Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PYRROLO [2, 3 -D]PYRIMIDINE DERIVATIVES AS INHIBITORS OF JANUS- RELATED KINASES (JAK)
Document Type and Number:
WIPO Patent Application WO/2014/128591
Kind Code:
A1
Abstract:
Described herein are pyrrolo{2,3-d}pyrimidine derivatives, their use as Janus Kinase (JAK) inhibitors, and pharmaceutical compositions containing them.

Inventors:
BROWN MATTHEW FRANK (US)
FENWICK ASHLEY EDWARD (US)
FLANAGAN MARK EDWARD (US)
GONZALES ANDREA (US)
JOHNSON TIMOTHY ALLAN (US)
KAILA NEELU (US)
MITTON-FRY MARK J (US)
STROHBACH JOSEPH WALTER (US)
TENBRINK RUTH E (US)
TRZUPEK JOHN DAVID (US)
UNWALLA RAYOMAND JAL (US)
VAZQUEZ MICHAEL L (US)
Application Number:
PCT/IB2014/058889
Publication Date:
August 28, 2014
Filing Date:
February 11, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
PFIZER (US)
International Classes:
C07D487/04; A61K31/519; A61P37/00; C07D519/00
Domestic Patent References:
WO2011045702A12011-04-21
WO2011075334A12011-06-23
WO2010020905A12010-02-25
WO2007012953A22007-02-01
WO1999065908A11999-12-23
WO1999065909A11999-12-23
WO2001042246A22001-06-14
WO2002000661A12002-01-03
WO2002096909A12002-12-05
WO2004046112A22004-06-03
WO2007012953A22007-02-01
WO2012075381A12012-06-07
WO2012009678A12012-01-19
WO2012117000A12012-09-07
WO2011086053A12011-07-21
WO2010032200A12010-03-25
WO2011068881A12011-06-09
WO2009060278A12009-05-14
WO2004032834A22004-04-22
Other References:
KISSELEVA ET AL., GENE, vol. 285, 2002, pages 1
YAMAOKA ET AL., GENOME BIOLOGY, vol. 5, 2004, pages 253
MURRAY, P.J., J. IMMUNOL., vol. 178, 2007, pages 2623 - 2629
KISSELEVA, T. ET AL., GENE, vol. 285, 2002, pages 1 - 24
O'SHEA, J. J. ET AL., CELL, vol. 109, 2002, pages S121 - S131
NEUBAUER H. ET AL., CELL, vol. 93, no. 3, 1998, pages 397 - 409
PARGANAS E. ET AL., CELL, vol. 93, no. 3, 1998, pages 385 - 95
STAHL; WERMUTH: "Handbook of Pharmaceutical Salts: Properties, Selection, and Use", 2002, WILEY-VCH
"Remington's Pharmaceutical Sciences", 1991, MACK PUB. CO.
T. W. GREENE; P. G. M. WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS INC.
J. MARCH: "Advanced Organic Chemistry", 1985, JOHN WILEY & SONS
W. R. BOWMAN; R. J. MARMON: "Comprehensive Organic Functional Group Transformations", vol. 2, 1995, PERGAMON
J. ORG. CHEM., vol. 72, 2007, pages 7423
Attorney, Agent or Firm:
KLEIMAN, Gabriel L. (235 East 42nd StreetNew York, NY, US)
Download PDF:
Claims:
CLAIMS

s claimed is:

1. A compound of formula I having the structure:

or a pharmaceutically acceptable salt thereof, wherein

R1 is hydrogen or C1-C4 alkyl, wherein said alkyl is further optionally substituted with one or more substituents selected from the group consisting of halo, hydroxy, methoxy, amino, CF , and C -C cycloalkyl;

R2 and R3 are each independently hydrogen, deuterium, C -C linear or branched chain alkyl, C3-C6 cycloalkyl, C^-C^ linear or branched chain perfluoroalkyl, C^-C^ linear or branched chain alkoxy, C1-C6 linear or branched chain perfluoroalkoxy, halogen, cyano, hydroxyl, amino, carboxy, aminocarbonyl, aryl, heteroaryl, (ary C^-C^ linear or branched chain alkyl,

(heteroaryDC^-C. linear or branched chain alkyl, (heterocyclic^-C. linear or branched chain alkyl, (C1-C6 linear or branched chain alkyl)aryl, (C^C. linear or branched chain alkyl)heteroaryl, (C1-C6 linear or branched chain alkyDheterocyclic, ( ^-C^ linear or branched chain

alkoxy carbonyl, (C^-C,. linear or branched chain alkyl)amino-carbonylamino, or (Cj-C linear or branched chain alkyl)aminocarbonyl;

R4 is selected from hydrogen, deuterium, C^-C^ linear or branched chain alkyl, C^-C^ lin¬ ear or branched chain perfluoroalkyl, aryl, and alkylaryl;

X is selected from— NH— and— RaRb— , where (a) Ra and Rb are independently hydrogen, deuterium, C^-C^. linear or branched chain alkyl, C3-C6 cycloalkyl, aryl, (aryOC^Cg linear or branched chain alkyl, heteroaryl, ( ^ ^ linear or branched chain alkyl)heteroaryl, (heteroaryl)^- C6 linear or branched chain alkyi, (heterocyclic)C1-C6 linear or branched chain alkyi, or (b) Ra and Rb together form a chain comprising— (CR Rd).— , where Rc and Rd are independently hydrogen, deuterium, C -C linear or branched chain alkyi, aryl, (C -C linear or branched chain alkyl)aryl, heteroaryl, (C1-C6 linear or branched chain alkyl)heteroaryl, halo, CN, CF3, hydroxyl, CONH2, or S02CH3;

Y is -A-R5, where A is a bond,— (CH2)k— or ~(CD2)k~ and R5 is C1-C6 linear or branched chain alkyi, C3-C6 cycloalkyi, aryl, or— N Ra.Rb,> or is an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyi, C3-C6 cycloalkyi, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, C1-C6 linear or branched chain alkyi, CN , hydroxyl, CF3,— ORe,— NReRf>— S(0)pRe and C3-C6 cycloalkyi, where said alkyi and cycloalkyi may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CONH2, and S02CH3, where (a) Ra, and Rb, are independently hydrogen, deuterium, C1-C6 linear or branched chain alkyi, C3-C6 cycloalkyi, aryl, (C1-C6 linear or branched chain alkyl)aryl, heteroaryl, or (C1-C6 linear or branched chain alkyl)heteroaryl, where said alkyi and cycloalkyi may be optionally su bstituted with one or more R ,, or (b) Ra, and Rb, together form a chain com- prising— (CRc,Rd,)— , where Rc, and Rd, are independently hydrogen, deuteri um, C1-C6 linear or branched chain alkyi, aryl, (C1-C6 linear or branched chain alkyl)aryl, heteroaryl, (C1-C6 linear or branched chain alkyl)heteroaryl, halo, CN , hydroxyl, CF3, CONH2,— ORe,— NReRf, or— S(0)pRe; where Re and Rf are independently hydrogen, deuterium, C1-C6 linear or branched chain alkyi, or C3-C6 cycloalkyi, where said alkyi and cycloalkyi may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF3, and CONH2; j is 2 , 3, 4 or 5 ; k is 1, 2; 3, or 4; p is 0, 1 or 2 ; and, n is 1 or 2.

2. The compound of claim 1 which is a compound of formula IA having the structure:

or a pharmaceutically acceptable salt thereof, wherein Y is -A-R5, where A is a bond,— (CH2)k— or— (CD2)k— and R5 is C1-C6 linear or branched chain alkyl, C3-C6 cycloalkyi, aryl, or— NR ,Rb,, or is an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure con- taining a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyl, C3-C6 cycloalkyi, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, C1-C6 linear or branched chain alkyl, CN, hydroxyl, CF3,— ORe,— NReRf>— S(0)pRe and C3-C6 cycloalkyi, where said alkyl and cycloalkyi may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CONH2, and S02CH3, where (a) Ra, and Rb, are independently hydrogen, deuterium, C^-C. linear or branched chain alkyl, C3-C6 cycloalkyi, aryl, (C^- C6 linear or branched chain alkyl)aryl, heteroaryl, or (C1-C6 linear or branched chain al- kyl)heteroaryl, where said alkyl and cycloalkyi may be optionally substituted with one or more Rc,, or (b) Ra, and Rb, together form a chain comprising— (CRc,Rd,)— , where Rc, and Rd, are inde¬ pendently hydrogen, deuterium, C1-C6 linear or branched chain alkyl, aryl, (C1-C6 linear or branched chain alkyl)aryl, heteroaryl, ( ^-C^ linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CF3, CONH2,— ORe,— NR Rf, or— S(0)pRe; where Re and Rf are independently hydrogen, deuterium, C1-C6 linear or branched chain alkyl, or C3-C6 cycloalkyi, where said alkyl and cycloalkyi may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF3, and CONH2; / is 2, 3, 4 or 5; k is 1, 2; 3, or 4; and, p is 0, 1 or 2. 3. A compound of claim 2 wherein A is a bond and R5 is a C -C^ linear or branched chain alkyl, C -C cycloalkyi or aryl.

4. A compound of claim 2 wherein A is a bond or— (CH )— , and R5 is C3-C6 cycloalkyi wherein said C3-C6 cycloalkyi is further optionally substituted with one or more substituents selected from the group consisting of halo, C1-C6 linear or branched chain alkyl, and CN where said alkyl and cycloalkyi may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CONH2, and S02CH3; where k is 1, 2, or 3.

5. A compound of claim 2 wherein A is a bond or— (CH2)k— , and R5 is an unsatu- rated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyl, C3-C6 cycloalkyi, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, C1-C6 linear or branched chain alkyl, CN, hydroxyl, CF3,— NRa,Rb„— ORe,— S(0)pR and C3-C6 cycloalkyi; where k is 1, 2, or 3.

6. The compound of claim 1 which is a compound of formula IB having the structure:

or a pharmaceutically acceptable salt thereof, wherein

(a) Ra, and Rb, are independently hydrogen, deuterium, C1-C6 linear or branched chain alkyl, C3-C6 cycloalkyi, aryl, (C -C linear or branched chain alkyl)aryl, heteroaryl, or (C -C linear or branched chain alkyl)heteroaryl, where said alkyl and cycloalkyi may be optionally substituted with one or more R ;

(b) Ra, and Rb, together form a chain comprising— (CRc,Rd,).— , where Rc, and Rd, are inde- pendently hydrogen, deuterium, C^C. linear or branched chain alkyl, aryl, (C1-C6 linear or branched chain alkyl)aryl, heteroaryl, (C1-C6 linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CF3, CONH2,— ORe,— NR,Rf> or— S(0)pRe; where Re and Rf are independently hydrogen, deuterium, C^C. linear or branched chain alkyl, or C3-C6 cycloalkyi, where said alkyl and cycloalkyi may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF3, and CONH2; or,

(c) Ra, and Rb, together form an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroa- toms independently selected from the group consisting of oxygen, nitrogen, and sulfur, where- in said monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, C1-C6 linear or branched chain alkyl, CN, hydroxyl, CF3,— NRa,Rb„—OR ,— S(0)pRe and C3-C6 cycloalkyi; j is 2, 3, 4 or 5; and, p is 0, 1 or 2. 7. The compound of claim 1 which is a compound of formula IC having the structure:

or a pharmaceutically acceptable salt thereof, wherein

(a) Ra, and Rb, are independently hydrogen, deuterium, C -C linear or branched chain alkyl, C3-C6 cycloalkyi, aryl, (C -C linear or branched chain alkyl)aryl, heteroaryl, or (C -C linear or branched chain alkyl)heteroaryl, where said alkyl and cycloalkyi may be optionally substituted with one or more R ;

(b) Ra, and Rb, together form a chain comprising— (CRc,Rd,).— , where Rc, and Rd, are independently hydrogen, deuterium, C^C. linear or branched chain alkyl, aryl, (C1-C6 linear or branched chain alkyl)aryl, heteroaryl, (C1-C6 linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CF3, CONH2,— ORe,— NR,Rf> or— S(0)pRe; where Re and Rf are independently hydrogen, deuterium, C^C. linear or branched chain alkyl, or C3-C6 cycloalkyi, where said alkyl and cycloalkyi may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF3, and CONH2; or,

(c) Ra, and Rb, together form an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroa- toms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, C1-C6 linear or branched chain alkyl, CN, hydroxyl, CF3,— NRa,Rb„—OR ,— S(0)pRe and C3-C6 cycloalkyi; j is 2, 3, 4 or 5; and, p is 0, 1 or 2.

8. The compound of claim 1 which is a compound of formula ID having the structure:

or a pharmaceutically acceptable salt thereof, wherein

Y is -AR5, where A is a bond or— (CH2)k— , and R5 is C1-C6 linear or branched chain alkyl, C3-C6 cycloalkyi, aryl, or is an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyl, C3-C6 cycloalkyi, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, C - C linear or branched chain alkyl, CN, hydroxyl, CF ,— NR R ,—OR ,— S(O) R and C -C cycloalkyi, where said alkyl and cycloalkyi may be optionally substituted with one or more sub- stituents selected from the group consisting of halo, CN, hydroxyl, CONH2, and S02CH3, where (a) Ra, and Rb, are independently hydrogen, deuterium, C^C^ linear or branched chain alkyl, C3- C6 cycloalkyi, aryl, (aryDC^C^ linear or branched chain alkyl, heteroaryl, (C1-C6 linear or branched chain alkyl)heteroaryl, (heteroary Cj-C. linear or branched chain alkyl, (heterocy- clic)C1-C6 linear or branched chain alkyl, where said alkyl and cycloalkyi may be optionally sub- stituted with one or more Rc„ or (b) Ra, and Rb, together form a chain comprising— (CRc,Rd,)— , where Rc, and Rd, are independently hydrogen, deuterium, C^C. linear or branched chain alkyl, aryl, (C1-C6 linear or branched chain alkyl)aryl, heteroaryl, (C^C. linear or branched chain al- kyDheteroaryl, halo, CN, hydroxyl, CF3, CONH2,— ORe,— NReRf, or— S(O) Re; where R and Rf where are independently hydrogen, deuterium, C1-C6 linear or branched chain alkyl, or C3-C6 cycloalkyi, where said alkyl and cycloalkyi may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF3, and CONH2; / is 2, 3, 4 or 5; k is 1, 2, or 3; and, p is 0, 1 or 2.

9. The compound of claim 1 selected from the group consisting of:

4-cyano-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}pyridine- 2-sulfonamide;

2,2,2-trifluoro-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}- ethanesulfonamide;

2- methyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}- propane-l-sulfonamide;

N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}propane-l- sulfonamide;

l-cyclopropyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}- methanesulfonamide;

N-{cis-3-[(butylsulfonyl)methyl]cyclobutyl}-N-methyl-7H-pyrrolo[2,3-d]pyrimidin-4- amine;

l-cyclopropyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}- azetidine-3-sulfonamide;

3- cyano-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}- azetidine-l-sulfonamide; (lR, 5S)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyri midin-4-yl)amino]cyclobutyl}-6-oxa-3- azabicyclo[3.1.1]heptane-3-sulfonamide;

(3R)-3-cyano-N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}- pyrrolidine- l-sulfonamide;

(3S)-3-cyano-N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}- pyrrol id ine- l-sulfonamide;

N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}- l-(oxetan-3- yl)methane-sulfonamide;

l-(3,3-d ifluorocyclobutyl)-N-{cis-3-[methyl(7H-pyrrolo[2 , 3-d]pyrimidin-4- yl)amino]cyclobutyl}methane-sulfonamide;

trans-3-(cyanomethyl)-N-{cis-3-[methyl(7H-pyrrolo[2 -d]pyrimidin-4-yl)amino]- cyclobutyl}cyclo-butanesulfonamide;

cis-3-(cyanomethyl)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyri midin-4-yl)amino]- cyclobutyljcyclobutane-sulfonamide;

N-[cis-3-({[(3 -difluorocyclobutyl)methyl]sulfonyl}methyl)cyclobutyl]-N-rnethyl-7H- pyrrolo[2 ,3-d]pyrimidin-4-amine;

(lS, 5S)- l-cyano-N-{cis-3-[methyl(7H-pyrrolo[2,3-d] pyrimidin-4-yl)amino]cyclobutyl}-

3- azabicyclo[3.1.0]hexane-3-sulfonamide;

(lR, 5 R)- l-cyano-N-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}- 3-azabicyclo[3.1.0]hexane-3-sulfonamide;

(3R)- l-[({cis-3-[methyl(7H-pyrrolo[2 -d]pyrimidin-4-yl)amino]cyclobutyl}meth- yl)sulfonyl] pyrrolidine- 3 -carbon itrile;

l-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}methyl)sulfonyl]-

4- (trifluoromethyl)piperidin-4-ol ;

N-(cis-3-{[(4,4-difluoropiperidin- l-yl)sulfonyl]methyl}cyclobutyl)-N-methyl-7H- pyrrolo[2 ,3-d]pyrimidin-4-amine;

(3S)- l-[({cis-3-[methyl(7H-pyrrolo[2 , 3-d]pyrimidin-4-yl)amino]cyclobutyl}meth- yl)sulfonyl] pyrrolidine- 3 -carbon itrile;

N-(cis-3-{[(3-chloro-4-fluorophenyl)sulfonyl]methyl}cyclobutyl)-N-methyl-7H- pyrrolo[2 ,3-d]pyrimidin-4-amine;

N-(cis-3-{[(2-cyclopropylethyl)sulfonyl]methyl}cyclobutyl)-N-methyl-7H-pyrrolo[2 ,3- d]pyrimidin-4-amine;

N-methyl-N-[cis-3-({[l-(propan-2-yl)pyrrolidin-3-yl]sulfonyl}methyl)cyclobutyl]-7H- pyrrolo[2 ,3-d]pyrimidin-4-amine; 3,3-difluoro-N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4- yl)amino]cyclobutyl}cyclobutane-sulfonamide;

1- [3-(cyanomethyl)oxetan-3-yl]-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}-methanesulfonamide;

cis-3-(cyanomethyl)-3-methyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}-cyclobutanesulfonamide;

trans-3-(cyanomethyl)-3-methyl-N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d] pyrimidin-4- yl)amino]cyclobutyl}cyclobutanesulfonamide;

N-(2-cyanoethyl)-N-methyl-N'-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidi n-4- yl)amino]cyclobutyl}sulfu ric diamide;

N^(lS R)-3-[methyl(7H-pyrrolo[2,3-d]pyri midin-4-yl)amino]cyclopentyl}propane- l- sulfonamide;

3-(2-hydroxypropan-2-yl)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}benzene-sulfonamide;

N-(cyclopropylmethyl)-N'-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4- yl)amino]cyclobutyl}sulfuric diamide;

N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}-4-(lH-pyrazol-3- yl)piperidine- l-sulfonamide;

2- methyl-N-{cis-3-[methyl(7H-pyrrolo[2 , 3-d]pyrimidin-4-yl)amino]cyclobutyl}-2 ,6- dihydropyrrolo[3,4-c]pyrazole-5(4H)-sulfonamide;

N-cyclopropyl- l-{trans-3-[methyl(7H-pyrrolo[2 , 3-d]pyrimidin-4- yl)amino]cyclobutyl}methane-sulfonamide;

2- [({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}methyl)sulfonyl]pyridine-4-carbonitrile;

(lS,3S)-3-[({cis-3-[methyl(7H-pyrrolo[2,3-d] pyri midin-4-yl)amino]cyclobutyl}methyl)- sulfonyl]cyclopentanecarbonitrile;

(lR,3R)-3-[({cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4- yl)amino]cyclobutyl}methyl)sulfonyl] cyclopentanecarbonitrile;

l-cyclopropyl-N-{trans-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4- yl)amino]cyclobutyl}methane sulfonamide;

3- cyano-N-{trans-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}pyrrolidine- l-sulfonamide;

N-methyl-N -{trans- 3-[(propylsulfonyl)methyl]cyclobutyl}-7H-pyrrolo[2 , 3-d] pyri mid in- 4-amine; and, 2-methyl-N-{cis-3-[methyl(7H-pyrrolo[2 -d]pyrimidin-4-yl)amino]cyclobutyl}- l,3- thiazole-5-sulfonamide; or, a pharmaceutically acceptable salt thereof.

10. A compound of clai m 1 wherein the compound is 2-methyl-N-{cis-3- [methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}- l,3-thiazole-5-sulfonamide, or a pharmaceutically acceptable salt thereof.

11. A compound of claim 1 wherein the compound is N-{cis-3-[methyl(7H- pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}-propane- l-sulfonamide, or a pharmaceutically acceptable salt thereof.

12. A compound of clai m 1 wherein the compound is trans-3-(cyanomethyl)-N- {cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}cyclobutanesulfonamide or a pharmaceutically acceptable salt thereof.

13. A compound of claim 1 wherein the compound is l-(3,3-difluorocyclobutyl)-N- {cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}methanesulfonamide or a pharmaceutically acceptable salt thereof.

14. A compound of claim 1 wherein the compound is N-{cis-3-[methyl(7H- pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}- l-(oxetan-3-yl)methanesulfonamide or a pharmaceutically acceptable salt thereof.

15. A compound of claim 1 wherein the compound is (3 R)- l-[({cis-3-[methyl(7H- pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}methyl)sulfonyl]pyrrolidine-3-carbonitrile or a pharmaceutically acceptable salt thereof.

16. A compound of claim 1 wherein the compound is 3 , 3-difluoro-N-{cis-3- [methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}cyclobutanesulfonamide or a pharmaceutically acceptable salt thereof.

17. A compound of claim 1 wherein the compound is (lS, 5S)- l-cyano-N-{cis-3- [methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}-3-azabicyclo[3.1.0]hexane-3- sulfonamide or a pharmaceutically acceptable salt thereof.

18. A pharmaceutical or a veterinary composition comprising a compound of claim 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

19. A method for treating or preventing a disorder or condition selected from rheu- matoid arthritis, myositis, vasculitis, pemphigus, Crohn's disease, , ulcerative colitis, Alzheimer's disease, lupus, nephritis, psoriasis, atopic dermatitis, autoimmune thyroid disorders, multiple sclerosis, major depression disorder, allergy, asthma, Sjogren's disease, dry eye syndrome, organ transplant rejection, xeno transplantation, Type I diabetes and complications from diabetes, cancer, leukemia, T cell acute lymphoblastic leukemia, adult T cell leukemia ac- tivated B-cell like, diffuse large B cell lymphoma, inflammatory bowel disease, septic shock, cardiopulmonary dysfunction, chronic pulmonary obstructive disorder, acute respiratory disease, and cachexia comprising the step of administering to a subject an effective amount of a composition comprising a compound of claim 1.

Description:
PYRROLO [2, 3 -D]PYRIMIDINE DERIVATIVES AS INHIBITORS OF JANUS- RELATED KINASES (JAK)

FIELD OF THE INVENTION

The present invention provides pharmaceutically active pyrrolo[2 ,3-d]pyri midine com- pounds and analogues. Such compounds are useful for inhibiting Janus Kinase (JAK). This invention also is directed to compositions comprisi ng methods for making such compounds, and methods for treating and preventing conditions mediated by JAK.

BACKGROUND OF THE INVENTION

Protein kinases are families of enzymes that catalyze the phosphorylation of specific residues in proteins, broadly classified into tyrosine and serine/threonine kinases. Inappropriate kinase activity, arising from mutation, over-expression, or inappropriate regulation, dys- regulation or de-regulation, as well as over- or under-production of growth factors or cytokines has been i mplicated in many diseases, including but not limited to cancer, cardiovascular diseases, allergies, asthma and other respiratory diseases, autoimmune d iseases, inflammatory diseases, bone diseases, metabolic disorders, and neurological and neurodegenerative disorders such as Alzheimer's disease. Inappropriate kinase activity triggers a variety of biological cellular responses relating to cell growth, cell differentiation , survival, apoptosis, mitogenesis, cell cycle control, and cel l mobility implicated in the aforementioned and related diseases.

Thus, protein kinases have emerged as an important class of enzymes as targets for therapeutic intervention. In particular, the JAK family of cellular protein tyrosine kinases (JAK1, JAK2, JAK3, and Tyk2) play a central role in cytoki ne signaling (Kisseleva et al., Gene, 2002, 285 , 1; Yamaoka et al. Genome Biology 2004, 5, 253)). Upon binding to their receptors, cytokines activate JAK which then phosphorylate the cytokine receptor, thereby creating docking sites for signaling molecules, notably, members of the signal transducer and activator of transcription (STAT) family that ultimately lead to gene expression. Numerous cytokines are known to activate the JAK family. These cytokines include, the IFN family (IFN-alpha, IFN-beta, IFN- omega, Limitin, IFN-gamma, IL- 10, IL- 19, IL-20, IL-22), the gp l30 family (IL-6, IL- 11, OSM, LIF, CNTF, NNT- l/BSF-3, G-CSF, CT- 1, Leptin, IL- 12 , I L-23), gamma C family (IL-2 , I L-7, TSLP, IL-9, IL- 15 , IL-21, IL-4, I L- 13), IL-3 family (IL-3 , IL-5 , GM-CSF), single chain family (EPO, GH, PRL, TPO), receptor tyrosine kinases (EGF, PDGF, CSF- 1, HGF), and G-protein coupled receptors (ATI).

There remains a need for new compounds that effectively and selectively inhibit specific JAK enzymes, and JAK1 in particular, vs. JAK2. JAK1 is a member of the Janus family of protein kinases composed of JAK1, JAK2, JAK3 and TYK2. JAK1 is expressed to various levels in all tissues. Many cytokine receptors signal through pairs of JAK kinases in the following combinations: JAK1/JAK2, JAK1/JAK3, JAK1/TYK2 , JAK2/TYK2 or JAK2/JAK2. JAK1 is the most broadly paired JAK kinase in this context and is required for signaling by γ-common (IL-2Ry) cytokine receptors, I L— 6 receptor family, Type I, II and III receptor families and IL-10 receptor family. Animal studies have shown that JAK1 is required for the development, function and homeostasis of the immune system. Modulation of immune activity through inhibition of JAK1 kinase ac- tivity can prove useful in the treatment of various immune disorders (Murray, P.J.

J. Immunol., 178, 2623-2629 (2007); Kisseleva, T., et al., Gene, 285, 1-24 (2002); O'Shea, J. J., et a!., Ceil, 109, (suppl.) S121-S131 (2002)) while avoiding JAK2 dependent erythropoietin (EPO) and thrombopoietin (TPO) signaling (Neubauer H., et al., Cell, 93(3), 397-409 (1998);

Parganas E., et al., Cell, 93(3), 385-95 (1998)).

SUMMARY OF THE INVENTION

The present invention provides a compound of formula I having the structure:

or a pharmaceutically acceptable salt thereof, wherein R 1 is hydrogen or C 1 -C 4 alkyl, wherein said alkyl is further optionally substituted with one or more substituents selected from the group consisting of halo, hydroxy, methoxy, amino, CF 3 , and C 3 -C 6 cycloalkyi; R 2 and R B are each independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, C 3 -C 6 cycloalkyi, C 1 -C 6 linear or branched chain perfluoroalkyl, t - 6 linear or branched chain alkoxy, C x -C 6 line- ar or branched chain perfluoroalkoxy, halogen, cyano, hydroxyl, amino, carboxy,

aminocarbonyl, aryl, heteroaryl, (aryl)C 1 -C 6 linear or branched chain alkyl, (heteroary C^C j . lin ¬ ear or branched chain alkyl, (heterocyclic)^-^ linear or branched chain alkyl, (C 1 -C 6 linear or branched chain alkyl)aryl, (C 1 -C 6 linear or branched chain alkyl)heteroaryl, (C 1 -C 6 linear or branched chain alkyl)heterocyclic, (C 1 -C 6 linear or branched chain alkoxyl)carbonyl, (C 1 -C 6 linear or branched chain alkyl)amino-carbonylamino, or (C 1 -C 6 linear or branched chain al- kyl)aminocarbonyl; R 4 is selected from hydrogen, deuterium, C -C linear or branched chain alkyl, C 1 -C 6 linear or branched chain perfluoroalkyl, aryl, and alkylaryl; X is selected from— NH— and— CR a R b — , where (a) R a and R b are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, C 3 -C 6 cycloalkyl, aryl, (aryDC^C. linear or branched chain alkyl, heteroaryl, (C 1 -C 6 linear or branched chain alkyl)heteroaryl, (heteroaryOC^-C^ linear or branched chain alkyl, (heterocyclic)^-^ linear or branched chain alkyl, or (b) R a and R b together form a chain comprising— (CR c R d ).— , where R and R d are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, aryl, (C 1 -C 6 linear or branched chain alkyl)aryl, heteroaryl, (C^^ linear or branched chain alkyl)heteroaryl, halo, CN, CF 3 , hydroxyl, CONH 2 , or S0 2 CH 3 ; Y is -A-R 5 , where A is a bond,— (CH 2 ) k — or— (CD 2 ) k — and R 5 is C^C. linear or branched chain alkyl, C 3 - C 6 cycloalkyl, aryl, or— NR,.R b , > or is an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroa- toms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyl, C 3 -C 6 cycloalkyl, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, C -C linear or branched chain alkyl, CN, hydroxyl, CF ,— OR ,— NR R ,— S(O) R and C -C cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CONH 2 , and S0 2 CH 3 , where (a) R a , and R b , are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, C 3 - C 6 cycloalkyl, aryl, (C 1 -C 6 linear or branched chain alkyl)aryl, heteroaryl, or (C 1 -C 6 linear or branched chain alkyl)heteroaryl, where said alkyl and cycloalkyl may be optionally substituted with one or more R c ,, or (b) R a , and R b , together form a chain comprising— (CR c ,R d ,).— , where R c , and R d , are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, aryl, (C 1 -C 6 linear or branched chain alkyl)aryl, heteroaryl, (C^C g linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CF 3 , CONH 2 ,— OR e ,— NR R f , or— S(O) R e ; where R and R f are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, or C 3 -C 6 cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF 3 , and CONH 2 ; / is 2, 3, 4 or 5; k is 1, 2; 3, or 4; p is 0, 1 or 2; and, n is 1 or 2.

In other aspects, the present invention also provides:

pharmaceutical compositions which comprise a pharmaceutically acceptable carrier and a compound of formula I;

methods for treating conditions or disorders including myositis, vasculitis, pemphigus, Crohn's disease, lupus, nephritis, psoriasis, multiple sclerosis, major depression disorder, al ¬ lergy, asthma, Sjogren's disease, dry eye syndrome, transplant rejection, cancer, inflammatory bowel disease, septic shock, cardiopulmonary dysfunction, acute respiratory disease, or ca- chexia by administering to a subject in need a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof;

methods for treating conditions or disorders including atopic dermatitis, eczema, psoriasis, scleroderma, lupus, pruritus, other pruritic conditions, allergic reactions including allergic dermatitis in mammal, horse allergic diseases including bite hypersensitivity, summer eczema, sweet itch in horses, heaves, inflammatory ai rway disease, recurrent airway obstruction, airway hyper-responsiveness, and chronic obstruction pulmonary disease by administering to a mammal in need a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof; and,

methods for the preparation of compounds of the present invention. The present invention will be further understood from the following description given by way of example only. The present invention is directed to a class of pyrrolo[2,3-d]pyri midine derivatives. In particular, the present invention is directed to pyrrolo[2,3-d]pyrimidine compounds useful as inhibitors of JAK, and particularly JAK1. While the present invention is not so limited, an appreciation of various aspects of the invention will be gained through the following discussion and the examples.

The term "alkyl", alone or in combination, means an acyclic, saturated hydrocarbon group of the formula C n H 2n+i which may be linear or branched. Examples of such groups include methyl, ethyl , n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl , tert-butyl, pentyl, iso-amyl and hexyl. Unless otherwise specified , an alkyl group comprises from 1 to 6 carbon atoms. The carbon atom content of alkyl and various other hydrocarbon-containing moieties is indicated by a prefix designating a lower and upper number of carbon atoms in the moiety, that is, the prefix C.-C indicates a moiety of the integer "i" to the integer "j" carbon atoms, inclusive. Thus, for example, C^-C 8 alkyl refers to alkyl of one to six carbon atoms, inclusive.

The term "hydroxy," as used herein, means an OH radical. The term "heterocyclic" refers to a saturated or partially saturated (i.e. non aromatic) heterocycle which may be attached via a ring nitrogen atom (when the heterocycle is attached to a carbon atom) or a ring carbon atom (in all cases). Equally, when substituted, the substituent may be located on a ring nitrogen atom (if the su bstituent is joined through a carbon atom) or a ring carbon atom (in all cases). Specific examples include oxiranyl, aziridinyl , oxetanyl, azetidinyl, tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, 1,4-dioxanyl, morpholinyl, piperazinyl, azepanyl, oxepanyl, oxazepanyl and diazepinyl.

The term "aryl" refers to an aromatic monocyclic or bicyclic hydrocarbon which may be attached via a ring carbon atom. Equally, when substituted, the substituent may be located on a ring carbon atom. Specific examples include phenyl, toluyl, xylyl, trimethylphenyl, and naphthyl. Examples of aryl substituents include alkyl, hydroxyl, halo, nitrile, alkoxy, trifluoromethyl, carboxamido, S0 2 Me, benzyl , and substituted benzyl.

The term "heteroaryl" refers to an aromatic heterocycle which may be attached via a ring carbon atom (in all cases) or a ring nitrogen atom with an appropriate valency (when the heterocycle is attached to a carbon atom). Equally, when substituted, the substituent may be located on a ring carbon atom (in all cases) or a ring nitrogen atom with an appropriate valency (if the su bstituent is joined through a carbon atom). Specific examples include thienyl, furanyl, pyrrolyl, pyrazolyl, imidazolyl, oxazolyl , isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, pyridyl, pyridazinyl, pyri midinyl and pyrazinyl . The term "cycloalkyl" means a monocyclic, saturated hydrocarbon group of the formula C n H 2n . Examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl. Unless otherwise specified, a cycloalkyl group comprises from 3 to 8 carbon atoms.

The terms "halo" and "halogen" refer to fluoride (F), chloride (CI), bromide (Br) or iodide

(I).

The term "mammal" refers to human, livestock or companion ani mals.

The term "companion animal" or "companion animals" refers to animals kept as pets or household animal. Examples of companion animals include dogs, cats, and rodents including hamsters, guinea pigs, gerbils and the like, rabbits, ferrets and birds.

The term "livestock" refers to animals reared or raised in an agricultural setting to make products such as food or fiber, or for its labor. In some embodiments, livestock are suitable for consumption by mammals, for example hu mans. Examples of livestock animals include cattle, goats, horses, pigs, sheep, including lambs, and rabbits, as well as birds, such as chickens, ducks and turkeys.

The term "treating" or "treatment" means an alleviation of symptoms associated with a disease, disorder or condition, or halt of further progression or worsening of those symptoms. Depending on the disease and cond ition of the patient, the term "treatment" as used herein may include one or more of curative, palliative and prophylactic treatment. Treatment can also include administering a pharmaceutical formulation of the present invention in combination with other therapies.

The term "therapeutically-effective" indicates the capability of an agent to prevent, or improve the severity of, the disorder, while avoidi ng adverse side effects typically associated with alternative therapies. The phrase "therapeutically-effective" is to be understood to be equivalent to the phrase "effective for the treatment, prevention, or amelioration", and both are intended to qualify the amount of each agent for use in the combination therapy which will achieve the goal of improvement in the severity of cancer, cardiovascular disease, or pain and inflammation and the frequency of incidence over treatment of each agent by itself, while avoiding adverse side effects typically associated with alternative therapies.

"Pharmaceutically acceptable" means suitable for use in mammals, companion animals or livestock animals.

If substituents are described as being "independently selected" from a group, each sub- stituent is selected independent of the other. Each substituent therefore may be identical to or different from the other substituent(s).

DETAILED DESCRIPTION OF THE INVENTION

The present invention is related to novel compounds which are selective JAK1 modulators useful for the treatment of diseases and conditions associated with dysregulation of the JAK1. The present invention further provides pharmaceutical compositions comprising such JAK1 modulators as well as methods of treating and/or preventing such diseases and conditions. Accordingly, the present invention provides a compound of formula I having the structure:

or a pharmaceutically acceptable salt thereof, wherein R 1 is hydrogen or C 1 -C 4 alkyl, wherein said alkyl is further optionally substituted with one or more substituents selected from the group consisting of halo, hydroxy, methoxy, amino, CF 3 , and C 3 -C 6 cycloalkyi; R 2 and R 3 are each independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, C 3 -C 6 cycloalkyi, C 1 -C fj linear or branched chain perfluoroalkyl, t - 6 linear or branched chain alkoxy, C 1 -C 6 linear or branched chain perfluoroalkoxy, halogen, cyano, hydroxyl, amino, carboxy,

aminocarbonyl, aryl, heteroaryl, (aryDC^C g linear or branched chain alkyl, (heteroaryl)^-^ linear or branched chain alkyl, (heterocyclic)^-^ linear or branched chain alkyl, (C^-C^ linear or branched chain alkyl)aryl, (C^-C j . linear or branched chain alkyl)heteroaryl, (C^-C j . linear or branched chain alkyl)heterocyclic, (C l -C 6 linear or branched chain alkoxyl)carbonyl, (C 1 -C 6 linear or branched chain alkyl)amino-carbonylamino, or (C -C linear or branched chain al- kyl)aminocarbonyl; R 4 is selected from hydrogen, deuterium, C -C linear or branched chain alkyi, C^-C. linear or branched chain perfluoroalkyl, aryl, and alkylaryl; X is selected from— NH— and— CR a R b — , where (a) R a and R b are independently hydrogen, deuterium, C^C. linear or branched chain alkyl, C 3 -C 6 cycloalkyl, aryl, (ary C^-C^ linear or branched chain alkyl, heteroaryl, (C 1 -C 6 linear or branched chain alkyl)heteroaryl, (heteroaryl)C 1 -C 6 linear or branched chain alkyl, (heterocyclic)^-^ linear or branched chain alkyl, or (b) R a and R b together form a chain comprising— (CR R d ).— , where R c and R d are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, aryl, (C 1 -C 6 linear or branched chain alkyl)aryl, heteroaryl, (C^-C^ linear or branched chain alkyl)heteroaryl, halo, CN, CF 3 , hydroxyl, CONH 2 , or S0 2 CH 3 ; Y is -A-R 5 , where A is a bond,— (CH 2 ) k — or— (CD 2 ) k — and R 5 is C^C. linear or branched chain alkyl, C 3 - C 6 cycloalkyl, aryl, or— NR ,R b ,, or is an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroa- toms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyl, C 3 -C 6 cycloalkyl, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, C -C linear or branched chain alkyl, CN, hydroxyl, CF ,— OR ,— NR R ,— S(O) R and C -C cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more sub- stituents selected from the group consisting of halo, CN, hydroxyl, CONH 2 , and S0 2 CH 3 , where (a) R a , and R b , are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, C 3 - C 6 cycloalkyl, aryl, (C 1 -C 6 linear or branched chain alkyl)aryl, heteroaryl, or (C 1 -C 6 linear or branched chain alkyl)heteroaryl, where said alkyl and cycloalkyl may be optionally substituted with one or more R c ,, or (b) R a , and R b , together form a chain comprising— (CR c ,R d ,).— , where R c , and R d , are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, aryl, (C 1 -C 6 linear or branched chain alkyl)aryl, heteroaryl, (C^^ linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CF 3 , CONH 2 ,— OR e ,— NR R f , or— S(O) R e ; where R and R f are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, or C 3 -C 6 cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF 3 , and CONH 2 ; / is 2, 3, 4 or 5; k is 1, 2; 3, or 4; p is 0, 1 or 2; and, n is 1 or 2. In one embodiment, the invention provides a compound of formula IA having the structure:

or a pharmaceutically acceptable salt thereof, wherein Y is -A-R 5 , where A is a bond,— (CH 2 ) k — or— (CD 2 ) k — and R 5 is C 1 -C 6 linear or branched chain alkyl, C 3 -C 6 cycloalkyi, aryl, or— NR ,R b ,, or is an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure con- taining a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyl, C 3 -C 6 cycloalkyi, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, C 1 -C 6 linear or branched chain alkyl, CN, hydroxyl, CF 3 ,— OR e ,— NR e R f> — S(0) p R e and C 3 -C 6 cycloalkyi, where said alkyl and cycloalkyi may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CONH 2 , and S0 2 CH 3 , where (a) R a , and R b , are independently hydrogen, deuterium, C^-C. linear or branched chain alkyl, C 3 -C 6 cycloalkyi, aryl, (C^- C 6 linear or branched chain alkyl)aryl, heteroaryl, or (C 1 -C 6 linear or branched chain al- kyl)heteroaryl, where said alkyl and cycloalkyi may be optionally substituted with one or more R c ,, or (b) R a , and R b , together form a chain comprising— (CR c ,R d ,)— , where R c , and R d , are inde ¬ pendently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, aryl, (C 1 -C 6 linear or branched chain alkyl)aryl, heteroaryl, ( ^-C^ linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CF 3 , CONH 2 ,— OR e ,— NR R f , or— S(0) p R e ; where R e and R f are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, or C 3 -C 6 cycloalkyi, where said alkyl and cycloalkyi may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF 3 , and CONH 2 ; / is 2, 3, 4 or 5; k is 1, 2; 3, or 4; and, p is 0, 1 or 2.

In one embodiment, the invention provides a compound of formula IA wherein A is a bond and R 5 is a C 1 -C 6 linear or branched chain alkyl, C 3 -C 6 cycloalkyi or aryl. In another em- bodiment, the invention provides a compound of formula IA wherein A is a bond or— (CH 2 ) k — , and R 5 is C -C cycloalkyi wherein said C -C cycloalkyi is further optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 6 linear or branched chain alkyl, and CN where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CONH 2 , and S0 2 CH 3 ; where k is 1, 2, or 3. In yet another embodiment, the invention provides a compound of for- mula IA wherein A is a bond or— (CH 2 ) k — , and R 5 is an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyl, C 3 -C 6 cycloalkyl, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, C 1 -C 6 linear or branched chain alkyl, CN, hydroxyl, CF 3 ,— NR,R b ,,— OR e ,— S(O) R and C -C cycloalkyl; where k is 1, 2, or 3.

In another embodiment, the invention provides the compound of formula IB having the structure:

or a pharmaceutically acceptable salt thereof, wherein (a) R a , and R b , are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, C 3 -C 6 cycloalkyl, aryl, {C -C^ linear or branched chain alkyl)aryl, heteroaryl, or (C^C g linear or branched chain alkyl)heteroaryl, where said alkyl and cycloalkyl may be optionally substituted with one or more R c , or (b) R a , and R b , together form a chain comprising— (CR c ,R d ,)— , where R c , and R d , are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, aryl, (C 1 -C 6 linear or branched chain alkyl)aryl, heteroaryl, (C 1 -C 6 linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CF 3 , CONH 2 ,— OR e ,— NR e R f , or— S(0) p R e ; where R c and R f are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, or C 3 -C 6 cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hy- droxyl, CF 3 , and CONH 2 ; or, (c) R a , and R b , together form an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, C x - C linear or branched chain alkyl, CN, hydroxyl, CF ,— NR R ,—OR ,— S(O) R and C -C cycloalkyl; j is 2, 3, 4 or 5; and, p is 0, 1 or 2.

In another embodiment, the invention provides the compound of formula IC having the structure:

or a pharmaceutically acceptable salt thereof, wherein (a) R a , and R b , are independently hydro ¬ gen, deuterium, C^C. linear or branched chain alkyl, C 3 -C 6 cycloalkyl, aryl, (C 1 -C 6 linear or branched chain alkyl)aryl, heteroaryl, or (C 1 -C 6 linear or branched chain alkyl)heteroaryl, where said alkyl and cycloalkyl may be optionally substituted with one or more R c , or (b) R a , and R b , together form a chain comprising— (CR ,R d ).— , where R c , and R d , are independently hydrogen, deuterium, C ± -C 6 linear or branched chain alkyl, aryl, (C 1 -C 6 linear or branched chain alkyl)aryl, heteroaryl, (C 1 -C 5 linear or branched chain alkyl)heteroaryl, halo, CN, hydroxyl, CF 3 , CONH 2 ,— OR e ,— NR e R f> or— S(0) p R e ; where R c and R f are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, or C 3 -C 6 cycloalkyl, where said alkyl and cycloalkyl may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF 3 , and CONH 2 ; or, (c) R a , and R b , together form an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said monocyclic or bicyclic ring structure is further optionally substi- tuted with one or more substituents selected from the group consisting of deuterium, halo, C x - C linear or branched chain alkyl, CN, hydroxyl, CF ,— NR R ,— OR ,— S(O) R and C -C cycloalkyi; / is 2, 3, 4 or 5; and, p is 0, 1 or 2.

In another embodiment, the invention provides the compound of formula ID having the structure:

or a pharmaceutically acceptable salt thereof, wherein

Y is -AR 5 , where A is a bond or— (CH 2 ) k — , and R 5 is C -C linear or branched chain alkyl, C 3 -C 6 cycloalkyi, aryl, or is an unsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyl, C 3 -C 6 cycloalkyi, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, C 1 -C e linear or branched chain alkyl, CN, hydroxyl, CF ,— NR R ,—OR ,— S(O) R and C -C cycloalkyi, where said alkyl and cycloalkyi may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CONH 2 , and S0 2 CH B , where (a) R a , and R b , are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, C 3 - C 6 cycloalkyi, aryl, (aryl)C 1 -C 6 linear or branched chain alkyl, heteroaryl, (C 1 -C 6 linear or branched chain alkyl)heteroaryl, (heteroaryl)C 1 -C 6 linear or branched chain alkyl, (heterocy- clic)C 1 -C 6 linear or branched chain alkyl, where said alkyl and cycloalkyi may be optionally substituted with one or more R ,, or (b) R a , and R b , together form a chain comprising— (CR c ,R d ).— , where R , and R d , are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, aryl, (C 1 -C 6 linear or branched chain alkyl)aryl, heteroaryl, (C 1 -C 6 linear or branched chain al- kyl)heteroaryl, halo, CN, hydroxyl, CF 3 , CONH 2 ,— OR e ,— NR e R f , or— S(0) p R e ; where R and R f where are independently hydrogen, deuterium, C 1 -C 6 linear or branched chain alkyl, or C 3 -C 6 cycloalkyi, where said alkyl and cycloalkyi may be optionally substituted with one or more sub- stituents selected from the group consisting of halo, CN, hydroxyl, CF 3 , and CONH 2 ; / is 2, 3 , 4 or 5 ; k is 1, 2 , or 3 ; and, p is 0, 1 or 2. In one embodiment, the invention provides the compound of formula ID wherein R 5 is a C 1 -C 6 linear or branched chain alkyi or C 3 -C 6 cycloalkyi.

In another embodi ment, the invention provides the compound of formula of formula ID wherein A is a bond or— (CH 2 ) k — , and R 5 is an u nsaturated, saturated or partially saturated monocyclic or bicyclic ring structure containing a total of five to eleven atoms having one to three heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur, wherein said alkyi, C 3 -C 6 cycloalkyi, aryl, or monocyclic or bicyclic ring structure is further optionally substituted with one or more substituents selected from the group consisting of deuterium, halo, C 1 -C 3 linear or branched chain alkyi, CN , hydroxyl, CF 3 ,— NR ,R b ,,— OR e ,—

S(0) p R c and C 3 -C 6 cycloalkyi; where R c and R f are independently hydrogen, deuteriu m, C 1 -C 6 linear or branched chain alkyi, or C 3 -C 6 cycloalkyi , where said alkyi and cycloalkyi may be optionally substituted with one or more substituents selected from the group consisting of halo, CN, hydroxyl, CF 3 , and CONH 2 ; k is 1, 2 , or 3 ; and, p is 0, 1 or 2. In another embodiment, the inven- tion provides the compound of formula I wherein R 5 is an unsaturated ring structure containing a total of five to eleven atoms having one or two heteroatoms independently selected from the group consisting of oxygen, nitrogen, and sulfur. In other embodiments, the invention provides the compound of formula I wherein R 5 is furyl, thiofuryl, pyrrolyl, pyrazolyl, oxazolyl , azetidinyl , piperidinyl or thiazolyl, optionally substituted by one or two methyl.

In another embodi ment, the invention provides the compound selected from the group consisting of:

4-cyano-N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}pyridine- 2-sulfonamide;

2 ,2,2-trifluoro-N-{cis-3-[methyl(7H-pyrrolo[2 , 3-d]pyrimidin-4-yl)amino]cyclobutyl}- ethanesulfonamide;

2-methyl-N-{cis-3-[methyl(7H-pyrrolo[2 , 3-d]pyrimidin-4-yl)amino]cyclobutyl}- propane-l-sulfonamide;

N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}propane- l- sulfonamide;

l-cyclopropyl-N-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}- methanesulfonamide;

N-{cis-3-[(butylsulfonyl)methyl]cyclobutyl}-N-methyl-7H-pyrr olo[2 ,3-d]pyri midin-4- amine;

l-cyclopropyl-N-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}- azetidine-3-sulfonamide; 3-cyano-N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}- azetidine- l-sulfonamide;

(lR, 5S)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyri midin-4-yl)amino]cyclobutyl}-6-oxa-3- azabicyclo[3.1.1]heptane-3-sulfonamide;

(3R)-3-cyano-N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}- pyrrol id ine- l-sulfonamide;

(3S)-3-cyano-N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}- pyrrol id ine- l-sulfonamide;

N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}- l-(oxetan-3- yl)methane-sulfonamide;

l-(3,3-d ifluorocyclobutyl)-N-{cis-3-[methyl(7H-pyrrolo[2 , 3-d]pyrimidin-4- yl)amino]cyclobutyl}methane-sulfonamide;

trans-3-(cyanomethyl)-N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]- cyclobutyljcyclo-butanesulfonamide;

cis-3-(cyanomethyl)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyri midin-4-yl)amino]- cyclobutyljcyclobutane-sulfonamide;

N-[cis-3-({[(3 ,3-difluorocyclobutyl)methyl]sulfonyl}methyl)cyclobutyl]-N-m ethyl-7H- pyrrolo[2 ,3-d]pyrimidin-4-amine;

(lS, 5S)- l-cyano-N-{cis-3-[methyl(7H-pyrrolo[2,3-d] pyrimidin-4-yl)amino]cyclobutyl}- 3-azabicyclo[3.1.0]hexane-3-sulfonamide;

(lR, 5 R)- l-cyano-N-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}-

3- azabicyclo[3.1.0]hexane-3-sulfonamide;

(3R)- l-[({cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}meth- yl)sulfonyl] pyrrolidine- 3 -carbon itrile;

l-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyc lobutyl}methyl)sulfonyl]-

4- (trifluoromethyl)piperidin-4-ol ;

N-(cis-3-{[(4,4-difluoropiperidin- l-yl)sulfonyl]methyl}cyclobutyl)-N-methyl-7H- pyrrolo[2 ,3-d]pyrimidin-4-amine;

(3S)- l-[({cis-3-[methyl(7H-pyrrolo[2 , 3-d]pyrimidin-4-yl)amino]cyclobutyl}meth- yl)sulfonyl] pyrrolidine- 3 -carbon itrile;

N-(cis-3-{[(3-chloro-4-fluorophenyl)sulfonyl]methyl}cyclobut yl)-N-methyl-7H- pyrrolo[2 ,3-d]pyrimidin-4-amine;

N-(cis-3-{[(2-cyclopropylethyl)sulfonyl]methyl}cyclobutyl)-N -methyl-7H-pyrrolo[2 ,3- d]pyrimidin-4-amine; N-methyl-N-[cis-3-({[l-(propan-2-yl)pyrrolidin-3-yl]sulfonyl }methyl)cyclobutyl]-7H- pyrrolo[2 ,3-d]pyrimidin-4-amine;

3,3-difluoro-N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}- cyclobutane-sulfonamide;

l-[3-(cyanomethyl)oxetan-3-yl]-N-{cis-3-[rnethyl(7H-pyrrolo[ 2,3-d]pyrimidin-4- yl)amino]cyclobutyl}-methanesulfonamide;

cis-3-(cyanomethyl)-3-methyl-N-{cis-3-[methyl(7H-pyrrolo[2,3 -d]pyri midin-4- yl)amino]cyclobutyl}-cyclobutanesulfonamide;

trans-3-(cyanomethyl)-3-methyl-N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d] pyrimidin-4- yl)amino]cyclobutyl}cyclobutanesulfonamide;

N-(2-cyanoethyl)-N-methyl-N'-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidi n-4- yl)amino]cyclobutyl}sulfuric diamide;

N-{(lS, 3R)-3-[methyl(7H-pyrrolo[2,3-d]pyri midin-4-yl)amino]cyclopentyl}propane- l- sulfonamide;

3-(2-hydroxypropan-2-yl)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]p yrimidin-4-yl)amino]- cyclobutyl}benzene-sulfonamide;

N-(cyclopropylmethyl)-N'-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]- cyclobutyljsulfuric diamide;

N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}-4-(lH-pyrazol-3- yl)piperidine- l-sulfonamide;

2-methyl-N-{cis-3-[methyl(7H-pyrrolo[2 , 3-d]pyrimidin-4-yl)amino]cyclobutyl}-2 ,6- dihydropyrrolo[3,4-c]pyrazole-5(4H)-sulfonamide;

2- [({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclo butyl}methyl)- sulfonyl]pyridine-4-carbonitrile;

(lS,3S)-3-[({cis-3-[methyl(7H-pyrrolo[2,3-d] pyri midin-4-yl)amino]cyclobutyl}methyl)- sulfonyl]cyclopentanecarbonitrile;

(lR,3R)-3-[({cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}- methyl)sulfonyl] cyclopentanecarbonitrile;

l-cyclopropyl-N-{trans-3-[methyl(7H-pyrrolo[2 , 3-d]pyrimidin-4-yl)amino]cyclobutyl}- methane sulfonamide;

3- cyano-N-{trans-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)ami no]cyclobutyl}- pyrrolidine- l-sulfonamide;

N-methyl-N -{trans- 3-[(propylsulfonyl)methyl]cyclobutyl}-7H-pyrrolo[2 , 3-d] pyri mid in- 4-amine; and, 2-methyl-N-{cis-3-[methyl(7H-pyrrolo[2 -d]pyrimidin-4-yl)amino]cyclobutyl}-1 - thiazole-5-sulfonamide; or, a pharmaceutically acceptable salt thereof.

In another embodi ment, the invention provides the compound selected from the group consisting of:

l-(3,3-d ifluorocyclobutyl)-N-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]- cyclo-butyl}methanesulfonamide;

trans-3-(cyanomethyl)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyri midin-4-yl)amino]- cyclobutyl}cyclo-butanesulfonamide;

N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclo butyl}propane- l- sulfonamide;

3,3-difluoro-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-y l)amino]cyclobutyl}- cyclobutane-sulfonamide; and,

N-{(lS, 3R)-3-[methyl(7H-pyrrolo[2,3-d]pyri midin-4-yl)amino]cyclopentyl}propane- l- sulfonamide; or, a pharmaceutically acceptable salt thereof.

In other embodiments, the invention provides the compound selected from the group consisting of:

(3 R)-3-cyano-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}pyrrolidine- l-sulfonamide;

(lR, 5S)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyri midin-4-yl)amino]cyclobutyl}-6-oxa-3- azabicyclo[3.1.1]heptane-3-sulfonamide;

(lS, 5S)- l-cyano-N-{cis-3-[methyl(7H-pyrrolo[2,3-d] pyrimidin-4-yl)amino]cyclobutyl}-

3- azabicyclo[3.1.0]hexane-3-sulfonamide;

N-(2-cyanoethyl)-N-methyl-N'-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidi n-4- yl)amino]cyclobutyl}sulfuric diamide; and,

2-methyl-N-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}-2,6- dihydropyrrolo[3,4-c]pyrazole-5(4H)-sulfonamide; or, a pharmaceutically acceptable salt thereof.

In another embodi ment, the invention provides the compound selected from the group consisting of:

(3 R)- l-[({cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}methyl)- sulfonyl] pyrrol id ine-3-carbonitrile;

l-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyc lobutyl}methyl)sulfonyl]-

4- (trifluoromethyl)piperidin-4-ol ;

N-(cis-3-{[(4,4-difluoropiperidin- l-yl)sulfonyl]methyl}cyclobutyl)-N-methyl-7H- pyrrolo[2,3-d]pyrimidin-4-amine; (3S)- l-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyc lobutyl}methyl) sulfonyl] pyrrol id ine-3-carbonitrile; and,

or, a pharmaceutically acceptable salt thereof.

In yet another embodiment, the invention provides the compound selected from the group consisting of:

(lR,3R)-3-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)a mino]cyclobutyl}methyl)- sulfonyl]cyclo-pentanecarbonitrile;

(lS,3S)-3-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)a mino]cyclobutyl}- methyl)sulfonyl]cyclo-pentanecarbonitrile;

2-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyc lobutyl}methyl)- sulfonyl]pyridine-4-carbonitrile;

N-[cis-3-({[(3,3-difluorocyclobutyl)methyl]sulfonyl}methyl)c yclobutyl]-N-methyl-7H- pyrrolo[2,3-d]pyrimidin-4-amine; and,

N-{cis-3-[(butylsulfonyl)methyl]cyclobutyl}-N-methyl-7H-pyrr olo[2,3-d]pyrimidin-4- amine; or, a pharmaceutically acceptable salt thereof.

Particularly preferred embodiments include 2-methyl-N-{cis-3-[methyl(7H-pyrrolo[2,3- d]pyri mid in-4-yl)amino]cyclobutyl}-l, 3-th iazole- 5 -sulfonamide, N-{cis-3-[methyl(7H- pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-propane- l-sulfonamide; N-{cis-3-[methyl(7H- pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-l-oxetan-3-yl methanesulfonamide; l-(3,3- difluorocyclobutyl)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimi din-4-yl)amino]cyclobutyl}- methanesulfonamide; 3,3-difluoro-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}cyclobutanesulfonamide ; trans-3-(cyanomethyl)-N-{cis-3-[methyl(7H- pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}-cyclobutanesu lfonamide; (lS, 5S)- l-cyano-N- {cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobu tyl}-3-azabi- cyclo[3.1.0]hexane-3-sulfonamide; and, (3S)-l-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin- 4-yl)amino]cyclobutyl}methyl)sulfonyl]pyrrolidine-3-carbonit rile; or, a pharmaceutically acceptable salt thereof.

The present invention also provides a pharmaceutical or a veterinary composition comprising a compound of formula I, IA, IB, IC or ID, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

The present invention also provides a method of treating a disorder or condition related to dysregulation of JAK, and particularly of JAKl, in a subject, comprising administering to the subject a therapeutically effective amount of the compound having the structure of formula I, IA, IB, IC or ID, or a pharmaceutically acceptable salt thereof. In certain embodiments, the dis- order or condition treated by the method is selected from among rheumatoid arthritis, myositis, vasculitis, pemphigus, Crohn's disease, , ulcerative colitis, Alzheimer's disease, lupus, nephritis, psoriasis, atopic dermatitis, autoimmune thyroid disorders, multiple sclerosis, major depression disorder, al lergy, asthma, Sjogren's disease, dry eye syndrome, organ transplant rejection, xeno transplantation, Type I diabetes and complications from diabetes, cancer, leukemia, T cell acute lymphoblastic leukemia, adult T cell leukemia activated B-cell like, diffuse large B cell lymphoma, inflammatory bowel disease, septic shock, cardiopulmonary dysfunction, chronic pulmonary obstructive disorder, acute respiratory disease, and cachexia comprising the step of administering to a subject an effective amount of a composition comprising a compound of formula I, IA, IB, IC or ID. In certain embodi ments, the therapeutically effective amount used in accord with the method is from 0.01 mg/kg of body weight/day to 100 mg/kg of body weight/day. In certain other embodi ments, the therapeutically effective amount used in accord with the method is the therapeutically effective amount is from 0.1 mg/kg of body weight/day to 10 mg/kg of body weight/day. In the practice of the method, the compound of formula I is preferably selected from N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}- propane- 1-sulfonamide, N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d] pyrimidin-4-yl)amino]cyclobutyl}- l-oxetan-3-yl methanes ulfonamide; l-(3 , 3-difluorocyclobutyl)-N-{cis-3-[methyl(7H- pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}-methanesulfonamide; 3 ,3-difluoro-N-{cis-3- [methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}cyclobutanesulfonamide ; trans-3- (cyanomethyl)-N-{cis-3-[methyl(7H-pyrrolo[2 , 3-d]pyrimidin-4-yl)amino]cyclobutyl}- cyclobutanesulfonamide; (lS, 5S)- l-cyano-N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4- yl)amino]cyclobutyl}-3-azabi-cyclo[3.1.0]hexane-3-sulfonamid e; and, (3S)- l-[({cis-3- [methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}methyl)sulfonyl]pyrrolid ine-3- carbonitrile;, or a pharmaceutically acceptable salt thereof.

The present invention further provides a method for treating or preventing a disorder or condition selected from atopic dermatitis, eczema, scleroderma, pruritus, other pruritic conditions, allergic reactions including allergic dermatitis in mammal, horse allergic diseases including bite hypersensitivity, summer eczema, sweet itch in horses, heaves, inflammatory airway disease, recurrent airway obstruction, and airway hyper-responsiveness by ad ministering to a mammal in need a therapeutically effective amount of a compound of formula I, IA, IB, IC or ID, or a pharmaceutically acceptable salt thereof.

In certain embodi ments, the therapeutically effective amount used in accord with the method is from 0.01 mg/kg of body weight/day to 100 mg/kg of body weight/day. In certain other embodiments, the therapeutically effective amount used in accord with the method is wherein the therapeutically effective amount is from 0.1 mg/kg of body weight/day to 10 mg/kg of body weight/day. In accord with the method, the mammal treated with the com- pound of the invention is selected from companion animals, dogs, and livestock. In certain embodi ments, the compound of formula I, IA, IB, IC or ID, or a pharmaceutically acceptable salt thereof, may be administered in accord with the method orally, parenterally, or topically.

In the practice of the method, the compound of formula I is preferably selected from N-{cis-3- [methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}-propane- l-sulfonamide; N-{cis-3- [methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}- l-oxetan-3- ylmethanesulfonamide; l-(3 ,3-difluorocyclobutyl)-N-{cis-3-[methyl(7H-pyrrolo[2,3- d]pyrimidin-4-yl)amino]cyclobutyl}methanesulfonamide; 3, 3-difluoro-N-{cis-3-[methyl(7H- pyrrolo[2 ,3-d]pyrimidin-4-yl)amino]cyclobutyl}cyclobutanesulfonamide ; trans-3- (cyanomethyl)-N-{cis-3-[methyl(7H-pyrrolo[2 , 3-d]pyrimidin-4-yl)amino]cyclobutyl}- cyclobutanesulfonamide; (lS, 5S)- l-cyano-N-{cis-3-[methyl(7H-pyrrolo[2 ,3-d]pyrimidin-4- yl)amino]cyclobutyl}-3-azabi-cyclo[3.1.0]hexane-3-sulfonamid e; and, (3S)- l-[({cis-3- [methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}methyl)sulfonyl]pyrrolid ine-3- carbonitrile; or, a pharmaceutically acceptable salt thereof.

Compounds that have the same molecular formula but differ in the nature or sequence of bonding of their atoms or the arrangement of their atoms in space are termed "isomers". Isomers that differ in the arrangement of their atoms in space are termed "stereoisomers". It will be appreciated by those skilled in the art that the compound of formula I, IA, IB, IC or ID can exist as cis- and trans- achiral diastereomers.

Included within the scope of the described compounds are all isomers (e.g. cis-, trans-, or diastereomers) of the compounds descri bed herein alone as well as any mixtures. All of these forms, including enantiomers, d iastereomers, cis, trans, syn, anti, solvates (including hydrates), tautomers, and mixtures thereof, are included in the described compounds.

Stereoisomeric mixtures, e.g . mixtures of diastereomers, can be separated into their corre- sponding isomers in a known manner by means of suitable separation methods.

Diastereomeric mixtures for example may be separated into their individual diastereomers by means of fractionated crystallization, chromatography, solvent distribution, and simi lar procedures. This separation may take place either at the level of one of the starting compounds or in a compound of formula I , IA, IB, IC or I D itself. Enantiomers may be separated through the for- mation of diastereomeric salts, for example by salt formation with an enantiomer-pure chiral acid, or by means of chromatography, for example by HPLC, using chromatographic substrates with chiral ligands.

In therapeutic use for treating disorders in a mammal, a compound of the present invention or its pharmaceutical compositions can be administered orally, parenterally, topically, rectally, transmucosally, or intestinally. Parenteral administrations include indirect injections to generate a systemic effect or direct injections to the afflicted area. Topical administrations include the treatment of skin or organs readily accessible by local application, for example, eyes or ears. It also includes transdermal delivery to generate a systemic effect. The rectal administration includes the form of suppositories. The preferred routes of administration are oral and parenteral.

Pharmaceutically acceptable salts of the compounds of formula I, IA, IB, IC or ID include the acid addition and base salts thereof. Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, adipate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulfate, borate, camsylate, citrate, cyclamate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulfate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phos- phate/dihydrogen phosphate, pyroglutamate, saccharate, stearate, succinate, tannate, tartrate, tosylate, trifluoroacetate and xinofoate salts.

Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sod ium, tromethamine and zinc salts.

Hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts. For a review on suitable salts, see Handboo of Pharmaceutical Salts: Properties, Selection, and Use by Stahl and Wermuth (Wiley-VCH, 2002).

Pharmaceutically acceptable salts of compounds of formula I, IA, IB, IC or ID may be prepared, respectively, by one or more of three methods: (i) by reacting the compound of formula I, IA, IB, IC or ID with the desired acid or base; (ii) by removing an acid- or base-labile protecting group from a suitable precursor of the compound of formula I, IA, IB, IC or ID or by ring-opening a suitable cyclic precursor, for example, a lactone or lactam, using the desired acid or base; or (iii) by converting one salt of the compound of formula I, IA, IB, IC or ID to another by reaction with an appropriate acid or base or by means of a suitable ion exchange column. All three reactions are typically carried out in solution . The resulting salt may precipitate out and be collected by filtration or may be recovered by evaporation of the solvent. The degree of ionization in the resulting salt may vary from completely ionized to almost non-ionized.

Pharmaceutical compositions of the present invention may be manufactured by methods well known in the art, e.g., by means of conventional mixing, dissolving, granulation, dragee- making, levigating, emulsifying, encapsulating, entrapping, lyophilizing processes or spray dry- ing. Pharmaceutical compositions for use in accordance with the present invention may be formulated in conventional manner using one or more pharmaceutically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active compound into preparations, which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. Pharmaceutically acceptable excipients and carriers are generally known to those skilled in the art and are thus included in the instant invention. Such excipients and carriers are described, for example, in "Remington's Pharmaceutical Sciences" Mack Pub. Co., New Jersey (1991). The formulations of the invention can be designed to be short-acting, fast-releasing, long-acting, and sustained-releasing. Thus, the pharmaceutical formulations can also be formulated for controlled release or for slow release.

Pharmaceutical compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an amount sufficient to achieve the intended purpose, i.e., control or the treatment of disorders or diseases. More specifically, a therapeutically effective amount means an amount of compound effective to prevent, alleviate or ameliorate symptoms/signs of disease or prolong the survival of the subject being treated.

The quantity of active component, which is the compound of this invention, in the pharmaceutical composition and unit dosage form thereof, may be varied or adjusted widely depending upon the manner of administration, the potency of the particular compound and the desired concentration. Determination of a therapeutically effective amount is well within the capability of those skilled in the art. Generally, the quantity of active component will range between 0.01% to 99% by weight of the composition.

Generally, a therapeutically effective amount of dosage of active component will be in the range of about 0.01 to about 100 mg/kg of body weight/day, preferably about 0.1 to about 10 mg/kg of body weight/day, more preferably about 0.3 to 3 mg/kg of body weight/day, even more preferably about 0.3 to 1.5 mg/kg of body weight/day It is to be understood that the dosages may vary depending upon the requirements of each subject and the severity of the disorders or diseases being treated.

The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day. The sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.

Also, it is to be understood that the initial dosage administered may be increased beyond the above upper level in order to rapidly achieve the desired plasma concentration. On the other hand, the initial dosage may be smaller than the optimum and the daily dosage may be progressively increased during the course of treatment depending on the particular situation. If desired, the daily dose may also be divided into multiple doses for administration, e.g., two to four times per day.

Compounds of the present invention are directed to pyrrolo[2,3-d] pyrimidi ne com- pounds useful as Janus Kinase inhibitors (JAK-i). They are useful as therapeutic agents in connection with the treating or preventing a disorder or condition selected from rheumatoid arthritis, myositis, vasculitis, pemphigus, Crohn's disease, ulcerative colitis, Alzheimer's disease, lupus, nephritis, psoriasis, atopic dermatitis, autoimmune thyroid disorders, multiple sclerosis, major depression disorder, allergy, asthma, Sjogren's disease, dry eye syndrome, organ trans- plant rejection, xeno transplantation, Type I diabetes and complications from diabetes, cancer, leukemia, T cell acute lymphoblastic leukemia, adult T cell leukemia activated B-cell like, diffuse large B cell lymphoma, inflammatory bowel disease, septic shock, cardiopulmonary dysfunction, chronic pulmonary obstructive disorder, acute respiratory disease, cachexia, and other indications where i mmunosuppression/immunomodulation would be desirable, comprising the step of administering to a subject an effective amount of a compound of the invention.

There are substantial needs for safe and efficacious agents to control d isorders related to JAK, such as atopic dermatitis, both in human and animals. The market for treating atopic dermatitis in ani mals is currently dominated by corticosteroids, which cause distressing and undesirable side effects in animals, specifically in companion animals such as dogs. Antihista- mines are also used, but are poorly effective. A canine formulation of cyclosporine (ATOPICA™) is currently being marketed for atopic dermatitis, but is expensive and has a slow onset of efficacy. In addition, there are Gl toleration issues with ATOPICA™. Compou nds of the present invention are JAK inhibitors with selective efficacy against JAK1. These compounds are expected to provide an alternative to steroid usage and provide resolution of chronic pruritus and in- flammation that would either persist in atopic dermatitis or slowly regress following removal of allergen or causative agent, such as fleas in flea-allergic dermatitis.

Compounds of the present invention may be administered in a pharmaceutically acceptable form either alone or in combination with one or more additional agents which modulate a mammalian immune system or with anti-inflammatory agents. These agents may include but are not limited to cyclosporin A (e.g., Sandimmune™ or Neoral™, rapamycin, FK-506

(tacrolimus), leflunomide, deoxyspergualin, mycophenolate (e.g., Cellcept™, azathioprine (e.g., Imuran™), daclizumab (e.g., Zenapax™), OKT3 (e.g., Orthocolone™), AtGam, aspirin, acetaminophen, ibuprofen, naproxen, piroxicam, and anti-inflammatory steroids (e.g., prednisolone or dexamethasone). These agents may be administered as part of the same or separate dosage forms, via the same or different routes of administration, and on the same or different admin- istration schedules according to standard pharmaceutical practice known to one skilled in the art.

Accordingly, the invention provides methods of treating or preventing a disease, condition or disorder associated with JAK in a subject, such as a hu man or non-human mammal, comprising administering an effective amount of one or more compounds described herein to the subject. Suitable subjects that can be treated include domestic or wild animals, companion animals, such as dogs, cats, horses and the like; livestock including, cows and other ruminants, pigs, poultry, rabbits and the like; primates, for example monkeys, such as rhesus monkeys and cynomolgus (also known as crab-eating or long-tailed) monkeys, marmosets, tamarins, chimpanzees, macaques and the like; and rodents, such as rats, mice, gerbils, guinea pigs and the like. In one embodiment, the compound is administered in a pharmaceutically acceptable form, optionally in a pharmaceutically acceptable carrier.

Conditions in which selective targeting of the JAK pathway or modulation of the JAK kinases, particularly JAK1, are contemplated to be therapeutically useful include, arthritis, asth- ma, autoimmune diseases, cancers or tumors, diabetes, certain eye diseases, disorders or conditions, inflammation, intestinal inflammations, allergies or conditions, neurodegenerative diseases, psoriasis, and transplant rejection . Conditions which can benefit from selective inhibition of JAK1 are discussed in greater detail below.

Accordingly, the compound of formula I, IA, IB, IC or ID, or its pharmaceutically accepta- ble salts, and pharmaceutical compositions thereof can be used to treat a variety of conditions or diseases such as the following:

Arthritis, including rheumatoid arthritis, juvenile arthritis, and psoriatic arthritis;

Autoimmune diseases or disorders, including those designated as single organ or single cell-type autoimmune disorders, for example Hashimoto's thyroiditis, autoimmune hemolytic anemia, autoi mmune atrophic gastritis of pernicious anemia, autoimmune encephalomyelitis, autoimmune orchitis, Goodpasture's disease, autoimmune thrombocytopenia, sympathetic ophthalmia, myasthenia gravis, Craves' disease, primary biliary cirrhosis, chronic aggressive hepatitis, ulcerative colitis and membranous glomerulopathy, those designated as involving systemic autoimmune disorder, for example systemic lu pus erythematosis, rheumatoid arthri- tis, Sjogren's syndrome, Reiter's syndrome, polymyositis-dermatomyositis, systemic sclerosis, polyarteritis nodosa, multiple sclerosis and bullous pemphigoid, and additional autoimmune diseases, which can be O-cell (humoral) based or T-cell based, including Cogan's syndrome, ankylosing spondylitis, Wegener's granulomatosis, autoimmune alopecia, Type I or juvenile onset diabetes, or thyroiditis; Cancers or tumors, including alimentary/gastrointestinal tract cancer, colon cancer, liver cancer, skin cancer including mast cell tumor and squamous cell carcinoma, breast and mammary cancer, ovarian cancer, prostate cancer, lymphoma, leukemia, including acute

myelogenous leukemia and chronic myelogenous leukemia, kidney cancer, lung cancer, muscle cancer, bone cancer, bladder cancer, brain cancer, melanoma including oral and metastatic melanoma, Kaposi's sarcoma, myelomas including multiple myeloma, myeloproliferative disorders, proliferative diabetic retinopathy, or angiogenic-associated disorders including solid tumors;

Diabetes, including Type I diabetes or complications from diabetes;

Eye diseases, disorders or conditions including autoimmune diseases of the eye, keratoconjunctivitis, vernal conjunctivitis, uveitis including uveitis associated with Behcet's disease and lens-induced uveitis, keratitis, herpetic keratitis, conical keratitis, corneal epithelial dystrophy, keratoleukoma, ocular premphigus, Mooren's ulcer, scleritis, Grave's

ophthalmopathy, Vogt-Koyanagi-Harada syndrome, keratoconjunctivitis sicca (dry eye), phlyctenule, iridocyclitis, sarcoidosis, endocrine ophthalmopathy, sympathetic ophthalmitis, allergic conjunctivitis, or ocular neovascularization;

Intestinal inflammations, allergies or conditions including Crohn's disease and/or ulcerative colitis, inflammatory bowel disease, celiac diseases, proctitis, eosinophilic gastroenteritis, or mastocytosis;

Neurodegenerative diseases including motor neuron disease, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, cerebral ischemia, or neurodegenerative disease caused by trau matic injury, strike, glutamate neurotoxicity or hypoxia; ischemic/reperfusion inj ury in stroke, myocardial ischemica, renal ischemia, heart attacks, cardiac hypertrophy, atherosclerosis and arteriosclerosis, organ hypoxia, or platelet aggregation;

Skin diseases, conditions or disorders including atopic dermatitis, eczema, psoriasis, scleroderma, pruritus or other pruritic conditions;

Allergic reactions including allergic dermatitis in mammal (including horse allergic diseases such as bite hypersensitivity), summer eczema, sweet itch in horses, heaves, inflammatory airway disease, recurrent airway obstruction, airway hyper-responsiveness, or chronic ob- struction pulmonary disease;

Asthma and other obstructive airways diseases, including chronic or inveterate asthma, late asthma, bronchitis, bronchial asth ma, allergic asthma, intrinsic asth ma, extrinsic asthma, or dust asthma;

Transplant rejection, including pancreas islet transplant rejection, bone marrow trans- plant rejection, graft-versus-host disease, organ and cell transplant rejection such as bone marrow, cartilage, cornea, heart, intervertebral disc, islet, kidney, limb, liver, lung, muscle, myoblast, nerve, pancreas, skin, small intestine, or trachea, or xeno transplantation; and

Another embodiment provides a method of selectively inhibiting a JAK1 enzyme, which includes contacting the JAK enzyme with either a non-therapeutic amount or a therapeutically effective amount of one or more of the presently taught compounds. Such methods can occur in vivo or in vitro. In vitro contact can involve a screening assay to determine the efficacy of the one or more compounds against a selected enzyme at various amounts or concentrations. In vivo contact with a therapeutically effective amount of the one or more compounds can involve treatment of a descri bed disease, disorder or condition or prophylaxis of organ transplant rejection in the ani mal in which the contact occurs. The effect of the one or more compounds on the JAK enzyme and/or host ani mal can also be determined or measured. Methods for determining JAK activity include those described in the Examples as well as those disclosed in WO99/65908, WO 99/65909, WO01/42246, WO02 /00661, WO02/096909, WO2004/046112 and WO2007/012953.

Chemical Synthesis

The following schemes and written descriptions provide general details regarding the preparation of the compounds of the invention.

SULFONAMIDES

Compounds of formula I, wherein p is 2, X is NH, Y is AR 5 ,and A is a bond, may be prepared according to Scheme 1.

Scheme 1

I (p = 2, X = NH, Y = AR 5 ; A = bond)

It will be apparent to those skilled in the art that sensitive functional groups (PC) may need to be protected and deprotected during the synthesis of a compound of the invention. Protection and deprotection may be achieved by conventional methods, as described, for example, in "Protective Groups in Organic Synthesis" by T. W. Greene and P. G. M. Wuts, John Wiley & Sons Inc. (1999), and references therein. Thus, in Scheme 1, Step 1, a compound of formula II, wherein Q 1 is halogen, is treated with a protecting agent to provide a compound of formula III, wherein PG s an arylsulfonyl protecting group such as benzenesulfonyl, or preferably para- toluenesulfonyl ("tosyl"). The protecting group may be installed by reaction of the compound of formula II with an arylsulfonyl chloride, preferably tosyl chloride, in the presence of a base such as aqueous sodium hydroxide solution and an organic solvent such as acetone. The reaction is typically run at 0 C C to about 50°C, preferably at about 23°C (room temperature). Alternatively, bases such as sodium hydride and potassium tert-butoxide may be used, employing a suitable solvent such as Ν,Ν-dimethylformamide or tetrahydrofuran. Several compounds of formula II are known in the literature and have been prepared by the methods above. For example, the synthesis of compound of formula II, wherein Q 1 is CI and R 2 and R 3 are hydrogen has been reported previously, for example in WO 2007 012953. In Scheme 1, Step 2 , the protected compound of formula III is combined with 1-2 equivalents of an amine of formula IV in the presence of a 1-3 equivalents of a base and a protic solvent to afford a compound of formula V. Suitable bases include triethylamine, diisopropylethylamine, and potassium carbonate while suitable solvents include methanol, eth- anol, diisopropyl alcohol and water or mixtures thereof. The reaction is typically run at about 23°C to about 150°C, preferably about 75°C. It will be noted that the amine of formula IV contains a second amino group that is protected with a protecting group PG 2 that can be removed under conditions that do not lead to loss of PG 1 . Suitable protecting groups PG 2 include t- butoxycarbonyl ("Boc") and ("Cbz"), preferably benzyloxycarbonyl.

In Scheme 1, Step 3, the protecting group PG 2 is removed from the compound of formula V under conditions that do not lead to loss of PG 1 to give a primary amine (or a salt thereof) of formula VI. When PG 2 is benzyloxycarbonyl, the benzyloxycarbonyl protecting group may be removed by hydrogenolysis wherein the compound of formula V is exposed to hydrogen or a hydrogen transfer reagent such as cyclohexene in the presence of a hydrogenation catalyst such as palladium hydroxide using a solvent such as methanol, acetic acid or, preferably, etha- nol. Alternatively, when PG 2 is benzyloxycarbonyl, the benzyloxycarbonyl protecting group may be removed by treatment of the compound of formula V with a solution of hydrogen bromide (about 6 equivalents) in acetic acid optionally in the presence of a suitable solvent such as ethyl acetate at a temperature from about minus 20°C to about 40°C, preferably less than 25°C. This latter deprotection method is preferred wherein n is 1, R 2 , R 3 and R 4 are hydrogen, R 1 is methyl, PG 1 is tosyl and PG 2 is benzyloxycarbonyl and provides the amine of formula VI as the dihydrobromide salt. When PG 2 is t-butoxycarbonyl, the t-butoxycarbonyl protecting group may be removed by treatment with an excess of an acid such as hydrochloric acid or trifluoroacetic acid in a solvent such as dichloromethane or 1,4-dioxane.

In Scheme 1, Step 4, the primary amine of formula VI (or salt thereof) is converted to a sulfonamide derivative of formula VII by treatment with an activated sulfon ic acid derivative of formula VIII, wherein Q 2 is halogen, O-alkyl or O-aryl in the presence of a base. Most commonly, VIII is a sulfonyl chloride derivative wherein Q 2 is CI. Many sulfonyl chlorides may be obtained from commercial sources. Also, several methods exist for the preparation of sulfonyl chlorides, which are well known to those skilled in the art and have been described in texts such as "Advanced Organic Chemistry" by J. March, John Wiley & Sons (1985). Typically, the amine of formula VI is treated with a sulfonyl chloride derivative of formula VIII wherein Q 2 is CI in the presence of at least one equivalent of a base such as triethylamine or diisopropylamine in a suitable solvent such as dichloromethane, tetrahydrofuran or acetonitrile. When a salt form of the amine is used, an additional equivalent of base is used for each equivalent of acid forming the salt. For example, using a dihydrobromide salt, two extra equivalents of base are used. The reaction may be run from about minus 20°C to about 50°C, preferably starting the reaction at about 0°C and then allowing it to warm to about 23°C (room temperature).

Finally, in Scheme 1, Step 5, the sulfonamide derivative of formula VII is deprotected to afford a compound of formula 1, wherein p is 2 , X is NH, Y is AR 5 and A is a bond. Two methods are typically employed , the choice of which is determined by the compatibility of the conditions with other functional groups on the molecule. The first method involves exposure of the compound of formula VII to an excess (about 4 equivalents) of a base such as lithium hydroxide or sodiu m hydroxide. The reaction is run in a solvent mixture containing water and an alcohol such as methanol or ethanol. It may also be run in a mixture of water and tetrahydrofuran, and, optionally an alcohol such as methanol or ethanol. The reaction may be ru n at a temperature of about 23 °C to about 100°C, typically about 60°C. The second method, which is preferred in instances where there is hydroxide-sensitive functionality such as nitrile present in the molecule, involves reaction of the compound of formula VII with an excess of tetrabutylammonium fluoride (4-25 equivalents) in a solvent such as 1,2-dimethoxyethane or, preferably

tetrahydrofuran, The deprotection is conducted at a temperature of about 0°C to about 60°C, preferably about 23°C.

Compounds of formula II, wherein Q 1 is halogen, are commercially available or are known in the chemical literature. For example, 4-chloro-7 - -pyrrolo[2,3-rf]pyrimidine, wherein Q 1 is CI and R 2 and R 3 are both hydrogen, is a read ily available commercial compound.

Compounds of formula IV are known in the chemical literature or may be prepared by standard chemical reactions well known to one skilled in the art.

An alternative method of preparing compounds of the invention wherein p is 2, X is NH, Y is AR 5 , A is a bond is shown in Scheme 2.

cheme 2

I (p = 2, X = NH, Y = AR 5 , A = bond) In Scheme 2, Step 1, a compound of formula IX is combined with a benzyloxycarbamate derivative of formula X in the presence of a base (1-5 equivalents) to provide a

benzyloxycarbamate derivative of formula XI. The reaction is carried out in a solvent such as water or an alcohol such as ethanol, optionally with addition of a miscible co-solvent such as tetrahydrofuran. Suitable bases include potassiu m carbonate, cesium carbonate, triethylamine and diisopropylethylamine. The reaction is run at about 23°C to about 100°C. When n is 1, R 2 , R 3 and R 4 are hydrogen, and R 1 is methyl, the preferred conditions are to run the reaction in water, using potassium carbonate (3 equivalents) as base, starting the reaction at about 23°C and then heating to about 95 °C.

In Scheme 2, Step 2 , the benzyloxycarbamate derivative of formula XI is deprotected by exposure to hydrogen or a hydrogen transfer reagent such as cyclohexene in the presence of a hydrogenation catalyst such as palladium hydroxide. At the same time, under the conditions of the deprotection, the chlorine atom at the 2-position of 7H-pyrrolo[2 ,3-d]pyrimidine ring is replaced with hydrogen to provide an amine hydrochloride salt of formula XII. The reaction is run in a solvent such as methanol or ethanol at a temperature of about 50°C to about 80°C. When R 2 , R 3 and R 4 are hydrogen, and R 1 is methyl, the preferred conditions are to run the reaction in ethanol at about 78°C using palladium hydroxide as catalyst, and cyclohexene (about 20 equivalents) as a hydrogen transfer reagent.

Finally, in Scheme 2, Step 3, the amine hydrochloride of formula XII is converted to a sulfonamide of formula I, wherein p is 2 , X is NH , Y is AR 5 , A is a bond by reaction with an activated sulfonic acid derivative of formula VIII, wherein Q 2 is halogen, O-alkyl or O-aryl in the presence of at least two equivalents of a base. Most commonly, VIII is a sulfonyl chloride derivative wherein Q 2 is CI. Suitable bases include triethylamine, diisopropylethylamine and potas- sium carbonate. Suitable solvents include Ν, Ν-dimethylformamide, and a mixture of tetrahydrofuran and water. The reaction may be run at a temperature of about minus 20°C to about 50°C preferably at about 23°C. Alternatively, the amine hydrochloride of formula XII is first treated with about 2 equivalents of trimethylchlorosilane in the presence of about 2-3 equivalents of a base such as lithiu m bis(di methylsilyl)amide or sodium bis(dimethylsilyl)amide in an suitable aprotic solvent such as tetrahydrofuran. Then, after about 1 hour, about 1.2 equivalents of the sulfonyl chloride of formula VIII, Q 2 is CI is added to provide, after workup, the sulfonamide of formula I, wherein p is 2 , X is NH, Y is AR 5 , A is a bond The reaction may be run at a temperature of about minus 20°C to about 50°C, preferably at about 23°C.

Compounds of formula IX, are commercially available or are known in the chemical literature. For example, 2 ,4-dichloro-7 - -pyrrolo[2 ,3-c ]pyrimidine, wherein R 2 and R 3 are both hydrogen, is commercially available. Its synthesis is described in PCT International Publication No. WO2007/012953.

SULFAMIDES

Compounds of formula I, wherein p is 2, X is NH, and Y is N R R b , may be prepared according to Scheme 3.

In Scheme 3, Step 1, an amine of formula VI (or salt thereof), wherein PG 1 is an arylsulfonyl protecting group such benzenesulfonyl, or preferably tosyl, is converted to oxazolidinone derivative of formula XIII. First, a solution of N-chlorosulfonylisocyanate (1 equivalent) is slowly added to a solution of 2-bromoethanol (1 equivalent) at a temperature of about -40°C to about 10°C, preferably about 0°C. Subsequently, after 0.5 to 2 hours, a solution of the amine of formula VI (1 equivalent) and a base such as triethylamine or

diisopropylethylamine (about 3 equivalents, plus one equivalent for each mole of acid forming a salt) is added slowly and the reaction is allowed to warm to about 23°C over a period of about 10 to 24 hours. Suitable solvents for the reaction include chloroform or preferably dichloro- methane.

In Scheme 3, Step 2 , the oxazolidinone derivative of formula XIII is reacted with 1-3 equivalents of an amine of the formula HNR R b , in the presence of a base (2-5 equivalents), to afford a sulfamide derivative of formula XIV. Suitable bases include triethylamine and diisopropylethylamine. The reaction is preferably carried out by heating to about 90°C to about 150°C in a pressure vessel using a suitable solvent such a Ν,Ν-di methylformamide or acetoni- trile.

In Scheme 3, Step 3, the compound of formula XIV is deprotected, removing the arylsulfonyl protecting group PG 1 to provide a sulfamide derivative of formula 1, wherein p is 2 , X is NH , and Y is NR R b . The reaction may be carried out by one of the two general methods described for Scheme 1, Step 5. Again, the choice of deprotection method is determined by the compati bility of the conditions with other functional groups on the molecule. Alternatively, the sulfamides of the formula XIV may be obtained directly from an amine of formula VI (or salt thereof). Thus, in Scheme 3, Step 4, the amine of formula VI (or salt thereof) is treated with a sulfamoyl chloride of the formula CI-S0 2 N R R b and a base such as triethylamine or

diisopropylethylamine as described for Scheme 1, Step 4. Sulfamoyl chlorides of the formula CI-S0 2 NR R b may be prepared, in turn, from amines of the formula HNR R b according to the procedures reviewed by W. R. Bowman and R. J. Marmon in "Comprehensive Organic Functional Group Transformations, Volume 2", Pergamon (1995).

The compounds of formula I, wherein p is 2 , X is NH, and Y is N R R b may also be obtained directly from an amine of formula XII (or salt thereof). Thus, in Scheme 3, Step 5, the amine of formula XII(or salt thereof) is treated with a sulfamoyl chloride of the formula Cl- S0 2 NR R b and a base such as triethylamine or diisopropylethylamine as described for Scheme 1, Step 4. Amines of formula XII are obtained as described for Scheme 2. Amines of formula XII (or salts thereof) may be obtained by the removal of the arylsulfonyl protecting group PG 1 from a compound of the formula VI (refer to Scheme 1). The deprotection may be carried out by one of the two general deprotection methods described for Scheme 1, Step 5. The choice of deprotection method is determined by the compatibility of the conditions with other functional groups on the molecule.

REVERSE SULFONAMIDES

Compounds of formula I, wherein p is 2, X is CH 2 , and Y is NR R b , may be prepared according to Scheme 4. Scheme 4

In Scheme 4, Step 1, a compound of formula III (refer to Scheme 1), is combined with an amino alcohol of formula XV in the presence of a base and a polar solvent to afford a com ¬ pound of formula XVI. Su itable bases include triethylamine and diisopropylethylamine while suitable solvents include methanol , diisopropyl alcohol and acetone. The reaction is typically run at about 23°C to about 70°C. Preferably, a catalytic amount (about 1 mole %) of potassium iodide is added to the reaction.

In Scheme 4, Step 2 , the compound of formula XVI is converted to a compound of formula XVII, wherein LG is a leaving group such as bromo, iodo, methanesulfonate or, preferably, pora-toluenesulfonate. Methods for installing such leaving groups are well-known to those skilled in the art and have been described in texts such as "Advanced Organic Chemistry" by j. March, John Wiley & Sons (1985). In the case where LG is p ra-toluenesulfonate, the compound of formula XVI is treated with pora-toluenesulfonyl chloride in the presence of a base such as triethylamine, diisopropylethylamine or Ν,Ν-dimethylaminopyridine in an aprotic solvent such as dichloromethane or tetrahydrofuran. The reaction is run at a temperature of about - 10°C to about 40°C, preferably beginning at around 0°C and allowing the reaction to warm to about 23°C.

In Scheme 4, Step 3, the compound of formula XVII is combined with a salt of thioacetic acid, preferably potassium thioacetate to yield a thioester derivative of formula XVIII. The reac- tion is carried out in a polar solvent such as Ν,Ν-dimethylformamide or N-methylpyrrol idine, at a temperature of about 23°C to about 80°C, preferably at about 55 °C.

In Scheme 4, Step 4, the thioester derivative of formula XVIII is converted to a sulfonic acid derivative of formula XIX by reaction with an aqueous solution of hydrogen peroxide, typically 30% by weight. The reaction is carried out in an acidic solvent such as formic or acetic acid at a temperature from about 0°C to about 40°C, preferably at about 23°C.

In Scheme 4, Step 5, the sulfonic acid derivative of formula XIX is converted to a sulfonyl chloride derivative of formula XX. Several methods for carrying out this functional group transformation are known in the literature. The preferred method is to treat the compound of formula XIX with an excess (3- 15 equivalents) of thionyl chloride in the presence of a catalytic amount of N, N-dimethylformamide in an aprotic solvent such as dichloromethane or chloroform. The reaction may be run from about minus 20°C to about 100°C, preferably beginning the reaction at about 0°C, and then warming to about 75°C.

Alternatively, in Scheme 4, Step 6, the thioester derivative of formula XVIII may be directly converted to the sulfonyl chloride derivative of formula XX by treatment with a chlorinat- ing agent. Several methods for carrying out this functional group transformation are known in the literature. Chlorinating agents include chlori ne gas and N-chlorosuccinimide, and the reaction is commonly run in the presence of an acid such as hydrochloric acid or acetic acid . Mixed aqueous solvents systems are often used, such as water and dichloromethane and water and acetonitrile. In Scheme 4, Step 7, the sulfonyl chloride derivative of formula XX is combined with 1- 3 equivalents of an amine of the formula HNR R b to form a sulfonamide derivative of formula XXI. The reaction is run in the presence of at least one equivalent of a base such as triethyla- mine or diiosopropylethylamine at a temperature from about minus 20°C to about 50°C, preferably starting the reaction at about 0°C and allowing the reaction to warm to about 23°C. The reaction is run in an aprotic solvent such as tetrahydrofuran or dichloromethane.

Finally in Scheme 4, Step 8, the arylsulfonyl protecting group PG 1 is removed to provide a compound of the formula I, wherein p is 2 , X is CH 2 , and Y is N R R b . The reaction may be carried out by one of the two general deprotection methods described for Scheme 1, Step 5. The choice of deprotection method is determined by the compatibi lity of the conditions with other functional groups on the molecule. Amino alcohols of formula XV are known in the chemical literature or may be prepared by methods well known to one skilled in the art.

SULFONES, Sulfoxides and Thioethers

Compounds of formula I, wherein p is 0, 1, or 2 , X is CH 2 , Y is AR 5 and A is a bond , may be prepared according to Scheme 5.

Scheme 5

In Scheme 5, Step 1, a compound of formula XVII (refer to Scheme 4) is treated with 1- 2 equivalents of a thiol of the formula R 5 SH in the presence of 1-2 equivalents of a base to give a sulfide of the formula XXIII. Suitable bases include sodium hydride, sodium

bis(trimethylsilyl)amide, l, 5-diazabicyclo[4.3.0]non-5-ene (DBN) and preferably, 1,8- diazabicyclo[5.4.0]undec-7-ene (DBU). The reaction is carried out in a solvent such as N ,N- dimethylformamide or N-methylpyrrol idinone at a temperature from about 0°C to about 50°C, preferably at about 23°C.

In Scheme 5, Step 2 , the compound of formula XXIII is deprotected , removing the arylsulfonyl protecting group PG 1 to provide a compound of formula XXIV. The reaction may be carried out by one of the two general deprotection methods described for Scheme 1, Step 5. The choice of deprotection method is determined by the compatibility of the conditions with other functional groups on the molecule. In Scheme 5, Step 3, a sulfide of formula XXIV is oxidized to yield a sulfone of formula I, wherein p is 2 , X is CH 2 , Y is AR 5 and A is a bond. Several methods are known in the literature and all involve the use of an oxidant such as meta-chloroperbenzoic acid , hydrogen peroxide, or potassium peroxymonosulfate (Oxone * ). A preferred method is to treat the compound of formula XXIV with 2 equivalents of potassium peroxymonosulfate (Oxone * ) in a solvent mixture of tetrahydrofuran, ethanol and water at a temperature of about 23°C. The sulfide of formula XXIV may also be oxidized under milder conditions, for example using 1 equivalent of meta- chlorobenzoic acid in a solvent such as dichloromethane at about 0°C to produce a sulfoxide of formula I, wherein p is 1, X is CH 2 , Y is AR 5 and A is a bond

It is noted that the order of Steps 2 and 3 in Scheme 5, may optionally be reversed such that the oxidation step is carried out prior to the deprotection step.

Compounds of the formula I, wherein p is 0, X is CH 2 , Y is AR 5 and A is a bond are prepared in Scheme 5, Step 4 by removing the arylsulfonyl protecting group PG 1 from a compound of formula XXIII. The reaction may be carried out by one of the two general deprotection meth- ods described for Scheme 1, Step 5. Again, the choice of deprotection method is determined by the compatibility of the conditions with other functional groups on the molecule.

In Scheme 5, Step 5, compounds of the formula XXIII are alternatively prepared from a thioacetate derivative of formula XVIII. First, the thioacetate of formula XVIII is dissolved in a solvent such as ethanol, methanol , or water (or a mixture thereof). A suitable base such as po- tassium carbonate or cesium carbonate (about 2 equivalents) is added and nitrogen is bubbled through the solution to remove oxygen. An alkylating agent of the formula R 5 -LG is then added, wherein LG is a leaving group such as bromo, iodo, methanesulfonate or, para-toluene- sulfonate. The reaction is conducted at a temperature from about minus 20°C to about 30°C. Preferably, the reaction is started at about 0°C and then allowed to warm to about 23°C.

Many thiols of the formula R 5 SH and alkylating agents of the formula R 5 -LG may be obtained from commercial sources. Also, several methods exist for the preparation of such compounds, which are well known to those skilled in the art and have been described in texts such as "Advanced Organic Chemistry" by J. March, John Wiley & Sons (1985).

It is noted that certain compounds of the invention can be obtained by functional group transformations at a late stage of the synthesis, for example, by chemical mod ification of the groups R 4 or R 5 after carrying out Steps 4 or 5 in Scheme 1, Step 3 in Scheme 2 , Steps 2, 3 or 4 in Scheme 3, Steps 7 or 8 Scheme 4 and Steps 2, 3, 4 or 5 in Scheme 5. Such functional group transformations may include one step or multiple steps, for example, reduction of an ester to an alcohol, reoxidation to an aldehyde, addition of an organomagesium reagent to form a sec- ondary alcohol, reoxidation to a ketone and, finally, addition of an organomagesium reagent to yield a tertiary alcohol.

In executing the synthesis of the compounds of the invention, one skilled in the art will recognize the need to sample and assay reaction mixtures prior to work up in order to monitor the progress of reactions and decide whether the reaction should be conti nued or whether it is ready to be worked up to obtain the desired prod uct. Common methods for assaying reaction mixtures include thin-layer chromatography (TLC), liquid chromatography/mass spectroscopy (LCMS), and nuclear magnetic resonance (NMR).

One skilled in the art will also recognize that the compounds of the invention may be prepared as mixtures of diastereomers or geometric isomers (e.g., cis and trans substitution on a cycloalkane ring). These isomers can be separated by standard chromatographic techniques, such as normal phase chromatography on silica gel, reverse phase preparative high pressure liquid chromatography or supercritical fluid chromatography. One skilled in the art will also recognize that some compounds of the invention are chiral and thus may be prepared as race- mic or scalemic mixtures of enantiomers. Several methods are available and are well known to those skilled in the art for the separation of enantiomers. A preferred method for the routine separation enantiomers is supercritical fluid chromatography employing a chiral stationary phase.

EXPERIMENTAL SECTION

Except where otherwise noted, reactions were run under an atmosphere of nitrogen. Chromatography on silica gel was carried out using 250-400 mesh silica gel using pressurized nitrogen (~10- 15 psi) to drive solvent through the column ("flash chromatography"). Where indicated, solutions and reaction mixtures were concentrated by rotary evaporation under vacuum.

Example 1: 2,2,2-Trifluoro-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin- 4- yl)amino]cyclobutyl}ethanesulfonamide

Step 1: Benzyl [c s-3-(methy\am\no)cyc\obuty\]carbamate and benzyl [trans-3- (methy\am no)cyc\obuty\]carbamate

A 33% solution of methylamine (1000 mL, 9.13 mol) in absolute ethanol was added to a mixture of benzyl (3-oxocyclobutyl)carbamate (WO2012/75381 Al and WO2012/09678 Al) (200 g, 0.913 mol) and acetic acid (88 mL) stirring in ethanol (1000 mL) at 0°C. The reaction mixture stirred for at 0 C C for 1.5 hours and then stirred at room temperature for 2 hours. Lithium borohydride (41 g, 2.05 mol) was added in portions to the reaction mixture at -70°C. After addition was complete, the reaction mixture was stirred at -70°C for 1 hour and then allowed to warm to room temperature over 12 hours. The reaction mixture was quenched with water (400 mL), and concentrated under vacuum to remove ethanol. The aqueous layer was acidified with concentrated hyd rochloric acid to pH 2, washed with ethyl acetate (2 x 1000 mL), basified with 10 % sodium hydroxide to pH 9- 10 and then extracted with dichloromethane (3 x 1000 mL). The combined organic layers were washed with brine (1000 mL), dried over sodium sulfate, and concentrated to obtain the crude product as a pale brown liquid. This was dissolved in dichloromethane (400 mL) and cooled to 0°C. To the resulting solution was added a solution of 4M HCI in dioxane (300 mL). The mixture was stirred at 0°C for 30 minutes, and then at room temperature for 12 hours. The reaction mixture was filtered and the remaining solid was re- crystallized from a mixtu re of methanol and methyl fert-butyl ether to afford the cis-isomer as a white solid (111.09 g , 52%). Ή NMR: (400 MHz, D 2 0): δ 7.33-7.38 (m, 5 H); 5.02 (s, 2H), 3.83- 3.87 (m, 1H), 3.89-3.41 (m, 1H), 2.66-2.70 (m, 2H), 2.56 (s, 3H), 2.03-2.05 (m, 2 H). LC/MS (exact mass) calculated for C i3 H ig N 2 0 2 ; 234.137, found (M + H + ); 235.1.

The trans isomer was isolated from the mother liquor using supercritical fluid chroma- tography.

Step 2 : Benzyl {cis-3-[(2-chloro-7H-pyrrolo[2,3-d]pyrimidin-4-yl)(methyl)am ino]- cyclobuty\}carbamate

To a solution of potassium carbonate (20.47 g, 148 mmol) in water (180 mL) was added benzyl [cis-3-(methylamino)cyclobutyl]carbamate (13.57 g, 50.2 mmol), followed by 2,4- dichloro-7H-pyrrolo(2,3-d)pyrimidine (9.0 g, 47.9 mmol) at room temperature. After addition was complete, the reaction mixture was stirred at 95°C overnight. The mixture was filtered to collect the solid. The filter cake was washed with water and dried under vacuum to afford the title compound (16.5 g, 89.7%) as a yellow solid. Ή NMR (400 MHz, DMSO-d ): δ 11.81 (sm 1 H), 7.65 (d, 1 H), 7.38 (m, 5 H), 7.16 (m, 1 H), 6.67 (d, 1 H), 5.02 (s, 2 H), 4.81 (m, 1 H), 3.85 (m, 1 H), 3.25 (s, 3 H), 2.53 (m, 2 H), 2.25 (m, 2 H). LC/MS (exact mass) calculated for

C i9 H 20 CIN 5 O 2 ; 385.131, found (M + H + ); 386.1.

Step 3 : cis-N-Methyl-N-7H-pyrrolo[2,3-d]pyrimidin-4-ylcyclobutcme- l , 3-diamm ' e hydrochloride

A mixture of {cis-3-[(2-chloro-7H-pyrrolo[2 ,3-d] pyrimidin-4-yl)(methyl)- amino]cyclobutyl}carbamate (13.0 g, 34.0 mmol), Pd(OH) 2 (40.3 g, 40.8 mmol) and cyclohexene (72.5 mL, 0.71 mol) in ethanol (300 mL) was stirred at reflux for 3 hours. The reaction mixture was filtered through a pad of Celite * and the pad was washed with methanol. The filtrate was concentrated under vacu um to afford the title compound (4.8 g, 66%) as a white solid. Ή N MR (400 MHz, DMSO-d 6 ): δ 11.68 (br, 1H), 8.11 (s, 1H), 7.67 (br, 2H), 7.17 (d, 1H), 6.65 (d, 1H), 5.08 (m, 1 H), 3.45 (m, 1 H), 3.26 (s, 3H), 2.31 (m, 4H). LC/ MS (exact mass) calculated for C n H i5 N 5 ; 217.133, found (M + H + ); 218.1.

Step 4: 2,2,2-Trifluoro^-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4 -yl)amino]- cyclobutyl}et anesulfonamide

To a solution of cis-N-methyl-N-7H-pyrrolo[2,3-d]pyrimidin-4-ylcyclobutane-l, 3- diamine hydrochloride (100 mg, 0.39 mmol) in tetrahydrofuran (0.8 mL) was added lithium bis(trimethylsilyl)amide (1M solution in tetrahydrofuran) (0.9 mL, 0.9 mmol) and

chlorotrimethylsilane (94 mg, 0.88 mmol) at room temperature. The reaction mixture was stirred for 45 minutes and then 2,2,2-trifluoroethanesulfonyl chloride (86 mg, 0.47 mmol) was added slowly. The mixture was stirred at room temperature for 18 hours and then partitioned between dichloromethane and water. The aqueous layer was and extracted twice with dichloromethane and the combined organic layers were concentrated to afford the crude product as a tan solid. The crude material was purified by chromatography on silica gel eluting with a mixture of dichloromethane and methanol (93: 7) to afford the title compound as a white solid (93 mg, 65%). Ή NMR (400 MHz, DMSO-d 6 ): δ 11.61 (br. s. , 1 H), 8.20 (d, 1 H), 8.08 (s, 1 H), 7.13 (d, 1 H), 6.60 (d, 1 H), 4.80-4.94 (m, 1 H), 4.34 (q, 2 H), 3.58-3.71 (m, 1 H), 3.23 (s, 3 H), 2.55-2.67 (m, 2 H), 2.17-2.30 (m, 2 H). LC/MS (exact mass) calculated for C 13 H 16 F 3 N 5 0 2 S;

363.098, found (M + H + ); 363.9. The following compounds, Examples 2-7, were prepared from cis-N-methyl-N-7H- pyrrolo[2 ,3-d]pyrimidin-4-ylcyclobutane- l,3-diamine hydrochloride (Example 1, Step 3) in a similar manner to that described in Example 1, Step 4, substituting the indicated sulfonyl chloride for 2,2,2-trifluoroethanesulfonyl chloride. Example 2 : N-{cis-3-[Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclo butyl}-p pane- l - sulfonamide

This compound was prepared using 1-propanesulfonyl chloride. The crude compound was purified by chromatography on silica gel eluting with a mixture of dichloromethane and methanol (93 : 7) to afford the title compound as a tan sol id (78% yield). Ή NMR (400 MHz, DMSO-d 6 ): δ 11.60 (br s, 1 H), 8.08 (s, 1 H), 7.46 (d, 1 H), 7.12 (d, 1 H), 6.61 (d, 1 H), 4.81-4.94 (m, 1 H), 3.47-3.62 (m, 1 H), 3.23 (s, 3 H), 2.87-2.96 (m, 2 H), 2.52-2.63 (m, 2 H), 2.14-2.27 (m, 2 H) 1.60- 1.73 (m, 2 H) 0.96 (t, 3 H). LC/MS (exact mass) calculated for C 14 H 21 N 5 0 2 S;

323.142, found (M + H + ); 324.1. Example 3: 2-Methyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)am ino]- cyclobutyljpropane- 1 -sulfonamide

This compound was prepared using 2-methyl- lpropanesulfonyl chloride. The crude compound was purified by chromatography on si lica gel eluting with a mixture of dichloro- methane and methanol (93 : 7) to afford the title compound as a white solid (52%). H NMR (400 MHz, DMSO-d 6 ): δ 11.64 (br s, 1 H), 8.12 (s, 1 H), 7.51 (d , 1 H), 7.03-7.26 (m, 1 H), 6.65 (d, 1 H), 4.82-5.02 (m, 1 H), 3.52-3.70 (m, 1 H), 3.26 (s, 3 H), 2.87 (d, 2 H), 2.55-2.67 (m, 2 H), 2.18-2.30 (m, 2 H), 2.11 (dt, 1 H), 1.04 (d, 6 H). LC/MS (exact mass) calculated for C 15 H 23 N 5 0 2 S; 337.157, found (M + H + ); 338.0.

Example 4A and Example 4B: cis- and trans-3-(Cyanomethyl)-N-{cis-3-[methyl(7H-pyrrolo[2,3- d]pyrimidin-4-yl)amino]cyclobutyl}cyclobutanesulfonamide

These compounds were prepared using a mixture (-1: 1) of cis- and trans-3- (cyanomethyl)cyclobutanesulfonyl chloride. The crude mixture of cis and trans isomers was pu- rified by chromatog raphy on silica gel eluting with a gradient of dichloromethane and methanol (100:0 to 10: 1) to afford a mixture (420 mg) of the title compounds as a white solid (67%). The cis and trans isomers were separated by supercritical fluid chromatography,

cis-isomer 4A: 160 mg (21%). Ή N M R (400 MHz, methanol-d 4 ): δ 8.12 (s, 1H), 7.13-7.12 (d, 1H), 6.69-6.69 (d, 1 H), 4.92-4.89 (m, 1 H), 3.84-3.78 (m, 1 H), 3.76-3.67 (m, 1 H), 3.36 (s, 3 H), 2.79-2.73 (m, 2 H), 2.65-2.64 (m, 3H), 2.58-2.52 (m, 2 H), 2.32-2.19 (m, 4 H). LC/MS (exact mass) calculated for C i? H 22 N 6 0 2 S; 374.152, found (M + H + ); 375.3.

trans-isomer 4B: 155 mg (20%). Ή NMR (400 MHz, methanol-d 4 ): δ 8.13 (s, 1H), 7.13 (d, 1H), 6.70 (d , 1H), 4.94-4.89 (m, 1H), 3.89-3.85 (m, 1H), 3.72-3.69 (m, 1H), 3.36 (s, 3H), 2.85-2.62 (m, 7H), 2.31-2.23 (m, 4H). LC/MS (exact mass) calculated for C 17 H 22 N 6 0 2 S; 374.152, found (M + H + ); 374.9.

The mixture of cis- and trans-3-(cyanomethyl)cyclobutanesulfonyl chlorides was prepared as follows:

Step 1: [3-(Benzyloxy)cyclobutylidene]acetonithle

To a cold suspension of sodium hydride (125 mg, 3.12 mmol) in tetrahydrofuran (12 mL) at 0°C was added diethyl cyanomethylphosphonate (1.21 g, 3.40 mmol). The mixture was stirred at room temperature for 1 hour before adding a solution of 3-(benzyloxy)cyclobutanone (500 mg, 2.84 mmol) in tetrahydrofuran (8 mL). The mixture was stirred at room temperature overnight, and was then quenched with water. The mixture was extracted with ethyl acetate (3 x 25 mL) and the combined organic layers were dried over sodium sulfate and concentrated. The residue was chromatographed on silica gel eluting with a gradient of petroleum ether and ethyl acetate (100:0 to 85 : 15) to afford the title compound (450 mg, 80%) as a yellow oil.

Step 2 : [3-(Benzyloxy)cyclobutyl]acetonitrile

A mixture of [3-(benzyloxy)cyclobutylidene]acetonitrile (10.2 g, 51 mmol,) and 10% Pd/C (2.0 g) in dry tetrahydrofuran was pressurized to 50 psi with hydrogen and stirred at room temperature for 3 days. The mixture was then filtered and concentrated under vacuum. The residue was chromatographed on silica gel eluting with a gradient of petroleum ether and ethyl acetate (100:0 to 80:20) to give the title compound (7 g, 70%) as a colorless οΜ. Ή NMR (400 MHz, CDCI 3 ): δ 7.36-7.28 (m, 5 H), 4.44-4.43 (m, 2 H), 4.30-4.09 (m, 1H), 3.98-3.95 (m, 1 H), 2.64-2.45 (m, 4 H), 1.81- 1.759 (m, 2 H).

Step 3 : (3-Hydroxycyclobutyl)acetonitrile

To a solution of [3-(benzyloxy)cyclobutyl]acetonitrile (1 g, 5.00 mmol) in acetonitrile (15 mL) was added d ropwise iodotrimethylsilane (1.5 g, 7.50 mmol) at 0°C. The mixture was sti rred at room temperature overnight. The mixture was quenched with triethylamine, concentrated and then purified by chromatography on si lica gel eluting with a gradient of petroleum ether and ethyl acetate (1 :0 to 1: 1) to afford the title compound (340 mg, 62%) as yellow oil . Ή N M R (400 MHz, CDCI 3 ): δ 4.55-4.15 (m, 1H), 2.49-2.46 (m, 2 H), 2.25-2.21 (m, 2H), 2.14-2.08 (m, 1H), 1.79- 1.72 (m, 2H).

Step 4: 3-(Cyanomethyl)cyclobutyl-4-methylbenzenesulfonate

To a solution of (3-hydroxycyclobutyl)acetonitrile (333 mg , 3.0 mmol) in dry dichloro- methane (25 mL) was added 4-di methylaminopyridine (732 mg, 6.0 mmol). The mixture stirred at room temperature for 5 minutes and then p-toluenesulfonyl chloride (859 mg, 4.5 mmol) was added. The resulting mixture was stirred at room temperature overnight. The mixture was washed with water (2 x 15 mL). The organic layer was dried over sodium sulfate and concentrat- ed. The residue was purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (10:0 to 7:3) to afford the title compound (520 mg, 65% yield) as colorless oil.

Step 5 : S-[3-(Cyanomethyl)cyclobutyl] ethanethioate

The mixture of 3-(cyanomethyl)cyclobutyl 4-methylbenzenesulfonate (1.5 g, 5.7 mmol) and potassiu m thioacetate (1.29 g, 3.00 mmol) in N,N-dimethylformamide (8 mL) was heated at 80°C overnight. The mixture was diluted with ethyl acetate (15 mL), washed with water (30 mL) and brine (2 x 30 mL), dried over sodium sulfate and concentrated. The residue was purified by preparative thin layer chromatography eluting with a mixture of petroleum ether and ethyl acetate (3 : 1) to afford the title compound (750 mg, 78%) as colorless oil . Ή N MR (400 MHz, CDCI ): δ 4.12-3.92 (m, 1 H), 2.86-2.77 (m, 2 H), 2.71-2.47 (m, 2 H), 2.42-2.37 (m, 2 H), 2.30-2.29 (m,3 H), 1.97- 1.90 (m, 1 H).

Step 6: 3-(Cyanomethyl)cyclobutanesulfoiiyl chloride

A mixture of N-chlorosuccinimide (1.6 g, 12.0 mmol) in concentrated HCI (3 mL) and acetonitrile (12 mL) was stirred at room temperature for 10 minutes. S-[3-

(cyanomethyl)cyclobutyl] ethanethioate (507 mg, 3.0 mmol) in acetonitrile (3 mL) was added at 0°C and stirred for 10 minutes. The mixture was diluted with aqueous sod ium bicarbonate (50 mL), and extracted with methyl tert-butyl ether (3 x 50 mL). The combined dried organic layers were dried over anhydrous sodium sulfate and concentrated. The crude product was purified by chromatography on silica gel eluting with a mixture of petroleum ether and ethyl acetate (100:0 to 50: 50) to afford the title compound (400 mg, 69%) as a yellow oil. Ή NMR (400 MHz, CDCI 3 ): δ 4.45-4.40 (m, 1H), 3.06-2.71 (m, 3H), 2.61-2.49 (m, 4H).

Example 5: / -[3-(Cyanomethyl)oxetan-3-yl]-N-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4- yl)amino]cyclobutyl}methanesulfonamide

This compound was prepared from [3-(cyanomethyl)oxetan-3-yl]methanesulfonyl chloride. The crude compound was purified using preparative thin layer chromatography eluting with ethyl acetate to afford the title compound as a white solid (32%). Ή N MR (400 MHz, methanol^): δ 8.13 (s, 1 H), 7.14-7.13 (m, 1 H), 6.71-6.70 (m, 1 H), 5.06-5.05 (m, 1 H), 4.85-4.81 (m, 2 H), 4.52-4.50 (m, 2 H), 3.77-3.75 (m, 1 H), 3.63 (m, 2 H), 3.39 (s, 3 H), 3.29-3.26 (m, 2 H), 2.85-2.78 (m, 2 H), 2.38-2.30 (m, 2 H). LC/MS (exact mass) calculated for C 1? H 22 N 6 0 3 S; 390.147, found (M + H + ); 391.0.

[3-(Cyano ethyl)oxet n-3-yl]methanesulfonyl chloride

Step 1: [3-(Cyano ethyl)oxetan-3-yl]methyl 4-methylbenzenesulfonate

This compound was prepared following the procedure of Example 4 Step 4, substituting

[3-(Hydroxymethyl)-3-oxetanyl]acetonitrile for (3-hydroxycyclobutyl)acetonitrile. The crude compound was purified by chromatography on si lica gel eluting with a mixture of petroleum ether and ethyl acetate (1:0 to 1 : 1) to afford the title compound as a white solid (10%). Ή NM R (400 MHz, CDCI 3 ): δ 7.82-7.80 (m, 2 H), 7.41-7.39 (m, 2 H), 4.54-4.35 (m, 4 H), 4.31 (s, 2 H), 2.79 (s, 2 H), 2.45 (s, 3H).

Step 2 : [3-(Cyanomethyl)oxetan-3-yl]methyl thiocyanate

A solution of [3-(cyanomethyl)oxetan-3-yl]methyl 4-methylbenzenesulfonate (150 mg, 0.53 mmol) and potassium thiocyanate (104 mg, 1.07 mmol) was stirred i n ethanol (10 mL). The reaction was heated to 85°C and stirred for 16 hours. The solvent was evaporated to afford the crude title compound as a white solid .

Step 3 : [3-(Cyanomethyl)oxetan-3-yl]methanesulfonyl chloride

Chlorine gas was bubbled through a solution of [3-(cyanomethyl)oxetan-3-yl]methyl thiocyanate (0.53 mmol, crude) in water (10 mL) at 0°C for 30 minutes. The reaction mixture was extracted with methyl tert-butyl ether (2 x 20 mL). The combined organic layers were dried over sodium sulfate and concentrated to afford the title compound (20 mg, 18%).

Example 6: N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclo butyl}- l -oxetan-3- ylmethanesulfonamide

This compound was prepared using oxetan-3-ylmethanesulfonyl chloride. The crude compound was purified by chromatography on si lica gel eluting with a mixture of dichloro- methane and methanol (85 : 15) to afford the title compound as a white sol id (23%). Ή NMR (400 MHz, methanol-d 4 ): δ 8.13 (m, 1 H), 7.13 (d, J = 4Hz, 1 H), 6.70-6.69 (m, J = 4Hz, 1 H), 4.93- 4.91 (m, l H), 4.84-4.83 (m, 2 H), 4.63-4.59 (m, 2 H), 3.74-3.68 (m, 1 H), 3.58-3.56 (m, 1 H), 3.47-3.45 (m, 2 H), 3.37 (s, 3 H), 2.79-2.77 (m, 2 H), 2.32-2.29 (m, 2 H). LC/MS (exact mass) calculated for C i5 H 2i N 50 3 S; 351.136, found (M + H + ); 352.1.

Oxetan-3-ylmethanesulfonyl chloride

Step 1: Oxetan-3-ylmethyl thiocyanate

This compound was prepared according to the procedure of Example 5 , Step 2, substituting oxetan-3-ylmethyl 4-methylbenzenesulfonate (WO2012/ 117000A1) for [3- (cyanomethyl)oxetan-3-yl] methyl 4-methylbenzenesulfonate to afford the crude title compound as a white solid . (100%).

Step 2 : Oxetan-3-ylmethanesulfonyl chloride

This compound was prepared in crude form (25% yield) following the procedure of Example 5 Step 3, substituting oxetan-3-ylmethyl thiocyanate for [3-(cyanomethyl)oxetan-3- yl]methyl thiocyanate.

Example 7A and 7B: cis- and trans-3-(Cyanomethyl)-3-methyl-N-{cis-3-[methyl(7H-pyrrolo[2 , 3- d]pyrimidin-4-yl)amino]cyclobutyl}cyclobutanesulfonamide

These compounds were prepared using a mixture (~1: 1) of cis- and trans-3- (cyanomethyl)-3-methylcyclobutanesulfonyl chloride. The crude mixture of cis- and trans iso- mers was purified by chromatography on si lica gel eluting with a gradient of petroleum ether:ethyl acetate (10: 1 to 1: 15) to afford a mixture (70 mg) of the title compounds as a light brown solid (28%). The cis and trans isomers were then separated by supercritical fluid chromatography (SFC).

cis-isomer (7A): 26 mg (10%); SFC retention time = 7.11 minutes; Ή NMR (400 MHz, methanol- d 4 ): δ 8.13 (s, 1 H), 7.13-7.13 (d , 1 H), 6.69 (d, 1 H), 4.93-4.86 (m, 1 H), 3.91-3.87 (m, 1 H),

3.71- 3.65 (m, 1 H), 3.37-3.33 (m, 3 H), 2.77-2.75 (m, 2 H), 2.68 (s, 2 H), 2.41-2.36 (m, 2 H), 2.26-2.21 (m, 2 H), 1.34 (m, 3 H). LC/MS (exact mass) calculated for C 1S H 24 N 6 0 2 S; 388.168, found (M + H + ); 389.1.

trans-isomer (7B) 24 mg (10%); SFC retention time = 11.35 minutes; Ή NMR (400 MHz, methanol^): δ 8.13 (s, 1 H), 7.14 (d, 1 H), 6.69 (d, 1 H), 4.93-4.86 (m, 1 H), 3.96-3.86 (m, 1 H),

3.72- 3.65 (m, 1 H), 3.36-3.31 (m, 3 H), 2.77-2.75 (m, 2 H), 2.71 (s, 2 H), 2.34-2.26 (m, 6 H), 1.33 (m, 3 H). LC/MS (exact mass) calculated for C i8 H 24 N 6 0 2 S; 388.168, found (M + H + ); 389.0.

The mixture of cis- and trans-3-(cyanomethyl)-3-methylcyclobutanesulfonyl chlorides was prepared as follows:

Step 1: 1 -Methyl-3-methylenecyclobut mecarbonitrile

To a solution of 3-methylenecyclobutanecarbonitrile (35.0 g, 373.0 mmol) in tetrahydrofuran (200 mL) was added dropwise lith ium bis(tri methylsilyl)amide (450 mL, 1M) at - 78°C. The solution was stirred for 1 hour at -78 ' C and iodomethane (30 mL, 448 mmol) was added to the reaction. After 1 hour, the mixture was warmed to room temperature and stirred overnight. The reaction mixture was quenched with aqueous ammonium chloride (380 mL) and extracted with methyl tert-butyl ether (3 x 400 mL). The combined organic layers were dried over sodium sulfate and concentrated. The crude product was purified by distillation under reduced pressure to afford the title compound (20 g, 50%) as clear oil. Ή N MR (400 MHz, CDCI 3 ): δ 4.90-4.89 (m, 2 H), 3.24-3.20 (m, 2 H), 2.67-2.62 (m, 2 H), 1.50 (s, 3 H).

Step 2 : 1 -Methyl-3-methylenecyclobutanecarboxylic acid

To a solution of l-methyl-3-methylenecyclobutanecarbonitrile (10.0 g, 93.3 mmol) in water (50 mL) and ethanol (50 mL) was added potassium hydroxide (25.6 g, 466.6 mmol). The reaction mixture was heated to reflux and stirred overnight. The ethanol was removed under reduced pressure, and the solution was cooled to below IO C, acidified with concentrated hydrochloric acid to pH 1. The aqueous phase was extracted with ethyl acetate (3 x 150 mL). The combined organic layers were dried over sodium sulfate and concentrated to afford the title compound (9 g, 77%). Ή NM R (400 MHz, CDCI 3 ): δ 11.90 (s, 1 H), 4.88-4.85 (m, 2 H), 3.23- 3.17 (m, 2 H), 2.53-2.41 (m, 2 H), 1.45 (s, 3 H). Step 3 : Ethyl 1 -methyl-3-tnethylenecyclobutaneccirboxylate

To a solution of l-methyl-3-methylenecyclobutanecarboxylic acid (6 g , 47.6 mmol) in dichloromethane (30 mL) at 0°C was added dropwise thionyl chloride (11.0 mL, 143 mmol). The solution was stirred at 0°C for 1 hour. Three drops of Ν,Ν-dimethylformamide were added to the solution. The solution was stirred at 0 ° C for 30 minutes. The solvent was evaporated and dichloromethane (20 mL) and ethanol (125 mL) were added to the residue. The resulting solution was stirred for 16 hours at room temperature. The solvent was evaporated and water (20 mL) was added to the residue. The aqueous layer was extracted with dichloromethane (4 x 20 mL). The combined organic layers were dried over sodium sulfate and concentrated. The crude product was purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (20: 1 to 10: 1) to afford the title compound (5 g, 68%). Ή NMR (400 MHz, CDCI 3 ): δ 4.85-4.83 (m, 2 H), 4.17-4.12 (m, 2 H), 3.18-3.12 (m, 2 H), 2.48-2.42 (m, 2 H), 1.41 (s, 3 H), 1.27- 1.23 (m, 3 H).

Step 4: (1 -Methyl-3-methylenecyclobutyl)methanol

A mixture of ethyl l-methyl-3-methylenecyclobutanecarboxylate (4.55 g, 29.5 mmol) lithium aluminum hydride (2.8 g, 72 mmol) in tetrahydrofuran (50 mL) was stirred overnight at room temperature. To the reaction mixture was added Na 2 SO 4 - 10H 2 O (3.7 g, 11.5 mmol) and the resulting mixture was stirred for 1 hour at room temperature. The solids were removed by filtration and the filtrate was concentrated under vacuum. The residue was extracted with di- chloromethane (3 x 50 mL). The combined organic extracts were dried over sodium sulfate and concentrated to afford the title compound (2.6 g, 79%) as a colorless oil. Ή NMR (400 MHz, CDCI 3 ): δ 4.79-4.78 (m, 2 H), 3.48 (s, 2 H), 2.53-2.48 (m, 2 H), 2.36-2.27 (m, 2 H), 1.16 (s, 3 H).

Step 5 : (1 -Methyl-3-methylenecyclobutyl)methyl 4-methylbenzenesulfonate

This compound was prepared following Example 4, Step 4, substituting (l-methyl-3- methylenecyclobutyl)methanol for (3-hydroxycyclobutyl)acetonitrile. The crude compound was purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (20: 1 to 4: 1) to afford the title compound (70%). Ή NM R (400 MHz, CDCI 3 ): δ 7.79 (d, 2 H), 7.34 (d, 2 H), 4.79-4.78 (m, 2 H), 3.90 (s, 2 H), 2.51-2.47 (m, 2 H), 2.44 (s, 3 H), 2.35-2.31 (m, 2 H), 1.15 (s, 3 H).

Step 6: (l -Methyl-3-methylenecyclobutyl)acetonitrile

A mixture of (l-methyl-3-methylenecyclobutyl)methyl 4-methylbenzenesulfonate (2.5 g, 9.4 mmol), potassiu m cyanide (1.3 g, 19 mmol) and Ν,Ν-di methylformamide (8mL) was stirred overnight at 70°C. Water (10 mL) and methyl tert-butyl ether (20 mL) were added to the mixture and the organic layer was separated. The aqueous phase was extracted with methyl tert-butyl ether (3 x 30 mL). The combined organic layers were washed with an aqueous saturated sodium bicarbonate solution (15 mL), dried over sodium sulfate, and concentrated. The crude product was purified by chromatography on silica gel eluting with a gradient of petrole- um ether and ethyl acetate (10: 1 to 5 : 1) to afford the title compound (1.1 g, 97%) as light brown oil. Ή NM R (400 MHz, CDCI 3 ): δ 4.88-4.87 (m, 2 H), 2.62-2.54 (m, 2 H), 2.50 (s, 2 H), 1.33 (s, 3 H).

Step 7: (1 -Methyl-3-oxocyclobutyl)acetonitrHe

Ozone gas was bubbled through a solution of (l-methyl-3-methylenecyclo- butyl)acetonitrile (1.08 g, 8.91 mmol) in dichloromethane (30 mL) -78 C for 10 minutes. After purging the solution with nitrogen gas, dimethylsulfide (10 mL) was added dropwise to the solution at -78°C. The solution was stirred for 30 minutes at -78 C and the solvent was removed under reduced pressure. The crude product was purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (20: 1 to 8: 1) to afford the title com- pound (920 mg , 84%) as a colorless oil. Ή NMR (400 MHz, CDCI 3 ): δ 3.11-3.06 (m, 2 H), 2.96- 2.91 (m, 2 H), 2.69 (s, 2 H), 1.53 (s, 3 H).

Step 8: (3-Hydroxy- l -methylcyclobutyl)acetonitrile

To a solution of (l-methyl-3-oxocyclobutyl)acetonitrile (400 mg, 3.25 mmol) in tetrahydrofuran (15 mL) was added sodium borohydride (246 mg, 6.5 mmol). The mixture was stirred for 3 hours at room temperature. Acetone (2 mL) was added and then the solvent was evaporated. Water (10 mL) was added to the residue and the aqueous phase was extracted with dichloromethane (4 x 15 mL). The combined organic layers were dried over sodium sulfate and concentrated. The crude product was purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (10: 1 to 1: 1) to afford the title compound (300 mg, 74%) as a colorless oil. Ή NMR (400 MHz, CDCI 3 ): δ 4.38-4.34 (m, 2 H), 2.46-2.27 (m, 4 H), 1.94-1.86 (m, 2 H), 1.33- 1.12 (m, 3 H).

Step 9: 3-(Cyanomethyl)-3-methylcyclobutyl 4-methylbenzenesulfonate

This compound was prepared following Example 7, Step 5 , substituting (3-hydroxy- l- methylcyclobutyl)acetonitrile for (l-methyl-3-methylenecyclo-butyl)methanol. The crude com- pound was purified by chromatography on silica gel eluting with a gradient of petroleu m ether and ethyl acetate (20: 1 to 4: 1) to afford the title compound (36%). Ή NMR (400 MHz, CDCI 3 ): δ 7.77 (d , 2 H), 7.35 (d, 2 H), 4.89-4.81 (m, 1 H), 2.45 (s, 3 H), 2.43-2.34 (m, 3 H), 2.26-2.21 (m, 1 H), 2.15-2.11 (m, 2 H), 1.33 (s, 3 H).

Step 10: S-[3-(Cyanomethyl)-3-methylcyclobutyl]ethanethioate This compound was prepared in 89% yield (crude) following the procedure of Example 4, Step 5 , substituting 3-(cyanomethyl)-3-methylcyclobutyl 4-methylbenzenesulfonate for 3- (cyanomethyl)cyclobutyl 4-methylbenzene-sulfonate. Ή NMR (400 MHz, CDCI 3 ): δ 3.12 (s, 1 H), 2.46-2.30 (m, 4 H), 2.19 (s, 2 H), 1.29 (s, 1 H) 1.26- 1.24 (m, 1 H), 1.18- 1.14 (m, 1 H), 1.13 (s, 3H).

Step 11: 3-(Cyanomethyl)-3- ethylcyclobutanesulfonyl chloride

This compound was prepared following Example 4 Step 6, substituting S-[3- (cyanomethyl)-3-methylcyclobutyl] ethanethioate for S-[3-(cyanomethyl)-cyclobutyl] ethanethioate. The crude compound was purified using chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (90: 10 to 30: 70) to afford the title compound as a yellow liquid (66%). Ή NMR (400 MHz, CDCI 3 ): δ 4.45-4.38 (m, 1 H), 2.67-2.55 (m, 4 H), 2.46-2.40 (m, 2 H), 1.42- 1.40 (m, 3 H).

Example 8: 4-Cyano^-{cis-3-[methyl(7H-pyrmlo[2,3-d]pyrimidin-4-yl)amino ]-cyclobutyl}pyridine- 2-sulfonamide

Step 1: 2-(Benzylthio)isonicotinonitrile

A 60% suspension of sodium hydride in mineral oi l (8.36 g, 210.0 mmol) was suspended in tetrahydrofuran (100 mL). A solution of benzyl mercaptan (21.5 g, 173 mmol) in

tetrahydrofuran (50 mL) was then added dropwise. A thick slurry formed during the addition. 4- Cyano-2-chloropyridine (12.5 g, 90.2 mmol) was added and the resulting mixture was stirred for 3 hours at room temperature. After carefully quenching with water, the mixture was partitioned between water and diethyl ether. The ether layer was washed with saturated aqueous sodiu m bicarbonate solution, dried over magnesi um sulfate and concentrated under reduced pressure. Heptane was added to the residue with solids forming rapidly. The solids were col- lected by filtration, washed with heptane, and dried to give (33.02 g, 84%) of the title compound as an off-white solid. Ή NMR (400 MHz, CDCI 3 ): δ 8.61 (d , 1 H), 7.25-7.46 (m, 6 H), 7.16-7.22 (m, 1 H), 4.47 (s, 2 H). LC/MS (exact mass) calculated for C i3 H io N 2 S; 226.056, found (M + H + ); 227.1.

Step 2: 4-cyanopyridine-2-sulfonyl chloride

To a mechanically-stirred mixture of 2-(benzylthio)isonicotinonitrile (8.92 g, 39.4 mmol) in dichloromethane (139 mL) and water (31 mL) was added dropwise sulfuryl chloride (22.5 mL, 278 mmol), keeping the temperature of the mixture below 3°C. After addition was complete, the mixture was stirred for 30 minutes with continued cooling in an ice bath. A slurry of water (50 mL) and ice (20 g) was added. The aqueous phase was extracted twice with di- chloromethane. The combined extracts were dried over magnesium sulfate and concentrated under reduced pressure to afford the crude title compound.

Step 3 : 4-Cyano-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)ami no]cyclobutyl}pyr-idme-2- sulfonamide

A solution of 4-cyanopyridine-2-sulfonyl chloride (9.7 g, 47.9 mmol) in N,N- dimethylformamide (10 mL) was added to a solution of cis-N-methyl-N-7H-pyrrolo[2,3- d]pyrimidin-4-ylcyclobutane- l,3-diamine hydrochloride (8.0 g, 36.8 mmol) and 4- dimethylaminopyridine (150 mg , 0.03 mmol) in N, N-dimethylformamide (90 mL) at room temperature. Diisopropylethylamine (13 mL, 77 mmol) was added and the resulting mixture was stirred at room temperature for 2 hours. The mixture was diluted with ethyl acetate (200 mL) and aqueous saturated sodium bicarbonate solution was added. Water was added to dissolve the precipitated solids. The aqueous phase was extracted three times with ethyl acetate. The combined organic extracts were washed four times with brine, d ried over anhydrous magnesium sulfate and concentrated under reduced pressure. A 1: 1 mixture of ethyl acetate and hex- anes was added to the residue. The solids were collected by filtration and then dissolved in di- chloromethane and a minimum amount of methanol. The resulting solution was passed through a silica gel plug eluting with a 5% solution of methanol in dichloromethane. The solvents were evaporated to afford a solid to which was added a solution of 10% methanol in dichloromethane. The mixture was briefly stirred and then let stand overnight. The solids were filtered, washed with dichloromethane and dried to afford the title compound (5.58 g, 39%) as an off- white solid. Ή NMR (400 MHz, DMSO-d 6 ): δ 11.62 (br. s., 1 H), 9.02 (d, 1 H), 8.52 (d, 1 H), 8.38 (s, 1 H), 8.17 (dd, 1 H), 8.07 (s, 1 H), 7.10-7.15 (m, 1 H), 6.59 (dd, 3.41 Hz, 1 H), 4.80-4.91 (m, 1 H), 3.58-3.71 (m, 1 H), 3.19 (s, 3 H), 2.25-2.36 (m, 2 H), 2.10 (m, 2 H). LC/MS (exact mass) calculated for C 17 H 17 N ? 0 2 S; 383.116, found (M + H + ); 384.1.

Example 9: 3-(l -Hydroxy- 1 -methylethyl)-N-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4- yl)amino]cyclobutyl}benzenesulfonamide

Step 1: Methyl 3-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)ammo]-cyc lobutyl}amino)- sulfonyljbenzoate

To a suspension of cis-N-methyl-N-7H-pyrrolo[2 ,3-d]pyri midin-4-ylcyclobutane- l,3- diamine hydrochloride (1.8 g, 8.29 mmol) in N,N-dimethylformamide (100 mL) was added portionwise triethylamine (6.7 mL, 49 mmol) at 0°C. Methyl 3-(chlorosulfonyl)benzoate (2.3 g, 9.9 mmol) was added at 0°C. The resulting mixture was stirred at room temperature for 3 hours. The solvent was removed under vacuum. The residue was chromatographed on silica gel eluting with a gradient of methanol in dichloromethane (3% to 10%) to afford the title compound (1.6 g, 47%) as a yellow solid.

Step 2 : 3-(Hydroxymethyl)^-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin -4-yl)amino]cyclo- butyl}benzenesulfonamide

To a solution of methyl 3-[({cis-3-[methyl(7H-pyrrolo[2,3-d]pyri midin-4- yl)amino]cyclobutyl}amino)sulfonyl]benzoate (800 mg, 1.92 mmol) in tetrahydrofuran (120 mL) was added lithium aluminum hydride (0.25 g, 6.7 mmol) at 0°C. The reaction was warmed to 25 °C and stirred for 3 hours. The reaction was quenched with water (2 mL) and stirred for 15 minutes. The reaction mixture was filtered. The filter cake was stirred in tetrahydrofuran (50 mL) and filtered again. The combined filtrate was concentrated to dryness to afford the title compound (430 mg, 58%) as a yellow solid.

Step 3 : 3-Formyl-N-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]- cyclobutyl}benzenesulfonamide

To a solution of 3-(hydroxymethyl)-N-{cis-3-[methyl(7H-pyrrolo[2 , 3-d]pyrimidin-4- yl)amino]cyclobutyl}benzenesulfonamide (400 mg, 1.03 mmol) in chloroform (50 mL) and methanol (5 mL) was added manganese dioxide (0.89 g, 10.0 mmol). The reaction mixture was stirred at 25°C overnight. The reaction mixture was filtered and the filter cake was washed with chloroform (3 x 25 mL). The combined filtrates were concentrated. The residue was chromato- graphed on silica gel eluting with a gradient of methanol in dichloromethane (2% to 8%) to af- ford the title compound (240 mg, 60%) as an oil.

Step 4: 3-(l -Hydroxyethyl)-N-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4- yl)amino]cyclobutyl}benzenesulfonamide

To a solution of 3-formyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}benzenesulfonamide (260 mg , 0.68 mmol) in tetrahydrofuran (20 mL) was added methyl magnesium bromide (1.8 mL, 5.4 mmol) at 0°C under nitrogen. The reaction was stirred at 25°C overnight and was then quenched with aqueous ammonium chloride (10 mL). The reaction mixture was extracted with ethyl acetate (3 x25 mL). The combined organic layers were dried over sodium sulfate and concentrated. The residue was purified by preparative high performance liquid chromatography to afford the title compound (60 mg, 22%) as a white solid. Ή NMR (400 MHz, DMSO-d 6 ): δ 11.6 (s, 1 H), 8.08 (s, 1 H), 8.03 (d, 1 H), 7.86 (s, 1 H), 7.70

(m, 1 H), 7.55 (m, 2 H), 7.15 (m, 1 H), 6.61 (m, 1 H), 5.44 (m, 1 H), 4.85 (m, 1 H), 3.56 (m, 1 H), 3.18 (s, 3 H), 2.18 (m, 2 H), 2.04 (m, 2 H), 1.32 (d, 3 H). LC/MS (exact mass) calculated for C H N O S; 401.152, found (M + H + ); 402.2. Step 5 : 3-Acetyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)am ino]- cyclobutyljbenzenesulfonamide

To a solution of 3-(l-hydroxyethyl)-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimid in-4- yl)amino]cyclobutyl}benzenesulfonamide (60 mg, 0.15 mmol) in chloroform (30 mL) and meth- anol (5 mL) was added manganese dioxide (190 mg, 2.2 mmol). The reaction mixture was stirred at 45°C overnight. Then the reaction mixture was filtered and the filter cake was washed with chloroform (3 x 25 mL). The combined filtrates were concentrated. The residue was purified by preparative high performance liquid chromatography to afford the title compound (15 mg, 25%) as a white solid. Ή NMR (400 MHz, DMSO-d 6 ): δ 11.58 (s, 1 H), 8.31 (s, 1 H), 8.21 (m, 2 H), 8.16 (m, 2 H), 7.76 (m, 1 H), 7.09 (d, 1 H), 6.56 (s, 1 H), 4.82 (m, 1 H), 3.54 (m, 1 H), 3.14 (s, 3 H), 2.81 (m, 3 H), 2.26 (m, 2 H), 1.98 (m, 2 H). LC/MS (exact mass) calculated for

C 19 H 21 N 5 0 3 S; 399.136, found (M + H + ); 400.1.

Step 6: 3-(l -Hydroxy- 1 -methylethyl)-N-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4- yl)amino]cyclobutyl}benzenesulfcmamide

To a solution of 3-acetyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyri midin-4- yl)amino]cyclobutyl}benzenesulfonamide (240 mg, 0.58 mmol) in tetrahydrofuran (20 mL) was added methyl magnesium bromide (2.4 mL, 7.2 mmol) at 0°C under nitrogen. The reaction was stirred at 25°C for 2 hours and was quenched with aqueous ammonium chloride solution (10 mL). The reaction mixture was extracted with ethyl acetate (3 x 25 mL). The combined organic layers were dried over sodium sulfate and concentrated . The residue was purified by preparative high performance liq uid chromatography to afford the title compound (101 mg, 42%) as a white solid. Ή NMR (400 MHz, DMSO-d 5 ): δ 11.6 (s, 1 H), 8.05 (s, 1 H), 7.97 (m, 2 H), 7.67 (m, 2 H), 7.52 (m, 1 H), 7.12 (m, 1 H), 6.57 (m, 1 H), 5.29 (s, 1 H), 4.85 (m, 1 H), 3.53 (m, 1 H), 3.15 (s, 3 H), 2.24 (m, 2 H), 1.98 (m, 2 H), 1.44 (s, 6 H). LC/MS (exact mass) calculated for

C 20 H 25 N 5 O S; 415.168, found (M + H + ); 416.0.

Example 10: 1 -Cyclopropyl-N-{trans-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4 - yl) imino]cyclobutyl}methanesulfoncimide

This compound was synthesized starting from benzyl [trans-3-(methylamino)- cyclobutyl]carbamate (Example 1, Step 1), followi ng procedures similar to those described for Example 1, Steps 2 and 3 to obtain trans-N-methyl-N-7H-pyrrolo[2,3-d]pyrimidin-4- ylcyclobutane- l ,3-diami ne hydrochloride. To the resulting hydrochloride (60 mg, 0.28 mmol) in TH F (10 mL) was added potassium carbonate (76 mg, 0.55 mmol), H20 (5 mL) and cyclopropylmethanesulfonyl chloride (52 mg, 0.33 mmol). The mixture stirred for two hours, was diluted with dichloromethane, washed with brine, dried over sodium sulfate, filtered, and concentrated. The crude product was purified by preparative high performance liquid chromatography to afford the title compound as a white solid (7 mg ; 8%). Ή NMR (400 MHz, methanol- d 4 ): δ 8.14 (s, 1 H), 7.16 (d, 1 H,), 6.72 (d, 1 H), 5.44-5.40 (m, 1 H), 4.07-4.06 (m, 1 H), 3.41 (s, 3 H), 3.01-2.99 (m, 2 H), 2.81-2.74 (m, 2 H), 2.54-2.49 (m, 2 H), 1.15- 1.13 (m, 1 H), 0.720- 0.69 (m, 2 H), 0.42-0.41(m, 2 H). LC/MS (exact mass) calculated for C 15 H 21 N 5 0 2 S; 335.142 , found (M + H + ); 336.1.

Example 11: N-{(1 S, 3R)-3-[Methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclopentyl}propane- l - sulfonamide

This compound was prepared following Example 10 substituting (lS,3R)-N-benzyl-N'- methylcyclopentane- l, 3-diamine for benzyl [trans-3-(methylamino)-cyclobutyl]carbamate, (lR,3S)-N-methyl-N-7H-pyrrolo[2, 3-d]pyrimidin-4-ylcyclopentane- l,3-diamine hydrochloride for trans-N-methyl-N-7H-pyrrolo[2 , 3-d] pyrim id in-4-ylcyclobutane- 1 ,3 -diamine hydrochlo- ride, and propane- l-sulfonyl chloride for cyclopropylmethanesulfonyl chloride to afford the title compound as an off-white solid (11%). The crude compound was purified using preparative high performance liquid chromatography. Ή NMR (400 MHz, methanol-d 4 ): δ 8.08 (s, 1 H), 7.08 (s, 1 H,), 6.65 (s, 1 H), 5.27-5.23 (m, 1 H), 3.81-3.76 (m, 1 H), 3.31 (s, 3 H), 2.33-2.29 (m, 1 H), 2.13-2.04 (m, 1 H), 1.98- 1.92 (m, 2 H), 1.82- 1.75 (m, 4 H), 1.06 (t, 3 H), 0.42-0.41(m, 2 H). LC/MS (exact mass) calculated for C i5 H 23 N 5 0 2 S; 337.157, found (M + H + ); 337.8.

(lS,3R)-N-benzyl-N'-methylcyclopentane- l,3-diamine was prepared as follows:

Step 1: Benzyl [(1 R,3S)-3-am nocyc\openty\]carbamate

Trifluoroacetic acid (15 mL, 190 mmol) was added to a solution of benzyl tert-butyl (1 ?,3S)- cyclopentane- l,3-diylbiscarbamate (prepared as descri bed in WO2011/086053A1) (5.02 g, 15.0 mmol) in dichloromethane (75 mL) at room temperature. The reaction was stirred for 2 hours and was then concentrated to afford the title compound as a light brown oil (6.70 g, crude)

Step 2 : Benzyl [(1 R,3S)-3-(benzy\am no)cyc\openty\]carbamate

Sodium triacetoxyhydroborate (4.38 g , 20.0 mmol) was added to a solution of benzyl [(l ?,35)-3-aminocyclopentyl]carbamate (5.23 g, 15.0 mmol) and benzaldehyde (1.7 mL, 16.0 mmol) in dichloromethane (75 mL) at room temperature. The mixture was stirred for 21 hours and then aqueous 1 M sodium hydroxide solution (75 mL) was added to make the solution basic. The aqueous layer was extracted with dichloromethane (2 x 25 mL). The combined organic layers were washed with brine (50 mL), dried over sodium sulfate and concentrated. The crude material was chromatographed on silica gel eluting with a mixture of dichloromethane and methanol (100:0 to 88: 12) to afford the title compound as a yellow oil (3.47 g, 71%). Ή NMR (400 MHz, CDCI 3 ): δ 7.35-7.31 (m, 5 H), 7.30-7.26 (m, 5 H), 5.07 (s, 2 H), 4.17-4.07 (m, 1 H) 3.76-3.68 (m, 2 H), 3.27-3.20 (m, 1 H), 2.02- 1.51 (m, 6 H).

Step 3 : (1 S,3R)-N-Benzyl-N'-methylcyclopentane- l , 3-diamine

Lithium aluminum hydride (1.02 g, 26.9 mmol) was added in portions to a solution of benzyl [(l R,3S)-3-(benzylamino)cyclopentyl]carbamate (3.47 g, 10.7 mmol) in tetrahydrofuran (70 mL) at room temperature. The reaction was heated to reflux for 3.5 hours. The mixture was then cooled in an ice bath and sequentially quenched with water (1.0 mL), aqueous 15% sodiu m hydroxide solution (1.0 mL) and water (3.0 mL). The suspension was diluted with ethyl acetate and filtered through Celite. The filtrate was concentrated and the residue was taken up in aqueous 0.5 M hydrochloric acid solution. The mixture was washed with diethyl ether (2 x 20 mL) and the aqueous solution was made basic (pH~ l l) with sodium hydroxide. The resulting mixture was extracted with dichloromethane (3 x 25 mL). The combined organic layers were dried over sodium sulfate and concentrated. The crude material was chromatographed on silica gel eluting with a mixture of dichloromethane and methanol (90: 10) to afford the title compound as a yellow oil (204 mg, 9%). Ή N MR (400 MHz, CDCI 3 ): δ 7.33-7.20 (m, 5 H), 3.74 (s, 2 H), 3.19-3.13 (m, 1 H), 3.08-3.02 (m, 1 H) 2.39 (s, 3 H), 2.09-2.03 (m, 1 H), 1.87- 1.81 (m, 2 H), 1.67-1.54 (m, 2 H), 1.46- 1.39 (m, 1 H). LC/MS (exact mass) calculated for C i3 H 2o N 2 ; 204.163 , found (M + H + ); 205.1.

Example 12 : l -(3,3-Difluorocyclobutyl)-N-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4- yl)amino]cyclobutyl}methcmesulfonamide

Step 1: Benzyl [cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2, 3-d]pyrimidin-4- y\}am no)cyc\obuty\]carbamate

4-Chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrim idine (15 g, 48.7 mmol) and benzyl [cis-3-(methylamino)cyclobutyl]carbamate (17.2 g, 63.5 mmol) were mixed with isopropyl alcohol (180 mL) and diisopropylethylamine (28 mL, 161 mmol). The resulting slurry was heated at 75°C for 6 hours. The reaction was cooled to room temperature, filtered, washed with isopropyl alcohol (150 mL) and dried in an oven at 50°C to give the title compound (23.5 g, 95%) as a white solid. Ή NMR (400 MHz, DMSO-d 6 ): δ 8.38 (s, 1 H), 8.03 (d, 2 H), 7.45 (d, 1 H), 7.38-7.28 (m, 4 H), 7.26 (s, 1 H), 7.25 (d, 1 H), 6.61 (d, 1 H), 5.08 (s, 2 H), 4.96 (d, 1 H), 4.77 (m, 1 H), 3.88 (m, 1 H), 3.23 (s, 3 H), 2.71 (m, 2 H), 2.36 (s, 3 H), 2.18 (m, 2 H). Step 2 : cis-N-Methyl-N-{7-[(4-methylphenyl)sulfonyl]-7H^yrmlo[2,3-d] pyrimidin-4-yl}cyclobut ne- 7 , 3-d ia mine dihydrobromide

Benzyl [cis-3-(methyl{7-[(4- methyl phenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyri mid in-4- yl}amino)cyclobutyl]carbamate (15.2 g, 30.1 mmol) was suspended in ethyl acetate (45 mL) and acetic acid (45 mL). To the slurry was slowly added a 4M solution of HBr i n acetic acid (45 mL, 180 mmol), maintaining the temperature below 25°C. The resulting slurry was stirred at room temperature for 2 hours. The solids were collected by filtration, washed with ethyl acetate (450 mL), and dried at 40°C to afford the title compound (16 g; 100%) as a white solid. Ή NM R (400 MHz, DMSO-d 6 ): δ 8.31 (s, 1 H), 8.20 (s, 2 H), 7.97 (d, 2 H), 7.72 (d, 1 H), 7.44 (d, 2 H), 7.08 (d, 1 H), 4.93 (m, 1 H), 3.54 (m, 1 H), 3.30 (s, 3 H), 2.50 (m, 4 H), 2.35 (s, 3 H). LC/MS (exact mass) calculated for C H N O S; 371.142, found (M + H + ); 372.1.

Step 3 : ({[(3,3-Difluorocyclobutyl)methyl]thio}methyl)benzene

A mixture of (3,3-difluorocyclobutyl)methyl 4-methylbenzenesulfonate (see

WO2010/032200A1) (4 g, 14.5 mmol), benzyl imidothiocarbamate (3.53g, 17.4 mmol), sodium hydroxide solution (1.45 g, 36.2 mmol, dissolved in 16 mL water) and N ,N-dimethylformamide (16 mL) was sti rred at 60°C for 16 hours. Water (40 mL) and ethyl acetate (150 mL) were added. The organic layer was washed with water (40 mL), separated, dried over sodium sulfate and concentrated. The residue was chromatographed on silica gel eluting with a gradient of petroleum ether and ethyl acetate (100:0 to 95 : 5) to afford the title compound as colorless oil (3.2 g, 81%). Ή NMR (400 MHz, CDCI 3 ): δ 7.34-7.24 (m, 5 H), 5.71 (s, 2 H), 2.71-2.61 (m, 2 H), 2.57- 2.55 (m, 2 H), 2.30-2.14 (m, 3 H).

Step 4: (3, 3-Difluorocyclobutyl)methanesulfonyl chloride

This compound was prepared following the procedure of Example 8 Step 2, substituting S-[3-(cyanomethyl)-3-methylcyclobutyl] ethaneth ioate for 2-(benzylthio)isonicotinonitrile to afford the title compound as a colorless oi l (93%). Ή NMR (400 MHz, CDCI 3 ,): δ 3.88-3.86 (m, 2 H), 3.03-2.94 (m, 3 H), 2.61-2.49 (m, 2 H).

Step 5 : 1 -(3,3-Difluorocyclobutyl)-N-[cis-3-(methyl{7-[(4-methylpheny l)sulfonyl]-7H-pyrrolo[2,3- d]pyrimidin-4-yl}amino)cyclobutyl]methcmesulfon imide

A solution of (3, 3-difluorocyclobutyl)methanesulfonyl chloride (2.5 g, 12.19 mmol) in 10 mL dichloromethane was added dropwise to a solution of cis-N-methyl-N-{7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2, 3-d] pyrimidi n-4-yl}cyclobutane- 1,3 -diamine

dihydrobromide (3.25 g, 6.10 mmol) and triethylamine (3.08 g, 30.49 mmol) in dichloromethane (150 mL) at 0°C over 15 minutes. The reaction was stirred at room temperature for 4 hours. Water (50 mL) was added and the organic layer was separated. The aqueous layer was extracted with dichloromethane (2 x 150 mL), and the combined organic layers were separated dried over sodium sulfate. The crude compound was purified by chromatography on silica gel eluting with a gradient of dichloromethane and methanol (100:0 to 90: 10) to afford the title compound as a white solid (2.0 g, 61%). LC/MS (exact mass) calculated for C 23 H 27 F 2 N 5 0 4 S 2 ;

539.147, found (M + H + ); 540.1.

Step 6: / -(3,3-Difluorocyclobutyl)-N-{cis-3 methyl(7H-pyrroloi2,3-d]pyrimidin-4- yl)amino]cyclobutyl}methcinesulfoncimide

A solution of l-(3 , 3-difluorocyclobutyl)-N-[cis-3-(methyl{7-[(4-methylphenyl)- sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)cyclobutyl]m ethanesulfonamide (2 g, 3.71 mmol) and lithium hydroxide monohydrate (780 mg, 18.6 mmol) in ethanol (40 mL) and water (20 mL) was sti rred at 60°C for 4 hours. The ethanol was evaporated and the remaining aqueous layer was neutralized to pH 7 with hydrochloric acid and subsequently extracted with dichloromethane (2 x 200 mL). The combined organic layers was dried over sod ium sulfate, filtered, concentrated, and purified by preparative high performance liquid ch romatography to afford the title compound (800 mg, 56%) as a white solid. Ή NM R (400 MHz, methanol-d 4 ): δ 8.15 (s, 1H), 8.13 (s, 1 H), 7.16-7.15 (m, 1 H), 6.73-6.62 (m, 1 H), 4.95-4.88 (m, 1 H), 3.73- 3.71 (m, 1 H), 3.38 (s, 3 H), 3.28-3.26 (m, 2 H), 2.87-2.78 (m, 4 H), 2.63-2.61 (m, 1 H), 2.56- 2.48 (m, 2 H), 2.35-2.28 (m, 2 H). LC/MS (exact mass) calculated for C i6 H F 2 N 5 0 2 S; 385.138, found (M + H + ); 386.1.

The following compounds, Examples 13-14, were prepared from cis-N-methyl-N-{7- [(4-methylphenyl)sulfonyl]-7H-pyrrolo[2 , 3-d]pyrimidin-4-yl}cyclobutane- l,3-diamine dihydrobromide (Example 12, Step 2) in a similar manner to that described in Example 12 , Step 5 , substituting the indicated sulfonyl chloride for (3,3-difluorocyclobutyl)methanesulfonyl chlo- ride and using the deprotection method illustrated in Example 12 , Step 6.

Example 13: 3, 3-Difluoro-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}cyclobutanesulfonamide

This compound was prepared using 3 ,3-difluorocyclobutanesulfonyl chloride using the procedure in PCT Publication No. WO2011/068881.The crude compound was purified by chromatography on silica gel eluting with a gradient of petroleum ether and ethyl acetate (80: 20 to 10:90) to afford the title compound as an off-white solid (22% over 2 steps). Ή N MR (400 MHz, methanol-d 4 ): δ 8.13 (s, 1 H), 7.13 (d, 1 H), 6.70 (d, 1 H), 4.86-4.81 (m, 1 H), 3.78-3.72 (m, 2 H), 3.35 (s, 3 H), 3.01-2.93 (m, 4 H), 2.78-2.76 (m, 2 H), 2.32-2.25 (m, 2 H). LC/MS (exact mass) calculated for C i5 H ig F 2 N 5 0 2 S; 371.123, found (M + H + ); 372.1.

Example 14: 1 -Cyclopropyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}methanesulfonamide

This compound was prepared as a white solid using cyclopropylmethanesulfonyl chloride (73% over 2 steps). Ή NMR (400 MHz, DMSO-d 6 ): δ 11.64 (br. s., 1 H), 8.11 (s, 1 H), 7.53 (d, 1 H), 7.12-7.19 (m, 1 H), 6.64 (m, 1 H), 4.84-4.97 (m, 1 H), 3.54-3.70 (m, 1 H), 3.26 (s, 3 H), 2.93 (d, 2 H), 2.55-2.66 (m, 2 H), 2.29-2.22 (m, 2 H), 0.96-1.09 (m, 1 H), 0.53-0.64 (m, 2 H), 0.29-0.39 (m, 2 H). LC/MS (exact mass) calculated for C i5 H ig F 2 N 5 0 2 S; 335.142, found (M + H + ); 336.0.

Example 15: 1 -Cyclopropyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}azetidine-3-sulfonamide

Step 1: tert-Butyl 3-({[cis-3-(methyl{7-[(4- ethylphenyl)sulfonyl]-7H-pyrmlo[2,3-d]pyrimidin-4- yl}amino)cyclobutyl]amino}sulfonyl)azetidine- l -carboxylate

cis-N-Methyl-N-{7-[(4- methyl phenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyri mid in-4- yl}cyclobutane-l,3-diamine dihydrobromide (7.0 g, 18.8 mmol) was free-based by stirring in excess aqueous IN sodium hydroxide solution for 3 minutes and then extracting into di- chloromethane. The organic layer was dried over sodium sulfate and concentrated. The remaining free base was taken up in dichloromethane (200 mL), cooled to 0°C and treated with and triethylamine (13 mL, 94 mmol) and tert-butyl 3-(chlorosulfonyl)azetidine-l-carboxylate. The reaction was allowed to stir at room temperature for 10 minutes. The crude mixture was washed with water and brine, then dried over sodium sulfate and concentrated to afford the crude product as a white solid. The solid was crystallized using a mixture of dichloromethane and diethyl ether to afford the title compound as a white solid (9.61 g, 90%). Ή NMR (400 MHz, methanol-d 4 ): δ 8.19 (s, 1 H), 7.92-8.01 (m, 2 H), 7.54 (d, 1 H), 7.35 (d, 2 H), 6.86 (d, 1 H), 4.76-4.65 (m, 1 H), 4.18 (br. s., 2 H), 3.99-4.10 (m, 3 H), 3.66-3.78 (m, 1 H), 3.25 (s, 3 H), 2.64-2.78 (m, 2 H), 2.37 (s, 3 H), 2.10-2.25 (m, 2 H), 1.41 (s, 9 H). LC/MS (exact mass) calcu- lated for C H N O S ; 590.198, found (M + H + ); 591.45.

Step 2: N-[cis-3-(Methyl{7-[(4-met ylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}amino)cyclobutyl]azetidine-3-sulfonamide

Acetyl chloride (0.20 mL, 2.8 mmol) was added to a solution of tert-butyl 3-({[cis-3- (methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimi din-4- yl}amino)cyclobutyl]amino}sulfonyl)azetidine- l-carboxylate (1.64 g, 2.78 mmol) in anhydrous dichloromethane (18 mL) and methanol (7 mL) at 0°C. The reaction mixture was stirred at room temperature for 16 hours. The white preci pitate was filtered off and taken up in saturated aqueous sodium bicarbonate solution (20 mL). The resulting solution was extracted with di- chloromethane (3 x 20 mL) and the combined organic layers were dried over sodium sulfate and concentrated to afford the title compound (810 mg, 60%) as a white solid. LC/MS (exact mass) calculated for C 21 H 26 N 6 0 4 S 2 ; 490.146, found (M + H + ); 491.0.

Step 3 : 1 -Cyclopmpyl^-[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H- pyrrolo[2,3-d]pyrimidin-4- yl}amino)cyclobutyl]azetidine-3-sulfonamide

N-[cis-3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2 ,3-d]pyrimidin-4- yl}amino)cyclobutyl]azetidine-3-sulfonamide (810 mg, 1.65 mmol), methanol (10 mL), molecular sieves, and [(l-ethoxycyclopropyl)oxy](trimethyl)silane (0.53 mL, 2.64 mmol) were combined in a sealable reaction vessel. The vessel was purged with nitrogen and acetic acid (1.28 mL, 8.26 mmol) was added. The vessel was sealed and then heated at 80°C for 2 hours. After the mixture was cooled to room temperature, sod ium cyanoborohydride (273 mg, 4.13 mmol) was added. The vessel was resealed and heated slowly to 40°C for 1.5 hou rs. The crude mixture was filtered over a bed of Celite, rinsing with methanol. The filtrate was concentrated and the residue was taken up an aqueous saturated sodium bicarbonate solution. The resulting solution was extracted with d ichloromethane (5 x 20 mL), and the combined organic layers were dried over sodium sulfate and concentrated to afford the title compound (576 mg, 74%) as a white solid. Ή NMR (400 MHz, methanol-d 4 ): δ 8.23 (s, 1 H), 7.95-8.05 (m, 2 H), 7.58 (d, 1 H), 7.39 (d , 2 H), 6.90 (d, 1 H), 4.69-4.83 (m, 1 H), 3.94-4.09 (m, 1 H), 3.65-3.75 (m, 3 H), 3.54- 3.64 (m, 2 H), 3.29 (s, 3 H), 2.67-2.79 (m, 2 H), 2.41 (s, 3 H), 2.15-2.29 (m, 2 H), 2.02-2.15 (m, 1 H), 0.43-0.51 (m, 2 H), 0.29-0.39 (m, 2 H). LC/ MS (exact mass) calculated for

C H N O S ; 530.177, found (M + H + ); 531.0.

Step 4: / -Cyclopropyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}azetidine-3-sulfonamide

A solution of cesium carbonate (976 mg, 3.0 mmol) in water (5 mL) was added to a solution of l-cyclopropyl-N-[cis-3-(methyl{7-[(4- methyl phenyl)sulfonyl]-7H-pyrrolo[2, 3- d]pyrimidin-4-yl}amino)cyclobutyl]azetidine-3-sulfonamide (530 mg, 1.0 mmol) in ethanol (10 mL). The reaction mixture was heated to reflux for 16 hours. After the solvent was removed, remaining material was taken up in water and extracted with a mixture of dichloromethane and methanol (96:4; 3 x 10 mL). The combined organic layers were dried over sodium sulfate and concentrated. The crude solid was crystallized from methanol to afford the title compound (225 mg, 59%) as a white solid. Ή N MR (400 M Hz, methanol-d 4 ): δ 8.10 (s, 1 H), 7.09 (d, 1 H), 6.66 (d, 1 H), 4.88-4.80 (m, 1 H), 4.03-3.96 (m, 1 H), 3.73-3.65 (m, 3 H), 3.61-3.57 (m, 2 H), 3.32 (s, 3 H), 2.77-2.68 (m, 2 H), 2.28-2.19 (m, 2 H), 2.08-2.03 (m, 1 H), 0.46-0.41 (m, 2 H), 0.34- 0.31 (m, 2 H). LC/MS (exact mass) calculated for C 17 H 24 N 6 0 2 S; 376.168, found (M + H + ); 377.0.

Example 16: N-(Cyclopropylmethyl)-N'-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyr imidin-4- yl)amino]cyclobutyl}sulf amide

Step 1: N-[cis-3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3 -d]pyrimidin-4- yl}amino)cyclobutyl]-2-oxo- 1 , 3-oxazolidine-3-sulfonamide

To a solution of chlorosulfonyl isocyanate (1.76 mL, 20.6 mmol) in dichloromethane

(150 mL) was added dropwise a solution of 2-bromoethanol (1.43 mL, 20.6 mmol) in dichloromethane (80 mL) at 0 C. After 30 minutes at 0 * C, a solution of cis-N-methyl-N-{7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2, 3-d] pyrimidi n-4-yl}cyclobutane- 1,3 -diamine

dihydrobromide (11.0 g, 20.6 mmol) and triethylamine (10.42 g, 103.2 mmol) in dry dichloro- methane (80 mL) was added dropwise, and the reaction mixture was allowed to warm to room temperature overnight. The reaction solution was dissolved in dichloromethane (1 L), washed with aqueous 1M hydrochloric acid solution (2 x 800 mL) and brine (500 mL). The solution was dried over sodium sulfate and concentrated to afford the title compound as white solid (8.5 g, 79%). Ή NMR (400 MHz, CD 3 OD): δ 8.22 (s, 1 H), 8.00 (d, 2 H), 7.58 (d, 1 H), 7.38 (d , 2 H), 6.91 (d, 1 H), 4.88 (m, 1 H), 4.45 -4.41 (m, 2 H), 4.06-4.02 (m, 2 H), 3.75 (m, 1 H), 3.29 (s, 3 H), 2.72-2.69 (m, 2 H), 2.40 (s, 3 H); 2.30-3.27 (m, 2 H). LC/MS (exact mass) calculated for C H N O S ; 520.120, found (M + H + ); 521.4.

Step 2 : N-(Cyclopmpylmethyl)-N'-[cis-3-(methyl{7-[(4-methylphenyl)su lfonyl]-7H-pyrrolo[2, 3- d]pyrimidin-4-yl}amino)cyclobutyl]sulfamide

A solution of N-[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2, 3- d]pyrimidin-4-yl}amino)cyclobutyl]-2-oxo- l,3-oxazolidine-3-sulfonamide (150 mg, 0.29 mmol), cyclopropanemethylamine (51 mg, 0.72 mmol) and triethylamine (116 mg, 1.15 mmol) in acetonitrile (3 mL) was stirred for 15 minutes at 100°C using microwave heating. The reaction mixture was concentrated to afford the crude title compound (146 mg, 100% crude yield) as a yellow oil. LC/ MS (exact mass) calculated for C 22 H 28 N 6 0 4 S 2 ; 504.161, found (M + H + ); 505.2. Step 3 : N-(Cyclopropylmethyl)-N'-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyr imidin-4- y\)amino]cyc\ob tyi}s \ amide

A solution of N-(cyclopropylmethyl)-N'-[cis-3-(methyl{7-[(4-methylphenyl)s ulfonyl]- 7H-pyrrolo[2 , 3-d]pyrimidin-4-yl}amino)cyclobutyl]sulfamide (146 mg , 0.29 mmol), lithium hy- droxide monohydrate (48 mg, 1.15 mmol) in ethanol (5 mL) and water (2.5 mL) was stirred at 100°C for 1 h. The reaction mixture was concentrated under vacuum and the crude product was purified by preparative high performance liquid chromatog raphy to afford the title compound (14 mg, 14%) as a white solid. Ή NM R (400 MHz, methanol-d 4 ): δ 8.12 (s, 1 H), 7.13 (d, 1 H), 6.90 (d , 1 H), 4.90-4.86 (m, 1 H), 3.63-3.59 (m, 1 H), 3.37 (s, 3 H), 2.85-2.83 (m, 2 H), 2.78- 2.71 (m, 2 H), 2.33-2.26 (m, 2 H), 1.05- 1.03 (m, 1 H), 0.57-0.52 (m, 2 H); 0.30-0.25 (m, 2 H). LC/ MS (exact mass) calculated for C 15 H 22 N 6 0 2 S; 350.152 , found (M + H + ); 351.2.

The following compounds, Examples 17-18, were prepared from N-[cis-3-(methyl{7- [(4-methylphenyl)sulfonyl]-7H-pyrrolo[2 , 3-d]pyrimidin-4-yl}amino)cyclobutyl]-2-oxo- l ,3- oxazolidine-3-sulfonamide (Example 16, Step 1) in a similar manner to that described in Example 16, Step 2 , substituting the indicated amine for cyclopropanemethylamine, and using the deprotection method illustrated in Example 16, Step 3. Example 17A and 17B: (R)- and (S)-3-Cyano-N-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4- yl)amino]cyclobutyl}pyrrolidine- 1 -sulfonamide

These compounds were prepared using racemic pyrrolidine-3-carbonitrile hydrochloride. The crude racemic mixture was purified by high performance liquid chromatography to afford a white solid (60 mg, 52% over 2 steps). The enantiomers were separated by supercritical fluid chromatography.

Enantiomer A (17A): 24 mg (21%): Ή NMR (400 MHz, CD 3 OD): δ 8.13 (s, 1 H), 7.13 (d, J = 3.2 Hz, 1 H), 6.69 (d, 1 H, J = 3.6 Hz, 1 H), 4.87-4.84 (m, 1 H), 3.73-3.67 (m, 1 H), 3.65-3.57 (m, 1 H), 3.53-3.50 (m, 2 H), 3.48-3.44 (m, 2 H), 3.405 (s, 3 H), 2.77-2.75 (m, 2 H), 2.42-2.20 (m, 4 H). LC/MS (exact mass) calculated for C 16 H 21 N 7 0 2 S; 375.148, found (M + H + ); 376.1. Chiral HPLC retention time = 5.97 mi nutes.

Enantiomer B (17B): 25 mg (21%). Ή NMR (400 M Hz, CD 3 OD): δ 8.13 (s, 1 H), 7.13 (d, J = 3.2 Hz, 1 H), 6.69 (d, 1 H, J = 3.6 Hz, 1 H), 4.87-4.84 (m, 1 H), 3.73-3.67 (m, 1 H), 3.65-3.57 (m, 1 H), 3.53-3.50 (m, 2 H), 3.48-3.44 (m, 2 H), 3.405 (s, 3 H), 2.77-2.75 (m, 2 H), 2.42-2.20 (m, 4 H). LC/MS (exact mass) calculated for C 16 H 21 N 7 0 2 S; 375.148, found (M + H + ); 376.1. Chiral HPLC retention time = 5.16 mi nutes.

Example 18: 2-Methyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)am ino]cyclobutyl}2, 6- dihydropyrrolo[3,4-c]pyrazole-5(4H)-sulfonamide This compound was prepared using 2-methyl-2 ,4, 5,6-tetrahydropyrrolo[3,4-c]pyrazole hydrochloride. The crude compound was purified by high performance liquid chromatography to afford the title compound as an off-white solid (24% over 2 steps). Ή N MR (400 MHz, methanol^): δ 8.08 (s, 1 H), 7.38 (s, 1 H), 7.10 (d, 1 H), 6.66 (d, 1 H), 4.87-4.86 (m, 1 H), 4.42- 4.41 (m, 4 H), 3.87 (s, 3 H), 3.71-3.67 (m, 1 H), 3.31 (s, 3 H), 2.68-2.61 (m, 2 H), 2.27-2.22 (m, 3 H). LC/MS (exact mass) calculated for C i7 H 22 N 8 0 2 S; 402.159, found (M + H + ); 403.2 and (M + Na); 425.1.

Example 19: N-{cis-3-lMethyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}-6-oxa-3- azab cyc\o[3. 1. 7 ]heptane-3-sulfonamide

Step 1: N-[cis-3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3 -d]pyrimidin-4- yl}amino)cyclobutyl]-6-ox -3-az bicyclo[3. 1. 7 ]heptane-3-sulfonamide

A mixture of N-[cis-3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2 ,3- d]pyrimidin-4-yl}amino)cyclobutyl]-2-oxo- l,3-oxazolidine-3-sulfonamide (208 mg, 0.40 mmol), 6-oxa-3-azabicyclo[3.1.1]heptane (50 mg, 0.50 mmol), triethylamine (220 u L, 1.58 mmol) in acetonitrile (15 mL), was heated in a 20 mL microwave vial in a microwave reactor for 1 hour at 120°C. Excess solvent was evaporated and the resulting oil was taken up in dichloromethane. The solution was washed with aq ueous ammonium chloride and brine. The crude material was d ried over sodium sulfate and concentrated to give an oil. This was chro- matographed on silica gel eluting with a gradient methanol in dichloromethane (0: 100 to 5 : 100) to afford the title compound as a foam (82 mg, 30%). Ή N MR (CDCI B ): δ 8.38 (s, 1 H), 8.04 (d, 2 H), 7.48 (d, 1 H), 7.28 (d, 2 H), 6.63 (d, 1 H), 4.78-4.69 (m, 1 H), 4.62 (d, 1 H), 4.47 (d, 1 H), 3.69-3.61 (m, 1 H), 3.58 (d, 3 H), 3.26-3.17 (m, 1 H), 3.24 (s, 3 H), 2.83-.275 (m, 2 H), 2.37 (s, 3 H), 2.18-2.11 (m, 2 H), 2.04 (d, 1 H), 1.18 (t, 1 H). LC/MS (exact mass) calculated for

C 2B H 28 N 6 0 5 S 2 ; 532.156, found (M + H + ); 533.

Step 2 : N-{cis-3-[Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclo butyl}-6-oxa-3- azab cyc\o[3. 7. 7 ]heptane-3-sulfonamide

N-[cis-3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2 ,3-d]pyrimidin-4- yl}amino)cyclobutyl]-6-oxa-3-azabicyclo[3.1.1]heptane-3-sulf onamide (229 mg, 0.43 mmol) was added to a solution of in 1 M tetrabutylammonium fluoride in tetrahydrofuran (6.5 mL, 6.4 mmol). The reaction was stirred at room temperature for 10 hours. The mixture was concentrated and the remaining material was chromatog raphed on silica gel eluting with a mixture of methanol in ethyl acetate (1 :9). A yellow oil was isolated that was triturated with a mixture of ethyl acetate and heptane to give a yellow solid. The solid was partitioned between ethyl acetate and water. The aqueous layer was extracted with ethyl acetate and the combined organic layers were washed with brine, dried over sodium sulfate and concentrated to afford an off-white solid. This was triturated with diethyl ether and then isopropyl alcohol to afford the title compound as a white solid (14 mg, 9%). Ή N M R (CD OD) δ 8.08 (s, 1 H), 7.09 (d, 1 H), 6.66 (s, 1 H), 4.90-4.81 (m, 1H), 4.62 (d, 2 H), 3.86-8.84 (m , 1 H), 3.66 (t, 1 H), 3.56-3.49 (m, 3 H), 3.33 (s, 3 H), 3.19-3.13 (m, 1 H), 2.75-2.70 (m, 2 H), 2.32-2.24 (m, 2 H), 2.05-2.03 (d, 1 H). LC/MS (exact mass) calculated for C 16 H 22 N 6 0 3 S; 378.147, found (M + H + ); 379.5.

The following compounds, Examples 20-24, were prepared from N-[cis-3-(methyl{7- [(4-methylphenyl)sulfonyl]-7H-pyrrolo[2 , 3-d]pyrimidin-4-yl}amino)cyclobutyl]-2-oxo- l ,3- oxazolidine-3-sulfonamide (Example 16, Step 1) in a similar manner to that described in Example 16, Step 2 , substituting the indicated amine for cyclopropanemethylamine, and using the deprotection method illustrated in Example 19, Step 2. Example 20: 3-Cyano-N-{cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]- cyclobutyljazetidine- 1 -sulfonamide

This compound was prepared using azetidine-3-carbonitrile. The crude compound was purified by high performance liquid chromatography to afford the title compound as a white solid (23% over 2 steps). Ή NM R (400 MHz, CD 3 OD): δ 8.10 (s, 1 H), 7.11 (d, 1 H), 6.68 (d, 1 H), 4.80 (m, 1 H), 4.02 (m, 2 H), 3.90 (m, 2 H), 3.58 (m, 2 H), 3.32 (s, 3 H), 2.72 (m, 2 H), 2.25 (m,

2 H). LC/ MS (exact mass) calculated for C i5 H ig N 7 0 2 S; 361.132 , found (M + H + ); 362.1.

Example 21: N-{cis-3-[Methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}-4-(l H-pyrazol-3- yl)piperidine- I -sulfonamide

This compound was prepared using 4-(lH-pyrazol-3-yl)piperidine. The crude compound was purified by chromatography on silica gel eluding with a mixture of dichloromethane and methanol (9: 1). The isolated material was triturated with diethyl ether and then ethyl acetate to afford the title compound as a white solid (10% over 2 steps). Ή NMR (300 MHz, CD 3 OD): δ 8.09 (s, 1 H), 7.48 (s, 1 H), 7.12-7.05 (m, 1 H), 6.71-6.60 (m, 1 H), 6.22-6.08 (m, 1 H), 4.92- 4.73 (m, 1H), 3.80-3.55 (m, 3 H), 3.41 (s, 3H), 2.90-2.65 (m, 5 H), 2.38-2.19 (m, 2 H), 2.09- 1.90 (m, 2 H) and 1.83- 1.65 (m, 2 H). LC/MS (exact mass) calculated for C ig H 26 N g 0 2 S; 430.190, found (M + H + ); 431.1. Example 22 : N-(2-Cyanoethyl)-N-methyl-N'-{cis-3-[methyl(7H-pyrrolo[2,3-d ]pyrimidin-4- yl)amino]cyclobutyl}sulf amide

This compound was prepared using 3-methylaminopropionitrile. The crude compound was purified by high performance l iquid chromatography to afford the title compound (7% over 2 steps). LC/MS (exact mass) calculated for C is H 2i N ? 0 2 S; 363.148, found (M + H + ); 364.0.

Example 23 and 27: (1 S, 5S)- 1 -Cyano-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}-3-azabicyclo[3. 1.0]hexane-3-sulfonamide and (1 R, 5R)- 7 -Cyano-N-{cis-3- [methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}-3-azabicyclo[3. 1.0]hexane-3- sulfonamide

These compounds were prepared using racemic 3-azabicyclo[3.1.0]hexane- l- carbonitrile. The crude racemic compound was purified as a white solid (92 mg, 21% over 2 steps) by chromatography on silica gel eluting with a gradient of dichloromethane and methanol (30: 1 to 5 : 1). The title enantiomers were separated by supercritical fluid chromatography. Enantiomer 23 : 41 mg (9%); SFC retention time = 4.28 minutes; Ή NM R (400 MHz, methanol- d 4 ): δ 8.13 (s, 1 H), 7.13 (d, 1 H), 6.70 (d, 1 H), 4.61 (s, 1 H), 3.57 - 3.72 (m, 2 H), 3.43 - 3.51 (m, 3 H), 3.36 (s, 3 H), 2.70 - 2.77 (m, 2 H), 2.24 - 2.38 (m, 3 H), 1.41 - 1.48 (m, 1 H), 1.32 (t, 1 H). LC/ MS (exact mass) calculated for C 17 H 21 N ? 0 2 S; 387.148, found (M + H + ); 388.1.

Enantiomer 27: 40 mg (9%); SFC retention ti me = 4.84 minutes Ή N M R (400 MHz, methanol- d 4 ): δ 8.13 (s, 1 H), 7.13 (d, 1 H), 6.70 (d, 1 H), 4.61 (s, 1 H), 3.57 - 3.72 (m, 2 H), 3.43 - 3.51 (m, 3 H), 3.36 (s, 3 H), 2.70 - 2.77 (m, 2 H), 2.24 - 2.38 (m, 3 H), 1.41 - 1.48 (m, 1 H), 1.32 (t, 1 H). LC/ MS (exact mass) calculated for C 17 H 21 N ? 0 2 S; 387.148, found (M + H + ); 388.1.

Racemic 3-azabicyclo[3.1.0]hexane- l-carbonitrile was prepared as follows.

Step 1: Racemic tert-butyl rac- 1 -formyl-3-azabicyclo[3. 1.0]hexane-3-carboxylate

l, l, l-Triacetoxy- l, l-dihydro- l,2-benziodoxol-3(lH)-one (6.5 g, 15.2 mmol) was added to a solution of racemic tert-butyl- l-(hydroxymethyl)-3-azabicyclo[3.1.0]hexane-3- carboxylate (Synlett 2009, 921) (2.5 g, 11.7 mmol) in anhydrous dichloromethane (60 mL). The reaction mixture stirred for 2 hours at room temperature. The mixture was diluted with dichloromethane (60 mL), washed with a saturated aqueous solution of sodi um sulfite, saturated sodiu m bicarbonate (30 mL) and brine (50 mL). The organic layer was dried over sodium sulfate and concentrated to give afford the title compound as a colorless oil (1.7 g, 68%). 'H NMR (400 MHz, CDCI 3 ): δ 9.01 (d, 1 H), 3.83 (d, 1 H), 3.68 (t, 1H), 3.59 (dd, 1 H), 3.50-3.36 (m, 1 H), 2.25-2.09 (m, 1 H), 1.63 (t, 1 H) , 1.43 (s, 9 H), 1.19- 1.06 (m, 1 H).

Step 2 : Racemic tert-butyl- 1 -[(hydroxyimino)methyl]-3-azabicyclo[3. 1.0]hexane-3-carboxylate Potassium carbonate (3.89 g, 28.2 mmol) and hydroxylamine hydrochloride (671 mg, 9.7 mmol) were added to a solution of tert-butyl racemic l-formyl-3-azabicyclo[3.1.0]hexane- 3-carboxylate (1.7 g , 8.05 mmol) in anhydrous dichloromethane (40 mL) at room temperature and then stirred for 16 hours. The mixture was diluted with ethyl acetate (80 mL), and washed with water (30 mL) and brine (30 mL). The organic layer was dried over sodium sulfate and concentrated. The residue was chromatographed on silica gel eluting with a gradient of petroleum ether and ethyl acetate (0: 100 to 83: 17) to afford the title compound as a yellow oil (1.6 g, 88%). Ή NMR (400 MHz, CDCI 3 ): δ 7.18 (s, 1 H), 3.74-3.55 (m, 3 H), 3.44-3.40 (m, 1 H), 1.74- 1.72 (m, 1 H), 1.44 (s, 9 H), 1.10 (t, 1 H), 0.86-0.83 (m, 1 H).

Step 3 : Racemic tert-butyl- 1 -cyano-3-azabicyclo[3. l .0]hexane-3-carboxylate

To a solution of racemic tert-butyl- l-[(hydroxyi mino)methyl]-3- azabicyclo[3.1.0]hexane-3-carboxylate (925 mg, 4.09 mmol) in anhydrous tetrahyd rofuran (100 mL) was added methyl /V-(triethylammonium sulfonyl)carbamate (2.92 g, 12.3 mmol). The reaction mixture was heated to reflux for 3 hours. After evaporation of the solvent, the residue was chromatographed on silica gel eluting with a mixture of petroleum ether and ethyl acetate (5 : 1) to afford the title compound as a colorless oil (570 mg, 67%). Ή NMR (400 MHz, CDCI 3 ): δ 3.84 (dd, 1 H), 3.64 (dd, 1 H), 3.50 (d, 1 H), 3.46 (dd, 1 H), 2.21-2.12 (m, 1 H), 1.44 (s, 9 H), 0.96 (t, 1 H).

Step 4: Racemic 3-azabicyclo[3. 7.0]hexane- l -carbonitrile

A solution of racemic tert-butyl- l-cyano-3-azabicyclo[3.1.0]hexane-3-carboxylate in trifluoroacetic acid (1 mL) and d ichloromethane (10 mL) was stirred at room temperature for 1 hour. The solvent was removed to afford the title compound (205 mg, 100%) as a brown oil.

Example 24: Racemic 3-cyano-N-{trans-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}pyrrolidine- 1 -sulfonamide

This compound was synthesized following the procedure of Example 10, substituting 3- cyanopyrrolidine- l-sulfonyl chloride for cyclopropylmethanesulfonyl chloride. The crude product was purified using preparative high performance liquid chromatography to afford the title compound as an off-white solid (5%). Ή NMR (400 MHz, CD 3 OD): δ 8.13 (s, 1 H), 7.14 (d, 1 H), 6.67 (d , 1 H), 5.45- 5.41 (m, 1 H), 4.00-3.64 (m, 1 H), 3.62-3.52 (m, 1 H), 3.5 1-3.47 (m, 2 H), 3.45-3.39 (m, 2 H), 3.369 (s, 3 H), 2.78-2.70 (m, 2 H), 2.53-2.47 (m, 2 H); 2.39-2.36 (m, 1 H); 2.27-2.24 (m, 1 H). LC/ MS (exact mass) calculated for C 16 H 21 N ? 0 2 S; 375.148, found (M + H + ); 375.9.

Racemic 3-cyanopyrrolidine- i -sulfonyl chloride A solution of racemic pyrrolidine-3-carbonitrile (53 mg, 0.4 mmol) and triethylamine (101 g, 1 mmol) in dry dichloromethane (1.0 mL) was added dropwise to a stirred solution of sulfuryl chloride (64.8 mg, 0.48 mmol) in dichloromethane (3.0 mL) was added at -78°C. The reaction was stirred at -78°C for 30 minutes, and then allowed to warm to room temperature over 1 hour. The reaction solution was washed with aqueous 1 M hydrochloric acid (5 mL) and brine (5 mL), dried over sodium sulfate and concentrated to afford the title compound as colorless oil (68 mg , crude).

Example 25: N-(cis-3-{[(4,4-Difluoropiperidin- l -yl)sulfonyl]methyl}cyclobutyl)-N-methyl-7H- pyrro\o[2, 3-d]pyrimidin-4-amine

Step 1: cis/tr ms-Ethyl 3-[(tert-butoxycarbonyl)ammo]cyclobutanecarboxyl ite

To a solution of a mixture of cis- and trans-ethyl 3-aminocyclobutanecarboxylate hydrochloride (cis/trans = 10: 1) (WO2009/60278) (10 g, 55.7 mmol) and triethylamine (19.4 mL, 139.1 mmol.) in dichloromethane (370 mL) at 0°C was added dropwise di-ferr-butyl dicarbonate (15.8 g, 72.3 mmol). After addition was complete, the mixture was stirred at room temperature overnight. The solvent was evaporated under reduced pressure and the resulting residue was chromatographed on silica gel eluting with a gradient of petroleum ether and ethyl acetate (10: 1 to 3 : 1) to afford the title mixture as a white solid (19 g, 92%). Ή NMR (400 MHz, CDCI 3 ): δ 4.77 (s, 1 H), 4.13 (q, 3 H), 2.68 - 2.82 (m, 1 H), 2.60 (d, 2 H), 1.99 - 2.17 (m, 2 H), 1.43 (s, 9 H), 1.25 (t, 3 H).

Step 2 : cis/trans-[3-(Met ylamino)cyclobutyl]methanol

Lithium aluminum hydride (9.14 g, 240.4 mmol) was suspended in dry tetrahydrofuran (350 mL). The mixture was cooled to 0°C and a solution of cis/trans ethyl 3-[(tert- butoxycarbonyl)amino]cyclobutanecarboxylate (cis/trans = 10: 1) (11.7 g, 48.1 mmol) in dry tetrahydrofuran (170 mL) was added drop wise. After addition was complete, the resulting mixture was heated to reflux overnight. After it was cooled to room temperature, the reaction was diluted with tetrahydrofuran (1.5 L) and then cooled to 0~5°C. Small portions of Na 2 SO 4 .10H 2 O were added until gas evolution had ceased. The mixture was filtered to remove the solids, which were washed with more tetrahydrofuran (500mL). The filtrate was concentrated to dry- ness affording the title mixture (cis/trans = 10: 1) as an oil (10 g, > 100%). Ή N M R (400 MHz, CDCI 3 ): δ 3.58 (d, J = 3.8 Hz, 2 H), 3.06-3.17 (m, 1 H), 2.34-2.43 (m, 3 H), 2.32 (s, 3 H), 1.48- 1.57 (m, 2 H).

Step 3 : cis/trans-[3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo [2,3-d]pyrimidm-4- yl}amino)cyclobutyl]methanol Potassium iodide (173 mg) and triethylamine (13 mL, 93.8 mmol) were added to a solution of cis/trans-[3-(methylamino)cyclobutyl]methanol (6.0 g, 52.1 mmol) in acetone (250 mL). 4-Chloro-7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2, 3-d]pyrimidine (14.4 g, 46.9 mmol) was then added and the resulting mixture was heated to reflux overnight. After evaporation of the solvent under reduced pressure, the residue was diluted with dichloromethane (500 mL). The solution was washed sequentially with water (300 mL), 2% aqueous citric acid (300 mL) and brine (300 mL), and then dried over sodium sulfate. After filtration, the solution was filtered and concentrated to afford the title mixture as a light solid (15.3 g, 85%). A portion (5.0 g) of the cis/trans-[3-(methyl{7-[(4- methyl phenyl)su If onyl]-7H-pyrrolo[2 , 3-d] pyri mid in-4- yl}amino)-cyclobutyl]methanol mixture was separated by supercritical fluid chromatography using a Chiralpak-AD col umn:

cis isomer, 4.6 g: Ή N M R (400 M Hz, methanol-d 4 ): δ 8.20 (s, 1 H), 7.98 (d , 2 H), 7.53 (d , 1 H),

7.34 (d , 2 H), 6.83 (d, 2 H), 4.99-4.95 (m, 1 H), 3.56 (d, J = 5.6 Hz, 1 H), 3.24 (s, 3 H), 2.36 (s, 3 H), 2.34-2.28 (m, 2 H), 2.24-2.19 (m, 1 H), 2.11-2.03 (m, 2 H). LC/MS (exact mass) calculat- ed for C H N O S: 386.14, found (M + H + ): 387.3

trans isomer, 0.4 g : Ή NMR (400 MHz, methanol-d 4 ): δ 8.20 (s, 1 H), 7.98 (d, 2 H), 7.55 (d, 1H),

7.35 (d , 2 H), 6.84 (d, 2H), 5.26- 5.22 (m, 1H), 3.69 (d, 1H), 3.30 (s, 3H), 2.46-2.41 (m, 3 H), 2.39 (s, 3H), 2.19-2.14 (m, 2H). LC/MS (exact mass) calculated for C ig H 22 N 4 0 3 S: 386.14, found (M + H + ): 387.3

Step 4: cis-[3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d ]pyrimidin-4- yl}amino)cyclobutyl]methyl 4- et ylbenzenesulfon ite

To a solution of cis-[3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3- d]pyrimidin-4-yl}amino)cyclobutyl]methanol (20 g, 51.8 mmol) and N,N-dimethylaminopyridine (12.6 g, 103.6 mmol) in dichloromethane (500 mL) at 0°C was added p-toluenesulfonyl chloride (14.8 g, 77.7 mmol). The reaction mixture was stirred at room temperature for 16 hours and then washed with water (500 mL). The combined aqueous washes were extracted with dichloromethane (2 x 800 mL). The combined organic layers were dried, filtered and concentrated under vacuum. The residue was chromatographed on silica gel eluting with a gradient of dichloromethane and methanol (100:0 to 95 : 5) to afford the title compound (23 g, 82%) as a white solid . LC/MS (exact mass) calculated for C 26 H 2g N 0,.S 2 : 540.150, found (M + H + ): 541.3. Step 5 : S-{[cis-3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2, 3-d]pyrimidin-4- yl}amino)cyclobutyl]methyl}ethanethioate

To a solution of potassium thioacetate (678 mg, 5.93 mmol) in N, N-dimethylformamide (5 mL) was added a solution of [cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2 , 3- d]pyrimidin-4-yl}arnino)cyclobutyl]methyl 4-methylbenzenesulfonate (2.0 g, 3.70 mmol) in N, N-dimethylformamide (6 mL) dropwise over 5 minutes at room temperature. The mixture was then heated to at 50-55°C overnight. The mixture was cooled to room temperature and quenched by pouring into aqueous saturated sod ium bicarbonate solution (60 mL). The mixture was extracted with ethyl acetate (3 x 30 mL) and the combined organic layers were washed with water (3 x 30 mL), brine (30 mL). After drying over Na 2 S0 4 the solution was concentrated. The residue was chromatographed on silica gel eluting with a gradient of dichloromethane and methanol (100:0 to 80:20) to afford the title compound (1.2 g, 73%) as a yellow solid. Ή N MR (400 MHz, CDCI ): δ 8.39 (s, 1H), 8.04 (d, 2 H), 7.45 (d , 1 H), 7.27 (d, 2 H), 6.63 (d, 1 H), 4.98- 4.88 (m, 1 H) 3.22 (s, 3 H) 3.02-3.00 (m, 2 H) 2.45-2.44 (m, 2 H), 2.47 (m, 3 H) 2.22 (m, 3 H) 2.21-2.24 (m, 1 H) 1.92- 1.87 (m, 2 H). LC/MS (exact mass) calculated for C 21 H 24 N 4 0 3 S 2 :

444.129, found (M + H + ): 445.1.

Step 6: [cis-3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d ]pyrimidin-4- yl}amino)cyclobutyl]methanesulfonic acid

To a solution of S-{[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2 ,3- d]pyrimidin-4-yl}amino)cyclobutyl]methyl} ethanethioate (580 mg, 1.31 mmol), in formic acid (10 mL) at room temperature was added 30% aqueous hydrogen peroxide solution (0.7 mL, 6.92 mmol). The resulting mixture was stirred at room temperature overnight. The reaction was poured into an aqueous 33% aqueous sodium bisulfate solution (1.12 mL) and then stirred for 10 minutes. Aqueous 33% sodium hydroxide solution (1.8 mL) was then added to adjust the pH to 5. The resulting mixture was stirred at room temperature for 1 hour. The solid was collected solid by filtration, washed with water (10 mL) and vacuum dried at about 60°C to afford the title compound (634 mg, crude) as a white solid. LC/MS (exact mass) calculated for C ig H 22 N 4 0 5 S 2 ; 450.103, found (M + H + ); 451.3.

Step 7: cis-[3-(Methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d ]pyrimidin-4- yl}amino)cyclobutyl]methanesulfonyl chloride

Thionyl chloride (0.3 ml, 3.33 mmol) was added dropwise over 5 minutes to a solution of cis-[3-(methyl{7-[(4- methyl phenyl)sulfonyl]-7H-pyrrolo[2, 3-d] pyrimidi n-4- yl}amino)cyclobutyl]methanesulfonic acid (150 mg, 0.33 mmol) in dichloromethane (20 mL) at 0°C. Two drops of Ν,Ν-dimethylformamide were added to the solution, which was then heated at 75°C for 2 hours. The mixture was cooled and the solvent was evaporated. The residue was washed with anhydrous dichloromethane (3 x 10 mL) to afford the crude title compound (170 mg) as a yellow solid. LC/MS (exact mass) calculated for C ig H 2i CIN 4 0 4 S 2 ; 468.069, found (M + H + ); 469.2. Step 8: N-(cis-3-{[(4,4-Difluoropiperidin- 1 -yl)sulfonyl]methyl}cyclobutyl)-N-methyl-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-amine

To a mixture of 4,4-difluoropiperidine (77 mg, 0.64 mmol) and triethylamine (97 mg, 0.96 mmol) in tetrahydrofuran (20 mL) at 0°C was added dropwise a solution of cis-[3- (methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2 , 3-d]pyrimidin-4- yl}amino)cyclobutyl]methanesulfonyl chloride (150 mg, 0.320 mmol) in tetrahydrofuran (10 ml_).The mixture was allowed to warm to room temperature overnight. The solvent was evaporated and the residue was taken up in ethyl acetate (80 mL). The solution was washed with brine (30 mL), dried over sodium sulfate and concentrated to afford the crude title compound (134 mg) as a white solid . LC/MS (exact mass) calculated for C 24 H 2g F 2 N ¾ 0 4 S 2 ; 553.651 , found (M + H + ); 554.3.

Step 9: N-(cis-3-{[(4,4-difluoropiperidin- 1 -yl)sulfonyl]methyl}cyclobutyl)-N-methyl-7H-pyrrolo[2,3- d]pyrimidin-4-amine

N-(cis-3-{[(4,4-Difluoropiperidin- l-yl)sulfonyl] methyl}cyclobutyl)-N-methyl-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidi n-4-amine (134 mg, 0.24 mmol) and lithium hydroxide monohydrate (51 mg, 1.21 mmol) were combined in a mixture of ethanol (14 mL) and water (7 mL) and then heated at 50°C overnight. The reaction was concentrated under vacuum and diluted with ethyl acetate (100 mL). The resulting solution was washed with brine (30 mL), dried over sodium sulfate and concentrated. The residue was chromatographed using pre- parative thin layer chromatography eluting with a mixture of ethyl acetate and methanol (20: 1) to afford the title compound (31 mg, 32.3 %) as a white solid. Ή NMR (400 MHz, methanol-d 4 ): δ 8.13 (s, 1H), 7.13 (d, J = 3.6, 1H), 6.70 (d, J = 3.6, 1H), 5.12 (m, 1H), 3.49-3.47 (m, 4H), 3.46 (m, 3 H), 3.33 (m, 2H), 2.62-2.54 (m, 3 H), 2.25-2.20 (m, 1H), 2.11-2.05 (m, 2H). LC/MS (exact mass) calculated for C 1? H 23 F 2 N 5 0 2 S; 399.154, found (M + H + ); 400.3.

Examples 26 to 29. The following compounds were made starting from cis-[3-(methyl{7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidi n-4-yl}amino)cyclobutyl]methanesulfonyl chloride (Example 25 , Step 7), according to the procedures of Example 25 , Step 8

(sulfonylation) and step 9 (deprotection), substituting the appropriate amine for 4,4- difluoropiperidine in Step 8.

Example 26: 1 -[({cis-3-[Methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}- methyl)sulfonyl]-4-(trifluoromethyl)piperidin-4-ol The title compound (31mg) was prepared using 4-(trifluoromethyl)piperidin-4-ol in the sulfonylation step and was deprotected using the method from Example 25 , Step 9. The compound was purified using preparative thin layer chromatography eluting with eluting with a mixture of ethyl acetate and methanol (20: 1). Ή NMR (400 MHz, methanol-d 4 ): δ 8.13 (s, 1H), 7.13 (d , J = 3.6, 1 H), 6.70 (d, J = 3.6, 1H), 5.10-5.08 (m, 1 H), 3.74-3.71 (m, 2 H), 3.36 (m, 3 H), 3.32-3.27 (m, 2 H), 3.19-3.13 (m, 2 H), 2.62-2.54 (m, 3 H), 2.25-2.21 (m, 2 H), 1.86- 1.84 (m, 4 H). LC/MS (exact mass) calculated for C 18 H 24 F 3 N 5 0 3 S; 447.155 , found (M + H + ); 448.3.

Example 28 and 29: (3R) and (3S)- 1 -[({cis-3-[Methyl(7H-pyrrolo[2, 3-d]pyrimidin-4- yl)amino]cyclobutyl}methyl)sulfonyl]pyrrolidine-3-carbonitri le

The title compounds (330 mg total) were prepared using pyrrolidine-3-carbonitrile enriched as an 80:20 scalemic mixture of (3R)-pyrrolidine-3-carbonitrile and (3S)-pyrrolidine-3- carbonitrile enantiomers in the sulfonylation step and was de-protected using the method from Example 19, Step 2. The compound was purified by chromatography on si lica gel eluting with gradient of petroleum ether and ethyl acetate (10: 1 to 1: 10). LC/MS m/z= 375.2 (M + 1). The enantiomers were separated by preparative supercritical fluid chromatography:

3 R-enantiomer(28): 178 mg. Ή NMR (400 MHz, CDCI 3 ): δ 8.31 (s, 1 H), 7.06 (d , 1 H), 6.58 (d, 1 H), 5.19-5.10 (m, 1 H), 3.77-3.75 (m, 1 H), 3.61-3.54 (m, 3 H), 3.33 (s, 3 H), 3.21-3.19 (m, 3 H), 2.69-2.66 (m, 3 H), 2.36-2.31 (m, 2 H), 2.14-2.11 (m, 2 H). LC/MS (exact mass) calculated for C i7 H 22 N 6 0 2 S; 374.15 , found (M + H + ); 375.2. Chiral HPLC retention time = 2.65 minutes

3S-enantiomer(29): 31 mg. Ή NMR (400 MHz, CDCI 3 ): δ 8.31 (s, 1H), 7.06 (d, 1 H), 6.58 (d, 1 H), 5.19-5.10 (m, 1 H), 3.77-3.75 (m, 1 H), 3.61-3.54 (m, 3 H), 3.33 (s, 3 H), 3.21-3.19 (m, 3 H), 2.69-2.66 (m, 3 H), 2.36-2.31 (m, 2 H), 2.14-2.11 (m, 2 H). LC/MS (exact mass) calculated for C i7 H 22 N 6 0 2 S; 374.15 , found (M + H + ); 375.2 Chiral HPLC retention time = 2.53 minutes

Example 30: N-{cis-3-[(Butylsulfonyl)methyl]cyclobutyl}-N-methyl-7H-pyrr olo[2,3-d]pyrimidin-4- amine

Step 1: N-{cis-3-[(Butylthio)methyl]cyclobutyl}-N-methyl-7H^yrmlo[2, 3-d]pyrimidiri-4-amiiie

A solution of cis-[3-(methyl{7-[(4-methyl phenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyri midin- 4-yl}amino)cyclobutyl]methyl 4-methylbenzenesulfonate from Example 25 , Step 4 (23 g, 42.6 mmol) was stirred in N-methylpyrrolidine (100 mL). Then l,8-d iazabicycloundec-7-ene (12.8 g, 85.2 mmol) and 1-butanethiol (7.8 g, 85.2 mmol) was added to the reaction mixture. The reaction was stirred at room temperature for 16 hours. Water (200 mL) and ethyl acetate (500 mL) were added. The aqueous layer was extracted with ethyl acetate (2 x 500 mL) and the com- bined organic layers were dried and concentrated. The residue was chromatographed on silica gel eluting with a gradient of dichloromethane and methanol (100:0 to 90: 10) to afford the title compound (11.8 g, 91%). LC/ MS (exact mass) calculated for C 16 H 24 N 4 S; 304.172, found (M + H + ); 305.3.

Step 2 : N-{cis-3-[(Butylsulfonyl)methyl]cyclobutyl}-N-methyl-7H-pyr lo[2,3-d]pyrimidiri-4-amine N-{cis-3-[(Butylthio)methyl]cyclobutyl}-N-methyl-7H-pyrrolo[ 2 , 3-d]pyrimidin-4-amine (12 g, 39.5 mmol) was dissolved in a mixture of tetrahydrofuran (200 mL), ethanol (200 mL) and water (200 mL). Potassium peroxomonosulfate (48.6 g, 79.0 mmol) was added and the reaction was stirred at room temperature for 1 hour. The mixture was then filtered ; the solids were washed with a mixture of tetrahydrofuran (40 mL), ethanol (40 mL) and water (20 mL). The filtrate was treated with aqueous 10% sodium bisulfite solution (200 mL) and stirred at room temperature for 20 minutes. A saturated solution of aqueous sodium bicarbonate was added to adjust the pH to ~7. The mixture was extracted with dichloromethane (3 x 800 mL) and the combined organic layers were dried and concentrated under vacuum. The crude residue was chromatographed on silica gel eluting with a g radient of dichloromethane and methanol (100:0 to 95 : 5) to obtain the title compound (11.4g, 86%). Ή NMR (400 MHz, methanol-d 4 ): δ 8.13 (s,

1 H), 7.13-7.12 (m, 1 H), 6.70-6.69 (m, 1 H), 5.13-5.10 (m, 1 H), 3.42 (s, 3 H), 3.33 (m, 2 H), 3.11-3.07 (m, 2 H), 2.65-2.63 (m, 3 H), 2.29-2.25 (m, 2 H), 1.86- 1.78 (m, 2 H), 1.55- 1.50 (m,

2 H), 1.03-0.99 (m, 3 H). LC/ MS (exact mass) calculated for C 16 H 24 N 4 0 2 S; 336.162, found (M + H + ); 337.3

Example 31: N-Methyl-N-(trans-3-((propylsulfonyl)methyl)cyclobutyl)-7H-p yrrolo[2,3-d]pyrimidin- 4-amine

The title compounds were made as a mixture of cis and trans isomers (50 mg) starting from cis and trans-[3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2 , 3-d]pyrimidin-4- yl}amino)cyclobutyl]methyl 4-methylbenzene sulfonate (cis/trans = 10: 1) (Example 25 , Step 4), according to procedures similar to those of Example 30, Steps 1 and 2, using propane- l-thiol instead of butane- l-thiol in Step 2. The mixture of cis and trans isomers was purified by reverse phase high performance liquid chromatography eluting with gradient of water and ace- tonitrile (95 : 5 to 5 :95). LC/MS (exact mass) calculated for C i5 H 22 N 4 0 2 S; 322.15 , found (M + H + ); 323.2

The cis and trans isomers were then separated by preparative supercritical fluid chromatography. trans isomer (31), 12 mg : Ή NMR (400 MHz, methanol-d 4 ): δ 8.12 (s, 1 H), 7.13-7.12 (m, 1 H), 6.69-6.66 (m, 1 H), 5.45-5.41 (m, 1 H), 3.46-3.44 (m, 2 H), 3.36 (s, 3 H), 3.11-3.09 (m, 2 H), 2.88-2.86 (m, 1 H), 2.75-2.67 (m, 2 H), 2.40-2.38 (m, 2 H), 1.91-1.86 (m, 2 H), 1.12- 1.10 (m, 3 H). LC/ MS (exact mass) calculated for C 15 H 22 N 4 0 2 S; 322.15 , found (M + H + ); 323.2

cis isomer, 36 mg : Ή NMR (400 MHz, methanol-d 4 ): δ 8.12 (s, 1 H), 7.13-7.12 (m, 1 H), 6.70- 6.69 (m, 1 H), 5.10- 5.20 (m, 1 H), 3.36 (s, 3 H), 3.33-3.32 (m, 2 H), 3.08-3.04 (m, 2 H), 2.64- 2.61 (m, 3 H), 2.24-2.22 (m, 2 H), 1.90- 1.84 (m, 2 H), 1.13- 1.09 (m, 3 H). LC/ MS (exact mass) calculated for C i5 H 22 N 4 0 2 S; 322.15 , found (M + H + ); 323.2 Example 32 : N-(cis-3-{[(2-Cyclopropylethyl)sulfomyl]methyl}cyclobutyl)-N -methyl-7H-pyrmlo[2,3- d]pyrimidin-4-amine

Step 1: N-(cis-3-{[(2-Cyclopropylethyl)sulfanyl]methyl}cyclobutyl)-N -methyl-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-amine

Nitrogen was bubbled through a mixture of S-{[cis-3-(methyl{7-[(4- methyl phenyl)sulfonyl]-7H-pyrrolo[2, 3-d] pyrimidi n-4-yl}amino)cyclobutyl] methyl}

ethanethioate Example 25 , Step 5 (190 mg, 0.43 mmol) and potassium carbonate (129 mg, 0.94 mmol) in methanol (10 mL) at 0 ° C for 2 minutes. 2-Cyclopropylethyl 4- methylbenzenesulfonate (159 mg, 1.53 mmol) was then added and the solution was stirred for 6 hours at room temperature. Dichloromethane (30 mL) and water (20 mL) were added and the aqueous layer was extracted with dichloromethane (2 x 20 mL). The combined organic layers were washed with brine, dried over sodium sulfate and concentrated. The crude product was purified by preparative thin layer chromatography using ethyl acetate-petroleum ether (1 :2) to give the title compound as a white solid (62 mg, 31%). LC/MS (exact mass) calculated for C H N O S ; 470.18, found (M + H + ); 471.1

Step 2 : N-(cis-3-{[(2-Cyclopropylethyl)sulfonyl]methyl}cyclobutyl)-N -methyl-7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-amine

The mixture of N-(cis-3-{[(2-cyclopropylethyl)sulfanyl]-methyl}cyclobutyl)- N-methyl-7- [(4-methylphenyl)-sulfonyl]-7H-pyrrolo[2,3-d] pyrimidin-4-amine (24 mg, 0.05 1 mmol) and potassiu m peroxomonosulfate (49 mg, 0.079 mmol) in tetrahydrofuran (1.2 mL), water (0.6 mL) and ethanol (1.2 mL) was stirred at room temperature for 20 minutes. Aqueous sodiu m bisulfite was added, followed by dichloromethane (20 mL). The aqueous layer was extracted with dichloromethane (2 x 20 mL) and the combined organic layers were extracted with brine, dried over sodium sulfate and concentrated. The crude material was used directly in next step. LC/MS (exact mass) calculated for C 24 H 30 N 4 O 4 S 2 ; 502.17, found (M + H + ); 503.3 Step 3 : N-(cis-3-{[(2-Cyclopmpylethyl)sulfonyl]methyl}cyclobutyl)-N- methyl-7H-pyrrolo[2, 3- d]pyrimidin-4-amine

The mixture of N-(cis-3-{[(2-cyclopropylethyl)sulfonyl]methyl}cyclobutyl)-N -methyl-7- [(4-methylphenyl)sulfonyl]-7H-pyrrolo[2 , 3-d]pyrimidin-4-amine (49 mg, 0.097 mmol) and lithium hydroxide (30 mg, 1.3 mmol) in water (5 mL) and ethanol (10 mL) was stirred at 50°C for 2 hours. Then, dichloromethane (20 mL) was added and the aqueous layer was extracted with dichloromethane. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated. The crude product was purified by reverse phase high performance liquid chromatography using a gradient of water and acetonitrile gradient (95 : 5 to 5 :95) to give the title compound (14 mg, 40%) as white solid. Ή NM R (400 MHz, methanol-d 4 ): δ 8.12 (s, 1

H), 7.13 (d, 2 H), 6.70 (d, 2 H), 5.12-5.09 (m, 1 H), 3.34 (s, 3 H), 3.34-3.33 (m, 2 H), 3.20-3.17 (m, 2 H), 2.64-2.61 (m, 3 H), 2.26-2.22 (m, 2 H), 1.75- 1.69 (m, 2 H), 0.89-0.86 (m, 2 H), 0.56- 0.52 (m, 2 H), 0.18-0.17 (m, 2 H). LC/MS (exact mass) calculated for C i7 H 24 N 4 0 2 S; 348.16, found (M + H + ); 349.1

Example 33: N-[cis-3-({[(3,3-Difluorocyclobutyl) ethyl]sulfonyl}-met yl)cyclobutyl]-N-niethyl-7H- pyrrolo[2, 3-d]pyrimidin-4-amine

Step 1: N-[cis-3-({[(3,3-Difluorocyclobutyl)methyl]sulfanyl}methyl)c yclobutyl]-N-methyl-7H- pyrrolo[2, 3-d]pyrimidin-4-amine

Nitrogen was bubbled through a mixture of S-{[cis-3-(methyl{7-[(4-methylphenyl)- sulfonyl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}amino)cyclobutyl]m ethyl} ethanethioate, Example 25 , Step 5 (250 mg, 0.56 mmol) and potassium carbonate (194 mg, 1.41 mmol) in methanol (100 mL) for two minutes at 0°C followed by addition of (3,3-difluorocyclobutyl)methyl 4- methylbenzenesulfonate (prepared as described i n WO2004/032834) (310 mg, 1.12 mmol). The mixture stirred for 6 hours at room temperature, filtered, and concentrated to give the title compound (270 mg, crude) as white solid. LC/ MS (exact mass) calculated for C 17 H 22 F 2 N 4 S;

352.15 , found (M + H + ); 353.2

Step 2 : N-[cis-3-({[(3,3-Difluomcyclobutyl)methyl]sulfonyl}methyl)cy clobutyl]-N- ethyl-7H- pyrrolo[2, 3-d]pyrimidin-4-amine

The mixture of N-[cis-3-({[(3 ,3-difluorocyclobutyl)methyl]sulfanyl}-methyl)cyclobutyl]-

N-methyl-7H-pyrrolo[2 ,3-d]pyrimidin-4-amine (45 mg, 0.13 mmol) and potassium peroxomonosulfate (157 mg, 0.26 mmol) in a mixture of tetrahydrofuran (20 mL), water (10 mL) and ethanol (20 mL) was stirred at room temperature for 20 minutes. Aqueous sodium bisulfite was then added , followed by dichloromethane (20 mL). The aqueous layer was extracted with dichloromethane (2 x 20 mL) and the combined organic layers were extracted with brine, dried over sodium sulfate, and concentrated. The crude product was purified by reverse phase high performance liquid chromatography using water-acetonitrile gradient (95 : 5 to 5 :95) to give the title compound as white solid (34 mg, 39%). Ή NMR (400 MHz, methanol-d 4 ): δ 8.29 (s, 1 H), 7.42 (d, 1 H), 7.03 (d, 1 H), 4.86 (m, 1 H), 3.51 (s, 3 H), 3.39-3.33 (m, 4 H), 2.84 (m, 1 H), 2.76-2.71 (m, 4 H), 2.53 (m, 2 H), 2.37-2.34 (m, 2 H). LC/MS (exact mass) calculated for C H F N O S; 384.14, found (M + FT); 385.1.

Example 34A and 34B: (I R, 3R) and (IS,

yl)amino]cyclobutyl}methyl)sulfonyl]cyclopentanecarbonitr ile

The title compound mixture of (1R, 3R) and (IS, 3S)-3-[({cis-3-[methyl(7H-pyrrolo[2,3- d]pyrimidin-4-yl)amino]cyclobutyl}-methyl)sulfonyl]cyclopent ane-carbonitrile was prepared from S-{[cis-3-(methyl{7-[(4-methyl phenyl)sulfonyl]-7H-pyrrolo[2 ,3-d]pyri midin-4- yl}amino)cyclobutyl]methyl} ethanethioate Example 25 , Step 5 , according to the procedure of Example 30, steps 1 and 2.

The title compound (180 mg) was separated by preparative supercritical fluid chromatography using a Chiralpak AS column:

(1R, 3R) enantiomer 34A: 60 mg, Ή NMR (400 MHz, methanol-d 4 ): δ 8.12 (s, 1 H), 7.13-7.12 (d, 1 H), 6.70-6.69 (d, 1 H), 5.17-5.11 (m, 1H), 3.86-3.78 (m, 1H), 3.41-3.36 (m, 5 H), 3.15- 3.11 (m, 1 H), 2.63-2.53 (m, 4 H), 2.37-2.13 (m, 6 H), 2.03- 1.91 (m, 1 H). LC/ MS (exact mass) calculated for C 17 H 22 F 2 N 4 0 2 S; 373.16, found (M + H + ); 374.1

(IS, 35) enantiomer 34B: 27 mg, LC/MS (exact mass) calculated for C 17 H 22 F 2 N 4 0 2 S; 373.16, found (M + H + ); 374.1

The intermed iate 3-cyanocyclopentyl 4-methylbenzenesulfonate used in step- 1 was prepared as shown below:

3-Cyanocyclopentyl 4-methylbenzenesulfonate

4-Methylbenzene- l-sulfonyl chloride (6.9 g, 36 mmol) and N,N-dimethyl pyridin-4- amine (100 mg) were added to a solution of compound 3-hydroxycyclopentane-carbonitrile (J. Org. Chem. 2007, 72 , 7423) (2 g, 18 mmol) and triethylamine ( 5.5 g, 54 mmol) in dichloro- methane (100 mL). The reaction was stirred at room temperature for 15 hours and then quenched mixture was quenched by addition of saturated aqueous sodium bicarbonate solution (20 mL). The mixture was extracted with dichloromethane (4 x 50 mL). The combined organic layers were dried over magnesium sulfate and concentrated. The residue was chromatographed on silica gel by eluting with a mixture of petroleum ether and ethyl acetate (1 : 1) to give the title compound as yellow oil (0.5 g, 11% yield). LC/MS (exact mass) calculated for C 13 H 15 N0 3 S;

265.08, found (M + 23); 287.9.

Example 35: Racemic N-methyl-N-[cis-3-({[l -(propcm-2-yl)pyrrolidin-3- yl]sulfonyl}methyl)cyclobutyl]-7H-pyrrolo[2, 3-d]pyrimidin-4-a ine

Step 1: ten-Butyl 3-({[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2 ,3-d]pyrimidiri-4- yl}amino)cyclobutyl]methyl}sulfcmyl)pyrrolidine- l -carboxylate

The solution of cis-[3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3- d]pyrimidin-4-yl}amino)cyclobutyl]methyl 4-methylbenzenesulfonate from Example 25 , Step 4 (2 g, 3.7 mmol) was stirred in N-methylpyrrol idine (40 mL). l,8-Diazabicycloundec-7-ene (1.13 g, 7.4 mmol) and 3-mercapto-pyrrolidine- l-carboxylic acid tert-butyl ester (1.13 g, 5.6 mmol) were then added to the reaction mixture. The reaction was stirred at room temperature for 16 hours. Water (200 mL) and ethyl acetate (500 mL) were added. The aqueous layer was extracted with ethyl acetate (2 x 500 mL).and the combined organic layers were dried and concentrated under vacuum to give the title compound as a wh ite solid (2.6 g, 118%). LC/MS (exact mass) calculated for C 28 H 3? N 5 0 4 S 2 ; 571.23, found (M + H + ): 572.1.

Step 2 : N-Methyl-7-[(4-methylphenyl)sulfonyl]-N-{cis-3-[(pyrrolidin- 3- ylsulfanyl)methyl]cyclobutyl}-7H-pyrwlo[2,3-d]pyrimidin-4-am ine

To a solution of tert-butyl 3-({[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H- pyrrolo[2 ,3-d]pyrimidin-4-yl}amino)cyclobutyl]methyl}sulfanyl)pyrroli dine-l-carboxylate (2.6 g, 4.5 mmol) in methanol (15 mL) was added 3M hydrochloric acid solution in methanol (40 mL). The resulting solution was stirred at room temperature for 1 hour. The solution was concentrated to give the crude product, which was purified by chromatography on silica gel eluting with a gradient of dichloromethane and methanol (100:0 to 85 : 15) to give the title compound as colorless oil (1.7 g, 52%). Ή NMR (400 MHz, methanol-d 4 ): δ 8.21 (s, 1 H), 7.99 (d, 2 H), 7.56 (d, 1 H), 7.37 (d, 2 H), 6.88 (d, 1 H), 4.95-4.87 (m, 1H), 3.29 (s, 1 H), 3.27 (s, 3 H), 3.21-3.17 (m, 1 H), 3.04-3.96 (m, 1 H), 2.92-29 (m, 1 H), 2.72-2.01 (m, 3 H), 2.50-2.43 (m, 2 H), 2.39 (s, 3 H), 2.29-2.15 (m, 2 H), 2.03-2.01 (m, 2 H), 1.98- 1.65 (m, 1 H). LC/MS (exact mass) calculat- ed for C 23 H 2g N 5 0 2 S 2 ; 471.18, found (M + 23): 494.

Step 3 : N-Methyl-7-[(4-methylphenyl)sulfonyl]-N-[cis-3-({[l -(propcm-2-yl)pyrrolidin-3- yl]sulfanyl}methyl)cyclobutyl]-7H-pyrrolo[2, 3-d]pyrimidin-4-amine

To a solution of N-methyl-7-[(4-methylphenyl)sulfonyl]-N-{cis-3-[(pyrrolidin- 3- ylsulfanyl)methyl]cyclobutyl}-7H-pyrrolo[2 , 3-d]pyrimidin-4-amine (472 mg, 1 mmol) in di- chloromethane (50 mL) was added acetone (174 mg, 3 mmol), 4A molecular sieves (40 mg) and sodiu m cyanoborohydride (189 mg , 3 mmol). The resulting solution was stirred at room temperature for 1 hour, then diluted with dichloromethane (70 mL) and water (70 mL). The aqueous layer was extracted with dichloromethane (2 x 50 mL) and the combined organic layers were washed with brine (100 mL). The organic layer was dried over anhydrous sodium sulfate and concentrated to give title compound (500 mg , 97% yield) as colorless oi l. LC/MS (exact mass) calculated for C 26 H 35 N 5 0 2 S 2 ; 513.22, found (M + H + ); 514.1.

Step 4: N-Methyl-7-[(4-methylphenyl)sulfonyl]-N-[cis-3-({[l -(propan-2-y\)pyrro\ d n-3- yl]sulfonyl}methyl)cyclobutyl]-7H-pyrrolo[2,3-d]pyri idin-4-a ine

The mixture of N-methyl-7-[(4-methylphenyl)sulfonyl]-N-[cis-3-({[l-(propan- 2- yl)pyrrolidin-3-yl]sulfanyl}methyl)cyclobutyl]-7H-pyrrolo[2, 3-d]pyrimidin-4-amine_(500 mg, 1.0 mmol) and potassium peroxomonosulfate (1.23 g, 2.0 mmol) in tetrahydrofuran (20 mL), water (10 mL), and ethanol (20 mL) was stirred at room temperature for 30 minutes. The reaction solution was diluted with ethyl acetate (100 mL) and water (50 mL). The aqueous layer was extracted with ethyl acetate (3 x 50 mL) and the combined organic layers were washed with brine (100 mL), dried over sodiu m sulfate and concentrated to give title compound as colorless oil (420 mg, 90%). LC/MS (exact mass) calculated for C 26 H 35 N 5 0 4 S 2 ; 545.21, found (M + H + ): 546.3.

Step 5 : N-Methyl-N-[cis-3-({[ 1 -(propcm-2-yl)pyrrolidin-3-yl]sulfonyl}methyl)-cyclobutyl]-7 H- pyrro\o[2, 3-d]pyrimidin-4-amine

The mixture of N-methyl-7-[(4-methylphenyl)sulfonyl]-N-[cis-3-({[l-(propan- 2- yl)pyrrolidin-3-yl]sulfonyl}methyl)cyclobutyl]-7H-pyrrolo[2, 3-d]pyrimidin-4-amine_(330 mg, 0.6 mmol) and lithium hydroxide (126 mg, 3 mmol) in a mixture of water (5 mL) and ethanol (10 mL) was sti rred at 50°C for 2 hours. The mixture was then concentrated and the residue was taken up in ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate and concentrated. The crude product was purified by reverse phase high performance liquid chromatography using a gradient of water and acetonitrile (95 : 5 to 5 :95) to give the title compound (89 mg, 38%) as white solid . Ή NMR (400 MHz, CDCI 3 ): δ 8.32 (s, 1 H), 7.07 (d, 1 H), 6.57 (d , 1 H), 5.13 (m, 1H), 3.57 (m, 1H), 3.33 (s, 3 H) 3.05-3.22 (m, 3 H), 2.92 (m, 1H), 2.78 - 2.87 (m, 1H), 2.58-2.77 (m, 4H), 2.50 (m, 1H), 2.19-2.34 (m, 2 H), 2.06-2.19 (m, 2 H), 1.12 (d, 6H). LC/MS (exact mass) calculated for C ig H 2g N 5 0 2 S; 391.20, found (M + H + ); 392.3

Example 36: N-(cis-3-{[(3-Chloro-4-fluomphenyl)sulfonyl]methyl}cyclobuty l)-N-methyl-7H- pyrrolo[2, 3-d]pyrimidin-4-amine Step 1: N-(cis-3-{[(3-Chloro-4-fluomphenyl)sulfanyl]methyl}cyclobuty l)-N-methyl-7H-pyrrolo[2,3- d]pyrimidin-4-amine

To a solution of 3-chloro-4-fluorothiophenol (93 mg, 0.55 mmol) in tetrahydrofuran (1.5 mL) was added 50% aqueous sodium hydroxide (44 mg, 0.55 mmol) and ethanol (1.5 mL). The mixture was stirred at room temperature for 1 hour. A solution of cis-[3-(methyl{7-[(4- methylphenyl)sulfonyl]-7H-pyrrolo[2, 3-d] pyrimidi n-4-yl}amino)cyclobutyl] methyl 4- methylbenzenesulfonate from Example 25 , Step 4 (200 mg, 0.37 mmol) in tetrahydrofuran (1.5 mL) was added to the reaction mixture. The combined mixture was heated at 40°C overnight. The reaction was concentrated and purified by silica column eluting with a gradient of heptanes and ethyl acetate (90: 10 to 0: 100) to afford the title compound (69 mg, 49.6%). Ή NMR (400 MHz, CDCI 3 ): δ 8.18 (s, 1 H), 7.39 (dd, 1 H), 7.28 (s, 1 H), 7.03-7.08 (m, 1 H), 7.00 (d , 1 H), 6.52 (d , 1 H), 4.97- 5.07 (m, 1 H), 3.35 (m, 2 H), 3.23 (s, 3 H), 2.89 (s, 1 H), 2.43-2.52 (m, 2 H), 2.19-2.30 (m, 2 H).

Step 2 : N-(cis-3-{[(3-Chloro-4-fluorophenyl)sulforiyl]methyl}cyclobu tyl)-N-methyl-7H-pyrrolo[2,3- d]pyrimidin-4-amine

To a solution of N-(cis-3-{[(3-chloro-4-fluorophenyl)sulfanyl] methyl}cyclobutyl)-N- methyl-7H-pyrrolo[2 , 3-d]pyrimidin-4-amine (75 mg, 0.2 mmol) in dichloromethane (10 mL) was added 3-chlorobenzoperoxoic acid (107 mg). The reaction was stirred at room temperature overnight and then concentrated. The crude residue was chromatographed on silica gel eluting with a gradient of dichloromethane and 2 M ammonia in methanol (80:20) to obtain the title compound (48 mg, 59.2%). Ή NMR (400 MHz, methanol-d 4 ): δ 8.31 (s, I H), 8.02 (m, IH), 7.81- 7.83 (m, IH) 7.37-7.27 (m, IH), 7.09 (d, IH), 6.65 (s, IH), 5.18-5.10 (m, 1 H), 4.15-4.09 (m, 1 H), 3.32 (m, 5 H), 2.59-2.54 (m, 2 H), 2.44-2.42 (m, 2 H). LC/MS (exact mass) calculated for C 18 H 18 CIFN 4 0 2 S; 408.08, found (M + H + ); 409

Example 37: 2-[({cis-3-[Methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)ammo]cyclobutyl}methyl) sulfonyl]pyridine-4-carbonitrile

Step 1: 2-({[cis-3-(Methyl{7-[(4-methylphenyl)sulfoiiyl]-7H-pyrrolo[ 2, 3-d]pyrimidin-4- yl} mino)cyclobutyl] ethyl}sulfanyl)pyridine-4-c rbonitrile

l,8-Diazabicycloundec-7-ene (24.6 g, 161 mmol) and 2-mercapto-isonicotinonitrile

(16.1 g, 118 mmol) were added to a solution of [cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]- 7H-pyrrolo[2 , 3-d]pyrimidin-4-yl}amino)cyclobutyl]-methyl methanesulfonate (50 g, 110 mmol) in N-methylpyrrolidine (250 mL). The reaction was heated at 50°C overnight. Additional 2- mercaptoisonicotinonitrile (8.1 g, 59 mmol) was added to drive the reaction to completion. The mixture was cooled to about 0°C and then the reaction was quenched by dropwise addition of water. The solids were collected by filtration, washed with water, and dried under vacuum at 50 C to give the title compound as a bright yellow solid (45.8 g, 82.8%). LC/MS (exact mass) calculated for C H N 0 S ; 504.14, found (M + H + ); 505.1

Step 2 : 2-[({cis-3-[Methyl(7H-pyrrolo[2,3-d]pyrimidin-4- yl)amino]cyclobutyl}methyl)sulfanyl]pyridine-4-carbonitrile

To a solution of 2-({[cis-3-(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2 ,3- d]pyrimidin-4-yl}amino)cyclobutyl]methyl}sulfanyl)pyridine-4 -carbonitrile (45.3 g, 89.8 mmol) in tetrahydrofuran (180 mL) was added a solution of 1M tetrabutylammonium fluoride in tetrahydrofuran (269 mL). The reaction mixture was heated to reflux for 6 hours and then cooled to room temperature. Water was added dropwise over 45 minutes. The solids were collected by filtration and washed with a mixture of 20% tetrahydrofuran (33 mL) and water (97 mL). The wet cake was dried under vacuum at 50 C to give the title compound as a tan solid (25 g, 79%). LC/MS (exact mass) calculated for C i8 H i8 N 6 S 2 ; 350.13, found (M + H + ); 351.1

Step 3 : 2-[({cis-3-[Met yl(7H-pyrrolo[2,3-d]py midin-4- yl)amino]cyclobutyl}methyl)sulfonyl]pyridine-4-carbonitrile

Potassium peroxomonosulfate (236.8 g, 385.2 mmol) was added slowly to a mixture of 2-[({cis-3-[methyl(7H-pyrrolo[2, 3-d]pyrimidin-4-yl)amino]cyclobutyl}- methyl)sulfanyl]pyridine-4-carbonitrile (22.5 g, 64.2 mmol) in methanol (337 mL) and water (56 mL) at 0°C. The reaction was stirred at 3°C for 20 hours. The reaction was quenched using 10% aqueous sodium bisulfate solution (40 mL). and the resulting slurry was stirred at room temperature for 2 hours. Aqueous 10% potassium carbonate solution was added until the pH was 4 to 5. The material was filtered and rinsed with water. The wet filter cake was dried under vacuum at 40°C to give an off-white solid. This material was taken up in tetrahydrofuran (50 mL) and heated to reflux for 3 hours. The mixture was cooled to room temperature and filtered to collect the solid, which was dried under vacuum at 40 C to the title compound as a light tan powder (17.3 g, 70.46%). Ή NMR (400 MHz, CDCI 3 ): δ 11.97 (s, 1 H), 8.95 (d, 1 H), 8.33-8.28 (m, 2 H), 7.81 (d, 1 H) 7.1 (d, 1 H), 6.54 (d, 1 H), 5.13-5.08 (m, 1 H), 3.63 (m, 2 H), 3.30 (s, 3 H), 2.54-2.48 (m, 3 H), 2.09-2.07 (m, 2 H). LC/MS (exact mass) calculated for C H N O S : 382.12, found (M + H + ); 383.1.

Example 38. 2-Met yl-N-{cis-3-[ ethyl(7H-pyrrolo[2,3-d]pyri idin-4-yl)a ino]cyclobutyl}- l ,3- thiazole-5-sulfonamide Step 1: 2-Methyl-N-[cis-3-( ethyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2,3-d]pyrimidi n-4- yl}amino)cyclobutyl]- 1 , 3-thiazole-5-sulfonamide

Triethylamine (62. Og, 0.613 mol) is added to a solution of cis-N-methyl-N-7H- pyrrolo[2 ,3-d]pyrimidin-4-ylcyclobutane- l,3-diamine hydrochloride (22.2. g, 0.102 mol) in di- chloromethane (250 mL). 2-Methylthiazole-5 -sulfonyl chloride (28. Og, 0.142 mol) in dichloro- methane (250 mL) is added over 30 minutes at room temperature to the reaction mixture. After 1.5 hours the solvent is removed under reduced pressure and the resultant solid dissolved in 4: 1 ethyacetate:dichloromethane (400 mL). The solution is filtered through a 40g silica plug, rinsing with ethylacetate (800 mL) and dichloromethane (100 mL). The solvent from the filtrate is removed under reduced pressure to give solid (59g). The solid is purified using silica gel column chromatography eluting from 1: 1 dichloromethane:ethylacetate to neat ethylacetate to give the title compound (44.4g, 81%); m/z (CI) 533 [M + H] + .

Step 2 : 2-Methyl-N-{cis-3-[methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl) ' o]cyclob tyl}- l ,3-thi zole- 5-sulfonamide

Lithium hydroxide (12. lg, 0.505 mol) in water (290 mL) is added to 2-methyl-N-[cis-3-

(methyl{7-[(4-methylphenyl)sulfonyl]-7H-pyrrolo[2 , 3-d]pyrimidin-4-yl}amino)cyclobutyl]-l,3- thiazole-5-sulfonamide (43.8g, 82.2 mmol) in isopropyl alcohol (435 mL) and the mixture heated to 60°C overnight. After cooling to room temperature the reaction mixture is filtered rinsing with water (145 mL). The filtrate is pH is adjusted to 6-7 using 6M aqueous hydrochloric acid. The reaction slurry is concentrated under reduced pressure. Water (370 mL) is added and the mixture cooled to 0°C. The solved is collected by filtration, washed with cold water (150 ml) then vacuum dried at 60°C overnight to give the title compound (25. Og, 80%); Ή NMR (DMSO- d6): δ 11.66- 11.71 (1 H), 8.44-8.47 (1 H), 8.11-8.08 (2 H), 7.16-7.17 (1 H),6.63-6.65 (1 H), 4.86-4.94 (1 H), 3.58-3.68 (1 H), 3.22 (3 H), 2.74 (3 H), 2.40-2.46 (2 H), 2.10-2.18 (2H). m/z (CI) 379 [M + H] + .

Biological Evaluation

JAK Caliper Enzyme Assay at ImM ATP

Test article was solubilized in dimethyl sulfoxide (DMSO) to a stock concentration of 30 mM. An 11-point half log dilution series was created in DMSO with a top concentration of 600 μΜ . The test compound plate also contained positive control wel ls containing a known inhibitor to define 100% inhibition and negative control wells containing DMSO to define no inhibition. The compound plates were di luted 1 to 60 resulting in a top final assay compound concentration of 10 μΜ and a 2% DMSO concentration. Test article and assay controls were added to a 384-well plate. Reaction mixtures contained 20 mM HEPES, pH 7.4, 10 mM magnesium chloride, 0.01% bovine serum albumin (BSA), 0.0005% Tween 20, 1 mM ATP and 1 μΜ peptide substrate. The JAKl and TYK2 assays contained 1 μΜ of the IRStide peptide (5 FAM-KKSRGDYMTMQID) and the JAK2 and JAK3 assays contained 1 μΜ of the JAKtide peptide (FITC-KGGEEEEYFELVKK). The assays were initiated by the addition of 20 nM JAKl, 1 nM JAK2 , 1 n M JAK3 or 1 nM TYK2 enzyme and were incubated at room temperature for three hours for JAKl, 60 minutes for JAK2 , 75 minutes for JAK3 or 135 minutes for TYK2. Enzyme concentrations and incubation times were optimized for each new enzyme preps and were modified slightly over time to ensure 20%-30% phosphorylation. The assays were stopped with a final concentration of 10 mM EDTA, 0.1% Coating Reagent and

100 mM HEPES, pH = 7.4. The assay plates were placed on a Caliper Life Science Lab Chip 3000 (LC3000) instru ment, and each well was sampled using appropriate separation conditions to measure the unphosphorylated and phosphorylated peptide. Table 1. Data for JAK Caliper™ Enzyme assay at 1 mM ATP.

HWB INF alpha induced STAT3 phosphorylation Assay

Test articles were prepared as 30 mM stocks in 100% DMSO, and then diluted to 5 mM. A 10-point 2.5 dilution series was created in DMSO with a top concentration of 5 mM. Further dilution was done by adding 4 μ Ι_ of the above test article solutions into 96 μί of PBS with a top concentration of 200 μ Μ .

To a 96-well polypropylene plate (VWR 82007-292) 90 μΙ of HWB was added per well, followed by addition of 5 μΙ test article solutions prepared above to give a top concentration of 10 μΜ. The plate was mixed and incubated for 45 minutes at 37°C. To each well was added 5 μΙ of human IFN alpha (Universal Type I IFN, R&D Systems #11200-2 ; final concentration of 5000U/ml) or D-PBS (unsti mulated control), mixed and incubated 15 minutes at 37°C. The reaction was quenched by adding Lyse/Fix Buffer [BD Phosflow 5x Lyse/Fix Buffer (BD #558049)] to all wells at 1000 μΙ/well and incubated for 20 minutes at 37°C; after washing with FACS buffer [D-PBS (Invitrogen cat# 14190) containing 0.1 % BSA and 0.1% sodium azide], 400 μΙ ice cold 90% methanol/water was added to each well and incubated on ice for 30 minutes. One more wash was done with cold FACS buffer and all samples were finally resuspended in 250 μΙ/well of the desired Alexa Fluor 647 conjugated anti-phospho-STAT3 (pY705) antibody (BD #557815) at 1: 125 dilution in FACS buffer. After overnight incubation at 4 degree all the samples were transferred into a 96-well polypropylene U-bottom plate (Falcon #353077) and checked by flow cytometry machine. IC 50 values obtained for examples 1 to 9, 11-23, 25-38 were in the range of 22 to 2610 nM.

Canine in i//Yro T-cell proliferation assay

T-cell activation plays a key role in a variety of inflammatory and autoimmune disorders as well as asthma, allergies and pruritus. Since T-cell activation can, in part, can be triggered by cytokines that signal through the JAK-STAT pathway, a JAK inhibitor could be effective against such diseases involving aberrant T-cell activation.

Methods: Canine whole blood was collected in sodium heparin tubes from 29 beagle dogs and 23 mixed breed dogs. Whole blood (20 μΙ_) was plated in 96-well plates (Costar 3598) with 180 μί of medium (RPMI 1640, Gibco #21870-076, with 1% heat inactivated fetal bovine serum, Gibco #10082-39, 292 μς/ιτιΙ L-glutamine, Gibco #250030-081, 100 u/ml penicillin and 100μg streptomycin per ml, Gi bco #15 140- 122) containing vehicle control or test compound (0.001 to 10 μΜ), concanavalin A (ConA; 1 μg/ml, Sigma C5275), and canine inter- leukin-2 (IL-2 ; 50 ng/ml , R&D Systems 1815-CL/CF). Wells containing whole blood, medium with vehicle control and no ConA or I L-2 were used as background controls. Plates were incubated at 37° C for 48 hours. Tritiated thymidine, 0.4 μα/well (Perkin Elmer, Net027A-005 MC), was added for 20 additional hours. Plates were frozen and then thawed, washed and filtered using a Brandel MLR-96 cell harvester and pre-wet filter mats (Wallac 1205-401, Perkin Elmer). Filters were d ried at 60° C for one hour (Precision 16EG convection oven) and placed into filter sample bags (Wallac 1205-411, Perkin Elmer) with 10 mL of scintillant (Wallac 1205-440, Perkin Elmer). Sealed filters were counted on a LKB Wallac 1205 Betaplate liquid scintillation counter. Data were collected via Gterm Betaplate program vl. l and transformed into percent inhibition, calculated using the following formula:

Data were graphically displayed as percent inhibition using GraphPad Prism 4.0, and IC curves were fitted using a point to point analysis. Example 38 had an IC 50 of 48.5 nM in this assay. This data suggests that the compounds of the present invention are effective in inhibiting T-cell proliferation, a key feature in diseases resulting from JAK dysregulation.