Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
THERAPEUTIC COMPOUNDS AND COMPOSITIONS, AND METHODS OF USE THEREOF
Document Type and Number:
WIPO Patent Application WO/2018/215389
Kind Code:
A1
Abstract:
Compounds and salts thereof that are useful as JAK kinse inhibitors are described herein. Also provided are pharmaceutical compositions that include such a JAK inhibitor and a pharmaceutically acceptable carrier, adjuvant or vehicle, and methods of treating or lessening the severity of a disease or condition responsive to the inhibition of a Janus kinase activity in a patient.

Inventors:
ZAK MARK (US)
ROMERO F ANTHONY (US)
CHENG YUN-XING (CN)
Application Number:
PCT/EP2018/063262
Publication Date:
November 29, 2018
Filing Date:
May 21, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
HOFFMANN LA ROCHE (CH)
GENENTECH INC (US)
International Classes:
C07D487/04; A61K31/519; A61P29/00
Domestic Patent References:
WO2015177326A12015-11-26
WO2011003065A22011-01-06
WO2013014567A12013-01-31
WO2010051549A12010-05-06
WO2011003065A22011-01-06
WO2015177326A12015-11-26
WO2017089390A12017-06-01
WO1996015111A11996-05-23
WO2014074775A12014-05-15
WO1997003094A11997-01-30
WO1997002289A11997-01-23
WO2005026124A12005-03-24
WO2003053930A12003-07-03
WO2006082412A22006-08-10
WO2002042298A12002-05-30
Foreign References:
US20100048557A12010-02-25
US5263475A1993-11-23
EP1052264A22000-11-15
EP1241176A12002-09-18
Other References:
SCHINDLER ET AL., J. BIOL. CHEM., vol. 282, 2007, pages 20059 - 20063
WILKS A.F., PROC. NATL. ACAD. SCI. U.S.A., vol. 86, 1989, pages 1603 - 1607
KISSELEVA ET AL., GENE, vol. 285, 2002, pages 1 - 24
LEVY ET AL., NAT. REV. MOL. CELL BIOL., vol. 3, 2005, pages 651 - 662
O'SHEA ET AL., CELL, vol. 109, 2002, pages S121 - S131
O'SHEA ET AL., CELL, vol. 109, 2002, pages S121 - S131
SCHEINECKER ET AL., NAT. REV. DRUG DISCOV., vol. 8, 2009, pages 273 - 274
COHN ET AL., ANNU. REV. IMMUNOL., vol. 22, 2004, pages 789 - 815
KASAIAN ET AL., BIOCHEM. PHARMACOL., vol. 76, no. 2, 2008, pages 147 - 155
KEARLEY ET AL., AM. J. RESP. CRIT. CARE MED., vol. 183, no. 7, 2011, pages 865 - 875
PERNIS ET AL., J. CLIN. INVEST., vol. 109, no. 10, 2002, pages 1279 - 1283
DEMOULIN ET AL., MOL. CELL BIOL., vol. 16, no. 9, 1996, pages 4710 - 4716
HAAN ET AL., CHEM. BIOL., vol. 18, no. 3, 2011, pages 314 - 323
MATHEW ET AL., J. EXP. MED., vol. 193, no. 9, 2001, pages 1087 - 1096
KUDLACZ, EUR. J. PHARMACOL., vol. 582, no. 1-3, 2008, pages 154 - 161
O'SHEA ET AL., CELL, vol. 109, 2002, pages S121 - S131
BASLUND ET AL., ARTHRITIS & RHEUMATISM, vol. 52, 2005, pages 2686 - 2692
CHANGELIAN ET AL., SCIENCE, vol. 302, 2003, pages 875 - 878
WATFORD, W.T.; O'SHEA, J.J., IMMUNITY, vol. 25, 2006, pages 695 - 697
KRUEGER ET AL., N. ENGL. J. MED., vol. 356, 2007, pages 580 - 592
REICH ET AL., NAT. REV. DRUG DISCOV., vol. 8, 2009, pages 355 - 356
MANNON ET AL., N. ENGL. J. MED., vol. 351, 2004, pages 2069 - 2079
DEAN, J. A: "Lang's Handbook of Chemistry", 1985, article "Table 7-2"
S. P. PARKER: "McGraw-Hill Dictionary of Chemical Terms", 1984, MCGRAW-HILL BOOK COMPANY
ELIEL, E.; WILEN, S.: "Stereochemistry of Organic Compounds", 1994, JOHN WILEY & SONS, INC.
WILMAN: "Prodrugs in Cancer Chemotherapy", BIOCHEMICAL SOCIETY TRANSACTIONS, vol. 14, 1986, pages 375 - 382
STELLA ET AL.: "Directed Drug Delivery", 1985, HUMANA PRESS, article "Prodrugs: A Chemical Approach to Targeted Drug Delivery", pages: 247 - 267
FLEISHER, D. ET AL.: "Improved oral drug delivery: solubility limitations overcome by the use of prodrugs", ADVANCED DRUG DELIVERY REVIEWS, vol. 19, 1996, pages 115, XP002478093, DOI: doi:10.1016/0169-409X(95)00103-E
J. MED. CHEM., vol. 39, 1996, pages 10
LOUIS F. FIESER; MARY FIESER: "Reagents for Organic Synthesis", vol. 1-19, 1967, WILEY
"Beilsteins Handbuch der organischen Chemie", SPRINGER-VERLAG
"Comprehensive Heterocyclic Chemistry", 1984, PERGAMON PRESS
T. W. GREENE: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS
TETRAHEDRON, vol. 61, 2005, pages 10827 - 10852
COMPREHENSIVE ORGANIC NAME REACTIONS AND REAGENTS, 2010, pages 575 - 581
CHEMICAL REVIEWS, vol. 95, no. 7, 1995, pages 2457 - 2483
J. AM. CHEM. SOC., vol. 136, 2014, pages 4149 - 4152
LOCHMULLER, C. H., J. CHROMATOGR., vol. 113, no. 3, 1975, pages 283 - 302
"Drug Stereochemistry, Analytical Methods and Pharmacology", 1993, MARCEL DEKKER, INC.
ELIEL, E.; WILEN, S.: "Stereochemistry of Organic Compounds", 1994, JOHN WILEY & SONS, INC., pages: 322
JACOB, J. ORG. CHEM., vol. 47, 1982, pages 4165
"Chiral Liquid Chromatography", 1989, CHAPMAN AND HALL
OKAMOTO, J. OF CHROMATOGR., vol. 513, 1990, pages 375 - 378
ANSEL, HOWARD C. ET AL.: "Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems", 2004, LIPPINCOTT, WILLIAMS & WILKINS
GENNARO, ALFONSO R. ET AL.: "Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT, WILLIAMS & WILKINS
ROWE, RAYMOND C.: "Handbook of Pharmaceutical Excipients.", 2005, PHARMACEUTICAL PRESS
REMINGTON'S PHARMACEUTICAL SCIENCES, 1990, pages 1289 - 1329
COOPER, A. E. ET AL., CURR. DRUG METAB., vol. 13, 2012, pages 457 - 473
HALLADAY ET AL., DRUG METAB. LETT., vol. 5, 2011, pages 220 - 230
MORRISON, J.F., BIOCHIM. BIOPHYS. ACTA, vol. 185, 1969, pages 269 - 296
WILLIAM, J.W.; MORRISON, J.F., METH. ENZYMOL., vol. 63, 1979, pages 437 - 467
MATHEW ET AL., J EXP MED, vol. 193, no. 9, 2001, pages 1087 - 1096
KUDLACZ, EUR J. PHARMACOL, vol. 582, no. 1-3, 2008, pages 154 - 161
Attorney, Agent or Firm:
SCHIRLIN, Julien (CH)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A compound of Formula (I)

or a pharmaceutically acceptable salt or stereoisomer thereof, wherein:

R1 is hydrogen or CH3;

R is halogen, Ci-Cealkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, or - ORa, wherein R2 is optionally substituted by one or more groups independently selected from the group consisting of halogen, Ci-C3alkyl, cyano, hydroxy and oxo;

Ra is Ci-C6alkyl, -phenyl-CORbRc, -phenyl-(3-6-membered heterocyclyl), or 3-11-membered heterocyclyl, wherein Ra is optionally substituted by one or more groups independently selected from the group consisting of halogen, Ci-C3alkyl, cyano, hydroxy and oxo;

Rb and Rc are each independently hydrogen or CH3;

R is hydrogen or NH2;

R4 is hydrogen or CH3; and

R5 is hydrogen or NH2.

2. The compound of claim 1 or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R1 is hydrogen.

3. The compound of claim 1 or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R1 is CH3.

4. The compound of any one of claims 1-3 or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R is hydrogen.

5. The compound of any one of claims 1-4 or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R4 and R5 are each hydrogen.

6. The compound of claim 1 or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R1, R3, R4 and R5 are each hydrogen.

7. The compound of any one of claims 1-6 or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R2 is Ci-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3- C6cycloalkyl, or -ORa, wherein R2 is optionally substituted by one or more groups independently selected from the group consisting of halogen, Ci-C3alkyl, cyano, hydroxy and oxo.

8. The compound of any one of claims 1-6 or a pharmaceutically acceptable salt or stereoisomer thereof, wherein R is selected from the group consisting of halogen, Ci-Cehaloalkyl and Ci-Cehaloalkoxy.

A compound selected from the group consisting of 1-18 below:

or a pharmaceutically acceptable salt or stereoisomer thereof.

1 1. A compound which is :

or a pharmaceutically acceptable salt thereof.

12. A compound which is :

or a pharmaceutically acceptable salt thereof.

13. A compound which is :

or a pharmaceutically acceptable salt thereof.

14. A compound which is :

or a pharmaceutically acceptable salt or stereoisomer thereof.

15. A compound which is :

or a pharmaceutically acceptable salt thereof.

16. A compound which is :

or a pharmaceutically acceptable salt or stereoisomer thereof.

17. A compound which is : or a pharmaceutically acceptable salt thereof.

18. A compound which is :

or a pharmaceutically acceptable salt thereof.

19. A compound which is :

or a pharmaceutically acceptable salt or stereoisomer thereof.

20. A compound which is:

or a pharmaceutically acceptable salt or stereoisomer thereof. 21. A compound which is :

or a pharmaceutically acceptable salt thereof.

22. A compound which is:

or a pharmaceutically acceptable salt thereof.

23. A compound which is :

or a pharmaceutically acceptable salt thereof.

24. A compound which is:

or a pharmaceutically acceptable salt thereof.

25. A compound which is :

or a pharmaceutically acceptable salt thereof.

26. A compound which is:

or a pharmaceutically acceptable salt thereof.

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

28. Use of a compound of any of claims 1-26 or a pharmaceutically acceptable salt or stereoisomer thereof in therapy.

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

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

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

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

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

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

35. The method of claim 33, wherein the Janus kinase is JAK1.

36. The invention as in hereinbefore described.

Description:
THERAPEUTIC COMPOUNDS AND COMPOSITIONS, AND

METHODS OF USE THEREOF

CROSS-REFERENCE TO RELATED APPLICATIONS This application claims the benefit of priority to U.S. Application Serial No.

62/640,865, filed March 9, 2018, and International Patent Application No.

PCT/CN2017/085276, filed May 22, 2017, each of which is incorporated by reference herein in its entirety.

FIELD OF THE INVENTION

The invention relates to compounds that are inhibitors of a Janus kinase, such as JAK1, as well as compositions containing these compounds, and methods of use including, but not limited to, diagnosis or treatment of patients suffering from a condition responsive to the inhibition of a JAK kinase. BACKGROUND OF INVENTION

Cytokine pathways mediate a broad range of biological functions, including many aspects of inflammation and immunity. Janus kinases (JAK), including JAK1, JAK2, JAK3 and TYK2, are cytoplasmic protein kinases that associate with type I and type II cytokine receptors and regulate cytokine signal transduction. Cytokine engagement with cognate receptors triggers activation of receptor associated JAKs and this leads to JAK-mediated tyrosine phosphorylation of signal transducer and activator of transcription (STAT) proteins and ultimately transcriptional activation of specific gene sets (Schindler et al, 2007, J. Biol. Chem. 282: 20059-63). JAK1, JAK2 and TYK2 exhibit broad patterns of gene expression, while JAK3 expression is limited to leukocytes. Cytokine receptors are typically functional as heterodimers, and as a result, more than one type of JAK kinase is usually associated with cytokine receptor complexes. The specific JAKs associated with different cytokine receptor complexes have been determined in many cases through genetic studies and corroborated by other experimental evidence. Exemplary therapeutic benefits of the inhibition of JAK enzymes are discussed, for example, in International Application No. WO 2013/014567.

JAK1 was initially identified in a screen for novel kinases (Wilks A.F., 1989, Proc. Natl. Acad. Sci. U.S.A. 86: 1603-1607). Genetic and biochemical studies have shown that JAK1 is functionally and physically associated with the type I interferon (e.g., IFNalpha), type II interferon (e.g., IFNgamma), and IL-2 and IL-6 cytokine receptor complexes (Kisseleva et al, 2002, Gene 285: 1-24; Levy et al, 2005, Nat. Rev. Mol. Cell Biol. 3:651-662; O'Shea et al, 2002, Cell, 109 (suppl): S121-S131). JAK1 knockout mice die perinatally due to defects in LIF receptor signaling

(Kisseleva et al, 2002, Gene 285: 1-24; O'Shea et al, 2002, Cell, 109 (suppl): S121- S131). Characterization of tissues derived from JAK1 knockout mice demonstrated critical roles for this kinase in the IFN, IL-10, IL-2/IL-4 and IL-6 pathways. A humanized monoclonal antibody targeting the IL-6 pathway (Tocilizumab) was approved by the European Commission for the treatment of moderate -to-severe rheumatoid arthritis (Scheinecker et al., 2009, Nat. Rev. Drug Discov. 8:273-274).

CD4 T cells play an important role in asthma pathogenesis through the production of TH2 cytokines within the lung, including IL-4, IL-9 and IL-13 (Cohn et al, 2004, Annu. Rev. Immunol. 22:789-815). IL-4 and IL-13 induce increased mucus production, recruitment of eosinophils to the lung, and increased production of IgE (Kasaian et al., 2008, Biochem. Pharmacol. 76(2): 147-155). IL-9 leads to mast cell activation, which exacerbates the asthma symptoms (Kearley et al., 2011, Am. J. Resp. Crit. Care Med., 183(7): 865-875). The IL-4Ra chain activates JAK1 and binds to either IL-4 or IL-13 when combined with the common gamma chain or the IL-13Ral chain respectively (Pernis et al, 2002, J. Clin. Invest. 109(10): 1279-1283). The common gamma chain can also combine with IL-9Ra to bind to IL-9, and IL- 9Ra activates JAK1 as well (Demoulin et al, 1996, Mol. Cell Biol. 16(9):4710- 4716). While the common gamma chain activates JAK3, it has been shown that JAK1 is dominant over JAK3, and inhibition of JAK1 is sufficient to inactivate signaling through the common gamma chain despite JAK3 activity (Haan et al., 2011, Chem. Biol. 18(3):314-323). Inhibition of IL-4, IL-13 and IL-9 signaling by blocking the JAK/STAT signaling pathway can alleviate asthmatic symptoms in pre-clinical lung inflammation models (Mathew et al, 2001, J. Exp. Med. 193(9): 1087-1096; Kudlacz et. al, 2008, Eur. J. Pharmacol. 582(1-3): 154-161).

Biochemical and genetic studies have shown an association between JAK2 and single-chain (e.g., EPO), IL-3 and interferon gamma cytokine receptor families (Kisseleva et al, 2002, Gene 285: 1-24; Levy et al, 2005, Nat. Rev. Mol. Cell Biol. 3:651-662; O'Shea et al, 2002, Cell, 109 (suppl): S121-S131). Consistent with this, JAK2 knockout mice die of anemia (O'Shea et al, 2002, Cell, 109 (suppl): S121- S131). Kinase activating mutations in JAK2 (e.g., JAK2 V617F) are associated with myeloproliferative disorders in humans.

JAK3 associates exclusively with the gamma common cytokine receptor chain, which is present in the IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21 cytokine receptor complexes. JAK3 is critical for lymphoid cell development and proliferation and mutations in JAK3 result in severe combined immunodeficiency (SCID) (O'Shea et al, 2002, Cell, 109 (suppl): S 121 -SI 31). Based on its role in regulating lymphocytes, JAK3 and JAK3-mediated pathways have been targeted for

immunosuppressive indications (e.g., transplantation rejection and rheumatoid arthritis) (Baslund et al., 2005, Arthritis & Rheumatism 52:2686-2692; Changelian et al, 2003, Science 302: 875-878).

TYK2 associates with the type I interferon (e.g., IFNalpha), IL-6, IL-10, IL-12 and IL-23 cytokine receptor complexes (Kisseleva et al, 2002, Gene 285: 1-24;

Watford, W.T. & O'Shea, J.J., 2006, Immunity 25:695-697). Consistent with this, primary cells derived from a TYK2 deficient human are defective in type I interferon, IL-6, IL-10, IL-12 and IL-23 signaling. A fully human monoclonal antibody targeting the shared p40 subunit of the IL-12 and IL-23 cytokines (Ustekinumab) was recently approved by the European Commission for the treatment of moderate -to-severe plaque psoriasis (Krueger et al, 2007, N. Engl. J. Med. 356:580-92; Reich et al, 2009, Nat. Rev. Drug Discov. 8:355-356). In addition, an antibody targeting the IL- 12 and IL-23 pathways underwent clinical trials for treating Crohn's Disease

(Mannon et al, 2004, N. Engl. J. Med. 351 :2069-79).

International Patent Application Publication Numbers WO 2010/051549, WO 2011/003065, WO 2015/177326 and WO 2017/089390 discuss certain

pyrazolopyrimidine compounds that are reported to useful as inhibitors of one or more Janus kinases. Data for certain specific compounds showing inhibition of JAKl as well as JAK2, JAK3, and/or TYK2 kinases is presented therein.

Currently there remains a need for additional compounds that are inhibitors of Janus kinases. For example, there is a need for compounds that possess useful potency as inhibitors of one or more Janus kinases (e.g., JAKl) in combination with other pharmacological properties that are necessary to achieve a useful therapeutic benefit. For example, there is a need for potent compounds that demonstrate selectivity for one Janus kinase over other kinases in general (e.g., selectivity for JAKl over other kinases such as leucine -rich repeat kinase 2 (LRRK2)). There is also a need for potent compounds that demonstrate selectivity for one Janus kinase over other Janus kinases (e.g., selectivity for JAKl over other Janus kinases). Kinases demonstrating selectivity for JAKl could provide a therapeutic benefit, with fewer side effects, in conditions responsive to the inhibition of JAKl . Additionally there is currently a need for potent JAKl inhibitors that possess other properties (e.g., melting point, pK, solubility, etc.) necessary for formulation and administration by inhalation. Such compounds would be particularly useful for treating conditions such as, for example, asthma.

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

SUMMARY OF THE INVENTION

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

One embodiment provides a compound of Formula (I):

or a stereoisomer or salt (e.g., a pharmaceutically acceptable salt) thereof, wherein:

R 1 is hydrogen or CH 3 ; R is halogen, Ci-Cealkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, or -

OR a , wherein R 2 is optionally substituted by one or more groups independently selected from the group consisting of halogen, Ci-C 3 alkyl, cyano, hydroxy and oxo;

R a is Ci-C 6 alkyl, -phenyl-COR b R c , -phenyl-(3-6-membered heterocyclyl), or 3-11-membered heterocyclyl, wherein R a is optionally substituted by one or more groups independently selected from the group consisting of halogen, Ci-C 3 alkyl, cyano, hydroxy and oxo; R b and R c are each independently hydrogen or CH 3 ; R is hydrogen or NH 2 ; R 4 is hydrogen or CH 3 ; and R 5 is hydrogen or NH 2 .

Also provided is a pharmaceutical composition comprising a JAK inhibitor as described herein, or a pharmaceutically acceptable salt thereof, and a

pharmaceutically acceptable carrier, dilient or excipient.

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

Certain compounds or salts thereof (e.g., pharmaceutically acceptable salts thereof) described herein possess beneficial potency as inhibitors of one or more Janus kinases (e.g., JAK1). Certain compounds or salts thereof (e.g.,

pharmaceutically acceptable salts thereof) are also a) selective for one Janus kinase over other kinases, b) selective for JAK1 over other Janus kinases, and/or c) possess other properties (e.g., melting point, pK, solubility, etc.) necessary for formulation and administration by inhalation. Certain compounds or salts thereof (e.g., pharmaceutically acceptable salts thereof) described herein may be particularly useful for treating conditions such as asthma.

DETAILED DESCRIPTION OF THE INVENTION

DEFINITIONS

"Halogen" or "halo" refers to F, CI, Br or I. Additionally, terms such as "haloalkyl," are meant to include monohaloalkyl and polyhaloalkyl, wherein one or more halogens replace a hydrogen(s) of an alkyl group.

The term "alkyl" refers to a saturated linear or branched-chain monovalent hydrocarbon radical, wherein the alkyl radical may be optionally substituted. In one example, the alkyl radical is one to eighteen carbon atoms (Ci-C 18 ). In other examples, the alkyl radical is Co-C 6 , C0-C5, C0-C3, C1-C12, Ci-Cio, Ci-C8, Ci-C 6 , Ci- C 5 , C 1 -C4, or C 1 -C 3 . Co alkyl refers to a bond. Examples of alkyl groups include methyl (Me, -CH 3 ), ethyl (Et, -CH 2 CH 3 ), 1 -propyl (n-Pr, n-propyl, -CH 2 CH 2 CH 3 ), 2- propyl (i-Pr, i-propyl, -CH(CH 3 ) 2 ), 1 -butyl (n-Bu, n-butyl, -CH 2 CH 2 CH 2 CH 3 ), 2- methyl-1 -propyl (i-Bu, i-butyl, -CH 2 CH(CH 3 ) 2 ), 2-butyl (s-Bu, s-butyl, - CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl (t-Bu, t-butyl, -C(CH 3 ) 3 ), 1-pentyl (n-pentyl, - CH 2 CH 2 CH 2 CH 2 CH 3 ), 2-pentyl (-CH(CH 3 )CH 2 CH 2 CH 3 ), 3-pentyl (-CH(CH 2 CH 3 ) 2 ),

2- methyl-2-butyl (-C(CH 3 ) 2 CH 2 CH 3 ), 3-methyl-2-butyl (-CH(CH 3 )CH(CH 3 ) 2 ), 3- methyl- 1 -butyl (-CH 2 CH 2 CH(CH 3 ) 2 ), 2-methyl- 1 -butyl (-CH 2 CH(CH 3 )CH 2 CH 3 ), 1 - hexyl (-CH 2 CH 2 CH 2 CH 2 CH 2 CH 3 ), 2-hexyl (-CH(CH 3 )CH 2 CH 2 CH 2 CH 3 ), 3-hexyl (- CH(CH 2 CH 3 )(CH 2 CH 2 CH 3 )), 2-methyl-2-pentyl (-C(CH 3 ) 2 CH 2 CH 2 CH 3 ), 3-methyl-2- pentyl (-CH(CH 3 )CH(CH 3 )CH 2 CH 3 ), 4-methyl-2-pentyl (-CH(CH 3 )CH 2 CH(CH 3 ) 2 ),

3- methyl-3-pentyl (-C(CH 3 )(CH 2 CH 3 ) 2 ), 2-methyl-3-pentyl (- CH(CH 2 CH 3 )CH(CH 3 ) 2 ), 2,3-dimethyl-2-butyl (-C(CH 3 ) 2 CH(CH 3 ) 2 ), 3,3-dimethyl-2- butyl (-CH(CH 3 )C(CH 3 )3, 1-heptyl and 1-octyl. In some embodiments, substituents for "optionally substituted alkyls" include one to four instances of F, CI, Br, I, OH, SH, CN, NH 2 , NHCH3, N(CH 3 ) 2 , N0 2 , N 3 , C(0)CH 3 , COOH, C0 2 CH 3 , methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methanesulfonylamino, SO, S0 2 , phenyl, piperidinyl, piperizinyl, and pyrimidinyl, wherein the alkyl, phenyl and heterocyclic portions thereof may be optionally substituted, such as by one to four instances of substituents selected from this same list.

The term "alkenyl" refers to linear or branched-chain monovalent hydrocarbon radical with at least one site of unsaturation, i.e., a carbon-carbon double bond, wherein the alkenyl radical may be optionally substituted, and includes radicals having "cis" and "trans" orientations, or alternatively, "E" and "Z" orientations. In one example, the alkenyl radical is two to eighteen carbon atoms (C 2 -Ci 8 ). In other examples, the alkenyl radical is C 2 -C 12 , C 2 -C 10 , C 2 -C 8 , C 2 -C 6 or C 2 -C3. Examples include, but are not limited to, ethenyl or vinyl (-CH=CH 2 ), prop-l-enyl (-

CH=CHCH 3 ), prop-2-enyl (-CH 2 CH=CH 2 ), 2-methylprop-l-enyl, but-l-enyl, but-2- enyl, but-3-enyl, buta-l,3-dienyl, 2-methylbuta-l,3-diene, hex-l-enyl, hex-2-enyl, hex-3-enyl, hex-4-enyl and hexa-l,3-dienyl. In some embodiments, substituents for "optionally substituted alkenyls" include one to four instances of F, CI, Br, I, OH, SH, CN, NH 2 , NHCH 3 , N(CH 3 ) 2 , NO 2 , N 3 , C(0)CH 3 , COOH, C0 2 CH 3 , methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methanesulfonylamino, SO, S0 2 , phenyl, piperidinyl, piperizinyl, and pyrimidinyl, wherein the alkyl, phenyl and heterocyclic portions thereof may be optionally substituted, such as by one to four instances of substituents selected from this same list.

The term "alkynyl" refers to a linear or branched monovalent hydrocarbon radical with at least one site of unsaturation, i.e., a carbon-carbon, triple bond, wherein the alkynyl radical may be optionally substituted. In one example, the alkynyl radical is two to eighteen carbon atoms (C 2 -Ci 8 ). In other examples, the alkynyl radical is C 2 -C 12 , C 2 -C 10) C 2 -Cg, C 2 -C6 or C 2 -C 3 . Examples include, but are not limited to, ethynyl (-C≡CH), prop-l-ynyl (-C≡CCH 3 ), prop-2-ynyl (propargyl, -CH 2 C≡CH), but-l-ynyl, but-2-ynyl and but-3-ynyl. In some embodiments, substituents for "optionally substituted alkynyls" include one to four instances of F, CI, Br, I, OH, SH, CN, NH 2 , NHCH 3 , N(CH 3 ) 2 , N0 2 , N 3 , C(0)CH 3 , COOH, C0 2 CH 3 , methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino,

methanesulfonylamino, SO, S0 2 , phenyl, piperidinyl, piperizinyl, and pyrimidinyl, wherein the alkyl, phenyl and heterocyclic portions thereof may be optionally substituted, such as by one to four instances of substituents selected from this same list.

"Alkylene" refers to a saturated, branched or straight chain hydrocarbon group having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of a parent alkane. In one example, the divalent alkylene group is one to eighteen carbon atoms (Ci-Cig). In other examples, the divalent alkylene group is Co-C 6 , C 0 -C5, C 0 -C 3 , C 1 -C 12 , C 1 -C 10 , Ci-Cg, Ci-C 6 , C \ - C 5 , C1-C4, or C1-C3. The group Co alkylene refers to a bond. Example alkylene groups include methylene (-CH 2 -), 1 ,1 -ethyl (-CH(CH 3 )-), (1 ,2-ethyl (-CH 2 CH 2 -), 1 ,1- propyl (-CH(CH 2 CH 3 )-), 2,2-propyl (-C(CH 3 ) 2 -), 1 ,2-propyl (-CH(CH 3 )CH 2 -), 1 ,3- propyl (-CH 2 CH 2 CH 2 -), l , l-dimethyleth-l ,2-yl (-C(CH 3 ) 2 CH 2 -), 1 ,4-butyl

(-CH 2 CH 2 CH 2 CH 2 -), and the like. The term "heteroalkyl" refers to a straight or branched chain monovalent hydrocarbon radical, consisting of the stated number of carbon atoms, or, if none are stated, up to 18 carbon atoms, and from one to five heteroatoms selected from the group consisting of O, N, Si and S, and wherein the nitrogen and sulfur atoms can optionally be oxidized and the nitrogen heteroatom can optionally be quatemized. In some embodiments, the heteroatom is selected from O, N and S, wherein the nitrogen and sulfur atoms can optionally be oxidized and the nitrogen heteroatom can optionally be quatemized. The heteroatom(s) can be placed at any interior position of the heteroalkyl group, including the position at which the alkyl group is attached to the remainder of the molecule (e.g., -O-CH 2 -CH 3 ). Examples include -CH 2 -CH 2 -0- CH 3 , -CH 2 -CH 2 -NH-CH 3 , -CH 2 -CH 2 -N(CH 3 )-CH 3 , -CH 2 -S-CH 2 -CH 3 , -S(0)-CH 3 , - CH 2 -CH 2 -S(0) 2 -CH 3 , -Si(CH 3 ) 3 and -CH 2 -CH=N-OCH 3 . Up to two heteroatoms can be consecutive, such as, for example, -CH 2 -NH-OCH 3 and -CH 2 -0-Si(CH 3 ) 3 .

Heteroalkyl groups can be optionally substituted. In some embodiments, substituents for "optionally substituted heteroalkyls" include one to four instances of F, CI, Br, I, OH, SH, CN, NH 2 , NHCH , N(CH 3 ) 2 , N0 2 , N 3 , C(0)CH 3 , COOH, C0 2 CH 3 , methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methanesulfonylamino, SO, S0 2 , phenyl, piperidinyl, piperizinyl, and pyrimidinyl, wherein the alkyl, phenyl and heterocyclic portions thereof may be optionally substituted, such as by one to four instances of substituents selected from this same list.

"Amino" means primary (i.e., -NH 2 ), secondary (i.e., -NRH), tertiary (i.e., - NPv ) and quaternary (i.e., -N(+)RRR) amines, that are optionally substituted, in which each R is the same or different and selected from alkyl, cycloalkyl, aryl, and heterocyclyl, wherein the alkyl, cycloalkyl, aryl and heterocyclyl groups are as defined herein. Particular secondary and tertiary amines are alkylamine,

dialkylamine, arylamine, diarylamine, aralkylamine and diaralkylamine, wherein the alkyl and aryl portions can be optionally substituted. Particular secondary and tertiary amines are methylamine, ethylamine, propylamine, isopropylamine, phenylamine, benzylamine, dimethylamine, diethylamine, dipropylamine and diisopropylamine. In some embodiments, R groups of a quarternary amine are each independently optionally substituted alkyl groups.

"Aryl" refers to a carbocyclic aromatic group, whether or not fused to one or more groups, having the number of carbon atoms designated, or if no number is designated, up to 14 carbon atoms. One example includes aryl groups having 6-14 carbon atoms. Another example includes aryl groups having 6-10 carbon atoms. Examples of aryl groups include phenyl, naphthyl, biphenyl, phenanthrenyl, naphthacenyl, 1,2,3,4-tetrahydronaphthalenyl, lH-indenyl, 2,3-dihydro-lH-indenyl, and the like (see, e.g., Lang's Handbook of Chemistry (Dean, J. A., ed.) 13 th ed. Table 7-2 [1985]). A particular aryl is phenyl. Substituted phenyl or substituted aryl means a phenyl group or aryl group substituted with one, two, three, four or five substituents, for example, 1-2, 1-3 or 1-4 substituents, such as chosen from groups specified herein (see "optionally substituted" definition), such as F, CI, Br, I, OH, SH, CN, NH 2 , NHCH 3 , N(CH 3 ) 2 , N0 2 , N 3 , C(0)CH 3 , COOH, C0 2 CH 3 , methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methanesulfonylamino, SO, S0 2 , phenyl, piperidinyl, piperizinyl, and pyrimidinyl, wherein the alkyl, phenyl and heterocyclic portions thereof may be optionally substituted, such as by one to four instances of substituents selected from this same list. Examples of the term

"substituted phenyl" include a mono- or di(halo)phenyl group such as 2-chlorophenyl,

2- bromophenyl, 4-chlorophenyl, 2,6-dichlorophenyl, 2,5-dichlorophenyl, 3,4- dichlorophenyl, 3-chlorophenyl, 3-bromophenyl, 4-bromophenyl, 3,4-dibromophenyl,

3- chloro-4-fluorophenyl, 2-fluorophenyl, 2,4-difluorophenyl and the like; a mono- or di(hydroxy)phenyl group such as 4-hydroxyphenyl, 3-hydroxyphenyl, 2,4- dihydroxyphenyl, the protected-hydroxy derivatives thereof and the like; a

nitrophenyl group such as 3- or 4-nitrophenyl; a cyanophenyl group, for example, 4- cyanophenyl; a mono- or di(alkyl)phenyl group such as 4-methylphenyl, 2,4- dimethylphenyl, 2-methylphenyl, 4-(isopropyl)phenyl, 4-ethylphenyl, 3-(n- propyl)phenyl and the like; a mono or di(alkoxy)phenyl group, for example, 3,4- dimethoxyphenyl, 3-methoxy-4-benzyloxyphenyl, 3-ethoxyphenyl, 4- (isopropoxy)phenyl, 4-(t-butoxy)phenyl, 3-ethoxy-4-methoxyphenyl and the like; 3- or 4- trifluoromethylphenyl; a mono- or dicarboxyphenyl or (protected

carboxy)phenyl group such 4-carboxyphenyl, a mono- or di(hydroxymethyl)phenyl or (protected hydroxymethyl)phenyl such as 3 -(protected hydroxymethyl)phenyl or 3,4- di(hydroxymethyl)phenyl; a mono- or di(aminomethyl)phenyl or (protected aminomethyl)phenyl such as 2-(aminomethyl)phenyl or 2,4-(protected

aminomethyl)phenyl; or a mono- or di(N-(methylsulfonylamino))phenyl such as 3- (N-methylsulfonylamino))phenyl. Also, the term "substituted phenyl" represents disubstituted phenyl groups where the substituents are different, for example, 3- methyl-4-hydroxyphenyl, 3-chloro-4-hydroxyphenyl, 2-methoxy-4-bromophenyl, 4- ethyl-2-hydroxyphenyl, 3-hydroxy-4-nitrophenyl, 2-hydroxy-4-chlorophenyl, 2- chloro-5-difluoromethoxy and the like, as well as trisubstituted phenyl groups where the substituents are different, for example 3-methoxy-4-benzyloxy-6-methyl sulfonylamino, 3-methoxy-4-benzyloxy-6-phenyl sulfonylamino, and tetrasubstituted phenyl groups where the substituents are different such as 3-methoxy-4-benzyloxy-5- methyl-6-phenyl sulfonylamino. In some embodiments, a substituent of an aryl, such as phenyl, comprises an amide. For example, an aryl (e.g., phenyl) substituent may be -(CH 2 )o- 4 CONR'R", wherein R' and R" each independently refer to groups including, for example, hydrogen; unsubstituted Ci_C 6 alkyl; Ci.Cealkyl substituted by halogen, OH, CN, unsubstituted Ci-C 6 alkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R";

unsubstituted Ci_C 6 heteroalkyl; Ci_C 6 heteroalkyl substituted by halogen, OH, CN, unsubstituted Ci-Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; unsubstituted C 6- Cio aryl; C 6 -Cio aryl substituted by halogen, OH, CN, unsubstituted Ci-C 6 alkyl, unsubstituted Ci-C 6 alkoxy, or NR'R"; unsubstituted 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S); and 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S) substituted by halogen, OH, CN, unsubstituted Ci-Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; or R' and R" can be combined with the nitrogen atom to form a 3-, 4-, 5-, 6-, or 7-membered ring wherein a ring atom is optionally substituted with N, O or S and wherein the ring is optionally substituted with halogen, OH, CN, unsubstituted Ci-Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R".

"Cycloalkyl" refers to a non-aromatic, saturated or partially unsaturated hydrocarbon ring group wherein the cycloalkyl group may be optionally substituted independently with one or more substituents described herein. In one example, the cycloalkyl group is 3 to 12 carbon atoms (C 3 -C 12 ). In other examples, cycloalkyl is C 3 -C 8 , C 3 -C 10 or C5-C 10 . In other examples, the cycloalkyl group, as a monocycle, is C 3 -C 8 , C3-C 6 or C5-C6. In another example, the cycloalkyl group, as a bicycle, is C 7 - C 12 . In another example, the cycloalkyl group, as a spiro system, is C5-C 12 . Examples of monocyclic cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent- 1-enyl, l-cyclopent-2-enyl, l-cyclopent-3-enyl, cyclohexyl, perdeuteriocyclohexyl, 1- cyclohex-l-enyl, l-cyclohex-2-enyl, l-cyclohex-3-enyl, cyclohexadienyl,

cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl and cyclododecyl. Exemplary arrangements of bicyclic cycloalkyls having 7 to 12 ring atoms include, but are not limited to, [4,4], [4,5], [5,5], [5,6] or [6,6] ring systems. Exemplary bridged bicyclic cycloalkyls include, but are not limited to, bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane and bicyclo[3.2.2]nonane. Examples of spiro cycloalkyl include, spiro[2.2]pentane, spiro[2.3]hexane, spiro[2.4]heptane, spiro[2.5]octane and spiro[4.5]decane. In some embodiments, substituents for "optionally substituted cycloalkyls" include one to four instances of F, CI, Br, I, OH, SH, CN, NH 2 , NHCH 3 , N(CH 3 ) 2 , N0 2 , N 3 , C(0)CH 3 , COOH, C0 2 CH 3 , methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methanesulfonylamino, SO, S0 2 , phenyl, piperidinyl, piperizinyl, and pyrimidinyl, wherein the alkyl, aryl and heterocyclic portions thereof may be optionally substituted, such as by one to four instances of substituents selected from this same list. In some embodiments, a substituent of a cycloalkyl comprises an amide. For example, a cycloalkyl substituent may be -(CH 2 )o_ 4 CONR'R", wherein R' and R" each independently refer to groups including, for example, hydrogen;

unsubstituted Ci_C 6 alkyl; Ci_C 6 alkyl substituted by halogen, OH, CN, unsubstituted Ci-C 6 alkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; unsubstituted Ci_C 6 heteroalkyl; Ci_C 6 heteroalkyl substituted by halogen, OH, CN, unsubstituted Ci- Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; unsubstituted C 6 -Cio aryl; C 6 -Cio aryl substituted by halogen, OH, CN, unsubstituted Ci-C 6 alkyl, unsubstituted Ci-C 6 alkoxy, or NR'R"; unsubstituted 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S); and 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S) substituted by halogen, OH, CN, unsubstituted Ci-C6alkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; or R and R" can be combined with the nitrogen atom to form a 3-, 4-, 5-, 6-, or 7-membered ring wherein a ring atom is optionally substituted with N, O or S and wherein the ring is optionally substituted with halogen, OH, CN, unsubstituted Ci-Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R". "Heterocyclic group", "heterocyclic", "heterocycle", "heterocyclyl", or "heterocyclo" are used interchangeably and refer to any mono-, bi-, tricyclic or spiro, saturated or unsaturated, aromatic (heteroaryl) or non-aromatic (e.g.,

heterocycloalkyl), ring system, having 3 to 20 ring atoms (e.g., 3-10 ring atoms), where the ring atoms are carbon, and at least one atom in the ring or ring system is a heteroatom selected from nitrogen, sulfur or oxygen. If any ring atom of a cyclic system is a heteroatom, that system is a heterocycle, regardless of the point of attachment of the cyclic system to the rest of the molecule. In one example, heterocyclyl includes 3-1 1 ring atoms ("members") and includes monocycles, bicycles, tricycles and spiro ring systems, wherein the ring atoms are carbon, where at least one atom in the ring or ring system is a heteroatom selected from nitrogen, sulfur or oxygen. In one example, heterocyclyl includes 1 to 4 heteroatoms. In one example, heterocyclyl includes 1 to 3 heteroatoms. In another example, heterocyclyl includes 3- to 7-membered monocycles having 1-2, 1-3 or 1-4 heteroatoms selected from nitrogen, sulfur or oxygen. In another example, heterocyclyl includes 4- to 6- membered monocycles having 1-2, 1-3 or 1-4 heteroatoms selected from nitrogen, sulfur or oxygen. In another example, heterocyclyl includes 3-membered

monocycles. In another example, heterocyclyl includes 4-membered monocycles. In another example, heterocyclyl includes 5-6 membered monocycles, e.g., 5-6 membered heteroaryl. In another example, heterocyclyl includes 3-11 membered heterocycloyalkyls, such as 4-11 membered heterocycloalkyls. In some

embodiments, a heterocycloalkyl includes at least one nitrogen. In one example, the heterocyclyl group includes 0 to 3 double bonds. Any nitrogen or sulfur heteroatom may optionally be oxidized (e.g., NO, SO, S0 2 ), and any nitrogen heteroatom may optionally be quaternized (e.g., [NR 4 ] + Cr, [NR 4 ] OH " ). Example heterocycles are oxiranyl, aziridinyl, thiiranyl, azetidinyl, oxetanyl, thietanyl, 1 ,2-dithietanyl, 1,3- dithietanyl, pyrrolidinyl, dihydro-lH-pyrrolyl, dihydrofuranyl, tetrahydrofuranyl, dihydrothienyl, tetrahydrothienyl, imidazolidinyl, piperidinyl, piperazinyl,

isoquinolinyl, tetrahydroisoquinolinyl, morpholinyl, thiomorpholinyl, 1,1-dioxo- thiomorpholinyl, dihydropyranyl, tetrahydropyranyl, hexahydrothiopyranyl, hexahydropyrimidinyl, oxazinanyl, thiazinanyl, thioxanyl, homopiperazinyl, homopiperidinyl, azepanyl, oxepanyl, thiepanyl, oxazepinyl, oxazepanyl, diazepanyl, 1 ,4-diazepanyl, diazepinyl, thiazepinyl, thiazepanyl, tetrahydrothiopyranyl, oxazolidinyl, thiazolidinyl, isothiazolidinyl, 1,1-dioxoisothiazolidinonyl, oxazolidinonyl, imidazolidinonyl, 4,5,6,7-tetrahydro[2H]indazolyl, tetrahydrobenzoimidazolyl, 4,5,6,7-tetrahydrobenzo[d]imidazolyl, 1 ,6- dihydroimidazol[4,5-d]pyrrolo[2,3-b]pyridinyl, thiazinyl, oxazinyl, thiadiazinyl, oxadiazinyl, dithiazinyl, dioxazinyl, oxathiazinyl, thiatriazinyl, oxatriazinyl, dithiadiazinyl, imidazolinyl, dihydropyrimidyl, tetrahydropyrimidyl, 1 -pyrrolinyl, 2- pyrrolinyl, 3 -pyrrolinyl, indolinyl, thiapyranyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, pyrazolidinyl, dithianyl, dithiolanyl, pyrimidinonyl, pyrimidindionyl, pyrimidin-2,4-dionyl, piperazinonyl, piperazindionyl,

pyrazolidinylimidazolinyl, 3-azabicyclo[3.1.0]hexanyl, 3,6- diazabicyclo[3.1.1]heptanyl, 6-azabicyclo[3.1.1]heptanyl, 3- azabicyclo[3.1.1]heptanyl, 3-azabicyclo[4.1.0]heptanyl, azabicyclo[2.2.2]hexanyl, 2- azabicyclo[3.2.1]octanyl, 8-azabicyclo[3.2.1]octanyl, 2-azabicyclo[2.2.2]octanyl, 8- azabicyclo[2.2.2]octanyl, 7-oxabicyclo[2.2. ljheptane, azaspiro[3.5]nonanyl, azaspiro[2.5]octanyl, azaspiro[4.5]decanyl, l-azaspiro[4.5]decan-2-only,

azaspiro[5.5]undecanyl, tetrahydroindolyl, octahydroindolyl, tetrahydroisoindolyl, tetrahydroindazolyl, 1,1-dioxohexahydrothiopyranyl. Examples of 5-membered heterocycles containing a sulfur or oxygen atom and one to three nitrogen atoms are thiazolyl, including thiazol-2-yl and thiazol-2-yl N-oxide, thiadiazolyl, including l,3,4-thiadiazol-5-yl and l,2,4-thiadiazol-5-yl, oxazolyl, for example oxazol-2-yl, and oxadiazolyl, such as l,3,4-oxadiazol-5-yl, and l,2,4-oxadiazol-5-yl. Example 5- membered ring heterocycles containing 2 to 4 nitrogen atoms include imidazolyl, such as imidazol-2-yl; triazolyl, such as l,3,4-triazol-5-yl; l,2,3-triazol-5-yl, 1,2,4-triazol-

5- yl, and tetrazolyl, such as lH-tetrazol-5-yl. Example benzo-fused 5-membered heterocycles are benzoxazol-2-yl, benzthiazol-2-yl and benzimidazol-2-yl. Example

6- membered heterocycles contain one to three nitrogen atoms and optionally a sulfur or oxygen atom, for example pyridyl, such as pyrid-2-yl, pyrid-3-yl, and pyrid-4-yl; pyrimidyl, such as pyrimid-2-yl and pyrimid-4-yl; triazinyl, such as l,3,4-triazin-2-yl and l,3,5-triazin-4-yl; pyridazinyl, in particular pyridazin-3-yl, and pyrazinyl. The pyridine N-oxides and pyridazine N-oxides and the pyridyl, pyrimid-2-yl, pyrimid-4- yl, pyridazinyl and the l,3,4-triazin-2-yl groups, are other example heterocycle groups. Heterocycles may be optionally substituted. For example, substituents for "optionally substituted heterocycles" include one to four instances of F, CI, Br, I, OH, SH, CN, NH 2 , NHCH 3 , N(CH 3 ) 2 , N0 2 , N 3 , C(0)CH 3 , COOH, C0 2 CH 3 , methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, oxo, trifluoromethyl, difluoromethyl, sulfonylamino, methanesulfonylamino, SO, S0 2 , phenyl, piperidinyl, piperizinyl, and pyrimidinyl, wherein the alkyl, aryl and heterocyclic portions thereof may be optionally substituted, such as by one to four instances of substituents selected from this same list. In some embodiments, a substituent of a heterocyclic group, such as a heteroaryl or heterocycloalkyl, comprises an amide. For example, a heterocyclic (e.g., heteroaryl or

heterocycloalkyl) substituent may be -(CH 2 )o_ 4 CONR'R", wherein R' and R" each independently refer to groups including, for example, hydrogen; unsubstituted Ci_ C 6 alkyl; Ci.Cealkyl substituted by halogen, OH, CN, unsubstituted Ci-Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; unsubstituted Ci_C 6 heteroalkyl; Ci_C 6 heteroalkyl substituted by halogen, OH, CN, unsubstituted Ci-Cealkyl, unsubstituted Ci-C alkoxy, oxo or NR'R"; unsubstituted C 6 -Cio aryl; C 6 -Cio aryl substituted by halogen, OH, CN, unsubstituted Ci-Cealkyl, unsubstituted Ci-C 6 alkoxy, or NR'R"; unsubstituted 3-1 1 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-1 1 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S); and 3-1 1 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-1 1 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S) substituted by halogen, OH, CN, unsubstituted Ci-C 6 alkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; or R' and R" can be combined with the nitrogen atom to form a 3-, 4-, 5-, 6-, or 7-membered ring wherein a ring atom is optionally substituted with N, O or S and wherein the ring is optionally substituted with halogen, OH, CN, unsubstituted Ci-C 6 alkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R".

"Heteroaryl" refers to any mono-, bi-, or tricyclic ring system where at least one ring is a 5- or 6-membered aromatic ring containing from 1 to 4 heteroatoms selected from nitrogen, oxygen, and sulfur, and in an example embodiment, at least one heteroatom is nitrogen. See, for example, Lang's Handbook of Chemistry (Dean, J. A., ed.) 13 th ed. Table 7-2 [1985]. Included in the definition are any bicyclic groups where any of the above heteroaryl rings are fused to an aryl ring, wherein the aryl ring or the heteroaryl ring is joined to the remainder of the molecule. In one embodiment, heteroaryl includes 5-6 membered monocyclic aromatic groups where one or more ring atoms is nitrogen, sulfur or oxygen. Example heteroaryl groups include thienyl, furyl, imidazolyl, pyrazolyl, thiazolyl, isothiazolyl, oxazolyl, isoxazolyl, triazolyl, thiadiazolyl, oxadiazolyl, tetrazolyl, thiatriazolyl, oxatriazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazinyl, tetrazinyl, tetrazolo[l,5-b]pyridazinyl, imidazol[l,2- ajpyrimidinyl and purinyl, as well as benzo-fused derivatives, for example benzoxazolyl, benzofuryl, benzothiazolyl, benzothiadiazolyl, benzotriazolyl, benzoimidazolyl and indolyl. Heteroaryl groups can be optionally substituted. In some embodiments, substituents for "optionally substituted heteroaryls" include one to four instances of F, CI, Br, I, OH, SH, CN, NH 2 , NHCH 3 , N(CH 3 ) 2 , N0 2 , N 3 , C(0)CH 3 , COOH, C0 2 CH 3 , methyl, ethyl, propyl, iso-propyl, butyl, isobutyl, cyclopropyl, methoxy, ethoxy, propoxy, trifluoromethyl, difluoromethyl,

sulfonylamino, methanesulfonylamino, SO, S0 2 , phenyl, piperidinyl, piperizinyl, and pyrimidinyl, wherein the alkyl, phenyl and heterocyclic portions thereof may be optionally substituted, such as by one to four instances of substituents selected from this same list. In some embodiments, a substituent of a heteroaryl comprises an amide. For example, a heteroaryl substituent may be -(CH 2 )o_ 4 CONR'R", wherein R' and R" each independently refer to groups including, for example, hydrogen;

unsubstituted Ci_C 6 alkyl; Ci_C 6 alkyl substituted by halogen, OH, CN, unsubstituted Ci-C6alkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; unsubstituted Ci_C 6 heteroalkyl; Ci_C 6 heteroalkyl substituted by halogen, OH, CN, unsubstituted Ci- Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; unsubstituted C 6 -Cio aryl; C 6 -Cio aryl substituted by halogen, OH, CN, unsubstituted Ci-C 6 alkyl, unsubstituted Ci-C 6 alkoxy, or NR'R"; unsubstituted 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S); and 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S) substituted by halogen, OH, CN, unsubstituted Ci-C 6 alkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; or R and R" can be combined with the nitrogen atom to form a 3-, 4-, 5-, 6-, or 7-membered ring wherein a ring atom is optionally substituted with N, O or S and wherein the ring is optionally substituted with halogen, OH, CN, unsubstituted Ci-Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R".

In particular embodiments, a heterocyclyl group is attached at a carbon atom of the heterocyclyl group. By way of example, carbon bonded heterocyclyl groups include bonding arrangements at position 2, 3, 4, 5, or 6 of a pyridine ring, position 3, 4, 5, or 6 of a pyridazine ring, position 2, 4, 5, or 6 of a pyrimidine ring, position 2, 3,

5, or 6 of a pyrazine ring, position 2, 3, 4, or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrole or tetrahydropyrrole ring, position 2, 4, or 5 of an oxazole, imidazole or thiazole ring, position 3, 4, or 5 of an isoxazole, pyrazole, or isothiazole ring, position 2 or 3 of an aziridine ring, position 2, 3, or 4 of an azetidine ring, position 2, 3, 4, 5, 6, 7, or 8 of a quinoline ring or position 1, 3, 4, 5, 6, 7, or 8 of an isoquinoline ring.

In certain embodiments, the heterocyclyl group is N-attached. By way of example, nitrogen bonded heterocyclyl or heteroaryl groups include bonding arrangements at position 1 of an aziridine, azetidine, pyrrole, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3 -imidazoline, pyrazole, pyrazoline, 2-pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, 1H- indazole, position 2 of a isoindole, or isoindoline, position 4 of a morpholine, and position 9 of a carbazole, or β-carboline.

The term "alkoxy" refers to a linear or branched monovalent radical represented by the formula -OR in which R is alkyl, as defined herein. Alkoxy groups include methoxy, ethoxy, propoxy, isopropoxy, mono-, di- and tri-fluoromethoxy and cyclopropoxy.

"Acyl" means a carbonyl containing substituent represented by the formula - C(0)-R in which R is hydrogen, alkyl, cycloalkyl, aryl or heterocyclyl, wherein the alkyl, cycloalkyl, aryl and heterocyclyl are as defined herein. Acyl groups include alkanoyl (e.g., acetyl), aroyl (e.g., benzoyl), and heteroaroyl (e.g., pyridinoyl).

"Optionally substituted" unless otherwise specified means that a group may be unsubstituted or substituted by one or more (e.g., 0, 1, 2, 3, 4, or 5 or more, or any range derivable therein) of the substituents listed for that group in which said substituents may be the same or different. In an embodiment, an optionally substituted group has 1 substituent. In another embodiment an optionally substituted group has 2 substituents. In another embodiment an optionally substituted group has 3 substituents. In another embodiment an optionally substituted group has 4 substituents. In another embodiment an optionally substituted group has 5

substituents.

Optional substituents for alkyl radicals, alone or as part of another substituent (e.g., alkoxy), as well as alkylenyl, alkenyl, alkynyl, heteroalkyl, heterocycloalkyl, and cycloalkyl, also each alone or as part of another substituent, can be a variety of groups, such as those described herein, as well as selected from the group consisting of halogen; oxo; CN; NO; N 3 ; -OR'; perfluoro-Ci_C4 alkoxy; unsubstituted C 3 -C 7 cycloalkyl; C 3 -C 7 cycloalkyl substituted by halogen, OH, CN, unsubstituted Ci- Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; unsubstituted C 6 -Cio aryl (e.g., phenyl); C 6 -Cio aryl substituted by halogen, OH, CN, unsubstituted Ci-Cealkyl, unsubstituted Ci-C 6 alkoxy, or NR'R"; unsubstituted 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S); 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S) substituted by halogen, OH, CN, unsubstituted Ci-C 6 alkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; -NR'R"; -SR; -SiR'R"R"'; -OC(0)R; -C(0)R * ; -C0 2 R * ; -CONR'R"; -OC(0)NR'R";

-NR"C(0)R'; -NR"'C(0)NR'R"; -NR"C(0) 2 R'; -S(0) 2 R; -S(0) 2 NR'R"; -NR'S(0) 2 R"; -NR"'S(0) 2 NR'R"; amidinyl; guanidinyl; -(CH 2 )i_ 4 -OR; -(CH 2 )i_ 4 -NR * R"; -(CH 2 )i_ 4 -SR; -(CH 2 ) 1 _ 4 -SiR'R"R"'; -(CH 2 ) 1 _ 4 -OC(0)R'; -(CH 2 ) 1 _ 4 -C(0)R; -(CH 2 ) 1 _ 4 -C0 2 R; and -(CH 2 )i_ 4 CONR'R", or combinations thereof, in a number ranging from zero to (2m'+l), where m' is the total number of carbon atoms in such radical. R', R" and R'" each independently refer to groups including, for example, hydrogen; unsubstituted Ci_C 6 alkyl; Ci_C 6 alkyl substituted by halogen, OH, CN, unsubstituted Ci-C 6 alkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; unsubstituted Ci_C 6 heteroalkyl; Ci_C 6 heteroalkyl substituted by halogen, OH, CN, unsubstituted Ci-Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; unsubstituted C 6 -Cio aryl; C 6 -Cio aryl substituted by halogen, OH, CN, unsubstituted Ci-Cealkyl, unsubstituted Ci-C 6 alkoxy, or NR'R"; unsubstituted 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S); and 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S) substituted by halogen, OH, CN, unsubstituted Ci-Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R". When R' and R" are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 3-, 4-, 5-, 6-, or 7-membered ring wherein a ring atom is optionally substituted with N, O or S and wherein the ring is optionally substituted with halogen, OH, CN, unsubstituted Ci- Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R". For example, -NR'R" is meant to include 1-pyrrolidinyl and 4-morpholinyl.

Similarly, optional substituents for the aryl and heteroaryl groups are varied. In some embodiments, substituents for aryl and heteroaryl groups are selected from the group consisting of halogen; CN; NO; N 3 ; -OR; perfluoro-Ci_C4 alkoxy;

unsubstituted C 3 -C 7 cycloalkyl; C 3 -C 7 cycloalkyl substituted by halogen, OH, CN, unsubstituted Ci-C 6 alkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; unsubstituted C 6 -Cio aryl (e.g., phenyl); C 6 -Cio aryl substituted by halogen, OH, CN, unsubstituted Ci-C6alkyl, unsubstituted Ci-C 6 alkoxy, or NR'R"; unsubstituted 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S); 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S) substituted by halogen, OH, CN, unsubstituted Ci-C 6 alkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; -NR'R"; -SR; -SiR'R'R'"; -OC(0)R * ; -C(0)R; -C0 2 R * ; -CONR'R";

-OC(0)NR'R"; -NR"C(0)R; -NR"'C(0)NR'R"; -NR"C(0) 2 R'; -S(0) 2 R; -S(0) 2 NR'R"; -NR'S(0) 2 R"; -NR"'S(0) 2 NR'R"; amidinyl; guanidinyl; -(CH 2 )i_ 4 -OR; -(CH 2 )i_ 4 -NR'R"; -(CH 2 ) 1 _ 4 -SR'; -(CH 2 ) 1 _ 4 -SiR'R"R"'; -(CH 2 ) 1 _ 4 -OC(0)R; -(CH 2 ) 1 _ 4 -C(0)R; -(CH 2 )i_ 4 -C0 2 R; and -(CH 2 )i_ 4 CONR'R", or combinations thereof, in a number ranging from zero to (2m'+l), where m' is the total number of carbon atoms in such radical. R', R" and R'" each independently refer to groups including, for example, hydrogen; unsubstituted Ci_C 6 alkyl; Ci.Cealkyl substituted by halogen, OH, CN, unsubstituted Ci-Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; unsubstituted Ci_ C 6 heteroalkyl; Ci_C 6 heteroalkyl substituted by halogen, OH, CN, unsubstituted Ci- Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R"; unsubstituted C 6 -Cio aryl; C 6 -Cio aryl substituted by halogen, OH, CN, unsubstituted Ci-C 6 alkyl, unsubstituted Ci-C 6 alkoxy, or NR'R"; unsubstituted 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S); and 3-11 membered heterocyclyl (e.g., 5-6 membered heteroaryl containing 1 to 4 heteroatoms selected from O, N and S or 4-11 membered heterocycloalkyl containing 1 to 4 heteroatoms selected from O, N and S) substituted by halogen, OH, CN, unsubstituted Ci-Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R". When R' and R" are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 3-, 4-, 5-, 6-, or 7-membered ring wherein a ring atom is optionally substituted with N, O or S and wherein the ring is optionally substituted with halogen, OH, CN,

unsubstituted Ci-Cealkyl, unsubstituted Ci-C 6 alkoxy, oxo or NR'R". For example, -NR'R" is meant to include 1 -pyrrolidinyl and 4-morpholinyl.

The term "oxo" refers to =0 or (=0) 2 .

As used herein a wavy line "·~*~ " that intersects a bond in a chemical structure indicate the point of attachment of the atom to which the wavy bond is connected in the chemical structure to the remainder of a molecule, or to the remainder of a fragment of a molecule. In some embodiments, an arrow together with an asterisk is used in the manner of a wavy line to indicate a point of attachment.

In certain embodiments, divalent groups are described generically without specific bonding configurations. It is understood that the generic description is meant to include both bonding configurations, unless specified otherwise. For example, in

1 2 3 * 2

the group R -R -R , if the group R is described as -CH 2 C(0)-, then it is understood that this group can be bonded both as R 1 -CH 2 C(0)-R 3 , and as R 1 -C(0)CH 2 -R 3 , unless specified otherwise.

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

The phrase "pharmaceutically acceptable" refers to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, such as, for example, a human, as appropriate.

Compounds of the present invention may be in the form of a salt, such as a pharmaceutically acceptable salt. "Pharmaceutically acceptable salts" include both acid and base addition salts. "Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, carbonic acid, phosphoric acid and the like, and organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic, and sulfonic classes of organic acids such as formic acid, acetic acid, propionic acid, glycolic acid, gluconic acid, lactic acid, pyruvic acid, oxalic acid, malic acid, maleic acid, maloneic acid, succinic acid, fumaric acid, tartaric acid, citric acid, aspartic acid, ascorbic acid, glutamic acid, anthranilic acid, benzoic acid, cinnamic acid, mandelic acid, embonic acid, phenylacetic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, salicyclic acid and the like.

"Pharmaceutically acceptable base addition salts" include those derived from inorganic bases such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Particular base addition salts are the ammonium, potassium, sodium, calcium and magnesium salts. Salts derived from pharmaceutically acceptable organic nontoxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-diethylaminoethanol, tromethamine, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine,

ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperizine, piperidine, N-ethylpiperidine, polyamine resins and the like. Particular organic non- toxic bases include isopropylamine, diethylamine, ethanolamine, tromethamine, dicyclohexylamine, choline, and caffeine.

In some embodiments, a salt is selected from a hydrochloride, hydrobromide, trifluoroacetate, sulphate, phosphate, acetate, fumarate, maleate, tartrate, lactate, citrate, pyruvate, succinate, oxalate, methanesulphonate, p-toluenesulphonate, bisulphate, benzenesulphonate, ethanesulphonate, malonate, xinafoate, ascorbate, oleate, nicotinate, saccharinate, adipate, formate, glycolate, palmitate, L-lactate, D- lactate, aspartate, malate, L-tartrate, D-tartrate, stearate, furoate (e.g., 2-furoate or 3- furoate), napadisylate (naphthalene-l,5-disulfonate or naphthalene- 1 -(sulfonic acid)- 5 -sulfonate), edisylate (ethane- 1 ,2-disulfonate or ethane- 1 -(sulfonic acid)-2- sulfonate), isethionate (2-hydroxyethylsulfonate), 2-mesitylenesulphonate, 2- naphthalenesulphonate, 2,5-dichlorobenzenesulphonate, D-mandelate, L-mandelate, cinnamate, benzoate, adipate, esylate, malonate, mesitylate (2-mesitylenesulphonate), napsylate (2-naphthalenesulfonate), camsylate (camphor- 10-sulphonate, for example (lS)-(+)-10-camphorsulfonic acid salt), glutamate, glutarate, hippurate (2- (benzoylamino)acetate), orotate, xylate (p-xylene-2-sulphonate), and pamoic (2,2'- dihydroxy- 1 , 1 '-dinaphthylmethane-3 ,3'-dicarboxylate).

A "sterile" formulation is aseptic or free from all living microorganisms and their spores.

"Stereoisomers" refer to compounds that have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.

Stereoisomers include diastereomers, enantiomers, conformers and the like.

"Chiral" refers to molecules which have the property of non-superimposability of the mirror image partner, while the term "achiral" refers to molecules which are superimposable on their mirror image partner.

"Diastereomer" refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g., melting points, boiling points, spectral properties or biological activities. Mixtures of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography such as HPLC.

"Enantiomers" refer to two stereoisomers of a compound which are non- superimposable mirror images of one another.

Stereochemical definitions and conventions used herein generally follow S. P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S., "Stereochemistry of Organic

Compounds", John Wiley & Sons, Inc., New York, 1994. Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane- polarized light. In describing an optically active compound, the prefixes D and L, or R and S, are used to denote the absolute configuration of the molecule about its chiral center(s). The prefixes d and 1 or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or 1 meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. For a given chemical structure, these stereoisomers are identical except that they are mirror images of one another. A specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process. The terms "racemic mixture" and "racemate" refer to an equimolar mixture of two enantiomeric species, devoid of optical activity. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies which are interconvertible via a low energy barrier. For example, proton tautomers (also known as prototropic tautomers) include interconversions via migration of a proton, such as keto-enol and imine-enamine isomerizations. Valence tautomers include interconversions by reorganization of some of the bonding electrons.

Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. A "solvate" refers to an association or complex of one or more solvent molecules and a compound of the present invention. Examples of solvents that form solvates include water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine. Certain compounds of the present invention can exist in multiple crystalline or amorphous forms. In general, all physical forms are intended to be within the scope of the present invention. The term "hydrate" refers to the complex where the solvent molecule is water.

A "metabolite" refers to a product produced through metabolism in the body of a specified compound or salt thereof. Such products can result, for example, from the oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, and the like, of the administered compound.

Metabolite products typically are identified by preparing a radiolabeled (e.g.,

14 C or 3 H) isotope of a compound of the invention, administering it in a detectable dose (e.g., greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to a human, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples. These products are easily isolated since they are labeled (others are isolated by the use of antibodies capable of binding epitopes surviving in the metabolite). The metabolite structures are determined in

conventional fashion, e.g., by MS, LC/MS or NMR analysis. In general, analysis of metabolites is done in the same way as conventional drug metabolism studies well known to those skilled in the art. The metabolite products, so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention.

A "subject," "individual," or "patient" is a vertebrate. In certain

embodiments, the vertebrate is a mammal. Mammals include, but are not limited to, farm animals (such as cows), sport animals, pets (such as guinea pigs, cats, dogs, rabbits and horses), primates, mice and rats. In certain embodiments, a mammal is a human. In embodiments comprising administration of a JAK inhibitor as described herein or a pharmaceutically acceptable salt thereof to a patient, the patient may be in need thereof.

The term "Janus kinase" refers to JAKl, JAK2, JAK3 and TYK2 protein kinases. In some embodiments, a Janus kinase may be further defined as one of JAKl, JAK2, JAK3 or TYK2. In any embodiment, any one of JAKl, JAK2, JAK3 and TYK2 may be specifically excluded as a Janus kinase. In some embodiments, a Janus kinase is JAKl . In some embodiments, a Janus kinase is a combination of JAKl and JAK2.

The terms "inhibiting" and "reducing," or any variation of these terms, includes any measurable decrease or complete inhibition to achieve a desired result. For example, there may be a decrease of about, at most about, or at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%), 85%o, 90%), 95%), 99%, or more, or any range derivable therein, reduction of activity (e.g., JAKl activity) compared to normal.

In some embodiments, a compound or a salt thereof (e.g., a pharmaceutically acceptable salt thereof) described herein is selective for inhibition of JAKl over JAK3 and TYK2. In some embodiments, a compound or a salt thereof (e.g., a

pharmaceutically acceptable salt thereof) is selective for inhibition of JAKl over JAK2, JAK3, or TYK2, or any combination of JAK2, JAK3, or TYK2. In some embodiments, a compound or a salt thereof (e.g., a pharmaceutically acceptable salt thereof) is selective for inhibition of JAKl and JAK2 over JAK3 and TYK2. In some embodiments, a compound or a salt thereof (e.g., a pharmaceutically acceptable salt thereof) is selective for inhibition of JAKl over JAK3. By "selective for inhibition" it is meant that the compound or a salt thereof (e.g., a pharmaceutically acceptable salt thereof) is at least a 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or more, or any range derivable therein, better inhibitor of a particular Janus kinase (e.g., JAKl) activity compared to another particular Janus kinase (e.g., JAK3) activity, or is at least a 2-, 3-, 4-, 5-, 10-, 25-, 50-, 100-, 250-, or 500-fold better inhibitor of a particular Janus kinase (e.g., JAKl) activity compared to another particular Janus kinase (e.g., JAK3) activity. "Therapeutically effective amount" means an amount of a compound or a salt thereof (e.g., a pharmaceutically acceptable salt thereof) of the present invention that (i) treats or prevents the particular disease, condition or disorder, or (ii) attenuates, ameliorates or eliminates one or more symptoms of the particular disease, condition, or disorder, and optionally (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition or disorder described herein. In some

embodiments, the therapeutically effective amount is an amount sufficient to decrease or alleviate the symptoms of an autoimmune or inflammatory disease (e.g., asthma). In some embodiments, a therapeutically effective amount is an amount of a chemical entity described herein sufficient to significantly decrease the activity or number of B- cells. In the case of cancer, the therapeutically effective amount of the drug may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; or relieve to some extent one or more of the symptoms associated with the cancer. To the extent the drug may prevent growth or kill existing cancer cells, it may be cytostatic or cytotoxic. For cancer therapy, efficacy can, for example, be measured by assessing the time to disease progression (TTP) or determining the response rate (RR).

"Treatment" (and variations such as "treat" or "treating") refers to clinical intervention in an attempt to alter the natural course of the individual or cell being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, stabilized (i.e., not worsening) state of disease, decreasing the rate of disease progression, amelioration or palliation of the disease state, prolonging survival as compared to expected survival if not receiving treatment and remission or improved prognosis. In some embodiments, a compound of the invention or a salt thereof (e.g., a pharmaceutically acceptable salt thereof), is used to delay development of a disease or disorder or to slow the progression of a disease or disorder. Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder, (for example, through a genetic mutation) or those in which the condition or disorder is to be prevented. "Inflammatory disorder" refers to any disease, disorder or syndrome in which an excessive or unregulated inflammatory response leads to excessive inflammatory symptoms, host tissue damage, or loss of tissue function. "Inflammatory disorder" also refers to a pathological state mediated by influx of leukocytes or neutrophil chemotaxis.

"Inflammation" refers to a localized, protective response elicited by injury or destruction of tissues, which serves to destroy, dilute, or wall off (sequester) both the injurious agent and the injured tissue. Inflammation is notably associated with influx of leukocytes or neutrophil chemotaxis. Inflammation can result from infection with pathogenic organisms and viruses and from noninfectious means such as trauma or reperfusion following myocardial infarction or stroke, immune responses to foreign antigens, and autoimmune responses. Accordingly, inflammatory disorders amenable to treatment with a compound or a salt thereof (e.g., a pharmaceutically acceptable salt thereof) of the present invention encompass disorders associated with reactions of the specific defense system as well as with reactions of the nonspecific defense system.

"Specific defense system" refers to the component of the immune system that reacts to the presence of specific antigens. Examples of inflammation resulting from a response of the specific defense system include the classical response to foreign antigens, autoimmune diseases, and delayed type hypersensitivity responses mediated by T-cells. Chronic inflammatory diseases, the rejection of solid transplanted tissue and organs, e.g., kidney and bone marrow transplants, and graft versus host disease (GVHD), are further examples of inflammatory reactions of the specific defense system.

The term "nonspecific defense system" refers to inflammatory disorders that are mediated by leukocytes that are incapable of immunological memory (e.g., granulocytes, and macrophages). Examples of inflammation that result, at least in part, from a reaction of the nonspecific defense system include inflammation associated with conditions such as adult (acute) respiratory distress syndrome (ARDS) or multiple organ injury syndromes; reperfusion injury; acute glomerulonephritis; reactive arthritis; dermatoses with acute inflammatory components; acute purulent meningitis or other central nervous system inflammatory disorders such as stroke; thermal injury; inflammatory bowel disease; granulocyte transfusion associated syndromes; and cytokine-induced toxicity. "Autoimmune disease" refers to any group of disorders in which tissue injury is associated with humoral or cell-mediated responses to the body's own constituents. Non-limiting examples of autoimmune diseases include rheumatoid arthritis, lupus and multiple sclerosis.

"Allergic disease" as used herein refers to any symptoms, tissue damage, or loss of tissue function resulting from allergy. "Arthritic disease" as used herein refers to any disease that is characterized by inflammatory lesions of the joints attributable to a variety of etiologies. "Dermatitis" as used herein refers to any of a large family of diseases of the skin that are characterized by inflammation of the skin attributable to a variety of etiologies. "Transplant rejection" as used herein refers to any immune reaction directed against grafted tissue, such as organs or cells (e.g., bone marrow), characterized by a loss of function of the grafted and surrounding tissues, pain, swelling, leukocytosis, and thrombocytopenia. The therapeutic methods of the present invention include methods for the treatment of disorders associated with inflammatory cell activation.

"Inflammatory cell activation" refers to the induction by a stimulus (including, but not limited to, cytokines, antigens or auto-antibodies) of a proliferative cellular response, the production of soluble mediators (including but not limited to cytokines, oxygen radicals, enzymes, prostanoids, or vasoactive amines), or cell surface expression of new or increased numbers of mediators (including, but not limited to, major histocompatability antigens or cell adhesion molecules) in inflammatory cells (including but not limited to monocytes, macrophages, T lymphocytes, B

lymphocytes, granulocytes (i.e., polymorphonuclear leukocytes such as neutrophils, basophils, and eosinophils), mast cells, dendritic cells, Langerhans cells, and endothelial cells). It will be appreciated by persons skilled in the art that the activation of one or a combination of these phenotypes in these cells can contribute to the initiation, perpetuation, or exacerbation of an inflammatory disorder.

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

The terms "cancer" and "cancerous", "neoplasm", and "tumor" and related terms refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. A "tumor" comprises one or more cancerous cells. Examples of cancer include carcinoma, blastoma, sarcoma, seminoma, glioblastoma, melanoma, leukemia, and myeloid or lymphoid

malignancies. More particular examples of such cancers include squamous cell cancer (e.g., epithelial squamous cell cancer) and lung cancer including small-cell lung cancer, non-small cell lung cancer ("NSCLC"), adenocarcinoma of the lung and squamous carcinoma of the lung. Other cancers include skin, kerato acanthoma, follicular carcinoma, hairy cell leukemia, buccal cavity, pharynx (oral), lip, tongue, mouth, salivary gland, esophageal, larynx, hepatocellular, gastric, stomach, gastrointestinal, small intestine, large intestine, pancreatic, cervical, ovarian, liver, bladder, hepatoma, breast, colon, rectal, colorectal, genitourinary, biliary passage, thyroid, papillary, hepatic, endometrial, uterine, salivary gland, kidney or renal, prostate, testis, vulval, peritoneum, anal, penile, bone, multiple myeloma, B-cell lymphoma, central nervous system, brain, head and neck, Hodgkin's, and associated metastases. Examples of neoplastic disorders include myeloproliferative disorders, such as polycythemia vera, essential thrombocytosis, myelofibrosis, such as primary myelofibrosis, and chronic myelogenous leukemia (CML).

A "chemotherapeutic agent" is an agent useful in the treatment of a given disorder, for example, cancer or inflammatory disorders. Examples of

chemotherapeutic agents are well-known in the art and include examples such as those disclosed in U.S. Publ. Appl. No. 2010/0048557, incorporated herein by reference. Additionally, chemotherapeutic agents include pharmaceutically acceptable salts, acids or derivatives of any of chemotherapeutic agents, as well as combinations of two or more of them.

"Package insert" is used to refer to instructions customarily included in commercial packages of therapeutic products that contain information about the indications, usage, dosage, administration, contraindications or warnings concerning the use of such therapeutic products.

Unless otherwise stated, structures depicted herein include compounds that differ only in the presence of one or more isotopically enriched atoms. Exemplary isotopes that can be incorporated into compounds of the present invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, such as 2 H, 3 H, U C, 13 C, 14 C, 13 N, 15 N, 15 0, 17 0, 18 0, 32 P, 33 P, 35 S, 18 F,

36 123 125

Cl, I, and I, respectively. Isotopically-labeled compounds (e.g., those labeled with 3 H and 14 C) can be useful in compound or substrate tissue distribution assays. Tritiated (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes can be useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements).

2 3

In some embodiments, one or more hydrogen atoms are replaced by H or H, or one or more carbon atoms are replaced by 13 C- or 14 C-enriched carbon. Positron emitting isotopes such as 15 O, 13 N, 11 C, and 18 F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy. Isotopically labeled compounds can generally be prepared by procedures analogous to those disclosed in the Schemes or in the Examples herein, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.

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

The use of the term "or" is used to mean "and/or" unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or."

Throughout this application, the term "about" is used to indicate that a value includes the standard deviation of error for the device or method being employed to determine the value.

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

Headings used herein are intended only for organizational purposes.

INHIBITORS OF JANUS KINASES

One embodiment provides a compound of Formula

or a salt (e.g., a pharmaceutically acceptable salt) thereof, wherein:

R 1 is hydrogen or CH 3 ;

R is halogen, Ci-Cealkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, or - OR a , wherein R 2 is optionally substituted by one or more groups independently selected from the group consisting of halogen, Ci-C 3 alkyl, cyano, hydroxy and oxo;

R a is Ci-Cealkyl, -phenyl-COR b R c , -phenyl-(3-6-membered heterocyclyl), or 3-11-membered heterocyclyl, wherein R a is optionally substituted by one or more groups independently selected from the group consisting of halogen, Ci-C 3 alkyl, cyano, hydroxy and oxo;

R b and R c are each independently hydrogen or CH 3 ;

R is hydrogen or NH 2 ;

R 4 is hydrogen or CH 3 ; and

R 5 is hydrogen or NH 2 .

In some embodiments, R 1 is hydrogen. In some embodiments, R 1 is CH 3 . In some embodiments, R 3 is hydrogen. In some embodiments, R 4 and R 5 are each hydrogen. In some embodiments, R 1 , R 3 , R 4 and R 5 are each hydrogen.

In some embodiments, R is selected from the group consisting of halogen, Ci- C 6 haloalkyl, and Ci-Cehaloalkyoxy. In some embodiments, R is selected from the group consisting of

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

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) or stereoisomer thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) or stereoisomer thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) or stereoisomer thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) or stereoisomer thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) thereof.

In some embodiments, the following compound is provided:

or a salt (e.g., a pharmaceutically acceptable salt) thereof.

In some embodiments, a compound selected from (a) - (v) below is provided, or a salt (e.g., a pharmaceutically acceptable salt) or stereoisomer thereof:



Also provided is a pharmaceutical composition comprising a JAK inhibitor as described herein, or a pharmaceutically acceptable salt thereof, and a

pharmaceutically acceptable carrier, dilient or excipient.

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

In one embodiment the disease or condition for therapy is cancer,

polycythemia vera, essential thrombocytosis, myelofibrosis, chronic myelogenous leukemia (CML), rheumatoid arthritis, inflammatory bowel syndrome, Crohn's disease, psoriasis, contact dermatitis or delayed hypersensitivity reactions.

In one embodiment the use of a JAK inhibitor as described herein or a pharmaceutically acceptable salt thereof, for the treatment of cancer, polycythemia vera, essential thrombocytosis, myelofibrosis, chronic myelogenous leukemia (CML), rheumatoid arthritis, inflammatory bowel syndrome, Crohn's disease, psoriasis, contact dermatitis or delayed hypersensitivity reactions is provided.

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

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

In one embodiment a JAK inhibitor as described herein or a pharmaceutically acceptable salt thereof is at least five-times more potent as an inhibitor of JAKl than as an inhibitor of LRRK2.

In one embodiment a JAK inhibitor as described herein or a pharmaceutically acceptable salt thereof is at least ten-times more potent as an inhibitor of JAKl than as an inhibitor of LRRK2.

In one embodiment a JAK inhibitor as described herein or a pharmaceutically acceptable salt thereof is at least five-times more potent as an inhibitor of JAKl than as an inhibitor of JAK2.

In one embodiment a JAK inhibitor as described herein or a pharmaceutically acceptable salt thereof is at least ten-times more potent as an inhibitor of JAKl than as an inhibitor of JAK2.

In one embodiment a JAK inhibitor as described herein or a pharmaceutically acceptable salt thereof is at least five-times more potent as an inhibitor of JAKl than as an inhibitor of JAK3.

In one embodiment a JAK inhibitor as described herein or a pharmaceutically acceptable salt thereof is at least ten-times more potent as an inhibitor of JAKl than as an inhibitor of JAK3.

In one embodiment a JAK inhibitor as described herein or a pharmaceutically acceptable salt thereof is at least five-times more potent as an inhibitor of JAKl than as an inhibitor of TYK2.

In one embodiment a JAK inhibitor as described herein or a pharmaceutically acceptable salt thereof is at least ten-times more potent as an inhibitor of JAKl than as an inhibitor of TYK2.

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

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

Compounds of the invention may contain one or more asymmetric carbon atoms. Accordingly, the compounds may exist as diastereomers, enantiomers or mixtures thereof. The syntheses of the compounds may employ racemates, diastereomers or enantiomers as starting materials or as intermediates. Mixtures of particular diastereomeric compounds may be separated, or enriched in one or more particular diastereomers, by chromatographic or crystallization methods. Similarly, enantiomeric mixtures may be separated, or enantiomerically enriched, using the same techniques or others known in the art. Each of the asymmetric carbon or nitrogen atoms may be in the R or S configuration and both of these configurations are within the scope of the invention.

In the structures shown herein, where the stereochemistry of any particular chiral atom is not specified, then all stereoisomers are contemplated and included as the compounds of the invention. Where stereochemistry is specified by a solid wedge or dashed line representing a particular configuration, then that stereoisomer is so specified and defined. Unless otherwise specified, if solid wedges or dashed lines are used, relative stereochemistry is intended.

Another aspect includes prodrugs of the compounds described herein, including known amino-protecting and carboxy-protecting groups which are released, for example hydrolyzed, to yield the compound of the present invention under physiologic conditions.

The term "prodrug" refers to a precursor or derivative form of a

pharmaceutically active substance that is less efficacious to the patient compared to the parent drug and is capable of being enzymatically or hydro lyrically activated or converted into the more active parent form. See, e.g., Wilman, "Prodrugs in Cancer Chemotherapy" Biochemical Society Transactions, 14, pp. 375-382, 615th Meeting Belfast (1986) and Stella et al., "Prodrugs: A Chemical Approach to Targeted Drug Delivery," Directed Drug Delivery, Borchardt et al., (ed.), pp. 247-267, Humana Press (1985). Prodrugs include, but are not limited to, phosphate-containing prodrugs, thiophosphate-containing prodrugs, sulfate-containing prodrugs, peptide-containing prodrugs, D-amino acid-modified prodrugs, glycosylated prodrugs, β-lactam- containing prodrugs, optionally substituted phenoxyacetamide-containing prodrugs or optionally substituted phenylacetamide-containing prodrugs, and 5-fluorocytosine and 5-fluorouridine prodrugs.

A particular class of prodrugs are compounds in which a nitrogen atom in an amino, amidino, aminoalkyleneamino, iminoalkyleneamino or guanidino group is substituted with a hydroxy group, an alkylcarbonyl (-CO-R) group, an alkoxycarbonyl (-CO-OR), or an acyloxyalkyl-alkoxycarbonyl (-CO-0-R-0-CO-R) group where R is a monovalent or divalent group, for example alkyl, alkylene or aryl, or a group having the Formula -C(0)-0-CPlP2-haloalkyl, where PI and P2 are the same or different and are hydrogen, alkyl, alkoxy, cyano, halogen, alkyl or aryl. In a particular embodiment, the nitrogen atom is one of the nitrogen atoms of the amidino group. Prodrugs may be prepared by reacting a compound with an activated group, such as acyl groups, to bond, for example, a nitrogen atom in the compound to the exemplary carbonyl of the activated acyl group. Examples of activated carbonyl compounds are those containing a leaving group bonded to the carbonyl group, and include, for example, acyl halides, acyl amines, acyl pyridinium salts, acyl alkoxides, acyl phenoxides such as p-nitrophenoxy acyl, dinitrophenoxy acyl, fluorophenoxy acyl, and difluorophenoxy acyl. The reactions are generally carried out in inert solvents at reduced temperatures such as -78 °C to about 50 °C. The reactions may also be carried out in the presence of an inorganic base, for example potassium carbonate or sodium bicarbonate, or an organic base such as an amine, including pyridine, trimethylamine, triethylamine, triethanolamine, or the like.

Additional types of prodrugs are also encompassed. For instance, a free carboxyl group of a JAK inhibitor as described herein can be derivatized as an amide or alkyl ester. As another example, compounds of the present invention comprising free hydroxy groups can be derivatized as prodrugs by converting the hydroxy group into a group such as, but not limited to, a phosphate ester, hemisuccinate,

dimethylaminoacetate, or phosphoryloxymethyloxycarbonyl group, as outlined in Fleisher, D. et al., (1996) Improved oral drug delivery: solubility limitations overcome by the use of prodrugs Advanced Drug Delivery Reviews, 19: 115.

Carbamate prodrugs of hydroxy and amino groups are also included, as are carbonate prodrugs, sulfonate esters and sulfate esters of hydroxy groups. Derivatization of hydroxy groups as (acyloxy)methyl and (acyloxy)ethyl ethers, wherein the acyl group can be an alkyl ester optionally substituted with groups including, but not limited to, ether, amine and carboxylic acid functionalities, or where the acyl group is an amino acid ester as described above, are also encompassed. Prodrugs of this type are described in J. Med. Chem., (1996), 39: 10. More specific examples include replacement of the hydrogen atom of the alcohol group with a group such as (Ci_ C 6 )alkanoyloxymethyl, 1 -((C i_C 6 )alkanoyloxy)ethyl, 1 -methyl- 1 -((Ci_

C 6 )alkanoyloxy)ethyl, (Ci_C 6 )alkoxycarbonyloxymethyl, N-(Ci_

C 6 )alkoxycarbonylaminomethyl, succinoyl, (Ci_C 6 )alkanoyl, alpha-amino(Ci_ C 4 )alkanoyl, arylacyl and alpha-aminoacyl, or alpha-aminoacyl-alpha-aminoacyl, where each alpha-aminoacyl group is independently selected from the naturally occurring L-amino acids, P(0)(OH) 2 , -P(0)(0(Ci_Ce)alkyl) 2 or glycosyl (the radical resulting from the removal of a hydroxyl group of the hemiacetal form of a carbohydrate).

"Leaving group" refers to a portion of a first reactant in a chemical reaction that is displaced from the first reactant in the chemical reaction. Examples of leaving groups include, but are not limited to, halogen atoms, alkoxy and sulfonyloxy groups. Example sulfonyloxy groups include, but are not limited to, alkylsulfonyloxy groups (for example methyl sulfonyloxy (mesylate group) and trif uoromethylsulfonyloxy (triflate group)) and arylsulfonyloxy groups (for example p-toluenesulfonyloxy (tosylate group) and p-nitrosulfonyloxy (nosylate group)).

SYNTHESIS OF JANUS KINASE INHIBITOR COMPOUNDS

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

Compounds may be prepared singly or as compound libraries comprising at least 2, for example 5 to 1,000 compounds, or 10 to 100 compounds. Libraries of compounds may be prepared by a combinatorial 'split and mix' approach or by multiple parallel syntheses using either solution phase or solid phase chemistry, by procedures known to those skilled in the art. Thus according to a further aspect of the invention there is provided a compound library comprising at least 2 compounds of the present invention.

For illustrative purposes, reaction Schemes depicted below provide routes for synthesizing the compounds of the present invention as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used. Although some specific starting materials and reagents are depicted in the Schemes and discussed below, other starting materials and reagents can be substituted to provide a variety of derivatives or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.

In the preparation of compounds of the present invention, protection of remote functionality (e.g., primary or secondary amine) of intermediates may be necessary. The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods. Suitable amino-protecting groups include acetyl, trifluoroacetyl, benzyl, phenylsulfonyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9-fluorenylmethyleneoxycarbonyl (Fmoc). The need for such protection is readily determined by one skilled in the art. For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.

Other conversions commonly used in the synthesis of compounds of the present invention, and which can be carried out using a variety of reagents and conditions, include the following:

(1) Reaction of a carboxylic acid with an amine to form an amide. Such a transformation can be achieved using various reagents known to those skilled in the art but a comprehensive review can be found in Tetrahedron, 2005, 61, 10827-10852.

(2) Reaction of a primary or secondary amine with an aryl halide or pseudo halide, e.g., a triflate, commonly known as a "Buchwald-Hartwig cross-coupling," can be achieved using a variety of catalysts, ligands and bases. A review of these methods is provided in Comprehensive Organic Name Reactions and Reagents, 2010, 575-581.

A palladium cross-coupling reaction between an aryl halide and a vinyl boronic acid or boronate ester. This transformation is a type of "Suzuki-Miyaura cross- coupling," a class of reaction that has been thoroughly reviewed in Chemical Reviews, 1995, 95(7), 2457-2483.

The hydrolysis of an ester to give the corresponding carboxylic acid is well known to those skilled in the art and conditions include: for methyl and ethyl esters, the use of a strong aqueous base such as lithium, sodium or potassium hydroxide or a strong aqueous mineral acid such as HC1; for a tert-butyl ester, hydrolysis would be carried out using acid, for example, HC1 in dioxane or trifluoroacetic acid (TFA) in dichloromethane (DCM).

Reaction Scheme 1

Reaction Scheme 1 illustrates a synthesis for compounds 6, 8 and 10 therein. Compound 1 can be arylated with 4-bromo-l-(difluoromethoxy)-2-iodobenzene under palladium catalyzed conditions to generate compound 2. The nitro group of compound 2 can be reduced with conditions such as iron and ammonium chloride to generate amino aniline 3. Amide bond coupling with commercially available pyrazolo[l,5-a]pyrimidine-3-carboxylic acid in the presence of a coupling reagent such as, but not limited to, PyAOP, with an organic base such as, but not limited to DIPEA, and DMAP in an organic solvent such as, but not limited to, DMF provides compound 4. Compound 4 can be converted to the corresponding iodide 5 using conditions such as sodium iodide and Cul with a base such as N,N-dimethylethane- 1,2-diamine in a solvent such as tBuOH. Compounds of formula 7 may be formed from treatment of compound 4 with a substituted boronic acid (or ester) or BF 3 K salt under palladium catalyzed conditions with a base such as, but not limited to, cesium carbonate in a solvent such as, but not limited to, 1,4-dioxane. Additionally, compounds of formula 9 may be synthesized by treatment of compound 4 with an appropriately substituted phenol under Pd catalyzed coupling conditions with a base, such as, but not limited to, cesium carbonate in a solvent such as, but not limited to, toluene. Removal of the SEM protecting group of compounds of formulas 5, 7 and 9 to generate compounds of formulas 6, 8 and 10 can be accomplished with an acid such as, but not limited to HC1 in a solvent such as, but not limited to, 1,4-dioxane.

Reaction Scheme 2

Reaction Scheme 2 illustrates a synthesis for compounds of Formulas I— III therein. Compounds of formulas 6, 8 and 10 can be treated with an appropriately substituted 2-bromo-N,N-dimethylacetamide with a base such as, but not limited to, cesium carbonate in a solvent such as, but not limited to, DMF to afford compounds of Formulas I— III.

Reaction Scheme 3

Reaction Scheme 3 illustrates a synthesis for compounds of Formulas IV- VI therein. Compounds of Formula II (Ri = Br) can be treated with an appropriately substituted phenol under Pd catalyzed coupling conditions with a base, such as, but not limited to, cesium carbonate in a solvent such as, but not limited to, toluene to afford compounds of Formula IV. Compounds of Formula V can be obtained from treatment of compound 4 with a substituted boronic acid (or ester) or BF 3 K salt under palladium catalyzed conditions with an base such as, but not limited to, cesium carbonate in a solvent such as, but not limited to, 1,4-dioxane. Difluoromethyl compounds of Formula VI can be obtained using methods described in J. Am. Chem. Soc, 2014, 136, 4149-4152. Additionally, when Ri of compounds of Formula V is an appropriately substituted olefin, further manipulation of this olefin using standard methods can be accomplished to furnish fluorinated alkanes.

Reaction Scheme 4

Reaction Scheme 4 illustrates a synthesis for compounds of Formula VII therein. Commercially available 4-(difluoromethoxy)phenol can be treated with a brominating agent such as, but not limited to, NBS in a solvent such as, but not limited, acetic acid to yield 12. Difluoromethylation of 12 to form compound 13 can be accomplished by treatment of compound 12 with diethyl (bromodifluoromethyl)phosphonate with a base such as, but not limited to, aqueous potassium hydroxide in a solvent such as, but not limited to, acetonitrile. Compound 13 can be treated with 4-nitro-l-[[2- (trimethylsilyl)ethoxy]methyl] - 1 H-pyrazole 4-bromo- 1 -(difluoromethoxy)-2- iodobenzene under palladium catalyzed conditions with a base such as, but not limited to, potassium carbonate in a solvent such as, but not limited to, DMA to generate compound 14. The nitro group of compound 14 can be reduced with conditions such as iron and ammonium chloride to generate amino aniline 15. Amide bond coupling of compound 15 with commercially available pyrazolo[l,5-a]pyrimidine-3-carboxylic acid in the presence of a coupling reagent such as, but not limited to, PyAOP, with an organic base such as, but not limited to, DIPEA and DMAP in a solvent such as, but not limited to, DMF provides compound 16. Removal of the SEM protecting group of compound 16 can be accomplished with an acid such as, but not limited to, HC1 in an organic solvent such as, but not limited to, 1,4-dioxane to generate compounds of Formula VII.

Reaction Scheme 5

Reaction Scheme 5 illustrates a synthesis for compounds of Formula VIII therein. Compound 17 can be treated with 2-bromo-N,N-dimethylacetamide with a base such as, but not limited to, cesium carbonate in a solvent such as, but not limited to, DMF to afford compound 18. Removal of the Boc protecting group of compound 18 can be accomplished with an acid such as, but not limited to, HCl in an organic solvent such as, but not limited to, 1,4-dioxane to generate compounds of Formula VIII.

It will be appreciated that where appropriate functional groups exist, compounds of various formulae or any intermediates used in their preparation may be further derivatised by one or more standard synthetic methods employing

condensation, substitution, oxidation, reduction, or cleavage reactions. Particular substitution approaches include conventional alkylation, arylation, heteroarylation, acylation, sulfonylation, halogenation, nitration, formylation and coupling procedures.

In a further example, primary amine or secondary amine groups may be converted into amide groups (-NHCOR' or -NRCOR') by acylation. Acylation may be achieved by reaction with an appropriate acid chloride in the presence of a base, such as triethylamine, in a suitable solvent, such as dichloromethane, or by reaction with an appropriate carboxylic acid in the presence of a suitable coupling agent such HATU (0-(7-azabenzotriazol- 1 -yl)-N,N,N ' ,N ' -tetramethyluronium

hexafluorophosphate) in a suitable solvent such as dichloromethane. Similarly, amine groups may be converted into sulphonamide groups (-NHSO2R' or -NR"S02R') groups by reaction with an appropriate sulphonyl chloride in the presence of a suitable base, such as triethylamine, in a suitable solvent such as dichloromethane. Primary or secondary amine groups can be converted into urea groups (-NHCONR'R" or - NRCONR'R") by reaction with an appropriate isocyanate in the presence of a suitable base such as triethylamine, in a suitable solvent, such as dichloromethane.

An amine (-NH 2 ) may be obtained by reduction of a nitro (-N0 2 ) group, for example by catalytic hydrogenation, using for example hydrogen in the presence of a metal catalyst, for example palladium on a support such as carbon in a solvent such as ethyl acetate or an alcohol e.g., methanol. Alternatively, the transformation may be carried out by chemical reduction using for example a metal, e.g., tin or iron, in the presence of an acid such as hydrochloric acid.

In a further example, amine (-CH 2 NH 2 ) groups may be obtained by reduction of nitriles (-CN), for example by catalytic hydrogenation using for example hydrogen in the presence of a metal catalyst, for example palladium on a support such as carbon, or Raney nickel, in a solvent such as an ether e.g., a cyclic ether such as tetrahydrofuran, at an appropriate temperature, for example from about -78 °C to the reflux temperature of the solvent.

In a further example, amine (-NH 2 ) groups may be obtained from carboxylic acid groups (-C0 2 H) by conversion to the corresponding acyl azide (-CON 3 ), Curtius rearrangement and hydrolysis of the resultant isocyanate (-N=C=0).

Aldehyde groups (-CHO) may be converted to amine groups (-CH 2 NR'R")) by reductive amination employing an amine and a borohydride, for example sodium triacetoxyborohydride or sodium cyanoborohydride, in a solvent such as a

halogenated hydrocarbon, for example dichloromethane, or an alcohol such as ethanol, where necessary in the presence of an acid such as acetic acid at around ambient temperature.

In a further example, aldehyde groups may be converted into alkenyl groups (- CH=CHR') by the use of a Wittig or Wadsworth-Emmons reaction using an appropriate phosphorane or phosphonate under standard conditions known to those skilled in the art.

Aldehyde groups may be obtained by reduction of ester groups (such as - C0 2 Et) or nitriles (-CN) using diisobutylaluminium hydride in a suitable solvent such as toluene. Alternatively, aldehyde groups may be obtained by the oxidation of alcohol groups using any suitable oxidising agent known to those skilled in the art.

Ester groups (-C0 2 R') may be converted into the corresponding acid group (- C0 2 H) by acid- or base-catalused hydrolysis, depending on the nature of R. If R is t- butyl, acid-catalysed hydrolysis can be achieved for example by treatment with an organic acid such as trifluoroacetic acid in an aqueous solvent, or by treatment with an inorganic acid such as hydrochloric acid in an aqueous solvent.

Carboxylic acid groups (-C0 2 H) may be converted into amides (CONHR' or - CONR'R") by reaction with an appropriate amine in the presence of a suitable coupling agent, such as HATU, in a suitable solvent such as dichloromethane.

In a further example, carboxylic acids may be homologated by one carbon (i.e

-C0 2 H to -CH 2 C0 2 H) by conversion to the corresponding acid chloride (-COC1) followed by Arndt-Eistert synthesis.

In a further example, -OH groups may be generated from the corresponding ester (e.g., -C0 2 R') ? or aldehyde (-CHO) by reduction, using for example a complex metal hydride such as lithium aluminium hydride in diethyl ether or tetrahydrofuran, or sodium borohydride in a solvent such as methanol. Alternatively, an alcohol may be prepared by reduction of the corresponding acid (-C0 2 H), using for example lithium aluminium hydride in a solvent such as tetrahydrofuran, or by using borane in a solvent such as tetrahydrofuran.

Alcohol groups may be converted into leaving groups, such as halogen atoms or sulfonyloxy groups such as an alkylsulfonyloxy, e.g., trifluoromethylsulfonyloxy or arylsulfonyloxy, e.g., /?-toluenesulfonyloxy group using conditions known to those skilled in the art. For example, an alcohol may be reacted with thioyl chloride in a halogenated hydrocarbon (e.g., dichloromethane) to yield the corresponding chloride. A base (e.g., triethylamine) may also be used in the reaction.

In another example, alcohol, phenol or amide groups may be alkylated by coupling a phenol or amide with an alcohol in a solvent such as tetrahydrofuran in the presence of a phosphine, e.g., triphenylphosphine and an activator such as diethyl-, diisopropyl, or dimethylazodicarboxylate. Alternatively alkylation may be achieved by deprotonation using a suitable base e.g., sodium hydride followed by subsequent addition of an alkylating agent, such as an alkyl halide.

Aromatic halogen substituents in the compounds may be subjected to halogen- metal exchange by treatment with a base, for example a lithium base such as n-butyl or t-butyl lithium, optionally at a low temperature, e.g., around -78 °C, in a solvent such as tetrahydrofuran, and then quenched with an electrophile to introduce a desired substituent. Thus, for example, a formyl group may be introduced by using N,N- dimethylformamide as the electrophile. Aromatic halogen substituents may alternatively be subjected to metal (e.g., palladium or copper) catalysed reactions, to introduce, for example, acid, ester, cyano, amide, aryl, heteraryl, alkenyl, alkynyl, thio- or amino substituents. Suitable procedures which may be employed include those described by Heck, Suzuki, Stille, Buchwald or Hartwig.

Aromatic halogen substituents may also undergo nucleophilic displacement following reaction with an appropriate nucleophile such as an amine or an alcohol. Advantageously, such a reaction may be carried out at elevated temperature in the presence of microwave irradiation.

METHODS OF SEPARATION

In each of the exemplary Schemes it may be advantageous to separate reaction products from one another or from starting materials. The desired products of each step or series of steps is separated or purified (hereinafter separated) to the desired degree of homogeneity by the techniques common in the art. Typically such separations involve multiphase extraction, crystallization or trituration from a solvent or solvent mixture, distillation, sublimation, or chromatography. Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; supercritical fluid; high, medium, and low pressure liquid chromatography methods and apparatus; small scale analytical;

simulated moving bed (SMB) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography.

Another class of separation methods involves treatment of a mixture with a reagent selected to bind to or render otherwise separable a desired product, unreacted starting material, reaction by product, or the like. Such reagents include adsorbents or absorbents such as activated carbon, molecular sieves, ion exchange media, or the like. Alternatively, the reagents can be acids in the case of a basic material, bases in the case of an acidic material, binding reagents such as antibodies, binding proteins, selective chelators such as crown ethers, liquid/liquid ion extraction reagents (LIX), or the like.

Selection of appropriate methods of separation depends on the nature of the materials involved. Example separation methods include boiling point, and molecular weight in distillation and sublimation, presence or absence of polar functional groups in chromatography, stability of materials in acidic and basic media in multiphase extraction, and the like. One skilled in the art will apply techniques most likely to achieve the desired separation.

Diastereomeric mixtures can be separated into their individual

diastereoisomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography or fractional

crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereoisomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers. Also, some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention. Enantiomers can also be separated by use of a chiral HPLC column or supercritical fluid chromatography.

A single stereoisomer, e.g., an enantiomer, substantially free of its

stereoisomer may be obtained by resolution of the racemic mixture using a method such as formation of diastereomers using optically active resolving agents (Eliel, E. and Wilen, S., Stereochemistry of Organic Compounds, John Wiley & Sons, Inc., New York, 1994; Lochmuller, C. H., J. Chromatogr., 113(3):283-302 (1975)).

Racemic mixtures of chiral compounds of the invention can be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the substantially pure or enriched stereoisomers directly under chiral conditions. See: Drug Stereochemistry, Analytical Methods and Pharmacology, Irving W. Wainer, Ed., Marcel Dekker, Inc., New York (1993). Diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, a-methyl- -phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid. The diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography. For separation of the optical isomers of amino compounds, addition of chiral carboxylic or sulfonic acids, such as camphorsulfonic acid, tartaric acid, mandelic acid, or lactic acid can result in formation of the diastereomeric salts.

Alternatively, the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair (Eliel, E. and Wilen, S.,

Stereochemistry of Organic Compounds, John Wiley & Sons, Inc., New York, 1994, p. 322). Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yield the pure or enriched enantiomer. A method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, a-methoxy-a-(trifluoromethyl)phenyl acetate (Jacob, J. Org. Chem. 47:4165 (1982)), of the racemic mixture, and analyzing the NMR spectrum for the presence of the two atropisomeric enantiomers or

diastereomers. Stable diastereomers of atropisomeric compounds can be separated and isolated by normal- and reverse-phase chromatography following methods for separation of atropisomeric naphthyl-isoquino lines (WO 96/15111, incorporated herein by reference). By method (3), a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase (Chiral Liquid

Chromatography W. J. Lough, Ed., Chapman and Hall, New York, (1989); Okamoto, J. of Chromatogr. 513:375-378 (1990)). Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism. The absolute stereochemistry of chiral centers and enatiomers can be determined by x-ray crystallography.

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

PHARMACEUTICAL COMPOSITIONS AND ADMINISTRATION

The compounds with which the invention is concerned are JAK kinase inhibitors, such as JAKl inhibitors, and are useful in the treatment of several diseases, for example, inflammatory diseases, such as asthma.

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

In one example, a compound of the invention or a pharmaceutically acceptable salt thereof may be formulated by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed into a galenical administration form. The pH of the formulation depends mainly on the particular use and the concentration of compound, but typically ranges anywhere from about 3 to about 8. In one example, a compound of the invention or a pharmaceutically acceptable salt thereof is formulated in an acetate buffer, at pH 5. In another embodiment, the compounds of the present invention are sterile. The compound may be stored, for example, as a solid or amorphous composition, as a lyophilized formulation or as an aqueous solution.

Compositions are formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.

It will be understood that the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination and the severity of the particular disease undergoing treatment. Optimum dose levels and frequency of dosing will be determined by clinical trial, as is required in the pharmaceutical art. In general, the daily dose range for oral administration will lie within the range of from about 0.001 mg to about 100 mg per kg body weight of a human, often 0.01 mg to about 50 mg per kg, for example 0.1 to 10 mg per kg, in single or divided doses. In general, the daily dose range for inhaled administration will lie within the range of from about 0.1 μg to about 1 mg per kg body weight of a human, preferably 0.1 μg to 50 μg per kg, in single or divided doses. On the other hand, it may be necessary to use dosages outside these limits in some cases.

The compounds of the invention or a pharmaceutically acceptable salt thereof, may be administered by any suitable means, including oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intrathecal, inhaled and epidural and intranasal, and, if desired for local treatment, intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous

administration. In some embodiments, inhaled administration is employed.

The compounds of the present invention or a pharmaceutically acceptable salt thereof, may be administered in any convenient administrative form, e.g., tablets, powders, capsules, lozenges, granules, solutions, dispersions, suspensions, syrups, sprays, vapors, suppositories, gels, emulsions, patches, etc. Such compositions may contain components conventional in pharmaceutical preparations, e.g., diluents (e.g., glucose, lactose or mannitol), carriers, pH modifiers, buffers, sweeteners, bulking agents, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, perfuming agents, flavoring agents, other known additives as well as further active agents.

Suitable carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel, Howard C, et al, Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005. For example, carriers include solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drugs, drug stabilizers, gels, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, such like materials and combinations thereof, as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, pp 1289-1329, 1990). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated. Exemplary excipients include dicalcium phosphate, mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate or combinations thereof. A pharmaceutical composition may comprise different types of carriers or excipients depending on whether it is to be administered in solid, liquid or aerosol form, and whether it need to be sterile for such routes of administration.

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

For topical application to the skin, a compound may be made up into a cream, lotion or ointment. Cream or ointment formulations which may be used for the drug are conventional formulations well known in the art, for example as described in standard textbooks of pharmaceutics such as the British Pharmacopoeia.

Compounds of the invention or a pharmaceutically acceptable salt thereof may also be formulated for inhalation, for example, as a nasal spray, or dry powder or aerosol inhalers. For delivery by inhalation, the compound is typically in the form of microparticles, which can be prepared by a variety of techniques, including spray- drying, freeze-drying and micronisation. Aerosol generation can be carried out using, for example, pressure-driven jet atomizers or ultrasonic atomizers, such as by using propellant-driven metered aerosols or propellant-free administration of micronized compounds from, for example, inhalation capsules or other "dry powder" delivery systems.

By way of example, a composition of the invention may be prepared as a suspension for delivery from a nebulizer or as an aerosol in a liquid propellant, for example, for use in a pressurized metered dose inhaler (PMDI). Propellants suitable for use in a PMDI are known to the skilled person, and include CFC-12, HFA-134a, HFA-227, HCFC-22 (CC1 2 F 2 ) and HFA-152 (CH 4 F 2 and isobutane).

In some embodiments, a composition of the invention is in dry powder form, for delivery using a dry powder inhaler (DPI). Many types of DPI are known.

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

In the case of an aerosol-based formulation, an example is:

Compound of the invention* 24 mg / canister

Lecithin, NF Liq. Cone. 1.2 mg / canister

Trichlorofluoromethane, NF 4.025 g / canister

Dichlorodifluoromethane, NF 12.15 g / canister.

* or a pharmaceutically acceptable salt thereof

A compound of the invention or a pharmaceutically acceptable salt thereof may be dosed as described depending on the inhaler system used. In addition to the compound, the administration forms may additionally contain excipients as described above, or, for example, propellants (e.g., Frigen in the case of metered aerosols), surface-active substances, emulsifiers, stabilizers, preservatives, flavorings, fillers (e.g., lactose in the case of powder inhalers) or, if appropriate, further active compounds. For the purposes of inhalation, a large number of systems are available with which aerosols of optimum particle size can be generated and administered, using an inhalation technique which is appropriate for the patient. In addition to the use of adaptors (spacers, expanders) and pear-shaped containers (e.g., Nebulator®,

Volumatic®), and automatic devices emitting a puffer spray (Autohaler®), for metered aerosols, in the case of powder inhalers in particular, a number of technical solutions are available (e.g., Diskhaler®, Rotadisk®, Turbohaler® or the inhalers, for example, as described in U.S. Patent No. 5,263,475, incorporated herein by reference). Additionally, compounds of the invention or a pharmaceutically acceptable salt thereof, may be delivered in multi-chamber devices thus allowing for delivery of combination agents.

The compound or a pharmaceutically acceptable salt thereof, may also be administered parenterally in a sterile medium. Depending on the vehicle and concentration used, the compound can either be suspended or dissolved in the vehicle. Advantageously, adjuvants such as a local anaesthetic, preservative or buffering agent can be dissolved in the vehicle.

TARGETED INHALED DRUG DELIVERY

Compounds of the present invention may be intended for targeted inhaled delivery. Optimisation of drugs for delivery to the lung by topical (inhaled) administration has been recently reviewed (Cooper, A. E. et al. Curr. Drug Metab. 2012, 13, 457-473).

Due to limitations in the delivery device, the dose of an inhaled drug is likely to be low (approximately <lmg/day) in humans, which necessitates highly potent molecules. High potency against the target of interest is especially important for an inhaled drug due to factors such as the limited amount of drug that can be delivered in a single puff from an inhaler, and the safety concerns related to a high aerosol burden in the lung (for example, cough or irritancy). For example, in some embodiments, a Ki of about 0.5 nM or less in a JAKl biochemical assay such as described herein, and an IC50 of about 20 nM or less in a JAKl dependent cell based assay such as described herein, may be desirable for an inhaled JAKl inhibitor. In other

embodiments, the projected human dose of a compound of the present invention, or a pharmaceutically acceptable salt thereof, is at least two times less than the projected human dose of a compound known in the art. Accordingly, in some embodiments, compounds (or a pharmaceutically acceptable salt thereof) described herein demonstrate such potency values.

IL13 signaling is strongly implicated in asthma pathogenesis. IL13 is a cytokine that requires active JAKl in order to signal. Thus, inhibition of JAKl also inhibits IL13 signaling, which may provide benefit to asthma patients. Inhibition of IL13 signaling in an animal model (e.g., a mouse model) may predict future benefit to human asthmatic patients. Thus, it may be beneficial for an inhaled JAKl inhibitor to show suppression of IL13 signaling in an animal model. Methods of measuring such suppression are known in the art. For example, as discussed herein and is known in the art, JAKl -dependent STAT6 phosphorylation is known downstream of IL13 stimulation. Accordingly, in some embodiments, compounds (or a pharmaceutically acceptable salt thereof) described herein demonstrate inhibition of lung pSTAT6 induction. To examine pharmacodynamic effects on pSTAT6 levels, compounds of the invention were co-dosed intra-nasally with 1 μg IL13 to female Balb/c mice. Compounds were formulated in 0.2% (v:v) Tween 80 in saline and mixed 1 : 1 (v:v) with IL13 immediately prior to administration. The intranasal doses were

administered to lightly anaesthetised (isoflurane) mice by dispensing a fixed volume (50 μί) directly into the nostrils by pipette to achieve the target dose level (3 mg/kg, 1 mg/kg, 0.3 mg/kg, 0.1 mg/kg). At 0.25 hr post dose, blood samples (ca 0.5 mL) were collected by cardiac puncture and plasma generated by centrifugation (1500g, 10 min, +4 °C). The lungs were perfused with chilled phosphate buffer saline (PBS), weighed and snap frozen in liquid nitrogen. All samples were stored at ca. -80 °C until analysis. Defrosted lung samples were weighed and homogenised following the addition of 2 mL HPLC grade water for each gram of tissue, using an Omni -Prep Bead Ruptor at 4°C. Plasma and lung samples were extracted by protein precipitation with three volumes of acetonitrile containing Tolbutamide (50 ng/mL) and Labetalol (25 ng/mL) as analytical internal standards. Following vortex mixing and

centrifugation for 30 minutes at 3200 g and 4 °C, the supernatants were diluted appropriately (e.g., 1 : 1 v:v) with HPLC grade water in a 96-well plate.

Representative aliquots of plasma and lung samples were assayed for the parent compound by LC-MS/MS, against a series of matrix matched calibration and quality control standards. The standards were prepared by spiking aliquots of control Balb/c mouse plasma or lung homogenate (2: 1 in HPLC grade water) with test compound and extracting as described for the experimental samples. A lung:plasma ratio was determined as the ratio of the mean lung concentration (μΜ) to the mean plasma concentration (μΜ) at the sampling time (0.25h). Theoretical target engagement was calculated with the following equation, assuming that all drug was within lung tissue and the fraction unbound was available to interact with the target:

(unbound tissue concentration/(unbound tissue concentration + in vitro cellular potency i.e., IC50))* 100

To measure pSTAT6 levels, mouse lungs were stored frozen at -80 °C until assay and homogenised in 0.6 ml ice-cold cell lysis buffer (Cell Signalling

Technologies, catalogue # 9803 S) supplemented with 1 mM PMSF and a cocktail of protease (Sigma Aldrich, catalogue # P8340) and phosphatase (Sigma Aldrich, catalogue # P5726 and P0044) inhibitors. Samples were centrifuged at 16060 x g for 4 minutes at 4 °C to remove tissue debris and protein concentration of homogenates determined using the Pierce BCA protein assay kit (catalogue # 23225). Samples were diluted to a protein concentration of 5 mg/ml in ice-cold distilled water and assayed for pSTAT6 levels by Meso Scale Discovery electro-chemiluminescent immuno-assay. Briefly, 5 μΐ/well 150 μg/ml STAT6 capture antibody (R&D

Systems, catalogue # MAB 2169) was coated onto 96 well Meso Scale Discovery High Binding Plates (catalogue # L15XB-3) and air-dried for 5 hours at room temperature. Plates were blocked by addition of 150μ1Λνε11 30mg/ml Meso Scale Discovery Blocker A (catalogue # R93BA-4) and incubation for 2 hours at room temperature on a microplate shaker. Blocked plates were washed 4 times with Meso Scale Discovery TRIS wash buffer (catalogue # R61TX-1), followed by transfer of 50 μΐ/well lung homogenate to achieve a protein loading of 250 μg/well. Assay plates were incubated overnight at 4 °C and washed 4 times with TRIS wash buffer before addition of 25 μΐ/well 2.5 μg/ml sulfotag-labelled pSTAT6 detection antibody (BD Pharmingen, catalogue # 558241) for 2 hours at room temperature on a microplate shaker. Plates were washed 4 times with TRIS wash buffer and 150 μΐ/well IX Meso Scale Discovery Read Buffer T (catalogue # R92TC-1) added. Lung homogenate pSTAT6 levels were quantified by detection of electro-chemiluminescence on a Meso Scale Discovery SECTOR S 600 instrument.

Selectivity between JAK1 and JAK2 may be important for an inhaled JAK1 inhibitor. For example, GMCSF (granulocyte -macrophage colony- stimulating factor) is a cytokine that signals through JAK2 exclusively. Neutralization of GMCSF activity is associated with pulmonary alveolar proteinosis (PAP) in the lung. However, submaximal JAK2 suppression does not appear to be associated with PAP. Thus, even modest JAKl vs JAK2 selectivity may be of benefit in avoiding full suppression of the GMCSF pathway and avoiding PAP. For example, compounds with about 2x-5x selectivity for JAKl over JAK2 may be of benefit for an inhaled JAKl inhibitor. Accordingly, in some embodiments, compounds (or a

pharmaceutically acceptable salt thereof) described herein demonstrate such selectivity. Methods of measuring JAKl and JAK2 selectivity are known in the art, and information can also be found in the Examples herein.

Additionally, it may be desirable for an inhaled JAKl inhibitor to be selective over one or more other kinases to reduce the likelihood of potential toxicity due to off-target kinase pathway suppression. Thus, it may also be of benefit for an inhaled JAKl inhibitor to be selective against a broad panel of non-JAK kinases, such as in protocols available from ThermoFisher Scientific's SelectScreen™ Biochemical Kinase Profiling Service using Adapta™ Screening Protocol Assay Conditions (Revised July 29, 2016), LanthaScreen™ Eu Kinase Binding Assay Screening

Protocol and Assay Conditions (Revised June 7, 2016), and/or Z'LYTE™ Screening Protocol and Assay Conditions (Revised September 16, 2016). For example, a compound of the present invention, or a pharmaceutically acceptable salt thereof, exhibits at least 50-fold selectivity for JAKl versus a panel of non-JAK kinases.

Accordingly, in some embodiments, compounds (or a pharmaceutically acceptable salt thereof) described herein demonstrate such selectivity.

Hepatocyte toxicity, general cytotoxicity or cytotoxicity of unknown mechanism is an undesirable feature for a potential drug, including inhaled drugs. It may be of benefit for an inhaled JAKl inhibitor to have low intrinsic cytotoxicity against various cell types. Typical cell types used to assess cytotoxicity include both primary cells such as human hepatocytes, and proliferating established cell lines such as Jurkat, HEK-293, and H23. For example, it may be of benefit for an inhaled JAKl inhibitor to have an IC 50 of greater than 50 μΜ or greater than 100 μΜ in cytotoxicity measurements against such cell types. Accordingly, in some embodiments, compounds (or a pharmaceutically acceptable salt thereof) described herein demonstrate such values. Methods of measuring cytotoxicity are known in the art. In some embodiments, compounds described herein were tested as follows:

(a) Jurkat, H23, and HEK293T cells were maintained at a sub confluent density in T175 flasks. Cells were plated at 450 cells/45 μΐ medium in Greiner 384 well black/clear tissue culture treated plates. (Greiner Catalog # 781091). After dispensing cells, plates were equilibrated at room temperature for 30 minutes. After 30 minutes at room temperature, cells were incubated overnight at 37 °C in a C0 2 and humidity controlled incubator. The following day, cells were treated with compounds diluted in 100% DMSO (final DMSO concentration on cells = 0.5%) with a 10 point dose-response curve with a top concentration of 50 μΜ. Cells and compounds were then incubated for 72 hours overnight at 37 °C in a C0 2 and humidity controlled incubator. After 72 hours of incubation, viability was measured using CellTiterGlo® (Promega Catalog# G7572) to all wells. After incubation at room temperature for 20 minutes, plates were read on EnVision™ (Perkin Elmer Life Sciences) using luminescence mode;

(b) using human primary hepatocytes: the test compound was prepared as a 10 mM solution in DMSO. Additionally, a positive control such as Chlorpromazine was prepared as a 10 mM solution in DMSO. Test compounds were typically assessed using a 7-point dose response curve with 2-fold dilutions. Typically, the maximum concentration tested was 50-100 μΜ. The top concentration was typically dictated by solubility of the test compound. Cryopreserved primary human hepatocytes (BioreclamationIVT)(lot IZT) were thawed in InVitroGro™ HT thawing media (BioreclamationlVT) at 37 °C, pelleted and resuspended. Hepatocyte viability was assessed by Trypan blue exclusion and cells were plated in black-walled, BioCoat™ collagen 384- well plates (Corning BD) at a density of 13,000 cells/well in InVitroGro™ CP plating media supplemented with 1% Torpedo™ Antibiotic Mix (BioreclamationlVT) and 5% fetal bovine serum. Cells were incubated overnight for 18 hours (37 °C, 5%> C0 2 ) prior to treatment. Following 18 hours incubation, plating media was removed and hepatocytes were treated with compounds diluted in InVitroGro™ HI incubation media containing 1% Torpedo™ Antibiotic Mix and 1% DMSO (serum-free conditions). Hepatocytes were treated with test compounds at concentrations such as 0.78, 1.56, 3.12, 6.25, 12.5, 25, and 50 μΜ at a final volume of 50 μΐ,. A positive control (e.g., Chlorpromazine) was included in the assay, typically at the same concentrations as the test compound. Additional cells were treated with 1% DMSO as a vehicle control. All treatments were for a 48 hour time period (at 37 °C, 5% C0 2 ) and each treatment condition was performed in triplicate. Following 48 hours of compound treatment, CellTiter-Glo® cell viability assay (Promega) was used as the endpoint assay to measure ATP content as a determination of cell viability. The assay was performed according to manufacture instructions. Luminescence was determined on an En Vision™ Muliplate Reader (PerkinElmer, Waltham, MA, USA). Luminescence data was normalized to vehicle (1% DMSO) control wells. Inhibition curves and IC 50 estimates were generated by non- linear regression of log-transformed inhibitor concentrations (7-point serial dilutions including vehicle) vs. normalized response with variable Hill slopes, with top and bottom constrained to constant values of 100 and 0, respectively (GraphPad Prism™, GraphPad Software, La Jolla, CA, USA).

Inhibition of the hERG (human ether-a-go-go-related gene) potassium channel may lead to long QT syndrome and cardiac arrhythmias. Although plasma levels of an inhaled JAK1 inhibitor are expected to be low, lung-deposited compound exiting the lung via pulmonary absorption into the bloodstream will circulate directly to the heart. Thus, local heart concentrations of an inhaled JAK1 inhibitor may be transiently higher than total plasma levels, particularly immediately after dosing.

Thus, it may be of benefit to minimize hERG inhibition of an inhaled JAK1 inhibitor. For example, in some embodiments, a hERG IC50 greater than 30x over the free-drug plasma Cmax is preferred. Accordingly, in some embodiments, compounds (or a pharmaceutically acceptable salt thereof) of the invention demonstrate minimized hERG inhibition under conditions such as:

(a) using hERG 2pt automatic patch clamp conditions to examine in vitro effects of a compound on hERG expressed in mammalizan cells, evaluated at room temperature using the QPatch HT® (Sophion Bioscience A/S, Denmark), an automatic parallel patch clamp system. In some cases, compounds were tested at only one or two concentrations such as 1 or 10 uM. In other cases a more extensive concentration response relationship was established to allow estimation of IC50. For example, test compound concentrations were selected to span the range of

approximately 10-90% inhibition in half-log increments. Each test article

concentration was tested in two or more cells (n > 2). The duration of exposure to each test article concentration was a minimum of 3 minutes; and/or

(b) those described in WO 2014/074775, in the Examples, under "Effect on Cloned hERG Potassium Channels Expressed in Mammalian Cells," a ChanTest™, a Charles River Company, protocol with the following changes: cells stably expressing hERG were held at -80 mV. Onset and steady state inhibition of hERG potassium current due to compound were measured using a pulse pattern with fixed amplitudes (conditioning prepulse: +20 mV for 1 s; repolarizing test ramepto -90 mV (-0.5 V/s) repeated at 5 s intervals). Each recording ended with a final application of a supramaximal concentration of a reference substance, E-4021 (500 nM) (Charles River Company). The remaining uninhibited current was subtracted off-line digitially from the data to determine the potency of the test substance for hERG inhibition.

CYP (cytochrome P450) inhibition may not be a desirable feature for an inhaled JAK1 inhibitor. For example, a reversible or time dependent CYP inhibitor may cause an undesired increase in its own plasma levels, or in the plasma levels of other co-administered drugs (drug-drug interactions). Additionally, time dependent CYP inhibition is sometimes caused by biotransformation of parent drug to a reactive metabolite. Such reactive metabolites may covalently modify proteins, potentially leading to toxicity. Thus, minimizing reversible and time dependent CYP inhibition may be of benefit to an inhaled JAK1 inhibitor. Accordingly, in some embodiments, compounds (or a pharmaceutically acceptable salt thereof) of the present invention demonstrate minimal or no reversible and/or time dependent CYP inhibition.

Methods of measuring CYP inhibition are known in the art. CYP inhibition of compounds described herein were assessed over a concentration range of 0.16 - 10 uM of compound using pooled (n=150) human liver microsomes (Corning,

Tewksbury, MA) using methods previously reported (Halladay et al, Drug Metab.

Lett. 2011, 5, 220-230). Incubation duration and protein concentration was dependent on the CYP isoform and the probe substrate/metabolites assessed. The following substrate/metabolites, and incubation times and protein concentrations for each CYP were used: CYP1A2, phenacetin/acetaminophen, 30 min, 0.03 mg/ml protein;

CYP2C9, warfarin/7-hydroxywarfarin, 30 min, 0.2 mg/ml protein; CYP2C19, mephenytoin/4-hydroxymephenytoin, 40 min, 0.2 mg/ml protein; CYP2D6, dextromethorphan/dextrorphan, 10 min, 0.03 mg/ml protein; CYP3A4, midazolam/1- hydroxymidazolam, 10 min, 0.03 mg/ml protein and CYP3A4 testosterone/ββ- hydroxytestosterone, 10 min, 0.06 mg/ml protein. These conditions were previously determined to be in the linear rate of formation for the CYP-specific metabolites. All reaction were initiated with ImM NADPH and terminated by the addition of 0.1% formic acid in acetonitrile containing appropriate stable labeled internal standard. Samples were analyzed by LC-MS/MS. For compounds destined to be delivered via dry powder inhalation there is also a requirement to be able to generate crystalline forms of the compound that can be micronized to 1-5 μιη in size. Particle size is an important determinant of lung deposition of an inhaled compound. Particles with a diameter of less than 5 microns (μηι) are typically defined as respirable. Particles with a diameter larger than 5 μιη are more likely to deposit in the oropharynx and are correspondingly less likely to be deposited in the lung. Additionally, fine particles with a diameter of less than 1 μιη are more likely than larger particles to remain suspended in air, and are

correspondingly more likely to be exhaled from the lung. Thus, a particle diameter of 1-5 μιη may be of benefit for an inhaled medication whose site of action is in the lung. Typical methods used to measure particle size include laser diffraction and cascade impaction. Typical values used to define particle size include:

• D10, D50, and D90. These are measurements of particle diameter that

indicate, respectively, 10%, 50%, or 90%> of the sample is below that value. For example a D50 of 3 μιη indicates that 50% of the sample is below 3 μιη in size.

• Mass mean aerodynamic diameter (MM AD). MM AD is the diameter at which 50% of the particles by mass are larger and 50% are smaller. MM AD is a measure of central tendency.

· Geometric Standard Deviation (GSD). GSD is a measure of the magnitude in dispersity from the MMAD, or the spread in aerodynamic particle size distribution.

A common formulation for inhaled medications is a dry powder preparation including the active pharmaceutical ingredient (API) blended with a carrier such as lactose with or without additional additives such as magnesium stearate. For this formulation and others, it may be beneficial for the API itself to possess properties that allow it to be milled to a respirable particle size of 1-5 μιη. Agglomeration of particles is to be avoided, which can be measured by methods known in the art, such as examining D90 values under different pressure conditions. Accordingly, in some embodiments, compounds (or a pharmaceutically acceptable salt thereof) of the present invention can be prepared with such a respirable particle size with little or no agglomeration.

As for crystallinity, for some formulations of inhaled drugs, including lactose blends, it is important that API of a specific crystalline form is used. Crystallinity and crystalline form may impact many parameters relevant to an inhaled drug including but not limited to: chemical and aerodynamic stability over time, compatibility with inhaled formulation components such as lactose, hygroscopicity, lung retention, and lung irritancy. Thus, a stable, reproducible crystalline form may be of benefit for an inhaled drug. Additionally, the techniques used to mill compounds to the desired particle size are often energetic and may cause low melting crystalline forms to convert to other crystalline forms, or to become fully or partially amorphous. A crystalline form with a melting point of less than 150 °C may be incompatible with milling, while a crystalline form with a melting point of less than 100 °C is likely to be non-compatible with milling. Thus, it may be beneficial for an inhaled medication to have a melting point of at least greater than 100 °C, and ideally greater than 150 °C. Accordingly, in some embodiments, compounds (or a pharmaceutically acceptable salt thereof) described herein demonstrate such properties.

Additionally, minimizing molecular weight may help to lower the efficacious dose of an inhaled JAK1 inhibitor. Lower molecular weight results in a

corresponding higher number of molecules per unit mass of the active pharmaceutical ingredient (API). Thus, it may be of benefit to find the smallest molecular weight inhaled JAK1 inhibitor that retains all the other desired properties of an inhaled drug.

Finally, the compound needs to maintain a sufficient concentration in the lung over a given time period so as to be able to exert a pharmacological effect of the desired duration, and for pharmacological targets where systemic inhibition of said target is undesired, to have a low systemic exposure. The lung has an inherently high permeability to both large molecules (proteins, peptides) as well as small molecules with concomitant short lung half-lives, thus it may be necessary to attenuate the lung absorption rate through modification of one or more features of the compounds: minimizing membrane permeability, increasing pKa, increasing cLogP, decreasing solubility, reducing dissolution rate, or introducing a degree of basicity (e.g., introducing an amine) into the compound to enhance binding to the phospholipid-rich lung tissue or through trapping in acidic sub-cellular compartments such as lysosomes (pH 5). Methods of measuring such properties are known in the art.

Accordingly, in some embodiments, a compound of the present invention (or a pharmaceutically acceptable salt thereof) exhibits one or more of the above features. Further, in some embodiments, a compound of the present invention favorably exhibits one or more of these features relative to a compound known in the art - this may be particularly true for compounds of the art intended as oral drugs versus inhaled. For example, compounds with rapid oral absorption are typically poorly retained in the lung on inhalation. METHODS OF TREATMENT WITH AND USES OF JANUS KINASE INHIBITORS

The compounds of the present invention or a pharmaceutically acceptable salt thereof, inhibit the activity of a Janus kinase, such as JAK1 kinase. For example, a compound or a pharmaceutically acceptable salt thereof inhibits the phosphorylation of signal transducers and activators of transcription (STATs) by JAK1 kinase as well as STAT mediated cytokine production. Compounds of the present invention are useful for inhibiting JAK1 kinase activity in cells through cytokine pathways, such as IL-6, IL-15, IL-7, IL-2, IL-4, IL-9, IL-10, IL-13, IL-21, G-CSF, IFNalpha, IFNbeta or IFNgamma pathways. Accordingly, in one embodiment is provided a method of contacting a cell with a compound of the present invention or a pharmaceutically acceptable salt thereof, to inhibit a Janus kinase activity in the cell (e.g., JAK1 activity).

The compounds can be used for the treatment of immunological disorders driven by aberrant IL-6, IL-15, IL-7, IL-2, IL-4, IL9, IL-10, IL-13, IL-21, G-CSF, IFNalpha, IFNbeta or IFNgamma cytokine signaling.

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

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

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

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

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

In another embodiment, a compound of the present invention or a

pharmaceutically acceptable salt thereof may be used to treat lung diseases such as a fibrotic lung disease or an interstitial lung disease (e.g., an interstitial pneumonia). In some embodiments, a compound of the present invention or a pharmaceutically acceptable salt thereof may be used to treat idiopathic pulmonary fibrosis (IPF), systemic sclerosis interstitial lung disease (SSc-ILD)), nonspecific interstitial pneumonia (NSIP), rheumatoid arthritis-associated interstitial lung disease (RA-ILD), sarcoidosis, hypersensitivity pneumonitis, or ILD secondary to connective tissue disease beyond scleroderma (e.g., polymyositis, dermatomyositis, rheumatoid arthritis, systemic lupus erythematosus (SLE), or mixed connective tissue disease).

In one embodiment, the cancer is breast, ovary, cervix, prostate, testis, penile, genitourinary tract, seminoma, esophagus, larynx, gastric, stomach, gastrointestinal, skin, keratoacanthoma, follicular carcinoma, melanoma, lung, small cell lung carcinoma, non-small cell lung carcinoma (NSCLC), lung adenocarcinoma, squamous carcinoma of the lung, colon, pancreas, thyroid, papillary, bladder, liver, biliary passage, kidney, bone, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, salivary gland, pharynx, small intestine, colon, rectum, anal, renal, prostate, vulval, thyroid, large intestine, endometrial, uterine, brain, central nervous system, cancer of the peritoneum, hepatocellular cancer, head cancer, neck cancer, Hodgkin's or leukemia.

In one embodiment, the disease is a myeloproliferative disorder. In one embodiment, the myeloproliferative disorder is polycythemia vera, essential thrombocytosis, myelofibrosis or chronic myelogenous leukemia (CML).

Another embodiment includes the use of a compound of the present invention or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a disease described herein (e.g., an inflammatory disorder, an immunological disorder or cancer). In one embodiment, the invention provides a method of treating a disease or condition as described herein e.g., an inflammatory disorder, an immunological disorder or cancer) by targeting inhibition of a JAK kinase, such as JAK1.

COMBINATION THERAPY

The compounds may be employed alone or in combination with other agents for treatment. The second or further (e.g., third) compound of a pharmaceutical composition or dosing regimen typically has complementary activities to the compound of this invention such that they do not adversely affect each other. Such agents are suitably present in combination in amounts that are effective for the purpose intended. The compounds may be administered together in a unitary pharmaceutical composition or separately and, when administered separately this may occur simultaneously or sequentially. Such sequential administration may be close or remote in time.

For example, other compounds may be combined with a compound of the present invention or a pharmaceutically acceptable salt thereof for the prevention or treatment of inflammatory diseases, such as asthma. Suitable therapeutic agents for a combination therapy include, but are not limited to: an adenosine A2A receptor antagonist; an anti-infective; a non-steroidal Glucocorticoid Receptor (GR Receptor) agonist; an antioxidant; a β2 adrenoceptor agonist; a CCRl antagonist; a chemokine antagonist (not CCRl); a corticosteroid; a CRTh2 antagonist; a DPI antagonist; a formyl peptide receptor antagonist; a histone deacetylase activator; a chloride channel hCLCAl blocker; an epithelial sodium channel blocker (ENAC blocker; an intercellular adhesion molecule 1 blocker (ICAM blocker); an IKK2 inhibitor; a TNK inhibitor; a transient receptor potential ankyrin 1 (TRPA1) inhibitor; a Bruton's tyrosine kinase (BTK) inhibitor (e.g., fenebrutinib); a spleen tyrosine kinase (SYK) inhibitor; a tryptase-beta antibody; an ST2 receptor antibody (e.g., AMG 282); a cyclooxygenase inhibitor (COX inhibitor); a lipoxygenase inhibitor; a leukotriene receptor antagonist; a dual β2 adrenoceptor agonist/M3 receptor antagonist (MABA compound); a MEK-1 inhibitor; a myeloperoxidase inhibitor (MPO inhibitor); a muscarinic antagonist; a p38 MAPK inhibitor; a phosphodiesterase PDE4 inhibitor; a phosphatidylinositol 3 -kinase δ inhibitor (PI3 -kinase δ inhibitor); a

phosphatidylinositol 3 -kinase γ inhibitor (PI3 -kinase γ inhibitor); a peroxisome proliferator activated receptor agonist (PPARy agonist); a protease inhibitor; a retinoic acid receptor modulator (RAR γ modulator); a statin; a thromboxane antagonist; a TLR7 receptor agonist; or a vasodilator.

In addition, a compound of the present invention or a pharmaceutically acceptable salt thereof, may be combined with: (1) corticosteroids, such as alclometasone dipropionate, amelometasone, beclomethasone dipropionate, budesonide, butixocort propionate, biclesonide, clobetasol propionate,

desisobutyrylciclesonide, dexamethasone, etiprednol dicloacetate, fluocinolone acetonide, fluticasone furoate, fluticasone propionate, loteprednol etabonate (topical) or mometasone furoate; (2) 2-adrenoreceptor agonists such as salbutamol, albuterol, terbutaline, fenoterol, bitolterol, carbuterol, clenbuterol, pirbuterol, rimoterol, terbutaline, tretoquinol, tulobuterol and long acting 2-adrenoreceptor agonists such as metaproterenol, isoproterenol, isoprenaline, salmeterol, indacaterol, formoterol (including formoterol fumarate), arformoterol, carmoterol, abediterol, vilanterol trifenate, or olodaterol; (3) corticosteroid/long acting β2 agonist combination products such as salmeterol/fiuticasone propionate (Advair®, also sold as Seretide®), formoterol/budesonide (Symbicort®), formoterol/fluticasone propionate

(Flutiform®), formoterol/ciclesonide, formoterol/mometasone furoate,

indacaterol/mometasone furoate, vilanterol trifenate/fiuticasone furoate (BREO ELLIPTA), or arformoterol/ciclesonide; (4) anticholinergic agents, for example, muscarinic-3 (M3) receptor antagonists such as ipratropium bromide, tiotropium bromide, aclidinium bromide (LAS-34273), glycopyrronium bromide, or

umeclidinium bromide; (5) M3-anticholinergic/ 2-adrenoreceptor agonist

combination products such as vilanterol /umeclidinium (Anoro® Ellipta®), olodaterol/tiotropium bromide, glycopyrronium bromide/indacaterol (Ultibro®, also sold as Xoterna®), fenoterol hydrobromide/ipratropium bromide (Berodual®), albuterol sulfate/ipratropium bromide (Combivent®), formoterol

fumarate/glycopyrrolate, or aclidinium bromide/formoterol; (6) dual pharmacology M3-anticholinergic/ 2-adrenoreceptor agonists such as batefenterol succinate, AZD- 2115 or LAS- 190792; (7) leukotriene modulators, for example, leukotriene antagonists such as montelukast, zafirulast or pranlukast or leukotriene biosynthesis inhibitors such as zileuton, or LTB4 antagonists such as amelubant, or FLAP inhibitors such as fibofiapon, GSK-2190915; (8) phosphodiesterase-IV (PDE-IV) inhibitors (oral or inhaled), such as roflumilast, cilomilast, oglemilast, rolipram, tetomilast, AVE-8112, revamilast, CHF 6001; (9) antihistamines, for example, selective histamine- 1 (HI) receptor antagonists such as fexofenadine, citirizine, loratidine or astemizole or dual H1/H3 receptor antagonists such as GSK 835726, or GSK 1004723; (10) antitussive agents, such as codeine or dextramorphan; (11) a mucolytic, for example, N-acetyl cysteine or fudostein; (12) a

expectorant/mucokinetic modulator, for example, ambroxol, hypertonic solutions (e.g., saline or mannitol) or surfactant; (13) a peptide mucolytic, for example, recombinant human deoxyribonoclease I (dornase-alpha and rhDNase) or helicidin; (14) antibiotics, for example azithromycin, tobramycin or aztreonam; (15) non- selective COX-l/COX-2 inhibitors, such as ibuprofen or ketoprofen; (16) COX-2 inhibitors, such as celecoxib and rofecoxib; (17) VLA-4 antagonists, such as those described in WO 97/03094 and WO 97/02289, each incorporated herein by reference; (18) TACE inhibitors and TNF-a inhibitors, for example anti-TNF monoclonal antibodies, such as Remicade® and CDP-870 and TNF receptor immunoglobulin molecules, such as Enbrel®; (19) inhibitors of matrix metalloprotease, for example MMP-12; (20) human neutrophil elastase inhibitors, such as BAY-85-8501 or those described in WO 2005/026124, WO 2003/053930 and WO 2006/082412, each incorporated herein by reference; (21) A2b antagonists such as those described in WO 2002/42298, incorporated herein by reference; (22) modulators of chemokine receptor function, for example antagonists of CCR3 and CCR8; (23) compounds which modulate the action of other prostanoid receptors, for example, a thromboxane A 2 antagonist; DPI antagonists such as laropiprant or asapiprant CRTH2 antagonists such as OC000459, fevipiprant, ADC 3680 or ARRY 502; (24) PPAR agonists including PPAR alpha agonists (such as fenofibrate), PPAR delta agonists, PPAR gamma agonists such as pioglitazone, rosiglitazone and balaglitazone; (25) methylxanthines such as theophylline or aminophylline and

methylxanthine/corticosteroid combinations such as theophylline/budesonide, theophylline/fluticasone propionate, theophylline/ciclesonide,

theophylline/mometasone furoate and theophylline/beclometasone dipropionate; (26) A2a agonists such as those described in EP 1052264 and EP 1241176; (27) CXCR2 or IL-8 antagonists such as AZD-5069, AZD-4721, or danirixin; (28) IL-R signalling modulators such as kineret and ACZ 885; (29) MCP-1 antagonists such as ABN-912; (30) a p38 MAPK inhibitor such as BCT197, JNJ49095397, losmapimod or PH- 797804; (31) TLR7 receptor agonists such as AZD 8848; (32) PI3-kinase inhibitors such as RV1729 or GSK2269557 (nemiralisib); (33) triple combination products such as TRELEGY ELLIPTA (fluticasone furoate, umeclidinium bromide, and vilanterol); or (34) small molecule inhibitors of TRPA1, BTK, or SYK.

In some embodiments a compound of the present invention or a

pharmaceutically acceptable salt thereof, can be used in combination with one or more additional drugs, for example anti-hyperproliferative, anti-cancer, cytostatic, cytotoxic, anti-inflammatory or chemotherapeutic agents, such as those agents disclosed in U.S. Publ. Appl. No. 2010/0048557, incorporated herein by reference. A compound of the present invention or a pharmaceutically acceptable salt thereof, can be also used in combination with radiation therapy or surgery, as is known in the art.

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

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

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

(b) instructions for use.

In another embodiment, the kit further comprises: (c) a second pharmaceutical composition, such as a pharmaceutical composition comprising an agent for treatment as described above, such as an agent for treatment of an inflammatory disorder, or a chemotherapeutic agent.

In one embodiment, the instructions describe the simultaneous, sequential or separate administration of said first and second pharmaceutical compositions to a patient in need thereof.

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

Containers for use include, for example, bottles, vials, syringes, blister pack, etc. The containers may be formed from a variety of materials such as glass or plastic. The container includes a compound of the present invention or a

pharmaceutically acceptable salt thereof, which is effective for treating the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The label or package insert indicates that the compound is used for treating the condition of choice, such as asthma or cancer. In one embodiment, the label or package inserts indicates that the compound can be used to treat a disorder. In addition, the label or package insert may indicate that the patient to be treated is one having a disorder characterized by overactive or irregular Janus kinase activity, such as overactive or irregular JAK1 activity. The label or package insert may also indicate that the compound can be used to treat other disorders.

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

In order to illustrate the invention, the following examples are included.

However, it is to be understood that these examples do not limit the invention and are only meant to suggest a method of practicing the invention. Persons skilled in the art will recognize that the chemical reactions described may be readily adapted to prepare other compounds of the present invention, and alternative methods for preparing the compounds are within the scope of this invention. For example, the synthesis of non- exemplified compounds according to the invention may be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by utilizing other suitable reagents known in the art other than those described, or by making routine modifications of reaction conditions.

Alternatively, other reactions disclosed herein or known in the art will be recognized as having applicability for preparing other compounds of the invention.

EXAMPLES

General Experimental Details

All solvents and commercial reagents were used as received unless otherwise stated. Where products were purified by chromatography on silica this was carried out using either a glass column manually packed with silica gel (Kieselgel 60, 220-440 mesh, 35-75 μιη) or an Isolute ® SPE Si II cartridge. 'Isolute SPE Si cartridge' refers to a pre-packed polypropylene column containing unbonded activated silica with irregular particles with average size of 50 μιη and nominal 60 A porosity. Where an Isolute ® SCX-2 cartridge was used, 'Isolute ® SCX-2 cartridge' refers to a pre-packed polypropylene column containing a non-end-capped propylsulphonic acid functionalised silica strong cation exchange sorbent.

LCMS Conditions

Method A

Experiments were performed on a SHIMADZU LCMS-2020 with a CIS-reverse- phase column (50 x 3 mm Shim-Pack XR-ODS, 2.2 μιη particle size), elution with solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Gradient - Time flow ml/min %A %B

0.00 1.2 95 5

2.00 1.2 5 95

2.70 1.2 5 95

2.75 1.2 95 5 Detection - UV (220 and 254 nm) and ELSD Method B

Experiments were performed on a SHIMADZU LCMS-2020 with a CIS-reverse- phase column (50 x 3 mm Shim-Pack XR-ODS, 2.2 μιη particle size), elution with solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Gradient - Time flow ml/min

0.00 1.2

3.60 1.2

4.00 1.2

4.70 1.2

4.75 1.2

Detection - UV (220 and 254 nm) and ELSD

Method C

Experiments were performed on a SHIMADZU LCMS-2020 with a CIS-reverse- phase column (50 x 3 mm Shim-Pack XR-ODS, 2.2 μιη particle size), elution with solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Gradient - Time flow ml/min %A %B

0.00 1.2 95 5

3.00 1.2 5 95

3.70 1.2 5 95

3.75 1.2 95 5

Detection - UV (220 and 254 nm) and ELSD Method D

Experiments were performed on a SHIMADZU LCMS-2020 with a C18-reverse- phase column (50 x 3 mm Shim-Pack XR-ODS, 2.2 μιη particle size), elution with solvent A: water + 0.05% trifluoroacetic acid; solvent B acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Gradient - Time flow ml/min %A %B

0.00 1.2 95 5

3.50 1.2 30 70

3.70 1.2 0 100

4.50 1.2 0 100

4.75 1.2 95 5

Detection - UV (220 and 254 nm) and ELSD

Method E

Experiments were performed on a SHIMADZU LCMS-2020 with a CIS-reverse- phase column (50 x 3 mm Shim-Pack XR-ODS, 2.2 μιη particle size), elution with solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Gradient - Time flow ml/min %A %B

0.00 1.2 95 5

3.50 1.2 40 60

3.70 1.2 0 100

4.70 1.2 0 100

4.75 1.2 95 5 Detection - UV (220 and 254 nm) and ELSD Method F

Experiments were performed on a SHIMADZU LCMS-2020 with a CIS-reverse- phase column (50 x 3 mm Shim-Pack XR-ODS, 2.2 μιη particle size), elution with solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Gradient - Time flow ml/min

0.00 1.2

3.50 1.2

3.70 1.2

4.50 1.2

4.75 1.2

Detection - UV (220 and 254 nm) and ELSD

Method G

Experiments were performed on a SHIMADZU 20A HPLC with a C 18-reverse-phase column (50 x 2.1 mm Ascentis Express CI 8, 2.7 μιη particle size), elution with solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Gradient - Time flow ml/min %A %B

0.00 1.0 95 5

1.10 1.0 0 100

1.60 1.0 0 100

1.70 1.0 95 5

Detection - UV (220 and 254 nm) and ELSD Method H

Experiments were performed on a SHIMADZU LCMS-2020 with a CIS-reverse- phase column (50 x 3 mm Shim-Pack XR-ODS, 2.2 μιη particle size), elution with solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Gradient - Time flow ml/min %A %B 0.00 1.2 95 5

1.10 1.2 0 100

1.70 1.2 0 100

1.75 1.2 95 5

Detection - UV (220 and 254 nm) and ELSD Method I

Experiments were performed on a SHIMADZU 20A HPLC with Poroshell HPH-Cig, column (50 x 3 mm, 2.7 μιη particle size), elution with solvent A: water/5 mM NH 4 HCO 3 ; solvent B: acetonitrile. Gradient:

Gradient - Time flow ml/min %A %B

0.00 1.2 90 10

1.10 1.2 5 95

1.60 1.2 5 95

1.70 1.2 90 10

Detection - UV (220 and 254 nm) and ELSD

Method J

Experiments were performed on a SHIMADZU LCMS-2020 with a CIS-reverse- phase column (50 x 3 mm Kinetex XB-Ci 8 , 2.6 μιη particle size), elution with solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05% trifluoroacetic acid. Gradient:

Gradient - Time flow ml/min

0.00 1.5

1.20 1.5

1.70 1.5

1.80 1.5

Detection - UV (220 and 254 nm) and ELSD Method K

Experiments were performed on a SHIMADZU LCMS-2020 with a CIS-reverse- phase column (50 x 3 mm Shim-Pack XR-ODS, 2.2 μιη particle size), elution with solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05%

5 trifluoroacetic acid. Gradient:

Gradient - Time flow ml/min %A %B

0.00 1.0 95 5

2.20 1.0 0 100

3.20 1.0 0 100

3.30 1.0 95 5

Detection - UV (220 and 254 nm) and ELSD

Method L

Experiments were performed on a SHIMADZU LCMS-2020 with a CIS-reverse- phase column (50 x 2.1 mm Kinetex XB-Cis 100A, 2.6 μιη particle size), elution with 0 solvent A: water + 0.05% trifluoroacetic acid; solvent B: acetonitrile + 0.05%

trifluoroacetic acid. Gradient:

Gradient - Time flow ml/min

0.00 1.0

1.10 1.0

1.60 1.0

1.70 1.0

Detection - UV (220 and 254 nm) and ELSD

List of Common Abbreviations 5 ACN Acetonitrile

Brine Saturated aqueous sodium chloride solution CH 3 OD Deuterated Methanol CDCI3 Deuterated Chloroform

DCM Dichloromethane

DIEA or DIPEA Diisopropylethylamine

DMA Dimethylacetamide

DMAP 4-Dimethylaminopyridine

DMF Dimethylformamide

DMSO Dimethylsulfoxide

OMSO-d6 Deuterated dimethylsulfoxide

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

EtOAc Ethyl acetate

EtOH Ethanol

FA Formic Acid

HOAc Acetic acid

g Gram

h hour

HATU (0-(7-azabenzotriazol- 1 -yl)-N,N,N ' ,N ' - tetramethyluronium hexafluorophosphate)

HC1 Hydrochloric acid

HOBt Hydroxybenzotriazole

HPLC High performance liquid chromatography

IMS Industrial methylated spirits

L Liter

LCMS Liquid chromatography-mass spectrometry

LiHMDS or LHMDS Lithium hexamethydisylazide

MDAP Mass directed automated purification

MeCN Acetonitrile

MeOH Methanol

min minute

mg Milligram

mL Millilitre

NMR Nuclear magnetic resonance spectroscopy

Pd 2 (dba) 3 .CHCl 3 Tris(dibenzylideneacetone)dipalladium(0)- chloroform adduct

PE Petroleum ether Prep-HP LC Preparative high perfomance liquid

chromatography

SCX-2 Strong cation exchange

TBAF Tetra-n-butylammonium fluoride

THF Tetrahydrofuran

TFA Trifluoroacetic acid

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

Intermediate 1

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

Synthesis of 4-bromo-l-(difluoromethoxy)-2-iodobenzene

To a solution of 4-bromo-2-iodophenol (282 g, 943 mmol) in N,N- dimethylformamide (2000 mL) and water (500 mL) was added sodium 2-chloro-2,2- difluoroacetate (216 g, 1.42 mol) and CS 2 CO 3 (617 g, 1.89 mol). The reaction vessel was equipped with a gas outlet for C0 2 release. The resulting mixture was stirred overnight at 120°C, allowed to cool to room temperature and poured into ice water (3000 mL). The resulting solution was extracted with ethyl acetate (3x1500 mL) and the organic layers were combined. The ethyl acetate extracts were washed with brine (1000 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with ethyl acetate/petroleum ether (1/10) to afford 300 g (91%) of 4-bromo-l- (dif uoromethoxy)-2-iodobenzene as a yellow oil. 1H NMR (300 MHz, CDC1 3 ) δ 7.96 (dd, J= 5.7 Hz, 2.4 Hz, 1H), 7.45 (dd, J = 8.7 Hz, 2.4 Hz, 1H), 7.03 (d, J= 8.7 Hz, 1H ), 6.39 (t, J = 72.9 Hz, 1H).

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

r

To a solution of 4-nitro-l-[[2-(trimethylsilyl)ethoxy]methyl]-lH-pyrazole (100 g, 411 mmol) in anhydrous THF (1000 mL) was added dropwise to a solution of LiHMDS (490 mL, 1.0 mol/L in THF) with stirring at -70°C under nitrogen. The resulting solution was stirred for 1 h at -50°C and then cooled to -70°C. ZnCl 2 (500 mL, 0.7 mol/L in THF) was added dropwise at -70°C. The resulting solution was allowed to warm to room temperature and stirred at room temperature for 1 h. To the mixture was added 4-bromo-l-(difluoromethoxy)-2-iodobenzene (150 g, 860 mmol), Pd(PPh 3 ) 4 (24.0 g, 20.8 mmol). The resulting solution was heated at reflux temperature overnight, allowed to cool to room temperature, and concentrated under reduced pressure. This reaction at this scale was repeated one more time, and the crude products from the two runs were combined for purification. The residue was purified by flash chromatography on silica gel eluting with ethyl acetate/petroleum ether (1/20). The appropriate fractions were combined and concentrated under reduced pressure. This resulted in 300 g (79%) of 5-[5-bromo-2- (difluoromethoxy)phenyl] -4-nitro- 1 -[ [2-(trimethylsilyl)ethoxy]methyl] - 1 H-pyrazole as a light yellow solid in all. 1H NMR (300 MHz, CDC1 3 ) δ 8.27 (s, 1H), 7.68 (dd, J = 8.7, 2.4 Hz, 1H), 7.62 (d, J= 2.4 Hz, 1H), 7.19 (d, J= 8.4 Hz, 1H), 6.39 (t, J= 72.5 Hz, 1H), 5.44 - 5.19 (m, 2H), 3.72 - 3.54 (m, 2H), 0.94 - 0.89 (m, 2H), 0.02 (s, 9H).

Step 3: Synthesis of 5-(5-bromo-2-(difluoromethoxy)phi

(trimethylsilyl)ethoxy)methyl)- 1 H-pyrazol-4-amine

r

To a solution of 5-(5-bromo-2-(difluoromethoxy)phenyl)-4-nitro-l-((2- (trimethylsilyl)ethoxy)methyl)-lH-pyrazole (50.1 g, 108 mmol) in ethanol (2000 mL) and water (200 mL) was added iron powder (60.1 g, 1.07 mol) and NH 4 C1 (28.0 g, 0.523 mol). The reaction mixture was stirred at reflux temperature for 3 h under nitrogen. The solids were filtered out, and washed with ethanol (100 mL). The filtrate was concentrated under reduced pressure. The residue was dissolved in 3000 mL of ethyl acetate. The ethyl acetate solution was washed with 1x500 mL of brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure to give 50.1 g of crude 5-(5-bromo-2-(difluoromethoxy)phenyl)-l-((2- (trimethylsilyl)ethoxy)methyl)-lH-pyrazol-4-amine as a yellow oil. The crude product was used for next step without further purification. LC/MS (Method G, ESI): [M+H] + = 434.2, R T = 0.93 min.

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

To a solution of 5-(5-bromo-2-(difluoromethoxy)phenyl)-l-((2- (trimethylsilyl)ethoxy)methyl)-lH-pyrazol-4-amine (50.1 g, 115 mmol) in DMA (1500 mL) was added pyrazolo[ 1,5 -a]pyrimidine-3-carboxylic acid (32.1 g, 196.0 mmol), PyAOP (102 g, 196 mmol), DMAP (1.41 g, 11.0 mmol) and DIPEA (44.1 g, 0.341 mol). The resulting solution was stirred for 3h at 60°C in an oil bath, and then allowed to cool to room temperature. The reaction mixture was then partitioned between water/ice (2000 mL) and ethyl acetate (2000 mL). The aqueous phase was extracted with ethyl acetate (2x). The organic layers were combined, washed with brine (1000 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with ethyl acetate/petroleum ether (4: 1). The appropriate fractions were combined and concentrated under reduced pressure. Water (150 mL) was added to the residue and the mixture was stirred in water for 1 h at room temperature. The solid was collected by filtration and air-dried to afford 60.1 g (91%) of N-(5-(5-bromo-2- (difluoromethoxy)phenyl)- 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 H-pyrazol-4- yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide as a light yellow solid. LC/MS (Method G, ESI): [M+H] + = 579.1 & 581.1, R T = 1.10 min. ! Η ΝΜΚ (300 MHz, CDC1 3 ) δ 9.62 (s, 1H), 8.80 (dd, J = 6.9, 1.7 Hz, 1H), 8.73 (s, 1H), 8.53 (dd, J= 4.2, 1.7 Hz, 1H), 8.38 (s, 1H), 7.79 (d, J= 2.4 Hz, 1H), 7.67 (dd, J = 8.8, 2.5 Hz, 1H), 7.29 (d, J= 1.4 Hz, 1H), 7.00 (dd, J= 6.9, 4.2 Hz, 1H), 6.43 (t, J= 72.6 Hz, 1H), 5.53 - 5.27 (m, 2H), 3.73 - 3.50 (m, 2H), 0.88 (ddd, J= 9.5, 6.4, 4.4 Hz, 2H), 0.00 (s, 9H).

Intermediate 2

N-[3-[5-bromo-2-(difluoromethoxy)phenyl]-lH-pyrazol-4-yl]pyr azolo[l,5- a]pyrimidine-3 -carboxamide N- [5 -[5 -bromo-2-(difluoromethoxy)phenyl] - 1 -[ [2-

(trimethylsilyl)ethoxy]methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (Intermediate 1, 5.00 g, 8.63 mmol) was treated with HCl/dioxane (150 mL, 4 M) overnight at room temperature. The resulting mixture was concentrated under reduced pressure. This resulted in 3.80 g of N-[3-[5-bromo-2- (difluoromethoxy)phenyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 -carboxamide as a yellow solid. The purity of the intermediate was sufficient for use in the next step without further purification. LC/MS (Method I, ESI): [M+H] + = 449.0, R T = 1.02 min. 1H NMR (400 MHz, CD 3 OD) δ 9.11 (dd, J = 6.8, 1.6 Hz, 1H), 8.67 - 8.64 (m, 2H), 8.32 (s, 1H), 7.80 (d, J = 2.4 Hz, 1H), 7.72 (dd, J = 8.8, 2.4 Hz, 1H), 7.37 (d, J = 8.8 Hz, 1H), 7.23 (dd, J= 7.0, 4.2 Hz, 1H), 6.81 (t, J= 73.2 Hz, 1H).

Intermediate 3

N-[3-[2-(difluoromethoxy)-5-iodophenyl]-lH-pyrazol-4-yl]pyra zolo[l,5- a]pyrimidine-3 -carboxamide

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

To a solution of N-[5-[5-bromo-2-(difluoromethoxy)phenyl]-l-[[2- (trimethylsilyl)ethoxy]methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (100 mg, 0.173 mmol) in t-BuOH (2 mL) was added N,N- dimethylethane-l,2-diamine (2.28 mg, 0.0259 mmol), Nal (155 mg, 1.04 mmol), Cul (4.93 mg, 0.026 mmol) under nitrogen. The resulting solution was stirred for 14 h at 120 °C in an oil bath under nitrogen before being cooled to room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with ethyl acetate/petroleum ether (1/1) to afford 80 mg (74%) of N-[5-[2-(difluoromethoxy)-5-iodophenyl]-l-[[2- (trimethylsilyl)ethoxy]methyl]-lH-pyrazol-4-yl]pyrazolo[l,5- a]pyrimidine-3- carboxamide as a yellow solid. LC/MS (Method J, ESI): [M+H] + = 627.1, R T = 1.31 mm.

Step 2: Synthesis of N-[3-[2-(difluoromethoxy)-5-iodophenyl]-lH-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide

N-[5-[2-(difluoromethoxy)-5-iodophenyl]-l-[[2-(trimethyls ilyl)ethoxy]methyl]- lH-pyrazol-4-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide (80.0 mg, 0.128 mmol) was treated with CF 3 CO 2 H (3.0 mL) for 30 min at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was dissolved in water. Saturated sodium bicarbonate was slowly added until the solution was adjusted pH ~ 8. The solid was collected by filtration. The solid was purified by flash chromatography on silica gel eluting with ethyl acetate/petroleum ether (2/1) to give 23.0 mg (36%) of N-[3-[2-(difluoromethoxy)-5-iodophenyl]-lH-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a light yellow solid. LC/MS (Method K, ESI): [M+H] + = 497.1, R T = 1.74 min. 1H NMR (300 MHz, CD 3 OD) δ 9.08 (dd, J = 6.9, 1.5 Hz, 1H), 8.65 - 8.61 (m, 2H), 8.27 (s, 1H), 7.94 (s, 1H), 7.87 (d, J= 8.7 Hz, 1H), 7.21 - 7.18 (m, 2H), 6.78 (t, J= 73.2 Hz, 1H).

Intermediate 4

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

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

To a solution of N-[5-[5-bromo-2-(difluoromethoxy)phenyl]-l-[[2- (trimethylsilyl)ethoxy]methyl]-lH-pyrazol-4-yl]pyrazolo[l,5- a]pyrimidine-3- carboxamide (Intermediate 1, 1.00 g, 1.73 mmol) in dioxane (15 mL) and water (3.0 mL) was added potassium (2-cyanocyclopropyl)trifluoroborate (449 mg, 2.60 mmol), Pd(dppf)Cl 2 .CH 2 Cl 2 (141 mg, 0.173 mmol, 0.10 equiv) and Cs 2 C0 3 (1.13 g, 3.47 mmol, 2.01 equiv) under nitrogen. The resulting solution was stirred overnight at 80 °C in an oil bath. The resulting mixture was concentrated under reduced pressure. The reaction at this scale was repeated five times, and the crude products from 5 operations were combined together for purification. The residue was passed through a short pad of silica gel eluting with ethyl acetate/petroleum ether (6/4). The appropriate fractions were combined and concentrated under reduced pressure to afford 2.5 g (purity = 85% by LCMS at 254nm) of N-[5-[5-(2-cyanocyclopropyl)-2- (difluoromethoxy)phenyl] - 1 - [ [2-(trimethylsilyl)ethoxy]methyl] - 1 H-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a yellow solid. The quality of the intermediate was sufficient for next step. No further purification was required. LC/MS (Method G, ESI): [M+H] + = 566.2, R T = 1.07 min.

Step 2: N-(3-[5-[(lR,2R)-2-cyanocyclopropyl]-2-(difluoromethoxy)phen yl]-lH- pyrazol-4-yl)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide

N-[5-[5-(2-cyanocyclopropyl)-2-(difluoromethoxy)phenyl]-l -[[2- (trimethylsilyl)ethoxy]methyl]-lH-pyrazol-4-yl]pyrazolo[l,5- a]pyrimidine-3- carboxamide (2.90 g, 5.13 mmol) was treated with trifluoroacetic acid (10 mL) in dichloromethane (20 mL) overnight at room temperature. The resulting mixture was concentrated under vacuum. The pH of the residue was adjusted to >7 with DIEA. The resulting mixture was concentrated under vacuum. Water was added and the mixture was stirred for 1 h. The solids were collected by filtration. The crude product (2.30 g) was purified by Prep-HP LC with the following conditions: Column, XBridge Shield RP18 OBD Column, 19* 150mm 5um 13nm; mobile phase, Water with 0.05% NH 4 OH and MeCN (30.0% MeCN up to 45.0% in 9 min); Detector, UV 254nm. The racemic product was separated by Prep-SFC with the following conditions: Column: CHIRALPAK-IC-SFC-02, 5cm*25cm(5um); Mobile Phase A:C0 2 :50, Mobile Phase B: EtOH :50; Flow rate: 180 mL/min; 220 nm ; R T1 = 13.97 min (the first peak); R-n = 17.84 min (the second peak) to afford two fractions:

Fraction 1 (R,R-isomer): The first peak was the desired fraction and further purified by re-crystallization from isopropanol. This resulted in 340 mg (15%) of N- (3-[5-[(lR,2R)-2-cyanocyclopropyl]-2-(difluoromethoxy)phenyl ]-lH-pyrazol-4- yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide as an off-white solid. 1H NMR (300 MHz, CD 3 OD) δ 9.09 (dd, J = 7.1, 1.7 Hz, 1H), 8.67 - 8.62 (m, 2H), 8.27 (s, 1H), 7.43 - 7.35 (m, 3H), 7.21 (dd, J = 6.9, 4.2 Hz, 1H), 6.76 (t, J = 73.8 Hz, 1H), 2.79 - 2.72 (m, 1H), 1.94 - 1.88 (m, 1H), 1.68 - 1.63 (m, 1H), 1.62 - 1.56 (m, 1H).

Fraction 2 (S,S-isomer): The second peak was got 474 mg (21%) of N-(3-[5- [(lS,2S)-2-cyanocyclopropyl]-2-(difluoromethoxy)phenyl]-lH-p yrazol-4- yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide as a white solid. 1H NMR (300 MHz, CD 3 OD) δ 9.09 (dd, J = 7.1, 1.7 Hz, 1H), 8.67 - 8.62 (m, 2H), 8.27 (s, 1H), 7.43 - 7.35 (m, 3H), 7.21 (dd, J = 6.9, 4.2 Hz, 1H), 6.76 (t, J= 73.8 Hz, 1H), 2.79 - 2.72 (m, 1H), 1.94 - 1.88 (m, 1H), 1.68 - 1.63 (m, 1H), 1.62 - 1.56 (m, 1H).

Intermediate 5

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

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

To a solution of N-(5-(5-bromo-2-(difluoromethoxy)phen5/ (trimethylsilyl)ethoxy)methyl)- 1 H-pyrazol-4-yl)pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (Intermediate 1, 1.40 g, 2.41 mmol) in dioxane (15 mL) and water (3.0mL) was added cyclopropylboronic acid (314 mg, 3.66 mmol), Pd(dppf)Cl 2 - CH 2 C1 2 (200 mg, 0.245 mmol) and Cs 2 C0 3 (1.56 g, 4.79 mmol) under nitrogen. The reaction mixture was stirred overnight at 80 °C under nitrogen. The resulting mixture was concentrated under reduced pressure. The residue was passed through a short pad of silica gel eluting with dichloromethane/methanol (94/6). The appropriate fractions were combined and concentrated under reduced pressure to give 1.40 g (purity = -85% at 254 nm) of N-[3-[5-cyclopropyl-2-(difluoromethoxy)phenyl]-l-[[2- (trimethylsilyl)ethoxy]methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide as a dark red solid. LC/MS (Method G, ESI): [M+H] + = 541.2, R T = 1.12 min. The intermediate was used without further purification.

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

N-[3-[5-cyclopropyl-2-(difluoromethoxy)phenyl]-l-[[2- (trimethylsilyl)ethoxy]methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (145 mg) from previous step was treated with HCl/dioxane (5.0 mL, 4 M) for 2 h at 25 °C. The solution was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions: Column: Xbridge CI 8, 19* 150 mm, 5 urn; Mobile Phase A: Water/0.05% NH 4 HC0 3 , Mobile Phase B: ACN; Flow rate: 30 mL/min; Gradient: 20%B to 85%B in 10 min; 254nm to obtain 44.9 mg (41%) of N-[3-[5-cyclopropyl-2-(difluoromethoxy)phenyl]-lH-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a yellow solid. LC/MS (Method H, ESI): [M+H] + = 411.2, R T = 1.14 min. 1 H NMR (300 MHz, CD 3 OD) δ 9.09 (dd, J = 6.9, 1.5 Hz, 1H), 8.63 - 8.61 (m, 2H), 8.27 (s, 1H), 7.28 - 7.25 (m, 3H), 7.20 (dd, J = 7.2, 4.2 Hz, 1H), 6.68 (t, J = 73.8 Hz, 1H), 2.04 - 1.95 (m, 1H), 1.03 - 0.97 (m, 2H), 0.79 - 0.71 (m, 2H)

Intermediate 6

N-[3-[2-(dinuoromethoxy)-5-[4-(dimethylcarbamoyl)phenoxy]phe nyl]-lH-pyrazol-4- yl]pyrazolo[ 1 ,5 -a]pyrimidine-3 -carboxamide

Step 1 : Synthesis of N-[5-[2-(difluoromethoxy)-5-[4-

(dimethylcarbamoyl)phenoxy]phenyl] - 1 -[ [2-(trimethylsilyl)ethoxy]methyl] - 1 H- pyrazol-4-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide

To a solution of N-5-[5-bromo-2-(difluoromethoxy)phenyl]-l-[2- (trimethylsilyl)ethoxy]methyl- 1 H-pyrazol-4-ylpyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (Intermediate 1, 1.17 g, 2.02 mmol) in toluene (10 mL) was added 4- hydroxy-N,N-dimethylbenzamide (0.400 g, 2.42 mmol), Cs 2 C0 3 (0.790 g, 2.43 mmol), [PdCl(allyl)] 2 (37.0 mg, 0.101 mmol) and t-BuBrettPhos (98.0 mg, 0.202 mmol) under nitrogen. The reaction mixture was stirred overnight at 90 °C. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with dichloromethane/methanol (95/5) to give 1.3 g (81%) of N-[5-[2-(difluoromethoxy)-5-[4- (dimethylcarbamoyl)phenoxy]phenyl] - 1 -[ [2-(trimethylsilyl)ethoxy]methyl] - 1 H- pyrazol-4-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a yellow oil. LC/MS (Method H, ESI): [M+H] + = 664.4, R T = 1.36 min.

Step 2: Synthesis of N-[3-[2-(difiuoromethoxy)-5-[4-

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

To a solution of N-[5-[2-(difluoromethoxy)-5-[4- (dimethylcarbamoyl)phenoxy]phenyl] - 1 -[ [2-(trimethylsilyl)ethoxy]methyl] - 1 H- pyrazol-4-yl]pyrazolo[l, 5 -a]pyrimidine-3 -carboxamide (139 mg, 0.209 mmol) in MeOH (3.0 mL) was added concentrated hydrochloric acid (1.5 mL, 12M). The reaction mixture was stirred for 2 h at 25 °C and concentrated under reduced pressure The residue was neutralized with DIPEA. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with dichloromethane/methanol (95/5) to obtain 28 mg (25%) of N-[3-[2- (difluoromethoxy)-5 -[4-(dimethylcarbamoyl)phenoxy]phenyl] - 1 H-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as an off-white solid. LC/MS (Method H, ESI): [M+H] + = 534.2, R T = 1.09 min. 1H NMR (300 MHz, DMSO^) δ 13.05 (s, 1H), 9.73 (s, 1H), 9.35 (dd, J= 7.1, 1.7 Hz, 1H), 8.70 (dd, J= 4.2, 1.5 Hz, 1H), 8.69 (s, 1H), 8.22 (s, 1H), 7.47 (d, J= 9.3 Hz, 1H), 7.39 (d, J= 8.4 Hz, 2H), 7.32 - 7.25 (m, 3H), 7.06 (d, J = 8.7 Hz, 2H), 7.18 (t, J= 73.8 Hz, 1H), 2.94 (s, 6H)

Intermediate 7

N-[3-[2,5-bis(difluoromethoxy)phenyl]-lH-pyrazol-4-yl]pyrazo lo[l,5-a]pyrimidine

3-carboxamide

Step 1 : Synthesis of l-(benzyloxy)-4-(difluoromethoxy)benzene

Into a 3000-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed N,N-dimethylformamide (1500 mL), 4-(benzyloxy)phenol (200 g, 999 mmol) and CS 2 CO 3 (651 g, 1.99 mol). The reaction vessel was equipped with an outlet for C0 2 release. This was followed by the addition of sodium 2-chloro- 2,2-difluoroacetate (228 g, 1.50 mol, 1.50 equiv) in several batches at 120 °C. After completion of the sodium 2-chloro-2,2-difluoroacetate addition, the reaction was stirred at 120 °C in an oil bath until gas evolution ceased (~1 h), and then allowed to cool to room temperature. The reaction mixture was slowly added to 3000 mL of water/ice with stirring. The resulting mixture was extracted with ethyl acetate (3x4000 mL). The organic layers were combined and dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with ethyl acetate/petroleum ether (1/19). The appropriate fractions were combined and concentrated under reduced pressure. This reaction was repeated four times. This resulted in 450 g (36%) of l-(benzyloxy)-4- (difluoromethoxy)benzene as a white solid in total.

Step 2: Synthesis of 4-(difluoromethoxy)phenol

Into a 3000-mL round-bottom flask was placed methanol (1500 mL), l-(benzyloxy)- 4-(difluoromethoxy)benzene (140 g, 559 mmol) and 10% Palladium carbon (15 g, 141 mmol). The resulting mixture was stirred under hydrogen (~45 psi) overnight at room temperature. The catalysts were filtered out. The filtrate was concentrated under reduced pressure. This reaction was repeated three times. This resulted in 300 g (78%) of 4-(difluoromethoxy)phenol as a yellow oil.

Step 3: Synthesis of 2-bromo-4-(difluoromethoxy)phenol

Into a 1000-mL round-bottom flask was placed acetic acid (500 mL), 4- (difluoromethoxy)phenol (50 g, 312 mmol) and NBS (55.6 g, 312 mmol). The reaction mixture was stirred for 1 h at 15 °C. The resulting mixture was then added slowly to 1000 mL of water/ice with stirring. The resulting solution was extracted with ethyl acetate (3x1000 mL). The organic layers were combined, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with dichloromethane/petroleum ether (30/70). The appropriate fractions were collected and concentrated under reduced pressure. This resulted in 50 g (67%) of 2-bromo-4- (difluoromethoxy)phenol as a light yellow oil.

Step 4: Synthesis of 2-bromo-l,4-bis(difluoromethoxy)benzene

Into a 2000-mL round-bottom flask, was placed CH 3 CN (500 mL), water (500 mL), 2-bromo-4-(difluoromethoxy)phenol (54 g, 226 mmol) and potassium hydroxide (94g, 1.68 mol). The flask was placed in an ice batch and the reaction mixture was stirred for 30 min in an ice batch. Diethyl (bromodifluoromethyl)phosphonate (120 g, 449 mmol) was then added dropwise to the reaction mixture at 0 °C. Upon completion of diethyl (bromodifiuoromethyl)phosphonate addition, the reaction mixture was stirred for 1 h in a water/ice bath. The resulting solution was extracted with ethyl acetate (3x300 mL). The organic layers were combined and dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with ethyl acetate/petroleum ether (1/19), and the appropriate fractions were collected and concentrated under reduced pressure. This resulted in 54 g (83%) of 2-bromo-l,4-bis(difiuoromethoxy)benzene as light yellow oil.

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

Into a 1000-mL round-bottom flask purged and maintained with an inert atmosphere of nitrogen, was placed DMA (500 mL), potassium carbonate (112 g, 810 mmol), 4- nitro-l-[[2-(trimethylsilyl)ethoxy]methyl]-lH-pyrazole (66 g, 271 mmol), 2-bromo- 1 ,4-bis(difiuoromethoxy)benzene (79 g, 273 mmol), 2,2-dimethylpropanoic acid (8.3 g, 81.3mmol), Pd(OAc) 2 (6.0 g, 26.7 mmol) and bis(adamantan-l- yl)(butyl)phosphane (19 g, 52.9 mmol, 0.195 equiv). The reaction mixture was stirred at 120 °C overnight in an oil bath, and allowed to cool to room temperature. The reaction mixture was then added to 1000 mL of water/ice with stirring. The resulting solution was extracted with ethyl acetate (3x1000 mL). The organic layers were combined and dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with ethyl acetate/petroleum ether (1/1). The appropriate fractions were collected and concentrated under reduced pressure. This resulted in 100 g (82%) of 5-[2,5- bis(difluoromethoxy)phenyl] -4-nitro- 1 -[ [2-(trimethylsilyl)ethoxy]methyl] - 1 H- pyrazole as a solid.

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

Into a 3000-mL 3-necked round-bottom flask was placed ethanol (1500 mL), water (150 mL), 5-[2,5-bis(difluoromethoxy)phenyl]-4-nitro-l-[[2-

(trimethylsilyl)ethoxy]methyl]-lH-pyrazole (100.00 g, 221 mmol), iron powder (124 g, 2.22 mol) and NH 4 C1 (59.2 g, 1.11 mol). The resulting mixture was stirred at reflux temperature in an oil bath for 2 h before being cooled to room temperature. The solids were filtered off, and washed with ethanol. The filtrate was concentrated under reduced pressure. The residue was dissolved in 3000 mL of ethyl acetate. The ethyl acetate solution was washed with 1x1000 mL of brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. This resulted in 100 g of crude 5- [2,5-bis(difluoromethoxy)phenyl]-l-[[2-(trimethylsilyl)ethox y]methyl]-lH-pyrazol-4- amine as light yellow oil, which was used directly without purification.

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

Into a 2000-mL round-bottom flask was placed DMA (1000 mL), 5-[2,5- bis(difluoromethoxy)phenyl]-l-[[2-(trimethylsilyl)ethoxy]met hyl]-lH-pyrazol-4- amine from previous step, pyrazolo[l,5-a]pyrimidine-3-carboxylic acid (58.06 g, 355.9 mmol), 7-Azabenzotriazol-l-yloxy)tripyrrolidinophosphonium hexafluorophosphate (PyAOP) (185.56 g, 355.9 mmol), 4-dimethylaminopyridine (2.90 g, 23.7 mmol) and DIPEA (92.0 g, 712 mmol). The resulting solution was stirred overnight at 65 °C in an oil bath. The reaction mixture was then added slowly to 2000 mL of water with stirring. The resulting solution was extracted with ethyl acetate (3x2000 mL). The combined organic phases were washed with 1000 mL of brine, dried over anhydrous sodium sulfate and concentrated in under pressure. The residue was purified by flash chromatography on silica gel eluting with ethyl acetate/petroleum ether (40/60). The appropriate fractions were combined and concentrated under reduced pressure to afford 120 g of N-[5-[2,5- bis(difluoromethoxy)phenyl]-l-[[2-(trimethylsilyl)ethoxy]met hyl]-lH-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a white solid.

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

Into a 2000-mL round-bottom flask was placed methanol (800 mL), concentrated hydrochloric acid (400 mL, 12N) and N-[5-[2,5-bis(difluoromethoxy)phenyl]-l-[[2- (trimethylsilyl)ethoxy]methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (80 g, 141mmol). The resulting solution was stirred for 4 h at 25 °C. The solids were collected by filtration. The solids were added to a 1 L flask and H 2 0 (200 mL) was added. A saturated NaHC0 3 aqueous solution was added dropwise with stirring until the solution reached pH~8. The solids were collected by filtration, washed with water and dried to afford 55 g (89%) of N-[3-[2,5- bis(difluoromethoxy)phenyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide as a light yellow solid. 1H NMR (300 MHz, CD 3 OD) δ 9.08 (dd, J= 7.2, 1.5 Hz, 1H), 8.65 - 8.61 (m, 2H), 8.28 (s, 1H), 7.46 (d, J= 9.0 Hz, 1H), 7.40 (d, J = 3.0 Hz, 1H), 7.34 (dd, J= 8.9, 2.9 Hz, 1H), 7.19 (dd, J= 6.7, 4.4 Hz, 1H), 6.87 (t, J = 73.7 Hz, 1H), 6.73 (t, J= 73.7 Hz, 1H).

N-(3-(5-((lR,2R)-2-cyanocyclopropyl)-2-(difluoromethoxy)p henyl)-l-(2- (dimethylamino)-2-oxoethyl)-lH-pyrazol-4-yl)pyrazolo[l,5-a]p yrimidine-3- carboxamide

To a solution of N-(3-(5-((lR,2R)-2-cyanocyclopropyl)-2-(difluoromethoxy)phen yl)- lH-pyrazol-4-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (Intermediate 4, 100 mg, 0.230 mmol) in DMF (10 mL) was added Cs 2 C0 3 (160 mg, 0.491 mmol). This was followed by the addition of 2-bromo-N,N-dimethylacetamide (80 mg, 0.482 mmol). The resulting mixture was stirred for 30 min at 60 °C in an oil bath. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with dichloromethane/methanol (8% MeOH) to give 20.9 mg (17%) of N-(3-(5-((lR,2R)-2-cyanocyclopropyl)-2- (difluoromethoxy)phenyl)- 1 -(2-(dimethylamino)-2-oxoethyl)- 1 H-pyrazol-4- yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide as a white solid. LC/MS (Method A, ESI): [M+H] + = 521.3, R T = 1.50 min; 1H NMR (400 MHz, DMSO-^) δ 9.76 (s, 1H), 9.35 (dd, J= 6.8, 1.6 Hz, 1H), 8.68 (s, 1H), 8.65 (dd, J= 4.0, 1.6 Hz, 1H), 8.28 (s, 1H), 7.42 - 7.38 (m, 3H), 7.29 (dd, J= 7.0, 4.2 Hz, 1H), 7.23 (t, J= 73.2 Hz, 1H), 5.20 (s, 2H), 3.06 (s, 3H), 2.89 (s, 3H), 2.84 - 2.80 (m, 1H), 2.12 - 2.09 (m, 1H), 1.66 - 1.61 (m, 1 H), 1.54 - 1.50 (m, 1H).

Example 2

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

To a solution of N-[3-[2-(difluoromethoxy)-5-iodophenyl]-lH-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide (Intermediate 3, 160 mg, 0.322 mmol) in DMF (4.0 mL) was added CS 2 CO 3 (210 mg, 0.645 mmol). To this mixture was added 2-bromo-N,N-dimethylacetamide (107 mg, 0.645 mmol). The resulting solution was stirred for 4 h at 60 °C in an oil bath. The reaction mixture was partitioned between water and ethyl acetate. The aqueous phase was extracted ethyl acetate (2x). The organic layers were combined, washed with water and brine successively, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was passed through a short pad of silica gel eluting with dichloromethane/methanol (90/10). The appropriate fractions were combined and concentrated under reduced pressure. The crude product was further purified by Prep-HP LC with the following conditions: Column, SunFire Prep CI 8 OBD Column, 19* 150mm 5um lOnm; mobile phase, Water (0.1% FA) and ACN (25.0% ACN up to 44.0% in 12 min); Detector, UV 254/220 nm to obtain 24.1 mg (13%) of N-[3-[2-(difluoromethoxy)-5- iodophenyl] - 1 - [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 - a]pyrimidine-3-carboxamide as a white solid. LC/MS (Method B, ESI): [M+H] + = 582.2, R T = 2.92; 1H NMR (400 MHz, CD 3 OD) 5 9.11 (dd, J= 6.8, 1.6 Hz, 1H), 8.67 (dd, J= 4.4, 1.6 Hz, 1H), 8.65 (s, 1H), 8.36 (s, 1H), 8.02 (d, J= 2.4 Hz, 1H), 7.90 (dd, J= 8.8, 2.4 Hz, 1H), 7.24 - 7.21 (m, 2H), 6.82 (t, J= 73.6 Hz, 1H), 5.24 (s, 2H), 3.18 (s, 3H), 3.03 (s, 3H).

Example 3

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

Step 1 : Synthesis of tert-butyl N-[3-([3-[5-chloro-2-(difluoromethoxy)phenyl]-l-

[(dimethylcarbamoyl)methyl]-lH-pyrazol-4-yl]carbamoyl)pyr azolo[l,5-a]pyrimidin- 2-yl] carbamate

To a solution of tert-butyl N-[3-([3-[5-chloro-2-(difluoromethoxy)phenyl]-lH- pyrazol-4-yl]carbamoyl)pyrazolo[l,5-a]pyrimidin-2-yl]carbama te (200 mg, 0.385 mmol) in DMF (5.0 mL) was added CS 2 CO 3 (251 mg, 0.770 mmol) and 2-bromo- N,N-dimethylacetamide (63.0 mg, 0.379 mmol). The reaction mixture was stirred for 16 h at 60 °C in an oil bath. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with dichloromethane/methanol (95/5). The appropriate fractions were combined and concentrated under reduced pressure to give 200 mg (86%) of tert-butyl N-[3-([3-[5- chloro-2-(difluoromethoxy)phenyl] - 1 - [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4- yl]carbamoyl)pyrazolo[l, 5 -a]pyrimidin-2-yl] carbamate as a yellow solid. LC/MS (Method H, ESI): [M+H] + = 605.3, R T = 1.32 min.

Step 2: Synthesis of 2-amino-N-[3-[5-chloro-2-(difluoromethoxy)phenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide

Tert-butyl N-[3-([3-[5-chloro-2-(difluoromethoxy)phenyl]-l-

[(dimethylcarbamoyl)methyl]-lH-pyrazol-4-yl]carbamoyl)pyr azolo[l,5-a]pyrimidin- 2-yl]carbamate (100 mg, 0.165 mmol) was treated with HC1 in dioxane (2.0 mL, 4M) for 2 h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HP LC with the following conditions: Column, XBridge Shield RP18 OBD Column, 5um, 19* 150mm; mobile phase, water (0.05%NH 3 H 2 O) and ACN (10% ACN up to 45% in 12 min); Detector, UV 220nm to give 27.4 mg (33%) of 2-amino-N-[3-[5-chloro-2- (difluoromethoxy)phenyl] - 1 - [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a white solid. LC/MS (Method A, ESI): [M+H] + = 505.2, R T = 1.58 min; 1H NMR (300 MHz, DMSO-^) δ 9.55 (s, 1H), 8.93 (dd, J= 6.8, 1.7 Hz, 1H), 8.36 (dd, J= 4.5, 1.5 Hz, 1H), 8.26 (s, 1H), 7.62 (dd, J = 8.7, 2.7 Hz, 1H), 7.54 (d, J= 2.7 Hz, 1H), 7.45 (J= 8.7 Hz, 1H), 7.26 (t, J= 73.2 Hz, 1H), 7.00 (dd, J= 6.6, 4.5 Hz, 1H), 6.57 (s, 2H), 5.18 (s, 2H), 3.04 (s, 3H), 2.87 (s, 3H). Examples 4 & 5

(R)-N-(3-(5-(l,2-difiuoroethyl)-2-(difluorom

oxoethyl)-lH-pyrazol-4-yl)pyrazolo[l,5-a]pyrimidine-3-car boxamide & (S)-N-(3-(5- (1 ,2-difluoroethyl)-2-(difluoromethoxy)phenyl)- 1 -(2-(dimethylamino)-2-oxoethyl)- lH-pyrazol-4-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide

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

To a solution of N-[3-[2-(difluoromethoxy)-5-ethenylphenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (1.70 g, 3.53 mmol) in tetrahydrofuran (20 mL) and water (10 mL) was added Os0 4 (1.82 g, 7.16 mmol) and NMO (831 mg, 7.09 mmol). The resulting solution was stirred for 2 h at room temperature. The resulting solution was diluted with 30 mL of H 2 0. The resulting solution was extracted with 3x50 mL of dichloromethane. The organic layers were combined, washed with brine, dried and concentrated under reduced pressure. The residue was purified by flash

chromatography on silica gel eluting with dichloromethane/methanol (9/1). The appropriate fractions were combined and concentrated under reduced pressure to give 700 mg (38%) of N-[3-[2-(difiuoromethoxy)-5-(l,2-dihydroxyethyl)phenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide as a brown solid.

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

To a solution of N-[3-[2-(difluoromethoxy)-5-(l,2-dihydroxyethyl)phenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (300 mg, 0.582 mmol) in dichloromethane was added DAST (374 mg, 2.32 mmol). The reaction mixture was stirred for 2 h at room temperature under nitrogen. The reaction was then quenched by the addition of 10 mL of water. The pH value of the solution was adjusted to 7 with a sodium bicarbonate aqueous solution (10 %). The resulting mixture was extracted with 3x10 mL of dichloromethane and the organic layers were combined and concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge BEH130 Prep C 18 OBD Column, 19* 150mm 5um; mobile phase, Water (0.05%NH 3 H 2 O) and ACN (30% ACN up to 34% in 7 min); Detector, UV 254/220nm to give 100 mg of racemic mixture. The racemic mixture was separated by Chiral-Prep-HPLC with the following conditions: Column, CHIRALPAK IA, 2.12* 15cm, 5um; mobile phase, Hexane/DCM = 5/1 and ethanol (hold 50.0% ethanol- in 13 min); flow rate 16 mL/min, Detector, UV 220/254 nm afford two fractions:

Isomer 1 : Eluted at 7.15 min, 12.4 mg (4%) of N-[3-[5-(l,2-difiuoroethyl)-2- (difluoromethoxy)phenyl] - 1 - [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a white solid. LC/MS (Method A, ESI): [M+H] + = 520.2, R T = 1.70 min; 1H NMR (400 MHz, DMSO-d 6 ) δ 9.72 (s, 1H), 9.34 (dd, J = 7.0, 1.4 Hz, 1H), 8.68 (s, 1H), 8.66 (dd, J = 4.0, 1.6 Hz, 1H), 8.29 (s, 1H), 7.65 - 7.61 (m, 2H), 7.51 (d, J = 8.0 Hz, 1H), 7.32 (t, J = 73.2 Hz, 1H), 7.29 (dd, J = 7.0, 4.2 Hz, 1H), 6.11 - 5.84 (m, 1H), 5.22 (s, 2H), 4.88 - 4.68 (m, 2H), 3.06 (s, 3H), 2.89 (s, 3H).

Isomer 2: Eluted at 9.66 min, 13.7 mg (5%) of N-[3-[5-(l,2-difluoroethyl)-2- (difluoromethoxy)phenyl] - 1 - [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a white solid. LC/MS (Method A, ESI): [M+H] + = 520.2, R T = 1.70 min; 1H NMR (400 MHz, DMSO-d 6 ) δ 9.72 (s, 1H), 9.34 (dd, J = 7.0, 1.4 Hz, 1H), 8.68 (s, 1H), 8.66 (dd, J = 4.0, 1.6 Hz, 1H), 8.29 (s, 1H), 7.65 - 7.61 (m, 2H), 7.51 (d, J = 8.0 Hz, 1H), 7.32 (t, J = 73.2 Hz, 1H), 7.29 (dd, J = 7.0, 4.2 Hz, 1H), 6.11 - 5.84 (m, 1H), 5.22 (s, 2H), 4.88 - 4.68 (m, 2H), 3.06 (s, 3H), 2.89 (s, 3H).

Example 6

N-(3-(2-(difluoromethoxy)-5-(difluoromethyl)phenyl)-l-(2-(di methylamino)-2- oxoethyl)-lH-pyrazol-4-yl)pyrazolo[l,5-a]pyrimidine-3-carbox amide

Step 1 : Synthesis of N-[3-[5-(l,l-difluoro-2-oxo-2-phenylethyl)-2- (difluoromethoxy)phenyl] - 1 - [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4- yl]pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide

To a solution of N-[3-[5-bromo-2-(difluoromethoxy)phenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (50.0 mg, 0.0936 mmol) in toluene (5.0 mL) was added 2,2-difluoro-l- phenylethan-l-one (29.0 mg, 0.186 mmol), [2-(2- aminophenyl)phenyl](chloro)palladium; tricyclohexylphosphane (12.0 mg, 0.0203 mmol) and K 3 PO 4 (80.0 mg, 0.377 mmol) under nitrogen. The resulting solution was stirred for 16 h at 100 °C in an oil bath. The reaction mixture was cooled to room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with

dichloromethane/methanol (92/8) to give 23 mg (40%) of N-[3-[5-(l,l-difiuoro-2- oxo-2-phenylethyl)-2-(difluoromethoxy)phenyl] - 1 - [(dimethylcarbamoyl)methyl] - 1 H- pyrazol-4-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a light yellow solid.

LC/MS (Method I, ESI): [M+H] + = 610.3, R T = 1.11 min.

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

To a solution of N-[3-[5-(l,l-difluoro-2-oxo-2-phenylethyl)-2- (difluoromethoxy)phenyl] - 1 - [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide (230 mg, 0.377 mmol) in toluene (30 mL) and water (5.0 mL) was added potassium hydroxide (43.0 mg, 0.766 mmol). The resulting solution was stirred for 16 h at 100 °C in an oil bath. The reaction mixture was cooled to room temperature. The resulting solution was diluted with 50 mL of H 2 0. The resulting solution was extracted with 2x30 mL of dichloromethane and the organic layers were combined and dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was passed through a short pad of silica gel eluting with dichloromethane/methanol (95/5). The appropriate fractions were combined and concentrated under reduced pressure. The residue was further purified by Prep-HPLC with the following conditions: Column, XBridge BEH130 Prep C18 OBD Column, 19* 150mm 5um; mobile phase, Water (0.05%NH 4 OH) and ACN (29.0% ACN up to 42.0% in 7 min); Detector, uv 254/220nm to obtain 23.7 mg ( 12%) of N-[3 - [2-(difluoromethoxy)-5 -(difluoromethyl)phenyl] - 1 - [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide as a white solid. LC/MS (Method A, ESI): [M+H] = 506.3, R T = 1.52 min. 1H NMR (400 MHz, DMSO^) δ 9.74 (s, 1H), 9.35 (dd, J= 6.8, 1.6 Hz, 1H), 8.68 (s,lH), 8.66 (dd, J= 4.4, 1.6 Hz, 1H), 8.32 (s, 1H), 7.79 (d, J= 8.8 Hz, 1H), 7.74 (s, 1H), 7.59 (d, J= 8.8 Hz, 1H), 7.30 (dd, J= 7.2, 4.4 Hz, 1H), 7.39 (t, J= 73.0 Hz, 1H), 7.14 (t, J= 55.8 Hz, 1H), 5.24 (s, 2H), 3.06 (s, 3H), 2.89 (s, 3H).

Example 7

N-(3-(5-(2,2-difluoroethyl)-2-(difluoromethoxy)phenyl)-l- (2-(dimethylamino)-2- oxoethyl)-lH-pyrazol-4-yl)pyrazolo[l,5-a]pyrimidine-3-carbox amide

To a solution of N-[3-[2-(difluoromethoxy)-5-(2-oxoethyl)phenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (90.0 mg, 0.181 mmol) in dichloromethane (2.0 mL) at 0 °C was added DAST (84 mg, 0.521 mmol) under nitrogen. The resulting solution was stirred for 2 h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions:

Column, XBridge Shield RP18 OBD Column, 5um, 19* 150mm; mobile phase, Water(0.05%NH 3 H 2 O) and ACN (10% ACN up to 45% in 12 min); Detector, UV 220nm. 27.4 mg product was obtained and concentrated under reduced pressure to give 36.2 mg (39%>) of N-[3-[5-(2,2-difluoroethyl)-2-(difluoromethoxy)phenyl]-l- [(dimethylcarbamoyl)methyl]-lH-pyrazol-4-yl]pyrazolo[l,5-a]p yrimidine-3- carboxamide as a white solid. LC/MS (Method C, ESI): [M+H] + = 520.2, R T = 1.89 min; 1H NMR (400 MHz, DMSO-^) δ 9.72 (s, 1H), 9.34 (dd, J= 6.8, 1.6 Hz, 1H), 8.67 (s, 1H), 8.64 (dd, J= 4.0, 1.6 Hz, 1H), 8.28 (s, 1H), 7.49 - 7.47 (m, 2H), 7.41 (d, J= 8.8 Hz, 1H), 7.28 (dd, J= 7.0, 4.2 Hz, 1H), 7.25 (t, J= 73.6 Hz, 1H), 6.46 - 6.09 (m, 1H), 5.20 (s, 2H), 3.26 - 3.19 (m, 2H), 3.06 (s, 3H), 2.88 (s, 3H).

Example 8

N-(3-(2-(difluoromethoxy)-5-(4-(dimethylcarbamoyl)phenoxy )phenyl)-l-(2- (dimethylamino)-2-oxoethyl)-lH-pyrazol-4-yl)pyrazolo[l,5-a]p yrimidine-3- carboxamide

To a solution of N-[3-[2-(difluoromethoxy)-5-[4-

(dimethylcarbamoyl)phenoxy]phenyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (Intermediate 6, 150 mg, 0.281 mmol) in DMF (4.0 mL) was added

Cs 2 C0 3 (183 mg, 0.562 mmol) and 2-bromo-N,N-dimethylacetamide (69.0 mg, 0.416 mmol). The reaction mixture was stirred for 2 h at 25 °C. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash

chromatography on silica gel eluting with dichloromethane/methanol (93/7) to give 22.0 mg (13%) of N-[3-[2-(difluoromethoxy)-5-[4- (dimethylcarbamoyl)phenoxy]phenyl] - 1 -[(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a white solid. LC/MS (Method A, ESI): [M+H] + = 619.3, R T = 1.50 min; 1H NMR (300 MHz, DMSO-^) δ 9.78 (s, 1H), 9.35 (dd, J= 7.1, 1.7 Hz, 1H), 8.70 (dd, J= 4.4, 1.7 Hz, 1H), 8.67 (s, 1H), 8.26 (s, 1H), 7.49 (d, J= 9.0 Hz, 1H), 7.42 (d, J= 8.7 Hz, 2H), 7.32 - 7.27 (m, 2H), 7.19 (t, J= 73.8 Hz, 1H), 7.18 (d, J= 3.0 Hz, 1H), 7.07 (d, J= 8.7 Hz, 2H), 5.18 (s, 2H), 3.03 (s, 3H), 2.94 (s, 6H), 2.86 (s, 3H).

Example 9

N-(3-(2-(difluoromethoxy)-5-(2-fluoroethyl)phenyl)-l-(2-( dimethylamino)-2- oxoethyl)-lH-pyrazol-4-yl)pyrazolo[l,5-a]pyrimidine-3-carbox amide

Step 1 : Synthesis of N-[3-[2-(difluoromethoxy)-5-(prop-2-en-l-yl)phenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide

To a solution of N-[3-[5-bromo-2-(difluoromethoxy)phenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (6.00 g, 11.23 mmol) in 1,4-dioxane (100 mL) was added 4,4,5,5- tetramethyl-2-(prop-2-en-l-yl)-l,3,2-dioxaborolane (3.02 g, 18.0 mmol, 1.600 equiv), Pd(dppf)Cl 2 dichloromethane (459 mg, 0.562 mmol) and a solution of CS 2 CO 3 (6.60 g, 20.3 mmol) in water (20 mL) under nitrogen. The resulting solution was stirred for 3 h at 85 °C. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with

dichloromethane/methanol (32/1) to give 4.10 g (74%) of N-[3-[2-(difluoromethoxy)- 5 -(prop-2-en- 1 -yl)phenyl] - 1 - [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a brown solid. LC/MS (Method L, ESI): [M+H] + = 496.1, R T = 0.83 min.

Step 2: Synthesis of N-[3-[2-(difluoromethoxy)-5-(2,3-dihydroxypropyl)phenyl]-l- [(dimethylcarbamoyl)methyl]-lH-pyrazol-4-yl]pyrazolo[l,5-a]p yrimidine-3- carboxamide

To a solution of N-[3-[2-(difluoromethoxy)-5-(prop-2-en-l-yl)phenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (7.06 g, 14.2 mmol) in tetrahydrofuran (140 mL) was added NMO (3.33 g, 28.4 mmol) and water (70 mL). This was followed by the addition of Os0 4 (7.23 g, 28.4 mmol) at room temperature. The resulting solution was stirred for 2 h at room temperature. The reaction was then quenched by the addition of 50 mL of Na 2 S 2 0 3 (sat.). The resulting mixture was extracted with ethyl acetate (3x). The combined organic phases were washed with water and brine successively, dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with dichloromethane/methanol (10/1) to obtain 4.02 g (53%) of N-[3-[2-(difluoromethoxy)-5-(2,3-dihydroxypropyl)phenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide as a light yellow solid. LC/MS (Method H, ESI): [M+H] + = 530.2, R T = 0.95 min.

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

To a solution of N-[3-[2-(difluoromethoxy)-5-(2,3-dihydroxypropyl)phenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (670 mg, 1.27 mmol) in CH 3 CN (15 mL) and water (3.0 mL) at 0 °C was added NaI0 4 (325 mg, 1.52 mmol). The resulting solution was stirred overnight at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was partitioned between water and ethyl acetate. The aqueous phase was extracted with ethyl acetate (2x). The combined organic phases were washed with brine, dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with

dichloromethane/methanol (92/8). The appropriate fractions were combined and concentrated under reduced pressure to give 482 mg (77%) of N-[3-[2-

(difluoromethoxy)-5 -(2-oxoethyl)phenyl] - 1 -[(dimethylcarbamoyl)methyl] - 1 H- pyrazol-4-yl]pyrazolo[l, 5 -a]pyrimidine-3 -carboxamide as a yellow solid. LC/MS (Method H, ESI): [M+H] + = 498.2, R T = 1.03 min.

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

To a solution of N-[3-[2-(difluoromethoxy)-5-(2-oxoethyl)phenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (487 mg, 0.979 mmol) in dichloromethane (20 mL) at 0 °C was added NaBH(OAc) 3 (300 mg, 1.42 mmol). The resulting solution was stirred for 3 h at room temperature. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with dichloromethane/methanol (93/7). The collected fractions were combined and concentrated under reduced pressure to obtain 430 mg (88%) of N-[3-[2- (difluoromethoxy)-5 -(2-hydroxyethyl)phenyl] - 1 -[(dimethylcarbamoyl)methyl] - 1 H- pyrazol-4-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a light yellow solid. LC/MS (Method H, ESI): [M+H] + = 500.3, R T = 1.00 min.

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

To a solution of N-[3-[2-(difluoromethoxy)-5-(2-hydroxyethyl)phi

[(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (200 mg, 0.400 mmol) in dichloromethane (10 mL) was added diethyl(trifluoro-4-sulfanyl)amine (96.6 mg, 0.599 mmol) at room temperature under nitrogen. The reaction mixture was stirred at room temperature for 3 h. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with dichloromethane/methanol (95/5). The appropriate fractions were combined and concentrated under reduced pressure. The crude product was further purified by Prep-HPLC with the following conditions: Column, silica gel; mobile phase, H 2 0 (NH 4 HC0 3 )/CH 3 CN = 90/10 increasing to H 2 0 (NH 4 HC0 3 )/CH 3 CN = 50/50 within 10 min; Detector, UV 254 nm to give 9.3 mg (5%) of N-[3-[2-(difluoromethoxy)-5-(2-fluoroethyl)phenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide as an off-white solid. LC/MS (Method A, ESI): [M+H] + = 502.3, R T = 1.48 min; 1H NMR (400 MHz, DMSO^) δ 9.74 (s, 1H), 9.34 (dd, J = 7.2, 1.6 Hz, 1H), 8.68 (s, 1H), 8.65 (dd, J = 4.2, 1.4 Hz, 1H), 8.28 (s, 1H), 7.45 - 7.40 (m, 2H), 7.39 (d, J = 8.8 Hz, 1H), 7.28 (dd, J= 7.0, 4.2 Hz, 1H), 7.23 (t, J= 73.6 Hz, 1H), 5.21 (s, 2H), 4.74 - 4.72 (m, 1H), 4.61 - 4.59 (m, 1H), 3.08 - 3.07 (m, 1H), 3.06 (s, 3H), 3.02 - 3.00 (m, 1H), 2.89 (s, 3H).

Examples 10 & 11

(S)-N-(3-(5-chloro-2-(difluoromethoxy)phenyl)- 1 -(1 -(dimethyl amino)- 1 -oxopropan- 2-yl)-lH-pyrazol-4-yl)pyrazolo[l, 5 -a]pyrimidine-3 -carboxamide & (R)-N-(3-(5- chloro-2-(difluoromethoxy)phenyl)- 1 -( 1 -(dimethylamino)- 1 -oxopropan-2-yl)- 1 H- pyrazol-4-yl)pyrazolo[ 1 ,5 -a]pyrimidine-3 -carboxamide

To a solution of N-[3-[5-chloro-2-(difluoromethoxy)phenyl]-lH-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide (200 mg, 0.494 mmol) in N,N- dimethylformamide (5.0 mL) was added 2-bromo-N,N-dimethylpropanamide (133 mg, 0.739 mmol) and CS 2 CO 3 (323 mg, 0.991 mmol). The resulting solution was stirred for 16 h at 60 °C in an oil bath. The resulting mixture was concentrated under reduced pressure. The residue was partitioned between ethyl acetate and water. The aqueous phase was extracted with ethyl acetate (2x). The combined organic phases were washed with brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by Prep-HP LC with the following conditions: Column, XBridge Shield RP18 OBD Column, 5um, 19* 150mm; mobile phase, water (10 mmol/L NH 4 HCO 3 ) and ACN (25.0% ACN up to 65.0% in 7 min); Detector, UV 220nm to give 120 mg of racemic mixture. The racemic mixture was separated by Chiral-Prep-HPLC with the following conditions: Column,

CHIRALPAK IF, 2*25cm, 5um; mobile phase, hexane and ethanol (hold 50.0% ethanol in 27 min); Detector, UV 220/254 nm to give two fractions: Isomer 1 (1 st peak): eluted at 18.08 min, 40.0 mg (16%) of N-[3-[5-chloro-2- (difluoromethoxy)phenyl] - 1 - [ 1 -(dimethylcarbamoyl)ethyl] - 1 H-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a white solid, LC/MS (Method A, ESI): [M+H] + = 504.2, R T = 1.65 min; 1H NMR (400 MHz, DMSO-d 6 ) δ 9.76 (s, 1H), 9.35 (dd, J = 7.2, 1.6 Hz, 1H), 8.68 (dd, J = 4.0, 1.6 Hz, 1H), 8.67 (s, 1H), 8.36 (s, 1H), 7.64 (dd, J = 8.8, 2.8 Hz, 1H), 7.61 (d, J = 2.8 Hz, 1H), 7.46 (d, J = 8.8 Hz, 1H), 7.30 (dd, J= 7.0, 4.2 Hz, 1H), 7.24 (t, J= 73.4 Hz, 1H), 5.65 (q, J= 6.8 Hz, 1H), 3.05 (s, 3H), 2.88 (s, 3H), 1.60 (d, J= 6.8 Hz, 3H).

Isomer 2 (2 nd peak): eluted at 22.84 min, 38.3 mg (15%) of N-[3-[5-chloro-2- (difluoromethoxy)phenyl] - 1 - [ 1 -(dimethylcarbamoyl)ethyl] - 1 H-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a white solid. LC/MS (Method A, ESI): [M+H] + = 504.2, R T = 1.65 min; 1H NMR (400 MHz, DMSO-d 6 ) δ 9.76 (s, 1H), 9.35 (dd, J = 7.2, 1.6 Hz, 1H), 8.68 (dd, J = 4.0, 1.6 Hz, 1H), 8.67 (s, 1H), 8.36 (s, 1H), 7.64 (dd, J = 8.8, 2.8 Hz, 1H), 7.61 (d, J = 2.8 Hz, 1H), 7.46 (d, J = 2.8 Hz, 1H), 7.30 (dd, J= 7.0, 4.2 Hz, 1H), 7.24 (t, J= 73.4 Hz, 1H), 5.65 (q, J= 6.8 Hz, 1H), 3.05 (s, 3H), 2.88 (s, 3H), 1.60 (d, J= 6.8 Hz, 3H).

Example 12

N-(3-(2-(difluoromethoxy)-5-((l,2,3,4-tetrahydroisoquinol in-7-yl)oxy)phenyl)-l-(2- (dimethylamino)-2-oxoethyl)- 1 H-pyrazol-4-yl)pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide

Step 1 : Synthesis of of tert-butyl 7-[4-(difluoromethoxy)-3-[l- [(dimethylcarbamoyl)methyl] -4- [pyrazolo [ 1 ,5 -a]pyrimidine-3 -amido] - 1 H-pyrazol-3- yl]phenoxy]-l,2,3,4-tetrahydroisoquinoline-2-carboxylate

To a solution of N-[3-[5-bromo-2-(difluoromethoxy)phenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (157 mg, 0.294 mmol) in toluene (10 mLl) was added CS 2 CO 3 (115 mg, 0.353 mmol), t-BuBrettPhos (14.3 mg, 0.0295 mmol), Pd 2 (allyl) 2 Cl 2 (5.38 mg, 0.0148 mmol) and tert-butyl 7-hydroxy-l, 2,3, 4-tetrahydroisoquino line -2-carboxylate (87.5 mg, 0.351 mmol) under nitrogen. The resulting solution was stirred overnight at 100 °C in an oil bath. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with

dichloromethane/methanol (10/1) to give 150 mg (73%) of tert-butyl 7-[4- (difluoromethoxy)-3-[ 1 -[(dimethylcarbamoyl)methyl]-4-[pyrazolo[ 1 ,5-a]pyrimidine- 3-amido]-lH-pyrazol-3-yl]phenoxy]-l, 2, 3, 4-tetrahydroisoquino line -2-carboxylate as a yellow solid. TLC: R f = 0.4; ethyl acetate/hexane = 4/1.

Step 2: Synthesis of N-[3-[2-(difluoromethoxy)-5-(l,2,3,4-tetrahydroisoquinolin-7 - yloxy)phenyl] - 1 -[(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [1,5- a]pyrimidine-3-carboxamide

Tert-butyl 7-[4-(difluoromethoxy)-3-[l-[(dimethylcarbamoyl)methyl]-4- [pyrazolo[l,5-a]pyrimidine-3-amido]-lH-pyrazol-3-yl]phenoxy] -l, 2,3,4- tetrahydroisoquinoline-2-carboxylate (150 mg, 0.213 mmol) was treated with HC1 in 1,4-dioxane (5.0 mL, 4M) for 1 h at room temperature. The resulting mixture was concentrated under reduced pressure. The crude product was purified by Prep-HPLC with the following conditions: Column, XBridge Shield RP18 OBD Column, 5um, 19* 150mm; mobile phase, Waters(0.05%NH 3 H 2 O) and ACN (15.0% ACN up to 55.0% in 10 min); Detector, UV 254/220 nm to afford 120 mg (93%) of N-[3-[2- (difluoromethoxy)-5-(l,2,3,4-tetrahydroisoquinolin-7-yloxy)p henyl]-l-

[(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide as a white solid. LC/MS (Method E, ESI): [M+H] + = 603.3, R T = 2.27 min; 1H NMR (300 MHz, DMSO-^) δ 9.74 (s, 1H), 9.35 (dd, J= 6.9, 1.5 Hz, 1H), 8.68 (dd, J= 4.4, 1.7 Hz, 1H), 8.67 (s, 1H), 8.25 (s, 1H), 7.43 (d, J= 9.0 Hz, 1H), 7.33 (dd, J= 7.2, 4.2 Hz, 1H), 7.18 (dd, J= 9.0, 3.0 Hz, 1H), 7.13 (t, J= 73.8 Hz,

1H), 7.06 (d, J= 8.4 Hz, 1H), 7.02 (d, J= 3.0 Hz, 1H), 6.82 (dd, J= 8.4, 2.4 Hz, 1H), 6.75 (d, J= 2.4 Hz, 1H), 5.17 (s, 2H), 3.77 (s, 2H), 3.03 (s, 3H), 2.93 (t, J= 5.7 Hz, 2H), 2.86 (s, 3H), 2.65 (t, J= 5.7 Hz, 2H).

Example 13

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

To a solution of N-[3-[5-chloro-2-(difluoromethoxy)phenyl]-l-

[(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (1.30 g, 2.65 mmol) in dioxane (30 mL) was added vinyl trifluoro- potassium borate (389 mg, 2.90 mmol), K 2 CO 3 (612 mg, 4.43 mmol), Pd(dppf)Cl 2 . CH 2 CI 2 (197 mg, 0.241 mmol) and water (4.0 mL) under nitrogen. The resulting solution was stirred for 4 h at 80 °C in an oil bath. The resulting mixture was concentrated under reduced pressure. The residue was partitioned between water and ethyl acetate. The aqueous phase was extracted with ethyl acetate (2x). The combined organic phases were washed with brine, dried, and concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with ethyl acetate to give 574 mg (45%) of N-[3-[2-(difluoromethoxy)-5-ethenylphenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide as a white solid. LC/MS (Method F, ESI): [M+H] + = 482.3, R T = 1.39 min; 1H NMR (400 MHz, DMSO-^) δ 9.77 (s, 1H), 9.35 (dd, J= 7.2, 1.6 Hz, 1H), 8.68 (s, 1H), 8.62 (dd, J= 4.4, 1.6 Hz, 1H), 8.30 (s, 1H), 7.69 (dd, J = 8.6, 2.2 Hz,

1H), 7.62 (d, J= 2.0 Hz, 1H), 7.41 (d, J= 8.4 Hz, 1H), 7.28 (dd, J= 6.7, 4.4 Hz, 1H), 7.27 (t, J= 73.6 Hz, 1H), 6.82 (dd, J= 17.6, 11.2 Hz, 1H), 5.87 (d, J= 17.6 Hz, 1H), 5.31 (d, J= 11.2 Hz, 1H), 5.22 (s, 2H), 3.06 (s, 3H), 2.89 (s, 3H).

Example 14

N-(3-(5-cyclopropyl-2-(dinuoromethoxy)phenyl)-l-(2-(dimet hylamino)-2-oxoethyl)- lH-pyrazol-4-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide

To a solution of N-[3-[5-cyclopropyl-2-(difluoromethoxy)phenyl]-lH-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide (Intermediate 5, 100 mg, 0.244 mmol) in DMF (10 mL) was added CS 2 CO 3 (160 mg, 0.491 mmol). To this mixture was added 2-bromo-N,N-dimethylacetamide (83 mg, 0.500 mmol). The resulting solution was stirred for 30 min at 60 °C in an oil bath. The resulting mixture was concentrated under reduced pressure. The residue was purified by flash chromatography on silica gel eluting with dichloromethane/methanol (8% MeOH) to obtain 63.8 mg (53%) of N- [3 -[5 -cyclopropyl-2-(difluoromethoxy)phenyl] - 1 - [(dimethylcarbamoyl)methyl] - lH-pyrazol-4-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a white solid. LC/MS (Method A, ESI): [M+H] + = 496.2, R T = 1.64 min; 1H NMR (400 MHz, DMSO-^) δ

9.74 (s, 1H), 9.35 (dd, J = 7.2, 4.0 Hz, 1H), 8.68 (s, 1H), 8.65 (dd, J= 4.0, 1.6 Hz, 1H), 8.27 (s, 1H), 7.32 - 7.23 (m, 4H), 7.16 (t, J= 74.0 Hz, 1H), 5.20 (s, 2H), 3.06 (s 3H), 2.89 (s, 3H), 2.03 - 1.97 (m, 1H), 0.99 - 0.94 (m, 2H), 0.73 - 0.68 (m, 2H).

Example 15

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

To a solution of crude N-[3-[5-bromo-2-(difluoromethoxy)phenyl]-lH-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide (Intermediate 2, 3.80 g, 8.46 mmol) in N,N-dimethylformamide (150 mL) was added CS 2 CO 3 (8.30 g, 25.5 mmol) and 2- bromo-N,N-dimethylacetamide (2.20 g, 13.3 mmol). The reaction mixture was stirred for 2 h at room temperature, and poured into 1.0 L of water with stirring. The solids were collected by filtration. The crude product was purified by re-crystallization from ethyl acetate once to afford 3.30 g (72% in two steps) of N-[3-[5-bromo-2- (difluoromethoxy)phenyl] - 1 - [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a yellow solid. LC/MS (Method D, ESI): [M+H] + = 536.1, R T = 2.72 min. 1H NMR (400 MHz, DMSO- ¾: δ (ppm) 9.75 (s, 1H), 9.36 (dd, J= 7.2, 1.6, 1H), 8.71 - 8.69 (m, 2H), 8.30 (s, 1H), 7.76 (dd, J= 8.8, 2.4, 1H), 7.68 (d, J= 2.8 Hz, 1H), 7.41 (d, J= 8.8 Hz, 1H), 7.31 (dd, J= 7.0, 4.2 Hz, 1H), 7.28 (t, J= 73.2 Hz, 1H), 5.22 (s, 2H), 3.06 (s, 3H), 2.89 (s, 3H).

Example 16

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

A stirred mixture of CS 2 CO 3 (31 g, 95 mmol), N,N-dimethylformamide (150 mL) and N- [3 -[5 -chloro-2-(difluoromethoxy)phenyl] - 1 H-pyrazol-4-yl]pyrazolo [1,5- a]pyrimidine-3-carboxamide (25 g, 62 mmol) was cooled to 0 °C using an ice water bath. 2-Bromo-N,N-dimethylacetamide (12.5 g, 75.3 mmol) was added dropwise. Upon the completion of the addition of 2-bromo-N,N-dimethylacetamide, the ice bath was removed, and the reaction mixture was allowed to warm to room temperature and stirred for 1.5 h at room temperature. The reaction mixture was gradually added to 2 L of H 2 0 with stirring. The precipitate was collected by filtration and dried under reduced pressure. MeOH (500 mL) was added to the crude product, and the mixture was heated to reflux for 0.5 h with stirring before being cooled to room temperature. The solids were collected by filtration, then an additional 500 mL of MeOH was added, and the mixture was heated to reflux for 30 min before being cooled to room temperature. The solids were collected by filtration to afford the title compound with 99.1% HPLC purity. The same reaction (at 25 g scale of N-[3-[5-chloro-2- (difluoromethoxy)phenyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide) was repeated 4 times and the samples of N-[3-[5-chloro-2- (difluoromethoxy)phenyl] - 1 - [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide from the 4 operations (81 g in total, all >99% HPLC purity) were combined for crystalline transformation.

N- [3 -[5 -chloro-2-(difluoromethoxy)phenyl] - 1 -[(dimethylcarbamoyl)methyl] - 1 H- pyrazol-4-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide (81 g, 165mmol) from the previous step was placed in a 1-L round bottom flask, and methanol (400 mL) was added. The mixture was stirred for 3 days at room temperature. The solids were collected by filtration and dried under reduced pressure. This resulted in 79.46 g (98%) of crystalline material of N-[3-[5-chloro-2-(difluoromethoxy)phenyl]-l- [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide as a yellow solid. LC/MS (Method D, ESI): [M+H] + = 490.2, R T = 2.63 min. 1H NMR (400 MHz, DMSO^) δ 9.77 (s, 1H), 9.36 (dd, J= 7.0, 1.4 Hz, 1H), 8.70 - 8.69 (m, 2H), 8.31 (s, 1H), 7.64 (dd, J= 8.8, 2.8 Hz, 1H), 7.57 (d, J= 2.4 Hz, 1H), 7.47 (d, J= 8.8 Hz, 1H), 7.30 (dd, J= 7.0, 4.2 Hz, 1H), 7.27 (t, J= 73.2 Hz, 1H), 5.22 (s, 2H), 3.06 (s, 3H), 2.89 (s, 3H).

Example 17

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

A stirring mixture of potassium carbonate (30 g, 217 mmol) and N-[3-[2,5- bis(difiuoromethoxy)phenyl] - 1 H-pyrazol-4-yl]pyrazolo [ 1 ,5 -a]pyrimidine-3 - carboxamide (Intermediate 7, 60 g, 137mmol) in N,N-dimethylformamide (400 mL) in a 1000-mL round-bottom flask was cooled to 0 °C using an ice water bath. 2- Bromo-N,N-dimethylacetamide (28 g, 169 mmol, 1.2 eq) was added dropwise, then the reaction was allowed to warm to room temperature, and stirring was continued overnight at room temperature. The reaction mixture was gradually added into 4 L of water with stirring. The solids were collected by filtration, then added to 2 L of ethyl acetate, and the mixture was heated to reflux temperature and stirred at this temperature for 30 min. The mixture was allowed to cool to room temperature and stirred at room temperature for two days. The solids were collected by filtration. The solids were divided into two equal portions and added to two 5-L round bottom flasks. Ethyl acetate (3 L each) was added to the two flasks, and the mixtures were heated to reflux. The mixture was stirred for 30 min at reflux temperature before being allowed to cool to room temperature and subsequently stirred for 48 h at room temperature. The solids were collected by filtration and dried under vacuum to afford 40 g (56%) of N-[3-[2,5-bis(difiuoromethoxy)phenyl]-l-[(dimethylcarbamoyl) methyl]-lH- pyrazol-4-yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide as a white solid (HPLC purity 99.0%). Crystalline transformation: EtOAc (200 mL) was added to 68 g of N-[3-[2,5- bis(difiuoromethoxy)phenyl] - 1 - [(dimethylcarbamoyl)methyl] - 1 H-pyrazol-4- yl]pyrazolo[l,5-a]pyrimidine-3-carboxamide (99.0% purity on HPLC ) in a 500 mL flask, and the mixture was stirred at room temperature for 2 days. The solid was filtered and dried under vacuum at room temperature for 6 h to afford the crystalline material (64.8 g ) (99.1% HPLC purity). LC/MS (Method D, ESI): [M+H] + = 522.2, R T = 2.62 min; 1H NMR (400 MHz, DMSO- ¾ δ 9.77 (s, 1H), 9.36 (dd, J= 6.8, 1.6 Hz, 1H), 8.69 (s, 1H), 8.67 (dd, J= 4.4, 1.6 Hz, 1H), 8.30 (s, 1H), 7.51 (d, J= 8.8 Hz, 1H), 7.39 (dd, J= 8.8, 2.8 Hz, 1H), 7.33 - 7.31 (m, 2H), 7.30 (t, J= 73.6 Hz, 1H), 7.24 (t, J= 73.2 Hz, 1H), 5.23 (s, 2H), 3.05 (s, 3H), 2.89 (s, 3H).

Example 18

N-(3 -(2-(difluoromethoxy)-5 -(3 -(3 -hydroxy- 1 -methylazetidin-3 -yl)phenoxy)phenyl)- l-(2-(dimethylamino)-2-oxoethyl)-lH-pyrazol-4-yl)pyrazolo[l, 5-a]pyrimidine-3- carboxamide

Step 1 : Synthesis of tert-butyl 3-[3-[4-(difluoromethoxy)-3- [4-(pyrazolo[l,5- a]pyrimidine-3 -carboxaniido)- 1 -((2-(trimethylsilyl)ethoxy)methyl)- 1 H-pyrazol-5 - yl]phenoxy]phenyl] -3 -hydroxyazetidine- 1 -carboxylate

A solution of N-[5-[5-bromo-2-(difluoromethoxy)phenyl]-l-(2- trimethylsilylethoxymethyl)pyrazol-4-yl]pyrazolo[l,5-a]pyrim idine-3-carboxami (300 mg, 0.518 mmol), tert-butyl 3-hydroxy-3-(3-hydroxyphenyl)azetidine-l- carboxylate (275 mg, 1.04 mmol), [PdCl(allyl)]2 (7.58 mg, 0.0207 mmol), RockPhos (24.3 mg, 0.0518 mmol), Cs2C03 (337 mg, 1.04 mmol), and toluene (4480 mg, 5.18 mL, 48.6 mmol) was stirred for 16 h at 100 °C under nitrogen. The resulting mixture was concentrated under vacuum and purified by flash chromatography on silica gel eluting with methanol/dichloromethane (0-10%). The appropriate fractions were combined and concentrated under reduced pressure to give tert-butyl 3-[3-[4- (difluoromethoxy)-3- [4-(pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamido)- 1 -((2- (trimethylsilyl)ethoxy)methyl)- 1 H-pyrazol-5 -yl]phenoxy]phenyl] -3 - hydroxyazetidine-l-carboxylate, 229 mg (59%) as a solid. LC/MS (Method L, ESI): [M+H]+ = 764.2.

Step 2: Synthesis of N-(3-(2-(difluoromethoxy)-5- (3-(3-hydroxyazetidin-3- yl)phenoxy)phenyl)-lH-pyrazol-4-yl)pyrazolo[l,5-a]pyrimidine -3-carboxamide

A solution of tert-butyl 3-[3-[4-(difluoromethoxy)-3-[4-(pyrazolo[l,5-a]pyrimidine-3 carbonylamino)-2-(2-trimethylsilylethoxymethyl)pyrazol-3 -yl]phenoxy]phenyl] -3 - hydroxy-azetidine-l-carboxylate (220 mg, 0.288 mmol), dichloromethane (3830 mg, 2.88 mL, 45.1 mmol), and trifluoroacetic acid (536 mg, 0.360 mL, 4.70 mmol) was stirred at ambient temperature for 16 h. The solution was concentrated under vacuum and used without further purification to afford N-(3-(2-(difluoromethoxy)-5- (3-(3- hydroxyazetidin-3 -yl)phenoxy)phenyl)- 1 H-pyrazol-4-yl)pyrazolo [ 1 ,5 -a]pyrimidine-3 carboxamide (220 mg, 1 13%), LC/MS (Method X, ESI): [M+H]+ = 534.1.

Step 3: Synthesis of N-(3-(2-(difluoromethoxy)-5- (3 -(3 -hydroxy- 1 -methyl azetidin-3- yl)phenoxy)phenyl)-lH-pyrazol-4-yl)pyrazolo[l, 5 -a]pyrimidine-3 -carboxamide

A solution of N-[3 - [2-(difluoromethoxy)-5 -[3 -(3 -hydroxyazetidin-3 - yl)phenoxy]phenyl]-lH-pyrazol-4-yl]pyrazolo[l,5-a]pyrimidine -3-carboxamide (220 mg, 0.326 mmol), 1,3,5-trioxane (39.1 mg, 0.434 mmol), dichloromethane (4320 mg, 3.26 mL, 50.9 mmol), methanol (516 mg, 0.652 mL, 15.8 mmol), and sodium tricacetoxyborohydride (207 mg, 0.979 mmol) was stirred at ambient temperature for 3 h. The resulting suspension was extracted with water and used without further purification to afford N-(3-(2-(difluoromethoxy)-5- (3-(3-hydroxy-l-methylazetidin- 3-yl)phenoxy)phenyl)-lH-pyrazol-4-yl)pyrazolo[l,5-a]pyrimidi ne-3-carboxamide (190 mg, 100%), LC/MS (Method X, ESI): [M+H]+ = 548.2.

Step 4: Synthesis of N-(3-(2-(difluoromethoxy)-5-(3-(3-hydroxy-l-methylazetidin-3 - yl)phenoxy)phenyl)- 1 -(2-(dimethylamino)-2-oxoethyl)- lH-pyrazol-4-yl)pyrazolo[ 1 ,5- a]pyrimidine-3 -carboxamide

A solution of N-[3 - [2-(difluoromethoxy)-5 -[3 -(3 -hydroxy- 1 -methyl-azetidin-3 - yl)phenoxy]phenyl]-lH-pyrazol-4-yl]pyrazolo[l, 5 -a]pyrimidine-3 -carboxamide (190 mg, 0.330 mmol), N,N-dimethylformamide (3110 mg, 3.30 mL, 42.6 mmol), 2- bromo-N,N-dimethyl-acetamide (65.7 mg, 0.0469 mL, 0.396 mmol), and N,N- diisopropylethylamine (174 mg, 0.230 mL, 1.32 mmol) was stirred at 50°C for 5 d. The residue was concentrated under reduced pressure and purified by Prep-HP LC with the following conditions: Column: Gemini NX C18 110A Column, 30x100 mm, 10 um; mobile phase: water (0.1% NH40H) and ACN (20% ACN up to 60% in 15 min); Detector, UV 254/220nm The appropriate fractions were combined and concentrated under reduced pressure to give N-(3-(2-(difluoromethoxy)-5-(3-(3- hydroxy- 1 -methylazetidin-3-yl)phenoxy)phenyl)- 1 -(2-(dimethylamino)-2-oxoethyl)- lH-pyrazol-4-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (2.4 mg, 1%). LC/MS (Method Y, ESI): [M+H] + = 633.2, R T = 3.42 min. 1H NMR (400 MHz, DMSO-d6) δ 13.01 (s, 1H), 9.75 (s, 1H), 9.36 (dd, J = 7.0, 1.7 Hz, 1H), 8.69 (dd, J = 4.2, 1.5 Hz, 1H), 8.65 (s, 1H), 8.25 (s, 1H), 7.44 (d, J = 8.9 Hz, 1H), 7.30 (dd, 1H), 7.38 - 6.89 (m, 7H), 3.32 (s, 2H), 3.20 (d, J = 13.7 Hz, 1H), 3.04 (d, J = 14.2 Hz, 1H), 2.72 - 2.62 (m, 7H), 2.47 - 2.39 (m, 1H), 2.33 (p, J = 1.8 Hz, 1H), 2.16 (s, 3H).

The following representative compounds of Table 1 were prepared using procedures similar to those described in the Schemes and Examples herein. Absolute stereochemistry of each compound below may not be depicted: therefore, structures may appear more than once, each representing a single stereoisomer.

Table 1 : Exem lary JAK Inhibitors of the Present Invention

Enzymatic Assays

JA Enzyme Assays were carried out as follows:

The activity of the isolated recombinant JAK1 and JAK2 kinase domain was measured by monitoring phosphorylation of a peptide derived from JAK3 (Val-Ala- Leu-Val-Asp-Gly-Tyr-Phe-Arg-Leu-Thr-Thr, fluorescently labeled on the N-terminus with 5-carboxyfluorescein) using the Caliper LabChip® technology (Caliper Life Sciences, Hopkinton, MA). To determine inhibition constants (K[), compounds were diluted serially in DMSO and added to 50 kinase reactions containing purified enzyme (1.5 nM JAK1 , or 0.2 nM JAK2), 100 mM HEPES buffer (pH 7.2), 0.015% Brij-35, 1.5 μΜ peptide substrate, ATP (25 μΜ), 10 mM MgCl 2 , 4 mM DTT at a final DMSO concentration of 2%. Reactions were incubated at 22 °C in 384-well polypropylene microtiter plates for 30 minutes and then stopped by addition of 25 μΐ, of an EDTA containing solution (100 mM HEPES buffer (pH 7.2), 0.015% Brij-35, 150 mM EDTA), resulting in a final EDTA concentration of 50 mM. After termination of the kinase reaction, the proportion of phosphorylated product was determined as a fraction of total peptide substrate using the Caliper LabChip® 3000 according to the manufacturer's specifications. K{ values were then determined using the Morrison tight binding model (Morrison, J.F., Biochim. Biophys. Acta. 185:269- 296 (1969); William, J.W. and Morrison, J.F., Meth. EnzymoL, 63:437-467 (1979)) modified for ATP-competitive inhibition [K \ = K avv l (1 + [ATP] / K m PP )]. Data for representative compounds is shown in Table 2.

JA 1 Pathway Assay in Cell Lines was carried out as follows:

Inhibitor potency (EC 50 ) was determined in cell-based assays designed to measure JAK1 dependent STAT phosphorylation. As noted above, inhibition of IL-4, IL-13, and IL-9 signaling by blocking the Jak/Stat signaling pathway can alleviate asthmatic symptoms in pre-clinical lung inflammation models (Mathew et al., 2001, J Exp Med 193(9): 1087-1096; Kudlacz et. al, 2008, Eur J. Pharmacol 582(1-3): 154- 161).

In one assay approach, TF-1 human erythroleukemia cells obtained from the American Type Culture Collection (ATCC; Manassas, VA) were used to measure JAK1 -dependent STAT6 phosphorylation downstream of IL-13 stimulation. Prior to use in the assays, TF-1 cells were starved of GM-CSF overnight in OptiMEM medium (Life Technologies, Grand Island, NY) supplemented with 0.5%> charcoal/dextran stripped fetal bovine serum (FBS), 0.1 mM non-essential amino acids (NEAA), and 1 mM sodium pyruvate. The assays were run in 384-well plates in serum-free OptiMEM medium using 300,000 cells per well. In a second assay approach, BEAS-2B human bronchial epithelial cells obtained from ATCC were plated at 100,000 cells per well of a 96-well plate one day prior to the experiment. The BEAS-2B assay was run in complete growth medium (bronchial epithelial basal medium plus bulletkit; Lonza; Basel, Switzerland).

Test compounds were serially diluted 1 :2 in DMSO and then diluted 1 :50 in medium just before use. Diluted compounds were added to the cells, for a final DMSO concentration of 0.2%, and incubated for 30 min (for the TF-1 assay) or 1 hr (for the BEAS-2B assay) at 37°C. Then, cells were stimulated with human recombinant cytokine at their respective EC90 concentrations, as previously determined for each individual lot. Cells were stimulated with IL-13 (R&D Systems, Minneapolis, MN) for 15 min at 37°C. The TF-1 cell reactions were stopped by the direct addition of lOx lysis buffer (Cell Signaling Technologies, Danvers, MA), whereas the BEAS-2B cell incubations were halted by the removal of medium and addition of lx lysis buffer. The resultant samples were frozen in the plates at -80°C. Compound mediated inhibition of STAT6 phosphorylation was measured in the cell lysates using MesoScale Discovery (MSD) technology (Gaithersburg, MD). EC50 values were determined as the concentration of compound required for 50% inhibition of STAT phosphorylation relative to that measured for the DMSO control. Data for representative compounds is shown in Table 2.

Table 2:

IL-13 p-STAT6

Ex JAKl Ki (uM) JAK2 Ki (uM) BEAS-2B

ICso (uM)

14 0.00080 0.00199 0.0473

15 0.00032 0.00043 0.0143

16 0.00065 0.00102 0.0334

17 0.00026 0.00072 0.0141

18 0.00318 0.00156 0.2850