Treatment of hidradenitis suppurativa with JAK inhibitors

文档序号:368706 发布日期:2021-12-10 浏览:19次 中文

阅读说明:本技术 使用jak抑制剂治疗化脓性汗腺炎 (Treatment of hidradenitis suppurativa with JAK inhibitors ) 是由 M·D·豪威尔 P·史密斯 于 2019-03-29 设计创作,主要内容包括:本申请提供了治疗有需要的患者的化脓性汗腺炎的方法,所述方法包括向所述患者施用治疗有效量的抑制JAK1和/或JAK2的化合物或其药学上可接受的盐。(The present application provides a method of treating hidradenitis suppurativa in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a compound that inhibits JAK1 and/or JAK2, or a pharmaceutically acceptable salt thereof.)

1. A method of treating hidradenitis suppurativa in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a compound that inhibits JAK1, or a pharmaceutically acceptable salt thereof, wherein the compound is:

R2is C1-6Alkyl radical, C1-6Haloalkyl, C3-6Cycloalkyl or C3-6cycloalkyl-C1-3Alkyl radical, wherein said C1-6Alkyl radical, C3-6Cycloalkyl and C3-6cycloalkyl-C1-3Each alkyl group is optionally selected from fluoro, -CF, 1,2 or 3 independently3And methyl;

R3is H or methyl;

R4h, F or Cl;

R5is H or F;

R6is H or F;

R7is H or F;

R8is H or methyl;

R9is H or methyl;

R10is H or methyl; and is

R11Is H or methyl;

((2R,5S) -5- {2- [ (1R) -1-hydroxyethyl ] -1H-imidazo [4,5-d ] thieno [3,2-b ] pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile;

3- [1- (6-chloropyridin-2-yl) pyrrolidin-3-yl ] -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] propionitrile;

3- (1- [1,3] oxazolo [5,4-b ] pyridin-2-ylpyrrolidin-3-yl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] propionitrile;

4- [ (4- { 3-cyano-2- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] propyl } piperazin-1-yl) carbonyl ] -3-fluorobenzonitrile;

4- [ (4- { 3-cyano-2- [3- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrrol-1-yl ] propyl } piperazin-1-yl) carbonyl ] -3-fluorobenzonitrile;

[ trans-1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] -3- (4- { [2- (trifluoromethyl) pyrimidin-4-yl ] carbonyl } piperazin-1-yl) cyclobutyl ] acetonitrile;

{ trans-3- (4- { [4- [ (3-hydroxyazetidin-1-yl) methyl ] -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- { [ (2S) -2- (hydroxymethyl) pyrrolidin-1-yl ] methyl } -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- { [ (2R) -2- (hydroxymethyl) pyrrolidin-1-yl ] methyl } -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

4- (4- {3- [ (dimethylamino) methyl ] -5-fluorophenoxy } piperidin-1-yl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] butanenitrile;

5- {3- (cyanomethyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-1-yl } -N-isopropylpyrazine-2-carboxamide;

4- {3- (cyanomethyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-1-yl } -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide;

5- {3- (cyanomethyl) -3- [4- (1H-pyrrolo [2,3-b ] pyridin-4-yl) -1H-pyrazol-1-yl ] azetidin-1-yl } -N-isopropylpyrazine-2-carboxamide;

{1- (cis-4- { [6- (2-hydroxyethyl) -2- (trifluoromethyl) pyrimidin-4-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{1- (cis-4- { [4- [ (ethylamino) methyl ] -6- (trifluoromethyl) pyridin-2-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{1- (cis-4- { [4- (1-hydroxy-1-methylethyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{1- (cis-4- { [4- { [ (3R) -3-hydroxypyrrolidin-1-yl ] methyl } -6- (trifluoromethyl) pyridin-2-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{1- (cis-4- { [4- { [ (3S) -3-hydroxypyrrolidin-1-yl ] methyl } -6- (trifluoromethyl) pyridin-2-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{ trans-3- (4- { [4- ({ [ (1S) -2-hydroxy-1-methylethyl ] amino } methyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- ({ [ (2R) -2-hydroxypropyl ] amino } methyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- ({ [ (2S) -2-hydroxypropyl ] amino } methyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- (2-hydroxyethyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

or a pharmaceutically acceptable salt of any of the above compounds.

2. The method of claim 1 wherein the compound or salt is selective for JAK1 over JAK2, JAK3 and TYK 2.

3. The method of claim 2, wherein the compound is of formula II or a pharmaceutically acceptable salt thereof:

R2is C1-6Alkyl radical, C1-6Haloalkyl, C3-6Cycloalkyl or C3-6cycloalkyl-C1-3Alkyl radical, wherein said C1-6Alkyl radical, C3-6Cycloalkyl and C3-6cycloalkyl-C1-3Each alkyl group is optionally selected from fluoro, -CF, 1,2 or 3 independently3And methyl;

R3is H or methyl;

R4h, F or Cl;

R5is H or F;

R6is H or F;

R7is H or F;

R8is H or methyl;

R9is H or methyl;

R10is H or methyl; and is

R11Is H or methyl.

4. The method of claim 3, wherein the compound is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide, or a pharmaceutically acceptable salt thereof.

5. The method of claim 3, wherein the salt is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide phosphate.

6. The method of claim 3, wherein the salt is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide hydrochloride.

7. The method of claim 3, wherein the salt is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide hydrobromide.

8. The method of claim 3, wherein the salt is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide sulfate.

9. The method of claim 1, wherein the compound is ((2R,5S) -5- {2- [ (1R) -1-hydroxyethyl ] -1H-imidazo [4,5-d ] thieno [3,2-b ] pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile or a pharmaceutically acceptable salt thereof.

10. The method of claim 1, wherein the compound is ((2R,5S) -5- {2- [ (1R) -1-hydroxyethyl ] -1H-imidazo [4,5-d ] thieno [3,2-b ] pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile monohydrate.

11. The method of claim 1, wherein the compound or salt is administered at a dose of 15mg, 30mg, 60mg, or 90mg as the free base.

12. The method of claim 1, further comprising administering an additional therapeutic agent.

13. The method of claim 12, wherein the other therapeutic agent is an antibiotic, a retinoid, a corticosteroid, an anti-TNF-a agent, or an immunosuppressive agent.

14. The method of claim 13, wherein the antibiotic is clindamycin, doxycycline, minocycline, trimethoprim-sulfamethoxazole, erythromycin, metronidazole, rifampin, moxifloxacin, dapsone, or a combination thereof.

15. The method of claim 13, wherein the retinoid is acitretin, or isotretinoin.

16. The method of claim 13, wherein the corticosteroid is triamcinolone, dexamethasone, fluocinolone, cortisone, prednisone, prednisolone, or fluorometholone.

17. The method of claim 13, wherein the anti-TNF-alpha agent is infliximab, etanercept, or adalimumab.

18. The method of claim 13, wherein the immunosuppressive agent is methotrexate, cyclosporine a, mycophenolate mofetil, or mycophenolate sodium.

19. The method of claim 12, wherein the additional therapeutic agent is finasteride, metformin, adapalene, or azelaic acid.

20. The method of any one of claims 1-19, wherein the administration of the compound or salt is topical administration.

21. The method of any one of claims 1-19, wherein the administration of the compound or salt is oral administration.

22. The method of any one of claims 1-19, wherein the method increases HiSCR (hidradenitis suppurativa clinical response) by 10%, 20%, 30%, 40%, or 50%.

Technical Field

The present application provides methods of treating Hidradenitis Suppurativa (HS) using compounds that modulate the activity of Janus kinases (JAK)1 and/or 2.

Background

Protein Kinases (PKs) regulate a variety of biological processes including, inter alia, cell growth, survival, differentiation, organ formation, morphogenesis, neovascularization, tissue repair and regeneration. Protein kinases also play a specialized role in many human diseases including cancer. Cytokines, low molecular weight polypeptides or glycoproteins regulate many of the pathways associated with the inflammatory response of the host to sepsis. Cytokines influence cell differentiation, proliferation and activation, and can modulate pro-inflammatory and anti-inflammatory responses to allow the host to respond appropriately to pathogens. Signaling of a large number of cytokines involves the Janus kinase family of protein tyrosine kinases (JAKs) and Signal Transducers and Activators of Transcription (STATs). There are four known mammalian JAKs: JAK1(Janus kinase-1), JAK2, JAK3 (also known as Janus kinase, white blood cells; JAKL; and L-JAK), and TYK2 (protein tyrosine kinase 2).

The pathogenesis of diseases is caused by cytokine-stimulated immune and inflammatory responses: pathologies such as Severe Combined Immunodeficiency Disease (SCID) result from a depression of the immune system, while hyperactive or inappropriate immune/inflammatory responses cause autoimmune diseases (e.g., asthma, systemic lupus erythematosus, thyroiditis, myocarditis) and pathologies such as scleroderma and osteoarthritis (Ortmann, r.a., t.cheng et al (2000) Arthritis Res 2(1): 16-32).

A lack of JAK expression is associated with a number of disease conditions. For example, Jak 1-/-mice are short at birth, unable to feed, and die perinatally (Rodig, S.J., M.A. Meraz et al, (1998) Cell93(3): 373-83). Jak 2-/-mouse embryos are anemic and die about 12.5 days post-coital due to lack of directed erythropoiesis.

The JAK/STAT pathway and in particular all four JAKs are believed to play a role in the pathogenesis of the asthmatic response, chronic obstructive pulmonary disease, bronchitis and other related inflammatory diseases of the lower respiratory tract. Various cytokines that signal through JAKs are associated with inflammatory diseases/disorders of the upper respiratory tract, such as diseases affecting the nose and sinuses (e.g., rhinitis and sinusitis), whether or not traditionally allergic reactions. The JAK/STAT pathway is also associated with inflammatory diseases/disorders of the eye and chronic allergic reactions.

JAK/STAT may be activated in cancer by stimulation of cytokines (e.g., IL-6 or GM-CSF) or by reduction of endogenous inhibitors of JAK signaling such as SOCS (suppressor of cytokine signaling) or PIAS (protein inhibitor of activated STAT) (Boudny, V. and Kovarik, J., Neoplasm.49:349-355, 2002). STAT signaling and activation of other pathways downstream of JAK (e.g., Akt) are associated with poor prognosis in many cancer types (Bowman, T. et al, Oncogene 19: 2474-. Elevated levels of circulating cytokines that signal through JAK/STAT are the cause of cachexia and/or chronic fatigue. Thus, JAK inhibition may benefit cancer patients for reasons that extend beyond potential anti-tumor activity.

JAK2 tyrosine kinase may be beneficial in patients with myeloproliferative disorders, such as Polycythemia Vera (PV), Essential Thrombocythemia (ET), myeloid metaplasia with myelofibrosis (MMM) (Levin ET al, Cancer Cell, Vol.7, 2005:387 397). Inhibition of JAK2V617F kinase reduces proliferation of hematopoietic cells, suggesting that JAK2 is a potential target for pharmacological inhibition in patients with PV, ET, and MMM.

Inhibition of JAK may be beneficial to patients suffering from skin immune disorders such as psoriasis and skin sensitization. It is believed that the maintenance of psoriasis is dependent on a number of inflammatory cytokines, in addition to the various chemokines and growth factors many of which signal through JAK (Adv Pharmacol.2000; 47:113-74) (JCI113: 1664-1675).

Therefore, there is a continuing need for new or improved agents that inhibit kinases such as JAK for the development of new and more effective drugs aimed at strengthening or inhibiting immune and inflammatory pathways, such as the treatment of hidradenitis suppurativa. This application is directed to these needs and others.

Disclosure of Invention

The present application provides a method of treating hidradenitis suppurativa in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a compound that inhibits JAK1 and/or JAK2, or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound or salt is selective for JAK1 and JAK2 as compared to selectivity for JAK3 and TYK 2.

In some embodiments, the compound or salt is selective for JAK1 as compared to JAK2, JAK3, and TYK 2.

In some embodiments, the compound is ruxolitinib (ruxolitinib), or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound is ruxolitinib, or a pharmaceutically acceptable salt thereof, wherein one or more hydrogen atoms are replaced with a deuterium atom.

In some embodiments, the salt is ruxolitinib phosphate.

In some embodiments, the compound is {1- {1- [ 3-fluoro-2- (trifluoromethyl) isonicotinyl ] piperidin-4-yl } -3[4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile or a pharmaceutically acceptable salt thereof.

In some embodiments, the salt is {1- {1- [ 3-fluoro-2- (trifluoromethyl) isonicotinyl ] piperidin-4-yl } -3[4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile adipate.

In some embodiments, the compound is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide, or a pharmaceutically acceptable salt thereof.

In some embodiments, the salt is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide phosphate.

In some embodiments, the compound or salt is administered at a dose of 15mg, 30mg, 60mg, or 90mg, as the free base.

In some embodiments, the compound is ((2R,5S) -5- {2- [ (1R) -1-hydroxyethyl ] -1H-imidazo [4,5-d ] thieno [3,2-b ] pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound is ((2R,5S) -5- {2- [ (1R) -1-hydroxyethyl ] -1H-imidazo [4,5-d ] thieno [3,2-b ] pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile monohydrate.

In some embodiments, the method further comprises administering an additional therapeutic agent (e.g., an antibiotic, a retinoid, a corticosteroid, an anti-TNF-a agent, or an immunosuppressive agent).

In some embodiments, the administration of the compound or salt is topical. In some embodiments, the administration of the compound or salt is oral administration.

In some embodiments, the method increases HiSCR (hidradenitis suppurativa clinical response) by 10%, 20%, 30%, 40% or 50%.

The present application also provides compounds that inhibit JAK1 and/or JAK2, or pharmaceutically acceptable salts thereof, for use in the treatment of hidradenitis suppurativa.

The present application further provides the use of a compound that inhibits JAK1 and/or JAK2, or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for the treatment of hidradenitis suppurativa.

Drawings

Figure 1 shows the individual gene expression values (MFI) of JAK1 for each replicate in the presence/absence of compounds a-D in TNF α and IFN- γ stimulated keratinocytes. Keratinocytes were stimulated with TNF α (25ng/mL) and IFN γ (25ng/mL) in the presence or absence of increasing concentrations of a JAK inhibitor. Data are presented as JAK1 expression levels for each group.

Figure 2 shows the individual gene expression values (MFI) of JAK2 for each replicate in the presence/absence of compounds a-D in TNF α and IFN- γ stimulated keratinocytes. Keratinocytes were stimulated with TNF α (25ng/mL) and IFN γ (25ng/mL) in the presence or absence of increasing concentrations of a JAK inhibitor. Data are presented as JAK2 expression levels for each group.

FIG. 3 shows the individual gene expression values (MFI) of IL-1. alpha. for each replicate in the presence/absence of compounds A-D in keratinocytes stimulated with TNF. alpha. and IFN-. gamma. Keratinocytes were stimulated with TNF α (25ng/mL) and IFN γ (25ng/mL) in the presence or absence of increasing concentrations of a JAK inhibitor. Data are presented as IL-1 α expression levels for each group.

FIG. 4 shows the individual gene expression values (MFI) of IL-6 for each replicate in the presence/absence of compounds A-D in keratinocytes stimulated with TNF α and IFN- γ. Keratinocytes were stimulated with TNF α (25ng/mL) and IFN γ (25ng/mL) in the presence or absence of increasing concentrations of a JAK inhibitor. Data are presented as IL-6 expression levels for each group.

FIG. 5 shows the individual protein concentrations (pg/mL) of IL-1 α in TNF α and IFN- γ stimulated keratinocytes in the presence/absence of compounds A-D for each replicate. Keratinocytes were stimulated with TNF α (25ng/mL) and IFN γ (25ng/mL) in the presence or absence of increasing concentrations of a JAK inhibitor. Data are presented as IL-1 α concentration for each group.

FIG. 6 shows the individual protein concentrations (pg/mL) of IL-6 in the presence/absence of compounds A-D in keratinocytes stimulated with TNF α and IFN- γ for each replicate. Keratinocytes were stimulated with TNF α (25ng/mL) and IFN γ (25ng/mL) in the presence or absence of increasing concentrations of a JAK inhibitor. Data are presented as IL-6 concentration for each group.

Figure 7 shows gene expression (MFI) of JAK1, JAK3 and TYK2 in the skin of healthy controls and subjects with hidradenitis suppurativa. Data are presented as JAK1, JAK3 or TYK2 gene expression levels for each healthy control (n ═ 4) and hidradenitis suppurativa (n ═ 41) subjects.

Fig. 8 shows gene expression (MFI) of STAT1, STAT2, and STAT3 in the skin of healthy controls and subjects with hidradenitis suppurativa. Data are presented as STAT1, STAT2, or STAT3 gene expression levels for each healthy control (n-4) and hidradenitis suppurativa (n-41) subjects.

Figure 9 shows gene expression (MFI) of IRAK1, IRAK2, and IRAK4 in the skin of healthy controls and subjects with hidradenitis suppurativa. Data are presented as IRAK1, IRAK2, or IRAK4 gene expression levels for each healthy control (n ═ 4) and hidradenitis suppurativa (n ═ 41) subjects.

Detailed Description

The present application provides, inter alia, a method of treating hidradenitis suppurativa in a patient in need thereof, the method comprising administering a therapeutically effective amount of a compound or a pharmaceutically acceptable salt thereof that inhibits JAK1 and/or JAK 2.

The methods described herein utilize compounds or salts that are inhibitors of JAK1 and/or JAK 2. In some embodiments, the compound is:

ruxolitinib;

ruxolitinib in which one or more hydrogen atoms are replaced with deuterium atoms;

{1- {1- [ 3-fluoro-2- (trifluoromethyl) isonicotinyl ] piperidin-4-yl } -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

4- {3- (cyanomethyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-1-yl } -N- [ 4-fluoro-2- (trifluoromethyl) phenyl ] piperidine-1-carboxamide;

[3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] -1- (1- { [2- (trifluoromethyl) pyrimidin-4-yl ] carbonyl } piperidin-4-yl) azetidin-3-yl ] acetonitrile;

4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide;

((2R,5S) -5- {2- [ (1R) -1-hydroxyethyl ] -1H-imidazo [4,5-d ] thieno [3,2-b ] pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile;

3- [1- (6-chloropyridin-2-yl) pyrrolidin-3-yl ] -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] propionitrile;

3- (1- [1,3] oxazolo [5,4-b ] pyridin-2-ylpyrrolidin-3-yl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] propionitrile;

4- [ (4- { 3-cyano-2- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] propyl } piperazin-1-yl) carbonyl ] -3-fluorobenzonitrile;

4- [ (4- { 3-cyano-2- [3- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrrol-1-yl ] propyl } piperazin-1-yl) carbonyl ] -3-fluorobenzonitrile;

[ trans-1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] -3- (4- { [2- (trifluoromethyl) pyrimidin-4-yl ] carbonyl } piperazin-1-yl) cyclobutyl ] acetonitrile;

{ trans-3- (4- { [4- [ (3-hydroxyazetidin-1-yl) methyl ] -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- { [ (2S) -2- (hydroxymethyl) pyrrolidin-1-yl ] methyl } -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- { [ (2R) -2- (hydroxymethyl) pyrrolidin-1-yl ] methyl } -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

4- (4- {3- [ (dimethylamino) methyl ] -5-fluorophenoxy } piperidin-1-yl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] butanenitrile;

5- {3- (cyanomethyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-1-yl } -N-isopropylpyrazine-2-carboxamide;

4- {3- (cyanomethyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-1-yl } -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide;

5- {3- (cyanomethyl) -3- [4- (1H-pyrrolo [2,3-b ] pyridin-4-yl) -1H-pyrazol-1-yl ] azetidin-1-yl } -N-isopropylpyrazine-2-carboxamide;

{1- (cis-4- { [6- (2-hydroxyethyl) -2- (trifluoromethyl) pyrimidin-4-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{1- (cis-4- { [4- [ (ethylamino) methyl ] -6- (trifluoromethyl) pyridin-2-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{1- (cis-4- { [4- (1-hydroxy-1-methylethyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{1- (cis-4- { [4- { [ (3R) -3-hydroxypyrrolidin-1-yl ] methyl } -6- (trifluoromethyl) pyridin-2-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{1- (cis-4- { [4- { [ (3S) -3-hydroxypyrrolidin-1-yl ] methyl } -6- (trifluoromethyl) pyridin-2-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{ trans-3- (4- { [4- ({ [ (1S) -2-hydroxy-1-methylethyl ] amino } methyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- ({ [ (2R) -2-hydroxypropyl ] amino } methyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- ({ [ (2S) -2-hydroxypropyl ] amino } methyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- (2-hydroxyethyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

or a pharmaceutically acceptable salt of any of the foregoing.

In some embodiments, the compound or salt is selective for JAK1 and JAK2 compared to JAK3 and TYK 2. In some embodiments, the compound is 3-cyclopentyl-3- [4- (7H-pyrrolo [2, 3-d)]Pyrimidin-4-yl) -1H-pyrazol-1-yl]Propionitrile or a pharmaceutical thereofA pharmaceutically acceptable salt. In some embodiments, the compound is (3R) -3-cyclopentyl-3- [4- (7H-pyrrolo [2, 3-d)]Pyrimidin-4-yl) -1H-pyrazol-1-yl]Propionitrile (ruxolitinib) or a pharmaceutically acceptable salt thereof. Luxolitinib with an IC of less than 10nM for JAK1 and JAK2 at 1mM ATP (assay A)50. 3-cyclopentyl-3- [4- (7H-pyrrolo [2,3-d ]]Pyrimidin-4-yl) -1H-pyrazol-1-yl]Propionitrile and ruxolitinib may be made by the procedure described in US 7,598,257 (example 67), filed on 12.12.2006, which is incorporated herein by reference in its entirety. In some embodiments, the inhibitor of JAK1 and/or JAK2 is (3R) -3-cyclopentyl-3- [4- (7H-pyrrolo [2,3-d ]]Pyrimidin-4-yl) -1H-pyrazol-1-yl]Propionitrile phosphate. The phosphate salt may be made as described in U.S. patent 8,722,693, which is incorporated herein by reference in its entirety.

In some embodiments, the compound or salt is a JAK1 inhibitor. In some embodiments, the compound or salt is selective for JAK1 as compared to JAK2, JAK3, and TYK 2. For example, some compounds described herein, or pharmaceutically acceptable salts thereof, preferentially inhibit JAK1 over one or more of JAK2, JAK3, and TYK 2. JAK1 plays an important role in a number of cytokine and growth factor signaling pathways that may cause or contribute to a disease condition when dysregulated. For example, IL-6 levels are elevated in rheumatoid arthritis, a disease that has been shown to have adverse effects (Fonesca et al, Autoimmiture Reviews,8:538-42, 2009). Because IL-6 signals at least in part via JAK1, IL-6 may produce potential clinical benefit indirectly via inhibition of JAK1 (Guschin et al, Embo J14: 1421,1995; Smolen et al, Lancet 371:987,2008). Furthermore, in some cancers, JAK1 is mutated, causing constitutively undesirable tumor cell growth and survival (Mullighan, Proc Natl Acad Sci U S A.106: 9414-. In other autoimmune diseases and cancers, elevated systemic levels of inflammatory cytokines that activate JAK1 can also cause the disease and/or associated symptoms. Thus, patients with such diseases may benefit from JAK1 inhibition. Selective inhibitors of JAK1 may be effective while avoiding the unnecessary and potentially undesirable effects of inhibiting other JAK kinases.

Hidradenitis suppurativa is characterized by marked skin inflammation; however, publications summarizing the inflammation are limited (Hoffman et al, PLOS One, 28.9.2018, https:// doi.org/10.1371/journal.bone.0203672). Presented herein are embodiments that support the following assumptions: inflammation is largely driven by JAK/STAT-mediated pathways. Examples C, D and E illustrate elevated JAK/STAT gene expression levels in the skin of HS patients compared to healthy skin. Furthermore, examples C, D and E illustrate that proinflammatory cytokines (TNF- α and IFN- γ) known to be elevated in HS induce JAK/STAT pathways in cultured keratinocytes and that this induction can be reduced by the addition of JAK inhibitors. Therefore, HS patients may benefit from JAK1 inhibition. Selective inhibitors of JAK1 may be effective while avoiding the unnecessary and potentially undesirable effects of inhibiting other JAK kinases.

In some embodiments, the compound or salt preferentially inhibits JAK1 (e.g., JAK2/JAK1 IC) as compared to JAK250Ratio of>1). In some embodiments, the compound or salt is about 10-fold selective for JAK1 as compared to JAK 2. In some embodiments, IC is measured, e.g., by measuring at 1mM ATP50(see example a) the compound or salt is about 3-fold, about 5-fold, about 10-fold, about 15-fold, or about 20-fold more selective for JAK1 than JAK 2.

In some embodiments, the JAK1 inhibitor is a compound of table 1 or a pharmaceutically acceptable salt thereof. The compounds in table 1 are selective JAK1 inhibitors (selective compared to JAK2, JAK3 and TYK 2). IC obtained by the method of example A at 1mM ATP50The values are shown in table 1.

TABLE 1

+ means <10nM (for assay conditions, see example A)

+ + means 100nM or less (for assay conditions, see example A)

+ + + + + means 300nM or less (for assay conditions, see example A)

aData for enantiomer 1

bData for enantiomer 2

In some embodiments, the JAK1 inhibitor is {1- {1- [ 3-fluoro-2- (trifluoromethyl) isonicotinyl ] piperidin-4-yl } -3[4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile or a pharmaceutically acceptable salt thereof.

In some embodiments, the JAK1 inhibitor is {1- {1- [ 3-fluoro-2- (trifluoromethyl) isonicotinyl ] piperidin-4-yl } -3[4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile adipate.

The synthesis and preparation of {1- {1- [ 3-fluoro-2- (trifluoromethyl) isonicotinoyl ] piperidin-4-yl } -3[4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile and its adipate can be found, for example, in U.S. patent publication No.2011/0224190 filed 3/9/2011, U.S. patent publication No.2013/0060026 filed 9/2012, 6/2014, U.S. patent publication No.2014/0256941 filed 3/5/2014, each of which is incorporated herein by reference in its entirety.

In some embodiments, the JAK1 inhibitor is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide, or a pharmaceutically acceptable salt thereof.

In some embodiments, the JAK1 inhibitor is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide phosphate.

In some embodiments, JAK1 is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide hydrochloride.

In some embodiments, JAK1 is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide hydrobromide.

In some embodiments, JAK1 is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide sulfate.

The synthesis and preparation of 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide and its phosphate salts can be found, for example, in U.S. patent publication No. us 2014/0343030, filed 5, 16 days 2014, which is incorporated herein by reference in its entirety.

In some embodiments, the JAK1 inhibitor is ((2R,5S) -5- {2- [ (1R) -1-hydroxyethyl ] -1H-imidazo [4,5-d ] thieno [3,2-b ] pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile or a pharmaceutically acceptable salt thereof.

In some embodiments, the JAK1 inhibitor is ((2R,5S) -5- {2- [ (1R) -1-hydroxyethyl ] -1H-imidazo [4,5-d ] thieno [3,2-b ] pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile monohydrate.

The synthesis of ((2R,5S) -5- {2- [ (1R) -1-hydroxyethyl ] -1H-imidazo [4,5-d ] thieno [3,2-b ] pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile and characterization of its anhydrous and monohydrate forms is described in U.S. patent publication nos. 2014/0121198 filed on 31.10.2013 and 2015/0344497 filed on 29.4.2015, each of which is incorporated herein by reference in its entirety.

In some embodiments, the compounds of table 1 are prepared by the synthetic procedures described in: U.S. patent publication No.2011/0224190 filed 3/9/2014, U.S. patent publication No.2014/0343030 filed 5/16/2014, U.S. patent publication No.2014/0121198 filed 10/31/2013, U.S. patent publication No.2010/0298334 filed 5/21/2010, U.S. patent publication No.2011/0059951 filed 8/31/2010, U.S. patent publication No.2012/0149681 filed 11/18/2011, U.S. patent publication No.2012/0149682 filed 11/18/2011, U.S. patent publication No. 2013/0018034 filed 6/19/2012, U.S. patent publication No.2013/0045963 filed 8/17/2012, and U.S. patent publication No.2014/0005166 filed 5/17/2013, each of which is incorporated herein by reference in its entirety.

In some embodiments, the JAK1 inhibitor is selected from a compound of: U.S. patent publication No.2011/0224190 filed 3/9/2014, U.S. patent publication No.2014/0343030 filed 5/16/2014, U.S. patent publication No.2014/0121198 filed 10/31/2013, U.S. patent publication No.2010/0298334 filed 5/21/2010, U.S. patent publication No.2011/0059951 filed 8/31/2010, U.S. patent publication No.2012/0149681 filed 11/18/2011, U.S. patent publication No.2012/0149682 filed 11/18/2011, U.S. patent publication No. 2013/0018034 filed 6/19/2012, U.S. patent publication No.2013/0045963 filed 8/17/2012, and U.S. patent publication No.2014/0005166 filed 5/17/2013, each of which is incorporated herein by reference in its entirety.

In some embodiments, the JAK1 inhibitor is a compound of formula I

Or a pharmaceutically acceptable salt thereof, wherein:

x is N or CH;

l is C (═ O) or C (═ O) NH;

a is phenyl, pyridinyl or pyrimidinyl, each optionally substituted with 1 or 2 independently selected R1Substituted by groups; and is

Each R1Independently fluorine or trifluoromethyl.

In some embodiments, the compound of formula I is {1- {1- [ 3-fluoro-2- (trifluoromethyl) isonicotinyl ] piperidin-4-yl } -3[4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of formula I is 4- {3- (cyanomethyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-1-yl } -N- [ 4-fluoro-2- (trifluoromethyl) phenyl ] piperidine-1-carboxamide or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound of formula I is [3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] -1- (1- { [2- (trifluoromethyl) pyrimidin-4-yl ] carbonyl } piperidin-4-yl) azetidin-3-yl ] acetonitrile or a pharmaceutically acceptable salt thereof.

In some embodiments, the JAK1 inhibitor is a compound of formula II

Or a pharmaceutically acceptable salt thereof, wherein:

R2is C1-6Alkyl radical, C1-6Haloalkyl, C3-6Cycloalkyl or C3-6cycloalkyl-C1-3Alkyl radical, wherein said C1-6Alkyl radical, C3-6Cycloalkyl and C3-6cycloalkyl-C1-3Each alkyl group is optionally selected from fluoro, -CF, 1,2 or 3 independently3And methyl;

R3is H or methyl;

R4h, F or Cl;

R5is H or F;

R6is H or F;

R7is H or F;

R8is H or methyl;

R9is H or methyl;

R10is H or methyl; and is

R11Is H or methyl.

In some embodiments, the compound of formula II is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide, or a pharmaceutically acceptable salt thereof.

In some embodiments, the JAK1 inhibitor is a compound of formula III

Or a pharmaceutically acceptable salt thereof, wherein:

Cy4is a tetrahydro-2H-pyran ring, optionally with 1 or 2 substituents independently selected from CN, OH, F, Cl, C1-3Alkyl radical, C1-3Haloalkyl, CN-C1-3Alkyl, HO-C1-3Alkyl, amino, C1-3Alkylamino and di (C)1-3Alkyl) amino, wherein said C is1-3Alkyl and di (C)1-3Alkyl) amino optionally selected from 1,2 or 3 independently selected from F, Cl, C1-3Alkylaminosulfonyl and C1-3Substituted with an alkylsulfonyl group; and is

R12is-CH2-OH、-CH(CH3) -OH or-CH2-NHSO2CH3

In some embodiments, the compound of formula III is ((2R,5S) -5- {2- [ (1R) -1-hydroxyethyl ] -1H-imidazo [4,5-d ] thieno [3,2-b ] pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile or a pharmaceutically acceptable salt thereof.

In some embodiments, the inhibitor of JAK1 and/or JAK2 is bartinib (barcetitinib), tofacitinib (tofacitinib), olatinib (oclacitinib), nonglutinib (filgonitinib), gandottinib (gandottinib), lestatinib (lestauurinb), momoritinib (momolotinib), bartinib (bacitinib), PF-04965842, upatatinib (upadacetitinib), pefinitib (peicitin), fiditinib (fedratinib), cucurbitacin i (curbitacin i), laratin-501 (acis), ATI-502(Aclaris), JTE052(Leo Pharma and Japan tobaco), or CHZ 868.

In some embodiments, the inhibitor of JAK1 and/or JAK2 may be an isotopically labeled compound or a pharmaceutically acceptable salt thereof. An "isotopically" or "radiolabeled" compound is a compound of the present disclosure in which one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature (i.e., naturally occurring). Suitable radionuclides that may be incorporated into the compounds of the present disclosure include, but are not limited to2H (deuterium, in turn, written as D),3H (tritium, also denoted as T),11C、13C、14C、13N、15N、15O、17O、18O、18F、35S、36Cl、82Br、75Br、76Br、77Br、123I、124I、125I and131I. for example, one or more hydrogen atoms in a compound of the disclosure may be replaced with deuterium atoms, e.g., -CD3By substitution into-CH3

One or more of the constituent atoms of the compounds described herein may be replaced or substituted with a natural or unnatural abundance of an atomic isotope. In some embodiments, the compound includes at least one deuterium atom. In some embodiments, the compound includes two or more deuterium atoms. In some embodiments, the compounds include 1-2, 1-3, 1-4, 1-5, or 1-6 deuterium atoms. In some embodiments, all hydrogen atoms in a compound may be replaced or substituted with deuterium atoms.

Synthetic methods for isotopes to be included in Organic compounds are known in The art (Deuterium laboratory in Organic Chemistry, Alan F. Thomas (New York, N.Y., Appleton-centre-Crofs, 1971; The Renaissance of H/D Exchange, Jens Atzrodt, Volker Derdau, Thorsten Fey and Jochen Zimmermann, Angel. chem. int. Ed.2007, 7744-7765; The Organic Chemistry of Isotropic laboratory, James R. Hanson, Royal Society of Chemistry, 2011.) isotopically labeled compounds can be used in a variety of studies, such as NMR, metabolic experiments and/or analytical methods.

Substitution with heavy isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements, and thus may be preferred in some circumstances. (see, e.g., A.Kerekes et al J.Med.chem.2011,54, 201-. In particular, substitution at one or more metabolic sites may provide one or more of the therapeutic advantages.

Thus, in some embodiments, the inhibitor of JAK1 and/or JAK2 is a compound wherein one or more hydrogen atoms in the compound are replaced with a deuterium atom, or a pharmaceutically acceptable salt thereof.

In some embodiments, the inhibitor of JAK1 and/or JAK2 is ruxolitinib in which one or more hydrogen atoms are replaced with a deuterium atom, or a pharmaceutically acceptable salt thereof. In some embodiments, the inhibitor of JAK1 and/or JAK2 is any of the compounds of U.S. patent 9249149 (which is incorporated herein by reference in its entirety), or a pharmaceutically acceptable salt thereof. In some embodiments, the inhibitor of JAK1 and/or JAK2 is CTP-543 or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound is a compound of formula I:

or a pharmaceutically acceptable salt thereof, wherein:

R1selected from H and D;

each R2Independently selected from H and D, provided that each R is attached to a common carbon2The same;

each R3Independently selected from H and D, provided that each R is attached to a common carbon3The same;

R4selected from H and D;

each R5Are the same and are selected from H and D; and is

R6、R7And R8Each is independently selected from H and D; with the proviso that when R1Is H, each R2And each R3Is H, R4Is H, and R6、R7And R8Each R is H5Is D.

In some embodiments, the inhibitor of JAK1 and/or JAK2 is a compound of formula I (wherein R is selected from the following compounds 100-130 in the table below) (wherein R is6、R7And R8Each H) or a pharmaceutically acceptable salt thereof. In some embodiments, the inhibitor of JAK1 and/or JAK2 is a compound of formula I (wherein R is selected from the group consisting of compounds 200-231 in the following table (wherein R is)6、R7And R8Each being D) or a pharmaceutically acceptable salt thereof.

In some embodiments, the inhibitor of JAK1 and/or JAK2 is baricitinib, or a pharmaceutically acceptable salt thereof, in which one or more hydrogen atoms are replaced with a deuterium atom. In some embodiments, the inhibitor of JAK1 and/or JAK2 is any of the compounds of U.S. patent 9540367 (which is incorporated herein by reference in its entirety), or a pharmaceutically acceptable salt thereof.

As used herein, the phrase "optionally substituted" means unsubstituted or substituted. As used herein, the term "substituted" means that a hydrogen atom is removed and replaced with a substituent. It will be appreciated that substitution on a given atom is limited by valence.

As used herein, the term "C" used alone or in combination with other termsn-mAlkyl "refers to a saturated hydrocarbon group having n to m carbon atoms that may be straight or branched. In some embodiments, the alkyl group contains 1 to 6 or 1 to 3 carbon atoms. Examples of alkyl moieties include (but are not limited to) chemical groups such as: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-methyl-1-butyl, 3-pentyl, n-hexyl, 1,2, 2-trimethylpropyl and the like.

As used herein, the term "alkylene" used alone or in combination with other terms refers to a divalent alkyl linking group that may be branched or straight chain, wherein two substituents may be attached at any position of the alkylene linking group. Examples of alkylene groups include, but are not limited to, ethyl-1, 2-diyl, propyl-1, 3-diyl, propyl-1, 2-diyl, and the like.

As used herein, the term "HO-C1-3Alkyl "refers to a group of the formula-alkylene-OH, wherein the alkylene has from 1 to 3 carbon atoms.

As used herein, the term "CN-C1-3Alkyl "means C substituted by cyano1-3An alkyl group.

As used herein, the term "amino" refers to the formula-NH2A group of (1).

As used herein, the term "di (C)1-3Alkyl) amino "refers to the formula-N (alkyl)2Wherein each of the two alkyl groups independently has 1 to 3 carbon atoms.

As used herein, the term "C1-3Alkylamino "refers to a group of the formula-NH (alkyl), wherein alkyl has 1 to 3 carbon atoms.

As used herein, the term "di (C)1-3Alkyl) aminosulfonyl "refers to the formula-S (O)2N (alkyl)2Wherein each alkyl group independently has 1 to 3 carbon atoms.

As used herein, the term "C1-3Alkylsulfonyl "means a group of the formula-S (O)2-a group of groups wherein the alkyl group has 1 to 3 carbon atoms.

As used herein, "halo" or "halogen," used alone or in combination with other terms, includes fluorine, chlorine, bromine, and iodine. In some embodiments, the halo group is fluoro or chloro.

As used herein, the term "C" used alone or in combination with other termsn-mHaloalkyl "means C having up to {2(n to m) +1} halogen atoms which may be the same or differentn-mAn alkyl group. In some embodiments, the halogen atom is a fluorine atom. In some embodiments, the alkyl group has 1 to 6 or 1 to 3 carbon atoms. Examples of haloalkyl groups include CF3、C2F5、CHF2、CCl3、CHCl2、C2Cl5And so on. In thatIn some embodiments, the haloalkyl is a fluoroalkyl.

As used herein, the term "C1-3Fluoroalkyl "means C which may be partially or completely substituted by fluorine atoms1-3An alkyl group.

As used herein, the term "C" used alone or in combination with other terms3-6Cycloalkyl "refers to a non-aromatic monocyclic hydrocarbon moiety having 3 to 6 carbon atoms, which may optionally contain one or more alkenylene groups as part of a ring structure. One or more ring-forming carbon atoms of a cycloalkyl group can be oxidized to form a carbonyl bond. Exemplary C3-6Cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, and the like. In some embodiments, the cycloalkyl group is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.

As used herein, the term "C3-6cycloalkyl-C1-3Alkyl "means a group of the formula-C1-3alkylene-C3-6A group of cycloalkyl groups.

The compounds described herein may be asymmetric (e.g., having one or more stereocenters). Unless otherwise indicated, all stereoisomers, such as enantiomers and diastereomers, are intended. Compounds containing asymmetrically substituted carbon atoms may be isolated in optically active or racemic forms. Methods for preparing optically active forms from optically inactive starting materials, for example by resolution of racemic mixtures or by stereoselective synthesis, are known in the art. Many geometric isomers of alkenes, C ═ N double bonds, and the like, may also be present in the compounds described herein, and all such stable isomers are contemplated herein. Cis and trans geometric isomers of the compounds of the present application are described and may be separated as mixtures of isomers or as separate isomers. In some embodiments, the compound has the (R) -configuration. In some embodiments, the compound has the (S) -configuration.

Resolution of racemic mixtures of compounds can be carried out by any of a number of methods known in the art. An exemplary method includes fractional crystallization using a chiral resolving acid as the optically active salt-forming organic acid. Resolving agents suitable for use in the fractional crystallisation process are for example the D and L forms of optically active acids such as tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or a variety of optically active camphorsulphonic acids (e.g. β -camphorsulphonic acid). Other resolving agents suitable for fractional crystallization include stereoisomerically pure forms of alpha-methylbenzylamine (e.g., S and R forms, or diastereomerically pure forms), 2-phenylglycinol, norephedrine, ephedrine, N-methylephedrine, cyclohexylethylamine, 1, 2-diaminocyclohexane, and the like.

The resolution of the racemic mixture can also be carried out by elution on a column packed with an optically active resolving agent, for example dinitrobenzoylphenylglycine. Suitable elution solvent compositions may be determined by one skilled in the art.

The compounds described herein include tautomeric forms. The tautomeric form results from the exchange of a single bond with an adjacent double bond with concomitant proton migration. Tautomeric forms include proton transfer tautomers, which are isomeric protonated states with the same empirical formula and total charge. Examples of proton transfer tautomers include keto-enol pairs, amide-imide pairs, lactam-lactam pairs, enamine-imine pairs and cyclic forms in which the proton may occupy two or more positions of the heterocyclic ring system, such as 1H-imidazole and 3H-imidazole, 1H-1,2, 4-triazole, 2H-1,2, 4-triazole and 4H-1,2, 4-triazole, 1H-isoindole and 2H-isoindole, as well as 1H-pyrazole and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution. For example, it will be recognized that the following pyrazole rings may form two tautomers:

it is intended that the claims cover both tautomers.

All compounds and pharmaceutically acceptable salts thereof may be found together with, or isolated from, other materials such as water and solvents (e.g., hydrates and solvates).

In some embodiments, a compound described herein or a salt thereof is substantially isolated. By "substantially isolated" is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected. Partial isolation may include, for example, compositions enriched for the compounds described herein. Substantial separation may include compositions containing at least about 50 wt.%, at least about 60 wt.%, at least about 70 wt.%, at least about 80 wt.%, at least about 90 wt.%, at least about 95 wt.%, at least about 97 wt.%, or at least about 99 wt.% of a compound described herein, or a salt thereof. Methods for isolating compounds and salts thereof are conventional in the art.

The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

The expressions "ambient temperature" and "room temperature" as used herein are understood in the art and generally refer to a temperature, e.g., a reaction temperature of about the temperature of the room in which the reaction is conducted, e.g., about 20 ℃ to about 30 ℃.

The present application also includes pharmaceutically acceptable salts of the compounds described herein. As used herein, "pharmaceutically acceptable salts" refer to derivatives of the disclosed compounds wherein the parent compound is modified by converting an acid or base moiety present into its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, inorganic or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and so on. Pharmaceutically acceptable salts herein include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present application can be synthesized from the parent compound, which contains a basic or acidic moiety, by conventional chemical methods. In general, such salts can be prepared by reacting the free acid or free base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or an organic solvent, or a mixture of the two; in general, nonaqueous media, such as diethyl ether, ethyl acetate, alcohols (e.g. methanol, ethanol, isopropanol or butanol) or Acetonitrile (ACN) are preferred. A list of suitable salts is found in Remington's Pharmaceutical Sciences, 17 th edition, Mack Publishing Company, Easton, Pa.,1985, page 1418 and Journal of Pharmaceutical Science,66,2(1977), each of which is incorporated herein by reference in its entirety.

As used herein, the term "contacting" refers to bringing the indicated moieties together in an in vitro system or in an in vivo system. For example, "contacting" JAK with a compound of the invention includes administering a compound of the present application to an individual or patient, e.g., a human, having JAK, and, for example, introducing a compound of the invention into a sample containing JAK-containing cells or purified preparations.

As used herein, the terms "subject," "individual," or "patient" are used interchangeably to refer to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, pigs, cows, sheep, horses, or primates, and most preferably humans. In some embodiments, a "subject", "individual" or "patient" is in need of such treatment.

In some embodiments, the inhibitor is administered in a therapeutically effective amount. As used herein, the phrase "therapeutically effective amount" refers to the amount of active compound or pharmaceutical agent that elicits the biological or medical response in a tissue, system, animal, subject, or human that is being sought by a researcher, veterinarian, medical doctor, or other clinician.

As used herein, the term "treating" or "treatment" refers to one or more of the following: (1) inhibiting the disease; e.g., inhibiting the disease, disorder or condition (i.e., arresting further development of the disorder and/or condition) in an individual who is experiencing or exhibiting the disorder or condition of the disease, disorder or condition; (2) improving the disease; for example, ameliorating a disease, disorder or condition (i.e., reversing the pathology and/or symptomatology) in an individual who is experiencing or exhibiting the pathology or symptomatology of the disease, disorder or condition, e.g., reducing the severity of the disease; or (3) preventing the disease, disorder or condition in an individual who is predisposed to the disease, disorder or condition but has not yet experienced or exhibited the pathology or symptomatology of the disease. In some embodiments, treating refers to inhibiting or ameliorating a disease. In some embodiments, the treatment is prophylaxis of a disease.

Combination therapy

The methods described herein may further comprise administering one or more additional therapeutic agents. One or more additional therapeutic agents may be administered to the patient simultaneously or sequentially.

In some embodiments, the other therapeutic agent is an antibiotic. In some embodiments, the antibiotic is clindamycin (clindamycin), doxycycline (doxycline), minocycline (minocycline), trimethoprim (trimethoprim) -sulfamethoxazole (sulfamethoxazole), erythromycin (erythromycin), metronidazole (metronidazole), rifampin (rifampin), moxifloxacin (moxifloxacin), dapsone (dapsone), or a combination thereof. In some embodiments, the antibiotic is clindamycin, doxycycline, minocycline, trimethoprim-sulfamethoxazole, or erythromycin in combination with metronidazole. In some embodiments, the antibiotic is a combination of rifampicin, moxifloxacin, and metronidazole. In some embodiments, the antibiotic is a combination of moxifloxacin and rifampicin.

In some embodiments, the other therapeutic agent is a retinoid. In some embodiments, the retinoid is acitretin (etretinate), acitretin (acitretin), or isotretinoin (isotretinoin).

In some embodiments, the additional therapeutic agent is a steroid. In some embodiments, the other therapeutic agent is a corticosteroid. In some embodiments, the steroid is, for example, triamcinolone (triamcinolone), dexamethasone (dexamethasone), fluocinolone (fluocinolone), cortisone (cortisone), prednisone (prednisone), prednisolone (prednisone), or fluorometholone (flumetholone).

In some embodiments, the other therapeutic agent is an anti-TNF- α agent. In some embodiments, the anti-TNF- α agent is an anti-TNF- α antibody. In some embodiments, the anti-TNF-alpha agent is infliximab (infliximab) or etanercept (etanercept) or adalimumab (adalimumab).

In some embodiments, the additional therapeutic agent is an immunosuppressive agent. In some embodiments, the immunosuppressive agent is methotrexate (methotrexate) or cyclosporin a. In some embodiments, the immunosuppressant is mycophenolate (mycophenolate mofetil) or sodium mycophenolate (mycophenolate sodium).

In some embodiments, the other therapeutic agent is finasteride (finasteride), metformin (metformin), adapalene (adapalene), or azelaic acid (azelaic acid).

In some embodiments, the method further comprises administering an additional therapeutic agent selected from the group consisting of an IMiD, an anti-IL-6 agent, a methylation reducing agent, and a Biological Response Modifier (BRM).

Generally, BRMs are substances made from living organisms for the treatment of disease, which occur naturally in the body, or which can be made in the laboratory. Examples of BRMs include IL-2, interferon, various types of colony stimulating factors (CSF, GM-CSF, G-CSF), monoclonal antibodies (e.g., abciximab, etanercept, infliximab, rituximab, trastuzumab (trastuzumab)), and high-dose ascorbic acid.

In some embodiments, the methylation reducing agent is a DNA methyltransferase inhibitor. In some embodiments, the DNA methyltransferase inhibitor is selected from 5-azacytidine (5azacytidine) and decitabine (decitabine).

Typically, IMiD acts as an immunomodulator. In some embodiments, the IMiD is selected from thalidomide (thalidomide), lenalidomide (lenalidomide), pomalidomide (pomalidomide), CC-11006, and CC-10015.

In some embodiments, the method further comprises administering an additional therapeutic agent selected from the group consisting of: anti-thymocyte globulin, recombinant human granulocyte-colony stimulating factor (G CSF), granulocyte-monocyte CSF (GM-CSF), Erythropoiesis Stimulating Agent (ESA), and cyclosporine.

In some embodiments, the method further comprises administering to the patient an additional JAK inhibitor. In some embodiments, the other JAK inhibitor is baricitinib, tofacitinib, olatinib, fegolitinib, gandotinib, lestatinib, momertinib, baritricitinib, PF-04965842, upatinib, pefinitib, feitinib, cucurbitacin I, or CHZ 868.

One or more other drugs, such as anti-inflammatory agents, immunosuppressive agents, and PI3K δ, mTor, Bcr-Abl, Flt-3, RAF, and FAK kinase inhibitors, such as those described in WO 2006/056399 (which is incorporated herein by reference in its entirety), or other agents, may be used in combination with the compounds described herein for the treatment of a JAK-associated disease, disorder, or condition. One or more other medications may be administered to the patient simultaneously or sequentially.

Examples of Bcr-Abl inhibitors include compounds of the class disclosed in U.S. patent No.5,521,184, WO 04/005281, and U.S. serial No. 60/578,491 (all incorporated herein by reference in their entirety), and pharmaceutically acceptable salts thereof.

Examples of suitable Flt-3 inhibitors include compounds as disclosed in WO 03/037347, WO 03/099771, and WO 04/046120 (all incorporated herein by reference in their entirety), and pharmaceutically acceptable salts thereof.

Examples of suitable RAF inhibitors include compounds as disclosed in WO 00/09495 and WO 05/028444 (both incorporated herein by reference in their entirety) and pharmaceutically acceptable salts thereof.

Examples of suitable FAK inhibitors include compounds as disclosed in WO 04/080980, WO 04/056786, WO 03/024967, WO 01/064655, WO 00/053595 and WO 01/014402 (all incorporated herein by reference in their entirety), and pharmaceutically acceptable salts thereof.

In some embodiments, one or more compounds of the invention may be used in combination with one or more other kinase inhibitors, including imatinib (imatinib), particularly for treating patients resistant to imatinib or other kinase inhibitors.

In some embodiments, the other therapeutic agent is fluocinolone acetonide (fluocinolone acetonide,) Or rimexolone (AL-2178, Vexol, Alcon).

In some embodiments, the other therapeutic agent is a cyclosporin

In some embodiments, the additional therapeutic agent is selected from DehydrexTM(Holles Labs), cervamide (Civamide) (Opko), sodium hyaluronate (Vismed, Lantibio/TRB Chemea), cyclosporine (ST-603, silicon Therapeutics), ARG101(T) (testosterone, Argentis), AGR1012(P) (Argentis), ecabet sodium (Senju-Ista), gefarnate (Santen), 15- (S) -hydroxyeicosatetraenoic acid (15(S) -HETE), cevimeline (cevilamine), doxycycline (ALTY-0501, Alacrity), minocycline, iDestrinTM(NP50301, sodium Pharmaceuticals), cyclosporin A (Nova22007, Novagali), oxytetracycline (duramycin, MOLI1901, lantibiio), CF101(2S,3S,4R,5R) -3, 4-dihydroxy-5- [6- [ (3-iodophenyl) methylamino)]Purin-9-yl]N-methyl-oxacyclopentane-2-carbamoyl, Can-Fine Biopharma), Volvosporin (voclosporin) (LX212 or LX214, Lux Biosciences), ARG103(Agentis), RX-10045 (synthetic resolvin analogue, Resolvyx), DYN15 (Dynamis Therapeutics), rivoglitazone (DE011, Daiichi Sanko), TB4(RegeneRx), OPH-01(Ophtalmis monoaco), PCS101(Pericor Science), REV1-31(Evolutec), tear protein (Lacritin) (Senju), rebamipide (Otsubaka-Novartis), Othera, PAI-2 (Unisituof Tenuifolia), Tetrakis (Pevolutid), Pentium (Klinesirolimus, Klinemia), Immunorubis (Klenofos), Tetrakis-Novartis (Klenofos), Tetrakis-551 (Othera), Pakis-2 (Unisitus of Tenuigrifficientolis), Tetrakis (Klinemia-1, Klinemia-novarus-ne (Klinemia, Piracoxis, Piracum (Piracum), Piracum) and Piracum (Piracum) for example, Piracum (Piracum) for the preparation of vaccine, Piracy, Piracum, Piracy, Piracum, Dehydroepiandrosterone, anakinra, efavirenzab, mycophenolate sodium, etanerceptHydroxychloroquine, NGX267(TorreyPines Therapeutics), Yamera (actemra), gemcitabine(gemcitabine), oxaliplatin (oxaliplatin), L-asparaginase or thalidomide.

In some embodiments, the other therapeutic agent is an anti-angiogenic agent, a cholinergic agonist, a TRP-1 receptor modulator, a calcium channel blocker, a mucin secretagogue, a MUC1 stimulator, a calcineurin inhibitor, a corticosteroid, a P2Y2 receptor agonist, a muscarinic receptor agonist, an mTOR inhibitor, another JAK inhibitor, a Bcr-Abl kinase inhibitor, a Flt-3 kinase inhibitor, a RAF kinase inhibitor, and a FAK kinase inhibitor, such as those described in WO 2006/056399, which is incorporated herein by reference in its entirety. In some embodiments, the other therapeutic agent is a tetracycline derivative (e.g., minocycline or doxycycline). In some embodiments, the additional therapeutic agent is conjugated to FKBP 12.

In some embodiments, the other therapeutic agent is an alkylating agent or a DNA cross-linking agent; antimetabolites/demethylating agents (e.g., 5-fluorouracil (5-flurouracil), capecitabine (capecitabine), or azacitidine); anti-hormone therapy (e.g., hormone receptor antagonists, SERMs, or aromatase inhibitors); mitotic inhibitors (e.g., vincristine (vincristine) or paclitaxel (paclitaxel)); topoisomerase (I or II) inhibitors (e.g., mitoxantrone (mitoxantrone) and irinotecan (irinotecan)); apoptosis inducers (e.g., ABT-737); nucleic acid therapy (e.g., antisense or RNAi); nuclear receptor ligands (e.g., agonists and/or antagonists: all trans retinoic acid or bexarotene); epigenetic targeting agents, such as histone deacetylase inhibitors (e.g., vorinostat), methylation reducing agents (e.g., decitabine); protein stability modulators, such as Hsp90 inhibitors, ubiquitin and/or ubiquitin-like binding or disaggregating molecules; or an EGFR inhibitor (erlotinib).

In some embodiments, the other therapeutic agents include antibiotics, antivirals, antifungals, anesthetics, anti-inflammatory agents (including steroidal and non-steroidal anti-inflammatory agents), and antiallergic agents. Examples of suitable agents include aminoglycosides such as amikacin (amikacin), gentamicin (gentamycin), tobramycin (tobramycin), streptomycin (streptmycin), netilmicin (netilmycin), and kanamycin (kanamycin); quinolones, such as ciprofloxacin (ciprofloxacin), norfloxacin (norfloxacin), ofloxacin (ofloxacin), trovafloxacin (trovafloxacin), lomefloxacin (lomefloxacin), levofloxacin (levofloxacin) and enoxacin (enoxacin); naphthyridine; a sulfonamide; polymyxin (polymyxin); chloramphenicol (chloramphenicol); neomycin (neomycin); paromomycin (paramomycin); colistin mesylate (colistimethate); bacitracin (bacitracin); vancomycin (vancomycin); a tetracycline; rifampin and its derivatives ("rifampins"); a cyclic serine; a beta-lactam; cephalosporins (cephalosporins); amphotericin (amphotericin); fluconazole (fluconazole); flucytosine (flucytosine); natamycin (natamycin); miconazole (miconazole); ketoconazole (ketoconazole); a corticosteroid; diclofenac (diclofenac); flurbiprofen (flurbiprofen); ketorolac (ketorolac); suprofen (suprofen); cromolyn (cromolyn); lodoxamide (lodoxamide); levocabastine (levocabastin); a naphazoline; antazoline (antazoline); non-nilamin (pheniramine); or an azalide antibiotic.

Pharmaceutical formulations and dosage forms

When used as a medicament, the compounds of the invention may be administered in the form of a pharmaceutical composition. These compositions may be prepared in a manner well known in the pharmaceutical art and may be administered by a variety of routes depending on whether local or systemic treatment is desired and the area to be treated. Administration can be topical (including transdermal, epidermal, ocular, and mucosal, including intranasal, vaginal, and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal), oral, or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal, intramuscular administration or injection or infusion; or intracranial, e.g., intrathecal or intraventricular administration. Parenteral administration may be in the form of a single bolus dose, or may be, for example, by a continuous infusion pump. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be desirable or necessary.

In some embodiments, the administration is topical administration. In some embodiments, the application is topical application to the skin.

In some embodiments, the administration is oral administration.

The invention also includes pharmaceutical compositions containing a compound of the invention, or a pharmaceutically acceptable salt thereof, as an active ingredient, in combination with one or more pharmaceutically acceptable carriers (excipients). In some embodiments, the composition is suitable for topical administration. In making the compositions of the present invention, the active ingredient is typically mixed with an excipient, diluted by an excipient, or enclosed within such a carrier, for example, in the form of a capsule, sachet, paper or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material that serves as a vehicle, carrier, or medium for the active ingredient. Thus, the compositions may be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders containing, for example, up to 10% by weight of the active compound.

In preparing the formulations, the active compound may be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it may be ground to a particle size of less than 200 mesh. If the active compound is substantially soluble in water, the particle size may be adjusted by milling to provide a substantially uniform distribution in the formulation, for example about 40 mesh.

The compounds of the invention may be milled using known milling procedures, such as wet milling, to obtain a particle size suitable for tablet formation and other formulation types. Finely powdered (nanoparticulate) formulations of the compounds of the invention may be prepared by methods known in the art, see for example international application No. wo 2002/000196.

Some examples of suitable excipients include lactose, glucose, sucrose, sorbitol, mannitol, starch, acacia, calcium phosphate, alginate, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methylcellulose. The formulation may additionally comprise: lubricants, such as talc, magnesium stearate and mineral oil; a wetting agent; emulsifying and suspending agents; preservatives, such as methyl-and propylhydroxy-benzoate; a sweetener; and a flavoring agent. The compositions of the present invention may be formulated so as to provide rapid, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.

In some embodiments, the pharmaceutical composition comprises silicified microcrystalline cellulose (SMCC) and at least one compound described herein, or a pharmaceutically acceptable salt thereof. In some embodiments, the silicified microcrystalline cellulose comprises about 98% microcrystalline cellulose and about 2% silicon dioxide w/w.

In some embodiments, the composition is a sustained release composition comprising at least one compound described herein or a pharmaceutically acceptable salt thereof and at least one pharmaceutically acceptable carrier. In some embodiments, the composition comprises at least one compound described herein or a pharmaceutically acceptable salt thereof and at least one component selected from the group consisting of microcrystalline cellulose, lactose monohydrate, hydroxypropyl methylcellulose, and polyethylene oxide. In some embodiments, the composition comprises at least one compound described herein or a pharmaceutically acceptable salt thereof and microcrystalline cellulose, lactose monohydrate, and hydroxypropyl methylcellulose. In some embodiments, the composition comprises at least one compound described herein or a pharmaceutically acceptable salt thereof and microcrystalline cellulose, lactose monohydrate, and polyethylene oxide. In some embodiments, the composition further comprises magnesium stearate or silicon dioxide. In some embodiments, the microcrystalline cellulose is Avicel PH102TM. In some embodiments, the lactose monohydrate is Fast-flo 316TM. In some embodiments, the hydroxypropyl methylcellulose is hydroxypropyl methylcellulose 2208K4M (e.g., Methocel K4 mpremiert (tm)) and/or hydroxypropyl methylcellulose 2208K100LV (e.g., Methocel K00 LV)TM). In some embodiments, polyoxyEthylene oxide is polyoxyethylene WSR 1105 (e.g., Polyox WSR 1105)TM)。

In some embodiments, a wet granulation process is used to produce the composition. In some embodiments, a dry granulation process is used to produce the composition.

The compositions may be formulated in unit dosage forms, each dose containing from about 1 to about 1,000mg, from about 1mg to about 100mg, from about 1mg to about 50mg, and from about 1mg to 10mg of the active ingredient. Preferably, the dose is from about 1mg to about 50mg or from about 1mg to about 10mg of the active ingredient. In some embodiments, each dose contains about 10mg of the active ingredient. In some embodiments, each dose contains about 50mg of the active ingredient. In some embodiments, each dose contains about 25mg of the active ingredient. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.

In some embodiments, the composition comprises from about 1 to about 1,000mg, from about 1mg to about 100mg, from 1mg to about 50mg, and from about 1mg to 10mg of the active ingredient. Preferably, the composition comprises from about 1mg to about 50mg or from about 1mg to about 10mg of the active ingredient. One of ordinary skill in the art will appreciate that this is embodied as a compound or composition containing from about 1mg to about 10mg, from about 1mg to about 20mg, from about 1mg to about 25mg, from about 1mg to about 50mg of the active ingredient.

In some embodiments, the dose of the compound or pharmaceutically acceptable salt thereof is 15mg, 30mg, 60mg, or 90mg, based on the free base. In some embodiments, the dose of compound 4, or a pharmaceutically acceptable salt thereof, is 15mg, 30mg, 60mg, or 90mg, based on the free base. In some embodiments, the dose of the compound or pharmaceutically acceptable salt thereof is 15mg, as the free base. In some embodiments, the dose of the compound or pharmaceutically acceptable salt thereof is 30mg, as the free base. In some embodiments, the dose of the compound or pharmaceutically acceptable salt thereof is 60mg, as the free base. In some embodiments, the dose of the compound or pharmaceutically acceptable salt thereof is 90mg, as the free base.

The active compounds can be effective over a wide dosage range and are generally administered in a pharmaceutically effective amount. It will be appreciated, however, that the amount of the compound actually administered will generally be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the compound actually administered, the age, weight and response of the individual patient, the severity of the patient's symptoms, and the like.

To prepare solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of the compounds of the present application. When referring to these preformulation compositions as homogeneous, the active ingredient is generally dispersed uniformly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid pre-formulation is then subdivided into unit dosage forms of the type described above containing, for example, from about 0.1mg to about 1000mg of the active ingredient of the present application.

The tablets or pills of the present application may be coated or otherwise compounded to provide a dosage form affording the benefit of prolonged action. For example, a tablet or pill may comprise an inner dosage component and an outer dosage component, the latter being in the form of an envelope over the former. The two components may be separated by an enteric layer that serves to resist disintegration in the stomach and allows the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials may be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with, for example, shellac, cetyl alcohol and cellulose acetate.

The compounds and compositions of the present application may be incorporated in liquid forms for oral administration or injection including aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils (e.g., cottonseed oil, sesame oil, coconut oil, or peanut oil), as well as elixirs and similar pharmaceutical vehicles.

Compositions for inhalation or insufflation include solutions and suspensions or mixtures thereof in pharmaceutically acceptable aqueous or organic solvents, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described above. In some embodiments, the composition is administered by the oral or nasal respiratory route to achieve a local or systemic effect. The composition may be sprayed by using an inert gas. The nebulized solution may be breathed directly from the nebulizing device, or the nebulizing device may be attached to a face mask, a tent, or an intermittent positive pressure ventilator. Solution, suspension or powder compositions may be administered orally or nasally from a device that delivers the formulation in a suitable manner.

The topical formulations may contain one or more conventional carriers. In some embodiments, the ointment may contain water and one or more hydrophobic carriers selected from, for example, liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white petrolatum, and the like. The carrier composition of the cream may be based on water in combination with glycerin and one or more other components (e.g., glyceryl monostearate, PEG-glyceryl monostearate, and cetearyl alcohol). Gels can be formulated using isopropanol and water, suitably in combination with other components such as glycerol, hydroxyethyl cellulose and the like. In some embodiments, the topical formulation contains at least about 0.1 wt%, at least about 0.25 wt%, at least about 0.5 wt%, at least about 1 wt%, at least about 2 wt%, or at least about 5 wt% of a compound of the present invention. The topical formulation may be suitably packaged, for example in 100g tubes, optionally associated with instructions for the selected indication for treatment of, for example, psoriasis or other skin conditions.

The amount of a compound or composition administered to a patient will vary depending on the article of administration, the purpose of administration (e.g., prophylaxis or treatment), the state of the patient, the mode of administration, and the like. In therapeutic applications, the composition may be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. The effective dosage will depend on the disease condition being treated and on factors such as the severity of the disease, the age, weight and general condition of the patient, and the attending physician.

The composition administered to the patient may be in the form of a pharmaceutical composition as described above. These compositions may be sterilized by conventional sterilization techniques, or may be sterile filtered. The aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized formulation being combined with a sterile aqueous carrier prior to administration. The pH of the compound formulation will generally be between 3 and 11, more preferably 5 to 9 and most preferably 7 to 8. It will be appreciated that the use of certain of the above excipients, carriers or stabilizers will form a pharmaceutical salt.

The therapeutic dosage of the compounds of the present application may vary depending, for example, on the particular therapeutic use, the mode of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of a compound of the invention in a pharmaceutical composition may vary depending on a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration. For example, the compounds of the invention may be provided in an aqueous physiological buffer solution containing from about 0.1% w/v to about 10% w/v of the compound for parenteral administration. Some typical dosages range from about 1 μ g to about 1g per kilogram of body weight per day. In some embodiments, the dosage range is from about 0.01mg to about 100mg per kg of body weight per day. The dosage will likely depend upon variables such as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the selected compound, the formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.

The compositions of the invention may also include one or more other drugs, examples of which are listed above.

Medicine box

The present application also includes kits useful, for example, in the treatment and/or prevention of cytokine-related diseases or disorders, such as CRS, comprising one or more containers containing pharmaceutical compositions comprising a therapeutically effective amount of a compound described herein. Such kits may also include, if desired, one or more of a variety of conventional kit components, e.g., containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be apparent to those of skill in the art. Instructions for the administration of the components, the guidelines for administration, and/or the guidelines for mixing the components may also be included in the kit, as an insert or as a label.

Examples

The present invention will be described in more detail by way of specific examples. The following examples are provided for illustrative purposes and are not intended to limit the invention in any way. Those skilled in the art will readily recognize a number of non-critical parameters that may be varied or modified to yield substantially the same results.

Example A: in vitro JAK kinase assay

Inhibitors of JAK1 useful for the treatment of cytokine-related diseases or disorders were tested for inhibitory activity against JAK targets according to the following in vitro assay described in Park et al, Analytical Biochemistry 1999,269, 94-104. Catalytic domains of human JAK1(a.a.837-1142), JAK2(a.a.828-1132) and JAK3(a.a.781-1124) with an N-terminal His-tag were expressed and purified in insect cells using baculovirus. The catalytic activity of JAK1, JAK2 or JAK3 was determined by measuring phosphorylation of biotinylated peptides. Phosphorylated peptides were detected by Homogeneous Time Resolved Fluorescence (HTRF). IC of compound for each kinase was measured in 40. mu.L of reaction50The reaction contained the enzyme, ATP and 500nM peptide in 50mM Tris (pH 7.8) buffer with 100mM NaCl, 5mM DTT and 0.1mg/mL (0.01%) BSA. For 1mM IC50ATP concentration in the reaction was measured to be 1 mM. The reaction was performed at room temperature for 1 hour, followed by termination in assay buffer (Perkin Elmer, Boston, MA) with 20. mu.L 45mM EDTA, 300nM SA-APC, 6nM Eu-Py 20. Binding to europium-labeled antibody was performed for 40 minutes and HTRF signal was measured on a Fusion plate reader (Perkin Elmer, Boston, MA). The compounds in Table 1 were tested in this assay and shown to have the IC's in Table 150The value is obtained.

Example B: studies of safety and efficacy of JAK1 and/or JAK2 inhibitors in subjects with moderate to severe hidradenitis suppurativa

Randomized, double-blind, placebo-controlled, multicenter studies were conducted for at least 6 months in men and women aged 18-75 years with moderate (Hurley) stage II) to severe (Hurley stage III) hidradenitis suppurativa. Stage helley I is associated with abscess formation (single or multiple), with no sinus and scarring. Stage II of helley is associated with recurrent abscesses, sinus tract formation and scarring; single or multiple widely separated lesions. The hercules stage III is associated with diffuse or near diffuse involvement throughout the area or multiple interconnected sinuses and abscesses. Study participants were randomized into 5 groups (about 50 participants per group) and treated with 15mg, 30mg, 60mg, or 90mg JAK1 and/or an inhibitor of JAK2 (e.g., ruxolitinib, compound 4, or compound 5, or a pharmaceutically acceptable salt thereof) or placebo. At week 16 (primary endpoint), participants in the placebo group were equally re-randomized to the active treatment group for 8 weeks. The blind approach is maintained. The primary endpoint was the proportion of subjects who achieved the hidradenitis suppurativa clinical response (HiSCR) at week 16.

The secondary endpoints included: (1) the proportion of subjects with HiSCR compared to baseline at each visit; (2) proportion of subjects who obtained 0 to 2 abscess and inflammatory nodule (AN) counts per visit; (3) mean change from baseline in HS pain numerical rating scale 1) at each visit; (4) changes in modified Sartorius scale (Sartorius scale) at weeks 16 and 24; (5) a change in the number of drainage fistulas counted per visit; (6) the proportion of subjects requiring damage rescue treatment by week 24; (7) the number of events that damaged rescue treatment by week 24; (8) a population PK of inhibitors of JAK1 and/or JAK2 (e.g. apparent clearance, apparent volume of distribution); (9) safety and tolerability assessed by monitoring the frequency, duration and severity of AEs, physical examinations, vital signs, and laboratory data of hematology, serum chemistry, and urinalysis; (10) a change in Dermatological Life Quality Index (DLQI) assessment; (11) change in disease severity from baseline at each visit as assessed by IHS43 score; (12) changes from baseline in hidradenitis suppurativa quality of life (HiSQOL) assessments at each visit; and (13) assessment of percent change in dose/exposure response from baseline during the treatment period in terms of efficacy and safety endpoints.

HiSCR was defined as at week 16, at least 50% decrease in abscess and inflammatory nodule (AN) counts relative to baseline, no increase in abscess counts and no increase in drainage fistula counts. The pain numerical rating scale was used to assess the worst skin pain and average skin pain caused by HS. The rating of 2 items ranged from 0 (no skin pain) to 10 (skin pain to as severe as you could imagine). The assessment was recorded on a diary by the participants prior to going to bed and based on a "24 hour before" recall period. A modified version of the sidoris scale was used to quantify HS severity. Scores were given to 12 body regions (left and right axilla, left and right inframammary/inframammary regions, intercalary region, left and right buttocks, left and right inguinal folds, perianal, perineum and others) with scores: scores were given to nodules (2 points each); abscess (4 points); fistula (4 min); scars (1 point); and the longest distance between two lesions (2-6 points, 0 if no lesions are present); and whether the lesions were separated from normal skin (score-0; score-6). The total sartorius scale is the sum of 12 area scores. Lesion rescue treatment: if a sharply painful lesion requires immediate intervention, the physician chooses to perform a rescue intervention. Only two types of intervention are allowed: (1) intralesional injection of triamcinolone acetonide suspension (up to 30mg total at the same visit) and/or (2) incision and pus evacuation. The intervention can be performed on a maximum of two different lesions at the same visit or on the same lesion at two different study visits. The same lesion may not be treated twice at the same visit. The study was discontinued if the subject required more than two interventions before week 16. International hidradenitis suppurativa severity scoring system (IHS 4): IH4 (min) ═ (number of nodules × 1) + (number of abscesses × 2) + (number of excretory tracts [ fistulas/sinuses ] × 4). Mild HS: less than or equal to 3 minutes; moderate HS: 4-10 min; severe HS: not less than 11 points.

Study treatment 1 (active agent) included an oral tablet containing 15mg of 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide. The administration levels included 15mg (1 tablet), 30mg (2 tablets), 60mg (4 tablets) and 90mg (6 tablets). Study treatment 2 (placebo) included an oral tablet placebo.

Blood samples for measuring plasma concentrations of inhibitors of JAK1 and/or JAK2 were taken at least at weeks 2, 12, 16, 20 and 24 at time points pre-, 1 and 2-5 hours post-dose before, after and after administration of study drug. If subjects discontinued before week 8, then at the time of premature discontinuation of visits, the cereal PK samples were collected (if applicable). The date/time of the previous dose administration is also recorded.

The tests for superiority of inhibitors of JAK1 and/or JAK2 at 90mg, 60mg, 30mg and 15mg compared to placebo were performed at a global 2-side α -0.05 level using the Hochberg procedure (Hochberg procedure). Comparisons between each active group and placebo at week 16 were made using logistic regression. At all dose levels, the superiority test was significant (e.g. a 10%, 20%, 30%, 40% or 50% improvement in HiSCR (hidradenitis suppurativa clinical response)) and demonstrated the efficacy of inhibitors of JAK1 and/or JAK2 in the treatment of HS. The tests showed a reduction in nodules and no disadvantages/advantages compared to placebo.

All secondary and exploratory efficacy measures were evaluated using descriptive statistics. Clinical safety data (vital signs, routine laboratory tests and AE) were analyzed using descriptive statistics. Determining an exposure-response (E-R) relationship between plasma JAK1 and/or JAK2 inhibitor PK exposure and efficacy/safety data. When at least half of the randomized subjects reached week 16, interim analyses were performed to estimate treatment response and drive future study plans.

Example C. Interferon-gamma and tumor necrosis-alpha induced Janus kinase expression in keratinocytes and subsequent production of inflammatory mediators

Transformed human keratinocytes (HaCaT) were purchased from AddexBio (Cat. No. T0020001) and cultured in Optimized Darbeck Modified Eagle's Medium (AddexBio, Cat. No. C0003-02) supplemented with 10% fetal bovine serum (Hyclone, Cat. No. 16140-. When the cells reached 80% -90% confluence, they were washed with 1 XDPBS followed by treatment with 0.25% trypsin (Gibco, Cat. 25200-056) at 37 ℃/5% CO2Incubated for 3-5 minutes and isolated from the tissue culture flasks. Cell culture medium was added to the trypsinized cells, and the cell suspension was transferred to a sterile 15mL centrifuge tube and briefly centrifuged at 1300rpm for 10 minutes. The trypsin-containing medium was aspirated from the cell pellet, followed by resuspension of the pellet in 10mL of cell culture medium. Counting Using Countess II automated cell counterCells and at 4X 104Individual cells/ml were seeded into tissue culture medium-treated 24-well plates and incubated at 37 deg.C/5% CO2Incubation was performed for 48 hours. After 48 hours, the medium was removed and replaced with 500uL of cell culture medium or recombinant human interferon gamma (R)&D Systems, Catalogue No. 285-IF-100) and recombinant human tumor necrosis factor alpha (R)&DSystems, catalog number 210-TA-020). HaCaT cells treated with combined cytokine stimulation were treated at final concentrations of each cytokine of 10ng/mL, 25ng/mL, 50ng/mL, or 100 ng/mL. The treated plates were mixed by gentle agitation for 30 seconds, followed by 37 deg.C/5% CO2Incubate for 24 hours. At the end of the 24 hour incubation, the medium was immediately removed from each disc.

RNA was isolated from HaCaT cells using the QuantiGene Plex assay reagents and protocol (Affymetrix, cat # QGP-232-M18042302). Cells were washed with 1x DPBS and then lysed by incubation with QuantiGene lysis buffer provided for 30 min at 50-55 ℃. The cell lysate is incubated with a set of capture beads and probes designed to specifically hybridize to mRNA from the target of interest for 18-24 hours at 55 ℃. This set of 32 targets of interest included housekeeping genes used to normalize the results. After 18-24 hours of incubation, the signal was amplified using the branched-chain DNA method according to the manufacturer's protocol (Affymetrix, cat. No. QGP-232-M18042302). After the hybridization and washing steps, assay plates were read on Luminex 200 and the data can be expressed as net median fluorescence intensity. The data were then normalized to the net median fluorescence intensity of the housekeeping gene HPRT1 (table 2).

TABLE 2 stimulation of human keratinocytes with TNF α and IFN γ induces JAK/STAT pathways and proinflammatory cytokines

Target proteins of interest in the medium were detected and quantified using ProCarta Multiplex immunoassay reagents and protocol (Invitrogen, Cat. No. EPX 450-12171-901). The medium is incubated with antibody-conjugated beads designed to bind to an epitope of a specific target protein and the bound protein is identified by the bead's unique spectrogram. Biotinylated detection antibody designed to bind to different epitopes of the same target protein and streptavidin-PE were added to the assay plate to quantify the amount of target protein. Assay plates were read on a Luminex 200 and data can be expressed as net median fluorescence intensity. The net median fluorescence intensity values of the antigen standard curves prepared according to the manufacturer's procedure (Invitrogen, catalog number EPX450-12171-901) were plotted against the expected concentration for each standard. The concentration of each protein was extrapolated from the antigen standard curve and the concentration can be expressed as pg/mL (table 3).

TABLE 3 stimulation of human keratinocytes with TNF α and IFN γ to induce proinflammatory cytokine production

Example D.Janus kinase inhibitors interfere with Interferon-gamma and tumor necrosis-alpha mediated inflammation in keratinocytes

Transformed human keratinocytes (HaCaT) were purchased from adexbio (catalog No. T0020001) and cultured as outlined in example C. Four compounds A-D (A: ruxolitinib, B: itatinib) ({1- {1- [ 3-fluoro-2- (trifluoromethyl) isonicotinyl)]Piperidin-4-yl } -3[4- (7H-pyrrolo [2, 3-d)]Pyrimidin-4-yl) -1H-pyrazol-1-yl]Azetidine-3-Methyl } acetonitrile), C: 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl]-2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl]Benzamide, D: ((2R,5S) -5- {2- [ (1R) -1-hydroxyethyl]-1H-imidazo [4,5-d]Thieno [3,2-b]Pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile) in DMSO, followed by serial dilution of each compound with cell culture medium to 400nM, 200nM, 100nM, and 50nM concentrations. After 48 hours, the cell culture medium was removed from the 24-well plate and replaced with 250uL of medium containing serially diluted drug, followed by 37 ℃/5% CO2Incubate for 15 minutes. After drug incubation, 250uL containing recombinant human interferon gamma (R)&D Systems, Catalogue No. 285-IF-100) and recombinant human tumor necrosis factor alpha (R)&D Systems, catalog number 210-TA-020) was added to the plate. The final concentration of recombinant human interferon gamma and recombinant human tumor necrosis factor alpha was 25ng/mL of each cytokine. Cytokine stimulation added to drug-containing wells resulted in final concentrations of 25nM, 50nM, 100nM and 200nM for each drug treatment. The treated plates were mixed by gentle agitation for 30 seconds, followed by 37 deg.C/5% CO2Incubate for 24 hours. At the end of the 24 hour incubation, the medium was immediately removed from each plate.

RNA was isolated from HaCaT cells using QuantiGene Plex assay reagents and protocols (Affymetrix, cat # QGP-232-M18042302) according to the manufacturer's guidelines. Cells were washed with 1x DPBS and then lysed by incubation with QuantiGene lysis buffer provided for 30 min at 50-55 ℃. The cell lysate is incubated with a set of capture beads and probes designed to specifically hybridize to mRNA from the target of interest for 18-24 hours at 55 ℃. Genes include housekeeping genes (e.g., HPRT1, GAPDH) used to normalize results. After 18-24 hours of incubation, the signal was amplified using the branched-chain DNA method according to the manufacturer's protocol (Affymetrix, cat. No. QGP-232-M18042302). After the hybridization and washing steps, assay plates were read on Luminex 200 and the data can be expressed as net median fluorescence intensity. The data were then normalized to the net median fluorescence intensity of the housekeeping gene HPRT1 (table 4).

TABLE 4 normalized expression of target genes in human keratinocytes stimulated with TNF α and IFN γ in the presence/absence of JAK inhibitors

FIGS. 1-4 illustrate the individual gene expression values (MFI) for JAK1, JAK2, IL-1 α, and IL-6 for each replicate in the presence/absence of JAK inhibitors in keratinocytes stimulated with TNF α and IFN- γ, respectively.

Target proteins of interest in the medium were detected and quantified using ProCarta Multiplex immunoassay reagents and protocol (Invitrogen, Cat. No. EPX 450-12171-901). The medium is incubated with antibody-conjugated beads designed to bind to an epitope of a specific target protein and the binding protein is identified by the bead's unique spectrogram. Biotinylated detection antibody designed to bind to different epitopes of the same target protein and streptavidin-PE were added to the assay plate to quantify the amount of target protein. Assay plates were read on a Luminex 200 and data can be expressed as net median fluorescence intensity. The net median fluorescence values of the antigen standard curves prepared according to the manufacturer's procedure (Invitrogen, catalog number EPX450-12171-901) were plotted against the expected concentration for each standard. The concentration of each protein was extrapolated from the antigen standard curve and the concentration can be expressed as pg/mL (table 5).

TABLE 5 concentration of inflammatory mediators produced by human keratinocytes stimulated with TNF α and IFN γ in the presence/absence of JAK inhibitors

FIGS. 5 and 6 illustrate the individual protein concentrations (pg/mL) of IL-1 α and IL-6 in each replicate in the presence/absence of JAK inhibitor in keratinocytes stimulated with TNF α and IFN- γ, respectively.

Example E: hidradenitis suppurativa skin biopsy characterized by increased Janus kinase expressionHealthy control skin total RNA from 3 single donors was purchased from Amsbio (cat # HR101 and R1234218-50). Healthy control skin total RNA from a pool of donors was purchased from Life Technologies (catalog number QS 0639). Hidradenitis suppurativa skin biopsies (41 donors) were purchased from Discovery Life Sciences as formalin fixed-paraffin embedded (FFPE) blocks from which total RNA was purified.

Gene expression was measured for healthy control (n-4) and hidradenitis suppurativa (n-41) skin total RNA samples using QuantiGene Plex assay reagents and protocols (Life Technologies, cat # QGP-277-M19012402) for the genes outlined in table 6. Purified RNA in the recommended assay range of 50ng to 500ng was used and incubated overnight with capture beads designed to specifically hybridize to mRNA from the selected gene (table 6). This set of targets included several housekeeping genes for normalizing the results. After overnight incubation, the signal was amplified using the branched-chain DNA method according to the manufacturer's procedure (Life Technologies). Assay plates were read on Luminex 200 and data can be expressed as net median fluorescence intensity (net MFI). Data were normalized to the geometric mean of the net MFI of the housekeeping genes ACTB and GAPDH. Figures 7-9 show gene expression of JAK1, JAK3, TYK2, STAT1, STAT2, STAT3, IRAK1, IRAK2, and IRAK4 in the skin of healthy controls and subjects with hidradenitis suppurativa.

TABLE 6 target genes

Various modifications of the invention in addition to those described herein will be apparent to those skilled in the art from the foregoing description. It is intended that such modifications be within the scope of the appended claims. Each reference cited in this application, including all patents, patent applications, and publications, is hereby incorporated by reference in its entirety.

In summary, the present invention includes, but is not limited to, the following technical items:

1. a method of treating hidradenitis suppurativa in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a compound that inhibits JAK1 and/or JAK2, or a pharmaceutically acceptable salt thereof, wherein the compound is:

ruxolitinib;

ruxolitinib in which one or more hydrogen atoms are replaced with deuterium atoms;

{1- {1- [ 3-fluoro-2- (trifluoromethyl) isonicotinyl ] piperidin-4-yl } -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

4- {3- (cyanomethyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-1-yl } -N- [ 4-fluoro-2- (trifluoromethyl) phenyl ] piperidine-1-carboxamide;

[3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] -1- (1- { [2- (trifluoromethyl) pyrimidin-4-yl ] carbonyl } piperidin-4-yl) azetidin-3-yl ] acetonitrile;

4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide;

((2R,5S) -5- {2- [ (1R) -1-hydroxyethyl ] -1H-imidazo [4,5-d ] thieno [3,2-b ] pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile;

3- [1- (6-chloropyridin-2-yl) pyrrolidin-3-yl ] -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] propionitrile;

3- (1- [1,3] oxazolo [5,4-b ] pyridin-2-ylpyrrolidin-3-yl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] propionitrile;

4- [ (4- { 3-cyano-2- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] propyl } piperazin-1-yl) carbonyl ] -3-fluorobenzonitrile;

4- [ (4- { 3-cyano-2- [3- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrrol-1-yl ] propyl } piperazin-1-yl) carbonyl ] -3-fluorobenzonitrile;

[ trans-1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] -3- (4- { [2- (trifluoromethyl) pyrimidin-4-yl ] carbonyl } piperazin-1-yl) cyclobutyl ] acetonitrile;

{ trans-3- (4- { [4- [ (3-hydroxyazetidin-1-yl) methyl ] -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- { [ (2S) -2- (hydroxymethyl) pyrrolidin-1-yl ] methyl } -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- { [ (2R) -2- (hydroxymethyl) pyrrolidin-1-yl ] methyl } -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

4- (4- {3- [ (dimethylamino) methyl ] -5-fluorophenoxy } piperidin-1-yl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] butanenitrile;

5- {3- (cyanomethyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-1-yl } -N-isopropylpyrazine-2-carboxamide;

4- {3- (cyanomethyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-1-yl } -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide;

5- {3- (cyanomethyl) -3- [4- (1H-pyrrolo [2,3-b ] pyridin-4-yl) -1H-pyrazol-1-yl ] azetidin-1-yl } -N-isopropylpyrazine-2-carboxamide;

{1- (cis-4- { [6- (2-hydroxyethyl) -2- (trifluoromethyl) pyrimidin-4-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{1- (cis-4- { [4- [ (ethylamino) methyl ] -6- (trifluoromethyl) pyridin-2-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{1- (cis-4- { [4- (1-hydroxy-1-methylethyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{1- (cis-4- { [4- { [ (3R) -3-hydroxypyrrolidin-1-yl ] methyl } -6- (trifluoromethyl) pyridin-2-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{1- (cis-4- { [4- { [ (3S) -3-hydroxypyrrolidin-1-yl ] methyl } -6- (trifluoromethyl) pyridin-2-yl ] oxy } cyclohexyl) -3- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile;

{ trans-3- (4- { [4- ({ [ (1S) -2-hydroxy-1-methylethyl ] amino } methyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- ({ [ (2R) -2-hydroxypropyl ] amino } methyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- ({ [ (2S) -2-hydroxypropyl ] amino } methyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

{ trans-3- (4- { [4- (2-hydroxyethyl) -6- (trifluoromethyl) pyridin-2-yl ] oxy } piperidin-1-yl) -1- [4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] cyclobutyl } acetonitrile;

or a pharmaceutically acceptable salt of any of the above compounds.

2. The method of claim 1, wherein the compound or salt is selective for JAK1 and JAK2 compared to JAK3 and TYK 2.

3. The method of claim 2, wherein the compound is ruxolitinib, or a pharmaceutically acceptable salt thereof.

4. The method of claim 3, wherein the compound is ruxolitinib, or a pharmaceutically acceptable salt thereof, wherein one or more hydrogen atoms are replaced with a deuterium atom.

5. The method of claim 3, wherein the salt is ruxolitinib phosphate.

6. The method of claim 1, wherein the compound or salt is selective for JAK1 as compared to JAK2, JAK3, and TYK 2.

7. The method of claim 6, wherein the compound is {1- {1- [ 3-fluoro-2- (trifluoromethyl) isonicotinyl ] piperidin-4-yl } -3[4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile or a pharmaceutically acceptable salt thereof.

8. The process of claim 7 wherein the salt is {1- {1- [ 3-fluoro-2- (trifluoromethyl) isonicotinoyl ] piperidin-4-yl } -3[4- (7H-pyrrolo [2,3-d ] pyrimidin-4-yl) -1H-pyrazol-1-yl ] azetidin-3-yl } acetonitrile adipate.

9. The method of claim 6, wherein the compound is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide, or a pharmaceutically acceptable salt thereof.

10. The method of claim 9, wherein the salt is 4- [3- (cyanomethyl) -3- (3',5' -dimethyl-1H, 1'H-4,4' -bipyrazol-1-yl) azetidin-1-yl ] -2, 5-difluoro-N- [ (1S) -2,2, 2-trifluoro-1-methylethyl ] benzamide phosphate.

11. The method of claim 6, wherein the compound is ((2R,5S) -5- {2- [ (1R) -1-hydroxyethyl ] -1H-imidazo [4,5-d ] thieno [3,2-b ] pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile or a pharmaceutically acceptable salt thereof.

12. The process according to claim 6, wherein the compound is ((2R,5S) -5- {2- [ (1R) -1-hydroxyethyl ] -1H-imidazo [4,5-d ] thieno [3,2-b ] pyridin-1-yl } tetrahydro-2H-pyran-2-yl) acetonitrile monohydrate.

13. The method of any one of claims 7-12, wherein the compound or salt is administered at a dose of 15mg, 30mg, 60mg, or 90mg as the free base.

14. The method of any one of claims 1-13, further comprising administering an additional therapeutic agent.

15. The method of claim 14, wherein the additional therapeutic agent is an antibiotic, a retinoid, a corticosteroid, an anti-TNF-a agent, or an immunosuppressive agent.

16. The method of claim 15, wherein the antibiotic is clindamycin, doxycycline, minocycline, trimethoprim-sulfamethoxazole, erythromycin, metronidazole, rifampin, moxifloxacin, dapsone, or a combination thereof.

17. The method of claim 15, wherein the retinoid is acitretin, or isotretinoin.

18. The method of claim 15, wherein the corticosteroid is triamcinolone, dexamethasone, fluocinolone, cortisone, prednisone, prednisolone, or fluorometholone.

19. The method of claim 15, wherein the anti-TNF-alpha agent is infliximab, etanercept, or adalimumab.

20. The method of claim 15, wherein the immunosuppressive agent is methotrexate, cyclosporine a, mycophenolate mofetil, or mycophenolate sodium.

21. The method of claim 14, wherein the additional therapeutic agent is finasteride, metformin, adapalene, or azelaic acid.

22. The method of any one of claims 1-21, wherein the administration of the compound or salt is topical administration.

23. The method of any one of claims 1-21, wherein the administration of the compound or salt is oral administration.

24. The method of any one of claims 1-23, wherein the method increases HiSCR (hidradenitis suppurativa clinical response) by 10%, 20%, 30%, 40%, or 50%.

69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:一种速溶型纳米级生物素微胶囊及其制备方法和应用

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!