide therapeutic benefits with fewer side effects. In addition, there is a need for effective JAK1 inhibitors with other properties (e.g., melting point, pK, solubility, etc.) that are essential for formulation and inhalation administration. Such compounds would be particularly useful in the treatment of disorders such as asthma.

There is a need in the art for additional or alternative treatments for conditions mediated by JAK kinases, such as those described previously.

Brief description of the invention

Provided herein are pyrazolopyrimidines that inhibit JAK kinases, such as selected from a compound of formula (I), a stereoisomer thereof, or a salt thereof, such as a pharmaceutically acceptable salt thereof. The JAK kinase may be JAK 1.

One embodiment provides a compound of formula (I) or a stereoisomer or salt (e.g., a pharmaceutically acceptable salt) thereof:

Figure BDA0002282880340000041

wherein:

R1is hydrogen or CH3

R2Is halogen, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl OR-ORaWherein R is2Optionally substituted by one or more groups independently selected from halogen, C1-C3Alkyl, cyano, hydroxy and oxo;

Rais C1-C6Alkyl, -phenyl-CORbRc-phenyl- (3-6-membered heterocyclyl) or 3-11-membered heterocyclyl, wherein R isaOptionally substituted by one or more groups independently selected from halogen, C1-C3Alkyl, cyano, hydroxy and oxo;

each RbAnd RcIndependently is hydrogen or CH3

R3Is hydrogen or NH2

R4Is hydrogen or CH3(ii) a And is

R5Is hydrogen or NH2

Also provided are pharmaceutical compositions comprising a JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent, or excipient.

Also provided is the use of a JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, in therapy, such as the treatment of an inflammatory disease (e.g., asthma). Also provided is the use of a JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of an inflammatory disease. Also provided are methods of preventing, treating, or lessening the severity of a disease or disorder in a patient responsive to inhibition of Janus kinase activity comprising administering to the patient a therapeutically effective amount of a JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof.

Certain compounds described herein or salts thereof (e.g., pharmaceutically acceptable salts thereof) have beneficial potency as inhibitors of one or more Janus kinases, such as JAK 1. Certain compounds or salts thereof (e.g., pharmaceutically acceptable salts thereof) also a) have a higher selectivity for one Janus kinase than other kinases, b) have a higher selectivity for JAK1 than other Janus kinases, and/or c) have other properties (e.g., melting point, pK, solubility, etc.) that are necessary for other formulations and inhalation administration. Certain compounds described herein or salts thereof (e.g., pharmaceutically acceptable salts thereof) may be particularly effective for treating conditions such as asthma.

Detailed Description

Definition of

"halogen" or "halo" means F, Cl, Br, or I. Furthermore, terms such as "haloalkyl" are intended to include monohaloalkyl and polyhaloalkyl, wherein one or more halogens replace one or more hydrogens in the alkyl.

The term "alkyl" means a saturated straight or branched chain monovalent hydrocarbon group, wherein the alkyl group may be optionally substituted. In one example, alkyl is 1 to 18 carbon atoms (C)1-C18). In other examples, alkyl is C0-C6、C0-C5、C0-C3、C1-C12、C1-C10、C1-C8、C1-C6、C1-C5、C1-C4Or C1-C3。C0Alkyl means a bond. Examples of alkyl groups include methyl (Me, -CH)3) Ethyl (Et-CH)2CH3) 1-propyl (n-Pr, n-propyl, -CH)2CH2CH3) 2-propyl (i-Pr, i-propyl, -CH (CH)3)2) 1-butyl (n-Bu, n-butyl, -CH)2CH2CH2CH3) 2-methyl-1-propyl (i-Bu, i-butyl, -CH)2CH(CH3)2) 2-butyl (s-Bu, s-butyl, -CH (CH)3)CH2CH3) 2-methyl-2-propyl (t-Bu, t-butyl, -C (CH)3)3) 1-pentyl (n-pentyl, -CH)2CH2CH2CH2CH3) 2-pentyl (-CH (CH)3)CH2CH2CH3) 3-pentyl (-CH (CH)2CH3)2) 2-methyl-2-butyl (-C (CH)3)2CH2CH3) 3-methyl-2-butyl (-CH (CH)3)CH(CH3)2) 3-methyl-1-butyl (-CH)2CH2CH(CH3)2) 2-methyl-1-butyl (-CH)2CH(CH3)CH2CH3) 1-hexyl (-CH)2CH2CH2CH2CH2CH3) 2-hexyl (-CH (CH)3)CH2CH2CH2CH3) 3-hexyl (-CH (CH)2CH3)(CH2CH2CH3) 2-methyl-2-pentyl (-C (CH))3)2CH2CH2CH3) 3-methyl-2-pentyl (-CH (CH)3)CH(CH3)CH2CH3) 4-methyl-2-pentyl (-CH (CH)3)CH2CH(CH3)2) 3-methyl-3-pentyl (-C (CH)3)(CH2CH3)2) 2-methyl-3-pentyl (-CH (CH)2CH3)CH(CH3)2)2, 3-dimethyl-2-butyl (-C (CH)3)2CH(CH3)2) 3, 3-dimethyl-2-butyl (-CH (CH)3)C(CH3)31-heptyl and 1-octyl. In some embodiments, substituents for "optionally substituted alkyl" include 1-4 of the following: F. cl, Br, I, OH, SH, CN, NH2、NHCH3、N(CH3)2、NO2、N3、C(O)CH3、COOH、CO2CH3Methyl, ethyl, propyl, isopropyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methylsulfonylamino, SO2Phenyl, piperidinyl, piperazinyl and pyrimidinyl, wherein the alkyl, phenyl and heterocyclic moieties thereof may be optionally substituted, such as with 1-4 examples of substituents selected from the same list.

The term "alkenyl" means a straight or branched chain monovalent hydrocarbon group having at least one site of unsaturation, i.e., a carbon-carbon double bond, wherein the alkenyl group may be optionally substituted and includes groups having "cis" and "trans" orientations, or alternatively "E" and "Z" orientations. In one example, alkenyl is 2 to 18 carbon atoms (C)2-C18). In other examples, alkenyl is C2-C12、C2-C10、C2-C8、C2-C6Or C2-C3. Examples include, but are not limited to, vinyl (ethenyl) or vinyl (vinyl) (-CH ═ CH2) Prop-1-enyl (-CH ═ CHCH)3) Prop-2-enyl (-CH)2CH=CH2) 2-methylpropan-1-enyl, but-2-enyl, but-3-enyl, but-1, 3-dienyl, 2-methylbut-1, 3-dienyl, hex-1-enyl, hex-2-enyl, hex-3-enyl, hex-4-enyl and hex-1, 3-dienyl. In some embodiments, substituents for "optionally substituted alkenyl" include 1-4 examples of the following: F. cl, Br, I, OH, SH, CN, NH2、NHCH3、N(CH3)2、NO2、N3、C(O)CH3、COOH、CO2CH3Methyl, ethyl, propyl, isopropyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methylsulfonylamino, SO2Phenyl, piperidinyl, piperazinyl and pyrimidinyl, wherein the alkyl, phenyl and heterocyclic moieties thereof may be optionally substituted, such as with 1-4 examples of substituents selected from the same list.

The term "alkynyl" means a straight or branched chain monovalent hydrocarbon group having at least one site of unsaturation, i.e., a carbon-carbon triple bond, wherein the alkynyl group may be optionally substituted. In one example, alkynyl is 2 to 18 carbon atoms (C)2-C18). In other examples, alkynyl is C2-C12、C2-C10、C2-C8、C2-C6Or C2-C3. Examples include, but are not limited to, ethynyl (-C ≡ CH), prop-1-ynyl (-C ≡ CCH)3) Prop-2-ynyl (propargyl, -CH)2C.ident.CH), but-1-ynyl, but-2-ynyl and but-3-ynyl. In some embodiments, substituents for "optionally substituted alkynyl" include 1-4 instances of the following: F. cl, Br, I, OH, SH, CN, NH2、NHCH3、N(CH3)2、NO2、N3、C(O)CH3、COOH、CO2CH3Methyl, ethyl, propyl, isopropyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methylsulfonylamino, trifluoromethyl, and mixtures thereof,SO、SO2Phenyl, piperidinyl, piperazinyl and pyrimidinyl, wherein the alkyl, phenyl and heterocyclic moieties thereof may be optionally substituted as exemplified by 1-4 substituents selected from the same list.

"alkylene" means a saturated branched or straight chain hydrocarbon radical having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of the parent alkane. In one example, the divalent alkylene group is 1 to 18 carbon atoms (C)1-C18). In other examples, the divalent alkylene is C0-C6、C0-C5、C0-C3、C1-C12、C1-C10、C1-C8、C1-C6、C1-C5、C1-C4Or C1-C3。C0Alkylene means a bond. Examples of alkylene groups include methylene (-CH)2-), 1-ethyl (-CH (CH)3) -, (1, 2-ethyl (-CH))2CH2-), 1-propyl (-CH (CH)2CH3) -), 2-propyl (-C (CH)3)2-), 1, 2-propyl (-CH (CH)3)CH2-), 1, 3-propyl (-CH)2CH2CH2-), 1-Dimethylethyl-1, 2-yl (-C (CH)3)2CH2-), 1, 4-butyl (-CH)2CH2CH2CH2-) and the like.

The term "heteroalkyl" means a straight or branched chain monovalent hydrocarbon radical consisting of the stated number of carbon atoms or up to 18 carbon atoms if not stated and 1 to 5 heteroatoms selected from O, N, Si and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. In some embodiments, the heteroatom is selected from O, N and S, where the nitrogen and sulfur atoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized. The heteroatom may be located at any internal position of the heteroalkyl group, including where the alkyl group is attached to the remainder of the molecule (e.g., -O-CH2-CH3). Examples include-CH2-CH2-O-CH3、-CH2-CH2-NH-CH3、-CH2-CH2-N(CH3)-CH3、-CH2-S-CH2-CH3、-S(O)-CH3、-CH2-CH2-S(O)2-CH3、-Si(CH3)3and-CH2-CH=N-OCH3. Up to two hetero atoms may be continuous, e.g. -CH2-NH-OCH3and-CH2-O-Si(CH3)3. Heteroalkyl groups may be optionally substituted. In some embodiments, the substituents of "optionally substituted heteroalkyl" include 1 to 4 examples of the following: F. cl, Br, I, OH, SH, CN, NH2、NHCH3、N(CH3)2、NO2、N3、C(O)CH3、COOH、CO2CH3Methyl, ethyl, propyl, isopropyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methylsulfonylamino, SO2Phenyl, piperidinyl, piperazinyl and pyrimidinyl, wherein the alkyl, phenyl and heterocyclic moieties thereof may be optionally substituted as exemplified by 1-4 substituents selected from the same list.

"amino" refers to a primary amine (i.e., -NH) that may be optionally substituted2) Secondary amines (i.e., -NRH), tertiary amines (i.e., -NRR), and quaternary amines (i.e., -N (+) RRR), wherein each R is the same or different and is selected from the group consisting of alkyl, cycloalkyl, aryl, and heterocyclyl, wherein alkyl, cycloalkyl, aryl, and heterocyclyl are defined herein. Specific secondary and tertiary amines are alkylamines, dialkylamines, arylamines, diarylamines, aralkylamines, and diarylalkylamines, wherein the alkyl and aryl moieties may be optionally substituted. Specific secondary and tertiary amines are methylamine, ethylamine, propylamine, isopropylamine, aniline, benzylamine dimethylamine, diethylamine, dipropylamine and diisopropylamine. In some embodiments, the R groups of the quaternary amine are each independently optionally substituted alkyl.

"aryl" means a carbocyclic aromatic group, whether fused to one or more groups, having the indicated number of carbon atoms or up to 14 carbon atoms if not indicated. One example includes aryl groups having 6 to 14 carbon atoms. Another example includesAryl groups having 6 to 10 carbon atoms. Examples of aryl groups include phenyl, naphthyl, biphenyl, phenanthryl, tetracenyl, 1,2,3, 4-tetrahydronaphthyl, 1H-indenyl, 2, 3-dihydro-1H-indenyl, and the like (see, e.g., Lang's Handbook of chemistry (Dean, J.A., ed.)13th ed.Table 7-2[1985]). A specific aryl group is phenyl. Substituted phenyl or substituted aryl refers to phenyl or aryl substituted with 1,2,3,4 or 5 substituents, for example 1-2, 1-3 or 1-4 substituents, as selected from the groups specified herein (see "optionally substituted" definitions), such as F, Cl, Br, I, OH, SH, CN, NH2、NHCH3、N(CH3)2、NO2、N3、C(O)CH3、COOH、CO2CH3Methyl, ethyl, propyl, isopropyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methylsulfonylamino, SO2Phenyl, piperidinyl, piperazinyl and pyrimidinyl, wherein the alkyl, phenyl and heterocyclic moieties thereof may be optionally substituted as exemplified by 1-4 substituents selected from the same list. Examples of the term "substituted phenyl group" include mono-or di (halo) phenyl groups such as 2-chlorophenyl, 2-bromophenyl, 4-chlorophenyl, 2, 6-dichlorophenyl, 2, 5-dichlorophenyl, 3, 4-dichlorophenyl, 3-chlorophenyl, 3-bromophenyl, 4-bromophenyl, 3, 4-dibromophenyl, 3-chloro-4-fluorophenyl, 2, 4-difluorophenyl and the like; mono-or di (hydroxy) phenyl, such as 4-hydroxyphenyl, 3-hydroxyphenyl, 2, 4-dihydroxyphenyl, protected hydroxy derivatives thereof, and the like; nitrophenyl, such as 3-or 4-nitrophenyl; cyanophenyl, such as 4-cyanophenyl; mono-or di (alkyl) phenyl such as 4-methylphenyl, 2, 4-dimethylphenyl, 2-methylphenyl, 4- (isopropyl) phenyl, 4-ethylphenyl, 3- (n-propyl) phenyl, and the like; mono-or di (alkoxy) phenyl such as 3, 4-dimethoxyphenyl, 3-methoxy-4-benzyloxyphenyl, 3-ethoxyphenyl, 4- (isopropoxy) phenyl, 4- (tert-butoxy) phenyl, 3-ethoxy-4-methoxyphenyl, etc.; 3-or 4-trifluoromethylphenyl; mono-or dicarboxyphenyl or (protected carboxy) phenyl, e.g. 4-carboxyphenyl, mono-or di (hydroxymethyl) phenylOr (protected hydroxymethyl) phenyl, such as 3- (protected hydroxymethyl) phenyl or 3, 4-bis (hydroxymethyl) phenyl; mono-or di (aminomethyl) phenyl or (protected aminomethyl) phenyl such as 2- (aminomethyl) phenyl or 2,4- (protected aminomethyl) phenyl; or mono-or di (N- (methylsulfonylamino)) phenyl, such as 3- (N-methylsulfonylamino)) phenyl. Likewise, the term "substituted phenyl" means a disubstituted phenyl group different in substituents, for example, 3-methyl-4-hydroxyphenyl, 3-chloro-4-hydroxyphenyl, 2-methoxy-4-bromophenyl, 4-ethyl-2-hydroxyphenyl, 3-hydroxy-4-nitrophenyl, 2-hydroxy-4-chlorophenyl, 2-chloro-5-difluoromethoxy and the like, and a trisubstituted phenyl group different in substituents, for example, 3-methoxy-4-benzyloxy-6-methylsulfonylamino, 3-methoxy-4-benzyloxy-6-phenylsulfonylamino, and a tetrasubstituted phenyl group different in substituents, for example, 3-methoxy-4-benzyloxy-5-methyl-6-phenylsulfonylamino A sulfonylamino group. In some embodiments, substituents for aryl groups, such as phenyl, include amides. For example, the aryl (e.g., phenyl) substituent may be- (CH)2)0-4CONR 'R ", wherein each R' and R" independently means the following groups including, for example, hydrogen; unsubstituted C1-C6An alkyl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R' substituted C1-C6An alkyl group; unsubstituted C1-C6A heteroalkyl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R' substituted C1-C6A heteroalkyl group; unsubstituted C6-C10An aryl group; c substituted by6-C10Aryl: halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy or NR' R "; an unsubstituted 3-11 membered heterocyclyl (e.g., a 5-6 membered heteroaryl group containing 1 to 4 heteroatoms selected from O, N and S, or a 4-11 membered heterocycloalkyl group containing 1 to 4 heteroatoms selected from O, N and S); and 3-11 membered heterocyclic group substituted with (e.g. containing 1 to 4 heteroatoms selected from O, N and S5-6 membered heteroaryl or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S): halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo, or NR' R "; or R 'and R' may combine with the nitrogen atom to form a 3-, 4-, 5-, 6-, or 7-membered ring, wherein the ring atoms are optionally substituted with N, O or S, and wherein the ring is optionally substituted with halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo, or NR 'R'.

"cycloalkyl" means a non-aromatic, saturated, or partially unsaturated cyclic hydrocarbon group, wherein the cycloalkyl group may be optionally independently substituted with one or more substituents described herein. In one example, cycloalkyl is 3 to 12 carbon atoms (C)3-C12). In other examples, cycloalkyl is C3-C8、C3-C10Or C5-C10. In other examples, cycloalkyl as a monocyclic ring is C3-C8、C3-C6Or C5-C6. In another example, cycloalkyl is C as bicyclic7-C12. In another example, cycloalkyl as a spiro ring system is C5-C12. Examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, perhydrocyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl and cyclododecyl. Exemplary arrangements of bicyclic cycloalkyl groups having 7 to 12 ring atoms include, but are not limited to, [4,4]、[4,5]、[5,5]、[5,6]Or [6,6 ]]A ring system. Exemplary bridged bicyclic cycloalkyls include, but are not limited to, bicyclo [ 2.2.1%]Heptane, bicyclo [2.2.2]Octane and bicyclo [3.2.2]Nonane. Examples of spirocycloalkyl include spiro [2.2]Pentane, spiro [2.3]Hexane, spiro [2.4 ]]Heptane, spiro [2.5 ]]Octane and spiro [4.5 ]]Decane. In some embodiments, the substituents of "optionally substituted cycloalkyl" include 1 to 4 examples selected from the group consisting of: F. cl, Br, I, OH, SH, CN, NH2、NHCH3、N(CH3)2、NO2、N3、C(O)CH3、COOH、CO2CH3Methyl, ethyl, propyl, isopropyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methylsulfonylamino, SO2Phenyl, piperidinyl, piperazinyl and pyrimidinyl, wherein the alkyl, aryl and heterocyclic moieties thereof may be optionally substituted, such as with 1 to 4 examples of substituents selected from the same list. In some embodiments, the substituents of the cycloalkyl group include amides. For example, the substituent of cycloalkyl may be- (CH)2)0-4CONR 'R ", wherein R' and R" each independently mean the following groups including, for example, hydrogen; unsubstituted C1-C6An alkyl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R' substituted C1-C6An alkyl group; unsubstituted C1-C6A heteroalkyl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R' substituted C1-C6A heteroalkyl group; unsubstituted C6-C10An aryl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy or NR 'R' substituted C6-C10An aryl group; an unsubstituted 3-11 membered heterocyclyl (e.g., a 5-6 membered heteroaryl group containing 1 to 4 heteroatoms selected from O, N and S or a 4-11 membered heterocycloalkyl group containing 1 to 4 heteroatoms selected from O, N and S); and 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl comprising 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl comprising 1 to 4 heteroatoms selected from O, N and S) substituted with: halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo, or NR' R "; or R 'and R' may combine with the nitrogen atom to form a 3-, 4-, 5-, 6-, or 7-membered ring, wherein the ring atom is optionally substituted with N, O or S, and wherein the ring is optionally substituted withGeneration: halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R'.

"heterocyclic group", "heterocyclic", "heterocyclyl" or "heterocyclic" are used interchangeably and mean any monocyclic, bicyclic, tricyclic or spirocyclic, saturated or unsaturated, aromatic (heteroaryl) or non-aromatic (e.g. heterocycloalkyl) ring system having from 3 to 20 ring atoms (e.g. 3-10 ring atoms) wherein the ring atoms are carbon and at least one atom in the ring or ring system is a heteroatom selected from nitrogen, sulfur or oxygen. If any ring atom in the ring system is a heteroatom, the system is heterocyclic, regardless of where the ring system is attached to the remainder of the molecule. In one example, heterocyclyl includes 3-11 ring atoms ("members") and includes monocyclic, bicyclic, tricyclic, and spiro ring systems in which the ring atoms are carbon and at least one atom in the ring or ring system is a heteroatom selected from nitrogen, sulfur, or oxygen. In one example, heterocyclyl includes 1 to 4 heteroatoms. In one example, heterocyclyl includes 1 to 3 heteroatoms. In another example, heterocyclyl includes 3-to 7-membered monocyclic rings having 1-2, 1-3, or 1-4 heteroatoms selected from nitrogen, sulfur, or oxygen. In another example, heterocyclyl includes 4-to 6-membered monocyclic rings having 1-2, 1-3, or 1-4 heteroatoms selected from nitrogen, sulfur, or oxygen. In another example, heterocyclyl includes 3-membered monocyclic rings. In another example, heterocyclyl includes a 4-membered monocyclic ring. In another example, heterocyclyl includes 5-6 membered monocyclic, e.g., 5-6 membered heteroaryl. In another example, heterocyclyl includes 3-11 membered heterocycloalkyl, such as 4-11 membered heterocycloalkyl. In some embodiments, the heterocycloalkyl group includes at least one nitrogen. In one example, heterocyclyl includes 0-3 double bonds. Any nitrogen or sulfur heteroatom may optionally be oxidized (e.g., NO, SO)2) And any nitrogen heteroatom may optionally be quaternized (e.g., [ NR ]4]+Cl-、[NR4]+OH-). Examples of heterocycles are oxiranyl, aziridinyl, thiepinyl, azetidinyl, oxetanyl, thiepinyl, 1, 2-dithiocyclobutyl, 1, 3-dithioheterocycloButyl, pyrrolidinyl, dihydro-1H-pyrrolyl, dihydrofuranyl, tetrahydrofuryl, dihydrothienyl, tetrahydrothienyl, imidazolidinyl, piperidinyl, piperazinyl, isoquinolinyl, tetrahydroisoquinolinyl, morpholinyl, thiomorpholinyl, 1-dioxo-thiomorpholinyl, dihydropyranyl, tetrahydropyranyl, hexahydrothiopyranyl, hexahydropyrimidyl, oxazinanyl (oxazinanyl), thiazinanyl, thiohexoxanyl, homopiperazinyl, homopiperidinyl, azepinyl, oxepinyl, thiepinyl, oxacycloheptyl, oxazepinyl, and oxazepinyl

Figure BDA0002282880340000101

A group, an oxazepinyl group, a diazepanyl group, a1, 4-diazepanyl group, a diazepine groupRadical, sulfur nitrogen hetero

Figure BDA0002282880340000103

A group, a thiazepanyl group, a tetrahydrothiopyranyl group, an oxazolidinyl group, a thiazolidinyl group, an isothiazolidinyl group, a1, 1-dioxoisothiazolidinonyl group, an oxazolidinonyl group, an imidazolidinonyl group, a4, 5,6, 7-tetrahydro [2H ] group]Indazolyl, tetrahydrobenzimidazolyl, 4,5,6, 7-tetrahydrobenzo [ d ]]Imidazolyl, 1, 6-dihydroimidazo [4,5-d]Pyrrolo [2,3-b]Pyridyl, thiazinyl, oxazinyl, thiadiazinyl, oxadiazinyl, dithiazinyl, dioxazinyl, oxathiazinyl, thiatriazinyl, oxatriazinyl (oxatrazinyl), dithiadiazinyl, imidazolinyl, dihydropyrimidyl, tetrahydropyrimidinyl, 1-pyrrolinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, thiopyranyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1, 3-dihydroheterocyclopentyl, pyrazolinyl, pyrazolidinyl, dithianyl, dithiopentyl (dithiolanyl), pyrimidonyl, pyrimidindionyl, pyrimidine-2, 4-diketo, piperazine, piperazinedionyl, pyrazolidinylimidazolinyl, 3-azabicyclo [ 3.1.0.3]Hexyl, 3, 6-diazabicyclo [3.1.1]Heptyl, 6-azabicyclo [3.1.1]Heptyl, 3-azabicyclo [3.1.1]Heptyl, 3-azabicyclo [4.1.0]Heptyl, azabicyclo [2.2.2]Hexyl radical,2-azabicyclo [3.2.1]Octyl, 8-azabicyclo [3.2.1]Octyl, 2-azabicyclo [2.2.2]Octyl, 8-azabicyclo [2.2.2]Octyl, 7-oxabicyclo [2.2.1]Heptane, azaspiro [3.5 ]]Nonyl, azaspiro [2.5 ]]Octyl, azaspiro [4.5 ]]Decyl, 1-azaspiro [4.5 ]]Decan-2-onyl, azaspiro [5.5 ]]Undecyl, tetrahydroindolyl, octahydroindolyl, tetrahydroisoindolyl, tetrahydroindazolyl, 1-dioxohexahydrothiopyranyl. Examples of 5-membered heterocyclic rings containing a sulfur or oxygen atom and 1 to 3 nitrogen atoms are thiazolyl, including thiazol-2-yl and thiazol-2-yl N-oxides; thiadiazolyl, including 1,3, 4-thiadiazol-5-yl and 1,2, 4-thiadiazol-5-yl, oxazolyl, e.g., oxazol-2-yl, and oxadiazolyl, e.g., 1,3, 4-oxadiazol-5-yl and 1,2, 4-oxadiazol-5-yl. Examples of the 5-membered heterocyclic ring having 2 to 4 nitrogen atoms include imidazolyl such as imidazol-2-yl; triazolyl such as 1,3, 4-triazol-5-yl; 1,2, 3-triazol-5-yl, 1,2, 4-triazol-5-yl, and tetrazolyl such as 1H-tetrazol-5-yl. Examples of benzo-fused 5-membered heterocycles are benzoxazol-2-yl, benzothiazol-2-yl and benzimidazol-2-yl. Examples of 6-membered heterocycles contain 1 to 3 nitrogen atoms and optionally sulphur or oxygen atoms, for example pyridyl, such as pyridin-2-yl, pyridin-3-yl and pyridin-4-yl; pyrimidinyl, such as pyrimidin-2-yl and pyrimidin-4-yl; triazinyl groups, such as 1,3, 4-triazin-2-yl and 1,3, 5-triazin-4-yl; pyridazinyl, especially pyridazin-3-yl and pyrazinyl. Examples of other heterocyclic groups are pyridine N-oxide and pyridazine N-oxide and pyridyl, pyrimidin-2-yl, pyrimidin-4-yl, pyridazinyl and 1,3, 4-triazin-2-yl. The heterocyclic ring may be optionally substituted. For example, substituents of "optionally substituted heterocycle" include 1 to 4 of the following examples: F. cl, Br, I, OH, SH, CN, NH2、NHCH3、N(CH3)2、NO2、N3、C(O)CH3、COOH、CO2CH3Methyl, ethyl, propyl, isopropyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methylsulfonylamino, SO2Phenyl, piperidinyl, piperazinyl and pyrimidinyl wherein the alkyl, aryl and heterocyclic moieties thereof may be optionally substituted, e.g. by 1 to 4 substituents selected from the same listExample substitutions. In some embodiments, substituents for heterocyclyl groups such as heteroaryl or heterocycloalkyl groups include amides. For example, a heterocyclic (e.g., heteroaryl or heterocycloalkyl) substituent may be- (CH)2)0-4CONR 'R ", wherein each R' and R" is independently meant to include groups such as hydrogen; unsubstituted C1-C6An alkyl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R' substituted C1-C6An alkyl group; unsubstituted C1-C6A heteroalkyl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R' substituted C1-C6A heteroalkyl group; unsubstituted C6-C10An aryl group; c substituted by6-C10Aryl: halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy or NR' R "; an unsubstituted 3-11 membered heterocyclyl (e.g., a 5-6 membered heteroaryl group containing 1 to 4 heteroatoms selected from O, N and S or a 4-11 membered heterocycloalkyl group containing 1 to 4 heteroatoms selected from O, N and S); and 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl comprising 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl comprising 1 to 4 heteroatoms selected from O, N and S) substituted with: halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo, or NR' R "; or R 'and R' may combine with the nitrogen atom to form a 3-, 4-, 5-, 6-, or 7-membered ring, wherein the ring atom is optionally substituted with N, O or S and wherein the ring is optionally substituted with: halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R'.

"heteroaryl" means any mono-, bi-, or tricyclic ring system in which at least one ring is a 5-or 6-membered aromatic ring containing 1 to 4 heteroatoms selected from nitrogen, oxygen, and sulfur, and in one example embodiment, at least one heteroatom is nitrogen. See, e.g., Lang's Handbook of Chemistry(Dean,J.A.,ed.)13thed. TABLE 7-2[1985 ]].. This definition includes any bicyclic group in which any of the above heteroaryl rings is fused to an aromatic ring, wherein the aromatic or heteroaryl ring is attached to the remainder of the molecule. In one embodiment, heteroaryl includes a 5-6 membered monocyclic aromatic group in which one or more ring atoms is nitrogen, sulfur, or oxygen. Examples of heteroaryl groups include thienyl, furyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, oxadiazolyl, tetrazolyl, thiatriazolyl, oxatriazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, tetrazinyl, tetrazolo [1,5-b ] and the like]Pyridazinyl, imidazo [1,2-a ]]Pyrimidinyl and purinyl groups, and benzo-fused derivatives, such as benzoxazolyl, benzofuranyl, benzothiazolyl, benzothiadiazolyl, benzotriazolyl, benzimidazolyl and indolyl groups. Heteroaryl groups may be optionally substituted. In some embodiments, the substituents of "optionally substituted heteroaryl" include 1 to 4 examples selected from the group consisting of: F. cl, Br, I, OH, SH, CN, NH2、NHCH3、N(CH3)2、NO2、N3、C(O)CH3、COOH、CO2CH3Methyl, ethyl, propyl, isopropyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, trifluoromethyl, difluoromethyl, sulfonylamino, methylsulfonylamino, SO2Phenyl, piperidinyl, piperazinyl and pyrimidinyl, wherein the alkyl, phenyl and heterocyclic moieties thereof may be optionally substituted, such as with 1 to 4 examples of substituents selected from the same list. In some embodiments, the substituent of the heteroaryl group comprises an amide. For example, the heteroaryl substituent may be- (CH)2)0-4CONR 'R ", wherein each R' and R" independently means the following groups including, for example, hydrogen; unsubstituted C1-C6An alkyl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R' substituted C1-C6An alkyl group; unsubstituted C1-C6A heteroalkyl group; by halogen, OH, CN, unsubstitutedC of (A)1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R' substituted C1-C6A heteroalkyl group; unsubstituted C6-C10An aryl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy or NR 'R' substituted C6-C10An aryl group; an unsubstituted 3-11 membered heterocyclyl (e.g., a 5-6 membered heteroaryl group containing 1 to 4 heteroatoms selected from O, N and S or a 4-11 membered heterocycloalkyl group containing 1 to 4 heteroatoms selected from O, N and S); and 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl comprising 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl comprising 1 to 4 heteroatoms selected from O, N and S) substituted with: halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo, or NR' R "; or R 'and R' may combine with the nitrogen atom to form a 3-, 4-, 5-, 6-, or 7-membered ring, wherein the ring atom is optionally substituted with N, O or S, and wherein the ring is optionally substituted with: halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R'.

In particular embodiments, the heterocyclyl group is attached at a carbon atom of the heterocyclyl group. For example, carbon-bonded heterocyclyl groups include the following bonding arrangements: 2,3,4, 5 or 6 positions of the pyridine ring, 3,4, 5 or 6 positions of the pyridazine ring, 2,4, 5 or 6 positions of the pyrimidine ring, 2,3, 5 or 6 positions of the pyrazine ring; 2,3,4 or 5 positions of furan, tetrahydrofuran, thiafuran (thiofuran), thiophene, pyrrole or tetrahydropyrrole rings; 2,4 or 5 position of oxazole, imidazole or thiazole ring; the 3,4 or 5 position of the isoxazole, pyrazole or isothiazole ring; the 2 or 3 position of the aziridine ring, the 2,3 or 4 position of the azetidine, the 2,3,4, 5,6,7 or 8 position of the quinoline ring or the 1,3,4, 5,6,7 or 8 position of the isoquinoline ring.

In certain embodiments, heterocyclyl is N-linked. For example, nitrogen-bonded heterocyclyl or heteroaryl groups include the following bonding arrangements: aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, 1-position of 1H-indazole; 2-position of isoindoline or isoindoline; the 4-position of morpholine, and the 9-position of carbazole or beta-carboline.

The term "alkoxy" means a straight OR branched chain monovalent hydrocarbon radical represented by the formula-OR, wherein R is alkyl, as defined herein. Alkoxy groups include methoxy, ethoxy, propoxy, isopropoxy, mono-, di-and tri-fluoromethoxy and cyclopropoxy.

"acyl" means a carbonyl-containing substituent represented by the formula-c (o) -R, wherein R is hydrogen, alkyl, cycloalkyl, aryl, or heterocyclyl, wherein alkyl, cycloalkyl, aryl, and heterocyclyl are defined herein. Acyl groups include alkanoyl (e.g., acetyl), aroyl (e.g., benzoyl), and heteroaroyl (e.g., pyridinoyl).

"optionally substituted" means, unless otherwise indicated, that a group may be unsubstituted or substituted with one or more substituents (e.g., 0, 1,2,3,4, or 5 or more, or any range derivable therein) set forth for that group, wherein the substituents may be the same or different. In one embodiment, the optionally substituted group has 1 substituent. In another embodiment, the optionally substituted group has 2 substituents. In another embodiment, the optionally substituted group has 3 substituents. In another embodiment, the optionally substituted group has 4 substituents. In another embodiment, the optionally substituted group has 5 substituents.

Alkyl (e.g., alkoxy) alone or as part of another substituent, and optional substituents for alkylene, alkenyl, alkynyl, heteroalkyl, heterocycloalkyl, and cycloalkyl, also alone or as part of another substituent, may be various groups as described herein, and selected from the group consisting of: halogen; an oxo group; CN; NO; n is a radical of3(ii) a -OR'; perfluoro-C1-C4An alkoxy group; unsubstituted C3-C7A cycloalkyl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstitutedC1-C6Alkoxy, oxo or NR 'R' substituted C3-C7A cycloalkyl group; unsubstituted C6-C10Aryl (e.g., phenyl); by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy or NR 'R' substituted C6-C10An aryl group; an unsubstituted 3-11 membered heterocyclyl (e.g., a 5-6 membered heteroaryl group containing 1 to 4 heteroatoms selected from O, N and S or a 4-11 membered heterocycloalkyl group containing 1 to 4 heteroatoms selected from O, N and S); a 3-11 membered heterocyclyl (e.g., a 5-6 membered heteroaryl comprising 1 to 4 heteroatoms selected from O, N and S or a 4-11 membered heterocycloalkyl comprising 1 to 4 heteroatoms selected from O, N and S) substituted with: halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo, or NR' R "; -NR' R "; -SR'; -SiR 'R "R'"; -OC (O) R'; -C (O) R'; -CO2R';-CONR'R”;-OC(O)NR'R”;-NR”C(O)R';-NR”'C(O)NR'R”;-NR”C(O)2R';-S(O)2R';-S(O)2NR'R”;-NR'S(O)2R”;-NR”'S(O)2NR' R "; an amidino group; guanidino; - (CH)2)1-4-OR';-(CH2)1-4-NR'R”;-(CH2)1-4-SR';-(CH2)1-4-SiR'R”R”';-(CH2)1-4-OC(O)R';-(CH2)1-4-C(O)R';-(CH2)1-4-CO2R'; and- (CH)2)1-4CONR ' R ", or combinations thereof, in an amount of from 0 to (2m ' +1), wherein m ' is the total number of carbon atoms in the group. Each R ', R "and R'" independently means a group including, for example, hydrogen; unsubstituted C1-C6An alkyl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R' substituted C1-C6An alkyl group; unsubstituted C1-C6A heteroalkyl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R' substituted C1-C6A heteroalkyl group; unsubstituted C6-C10An aryl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy or NR 'R' substituted C6-C10An aryl group; an unsubstituted 3-11 membered heterocyclyl (e.g., a 5-6 membered heteroaryl group containing 1 to 4 heteroatoms selected from O, N and S or a 4-11 membered heterocycloalkyl group containing 1 to 4 heteroatoms selected from O, N and S); and 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl comprising 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl comprising 1 to 4 heteroatoms selected from O, N and S) substituted with: halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R'. When R 'and R' are attached to the same nitrogen atom, they may combine with the nitrogen atom to form a 3-, 4-, 5-, 6-, or 7-membered ring, wherein the ring atom is optionally substituted with N, O or S and wherein the ring is optionally substituted with halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo, or NR 'R'. For example, -NR' R "is intended to include 1-pyrrolidinyl and 4-morpholinyl.

Similarly, the optional substituents for aryl and heteroaryl groups are widely varied. In some embodiments, the substituents for aryl and heteroaryl are selected from halogen; CN; NO; n is a radical of3(ii) a -OR'; perfluoro-C1-C4An alkoxy group; unsubstituted C3-C7A cycloalkyl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R' substituted C3-C7A cycloalkyl group; unsubstituted C6-C10Aryl (e.g., phenyl); by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy or NR 'R' substituted C6-C10An aryl group; an unsubstituted 3-11 membered heterocyclyl (e.g., a 5-6 membered heteroaryl group containing 1 to 4 heteroatoms selected from O, N and S or a 4-11 membered heterocycloalkyl group containing 1 to 4 heteroatoms selected from O, N and S); 3-11 membered heterocyclic group substituted with the following group (examples)Such as a 5-6 membered heteroaryl group containing 1 to 4 heteroatoms selected from O, N and S or a 4-11 membered heterocycloalkyl group containing 1 to 4 heteroatoms selected from O, N and S): halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo, or NR' R "; -NR' R "; -SR'; -SiR 'R "R'"; -OC (O) R'; -C (O) R'; -CO2R';-CONR'R”;-OC(O)NR'R”;-NR”C(O)R';-NR”'C(O)NR'R”;-NR”C(O)2R';-S(O)2R';-S(O)2NR'R”;-NR'S(O)2R”;-NR”'S(O)2NR' R "; an amidino group; guanidino; - (CH)2)1-4-OR';-(CH2)1-4-NR'R”;-(CH2)1-4-SR';-(CH2)1-4-SiR'R”R”';-(CH2)1-4-OC(O)R';-(CH2)1-4-C(O)R';-(CH2)1-4-CO2R'; and- (CH)2)1-4CONR ' R ", or combinations thereof, in an amount of from 0 to (2m ' +1), wherein m ' is the total number of carbon atoms in the group. Each R ', R "and R'" independently means a group including, for example, hydrogen; unsubstituted C1-C6An alkyl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R' substituted C1-C6An alkyl group; unsubstituted C1-C6A heteroalkyl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R' substituted C1-C6A heteroalkyl group; unsubstituted C6-C10An aryl group; by halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy or NR 'R' substituted C6-C10An aryl group; an unsubstituted 3-11 membered heterocyclyl (e.g., a 5-6 membered heteroaryl group containing 1 to 4 heteroatoms selected from O, N and S or a 4-11 membered heterocycloalkyl group containing 1 to 4 heteroatoms selected from O, N and S); and 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl or bag containing 1 to 4 heteroatoms selected from O, N and S) substituted with4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S): halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo or NR 'R'. When R 'and R' are attached to the same nitrogen atom, they may combine with the nitrogen atom to form a 3-, 4-, 5-, 6-, or 7-membered ring, wherein the ring atom is optionally substituted with N, O or S, and wherein the ring is optionally substituted with halogen, OH, CN, unsubstituted C1-C6Alkyl, unsubstituted C1-C6Alkoxy, oxo, or NR 'R'. For example, -NR' R "is intended to include 1-pyrrolidinyl and 4-morpholinyl.

The term "oxo" means O or (═ O)2

As used herein, a wavy line intersecting a bond in a chemical structure

Figure BDA0002282880340000151

Denotes the point in the chemical structure at which the atom attached to the wavy bond is attached to the remainder of the molecule or to the remainder of the fragment of the molecule. In some embodiments, arrows and asterisks are used in the manner of wavy lines to indicate points of attachment.

In certain embodiments, divalent groups generally do not depict a specific bonding configuration. It should be understood that, unless otherwise indicated, the generic description is intended to include both bonding configurations. For example, in the radical R1–R2–R3In case of the group R2Is described as-CH2C (O) -, it is to be understood that this group may be referred to as R unless otherwise indicated1–CH2C(O)–R3And R1–C(O)CH2–R3And (4) bonding.

The terms "compound of the invention" and the like, unless otherwise indicated, include compounds of formula (I) herein such as compounds 1-18, sometimes referred to as JAK inhibitors, including stereoisomers (including atropisomers), geometric isomers, tautomers, solvates, metabolites, isotopes, salts (e.g., pharmaceutically acceptable salts) and prodrugs thereof. In some embodiments, solvates, metabolites, isotopes or prodrugs, and any combination thereof are excluded.

The phrase "pharmaceutically acceptable" means molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered in suitable amounts to an animal such as a human.

The compounds of the invention may be in the form of a salt, such as a pharmaceutically acceptable salt. "pharmaceutically acceptable salts" include acid addition salts and base addition salts. By "pharmaceutically acceptable acid addition salt" is meant a salt which retains the biological effectiveness and properties of the free base and which is not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric, hydrobromic, sulfuric, nitric, carbonic, phosphoric and the like, which may be selected from aliphatic, alicyclic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic organic acids such as formic, acetic, propionic, glycolic, gluconic, lactic, pyruvic, oxalic, malic, maleic, malonic, succinic, fumaric, tartaric, citric, aspartic, ascorbic, glutamic, anthranilic, benzoic, cinnamic, mandelic, pamoic, phenylacetic, methanesulfonic, ethanesulfonic, benzenesulfonic, p-toluenesulfonic, salicylic and the like.

"pharmaceutically acceptable base addition salts" include those derived from inorganic bases such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Specific base addition salts are ammonium, potassium, sodium, calcium and magnesium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include the following salts: primary, secondary and tertiary amines, substituted ammonium, including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-diethylaminoethanol, tromethamine, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine (theobromamine), purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins, and the like. Specific organic non-toxic bases include isopropylamine, diethylamine, ethanolamine, tromethamine, dicyclohexylamine, choline, and caffeine.

In some embodiments, the salt is selected from the group consisting of hydrochloride, hydrobromide, trifluoroacetate, sulfate, phosphate, acetate, fumarate, maleate, tartrate, lactate, citrate, pyruvate, succinate, oxalate, methanesulfonate, p-toluenesulfonate, bisulfate, benzenesulfonate, ethanesulfonate, malonate, xinafoate (xinafoate), ascorbate, oleate, nicotinate, saccharinate, adipate, formate, glycolate, palmitate, L-lactate, D-lactate, aspartate, malate, L-tartrate, D-tartrate, stearate, furoate (e.g., 2-furoate or 3-furoate), naphthalenedisulfonate (naphthalene-1, 5-disulfonate or naphthalene-1- (sulfonic) -5-sulfonate), Ethanedisulfonate (ethane-1, 2-disulfonate or ethane-1- (sulfonic acid) -2-sulfonate), isethionate (2-isethionate), 2,4, 6-trimethylbenzenesulfonate (2-mesitylenesulphonate), 2-naphthalenesulfonate (2-naphthalenesulfonate), 2, 5-dichlorobenzenesulfonate, D-mandelate, L-mandelate, cinnamate, benzoate, adipate, ethanesulfonate, malonate, mesitylate (2,4, 6-trimethylbenzenesulfonate), naphthalenesulfonate (2-naphthalenesulfonate), camphorsulfonate (camphorate) (camphor-10-sulfonate, e.g., (1S) - (+) -10-camphorsulfonate), glutamate, glutarate, hippurate (2- (benzoylamino) acetate), Orotate (orotate), xylate (p-xylene-2-sulfonate) and pamoate (2,2 ' -dihydroxy-1, 1 ' -dinaphthylmethane-3, 3 ' -dicarboxylate).

"sterile" preparations are sterile or free of all living microorganisms and spores thereof.

"stereoisomers" means compounds having the same chemical composition but differing in the arrangement of atoms or groups in space. Stereoisomers include diastereomers, enantiomers, conformers, and the like. .

"chiral" means a molecule having the property that its mirror image counterparts are not superimposable, while the term "achiral" means a molecule whose mirror image counterparts are superimposable.

"diastereomer" means a stereoisomer having two or more chiral centers and whose molecules are not mirror images of one another. Diastereomers have different physical properties, such as melting points, boiling points, spectral characteristics, or biological activities. Mixtures of diastereomers may be separated under high resolution analytical methods such as electrophoresis and chromatography such as HPLC.

"enantiomer" means two stereoisomers of a compound that are non-overlapping mirror images of each other.

The stereochemical definitions and conventions used herein generally follow the general definitions of S.P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S., "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., New York, 1994. Many organic compounds exist in an optically active form, i.e., they have the ability to rotate a plane of plane polarized light. In describing optically active compounds, the prefixes D and L or R and S are used to denote the absolute configuration of a molecule with respect to its chiral center. The prefixes d and l or (+) and (-) are used to indicate rotation of the compound to plane polarized light, with (-) or l indicating that the compound is left-handed. Compounds prefixed with (+) or d are dextrorotatory. For a given chemical structure, unless mirror images of each other, the stereoisomers are identical. A particular stereoisomer may also be referred to as an enantiomer, and mixtures of such isomers are often referred to as enantiomeric mixtures. A 50:50 enantiomeric mixture is referred to as a racemic mixture or racemate, which may occur when there is no stereoselectivity or stereospecificity in the chemical reaction or process. The terms "racemic mixture" and "racemate" mean an equimolar mixture of two enantiomers, free of optical activity.

The term "tautomer" or "tautomeric form" means structural isomers of different energies that may be interchanged via a low energy barrier. For example, proton tautomers (also referred to as protic tautomers) include proton transfer interconversions, such as keto-enol and imine-enamine isomers. Valence tautomers include interconversions resulting from recombination of certain bonded electrons.

Certain compounds of the present invention may exist in unsolvated forms as well as solvated forms, including hydrated forms. By "solvate" is meant the association or complexation of one or more solvent molecules with a compound of the invention. Examples of the solvate-forming solvent include water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine. Certain compounds of the present invention may exist in polycrystalline or amorphous form. In general, all physical forms are encompassed within the scope of the present invention. The term "hydrate" means a complex in which the solvent molecule is water.

By "metabolite" is meant the product of a particular compound or salt thereof produced via in vivo metabolism. Such products may, for example, result from oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, etc., of the administered compound.

Metabolite products are typically identified as follows: preparation of radiolabeled isotopes of the compounds of the invention (e.g.14C or3H) Administered to an animal such as rat, mouse, guinea pig, monkey or human at a detectable dose (e.g., greater than about 0.5mg/kg) for a time sufficient for metabolism to occur (typically about 30 seconds to 30 hours) and the conversion products to be isolated from urine, blood or other biological samples. These products are easily separated by being labeled (the remaining products are separated by using antibodies capable of binding to epitopes that survive in the metabolite). Metabolite structure is determined in a conventional manner, e.g. MS, LC/MS or NMR analysis. Typically, metabolite analysis is performed in the same manner as conventional pharmacokinetic studies well known to those skilled in the art. Metabolite products, so long as they are not otherwise found in vivo, can be used in diagnostic assays for therapeutic dosages of the compounds of the present invention.

A "subject," "individual," or "patient" is a vertebrate. In certain embodiments, the vertebrate is a mammal. Mammals include, but are not limited to, farm animals (e.g., cattle), sport animals, pets (e.g., guinea pigs, cats, dogs, rabbits, and horses), primates, mice, and rats. In certain embodiments, the mammal is a human. In embodiments comprising administering to a patient a JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, the patient may have a therapeutic need.

The term "Janus kinase" means JAK1, JAK2, JAK3 and TYK2 protein kinases. In some embodiments, the Janus kinase may be further defined as one of JAK1, JAK2, JAK3, or TYK 2. In any embodiment, any one of JAK1, JAK2, JAK3, and TYK2 can be specifically excluded as a Janus kinase. In some embodiments, the Janus kinase is JAK 1. In some embodiments, the Janus kinase is a combination of JAK1 and JAK 2.

The terms "inhibit" and "reduce," or any variation of these terms, include any measurable reduction or complete inhibition to achieve a desired result. For example, it may be about, up to about, or at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or more, or any range derivable therein, that the activity (e.g., JAK1 activity) is reduced as compared to normal.

In some embodiments, a compound described herein or a salt thereof (e.g., a pharmaceutically acceptable salt thereof) has a selective effect in inhibiting JAK1 over JAK3 and TYK 2. In some embodiments, the compound or salt thereof (e.g., a pharmaceutically acceptable salt thereof) has a selective inhibition of JAK1 relative to JAK2, JAK3 or TYK2 or any combination of JAK2, JAK3 or TYK 2. In some embodiments, the compound or salt thereof (e.g., a pharmaceutically acceptable salt thereof) has a selective effect in inhibiting JAK1 and JAK2 relative to JAK3 and TYK 2. In some embodiments, the compound or salt thereof (e.g., a pharmaceutically acceptable salt thereof) has a selective inhibition effect on JAK1 relative to JAK 3. By "selectively inhibit" is meant that the compound or salt thereof (e.g., a pharmaceutically acceptable salt thereof) is an inhibitor that has at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% or more, or any range derivable therein, better activity against a particular Janus kinase (e.g., JAK1) than against another particular Janus kinase (e.g., JAK3), or at least 2-, 3-, 4-, 5-, 10-, 25-, 50-, 100-, 250-, or 500-fold better activity against a particular Janus kinase (e.g., JAK1) than against another particular Janus kinase (e.g., JAK 3).

By "therapeutically effective amount" is meant an amount of a compound of the present invention, or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), that (i) treats or prevents a particular disease, condition, or disorder, or (ii) attenuates, alleviates, or eliminates one or more symptoms of a particular disease, condition, or disorder, and optionally (iii) prevents or delays the onset of one or more symptoms of a particular disease, condition, or disorder described herein. In some embodiments, a therapeutically effective amount is an amount sufficient to reduce or alleviate symptoms of an autoimmune or inflammatory disease (e.g., asthma). In some embodiments, a therapeutically effective amount is an amount of a chemical entity described herein sufficient to significantly reduce B-cell activity or number. For cancer, a therapeutically effective amount of the drug may reduce the number of cancer cells; reducing the size of the tumor; inhibit (i.e., slow to some extent, preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent, preferably stop) tumor metastasis; inhibit tumor growth to some extent; or to alleviate one or more symptoms associated with cancer to some extent. To the extent that the drug can prevent growth or kill existing cancer cells, it may be cytostatic or cytotoxic. For cancer treatment, efficacy can be measured, for example, by assessing time to disease progression (TTP) or determining Response Rate (RR).

"treatment" (and variants such as "treatment" or "treating") means clinical intervention in an attempt to alter the natural course of the individual or cell being treated, and may be used prophylactically or during clinical pathology. Desired effects of treatment include preventing the occurrence or recurrence of disease, alleviating symptoms, reducing any direct or indirect pathological consequences of the disease, stabilizing (i.e., not worsening) the disease state, reducing the rate of disease progression, alleviating or ameliorating the disease state, prolonging survival as compared to expected survival without treatment, and ameliorating or improving prognosis. In some embodiments, a compound of the invention or a salt thereof (e.g., a pharmaceutically acceptable salt thereof) is used to delay the onset of, or slow the progression of, a disease or disorder. Those in need of treatment include those already with the condition or disorder, as well as those predisposed to having the condition or disorder (e.g., by genetic variation), or those in need of prevention of the condition or disorder.

By "inflammatory disorder" is meant any disease, disorder or syndrome in which an excessive or unregulated inflammatory response results in excessive inflammatory symptoms, host tissue damage or loss of tissue function. By "inflammatory disorder" is also meant a pathological condition mediated by leukocyte influx or neutrophil chemotaxis.

By "inflammation" is meant a local protective response caused by tissue damage or destruction that serves to destroy, dilute, or separate (sequester) the injurious agent from the injured tissue. Inflammation is particularly strongly associated with leukocyte influx or neutrophil chemotaxis. Inflammation may be caused by infection with pathogenic organisms and viruses or by non-infectious means, such as reperfusion following trauma or myocardial infarction or stroke, immune and autoimmune responses to external antigens. Thus, inflammatory disorders suitable for treatment with a compound of the invention or a salt thereof (e.g., a pharmaceutically acceptable salt thereof) include diseases associated with a reaction of a specific defense system as well as a non-specific defense system.

By "specific defence system" is meant the components of the immune system that react to the presence of specific antigens. Examples of inflammation caused by a response of a specific defense system include a typical response to a foreign antigen, autoimmune diseases, and delayed hypersensitivity mediated by T-cells. Chronic inflammatory diseases, rejection of solid transplanted tissues and organs such as kidney and bone marrow transplantation, and Graft Versus Host Disease (GVHD) are further examples of specific defense system inflammatory responses.

The term "non-specific defense system" means inflammatory disorders mediated by white blood cells (e.g. granulocytes and macrophages) that fail immunological memory. Examples of inflammation caused at least in part by non-specific defense system reactions include inflammation associated with: such as adult (acute) respiratory distress syndrome (ARDS) or multiple organ injury syndrome; reperfusion injury; acute glomerulonephritis; reactive arthritis; skin diseases with acute inflammatory components; acute purulent meningitis or other central nervous system inflammatory disorders such as stroke; heat damage; inflammatory bowel disease; -granulocyte transfusion-related syndrome; and cytokine-induced toxicity.

By "autoimmune disease" is meant any group of disorders in which tissue damage is associated with a humoral or cell-mediated response to the body's own components. Non-limiting examples of autoimmune diseases include rheumatoid arthritis, lupus and multiple sclerosis.

"allergic disease" as used herein means any symptom, tissue damage or loss of tissue function caused by an allergy. "arthritic disease" as used herein, means any disease characterized by inflammatory damage to joints attributable to multiple etiologies. "dermatitis" as used herein means any of a large family of skin diseases characterized by skin inflammation due to a variety of etiologies. "transplant rejection" as used herein means any immune response directed against transplanted tissue such as an organ or cells (e.g., bone marrow) characterized by loss of function of the transplanted and surrounding tissues, pain, swelling, leukocytosis, and thrombocytopenia. The therapeutic methods of the present invention include methods of treating disorders associated with inflammatory cell activation.

By "inflammatory cell activation" is meant cell surface expression of a new or increased number of mediators (including but not limited to major histocompatibility antigens or cell adhesion molecules) in a proliferative cell response induced by a stimulus (including but not limited to cytokines, antigens, or autoantibodies), production of soluble mediators (including but not limited to cytokines, oxygen radicals, enzymes, prostanoids, or vasoactive amines), or inflammatory cells (including but not limited to monocytes, macrophages, T lymphocytes, B lymphocytes, granulocytes (i.e., polymorphonuclear leukocytes such as neutrophils, basophils, and eosinophils), mast cells, dendritic cells, langerhans cells, and endothelial cells). One skilled in the art will recognize that activation of one or a combination of these phenotypes in the cell can promote the initiation, perpetuation, or exacerbation of an inflammatory disease.

In some embodiments, inflammatory disorders that may be treated according to the methods of the present invention include, but are not limited to, asthma, rhinitis (e.g., allergic rhinitis), allergic airway syndrome, atopic dermatitis, bronchitis, rheumatoid arthritis, psoriasis, contact dermatitis, chronic obstructive pulmonary disease, and delayed type hypersensitivity.

The terms "cancer" and "cancerous," "tumor (neoplasms)" and "tumor (tumor)" and related terms mean or describe the physiological condition of a mammal, which is typically characterized by uncontrolled cell growth. A "tumor" includes one or more cancer cells. Examples of cancers include carcinomas, blastomas, sarcomas, seminomas, glioblastomas, melanomas, leukemias, and myeloid or lymphoid malignancies. More specific examples of such cancers include squamous cell cancer (e.g., epithelial squamous cell cancer) and lung cancer, including small-cell lung cancer, non-small cell lung cancer ("NSCLC"), adenocarcinoma of the lung, and squamous carcinoma of the lung. Other cancers include skin cancer, keratoacanthoma (keratoacanthoma), follicular cancer, hairy cell leukemia, oral cancer, pharyngeal (oral) cancer, lip cancer, tongue cancer, oral (mouth) cancer, salivary gland cancer, esophageal cancer, laryngeal cancer, hepatocellular (hepatocellulara), gastric (gastric cancer), gastric (stomach), gastrointestinal (gastrointestinal) cancer, small intestinal cancer, large intestinal cancer, pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatocellular (hepatoma), breast cancer, colon cancer, rectal cancer, colorectal cancer, genitourinary cancer, biliary tract cancer, thyroid cancer, papillary (papillary), liver (hepatotic), endometrial cancer, uterine cancer, salivary gland cancer, kidney (kidney) or kidney (renal) cancer, prostate cancer, testicular cancer, vulval cancer, peritoneal cancer, anal cancer, penile cancer, bone cancer, multiple myeloma, B-cell lymphoma, central nervous system cancer, brain cancer, head and neck cancer, renal (renal) cancer, prostate cancer, testicular cancer, vulval cancer, peritoneal cancer, renal cancer, penile cancer, multiple myeloma, B-cell, Hodgkin's disease and related metastases. Examples of neoplastic diseases (neoplasic disorders) include myeloproliferative disorders, such as polycythemia vera, essential thrombocythemia, myelofibrosis, such as essential myelofibrosis, and Chronic Myelogenous Leukemia (CML).

A "chemotherapeutic agent" is an agent that can be used to treat a given disorder, such as cancer or an inflammatory disease. Examples of chemotherapeutic agents are well known in the art and include, for example, those disclosed in U.S. published application No. 2010/0048557, incorporated herein by reference. In addition, the chemotherapeutic agent includes a pharmaceutically acceptable salt, acid or derivative of any of the chemotherapeutic agents, as well as combinations of two or more thereof.

"package insert" is used to mean an insert typically included in commercial packaging for a therapeutic product that contains indications, usage, dosages, administration, contraindications, or warning information for use of the therapeutic product.

Unless otherwise indicated, structures described herein include compounds that differ only in the presence of one or more isotopically enriched atoms. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, such as2H、3H、11C、13C、14C、13N、15N、15O、17O、18O、32P、33P、35S、18F、36Cl、123I and125I. isotopically-labelled compounds (e.g. with3H and14c-labeled ones) can be used in compound or substrate tissue distribution assays. Tritium (i.e. tritium3H) And carbon-14 (i.e.14C) Isotopes are useful for their ease of preparation and detection. Further, with heavier isotopes such as deuterium (i.e., deuterium)2H) Substitution may provide certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements). In some embodiments, one or more hydrogen atoms are replaced with2H or3H, or one or more carbon atoms substituted by13C-or14C-enriched carbon substitution. Positron emitting isotopes such as15O、13N、11C and18f can be used in Positron Emission Tomography (PET) studies to detect substrate receptor occupancy. Isotopically labeled compounds can generally be prepared by following procedures analogous to those disclosed in the schemes and examples herein, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.

It is specifically contemplated that any limitation discussed with respect to one embodiment of the invention may apply to any other embodiment of the invention. In addition, any compound or salt thereof (e.g., a pharmaceutically acceptable salt thereof) or composition of the invention can be used in any method of the invention, and any method of the invention can be used to produce or use any compound or salt thereof (e.g., a pharmaceutically acceptable salt thereof) or composition of the invention.

The use of the term "or" is used to indicate "and/or" unless clearly indicated to refer only to alternatives or mutual exclusions between alternatives, but this disclosure supports the definition of alternatives and "and/or" only.

In this application, the term "about" is used to indicate that a numerical value includes the standard deviation of the equipment or method used to determine the value.

"A" or "an" as used herein means one or more unless clearly indicated otherwise. "another", as used herein, means at least a second or more.

Headings are used herein for typesetting purposes only.

Janus kinase inhibitors

One embodiment provides a compound of formula (I) or a salt (e.g., a pharmaceutically acceptable salt) thereof:

Figure BDA0002282880340000221

wherein:

R1is hydrogen or CH3

R2Is halogen, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C3-C6Cycloalkyl OR-ORaWherein R is2Optionally substituted by one or more groups independently selected from halogen, C1-C3Alkyl, cyano, hydroxy and oxo;

Rais C1-C6Alkyl, -phenyl-CORbRc-phenyl- (3-6-membered heterocyclyl) or 3-11-membered heterocyclyl, wherein R isaOptionally substituted by one or more groups independently selected from halogen, C1-C3Alkyl, cyano, hydroxy and oxo;

each RbAnd RcIndependently is hydrogen or CH3

R3Is hydrogen or NH2

R4Is hydrogen or CH3(ii) a And is

R5Is hydrogen or NH2

In some embodiments, R1Is hydrogen. In some embodiments, R1Is CH3. In some embodiments, R3Is hydrogen. In some embodiments, each R is4And R5Is hydrogen. In some embodiments, each R is1、R3、R4And R5Is hydrogen.

In some embodiments, R2Selected from halogen, C1-C6Haloalkyl and C1-C6A haloalkoxy group. In some embodiments, R2Is selected from

In some embodiments, there is provided a compound selected from the following 1-18, or a salt (e.g., a pharmaceutically acceptable salt) or stereoisomer thereof:

Figure BDA0002282880340000232

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) thereof are provided:

Figure BDA0002282880340000252

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) thereof are provided:

Figure BDA0002282880340000261

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) thereof are provided:

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) or stereoisomers thereof are provided:

Figure BDA0002282880340000263

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) thereof are provided:

Figure BDA0002282880340000264

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) thereof, or stereoisomers thereof, are provided:

Figure BDA0002282880340000271

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) thereof are provided:

Figure BDA0002282880340000272

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) thereof are provided:

Figure BDA0002282880340000273

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) or stereoisomers thereof are provided:

Figure BDA0002282880340000274

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) or stereoisomers thereof are provided:

Figure BDA0002282880340000281

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) thereof are provided:

Figure BDA0002282880340000282

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) thereof are provided:

Figure BDA0002282880340000283

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) thereof are provided:

Figure BDA0002282880340000284

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) thereof are provided:

Figure BDA0002282880340000291

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) thereof are provided:

Figure BDA0002282880340000292

in some embodiments, the following compounds or salts (e.g., pharmaceutically acceptable salts) thereof are provided:

Figure BDA0002282880340000293

in some embodiments, there is provided a compound selected from the following (a) - (v), or a salt (e.g., a pharmaceutically acceptable salt) or stereoisomer thereof:

Figure BDA0002282880340000294

Figure BDA0002282880340000301

also provided are pharmaceutical compositions comprising a JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent, or excipient.

Also provided is the use of a JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, in therapy, such as the treatment of inflammatory diseases (e.g., asthma). Also provided is the use of a JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of an inflammatory disease. Also provided are methods of preventing, treating, or lessening the severity in a patient of a disease or disorder responsive to inhibition of Janus kinase activity, comprising administering to the patient a therapeutically effective amount of a JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof.

In one embodiment, the disease or disorder treated is cancer, polycythemia vera, essential thrombocythemia, myelofibrosis, Chronic Myelogenous Leukemia (CML), rheumatoid arthritis, inflammatory bowel disease, Crohn's disease, psoriasis, contact dermatitis, or delayed-type hypersensitivity.

In one embodiment, there is provided a use of a JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, for treating: cancer, polycythemia vera, essential thrombocythemia, myelofibrosis, Chronic Myelogenous Leukemia (CML), rheumatoid arthritis, inflammatory bowel disease, Crohn's disease, psoriasis, contact dermatitis or delayed type hypersensitivity reactions.

In one embodiment, a composition formulated for administration by inhalation is provided.

In one embodiment, there is provided a metered dose inhaler comprising a compound of the present invention or a pharmaceutically acceptable salt thereof.

In one embodiment, the JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, has at least five times the potency as a JAK1 inhibitor as a LRRK2 inhibitor.

In one embodiment, the JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, has at least ten times the potency as a JAK1 inhibitor as a LRRK2 inhibitor.

In one embodiment, the JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, has at least five times greater potency as an inhibitor of JAK1 than as an inhibitor of JAK 2.

In one embodiment, the JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, has at least ten times greater potency as a JAK1 inhibitor than as a JAK2 inhibitor.

In one embodiment, the JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, has at least five times greater potency as an inhibitor of JAK1 than as an inhibitor of JAK 3.

In one embodiment, the JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, has at least ten times greater potency as a JAK1 inhibitor than as a JAK3 inhibitor.

In one embodiment, the JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, is at least five times more potent as a JAK1 inhibitor than as a TYK2 inhibitor.

In one embodiment, the JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, has at least ten times the potency as a JAK1 inhibitor as a TYK2 inhibitor.

In one embodiment, there is provided a method of treating hair loss in a mammal comprising administering to the mammal a JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof.

In one embodiment, there is provided the use of a JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, for treating alopecia.

In one embodiment, there is provided a use of a JAK inhibitor described herein, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating hair loss in a mammal.

The compounds of the present invention may contain one or more asymmetric carbon atoms. Thus, the compounds may exist as diastereomers, enantiomers, or mixtures thereof. Compound synthesis racemates, diastereomers or enantiomers can be used as starting materials or intermediates. Mixtures of specific diastereomeric compounds can be separated or enriched in one or more specific diastereomers by chromatography or crystallization. Similarly, enantiomeric mixtures or enantiomeric enrichments can be separated using the same techniques or other techniques known in the art. Each asymmetric carbon or nitrogen atom may be in either the R or S configuration, and both configurations are included within the scope of the present invention.

In the structures shown herein, if the stereochemistry of any particular chiral atom is unspecified, all stereoisomers are contemplated and are included as compounds of the present invention. When stereochemistry is defined by a solid wedge or dashed line representing a particular configuration, then the stereoisomer is so defined and defined. Unless otherwise indicated, if solid wedges or dashed lines are used, it is intended to indicate relative stereochemistry.

Another aspect includes prodrugs of the compounds described herein, including known amino protecting groups and carboxy protecting groups, which are released, e.g., hydrolyzed, under physiological conditions to yield the compounds of the invention.

The term "prodrug" means a precursor or derivative form of a pharmaceutically active substance that is less effective in a patient than the parent drug and is capable of being activated by enzyme or hydrolysis or converted to the more active parent form. See, for example, Wilman, "Prodrug Cancer therapy" Biochemical Society Transactions,14, pp.375-382,615 through Belfast (1986) and Stella et al, "Prodrugs: A Chemical Approach to targeted Drug Delivery," Directed Drug Delivery, Borchardt et al, (ed.), pp.247-267, HumanaPress (1985). Prodrugs include, but are not limited to, phosphate-containing prodrugs, thiophosphate-containing prodrugs, sulfate-containing prodrugs, peptide-containing prodrugs, D-amino acid-modified prodrugs, glycosylated prodrugs, β -lactam-containing prodrugs, optionally substituted phenoxyacetamide-containing prodrugs or optionally substituted phenylacetamide-containing prodrugs, and 5-fluorocytosine and 5-fluorouridine prodrugs.

A particular class of prodrugs are compounds wherein the nitrogen atom in the amino, amidino, aminoalkyleneamino, iminoalkyleneamino or guanidino group is substituted by: hydroxy (-OH), alkylcarbonyl (-CO-R), alkoxycarbonyl (-CO-OR), OR acyloxyalkyl-alkoxycarbonyl (-CO-O-R-O-CO-R), wherein R is a monovalent OR divalent group such as alkyl, alkylene OR aryl, OR a group having the formula-C (O) -O-CP1P 2-haloalkyl in which P1 and P2 are the same OR different and are hydrogen, alkyl, alkoxy, cyano, halogen, alkyl OR aryl. In a particular embodiment, the nitrogen atom is one of the nitrogen atoms of the amidino group. Prodrugs can be prepared by reacting a compound with an activated group, such as an acyl group, to, for example, bond a nitrogen atom in the compound to the exemplary carbonyl group activating the acyl group. Examples of activated carbonyl compounds are those containing a leaving group bonded to a carbonyl group, and include, for example, acid halides, acyl amines, acyl pyridinium salts, acyl alkoxides, acyl phenolates such as p-nitrophenoxyacyl, dinitrophenoxyacyl, fluorophenoxyacyl, and difluorophenoxyacyl. The reaction is generally carried out in an inert solvent at reduced temperatures, e.g., -78 ℃ to about 50 ℃. The reaction may likewise be carried out in the presence of an inorganic base such as potassium carbonate or sodium bicarbonate or an organic base such as an amine, including pyridine, trimethylamine, triethylamine, triethanolamine and the like.

Other types of prodrugs are also included. For example, the free carboxyl group of the JAK inhibitors described herein can be derivatized as an amide or alkyl ester. As a further example of the use of the coating,compounds of the present invention which include a free hydroxyl group may be derivatized into prodrugs by converting the hydroxyl group to, for example, but not limited to, phosphate, hemisuccinate, dimethylaminoacetate or phosphoryloxymethoxymethoxycarbonyl, as outlined in Fleisher, D.et al, (1996) Improved oral Drug Delivery by the use of Advanced Drug Delivery Reviews,19:115. Carbamate prodrugs of hydroxy and amino groups are also included, such as carbonate prodrugs of hydroxy groups, sulfonates and sulfates. Hydroxyl groups are derivatized as (acyloxy) methyl and (acyloxy) ethyl ethers wherein the acyl group may be an alkyl ester optionally substituted with groups including, but not limited to, ether, amine and carboxylic acid functional groups or wherein the acyl group is an amino acid ester as described previously are also included. Prodrugs of this type are described in j.med.chem., (1996),39:10. More specific examples include replacement of the hydrogen atom on the alcohol group by a group such as (C)1-C6) Alkanoyloxymethyl, 1- ((C)1-C6) Alkanoyloxy) ethyl, 1-methyl-1- ((C)1-C6) Alkanoyloxy) ethyl group, (C)1-C6) Alkoxycarbonyloxymethyl, N- (C)1-C6) Alkoxycarbonylaminomethyl, succinyl, (C)1-C6) Alkanoyl, alpha-amino (C)1-C4) Alkanoyl, arylacyl and alpha-aminoacyl or alpha-aminoacyl-alpha-aminoacyl wherein each alpha-aminoacyl is independently selected from the group consisting of a naturally occurring L-amino acid, P (O) (OH)2、-P(O)(O(C1-C6) Alkyl radical)2Or a glycosyl (a group obtained by removing the hydroxyl group of the hemiacetal form of a carbohydrate).

By "leaving group" is meant a portion of a first reactant in a chemical reaction that is displaced from the first reactant in the chemical reaction. Examples of leaving groups include, but are not limited to, halogen atoms, alkoxy groups, and sulfonyloxy groups. Examples of sulfonyloxy groups include, but are not limited to, alkylsulfonyloxy groups such as methylsulfonyloxy (mesylate) and trifluoromethylsulfonyloxy (triflate) groups, and arylsulfonyloxy groups such as p-toluenesulfonyloxy (tosylate) and p-nitrobenzenesulfonyloxy (nosylate group).

Synthesis of Janus kinase inhibitor compounds

The compounds may be synthesized by the synthetic routes described herein. In certain embodiments, methods well known in the chemical arts can be used in addition to or in accordance with the descriptions contained herein. Starting materials are generally commercially available from commercial sources such as Aldrich Chemicals (Milwaukee, Wis.) or are readily prepared using methods well known to those skilled in the art (e.g., as generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v.1-19, Wiley, N.Y. (1967. 1999.), Beilsteins handbuch der organischen Chemie,4, autofl. ed. Springer-Verlag, Berlin, including supples (also available from Beilstein online databases)), or Comprehensive Heterocyclic Chemistry, Editors Katrizky and Rees, Pergamon Press, 1984.

The compounds may be prepared individually or as compound libraries comprising at least 2, e.g. 5 to 1,000 or 10 to 100 compounds. Libraries of compounds can be prepared by a combinatorial "separation and mixing" approach or by multiple parallel syntheses using solution phase or solid phase chemistry, by methods known to those skilled in the art. Thus, according to a further aspect of the invention, there is provided a library of compounds comprising at least 2 compounds of the invention.

For illustrative purposes, the following reaction schemes provide synthetic routes to the compounds of the present invention as well as key intermediates. See the examples section below for a more detailed description of the individual reaction steps. Those skilled in the art will appreciate that other synthetic routes may also be employed. Although some specific starting materials and reagents are described and discussed in the schemes below, other starting materials and reagents may be substituted to provide different derivatives or reaction conditions. In addition, many of the compounds prepared according to the methods described below can be further modified according to the present disclosure using conventional chemistry well known to those skilled in the art.

In preparing the compounds of the present invention, it may be necessary to protect remote functional groups (e.g., primary or secondary amines) of intermediates. The need for such protection may vary depending on the nature of the remote functional group and the conditions of the preparation process. Suitable amino-protecting groups include acetyl, trifluoroacetyl, benzyl, phenylsulfonyl, tert-Butyloxycarbonyl (BOC), benzyloxycarbonyl (CBZ) and 9-fluorenylmethoxycarbonyl (Fmoc). The need for such protection is readily determined by those skilled in the art. General descriptions of protecting Groups and their uses are found in T.W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.

Other transformations commonly used in the synthesis of the compounds of the present invention may be performed using a variety of reagents and conditions, including the following:

(1) the carboxylic acid reacts with the amine to form an amide. The conversion can be achieved using a variety of agents known to those skilled in the art, but for a comprehensive review see Tetrahedron,2005,61,10827, 10852.

(2) The reaction of primary or secondary amines with aryl halides or pseudohalides such as triflates, commonly referred to as "Buchwald-Hartwig cross-coupling reactions," can be achieved by using a variety of catalysts, ligands, and bases. A summary of these methods is provided by Comprehensive Organic Name Reactions and Reagents,2010, 575-.

(3) Palladium cross-coupling reactions between aryl halides and vinyl boronic acids or esters. This transformation is one of the "Suzuki-Miyaura cross-coupling reactions", of the type well reviewed in Chemical Reviews,1995,95(7),2457-2483.

(4) Ester hydrolysis to produce the corresponding carboxylic acid is well known to those skilled in the art and conditions include: for the methyl and ethyl esters, strong aqueous alkaline solutions such as lithium hydroxide, sodium hydroxide or potassium hydroxide or strong aqueous mineral acids such as HCl; for the tert-butyl ester, hydrolysis is achieved using an acid, for example, HCl in dioxane or trifluoroacetic acid (TFA) in Dichloromethane (DCM).

Reaction scheme 1

Figure BDA0002282880340000361

Reaction scheme 1 illustrates the synthesis of compounds 6, 8 and 10 therein. Compound 1 can be arylated with 4-bromo-1- (difluoromethoxy) -2-iodobenzene under palladium catalysis to produce compound 2. Process for preparation of Compound 2The nitro group can be reduced using conditions such as iron and ammonium chloride to produce the aminoaniline 3. Amide bond with commercially available pyrazole [1,5-a ]]Pyrimidine-3-carboxylic acid is coupled in an organic solution such as but not limited to DMF in the presence of a coupling reagent such as but not limited to PyAOP and an organic base such as but not limited to DIPEA and DMAP to provide compound 4. Compound 4 can be converted to the corresponding iodide 5 using conditions such as sodium iodide and CuI with a base such as N, N-dimethylethane-1, 2-diamine in a solvent such as tBuOH. By using substituted boric acid (or ester) or BF3Compound 7 can be formed by treating compound 4 with a salt of K under palladium catalyzed conditions and a base such as, but not limited to, cesium carbonate in a solvent such as, but not limited to, 1, 4-dioxane. In addition, compound 9 can be synthesized by treating compound 4 with an appropriately substituted phenol under Pd-catalyzed coupling conditions with a base such as, but not limited to, cesium carbonate in a solvent such as, but not limited to, toluene. Removal of the SEM protecting groups in compounds of formulae 5, 7 and 9 to yield compounds of formulae 6, 8 and 10 can be accomplished by using an acid such as, but not limited to, HCl in a solvent such as, but not limited to, 1, 4-dioxane.

Reaction scheme 2

Figure BDA0002282880340000371

Scheme 2 illustrates the synthesis of compounds of formulae I-III therein. Compounds of formulae 6, 8 and 10 may be treated with appropriately substituted 2-bromo-N, N-dimethylacetamide with a base such as, but not limited to, cesium carbonate in a solvent such as, but not limited to, DMF to give compounds of formulae I-III.

Reaction scheme 3

Figure BDA0002282880340000381

Reaction scheme 3 illustrates the synthesis of compounds of formulae IV-VI therein. The compound of formula II (R) may be treated with an appropriately substituted phenol under Pd-catalyzed coupling conditions with a base such as, but not limited to, cesium carbonate, in a solvent such as, but not limited to, toluene1Br) to give a compound of formula IV. With boric acid (or ester) or BF optionally substituted3K salt with a base such as, but not limited to, palladium catalyzedCompound 4 is treated in cesium carbonate in a solvent such as, but not limited to, 1, 4-dioxane to give the compound of formula V. Difluoromethyl compounds of formula VI can be obtained using the methods described in j.am.chem.soc.,2014,136, 4149-4152. Furthermore, when R of the compound of formula V1When appropriately substituted, further treatment of the alkene using standard procedures can be effected to provide the fluoroalkane.

Reaction scheme 4

Figure BDA0002282880340000391

Scheme 4 illustrates the synthesis of compounds of formula VII therein. Commercially available 4- (difluoromethoxy) phenol can be treated with a brominating agent such as, but not limited to, NBS in a solvent such as, but not limited to, acetic acid to afford compound 12. Difluoromethylation of compound 12 can be achieved by treatment of compound 12 with diethyl (bromodifluoromethyl) phosphate and a base such as, but not limited to, potassium hydroxide in a solvent such as, but not limited to, acetonitrile to form compound 13. Compound 13 can be treated with 4-nitro-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrazole 4-bromo-1- (difluoromethoxy) -2-iodobenzene under palladium catalyzed conditions with a base such as, but not limited to, potassium carbonate in a solvent such as, but not limited to, DMA to produce compound 14. The nitro group of compound 14 can be reduced using conditions such as iron and ammonium chloride to produce the aminoaniline 15. Coupling of the amide bond of compound 15 with commercially available pyrazolo [1,5-a ] pyrimidine-3-carboxylic acid in the presence of a coupling reagent such as, but not limited to, PyAOP and an organic base such as, but not limited to, DIPEA and DMAP in a solvent such as, but not limited to, DMF provides compound 16. SEM protecting groups of compound 16 can be removed with an acid such as, but not limited to, HCl in an organic solvent such as, but not limited to, 1, 4-dioxane to yield the compound of formula VII.

Reaction scheme 5

Figure BDA0002282880340000401

Reaction scheme 5 illustrates the synthesis of compounds of formula VIII therein. Compound 17 can be treated with 2-bromo-N, N-dimethylacetamide with a base such as, but not limited to, cesium carbonate in a solvent such as, but not limited to, DMF to provide compound 18. The BOC protecting group of compound 18 can be removed with an acid such as, but not limited to, HCl in an organic solvent such as, but not limited to, 1, 4-dioxane to yield the compound of formula VIII.

It will be appreciated that the various compounds of formula (la) or any intermediate used in their preparation, when a suitable functional group is present, may be further derivatized by one or more standard synthetic methods, using condensation, substitution, oxidation, reduction or shear reactions. Specific substitution methods include conventional alkylation, arylation, heteroarylation, acylation, sulfonylation, halogenation, nitration, formylation, and coupling methods.

In another example, a primary or secondary amine group can be converted to an amide group (-NHCOR 'or-NRCOR') by acylation. Acylation may be achieved by reaction with the appropriate acid chloride in the presence of a base such as triethylamine in a suitable solvent such as dichloromethane, or by reaction with the appropriate carboxylic acid in the presence of a suitable coupling reagent such as HATU (O- (7-azabenzotriazol-1-yl) -N, N' -tetramethyluronium hexafluorophosphate) in a suitable solvent such as dichloromethane. Similarly, an amine group can be converted to a sulfonamide group (-NHSO) by reaction with a suitable sulfonyl chloride in the presence of a suitable base such as triethylamine in a suitable solvent such as dichloromethane2R 'or-NR' SO2R'). The primary or secondary amine group can be converted to a ureido group (-NHCONR 'R' or-NRCONR 'R') by reaction with a suitable isocyanate in the presence of a suitable base such as triethylamine in a suitable solvent such as dichloromethane.

Can pass through nitro (-NO)2) Reduction to give amine (-NH)2) For example by catalytic hydrogenation using, for example, hydrogen in the presence of a metal catalyst, for example palladium on carbon, in a solvent such as ethyl acetate or an alcohol such as methanol. Alternatively, conversion may be achieved by chemical reduction using, for example, a metal, such as tin or iron, in the presence of an acid, such as hydrochloric acid.

In another example, a nitrile (-CN) may be reduced to give an amino (-CH)2NH2) E.g. catalytic hydrogenation using, e.g. hydrogen on a metal catalyst, e.g. palladium on a support such as carbon or raneyIn the presence of nickel in a solvent such as an ether, for example a cyclic ether such as tetrahydrofuran, at a suitable temperature, for example from about-78 ℃ to the reflux temperature of the solvent.

In another example, amino (-NH)2) May be derived from carboxylic acid groups (-CO)2H) By conversion to the corresponding acyl azide (-CON)3) Curtius rearrangement and hydrolysis of the resulting isocyanate (-N ═ C ═ O).

Aldehyde groups (-CHO) can be converted to amine groups (-CH) by reductive amination2NR' R "), using amines and borohydrides, for example sodium triacetoxyborohydride or sodium cyanoborohydride, in solvents such as halogenated hydrocarbons, for example dichloromethane, or alcohols such as ethanol, if desired in the presence of acids such as acetic acid, at around ambient temperature.

In another example, the aldehyde group can be converted to an alkenyl (-CH ═ CHR') group using a Wittig or wadswworth-Emmons reaction using a suitable phosphorane or phosphonate under standard conditions known to those skilled in the art.

The aldehyde group can be reduced by using diisobutylaluminum hydride in a suitable solvent such as toluene (e.g., -CO)2Et) or a nitrile group (-CN). Alternatively, the alcohol group may be oxidized to give an aldehyde group using any suitable oxidizing agent known to those skilled in the art.

Depending on the nature of R, the ester group (-CO) can be hydrolyzed by acid or base catalysis2R') into the corresponding acid group (-CO)2H) In that respect If R is t-butyl, acid-catalyzed hydrolysis may be effected, for example, by treatment with an organic acid, such as trifluoroacetic acid, in an aqueous solvent or by treatment with a mineral acid, such as hydrochloric acid, in an aqueous solvent.

Carboxylic acid group (-CO)2H) Can be converted to an amide group (CONHR 'or-CONR' R ") by reaction with the appropriate amine in the presence of a suitable coupling reagent such as HATU in a suitable solvent such as dichloromethane.

In another example, a carboxylic acid can be homologated to one carbon (i.e., -CO) by conversion to the corresponding acid chloride (-COCl) and subsequent synthesis by Arndt-Eistert2H to-CH2CO2H)。

In another example, the-OH group can be formed from the corresponding ester (e.g.-CO2R') or an aldehyde (-CHO) is produced by reduction using, for example, a complex metal hydride such as lithium aluminum hydride in diethyl ether or tetrahydrofuran, or sodium borohydride in a solvent such as methanol. Alternatively, the alcohol may be prepared by reducing the corresponding acid (-CO)2H) The preparation is carried out, for example, using lithium aluminum hydride in a solvent such as tetrahydrofuran, or using borane in a solvent such as tetrahydrofuran.

The alcohol group may be converted to a leaving group, such as a halogen atom or a sulfonyloxy group, such as an alkylsulfonyloxy group, e.g. a trifluoromethylsulfonyloxy group or an arylsulfonyloxy group, e.g. a p-toluenesulfonyloxy group, using conditions known to the person skilled in the art. For example, an alcohol may be reacted with thionyl chloride in a halogenated hydrocarbon (e.g., dichloromethane) to give the corresponding chloride. A base (e.g., triethylamine) may also be used in the reaction.

In another example, the alcohol, phenol, or amide group may be alkylated by coupling the diphenol or amide with the alcohol in a solvent such as tetrahydrofuran in the presence of a phosphine such as triphenylphosphine and an activator such as diethyl, diisopropyl, or dimethyl azodicarboxylate. Alternatively, alkylation can be achieved using a suitable base such as sodium hydride to deprotonate followed by the addition of an alkylating agent such as an alkyl halide.

The aromatic halogen substituents in the compounds may be subjected to a halogen-metal exchange by treatment with a base, e.g. a lithium base such as n-butyl or t-butyl lithium, optionally at low temperature, e.g. around-78 ℃, in a solvent such as tetrahydrofuran, followed by quenching with an electrophile to introduce the desired substituent. Thus, for example, a formyl group can be introduced by using N, N-dimethylformamide as electrophile. Alternatively, the aromatic halogen substituent may undergo a metal (e.g., palladium or copper) catalyzed reaction to introduce, for example, an acid, ester, cyano, amide, aryl, heteroaryl, alkenyl, alkynyl, thio-or amino substituent. Suitable methods that may be employed include those described by Heck, Suzuki, Stille, Buchwald or Hartwig.

The aromatic halogen substituents may also undergo nucleophilic displacement after reaction with a suitable nucleophile such as an amine or alcohol. Advantageously, such reactions can be carried out in the presence of microwave radiation at elevated temperatures.

Separation method

In each exemplary embodiment, it may be advantageous to separate the reaction products from each other or from the starting materials. The desired product of each step or series of steps is isolated or purified (hereinafter referred to as isolated) to the desired degree of homogeneity by techniques conventional in the art. Typically the separation involves heterogeneous extraction, crystallization or trituration from a solvent or solvent mixture, distillation, sublimation or chromatography. Chromatography may involve a variety of methods including, for example, reverse phase and normal phase chromatography; size exclusion chromatography; ion exchange chromatography; supercritical fluid chromatography; high, medium and low pressure liquid chromatography methods and apparatus; small scale analytical chromatography; simulated moving bed chromatography (SMB) and preparative thin or thick layer chromatography, as well as small scale thin layer chromatography and flash chromatography techniques.

Another class of separation methods involves treating the mixture with reagents selected to bind or allow it to separate desired products, unreacted starting materials, reaction byproducts, and the like. The reagents include adsorbents or absorbents such as activated carbon, molecular sieves, ion exchange media, and the like. Alternatively, the reagent may be an acid in the case of a basic material and a base in the case of an acidic material, binding reagents such as antibodies, binding proteins, selective chelators such as crown ethers, liquid/liquid ion extractants (LIX), and the like.

The choice of a suitable separation method depends on the nature of the materials involved. Examples of separation methods include boiling point, molecular weight in distillation and sublimation, presence or absence of polar functional groups in chromatography, stability of materials in acidic or basic media in heterogeneous extraction, and the like. Those skilled in the art will apply the techniques most likely to achieve the desired separation.

Mixtures of diastereomers may be separated into the respective diastereomers on the basis of their physicochemical differences by methods well known to those skilled in the art, such as chromatography or fractional crystallization. Enantiomers can be separated as follows: by reaction with a suitable optically active compound, for example a chiral auxiliary such as a chiral alcohol or Mosher's acid chloride, to convert the enantiomeric mixture into a diastereomeric mixture, separation of the diastereomers and conversion (e.g. hydrolysis) of the individual diastereomers to the corresponding pure enantiomers. Likewise, some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are also considered part of the present invention. Enantiomers can also be separated using chiral HPLC columns or supercritical fluid chromatography.

Single stereoisomers, e.g., enantiomers, which are substantially free of their stereoisomers, can be obtained by resolution of racemic mixtures using methods such as formation of diastereomers using optically active resolving agents (Eliel, E. and Wilen, S., Stereochemistry of Organic Compounds, John Wiley & Sons, Inc., New York, 1994; Lochmuller, C.H., J.Chromatogr.,113(3): 283) -302 (1975)). The racemic mixture of the chiral compounds of the present invention can be separated and isolated via any suitable method, including: (1) the ionic diastereomeric salts formed with chiral compounds are separated by fractional crystallization or other means, (2) the diastereomeric compounds formed with a chiral derivatizing reagent, the diastereomers separated and converted to the pure stereoisomers, and (3) the substantially pure or enriched stereoisomers are separated directly under chiral conditions. See: drug Stereochemistry, Analytical Methods and Pharmacology, IrvingW.Wainer, Ed., Marcel Dekker, Inc., New York (1993).

Diastereomeric salts can be formed by reaction of an enantiomerically pure chiral base such as brucine, quinine, ephedrine, strychnine, alpha-methyl-beta-phenylethylamine (amphetamines), and the like, with an asymmetric compound bearing an acidic functional group such as a carboxylic acid and a sulfonic acid. Diastereomeric salts can be induced for separation by fractional crystallization or ion chromatography. For the separation of optical isomers of amino compounds, addition of chiral carboxylic or sulfonic acids such as camphorsulfonic, tartaric, mandelic or lactic acids can form diastereomeric salts.

Alternatively, the substrate to be resolved is reacted with one enantiomer of a chiral compound to produce a diastereomer pair (Eliel, E. and Wilen, S., Stereochemistry of Organic Compounds, John Wiley & Sons, Inc., New York,1994, p.322). Diastereomeric compounds can be formed by reacting an asymmetric compound with an enantiomerically pure chiral derivatizing reagent, such as a menthyl derivative, followed by separation of the diastereomers and hydrolysis to give the pure or enriched enantiomer. The method for determining optical purity involves the preparation of a chiral ester of a racemic mixture, such as menthyl ester, for example, (-) menthyl chloroformate, or Mosher ester, alpha-methoxy-alpha- (trifluoromethyl) phenyl acetate (Jacob, j.org. chem.47:4165(1982)) in the presence of a base, and NMR spectroscopic analysis of the presence of the two diastereomeric enantiomers or diastereomers. The stable diastereoisomers of the atropisomeric compounds can be separated and isolated by normal-and reverse-phase chromatography, following the method for separating the atropisomeric naphthyl-isoquinoline (WO 96/15111, incorporated herein by reference). By the method (3), racemic mixtures of the two enantiomers can be separated by chromatography on a Chiral stationary phase (Chiral Liquid chromatography W.J.Lough, Ed., Chapman and Hall, New York, (1989); Okamoto, J.of chromatography.513: 375-. Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules containing asymmetric carbon atoms, such as optical rotation and circular dichroism. The absolute stereochemistry of chiral centers and enantiomers can be determined by x-ray crystallography.

Positional isomers and intermediates for their synthesis can be observed by characterization methods such as NMR and analytical HPLC. For certain compounds with sufficiently high interconversion energy barriers, the E and Z isomers may be separated, for example by preparative HPLC.

Pharmaceutical compositions and administration

The compounds to which the present invention relates are JAK kinase inhibitors, such as JAK1 inhibitors, and may be useful in the treatment of a variety of diseases, for example inflammatory diseases, such as asthma.

Accordingly, another embodiment provides a pharmaceutical composition or medicament comprising a compound of the invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent or excipient, and methods of using the compounds of the invention to prepare such compositions and medicaments.

In one example, a compound of the invention or a pharmaceutically acceptable salt thereof can be formulated for galenic administration by admixture with a physiologically acceptable carrier at ambient temperature, at a suitable pH and in the desired purity, i.e. a carrier which is non-toxic to the recipient at the dosages and concentrations employed. The pH of the formulation depends primarily on the particular use and concentration of the compound, but is typically in any range from about 3 to about 8. In one example, a compound of the invention or a pharmaceutically acceptable salt thereof is formulated in acetate buffer at pH 5. In another embodiment, the compounds of the invention are sterile. The compounds may be stored, for example, as solid or amorphous compositions, lyophilized formulations, or aqueous solutions.

The formulation, administration and mode of administration of the compositions are in accordance with good medical practice. Factors to be considered in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of agent delivery, the method of administration, the schedule of administration, and other factors well known to practitioners of the physician.

It will be understood that the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination and the severity of the particular disease undergoing therapy. The optimal dose level and frequency of administration will be determined by clinical trials, as required in the pharmaceutical arts. Typically, the daily dose for oral administration will range from about 0.001mg to about 100mg per kg of body weight of the patient, often from 0.01mg to about 50mg per kg, for example from 0.1 to 10mg per kg, taken in single or divided doses. In general, the daily dose for inhalation will range from about 0.1 μ g to about 1mg per kg of patient body weight, preferably 0.1 μ g to 50 μ g per kg, in single or divided doses. On the other hand, it may be necessary in certain cases to use dosages which exceed these limits.

The compounds of the invention or pharmaceutically acceptable salts thereof may be administered by any suitable means including oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intrathecal, inhalation and epidural and intranasal, and if desired for topical treatment, intralesional administration. Parenteral infusion includes intramuscular, intravenous, intraarterial, intraperitoneal or subcutaneous administration. In some embodiments, administration is by inhalation.

The compounds of the present invention or pharmaceutically acceptable salts thereof may be administered in any convenient form of administration, such as tablets, powders, capsules, lozenges, granules, solutions, dispersions, suspensions, syrups, sprays, vapors (vapors), suppositories, gels, emulsions, patches and the like. The compositions may contain conventional ingredients for pharmaceutical formulations such as diluents (e.g., glucose, lactose or mannitol), carriers, pH adjusting agents, buffers, sweeteners, fillers, stabilizers, surfactants, wetting agents, lubricants, emulsifiers, suspending agents, preservatives, antioxidants, opacifiers, glidants, processing aids, colorants, flavoring agents, other known additives and other active agents.

Suitable carriers and excipients are well known to those skilled in the art and are described in detail, for example, in Ansel, Howard C, et al, Ansel's Pharmaceutical Delivery Forms and Drug Delivery systems, Philadelphia, Lippincott, Williams & Wilkins, 2004; gennaro, Alfonso R., et al, Remington, The Science and Practice of pharmacy Philadelphia, Lippincott, Williams & Wilkins, 2000; and Rowe, Raymond C.handbook of Pharmaceutical excipients Chicago, Pharmaceutical Press, 2005. For example, carriers include solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drugs, drug stabilizers, gels, binders, excipients, disintegrants, lubricants, sweeteners, flavoring agents, dyes, and the like, and combinations thereof, as known to those skilled in the art (see, e.g., Remington's Pharmaceutical Sciences, pp 1289-1329, 1990). Unless any conventional carrier is incompatible with the active ingredient, its use in therapeutic or pharmaceutical compositions is contemplated. Exemplary excipients include dicalcium phosphate, mannitol, lactose, starch, magnesium stearate, sodium saccharin, cellulose, magnesium carbonate, or combinations thereof. Pharmaceutical compositions may include different types of carriers or excipients depending on whether they are to be administered in solid, liquid or aerosol form or whether the route of administration requires sterile administration.

For example, tablets and capsules for oral administration may be presented in unit dosage form and may contain conventional excipients such as binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers, for example lactose, sugar, corn starch, calcium phosphate, sorbitol or glycine; tabletting lubricants, for example magnesium stearate, talc, polyethylene glycol or silica; disintegrants for example potato starch or acceptable wetting agents such as sodium lauryl sulphate. The tablets may be coated according to methods well known in conventional pharmaceutical practice. Oral liquid preparations may be in the form of, for example, aqueous or oily suspensions, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for constitution with water or other suitable vehicle before use. The liquid preparations may contain conventional additives such as suspending agents, for example sorbitol, syrup, methyl cellulose, glucose syrup, gelatin hydrogenated edible fats; emulsifying agents, for example lecithin, sorbitan monooleate or acacia; non-aqueous vehicles (which may include edible oils), for example almond oil, fractionated coconut oil, oily esters such as glycerol, propylene glycol or ethanol; preservatives, for example methyl or propyl p-hydroxybenzoate or sorbic acid, and, if desired, conventional flavouring or colouring agents.

For topical application to the skin, the compounds may be formulated as a cream, lotion or ointment. Cream or ointment formulations which may be used in medicine are conventional formulations well known in the art, for example as described in standard textbooks of pharmacy, such as British Pharmacopoeia.

The compounds of the invention or pharmaceutically acceptable salts thereof may also be formulated for inhalation, for example as a nasal spray, or as a dry powder or aerosol inhaler. For inhalation delivery, the compounds are typically in particulate form, which can be prepared by a variety of techniques, including spray drying, freeze drying and micronization. Aerosols may be generated by using, for example, a pressure-driven jet nebulizer or an ultrasonic nebulizer, such as using a propellant-driven metered aerosol, or propellant-free administration of micronized compounds, for example, from an inhalation capsule or other "dry powder" delivery system.

For example, the compositions of the present invention may be prepared as suspensions for nebulizer delivery, or as aerosols in liquid propellants, such as for Pressurized Metered Dose Inhalers (PMDI). Skilled personPropellants suitable for use in PMDI are known and include CFC-12, HFA-134a, HFA-227, HCFC-22 (CCl)2F2) And HFA-152 (CH)4F2And isobutane).

In some embodiments, the compositions of the present invention are in a dry powder form for delivery using a Dry Powder Inhaler (DPI). Various types of DPIs are known.

The microparticles for administration delivery may be formulated with excipients to aid delivery and release. For example, in a dry powder formulation, the microparticles may be formulated with large carrier particles that aid in flow from the DPI into the lung. Suitable carrier particles are known and include lactose particles; its mass median aerodynamic diameter may be, for example, greater than 90 μm.

In the case of aerosol-based formulations, one example is:

Figure BDA0002282880340000461

or a pharmaceutically acceptable salt thereof

The compounds of the present invention or pharmaceutically acceptable salts thereof may be administered as described depending on the inhaler system used. In addition to the compounds, the administration forms can additionally comprise excipients as described above or, for example, propellants (e.g. Frigen in the case of metered aerosols), surface-active substances, emulsifiers, stabilizers, preservatives, flavorings, fillers (e.g. lactose in the case of powder inhalers) or, where appropriate, other active compounds.

For inhalation purposes, a number of systems are available for generating and administering aerosols of optimal particle size, using inhalation techniques appropriate to the patient. Except for using adapters (gaskets, expanders) and pear-shaped containers (e.g. for use in the production of containers for ice-making) And an automatic device for projecting a spray (pump spray)

Figure BDA0002282880340000463

Furthermore, for metered aerosols, especially in the case of dry powder inhalers, a wide variety of formulations may be usedTechnical solution (for example)

Figure BDA0002282880340000464

Or an inhaler, such as described in U.S. patent No. 5,263,475, incorporated herein by reference). Furthermore, the compounds of the present invention or pharmaceutically acceptable salts thereof may be delivered in a multi-compartment device to allow delivery of the combined active agents.

The compounds or pharmaceutically acceptable salts thereof may also be administered parenterally in a sterile medium. Depending on the vehicle and concentration used, the compound may be suspended or dissolved in the vehicle. Advantageously, adjuvants such as local anesthetics, preservatives or buffers can be dissolved in the vehicle.

Targeted inhalation drug delivery

The compounds of the invention are useful for targeted inhalation delivery. Drug optimization for delivery to the lungs by local (inhalation) administration has recently been reviewed (Cooper, a.e. et al curr. drug meta-2012, 13, 457-.

Due to limitations of the delivery device, the dose of inhaled drug in humans can be low (approximately)<1 mg/day) which requires a highly effective molecule. High efficacy against a target of interest is particularly important for inhaling drugs because of factors such as the limited number of drugs that can be delivered per puff from the inhaler and safety concerns (e.g., coughing or irritation) associated with high aerosol load in the lungs. For example, in some embodiments, a Ki of about 0.5nM or less in a biochemical assay for JAK1 as described herein and an IC of about 20nM or less in a cell-based assay for JAK1 as described herein50May be desirable for inhalation JAK1 inhibitors. In other embodiments, the predicted human dose of a compound of the invention, or a pharmaceutically acceptable salt thereof, is at least 1/2 less than the predicted human dose of a compound known in the art. Thus, in some embodiments, the compounds described herein (or pharmaceutically acceptable salts thereof) exhibit the potency value.

IL13 signaling is intimately involved in asthma pathogenesis. IL13 is a cytokine that requires active JAK1 to signal. Thus, inhibition of JAK1 may also inhibit IL13 signaling, which may provide benefits to asthmatic patients. Inhibition of IL13 signaling in animal models (e.g., mouse models) may predict future benefit to human asthma patients. Therefore, inhaled JAK1 inhibitors show that inhibition of IL13 signaling may be beneficial in animal models. Methods for measuring such inhibition are known in the art. For example, as discussed herein and known in the art, JAK 1-dependent STAT6 phosphorylation is known downstream of IL13 stimulation. Thus, in some embodiments, the compounds described herein (or pharmaceutically acceptable salts thereof) exhibit inhibition of lung pSTAT6 induction. To examine the pharmacodynamic effect at the level of pSTAT6, a compound of the invention was co-administered intranasally to female Balb/c mice with 1 μ g IL 13. Immediately prior to administration, the compounds were formulated in 0.2% (v: v) Tween80 saline solution and mixed with IL 131: 1(v: v). An intranasal dose was administered to light anesthetized (isoflurane) mice by pipetting a fixed volume (50 μ L) directly into the nostrils to reach the target dose level (3mg/kg, 1mg/kg, 0.3mg/kg, 0.1 mg/kg). At 0.25 hours post-dose, blood samples (approximately 0.5mL) were collected by cardiac puncture and centrifuged (1500g, 10min, +4 ℃) to give plasma. Lungs were perfused with cold Phosphate Buffered Saline (PBS), weighed, and snap frozen in liquid nitrogen. All samples were stored at about-80 ℃ until analysis. After addition of 2mL HPLC grade water per gram of tissue, thawed lung samples were weighed and homogenized at 4 ℃ using Omni-Prep Bead raptor. Plasma and lung samples were extracted by protein precipitation with three volumes of acetonitrile containing tolbutamide (50ng/mL) and labetalol (25ng/mL) as internal analytical standards. After vortex mixing and centrifugation at 3200g and 4 ℃ for 30 minutes, the supernatant is diluted with HPLC grade water in a suitable amount (e.g., 1:1v: v) in a 96-well plate. Representative aliquots of plasma and lung samples were analyzed for parent compounds by LC-MS/MS for a series of matrix-matched calibration and quality control standards. Standards were prepared by spiking aliquots of control Balb/c mouse plasma or lung homogenate (2: 1 in HPLC grade water) with the test compound and extracting as described for the experimental samples. The lung to plasma ratio was determined as the ratio of the mean lung concentration (μ M) to the mean plasma concentration (μ M) at the sampling time (0.25 h). Theoretical target occupancy was calculated by the following equation, assuming that all drugs are in the lung tissue and the unbound fraction can interact with the target:

(unbound tissue concentration/(unbound tissue concentration + in vitro cell potency, i.e. IC)50))*100

To measure pSTAT6 levels, mouse lungs were cryopreserved at-80 ℃ until assayed and homogenized in 0.6ml of ice-cold Cell lysis buffer (Cell Signalling Technologies, catalog #9803S) supplemented with 1mm pmsf and a cocktail of protease (Sigma Aldrich, catalog # P8340) and phosphatase (Sigma Aldrich, catalog # P5726 and P0044) inhibitors. Samples were centrifuged at 16060Xg for 4min at 4 ℃ to remove tissue debris and the protein concentration of the homogenate was determined using the PierceBCA protein assay kit (catalog # 23225). Samples were diluted to a concentration of 5mg/ml in ice cold distilled water and pSTAT6 levels were determined by Meso Scale Discovery electrochemiluminescence immunoassay. Briefly, 5 μ L/well of 150 μ g/ml STAT6 capture antibody (R & D Systems, catalog # MAB 2169) was coated onto 96-well Meso-Scale Discovery high binding plates (catalog # L15XB-3) and air dried at room temperature for 5 hours. The well plate was blocked by adding 150. mu.l/well of 30mg/ml Meso Scale Discovery Block A (Cat # R93BA-4) and incubated for 2 hours at room temperature on a microplate shaker. Blocked plates were washed 4 times with Meso Scale Discovery TRIS wash buffer (catalog # R61TX-1) followed by transfer of lung homogenate at 50 μ l/well to achieve a protein loading of 250 μ g/well. The assay plates were incubated overnight at 4 ℃ and washed 4 times with TRIS wash buffer, followed by addition of 25. mu.l/well of 2.5. mu.g/ml sulfotag-labeled detection antibody pSTAT6 (BD Pharmingen, Cat #558241) for 2 hours on a microplate shaker at room temperature. Plates were washed 4 times with TRIS wash Buffer and 150. mu.l/well 1 Xmeso Scale Discovery Read Buffer T (Cat # R92TC-1) was added. The level of pSTAT6 was quantified for lung homogenates by measuring electrochemiluminescence using a Meso Scale Discovery SECTOR S600 instrument.

Selectivity between JAK1 and JAK2 may be important for inhalation of JAK1 inhibitors. For example, GMCSF (granulocyte macrophage colony stimulating factor) is a cytokine that signals exclusively through JAK 2. Neutralization of GMCSF activity was associated with alveolar proteinosis (PAP) in the lung. However, the second largest JAK2 inhibition appears to be PAP independent. Therefore, even modest JAK1 vsJAK2 selectivity may be beneficial in avoiding complete inhibition of the GMCSF pathway and avoiding PAP. For example, compounds that are approximately 2x-5x selective for JAK1 versus JAK2 may be beneficial for the uptake of JAK1 inhibitors. Thus, in some embodiments, the compounds described herein (or pharmaceutically acceptable salts thereof) exhibit such selectivity. Methods for measuring JAK1 and JAK2 selectivity are known in the art, and information can also be found in the examples herein.

In addition, it may be desirable for an inhaled JAK1 inhibitor to be selective for one or more other kinases to reduce the potential for potential toxicity from off-target kinase pathway inhibition. Therefore, it would also be beneficial for inhaled JAK1 inhibitors to be selective for a wide range of non-JAK kinases, such as in the SelectScreen available from ThermoFisher ScientificTMBiochemical kinase assay service using AdaptaTMScreening protocol in protocol determination conditions (revision of 29/7/2016), lantha ScreenTMEu kinase binding assay screening protocol and assay conditions (revision 6/7/2016) and/or Z' LYTETMScreening protocol and assay conditions (revised 2016, 9, 16). For example, a compound of the invention, or a pharmaceutically acceptable salt thereof, exhibits at least 50-fold selectivity for JAK1 over a group of non-JAK kinases. Thus, in some embodiments, the compounds described herein (or pharmaceutically acceptable salts thereof) exhibit such selectivity.

Hepatotoxicity, general cytotoxicity or cytotoxicity of unknown mechanisms are undesirable characteristics for potential drugs, including inhaled drugs. It may be beneficial for inhaled JAK1 inhibitors to have low intrinsic cytotoxicity against a variety of cell types. Typical cell types used to assess cytotoxicity include primary cells such as human hepatocytes, and proliferation-determining cell lines such as Jurkat, HEK-293, and H23. For example, an inhaled JAK1 inhibitor has an IC of greater than 50 μ Μ or greater than 100 μ Μ in a measure of cytotoxicity to the cell type50May be beneficial. Thus, in some embodiments, the compounds described herein (or pharmaceutically acceptable salts thereof) exhibit the stated values. Methods for measuring cytotoxicity are known in the art. In some implementationsIn this scheme, the compounds described herein were tested as follows:

(a) jurkat, H23 and HEK293T cells were maintained at sub-confluent density in T175 flasks. Cells were plated at 450 cells/45 μ l medium in Greiner384 well black/clear tissue culture treated plates (Greiner catalog # 781091). After dispensing the cells, the plates were equilibrated at room temperature for 30 minutes. After 30 minutes at room temperature, the cells were placed in CO2And incubated overnight at 37 ℃ in a humidity controlled incubator. The next day, cells were treated with compounds diluted in 100% DMSO (final DMSO concentration on cells ═ 0.5%) and the maximum concentration was 50 μ M at a 10-point dose-response curve. Thereafter exposing the cells and compounds to CO2And incubated overnight in a humidity controlled incubator at 37 ℃ for 72 hours. After 72 hours incubation, use

Figure BDA0002282880340000491

(Promega catalog # G7572) viability was measured for all wells. After incubation at room temperature for 20 minutes, at EnVisionTM(Perkin Elmer Life Sciences) read the plate using the light emitting mode;

(b) human primary hepatocytes: test compounds were prepared as 10mM DMSO solutions. In addition, positive controls such as chlorpromazine were made as 10mM DMSO solutions. Test compounds are typically evaluated at 2-fold dilutions using a 7-point dose-response curve. Typically, the maximum concentration tested is 50-100. mu.M. The maximum concentration is generally determined by the solubility of the compound being tested. Cryopreserved primary human hepatocytes (BiorecamationIVT) (batch IZT) were plated in InVitroGroTMHT thawing medium (BiorecamationIVT), thawing at 37 ℃, precipitating and resuspending. Hepatocyte viability was assessed by trypan blue exclusion and cells were seeded at a density of 13,000 cells/well on black-wall BioCoatTMCollagen 384-well plate (Corning BD) supplemented with 1% TorpedoTMAntibiotic cocktail (BiorecomationIVT) and InVitroGro with 5% fetal bovine serumTMCP plate medium. Cells were incubated overnight for 18 hours (37 ℃, 5% CO) before treatment2). After 18 hours of incubation, the plating medium was removed and the hepatocytes were treated with a compound diluted to contain 1% TorpedoTMAntibiotic mixtureInVitroGro of the Compound and 1% DMSO (serum-free conditions)TMHI incubation medium. Hepatocytes are treated with test compounds at concentrations of, e.g., 0.78, 1.56, 3.12, 6.25, 12.5, 25 and 50 μ M, with a final volume of 50 μ L. A positive control (e.g., chlorpromazine) is included in the assay, typically at the same concentration as the compound being tested. Additional cells were treated with 1% DMSO as vehicle control. All treatments were carried out for 48 hours (5% CO at 37 ℃)2) And each treatment condition was performed in triplicate. After 48 hours of treatment of the compound, use

Figure BDA0002282880340000501

Cell viability assay (Promega) as an end-point assay, ATP content was measured to determine cell viability. The measurements were made according to the manufacturing instructions. Using EnVisionTMLuminescence was determined by a Muliplate reader (PerkinElmer, Waltham, MA, USA). Luminescence data were normalized to vehicle (1% DMSO) control wells. Inhibition curve and IC50Estimates were generated by non-linear regression of the logarithmically transformed normalized response of inhibitor concentration (7-point serial dilutions including vehicle) against variable Hill slope, top and bottom limited to constant values of 100 and 0, respectively (GraphPad Prism)TM,GraphPad Software,La Jolla,CA,USA)。

Inhibition of the hERG (human Ether-a-go-go related gene) potassium channel may lead to long QT syndrome and arrhythmia. Although plasma levels of inhaled JAK1 inhibitor are expected to be low, pulmonary deposition of compounds that exit the lungs into the bloodstream through pulmonary absorption will circulate directly to the heart. Thus, local cardiac concentrations of inhaled JAK1 inhibitor can be transiently higher than total plasma levels, especially immediately after administration. Therefore, it may be beneficial to minimize hERG inhibition of inhaled JAK1 inhibitors. For example, in some embodiments, hERG IC is preferred50Is 30 times the Cmax of free drug plasma. Thus, in some embodiments, the compounds of the invention (or pharmaceutically acceptable salts thereof) exhibit minimized hERG inhibition in conditions such as:

(a) in vitro effect of compounds on hERG expressed in mammalian cells was examined using hERG 2pt automated patch clamp conditions, and allowed to stand at room temperatureUsing QPatch

Figure BDA0002282880340000502

(Sophion Bioscience A/S, Denmark), an automated parallel patch clamp system. In some cases, the compounds are tested at only one or two concentrations, such as 1 or 10 μ M. In other cases, a broader concentration response relationship is established to allow IC50And (6) evaluating. For example, the concentration of test compound is selected to span an inhibition range of about 10-90% in half log increments. Each test article concentration is measured in two or more cells (n.gtoreq.2). The exposure time for each test article concentration is at least 3 minutes; and/or

(b) Those described in the examples of WO 2014/074775, under "Effect on closed hERG Potasinterstitial Channels Expressed in Mammalian Cells", ChanTestTMCharles River Company, protocol with the following variations: cells stably expressing hERG were maintained at-80 mV. The initial and steady state inhibition of hERG potassium current due to the compound was measured using a pulse mode of fixed amplitude (pre-pulse was adjusted: +20mV for 1 second; repolarization test ramp to)90mV (-0.5V/s), repeated at 5 second intervals). Each recording was concluded with the final application of a super-concentrated reference substance E-4021(500nM) (Charles River, Inc.). The remaining uninhibited current was subtracted off-line from the data to determine the hERG inhibitory potency of the test substance.

CYP (cytochrome P450) inhibition may not be a desirable trait for the uptake of JAK1 inhibitors. For example, reversible or time-dependent CYP inhibitors may cause an undesirable increase in their own plasma levels or plasma levels of other co-administered drugs (drug interactions). Furthermore, time-dependent CYP inhibition is sometimes caused by the biotransformation of the parent drug to reactive metabolites. The reactive metabolites may covalently modify the protein, potentially leading to toxicity. Therefore, minimizing reversible and time-dependent CYP inhibition may be beneficial for inhalation of JAK1 inhibitors. Thus, in some embodiments, the compounds of the present invention (or pharmaceutically acceptable salts thereof) exhibit minimal or no reversible and/or time-dependent CYP inhibition. Methods of measuring CYP inhibition are known in the art. CYP inhibition of the compounds described herein was assessed using pooled (n 150) human liver microsomes (Corning, Tewksbury, MA) in the concentration range of 0.16-10 μ M using the previously reported method (haladay et al, Drug metal. lett.2011,5,220-. The incubation time and protein concentration are based on the CYP isoform (isoform) and the probe substrate/metabolite analyzed. The following substrates/metabolites and incubation times and protein concentrations for each CYP were used: CYP1A2, phenacetin/acetaminophen, 30 minutes, 0.03mg/ml protein; CYP2C9, warfarin/7-hydroxy warfarin, 30 min, 0.2mg/ml protein; CYP2C19, metrafenone/4-hydroxymetrafenone, 40 min, 0.2mg/ml protein; CYP2D6, dextromethorphan/Dextrorphan (dextrorhan), 10min, 0.03mg/ml protein; CYP3a4, midazolam/1-hydroxymidazolam, 10 minutes, 0.03mg/ml protein and CYP3a4 testosterone/6 β -hydroxytestosterone, 10 minutes, 0.06mg/ml protein. These conditions have previously been determined to be linear rates of CYP-specific metabolite formation. All reactions were initiated with 1mM NADPH and stopped by the addition of a 0.1% formic acid in acetonitrile containing the appropriate stable labeled internal standard. Samples were analyzed by LC-MS/MS.

For compounds intended to be delivered by dry powder inhalation, it is also desirable to be able to produce crystalline forms of the compound that can be micronized to 1-5 μm. Particle size is an important determinant of pulmonary deposition of inhaled compounds. Particles less than 5 micrometers (μm) in diameter are generally defined as respirable. Particles larger than 5 μm in diameter are more likely to deposit in the oropharynx and correspondingly less likely to deposit in the lungs. Furthermore, fine particles less than 1 μm in diameter are more likely to remain suspended in air than large particles and are correspondingly more likely to be exhaled from the lungs. Thus, for inhaled drugs with a site of action in the lung, a particle diameter of 1-5 μm may be beneficial. Typical methods for measuring particle size include laser diffraction and cascade impaction (cascade impaction). Typical values for defining particle size include:

d10, D50 and D90. These are measures of particle diameter, indicating that 10%, 50% or 90% of the samples are below this value, respectively. For example, a D50 of 3 μm indicates that 50% of the sample is less than 3 μm in size.

Mass Mean Aerodynamic Diameter (MMAD). MMAD is the diameter where 50% of the particles are larger and 50% smaller by mass. MMAD is an index that measures central tendency.

Geometric Standard Deviation (GSD). GSD is a measure of the size of the dispersion of MMAD, or a measure of the aerodynamic particle size distribution spread.

Common formulations for inhaled pharmaceuticals are dry powder formulations comprising the Active Pharmaceutical Ingredient (API) in admixture with a carrier, such as lactose, with or without additional additives such as magnesium stearate. For these or other formulations, it may be beneficial for the API itself to have properties that allow it to be ground to an inhalable particle size of 1-5 μm. Agglomeration of particles should be avoided and can be measured by methods known in the art, such as examining the D90 values under different pressure conditions. Accordingly, in some embodiments, the compounds of the present invention (or pharmaceutically acceptable salts thereof) may be prepared with such respirable particle sizes, with little or no agglomeration.

With respect to crystallinity, for some inhaled pharmaceutical formulations, including lactose blends, it is important to use a particular crystalline form of the API. Crystallinity and crystalline form may affect many parameters associated with inhaled drugs including, but not limited to: chemical and aerodynamic stability over time, compatibility with inhaled formulation ingredients such as lactose, hygroscopicity, lung retention, and lung irritation. Thus, a stably reproducible crystalline form may be beneficial for inhalation of a drug. Furthermore, the techniques used to grind the compound to the desired particle size are generally energetic and may cause the low melting crystalline form to convert to other crystalline forms, or to a fully or partially amorphous form. Crystalline forms with melting points below 150 ℃ may be incompatible with milling, while crystalline forms with melting points below 100 ℃ may be incompatible with milling. It may therefore be beneficial for the inhaled drug to have a melting point at least above 100 ℃ and ideally above 150 ℃. Accordingly, in some embodiments, the compounds herein (or pharmaceutically acceptable salts thereof) exhibit such properties.

In addition, minimizing molecular weight may help reduce the effective dose of inhaled JAK1 inhibitor. Lower molecular weights result in correspondingly higher numbers of molecules of the Active Pharmaceutical Ingredient (API) per unit mass. It may therefore be beneficial to find a minimum molecular weight for an inhaled JAK1 inhibitor that retains all other desirable properties of the inhaled drug.

Finally, the compound needs to be maintained in sufficient concentration in the lung for a given period of time to be able to exert a pharmacological effect of the desired duration, and should have low systemic exposure to the pharmacological target that does not require systemic inhibition of that target. The lung has an inherently high permeability to large molecules (proteins, peptides) as well as small molecules with a concurrent short lung half-life, and therefore it may be necessary to reduce the lung absorption rate by modifying one or more properties of the compound: minimizing membrane permeability, increasing pKa, increasing cLogP, decreasing solubility, decreasing dissolution rate, or introducing a degree of basicity to the compound (e.g., introducing an amine) to promote binding to phospholipid-rich lung tissue, or by entrapment in acidic subcellular compartments such as lysosomes (pH 5). Methods of measuring such properties are known in the art.

Thus, in some embodiments, a compound of the invention (or a pharmaceutically acceptable salt thereof) exhibits one or more of the above-described properties. Furthermore, in some embodiments, the compounds of the present invention advantageously exhibit one or more of the described properties relative to compounds known in the art, particularly with respect to inhalation drugs as compared to compounds of the art intended as oral drugs. For example, compounds with rapid oral absorption are often difficult to retain in the lungs upon inhalation.

Treatment method and application of Janus kinase inhibitor

The compounds of the invention, or pharmaceutically acceptable salts thereof, inhibit the activity of Janus kinases such as JAK1 kinase. For example, the compounds or pharmaceutically acceptable salts thereof inhibit JAK1 kinase-induced phosphorylation of Signal Transducers and Activators of Transcription (STATs) and STAT-mediated cytokine production. The compounds of the invention may be used to inhibit JAK1 kinase activity in cells via a cytokine pathway, such as the IL-6, IL-15, IL-7, IL-2, IL-4, IL-9, IL-10, IL-13, IL-21, G-CSF, IFN α, IFN β or IFN γ pathway. Thus, in one embodiment, a method is provided for contacting a cell with a compound of the invention, or a pharmaceutically acceptable salt thereof, to inhibit Janus kinase activity (e.g., JAK1 activity) in the cell.

The compounds are useful for treating immunological disorders caused by aberrant IL-6, IL-15, IL-7, IL-2, IL-4, IL9, IL-10, IL-13, IL-21, G-CSF, IFN α, IFN β or IFN γ cytokine signaling.

Accordingly, one embodiment includes a compound of the present invention, or a pharmaceutically acceptable salt thereof, for use in therapy.

In some embodiments, there is provided the use of a compound of the invention, or a pharmaceutically acceptable salt thereof, in the treatment of an inflammatory disease. Further provided is the use of a compound of the invention, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of an inflammatory disease such as asthma. Also provided are compounds of the invention, or pharmaceutically acceptable salts thereof, for use in the treatment of inflammatory diseases such as asthma.

Another embodiment includes a method of preventing, treating or lessening the severity of a disease or condition in a patient that is responsive to inhibition of Janus kinase activity, such as JAK1 kinase activity, such as asthma. The method may comprise the step of administering to the patient a therapeutically effective amount of a compound of the present invention, or a pharmaceutically acceptable salt thereof. In one embodiment, the disease or disorder responsive to inhibition of a Janus kinase, such as JAK1 kinase, is asthma.

In one embodiment, the disease or disorder is cancer, stroke, diabetes, hepatomegaly, cardiovascular disease, multiple sclerosis, alzheimer's disease, cystic fibrosis, viral disease, autoimmune disease, atherosclerosis, restenosis, psoriasis, rheumatoid arthritis, inflammatory bowel disease, asthma, allergic diseases, inflammation, nervous system disease, hormone-related disease, organ transplant-related disorder (e.g., transplant rejection), immunodeficiency disorder, destructive bone disorder, proliferative disorder, infectious disease, cell death-related disorder, thrombin-induced platelet aggregation, liver disease, pathological immune disorder involving T cell activation, CNS disorder, or myeloproliferative disorder.

In one embodiment, the inflammatory disease is rheumatoid arthritis, psoriasis, asthma, inflammatory bowel disease, contact dermatitis, or delayed type hypersensitivity. In one embodiment, the autoimmune disease is rheumatoid arthritis, lupus or multiple sclerosis.

In another embodiment, a compound of the invention, or a pharmaceutically acceptable salt thereof, can be used to treat a pulmonary disease such as fibrotic lung disease or an interstitial lung disease (e.g., interstitial pneumonia). In some embodiments, a compound of the invention, or a pharmaceutically acceptable salt thereof, can be used to treat Idiopathic Pulmonary Fibrosis (IPF), systemic sclerosis interstitial lung disease (SSc-ILD), non-specific interstitial pneumonia (NSIP), rheumatoid arthritis-associated interstitial lung disease (RA-ILD), sarcoidosis, hypersensitivity pneumonitis, or an ILD secondary to a connective tissue disease other than scleroderma, such as polymyositis, dermatomyositis, rheumatoid arthritis, Systemic Lupus Erythematosus (SLE), or a mixed connective tissue disease.

In one embodiment, the cancer is breast cancer, ovarian cancer, cervical cancer, prostate cancer, testicular cancer, penile cancer, genitourinary tract cancer, seminoma, esophageal cancer, laryngeal cancer, gastric cancer (gasteric), gastric cancer (stomach), gastrointestinal cancer, skin cancer, keratoacanthoma (keratoacanthoma), follicular cancer, melanoma, lung cancer, small cell lung cancer, non-small cell cancer (NSCLC), lung adenocarcinoma, lung squamous cancer, colon cancer, pancreatic cancer, thyroid cancer, papillary cancer (papillary), bladder cancer, liver cancer, biliary tract cancer, kidney (kidney) cancer, bone cancer, myeloid disorder (myeloid disorder), lymphoid disorder (lymphodidorder), hairy cell cancer, oral and pharyngeal (oral) cancer, lip cancer, tongue cancer, mouth (mouth) cancer, salivary gland cancer, pharyngeal cancer, small intestine cancer, colon cancer, rectal cancer, anal cancer, renal (renal) cancer, prostate cancer, thyroid cancer, large intestine cancer, vulval cancer, or a combination thereof, Endometrial cancer, uterine cancer, brain cancer, central nervous system cancer, peritoneal cancer, hepatocellular cancer, head cancer, neck cancer, Hodgkin's disease, or leukemia.

In one embodiment, the disease is a myeloproliferative disorder. In one embodiment, the myeloproliferative disorder is polycythemia vera, essential thrombocythemia, myelofibrosis or Chronic Myelogenous Leukemia (CML).

Another embodiment includes the use of a compound of the present invention, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a disease described herein (e.g., an inflammatory disorder, an immune disorder, or cancer). In one embodiment, the invention provides methods of treating a disease or disorder as described herein, e.g., an inflammatory disorder, an immune disorder, or cancer, by targeted inhibition of a JAK kinase, such as JAK 1.

Combination therapy

The compounds may be administered alone or in combination with other active agents for therapy. The second or additional (e.g., third) compound of a pharmaceutical composition or dosage regimen will generally have complementary activities to the compounds of the present invention such that they do not adversely affect each other. Such active agents are suitably present in combination in an effective amount to achieve the intended purpose. The compounds may be co-administered or administered separately in a single pharmaceutical composition, which when administered separately may be simultaneous or sequential. The sequential administration may be close in time or remote in time.

For example, other compounds may be used in combination with the compounds of the present invention or pharmaceutically acceptable salts thereof for the prevention or treatment of inflammatory diseases such as asthma. Suitable therapeutic agents for combination therapy include, but are not limited to: adenosine A2A receptor antagonists; an anti-infective agent; a non-steroidal glucocorticoid receptor (GR receptor) agonist; an antioxidant; a β 2 adrenoceptor agonist; CCR1 antagonists; chemokine antagonists (non-CCR 1); a corticosteroid; CRTh2 antagonists; DP1 antagonists; formyl peptide receptor antagonists; a histone deacetylase activator; a chloride channel hCLCA1 blocker; epithelial sodium channel blockers (ENAC blockers; intercellular adhesion molecule 1 blockers (ICAM blockers); IKK2 inhibitors; JNK inhibitors; transient receptor potential ankyrin 1(TRPA1) inhibitors; Bruton's Tyrosine Kinase (BTK) inhibitors (e.g. non-nintinib (fennebruntinib)); spleen tyrosine kinase (SYK) inhibitors; trypsin-like-beta antibodies; ST2 receptor antibodies (e.g. AMG282), cyclooxygenase inhibitors (COX inhibitors), lipoxygenase inhibitors; leukotriene receptor antagonists; dual beta 2-adrenoceptor agonists/M3 receptor antagonists (MABA compounds); MEK-1 inhibitors; myeloperoxidase inhibitors (MPO inhibitors), muscarinic antagonists; p38MAPK inhibitors; phosphodiesterase PDE4 inhibitors; phosphatidylinositol 3-kinase delta inhibitors (PI 3-kinase delta inhibitors); phosphatidylinositol 3-kinase gamma inhibitors (PI 3-gamma kinase inhibitors); peroxidizes An agonist of the enzyme proliferator-activated receptor (PPAR γ agonist); a protease inhibitor; retinoic acid receptor modulators (RAR γ modulators); a statin drug; a thromboxane antagonist; TLR7 receptor agonists; or a vasodilator.

Furthermore, a compound of the present invention or a pharmaceutically acceptable salt thereof may be combined with: (1) corticosteroids such as alclometasone dipropionate, almometasone (amelomeasone), beclomethasone dipropionate, budesonide propionate (butixocort propionate), bisopronide (biclonide), clobetasol propionate, ciclesonide de-isobutyrate (dessobutyryl ciclesonide), dexamethasone, eprinondione dihydrochloride (etiprenoldicloacetate), fluocinonide, fluticasone furoate, fluticasone propionate, loteprednol (local) or mometasone furoate; (2) beta 2-adrenoceptor agonists such as salbutamol, albuterol, terbutaline, fenoterol, bitolterol, carbbuterol, clenbuterol, pirbuterol, ritodrol, rimoterol, terbutaline, troquinol, tulobuterol, and long-acting beta 2-adrenoceptor agonists such as metaproterenol, isoproterenol, salmeterol, indacaterol, formoterol (including formoterol fumarate), arformoterol, carmofibrinole, carpoterol, acloterol, aclidinol, aclatorol, aclidinoterol, albuterol acetate, or lanoteronate; (3) corticosteroid/long-acting beta 2 agonist combinations, e.g. salmeterol/fluticasone propionate (f)

Figure BDA0002282880340000551

Also can be used forSold), formoterol/budesonide

Figure BDA0002282880340000553

Formoterol/fluticasone propionate

Figure BDA0002282880340000554

Formoterol/ciclesonide, formoterol/mometasone furoate, indacaterol/mometasone furoate, vilanterol triphenacetate/fluticasone furoate (BREO elipta), or arformoterol/ciclesonide; (4) anticholinergics, for example muscarinic-3 (M3) receptor antagonists such as ipratropium bromide, tiotropium bromide, aclidinium bromide (LAS-34273), glycopyrronium bromide or umeclidinium bromide; (5) m3-anticholinergic/beta 2-adrenoceptor agonist combinations, e.g. vilanterol/umeclidinium bromide Oldaterol/tiotropium bromide, glycopyrronium bromide/indacaterol: (

Figure BDA0002282880340000563

Also can be used for

Figure BDA0002282880340000564

Sold), fenoterol hydrobromide/ipratropium bromide

Figure BDA0002282880340000565

Salbutamol sulfate/ipratropium bromide

Figure BDA0002282880340000566

Formoterol fumarate/glycopyrronium bromide or aclidinium bromide/formoterol; (6) dual pharmacological M3-anticholinergic/beta 2-adrenoceptor agonists, e.g. batefentrol succinate, AZD-2115 or LAS-190792; (7) leukotriene modulators, e.g., leukotriene antagonists such as montelukast, zafirlukast or pranlukast (pranlukast), or leukotriene biosynthesis inhibitors such as zileuton, or LTB4 antagonists such as ameluban, or FLAP inhibitors such as fiboflapon, GSK-2190915; (8) phosphodiesterase-IV (PDE-IV) inhibitors (oral or inhaled), such as roflumilast (roflumilast), cilomilast (cilomilast), ogermorite (oglemilast), rolipram (rolipram), tetomilast (tetomilast), AVE-8112, revamilast, CHF 6001; (9) antihistamines, for example selective histamine-1 (H1) receptor antagonists such as fexofenadine, cetirizine, loratadine, or astemizole, or dual H1/H3 receptor antagonists such as GSK 835726 or GSK 1004723; (10) antitussives, such as codeine or dextromethorphan (dextromethorphan); (11) mucolytics, such as N-acetylcysteine or fudostein; (12) expectorants/mucus actuation modulators (mucokineticimodulators), such as ambroxol (ambroxol), hypertonic solutions (e.g. saline or mannitol), or surfactants; (13) mucolytic peptides, such as recombinant human deoxyribonuclease I (streptodornase-alpha and rhDNase) or Spiromycins; (14) antibiotics, such as azithromycin, tobramycin or aztreonam; (15) non-selective COX-1/COX-2 inhibitors, such as ibuprofen or ketoprofen; (16) COX-2 inhibitors, such as celecoxib and rofecoxib; (17) VLA-4 antagonists, such as those described in WO 97/03094 and WO 97/02289, both incorporated herein by reference; (18) TACE inhibitors and TNF-alpha inhibitors, e.g. anti-TNF monoclonal antibodies, e.g.And CDP-870, and TNF receptor immunoglobulin molecules, e.g.

Figure BDA0002282880340000568

(19) Matrix metalloproteinase inhibitors, such as MMP-12; (20) human neutrophil elastase inhibitors, such as those described in BAY-85-8501 or WO 2005/026124, WO 2003/053930 and WO 2006/082412, all of which are incorporated herein by reference; (21) a2b antagonists such as those described in WO 2002/42298 toIncorporated herein by reference; (22) modulators of chemokine receptor function, such as antagonists of CCR3 and CCR 8; (23) compounds which modulate other prostanoid receptors, e.g. thromboxane A2An antagonist; DP1 antagonists such as laropiprant (laropiprant) or asapirant, CRTH2 antagonists such as OC000459, non-veppiren (fevipiprant), ADC 3680 or ARRY 502; (24) PPAR agonists include PPAR α agonists (such as fenofibrate), PPAR δ agonists, PPAR γ agonists such as pioglitazone, rosiglitazone and balaglitazone (balaglitazone); (25) methylxanthines such as theophylline or aminophylline, and methylxanthine/corticosteroid combinations such as theophylline/budesonide, theophylline/fluticasone propionate, theophylline/ciclesonide, theophylline/mometasone furoate and theophylline/beclomethasone dipropionate; (26) a2a agonists such as those described in EP1052264 and EP 1241176; (27) CXCR2 or IL-8 antagonists such as AZD-5069, AZD-4721 or danirixin; (28) IL-R signal modulators such as anakinra (kineret) and ACZ 885; (29) MCP-1 antagonists such as ABN-912; (30) p38MAPK inhibitors such as BCT197, JNJ49095397, loshapimod or PH-797804; (31) TLR7 receptor agonists such as AZD 8848; (32) PI 3-kinase inhibitors such as RV1729 or GSK2269557 (nemiralisib); (33) triple combination products such as TRELEGY ELLIPTA (fluticasone furoate, umeclidinium bromide and vilanterol); or (34) a small molecule inhibitor of TRPA1, BTK or SYK.

In some embodiments, a compound of the present invention, or a pharmaceutically acceptable salt thereof, may be used in combination with one or more other drugs, such as an anti-hyperproliferative agent, an anti-cancer agent, a cytostatic agent, a cytotoxic agent, an anti-inflammatory agent, or a chemotherapeutic agent, such as those active agents disclosed in U.S. published application No. 2010/0048557, which is incorporated herein by reference. The compounds of the present invention or pharmaceutically acceptable salts thereof may also be used in combination with radiation therapy or surgery, as is known in the art.

Combinations of any of the foregoing with a compound of the present invention or a pharmaceutically acceptable salt thereof are specifically contemplated.

Article of manufacture

Another embodiment includes an article of manufacture (e.g., a kit) for treating a disease or disorder responsive to the inhibition of a Janus kinase, such as JAK1 kinase. The kit may comprise:

(a) a first pharmaceutical composition comprising a compound of the invention or a pharmaceutically acceptable salt thereof; and

(b) and (5) instructions for use.

In another embodiment, the kit further comprises:

(c) a second pharmaceutical composition, such as a pharmaceutical composition, comprises an agent for use in the aforementioned treatment, such as an agent for use in the treatment of an inflammatory disease, or a chemotherapeutic agent.

In one embodiment, the instructions describe administering said first and second pharmaceutical compositions to a patient in need thereof simultaneously, sequentially or separately.

In one embodiment, the first and second compositions are contained in separate containers. In another embodiment, the first and second compositions are contained in the same container.

Containers used include, for example, bottles, vials, syringes, blister packs, and the like. The container may be formed from a variety of materials, such as glass or plastic. The container comprises a compound of the invention or a pharmaceutically acceptable salt thereof, which is effective for treating the condition and may have a sterile access port (e.g., the container may be an intravenous bag or a vial having a stopper pierceable by a hypodermic injection needle). The label or package insert indicates that the compound is useful for treating a selected disease, such as asthma or cancer. In one embodiment, the label or package insert indicates that the compound is useful for treating a disorder. In addition, the label or package insert may indicate that the patient to be treated suffers from a disorder characterized by overactive or irregular Janus kinase activity, such as overactive or irregular JAK1 activity. The label or package insert may also indicate that the compound is useful for treating other disorders.

Alternatively, or in addition, the kit may further comprise a second (or third) container comprising a pharmaceutically acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate buffered saline, Ringer's solution, or dextrose solution. It may further include other materials as desired from a commercial and user standpoint, including other buffers, diluents, filters, needles and syringes.

The following examples are included to illustrate the invention. It should be understood, however, that these examples are not limiting and are intended merely to suggest a method of practicing the invention. One skilled in the art will recognize that the above chemical reactions can be readily adapted to prepare some of the other compounds of the present invention, and that alternative methods of preparing the compounds of the present invention are considered to be within the scope of the present invention. For example, the synthesis of non-exemplified compounds according to the invention may be successfully carried out by modifications apparent to those skilled in the art, such as appropriate protection of interfering groups, the use of other suitable reagents known in the art in addition to those described herein, or routine modification of reaction conditions. Alternatively, other reactions disclosed herein or known in the art are deemed to have applicability for the preparation of other compounds of the invention.

Examples

General experimental details

All solvents and commercial reagents were used as received unless otherwise indicated. When the product is purified by chromatography on silica gel, a glass column (Kieselgel 60, 220-440 mesh, 35-75 μm) packed manually on silica gel is used or

Figure BDA0002282880340000582

SPE Si II column. By 'Isolute SPE Si column' is meant a pre-packed polypropylene chromatography column comprising unbound active silica and a nominal porosity with an average size of 50 μm

Figure BDA0002282880340000585

The irregular particle of (4). When in use

Figure BDA0002282880340000583

SCX-2 column time'

Figure BDA0002282880340000584

SCX-2 column' means a pre-packed polypropylene chromatography column comprising a non-blocked propane sulfonic acid functionalized silica gel strong cation exchange adsorbent.

LCMS conditions

Method A

The experiment was performed on a SHIMADZU LCMS-2020 equipped C18 reverse phase column (50X3mm Shim-Pack XR-ODS, 2.2 μm particle size) eluting solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Figure BDA0002282880340000581

Figure BDA0002282880340000591

detection-UV (220 and 254nm) and ELSD

Method B

The experiment was performed on a SHIMADZU LCMS-2020 equipped C18 reverse phase column (50X3mm Shim-Pack XR-ODS, 2.2 μm particle size) eluting solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Figure BDA0002282880340000592

detection-UV (220 and 254nm) and ELSD

Method C

The experiment was performed on a SHIMADZU LCMS-2020 equipped C18 reverse phase column (50X3mm Shim-Pack XR-ODS, 2.2 μm particle size) eluting solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Figure BDA0002282880340000593

detection-UV (220 and 254nm) and ELSD

Method D

The experiment was performed on a SHIMADZU LCMS-2020 equipped C18 reverse phase column (50X3mm Shim-Pack XR-ODS, 2.2 μm particle size) eluting solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Figure BDA0002282880340000594

Figure BDA0002282880340000601

detection-UV (220 and 254nm) and ELSD

Method E

The experiment was performed on a SHIMADZU LCMS-2020 equipped C18 reverse phase column (50X3mm Shim-Pack XR-ODS, 2.2 μm particle size) eluting solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Figure BDA0002282880340000602

detection-UV (220 and 254nm) and ELSD

Method F

The experiment was performed on a SHIMADZU LCMS-2020 equipped C18 reverse phase column (50X3mm Shim-Pack XR-ODS, 2.2 μm particle size) eluting solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Figure BDA0002282880340000603

detection-UV (220 and 254nm) and ELSD

Method G

The experiment was performed on a SHIMADZU 20A HPLC equipped with a C18 reverse phase column (50 × 2.1mm Ascentis Express C18, 2.7 μm particle size), eluting solvent a: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Figure BDA0002282880340000611

detection-UV (220 and 254nm) and ELSD

Method H

The experiment was performed on a SHIMADZU LCMS-2020 equipped C18 reverse phase column (50X3mm Shim-Pack XR-ODS, 2.2 μm particle size) eluting solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Figure BDA0002282880340000612

detection-UV (220 and 254nm) and ELSD

Method I

The experiment was performed on a SHIMADZU 20A HPLC equipped with a Poroshell HPH-C18 column (50x3mm, 2.7 μm particle size) eluting solvent a: water/5 mM NH4HCO3(ii) a Solvent B: and (3) acetonitrile. Gradient:

Figure BDA0002282880340000613

detection-UV (220 and 254nm) and ELSD

Method J

The experiment was carried out on a ShimadZU LCMS-2020 equipped with a C18 reverse phase column (50X3mm Kinetex XB-C18, 2.6 μm particle size) eluting solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Figure BDA0002282880340000614

Figure BDA0002282880340000621

detection-UV (220 and 254nm) and ELSD

Method K

The experiment was performed on a SHIMADZU LCMS-2020 equipped C18 reverse phase column (50X3mm Shim-Pack XR-ODS, 2.2 μm particle size) eluting solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Figure BDA0002282880340000622

detection-UV (220 and 254nm) and ELSD

Method L

Equipped with a C18 reverse phase column (50x2.1mm Kinetex XB-C)18100A, 2.6 μm particle size) was run on SHIMADZU LCMS-2020 eluting solvent a: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Figure BDA0002282880340000623

detection-UV (220 and 254nm) and ELSD

List of common abbreviations

ACN acetonitrile

Brine saturated aqueous sodium chloride solution

CH3OD deuterated methanol

CDCl3Deuterated chloroform

DCM dichloromethane

DIEA or DIPEA diisopropylethylamine

DMA dimethyl acetamide

DMAP 4-dimethylaminopyridine

DMF dimethyl formamide

DMSO dimethyl sulfoxide

DMSO-d6 deuterated dimethyl sulfoxide

EDC or EDCI 1-ethyl-3- (3-dimethylaminopropyl) carbodiimide

EtOAc ethyl acetate

EtOH ethanol

FA formic acid

HOAc acetic acid

g

h hours

HATU (O- (7-azabenzotriazol-1-yl) -N, N, N ', N' -tetramethyluronium hexafluorophosphate)

HCl hydrochloric acid

HOBt hydroxybenzotriazole

HPLC high performance liquid chromatography

IMS Industrial methylated spirits (Industrial methylated spirits)

L liter

LCMS liquid chromatography-mass spectrometry

LiHMDS or LHMDS lithium hexamethyldisilazide (lithium hexamethyldisilazide)

MDAP quality-oriented automatic purification

MeCN acetonitrile

MeOH methanol

min for

mg of

mL of

NMR nuclear magnetic resonance spectrum

Pd2(dba)3.CHCl3Tris (dibenzylideneacetone) dipalladium (0) -chloroform adduct

PE Petroleum Ether

Prep-HPLC preparative high performance liquid chromatography

SCX-2 Strong cation exchange

TBAF tetra-n-butylammonium fluoride

THF tetrahydrofuran

TFA trifluoroacetic acid

Xantphos 4, 5-bis (diphenylphosphino) -9, 9-dimethylxanthene

Intermediate 1

Figure BDA0002282880340000641

N- (5- (5-bromo-2- (difluoromethoxy) phenyl) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazol-4-yl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Step 1: synthesis of 4-bromo-1- (difluoromethoxy) -2-iodobenzene

Figure BDA0002282880340000642

To a solution of 4-bromo-2-iodophenol (282g, 943mmol) in N, N-dimethylformamide (2000mL) and water (500mL) were added sodium 2-chloro-2, 2-difluoroacetate (216g, 1.42mol) and Cs2CO3(617g, 1.89 mol). The reaction vessel is equipped with a device for releasing CO2Is provided. The resulting mixture was stirred at 120 ℃ overnight, allowed to cool to room temperature and poured into ice water (3000 mL). The resulting solution was extracted with ethyl acetate (3 × 1500mL) and the organic layers were combined. The ethyl acetate extract was washed with brine (1000mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with ethyl acetate/petroleum ether (1/10) to give 300g (91%) of 4-bromo-1- (difluoromethoxy) -2-iodobenzene as a pale yellow oil.1H NMR(300MHz,CDCl3)δ7.96(dd,J=5.7Hz,2.4Hz,1H),7.45(dd,J=8.7Hz,2.4Hz,1H),7.03(d,J=8.7Hz,1H),6.39(t,J=72.9Hz,1H)。

Step 2: synthesis of 5- [ 5-bromo-2- (difluoromethoxy) phenyl ] -4-nitro-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrazole

Figure BDA0002282880340000651

To 4-nitro-1- [ [2- (trimethylsilyl) ethoxy ] at-70 ℃ under nitrogen with stirring]Methyl radical]To a solution of-1H-pyrazole (100g, 411mmol) in anhydrous THF (1000mL) was added dropwise a LiHMDS solution (490mL, 1.0mol/L in THF). The resulting solution was stirred at-50 ℃ for 1 hour and then cooled to-70 ℃. Reacting ZnCl2(500mL, 0.7mol/L THF solution) was added dropwise at-70 ℃. The resulting solution was allowed to warm to room temperature and stirred at room temperature for 1 hour. To the mixture was added 4-bromo-1- (difluoromethoxy) -2-iodobenzene (150g, 860mmol), Pd (PPh)3)4(24.0g, 20.8 mmol). The resulting solution was heated at reflux temperature overnight, allowed to cool to room temperature and concentrated under reduced pressure. The reaction was repeated once at this scale and the two resulting crude products were combined for purification. Removing residuesThe residue was purified by flash chromatography on silica eluting with ethyl acetate/petroleum ether (1/20). The appropriate fractions were combined and concentrated under reduced pressure. This gave a total of 300g (79%) of 5- [ 5-bromo-2- (difluoromethoxy) phenyl]-4-nitro-1- [ [2- (trimethylsilyl) ethoxy ] ethanol]Methyl radical]-1H-pyrazole as a pale yellow solid.1H NMR(300MHz,CDCl3)δ8.27(s,1H),7.68(dd,J=8.7,2.4Hz,1H),7.62(d,J=2.4Hz,1H),7.19(d,J=8.4Hz,1H),6.39(t,J=72.5Hz,1H),5.44–5.19(m,2H),3.72–3.54(m,2H),0.94–0.89(m,2H),0.02(s,9H)。

And step 3: synthesis of 5- (5-bromo-2- (difluoromethoxy) phenyl) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazol-4-amine

Figure BDA0002282880340000652

To a solution of 5- (5-bromo-2- (difluoromethoxy) phenyl) -4-nitro-1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazole (50.1g, 108mmol) in ethanol (2000mL) and water (200mL) were added iron powder (60.1g, 1.07mol) and NH4Cl (28.0g, 0.523 mol). The reaction mixture was stirred at reflux temperature under nitrogen for 3 hours. The solid was filtered off and washed with ethanol (100 mL). The filtrate was concentrated under reduced pressure. The residue was dissolved in 3000mL of ethyl acetate. The ethyl acetate solution was washed with 1x500mL brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give 50.1g of crude 5- (5-bromo-2- (difluoromethoxy) phenyl) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazol-4-amine as a yellow oil. The crude product was used in the next step without purification. LC/MS (method G, ESI): [ M + H ]]+=434.2,RT=0.93min。

And 4, step 4: synthesis of N- (5- (5-bromo-2- (difluoromethoxy) phenyl) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazol-4-yl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Figure BDA0002282880340000661

To 5- (5-bromo-2- (difluoromethoxy) phenyl) -1- ((2-, (Trimethylsilyl) ethoxy) methyl) -1H-pyrazol-4-amine (50.1g, 115mmol) in DMA (1500mL) was added pyrazolo [1,5-a]Pyrimidine-3-carboxylic acid (32.1g, 196.0mmol), PyAOP (102g, 196mmol), DMAP (1.41g, 11.0mmol), and DIPEA (44.1g, 0.341 mol). The resulting solution was stirred in a 60 ℃ oil bath for 3 hours and then allowed to cool to room temperature. The reaction mixture was then partitioned between water/ice (2000mL) and ethyl acetate (2000 mL). The aqueous phase was extracted with ethyl acetate (2 ×). The organic layers were combined, washed with brine (1000mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica eluting with ethyl acetate/petroleum ether (4: 1). The appropriate fractions were combined and concentrated under reduced pressure. To the residue was added water (150mL) and the mixture was stirred in water at room temperature for 1 hour. The solid was collected by filtration and air-dried to give 60.1g (91%) of N- (5- (5-bromo-2- (difluoromethoxy) phenyl) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazol-4-yl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide, as a pale yellow solid. LC/MS (method G, ESI): [ M + H ]]+=579.1&581.1,RT=1.10min.1H NMR(300MHz,CDCl3)δ9.62(s,1H),8.80(dd,J=6.9,1.7Hz,1H),8.73(s,1H),8.53(dd,J=4.2,1.7Hz,1H),8.38(s,1H),7.79(d,J=2.4Hz,1H),7.67(dd,J=8.8,2.5Hz,1H),7.29(d,J=1.4Hz,1H),7.00(dd,J=6.9,4.2Hz,1H),6.43(t,J=72.6Hz,1H),5.53-5.27(m,2H),3.73-3.50(m,2H),0.88(ddd,J=9.5,6.4,4.4Hz,2H),0.00(s,9H).

Intermediate 2

Figure BDA0002282880340000662

N- [3- [ 5-bromo-2- (difluoromethoxy) phenyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Reacting N- [5- [ 5-bromo-2- (difluoromethoxy) phenyl]-1- [ [2- (trimethylsilyl) ethoxy ] group]Methyl radical]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]Pyrimidine-3-carboxamide (intermediate 1, 5.00g, 8.63mmol) was treated with HCl/dioxane (150mL, 4M) at room temperature overnight. The resulting mixture was concentrated under reduced pressure. To obtain 3.80g N- [3- [ 5-bromo-2- (difluoromethoxy) phenyl group]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]Pyrimidine-3-carboxamide, as a yellow solid. The intermediate was pure enough to be used in the next step without further purification. LC/MS (method I, ESI): [ M + H ]]+=449.0,RT=1.02min.1H NMR(400MHz,CD3OD)δ9.11(dd,J=6.8,1.6Hz,1H),8.67–8.64(m,2H),8.32(s,1H),7.80(d,J=2.4Hz,1H),7.72(dd,J=8.8,2.4Hz,1H),7.37(d,J=8.8Hz,1H),7.23(dd,J=7.0,4.2Hz,1H),6.81(t,J=73.2Hz,1H)。

Intermediate 3

Figure BDA0002282880340000671

N- [3- [2- (difluoromethoxy) -5-iodophenyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Step 1: synthesis of N- [5- [2- (difluoromethoxy) -5-iodophenyl ] -1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Figure BDA0002282880340000672

To N- [5- [ 5-bromo-2- (difluoromethoxy) phenyl ] under nitrogen]-1- [ [2- (trimethylsilyl) ethoxy ] group]Methyl radical]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]To a solution of pyrimidine-3-carboxamide (100mg, 0.173mmol) in t-BuOH (2mL) were added N, N-dimethylethane-1, 2-diamine (2.28mg, 0.0259mmol), NaI (155mg, 1.04mmol), and CuI (4.93mg, 0.026 mmol). The resulting solution was stirred in an oil bath at 120 ℃ under nitrogen for 14 hours, after which it was cooled to room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel, eluting with ethyl acetate/petroleum ether (1/1), to give 80mg (74%) of N- [5- [2- (difluoromethoxy) -5-iodophenyl]-1- [ [2- (trimethylsilyl) ethoxy ] group]Methyl radical]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]Pyrimidine-3-carboxamide, as a yellow solid. LC/MS (method J, ESI): [ M + H ]]+=627.1,RT=1.31min.

Step 2: synthesis of N- [3- [2- (difluoromethoxy) -5-iodophenyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Reacting N- [5- [2- (difluoromethoxy) -5-iodophenyl]-1- [ [2- (trimethylsilyl) ethoxy ] group]Methyl radical]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]Pyrimidine-3-carboxamide (80.0mg, 0.128mmol) with CF3CO2H (3.0mL) was treated at room temperature for 30 min. The resulting mixture was concentrated under reduced pressure. The residue was dissolved in water. Saturated sodium bicarbonate was slowly added until the solution pH-8. The solid was collected by filtration. The solid was purified by flash chromatography on silica gel, eluting with ethyl acetate/petroleum ether (2/1), to give 23.0mg (36%) of N- [3- [2- (difluoromethoxy) -5-iodophenyl]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]Pyrimidine-3-carboxamide, as a pale yellow solid. LC/MS (method K, ESI): [ M + H ]]+=497.1,RT=1.74min.1H NMR(300MHz,CD3OD)δ9.08(dd,J=6.9,1.5Hz,1H),8.65–8.61(m,2H),8.27(s,1H),7.94(s,1H),7.87(d,J=8.7Hz,1H),7.21–7.18(m,2H),6.78(t,J=73.2Hz,1H)。

Intermediate 4

Figure BDA0002282880340000682

N- (3- (5- ((1R,2R) -2-cyanocyclopropyl) -2- (difluoromethoxy) phenyl) -1H-pyrazol-4-yl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Step 1: synthesis of N- [5- [5- (2-cyanocyclopropyl) -2- (difluoromethoxy) phenyl ] -1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Figure BDA0002282880340000691

To N- [5- [ 5-bromo-2- (difluoromethoxy) phenyl ] under nitrogen]-1- [ [2- (trimethylsilyl) ethoxy ] group]Methyl radical]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]To a solution of pyrimidine-3-carboxamide (intermediate 1, 1.00g, 1.73mmol) in dioxane (15mL) and water (3.0mL) was added potassium (2-cyanocyclopropyl) trifluoroborate (449mg,2.60mmol)、Pd(dppf)Cl2.CH2Cl2(141mg, 0.173mmol, 0.10equiv) and Cs2CO3(1.13g, 3.47mmol, 2.01 equiv). The resulting solution was stirred in an oil bath at 80 ℃ overnight. The resulting mixture was concentrated under reduced pressure. The reaction on this scale was repeated five times and the crude products of 5 runs were combined for purification. The residue was passed through a short pad of silica gel, eluting with ethyl acetate/petroleum ether (6/4). The appropriate fractions were combined and concentrated under reduced pressure to give 2.5g (85% purity, LCMS 254nm) of N- [5- [5- (2-cyanocyclopropyl) -2- (difluoromethoxy) phenyl]-1- [ [2- (trimethylsilyl) ethoxy ] group]Methyl radical]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]Pyrimidine-3-carboxamide, as a yellow solid. The intermediate was of sufficient quality to proceed to the next step. No further purification was required. LC/MS (method G, ESI): [ M + H ]]+=566.2,RT=1.07min。

Step 2: n- (3- [5- [ (1R,2R) -2-cyanocyclopropyl ] -2- (difluoromethoxy) phenyl ] -1H-pyrazol-4-yl) pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Reacting N- [5- [5- (2-cyanocyclopropyl) -2- (difluoromethoxy) phenyl]-1- [ [2- (trimethylsilyl) ethoxy ] group]Methyl radical]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]Pyrimidine-3-carboxamide (2.90g, 5.13mmol) was treated with trifluoroacetic acid (10mL) in dichloromethane (20mL) overnight at room temperature. The resulting mixture was concentrated in vacuo. The pH of the residue was adjusted to DIEA>7. The resulting mixture was concentrated in vacuo. Water was added and the mixture was stirred for 1 hour. The solid was collected by filtration. The crude product (2.30g) was purified by preparative-HPLC under the following conditions: column, XBridge Shield RP18 OBD column, 19 x150 mm5um 13 nm; mobile phase of 0.05% NH4OH water and MeCN (30.0% MeCN rises to 45.0% in 9 minutes); detector, UV 254 nm. The racemic product was isolated by preparative-SFC under the following conditions: column: CHIRALPAKIC-SFC-02, 5cm 25cm (5 um); mobile phase A: CO 2250, mobile phase B: 50 parts of EtOH; flow rate: 180 mL/min; 220 nm; rT113.97min (first peak); rT2=17.84min (second peak) to obtain two fractions:

fraction 1(R, R-isomer): the first peak is the desired fraction and is further purified by recrystallization from isopropanol. 340mg (15%) of N- (3- [5- [ (1R,2R) -2-cyanocyclopropyl) -were obtained]-2- (difluoromethoxy) phenyl]-1H-pyrazol-4-yl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide, as an off-white solid.1H NMR(300MHz,CD3OD)δ9.09(dd,J=7.1,1.7Hz,1H),8.67–8.62(m,2H),8.27(s,1H),7.43–7.35(m,3H),7.21(dd,J=6.9,4.2Hz,1H),6.76(t,J=73.8Hz,1H),2.79–2.72(m,1H),1.94–1.88(m,1H),1.68–1.63(m,1H),1.62–1.56(m,1H).

Fraction 2(S, S-isomer): the second peak gave 474mg (21%) of N- (3- [5- [ (1S,2S) -2-cyanocyclopropyl]-2- (difluoromethoxy) phenyl]-1H-pyrazol-4-yl) pyrazolo [1,5-a]Pyrimidine-3-carboxamide, as a white solid.1H NMR(300MHz,CD3OD)δ9.09(dd,J=7.1,1.7Hz,1H),8.67–8.62(m,2H),8.27(s,1H),7.43–7.35(m,3H),7.21(dd,J=6.9,4.2Hz,1H),6.76(t,J=73.8Hz,1H),2.79–2.72(m,1H),1.94–1.88(m,1H),1.68–1.63(m,1H),1.62–1.56(m,1H).

Intermediate 5

Figure BDA0002282880340000701

N- [3- [ 5-cyclopropyl-2- (difluoromethoxy) phenyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Step 1: synthesis of N- [3- [ 5-cyclopropyl-2- (difluoromethoxy) phenyl ] -1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Figure BDA0002282880340000711

To N- (5- (5-bromo-2- (difluoromethoxy) phenyl) -1- ((2- (trimethylsilyl) ethoxy) methyl) -1H-pyrazol-4-yl) pyrazolo [1,5-a ] under nitrogen]To a solution of pyrimidine-3-carboxamide (intermediate 1, 1.40g, 2.41mmol) in dioxane (15mL) and water (3.0mL) was added cyclopropylboronic acid (314mg, 3.66 m)mol)、Pd(dppf)Cl2-CH2Cl2(200mg, 0.245mmol) and Cs2CO3(1.56g, 4.79 mmol). The reaction mixture was stirred at 80 ℃ under nitrogen overnight. The resulting mixture was concentrated under reduced pressure. The residue was passed through a short pad of silica gel, eluting with dichloromethane/methanol (94/6). The appropriate fractions were combined and concentrated under reduced pressure to give 1.40g (purity at 254nm ═ 85%) of N- [3- [ 5-cyclopropyl-2- (difluoromethoxy) phenyl]-1- [ [2- (trimethylsilyl) ethoxy ] group]Methyl radical]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]Pyrimidine-3-carboxamide, as a dark red solid. LC/MS (method G, ESI): [ M + H ]]+=541.2,RT1.12 min. The intermediate was used without further purification.

Step 2: synthesis of N- [3- [ 5-cyclopropyl-2- (difluoromethoxy) phenyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Figure BDA0002282880340000712

The N- [3- [ 5-cyclopropyl-2- (difluoromethoxy) phenyl group obtained in the previous step]-1- [ [2- (trimethylsilyl) ethoxy ] group]Methyl radical]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]Pyrimidine-3-carboxamide (145mg) was treated with HCl/dioxane (5.0mL, 4M) for 2 hours at 25 ℃. The solution was concentrated under reduced pressure. The residue was purified by preparative-HPLC under the following conditions: column: xbridge C18, 19 x150 mm, 5 um; mobile phase A: water/0.05% NH4HCO3And the mobile phase B: ACN; flow rate: 30 mL/min; gradient: 20% B to 85% B in 10 minutes; 254nm to give 44.9mg (41%) of N- [3- [ 5-cyclopropyl-2- (difluoromethoxy) phenyl]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]Pyrimidine-3-carboxamide, as a yellow solid. LC/MS (method H, ESI): [ M + H ]]+=411.2,RT=1.14min.1H NMR(300MHz,CD3OD)δ9.09(dd,J=6.9,1.5Hz,1H),8.63–8.61(m,2H),8.27(s,1H),7.28–7.25(m,3H),7.20(dd,J=7.2,4.2Hz,1H),6.68(t,J=73.8Hz,1H),2.04–1.95(m,1H),1.03–0.97(m,2H),0.79–0.71(m,2H)

Intermediate 6

Figure BDA0002282880340000721

N- [3- [2- (difluoromethoxy) -5- [4- (dimethylcarbamoyl) phenoxy ] phenyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Step 1: synthesis of N- [5- [2- (difluoromethoxy) -5- [4- (dimethylcarbamoyl) phenoxy ] phenyl ] -1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Figure BDA0002282880340000722

To N-5- [ 5-bromo-2- (difluoromethoxy) phenyl under nitrogen]-1- [2- (trimethylsilyl) ethoxy]methyl-1H-pyrazol-4-yl pyrazolo [1,5-a]To a solution of pyrimidine-3-carboxamide (intermediate 1, 1.17g, 2.02mmol) in toluene (10mL) was added 4-hydroxy-N, N-dimethylbenzamide (0.400g, 2.42mmol), Cs2CO3(0.790g,2.43mmol)、[PdCl(allyl)]2(37.0mg, 0.101mmol) and t-BuBrettPhos (98.0mg, 0.202 mmol). The reaction mixture was stirred at 90 ℃ overnight. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica eluting with methylene chloride/methanol (95/5) to give 1.3g (81%) of N- [5- [2- (difluoromethoxy) -5- [4- (dimethylcarbamoyl) phenoxy ] ethyl acetate]Phenyl radical]-1- [ [2- (trimethylsilyl) ethoxy ] group]Methyl radical]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]Pyrimidine-3-carboxamide, as a yellow oil. LC/MS (method H, ESI): [ M + H ]]+=664.4,RT=1.36min.

Step 2: synthesis of N- [3- [2- (difluoromethoxy) -5- [4- (dimethylcarbamoyl) phenoxy ] phenyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Figure BDA0002282880340000731

To the N- [5- [2- (difluoromethoxy) -5- [4- (dimethylcarbamoyl) phenoxy group]Phenyl radical]-1- [ [2- (trimethylsilyl) ethoxy ] group]Methyl radical]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]To a solution of pyrimidine-3-carboxamide (139mg, 0.209mmol) in MeOH (3.0mL) was added concentrated hydrochloric acid (1.5mL, 12M). The reaction mixture was stirred at 25 ℃ for 2 hours and concentrated under reduced pressure. The residue was neutralized with DIPEA. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica eluting with methylene chloride/methanol (95/5) to give 28mg (25%) of N- [3- [2- (difluoromethoxy) -5- [4- (dimethylcarbamoyl) phenoxy ] p]Phenyl radical]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]Pyrimidine-3-carboxamide, as an off-white solid. LC/MS (method H, ESI): [ M + H ]]+=534.2,RT=1.09min。1H NMR(300MHz,DMSO-d6)δ13.05(s,1H),9.73(s,1H),9.35(dd,J=7.1,1.7Hz,1H),8.70(dd,J=4.2,1.5Hz,1H),8.69(s,1H),8.22(s,1H),7.47(d,J=9.3Hz,1H),7.39(d,J=8.4Hz,2H),7.32–7.25(m,3H),7.06(d,J=8.7Hz,2H),7.18(t,J=73.8Hz,1H),2.94(s,6H).

Intermediate 7

N- [3- [2, 5-bis (difluoromethoxy) phenyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide

Step 1: synthesis of 1- (benzyloxy) -4- (difluoromethoxy) benzene

Figure BDA0002282880340000741

To a 3000-mL round-bottomed flask purged and maintained under a nitrogen inert atmosphere were added N, N-dimethylformamide (1500mL), 4- (benzyloxy) phenol (200g, 999mmol) and Cs2CO3(651g, 1.99 mol). The reaction vessel is provided with CO2An outlet for release. Subsequently, sodium 2-chloro-2, 2-difluoroacetate (228g, 1.50mol, 1.50equiv) was added in several portions at 120 ℃. After the addition of the 2-chloro-2, 2-difluorosodium acetate was complete, the reaction was stirred in a 120 ℃ oil bath until no gas escaped (. about.1 h) and then allowed to cool to room temperature. The reaction mixture was added slowly to 3000mL of water/ice with stirring. The resulting mixture was extracted with ethyl acetate (3x4000 mL). Combining the organic layers with anhydrous sodium sulfateDried and concentrated under reduced pressure. The residue was purified by flash chromatography on silica eluting with ethyl acetate/petroleum ether (1/19). The appropriate fractions were combined and concentrated under reduced pressure. The reaction was repeated four times. A total of 450g (36%) of 1- (benzyloxy) -4- (difluoromethoxy) benzene was obtained as a white solid.

Step 2: synthesis of 4- (difluoromethoxy) phenol

Figure BDA0002282880340000742

To a 3000-mL round bottom flask were added methanol (1500mL), 1- (benzyloxy) -4- (difluoromethoxy) benzene (140g, 559mmol), and 10% palladium on carbon (15g, 141 mmol). The resulting mixture was stirred overnight at room temperature under hydrogen (-45 psi). The catalyst is filtered off. The filtrate was concentrated under reduced pressure. The reaction was repeated three times. 300g (78%) of 4- (difluoromethoxy) phenol were obtained as a yellow oil.

And step 3: synthesis of 2-bromo-4- (difluoromethoxy) phenol

Figure BDA0002282880340000743

To a 1000-mL round bottom flask was added acetic acid (500mL), 4- (difluoromethoxy) phenol (50g, 312mmol) and NBS (55.6g, 312 mmol). The reaction mixture was stirred at 15 ℃ for 1 hour. The resulting mixture was then slowly added to 1000mL of water/ice with stirring. The resulting solution was extracted with ethyl acetate (3 × 1000 mL). The organic layers were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica eluting with methylene chloride/petroleum ether (30/70). The appropriate fractions were collected and concentrated under reduced pressure. 50g (67%) of 2-bromo-4- (difluoromethoxy) phenol were obtained as a pale yellow oil.

And 4, step 4: synthesis of 2-bromo-1, 4-bis (difluoromethoxy) benzene

Figure BDA0002282880340000751

To a 2000-mL round bottom flask was added CH3CN (500mL), water (500mL), 2-bromo-4- (difluoromethyl)Oxy) phenol (54g, 226mmol) and potassium hydroxide (94g, 1.68 mol). The flask was placed in an ice bath and the reaction mixture was stirred in the ice bath for 30 minutes. Diethyl (bromodifluoromethyl) phosphonate (120g, 449mmol) was then added dropwise to the reaction mixture at 0 ℃. After the addition of diethyl (bromodifluoromethyl) phosphonate was complete, the reaction mixture was stirred in a water/ice bath for 1 hour. The resulting solution was extracted with ethyl acetate (3 × 300 mL). The organic layers were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica eluting with ethyl acetate/petroleum ether (1/19) and the appropriate fractions were collected and concentrated under reduced pressure. 54g (83%) of 2-bromo-1, 4-bis (difluoromethoxy) benzene were obtained as a pale yellow oil.

And 5: synthesis of 5- [2, 5-bis (difluoromethoxy) phenyl ] -4-nitro-1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrazole

Figure BDA0002282880340000752

To a 1000-mL round bottom flask purged and maintained under a nitrogen inert atmosphere was added DMA (500mL), potassium carbonate (112g, 810mmol), 4-nitro-1- [ [2- (trimethylsilyl) ethoxy ] ethanol]Methyl radical]-1H-pyrazole (66g, 271mmol), 2-bromo-1, 4-bis (difluoromethoxy) benzene (79g, 273mmol), 2-dimethylpropionic acid (8.3g, 81.3mmol), Pd (OAc)2(6.0g, 26.7mmol) and bis (adamantan-1-yl) (butyl) phosphine (19g, 52.9mmol, 0.195 equiv). The reaction mixture was stirred in a 120 ℃ oil bath overnight and allowed to cool to room temperature. The reaction mixture was then added to 1000mL of water/ice with stirring. The resulting solution was extracted with ethyl acetate (3 × 1000 mL). The organic layers were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica eluting with ethyl acetate/petroleum ether (1/1). The appropriate fractions were collected and concentrated under reduced pressure. 100g (82%) of 5- [2, 5-bis (difluoromethoxy) phenyl are obtained]-4-nitro-1- [ [2- (trimethylsilyl) ethoxy ] ethanol]Methyl radical]-1H-pyrazole, as a solid.

Step 6: synthesis of 5- [2, 5-bis (difluoromethoxy) phenyl ] -1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrazol-4-amine

To a 3000-mL 3-necked round bottom flask was added ethanol (1500mL), water (150mL), 5- [2, 5-bis (difluoromethoxy) phenyl]-4-nitro-1- [ [2- (trimethylsilyl) ethoxy ] ethanol]Methyl radical]-1H-pyrazole (100.00g, 221mmol), iron powder (124g, 2.22mol) and NH4Cl (59.2g, 1.11 mol). The resulting mixture was stirred in a refluxing temperature oil bath for 2 hours and then cooled to room temperature. The solid was filtered off and washed with ethanol. The filtrate was concentrated under reduced pressure. The residue was dissolved in 3000mL of ethyl acetate. The ethyl acetate solution was washed with 1x1000mL brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. 100g of crude 5- [2, 5-bis (difluoromethoxy) phenyl]-1- [ [2- (trimethylsilyl) ethoxy ] group]Methyl radical]-1H-pyrazol-4-amine, as a pale yellow oil, not purified for direct use.

And 7: synthesis of N- [5- [2, 5-bis (difluoromethoxy) phenyl ] -1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide

To a 2000-mL round bottom flask was added DMA (1000mL), 5- [2, 5-bis (difluoromethoxy) phenyl ] -1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrazol-4-amine obtained in the previous step, pyrazolo [1,5-a ] pyrimidine-3-carboxylic acid (58.06g, 355.9mmol), 7-azabenzotriazol-1-yloxy) tripyrrolidinophosphonium hexafluorophosphate (PyAOP) (185.56g, 355.9mmol), 4-dimethylaminopyridine (2.90g, 23.7mmol), and DIPEA (92.0g, 712 mmol). The resulting solution was stirred in a 65 ℃ oil bath overnight. The reaction mixture was then slowly added to 2000mL of water with stirring. The reaction solution was extracted with ethyl acetate (3 × 2000 mL). The combined organic phases were washed with 1000mL brine, dried over anhydrous sodium sulfate and concentrated under pressure. The residue was purified by flash chromatography on silica eluting with ethyl acetate/petroleum ether (40/60). The appropriate fractions were combined and concentrated under reduced pressure to give 120g N- [5- [2, 5-bis (difluoromethoxy) phenyl ] -1- [ [2- (trimethylsilyl) ethoxy ] methyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide as a white solid.

And 8: synthesis of N- [3- [2, 5-bis (difluoromethoxy) phenyl ] -1H-pyrazol-4-yl ] pyrazolo [1,5-a ] pyrimidine-3-carboxamide

To a 2000-mL round bottom flask was added methanol (800mL), concentrated hydrochloric acid (400mL, 12N) and N- [5- [2, 5-bis (difluoromethoxy) phenyl]-1- [ [2- (trimethylsilyl) ethoxy ] group]Methyl radical]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]Pyrimidine-3-carboxamide (80g, 141 mmol). The resulting solution was stirred at 25 ℃ for 4 hours. The solid was collected by filtration. Put the solid into a 1L flask and add H2O (200 mL). Saturated NaHCO was added dropwise with stirring3The aqueous solution was allowed to reach pH 8. The solid was collected by filtration, washed with water and dried to give 55g (89%) of N- [3- [2, 5-bis (difluoromethoxy) phenyl]-1H-pyrazol-4-yl]Pyrazolo [1,5-a]Pyrimidine-3-carboxamide, as a pale yellow solid.1H NMR(300MHz,CD3OD)δ9.08(dd,J=7.2,1.5Hz,1H),8.65–8.61(m,2H),8.28(s,1H),7.46(d,J=9.0Hz,1H),7.40(d,J=3.0Hz,1H),7.34(dd,J=8.9,2.9Hz,1H),7.19(dd,J=6.7,4.4Hz,1H),6.87(t,J=73.7Hz,1H),6.73(t,J=73.7Hz,1H).

96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:Acalabrutinib的新晶型及其制备方法和用途

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!