<<

USOO9365579B2

(12) United States Patent (10) Patent No.: US 9,365,579 B2 Wishart et al. (45) Date of Patent: Jun. 14, 2016

(54) TRICYCLIC COMPOUNDS 4,053,474 A 10, 1977 Treuner et al. 5,212,310 A 5/1993 Thurkaufetal. 5,266,698 A 11/1993 Shaw et al. (71) Applicant: AbbVie Inc., North Chicago, IL (US) 5,521, 173 A 5/1996 Venkatesan et al. 5,605,690 A 2f1997 Jacobs et al. (72) Inventors: Neil Wishart, Jefferson, MA (US); 5,693,801 A 12/1997 Shaw et al. Maria A. Argiriadi, Wayland, MA (US); 5,733,905 A 3/1998 Albright et al. David J. Calderwood, Framingham, 5,736,540 A 4/1998 Albright et al. 5,753,648 A 5/1998 Albright et al. MA (US); Anna M. Ericsson, 5,763,137 A 6/1998 Deprez et al. Shrewsbury, MA (US); Bryan A. 5,840,888 A 11/1998 Shaw et al. Fiamengo, Worcester, MA (US); 5.990,109 A 11/1999 Chen et al. Kristine E. Frank, Grayslake, IL (US); 6,090,382 A 7/2000 Salfeld et al. Michael M. Friedman, Brookline, MA 6,262,241 B1 7/2001 Cook et al. 6,653,471 B2 11/2003 Yohannes et al. (US); Dawn M. George, Charlton, MA 6,949,562 B2 9, 2005 Yohannes et al. (US); Eric R. Goedken, Worcester, MA 7,169,926 B1 1/2007 Burgess et al. (US); Nathan S. Josephsohn, Boston, 7.593,820 B2 9, 2009 Wilks et al. MA (US); Biqin C. Li, Southborough, 7,772,231 B2 8/2010 Sheppard et al. MA (US); Michael J. Morytko, 8,426,411 B2 4/2013 Wishart et al. Framingham, MA (US); Kent D. 8,637,529 B2 1/2014 Woller et al. Stewart, Gurnee, IL (US); Jeffrey W. (Continued) Voss, Holden, MA (US); Grier A. Wallace, Sterling, MA (US); Lu Wang, FOREIGN PATENT DOCUMENTS Northborough, MA (US); Kevin R. CA 2675288 A1 T 2008 Woller, Antioch, IL (US) EA 007415 B1 10, 2006 (73) Assignee: AbbVie Inc., North Chicago, IL (US) (Continued) OTHER PUBLICATIONS (*) Notice: Subject to any disclaimer, the term of this Banker, et al., (1996), Modern Pharmaceuticals, p. 596. patent is extended or adjusted under 35 Dorwal, F. Z., Side Reactions in Organic Synthesis : A Guide to U.S.C. 154(b) by 0 days. Successful Sythesis Design, Weinheim: WILEY-VCH Verlag GmbH & Co. KGaA, 2005, Preface. (21) Appl. No.: 14/610,119 Epps, S.V. et al., “Design and Synthesis of Tricyclic Cores for Kinase Inhibition.” Bioorganic & Medicinal Chemistry Letters, 2013, vol. (22) Filed: Jan. 30, 2015 23, p. 693-698. Hauser, et al., Journal of Organic Chemistry (1961), 26, 451-5. (65) Prior Publication Data Hisham A. Abd El-Nabi. 1-Aryl-2-Chloro-5-Methoxy-1H-3-Pyr rolecarbaldehyde as Synthons for Fused Heterocycles: Synthesis of US 2015/0210708 A1 Jul. 30, 2015 Pyrazolo 3,4-D Pyridine Derivatives, Journal of Chemical Research, May 2004, pp. 325-327, vol. 5. Related U.S. Application Data (Continued) (63) Continuation of application No. 12/481,028, filed on Primary Examiner — Wu-Cheng Winston Shen Jun. 9, 2009, now Pat. No. 8,962,629. Assistant Examiner — Christopher R Stone (60) Provisional application No. 61/201,064, filed on Dec. (74) Attorney, Agent, or Firm — McCarter & English, LLP: 5, 2008, provisional application No. 61/190,159, filed Elizabeth A. Hanley: Yu Lu on Aug. 26, 2008, provisional application No. 61/131,599, filed on Jun. 10, 2008, provisional (57) ABSTRACT application No. 61/131,602, filed on Jun. 10, 2008. The invention provide a compound of Formula (I) (51) Int. Cl. CO7D 487/4 (2006.01) Formula (I) CO7D 498/4 (2006.01) UsX CO7D 51.3/4 (2006.01) (O / (52) U.S. Cl. CPC ...... C07D 487/14 (2013.01); C07D498/14 (2013.01); C07D 513/14 (2013.01) (58) Field of Classification Search CPC ... C07D 487/14: CO7D 498/14: CO7D 513/14 See application file for complete search history. pharmaceutically acceptable salts, pro-drugs, biologically (56) References Cited active metabolites, stereoisomer and isomer thereof wherein the variable are defined herein. The compound of the inven U.S. PATENT DOCUMENTS tion are useful for treating immunological and oncological conditions. 3,663,559 A 5/1972 Derijckere et al. 3,929,992 A 12/1975 Sehgal et al. 15 Claims, No Drawings US 9,365,579 B2 Page 2

(56) References Cited WO 2008/063287 A2 5, 2008 WO 2008/084861 A1 T 2008 U.S. PATENT DOCUMENTS WO 2008/094602 A2 8, 2008 WO 2008/112695 A2 9, 2008 8,785,639 B2 7/2014 Wishartet al. WO 2008121748 A2 10, 2008 2003/0078277 A1 4, 2003 Hibi et al. WO 2009/005.675 A1 1/2009 2004/0023992 A1 2, 2004 Das et al. WO 2009/108827 A1 9, 2009 2005/0176796 A1 8, 2005 D’Alessio et al. WO 2009,152133 A1 12/2009 2006, O183758 A1 8, 2006 Beard et al. WO 2010003133 A2 1/2010 2008/0070914 A1 3/2008 Freyne et al. WO 2011068899 A1 6, 2011 2009/0215724 A1 8, 2009 DuBois et al. 2009/0215750 A1 8/2009 Bamberg et al. OTHER PUBLICATIONS 38885 A. 858 Esty al. Jain, Sanjay et al., A Novel Synthesis of Di(I-Methylazacycloalkeno) 2009.0312338 A1 12, 2009 Wishart et al. 2,3-b:2',3'-dPyridines Through Annulation on Lactam 2011, 0021425 A1 1/2011 Billedeau Acetals;Tetrahedron Letters, 1990 , pp. 13 1-134, vol. 31 No. 1. 2011/O190489 A1 8, 2011 Wishart et al. Jordan, V. C. “Tamoxifen: A most Unlikely Pionering Medicine.” 2012/0034250 A1 2/2012 Shirakami et al. Nature Reviews: Drug Discovery, 2003, 2: 205-213. 2012/0330012 A1 12/2012 Frank et al. Kempson, J. et al., “Synthesis, initial SAR and biological evaluation 2013, OO72470 A1 3/2013 Wishart et al. of 1.6-dihydroimidazo[4, 5-dipyrrolo[2,3-bipyridin-4-amine 2013/0216497 A1 8, 2013 Wishart et all derived inhibtors of IkB kinase”, Bioorganic & Medicinal Chemistry Letters, 2009, vol. 19, pp. 2646-2649. Metabolite. Merriam-Webstercom. Merriam-Webster, n.d. Web. FOREIGN PATENT DOCUMENTS Dec. 4, 2013.

or N(R)C(O)N(R) R ; D is an optionally Substituted (C-Cs)alkylene, optionally Substituted bridged (C-C)cycloalkylene, optionally substi tuted (C-C)cycloalkylene, optionally Substituted (C-Co) arylene, optionally substituted (C-C)heteroarylene, optionally Substituted bridged (C-Co)heterocyclylene or optionally substituted (C-C)heterocyclylene; 10 E is a bond, —R , —C(O)—R , —C(O)C(O)—R ,

when R is H, CH, or—C(O)CH then R is not H, C(O) OCHCH, -C(O)NH-optionally substituted phenyl-NHC 15 (O)-optionally substituted phenyl or —S(O)-phenyl. In a second embodiment the invention provides a com pound of Formula (II)

Formula (II) N- Ra; I Ré 25 where in all cases, E is linked to either a carbon or a nitrogen atom in D; G is hydrogen, deuterium, N(R)(R), halogen, OR", - SR", S(O)R', S(O).R, NO, C(O)OR, CN, pharmaceutically acceptable salts, pro-drugs, biologically 30 C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, active metabolites, stereoisomers and isomers thereof N(R)C(O)N(R), C(O-R)(R), C(O)R, CF, wherein OCF, N(R)S(O).R., S(O)N(R)(R), S(O)N when T is NR, U is N, X is CR and there is a double bond (R)C(O)R’, an optionally substituted (C-C)alkyl, option between U and X: ally substituted (C-C)alkenyl, optionally substituted (C- when T is O, U is N, X is CR and there is a double bond 35 C.)alkynyl, optionally substituted (C-C)cycloalkyl, between U and X: optionally substituted (C-C)heteroaryl, optionally substi when T is CR, U is N, X is NR and there is a double bond tuted (C-Co)heterocyclyl, optionally Substituted (C-Co) between T and U; aryl, optionally substituted —(C-C)alkylene-(C-C)cy when T is CR, U is CR, X is NR and there is a double cloalkyl, optionally Substituted —(C-C)alkyl-(Co-Co) bond between T and U; 40 aryl, optionally substituted —(C-C)alkylene-(C-C) R", R and R are independently hydrogen, deuterium, heteroaryl or optionally Substituted —(C-C)alkylene-(C- -N(R)(R), halogen, OR, - SR", S(O)R’, -S(O), Co)heterocyclyl: R, NO, C(O)OR, CN, C(O)N(R)(R), N(R) wherein in a moiety comprising N(R)(R'), the nitrogen, C(O)(R), —C(O)R", N(R)S(O) -, -S(O)N(R)- R" and R may form a ring such that N(R)(R') rep —CF, —OCF, optionally substituted (C-C)alkyl, option 45 resents an optionally substituted (C-Co)heterocyclyl ally Substituted (C-C)alkenyl, optionally substituted (C- linked through a nitrogen; C.)alkynyl, optionally substituted (C-C)cycloalkyl, R is a hydrogen, deuterium, an optionally substituted optionally substituted (C-Co)heteroaryl, optionally Substi bridged (C-C)cycloalkyl group, optionally substituted tuted (C-C) heterocyclyl, or optionally substituted (C- bridged (C-C)heterocyclyl group, optionally substituted Co)aryl; 50 adamantyl, optionally Substituted (C-Cs)alkyl, optionally wherein in a moiety comprising N(R)(R), the nitrogen, Substituted (C-C)cycloalkyl, optionally Substituted (C- R" and R may form a ring such that N(R)(R) represents Cs)cycloalkenyl, optionally Substituted (C-C)aryl, option an optionally substituted (C-Co)heterocyclyl linked ally substituted (C-C)heteroaryl or optionally substituted through a nitrogen; (C-C)heterocyclyl: or Ris-J-L-M-Q, wherein: R is an optionally substituted bridged (Cs-C)cycloalkyl 55 J is a bond, —C(O)—, optionally Substituted (C-C)alky group, optionally substituted bridged (C-C)heterocyclyl lene, optionally substituted (C-C)alkenylene, optionally group, optionally Substituted adamantyl, optionally Substi Substituted (C-C)alkynylene, optionally Substituted (C- tuted (C-Cs)alkyl, optionally substituted (C-C)cy C)cycloalkylene, optionally Substituted (C-C)heterocy cloalkyl, optionally Substituted (C-C)cycloalkenyl, option clylene, -C(O)N(R)-R , —C(O-R)(R)-R , or ally Substituted (C-Co.)aryl, optionally Substituted (C-Co) 60 S(O)N(R)R ; heteroaryl, optionally substituted (C-C)heterocyclyl or L is a bond, an optionally substituted (C-C)alkylene, -A-D-E-G optionally Substituted bridged (C-C)cycloalkylene, wherein: optionally substituted (C-C)cycloalkylene, optionally A is a bond, —C(O)—, optionally substituted (C-C) Substituted (C-Co.)arylene, optionally Substituted (C-Co) alkylene, optionally Substituted (C-C)alkenylene, option 65 heteroarylene, optionally Substituted bridged (C-Co)het ally Substituted (C-C)alkynylene, optionally Substituted erocyclylene or an optionally Substituted (C-C)heterocy (C-C)cycloalkylene, optionally Substituted (C-C)het clylene; or US 9,365,579 B2 7 8 L is W is

O O O O 5

N-R N-R t l 10 Y is a bond, —R , —C(O)—R , —C(O)C(O)—R , M is a bond, -R-, -C(O)—R , —C(O)C(O)— C(O)O Re C(O)C(O)N(R) Re O Re . R C(O)O R C(O)C(O)N(R) R. , S(O), RS(Q) R' SR N(R) O Re , S(O), R , S(O) R' , S R , BNCR C(O)R C(O)N(RROC N(Ra) Re N(Ra)C(O) Re C(O)N(Ra) R(9)N(RROCO) - N(R')S(O), R orR' - S(O)N(R)-RNCROC(O)N(R') ; Z is hydrogen, deuterium, N(R)(R), halogen, OR", - SR", S(O)R', S(O).R, NO, C(O)OR, CN, C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, N(R)C(O)N(R), C(O R)(R), C(O)R, CF, Q is hydrogen, deuterium, N(R)(R), halogen, OR", OCF, N(R)S(O).R., S(O)N(R)(R), S(O)N - SR", S(O)R', S(O).R, NO, C(O)OR, CN, (R)C(O)R’, an optionally substituted (C-C)alkyl, an C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, optionally substituted (C-C)alkenyl, an optionally substi N(R)C(O)N(R), C(O-R)(R), C(O)R, CF, tuted (C-C)alkynyl, an optionally Substituted (C-C)cy 25 cloalkyl, an optionally substituted (C-C)heteroaryl, an OCF, N(R)S(O).R., S(O)N(R)(R), S(O)N optionally Substituted (C-Co)heterocyclyl, an optionally (R)C(O)R’, optionally substituted (C-C)alkyl, optionally Substituted (C-C)aryl, an optionally substituted—(C-C) Substituted (C-C)alkenyl, optionally Substituted (C-C) alkylene-(C-C)cycloalkyl, an optionally Substituted alkynyl, optionally substituted (C-C)cycloalkyl, option —(C-C)alkyl-(C-C)aryl, an optionally substituted ally substituted (C-C)heteroaryl, optionally substituted 30 —(C-C)alkylene-(C-Co)heteroaryl or an optionally Sub stituted —(C-C)alkylene-(C-C)heterocyclyl: (C-Co)heterocyclyl, optionally Substituted (C-Co)aryl, whereinina moiety comprising N(R)(R), the nitrogen, optionally substituted —(C-C)alkylene-(C-Co)cy R" and R may form a ring such that N(R)(R) represents cloalkyl, optionally Substituted —(C-C)alkylene-(Co-Co) an optionally substituted (C-Co)heterocyclyl linked aryl, optionally substituted —(C-C)alkyl-(C-C)het through a nitrogen; eroaryl or optionally substituted—(C-C)alkylene-(C-C) 35 R" and Rare independently hydrogen, deuterium, option ally substituted (C-C)alkyl, optionally substituted (C- heterocyclyl: Co)alkenyl, optionally Substituted (C-C)alkynyl, option whereininamoiety comprising N(R)(R), the nitrogen, ally substituted —(C-Co.)alkylene-O-(C-Co.)alkyl, R" and R may form a ring such that N(R)(R') rep optionally substituted (C-C)cycloalkyl, optionally Substi resents an optionally substituted (C-Co)heterocyclyl 40 tuted (C-Co)aryl, optionally Substituted (C-Co)het linked through a nitrogen; eroaryl, optionally substituted (C-C)heterocyclyl, option ally Substituted —(C-C)alkylene-(C-C)cycloalkyl, R is hydrogen, deuterium, optionally substituted bridged optionally substituted —(C-C)alkylene-(C-C)aryl, (C-C) cycloalkyl group, optionally substituted bridged optionally substituted —(C-C)alkylene-(C-Co)het (C-Co)heterocyclyl group, optionally substituted adaman 45 eroaryl or optionally Substituted —(C-C)alkylene-(C-Co) tyl, optionally Substituted (C-Cs)alkyl, optionally Substi heterocyclyl; and tuted (C-C)cycloalkyl, optionally substituted (C-C)cy R is a bond or is independently selected from optionally Substituted (C-C)alkylene, optionally substituted (C-C) cloalkenyl, optionally substituted (C-C)aryl, optionally alkenylene, optionally Substituted (C-Co.)alkynylene, Substituted (C-Co)heteroaryl or optionally substituted (C- 50 optionally substituted —(C-C)alkylene-O-(C-C) Co)heterocyclyl; or alkylene-group, optionally Substituted (C-C)cycloalky Ris-V-W-Y-Z wherein: lene, optionally Substituted (C-Co.)arylene, optionally Sub stituted (C-Co)heteroarylene, or optionally Substituted (C- V is a bond, —C(O)—, optionally substituted (C-C) Co)heterocyclylene; alkyl, optionally substituted (C-C)alkenyl, optionally Sub 55 provided that when the compound is stituted (C-C)alkynyl, optionally Substituted (C-C)cy cloalkyl, optionally substituted (C-C)heterocyclyl. —C(O) N(R)-R , —C(O-R)(R)-R, , or -S(O)N(R) R3 R ; 60 - W is a bond, an optionally substituted (C-C)alkyl, R6 N optionally Substituted bridged (C-C)cycloalkyl, optionally N Substituted (C-C)cycloalkyl, optionally substituted (C- 2 N N Co)aryl, optionally substituted (C-Co)heteroaryl, option 65 ally Substituted bridged (C-C)heterocyclyl or an option H ally Substituted (C-Co)heterocyclyl; or US 9,365,579 B2 10 R is not linked to the pyrazole ring by a nitrogen or oxygen tuted (C-C)cycloalkylene, optionally Substituted (C-Co) atom; and provided the compound is not arylene, optionally substituted (C-C)heteroarylene, optionally substituted bridged (C-C)heterocyclylene oran

optionally substituted (C-Co)heterocyclylene;

10

or E is

wherein when R is H, CH or C(O)OH and R is not H, 15 —C(O)CCHCH. —C(O)NH optionally substituted phe nyl-NHC(O)-optionally substituted phenyl or —S(O)-phe nyl. N- Ra; In a third embodiment the invention provides a compound I of formula (Ig) R

Formula (Ig) where in all cases, E is linked to either a carbon or a nitrogen atom in D; 25 G is hydrogen, deuterium, N(R)(R'), halogen, OR", - SR", S(O)R', S(O),R, NO,-C(O)OR, CN, C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, N(R)C(O)N(R), C(O R)(R), C(O)R, CF, OCF, N(R)S(O).R., S(O)N(R)(R), S(O)N 30 (R)C(O)R’, an optionally substituted (C-C)alkyl, an optionally substituted (C-C)alkenyl, an optionally substi tuted (C-C)alkynyl, an optionally substituted (C-Co)cy pharmaceutically acceptable salts, pro-drugs, biologically cloalkyl, an optionally Substituted (C-Co)heteroaryl, an active metabolites, stereoisomers and isomers thereof optionally Substituted (C-C) heterocyclyl, an optionally wherein 35 Substituted (C-C)aryl, an optionally Substituted —(C-C) R", R and Rare each independently hydrogen, deute alkylene-(C-C)cycloalkyl, an optionally substituted rium, N(R)(R), halogen, —OR", -SR", -S(O)R", —(C-C)alkylene-(Co-Co.)aryl, an optionally substituted S(O),R, NO, C(O)OR, CN, C(O)N(R)(R), —(C-C)alkylene-(C-C)heteroaryl or an optionally sub N(R)C(O)(R), C(O)R, N(R)S(O) , S(O)N stituted —(C-C)alkylene-(C-C)heterocyclyl; (R)—, —CF, —OCF, optionally substituted —(C-C) 40 alkyl, optionally substituted (C-C)alkenyl, optionally Sub wherein in a moiety containing N(R)(R), the nitrogen, stituted —(C-C)alkynyl, optionally substituted —(C-C) R" and R may form a ring such that N(R)(R') rep cycloalkyl, optionally Substituted —(C-Co)heteroaryl, resents an optionally substituted (C-Co)heterocyclyl optionally Substituted —(C-C) heterocyclyl, or optionally linked through a nitrogen; Substituted —(C-C)aryl; 45 R" is a hydrogen, deuterium, an optionally substituted wherein in a moiety containing N(R)(R'), the nitrogen, bridged (C-C) cycloalkyl group, optionally substituted R" and R may form a ring such that N(R)(R') rep bridged (C-Co)heterocyclyl group, optionally substituted resents an optionally substituted (C-Co)heterocyclyl adamantyl, optionally Substituted (C-C) alkyl, optionally linked through a nitrogen; Substituted (C-C)cycloalkyl, optionally Substituted (C- R is an optionally substituted bridged (Cs-C) cycloalkyl 50 Cs)cycloalkenyl, optionally Substituted (C-C)aryl, option group, optionally Substituted bridged (C-C) heterocyclyl ally substituted (C-C)heteroaryl or optionally substituted group, optionally Substituted adamantyl, optionally Substi (C-Co)heterocyclyl; or tuted (C-Cs) alkyl, optionally Substituted (C-C)cy Ris-J-L-M-Q, wherein: cloalkyl, optionally substituted (C-C)cycloalkenyl, option J is a bond, —C(O)—, optionally Substituted (C-C)alky ally Substituted (C-Co.)aryl, optionally Substituted (C-Co) 55 lene, optionally substituted (C-C)alkenylene, optionally heteroaryl or optionally substituted (C-C)heterocyclyl; or Substituted (C-C)alkynylene, optionally Substituted (C- Ris-A-D-E-G, wherein: C)cycloalkylene, optionally Substituted (C-C)heterocy A is a bond, —C(O)—, optionally substituted (C-C) clylene, -C(O)N(R)-R , —C(O-R)(R)-R , or alkylene, optionally Substituted (C-C)alkenylene, option S(O)N(R)R ; ally Substituted (C-C)alkynylene, optionally Substituted 60 (C-C)cycloalkylene, optionally substituted (C-C)het L is a bond or an optionally substituted (C-C) alkylene, optionally substituted bridged (C-C)cycloalkylene, optionally substituted (C-C)cy cloalkylene, optionally Substituted (C-C)arylene, option or N(R)C(O)N(R) R. ; 65 ally substituted (C-C)heteroarylene, optionally Substi D is an optionally Substituted (C-Cs)alkylene, optionally tuted bridged (C-Co)heterocyclylene or an optionally Substituted bridged (C-C)cycloalkylene, optionally Substi Substituted (C-Co)heterocyclylene; US 9,365,579 B2 12

3 Formula (III) R /Ys -( R2 *N N

N Y-R R 5 N NV 10 H

pharmaceutically acceptable salts, pro-drugs, biologically active metabolites, stereoisomers and isomers thereof 15 wherein R OC(O)N(R) R OC(O) R. , N(R)C X is CR or N;Y is CR or N: (O)N(R) R - N(R)S(O), R or S(O)N(R)- R", R and R are each independently hydrogen, deute R ; rium, N(R)(R), halogen, -OR. - SR", S(O)R", S(O),R, NO, C(O)OR, CN, C(O)N(R)(R), Q is hydrogen, deuterium, N(R)(R), halogen, OR", N(R)C(O)(R), C(O)R", C(OH)RR, N(R)S - SR", S(O)R', S(O),R, NO,-C(O)OR, CN, (O) R' , S(O)N(R)(R), CFs, OCF, optionally C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, Substituted —(C-C)alkyl, optionally Substituted (C-C) N(R)C(O)N(R), C(O R)(R), C(O)R, CF, alkenyl, optionally substituted (C-C)alkynyl, optionally –OCF, N(R)S(O),R), S(O)N(R)(R), S(O)N Substituted (C-C)cycloalkyl, optionally Substituted (C- (R)C(O)R’, an optionally substituted (C-C)alkyl, an 25 optionally Substituted (C-C)alkenyl, an optionally Substi Co)heteroaryl, optionally Substituted (C-Co) heterocyclyl, tuted (C-C)alkynyl, an optionally substituted (C-C)cy or optionally substituted (C-C)aryl; cloalkyl, an optionally Substituted (C-Co)heteroaryl, an wherein in a moiety containing N(R)(R), the nitrogen, optionally substituted (C-Co)heterocyclyl, an optionally R" and R may form a ring such that N(R)(R') rep Substituted (C-C)aryl, an optionally substituted—(C-C) 30 resents an optionally substituted (C-Co)heterocyclyl alkylene-(C-C)cycloalkyl, an optionally substituted or optionally substituted (C-C)heteroaryl linked —(C-C)alkylene-(Co-Co)aryl, an optionally substituted through a nitrogen; —(C-C)alkylene-(C-C)heteroaryl or an optionally sub R is an optionally substituted bridged (C-C)cycloalkyl stituted —(C-C)alkylene-(C-C)heterocyclyl; 35 group, optionally Substituted bridged (C-C)heterocyclyl wherein in a moiety containing N(R)(R'), the nitrogen, group, optionally Substituted (C-Cs)alkyl, optionally Substi R" and R may form a ring such that N(R)(R') rep tuted (C-C)cycloalkyl, optionally substituted (C-C)cy resents an optionally substituted (C-C)heterocyclyl cloalkenyl, optionally Substituted (C-C)aryl, optionally linked through a nitrogen; Substituted (C-C)heteroaryl or optionally substituted (C- 40 Co)heterocyclyl; or R is -A-D-E-G, wherein: R" and R are independently hydrogen, deuterium, an A is a bond, —C(O)—, optionally Substituted (C-C) optionally Substituted (C-Co.)alkyl, an optionally Substi alkylene, optionally Substituted (C-C)alkenylene, option tuted (C-C)alkenyl, an optionally substituted (C-C) ally Substituted (C-C)alkynylene, optionally Substituted alkynyl, an optionally substituted —(C-Co.)alkylene-O- (C-C)cycloalkylene, optionally Substituted (C-C)het (C-Co.)alkyl, an optionally Substituted (C-Co)cycloalkyl, 45 an optionally substituted (C-C)aryl, an optionally substi tuted (C-C)heteroaryl, an optionally Substituted (C-Co) heterocyclyl, an optionally substituted —(C-C)alkylene (C-C)cycloalkyl, an optionally substituted —(C-C) D is an optionally substituted (C-Cs) alkylene, optionally alkylene-(C-C)aryl, an optionally Substituted —(C-C) 50 Substituted bridged (C-C)cycloalkylene, optionally substi alkylene-(C-Co)heteroaryl or an optionally Substituted tuted (C-C)cycloalkylene, optionally Substituted bridged —(C-C)alkylene-(C-C)heterocyclyl; and (Cs-Co)cycloalkenylene, optionally Substituted (C-C)cy R is a bond, an optionally substituted (C-C)alkylene, an cloalkenylene, optionally substituted (C-C)arylene, optionally substituted (C-Co.)alkenylene, an optionally Sub optionally Substituted (C-C)heteroarylene, optionally Sub stituted (C-Co.)alkynylene, an optionally Substituted—(C- 55 stituted bridged (C-C)heterocyclylene or an optionally Co)alkylene-O-(C-C)alkylene-group, an optionally Substituted (C-Co)heterocyclylene; Substituted (C-C)cycloalkylene, an optionally Substituted (Co-Co.)arylene, an optionally Substituted (C-C)het eroarylene, or an optionally substituted (C-C)heterocy clylene. 60 In a fourth embodiment the invention provides a compound of Formula (III)

65 (R')S(O), R , or - R S(O)N(R) R ; or E is US 9,365,579 B2 13 14 M is

N- Ra; I Ré N- Ra; I Ré where in all cases, E is linked to either a carbon or a 10 nitrogen atom in D; where in all cases, Mis linked to either a carbon or a nitrogen G is hydrogen, deuterium, N(R)(R), halogen, OR", atom in L. - SR", S(O)R', S(O),R, NO,-C(O)OR, CN, Q is hydrogen, deuterium, N(R)(R'), halogen, OR", C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, 15 - SR", S(O)R', S(O).R, NO, C(O)OR, CN, N(R)C(O)N(R), C(O R)(R), C(O)R, CF, C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, –OCF, N(R)S(O).R., S(O)N(R)(R), S(O)N N(R)C(O)N(R), C(O R)(R), C(O)R, CF, (R)C(O)R’, an optionally substituted (C-C)alkyl, an OCF, N(R)S(O).R., S(O)N(R)(R), S(O)N optionally Substituted (C-C)alkenyl, an optionally Substi (R)C(O)R’, an optionally substituted (C-C)alkyl, an tuted (C-C)alkynyl, an optionally substituted (C-C)cy optionally Substituted (C-C)alkenyl, an optionally Substi cloalkyl, an optionally Substituted (C-Co)heteroaryl, an tuted (C-C)alkynyl, an optionally substituted (C-C)cy optionally Substituted (C-C) heterocyclyl, an optionally cloalkyl, an optionally Substituted —(C-Co)heteroaryl, an Substituted (C-C)aryl, an optionally substituted—(C-C) optionally Substituted —(C-Co) heterocyclyl, an optionally alkylene-(C-C)cycloalkyl, an optionally Substituted Substituted (C-C)aryl, an optionally substituted—(C-C) —(C-C)alkylene-(Co-Co)aryl, an optionally substituted 25 alkylene-(C-C)cycloalkyl, an optionally Substituted —(C-C)alkylene-(C-C)heteroaryl, or an optionally sub —(C-C)alkylene-(Co-Co.)aryl, an optionally substituted stituted —(C-C)alkylene-(C-C)heterocyclyl; —(C-C)alkylene-(C-C)heteroaryl, or an optionally sub wherein in a moiety containing N(R)(R), the nitrogen, stituted —(C-C)alkylene-(C-C)heterocyclyl; R" and R may form a ring such that N(R)(R') rep wherein in a moiety containing N(R)(R), the nitrogen, resents an optionally substituted (C-C)heterocyclyl 30 R" and R may form a ring such that N(R)(R') rep or an optionally substituted (C-Co) heteroaryl linked resents an optionally substituted (C-Co)heterocyclyl through a nitrogen; or an optionally Substituted (C-C) heteroaryl linked R is a hydrogen, deuterium, an optionally substituted through a nitrogen; bridged (C-C)cycloalkyl group, optionally substituted 35 R" is a hydrogen, deuterium, an optionally substituted bridged (C-Co)heterocyclyl group, optionally substituted bridged (C-C)cycloalkyl group, optionally substituted (C-Cs)alkyl, optionally Substituted (C-Co)cycloalkyl, bridged (C-C)heterocyclyl group, optionally substituted optionally substituted (C-C)cycloalkenyl, optionally sub (C-Cs)alkyl, optionally Substituted (C-Co)cycloalkyl, stituted (C-Co)aryl, optionally Substituted (C-C)het optionally Substituted (C-C)cycloalkenyl, optionally Sub eroaryl or optionally substituted (C-C)heterocyclyl: or R 40 stituted (C-C)aryl, optionally substituted (C-C)het is -J-L-M-Q, wherein: eroarylor optionally substituted (C-Co)heterocyclyl; or R' J is a bond, —C(O)—, optionally Substituted (C-C)alky is -U-V-W-Z wherein: lene, optionally Substituted (C-C)alkenylene, optionally U is a bond, —C(O)—, optionally substituted (C-C) Substituted (C-C)alkynylene, optionally Substituted (C- alkylene, optionally Substituted (C-C)alkenylene, option C)cycloalkylene, optionally Substituted (C-C)heterocy 45 ally Substituted (C-C)alkynylene, optionally Substituted (C-C)cycloalkylene, optionally Substituted (C-C)het

50 R - N(R)S(O), R or N(R)C(O)N(R) R. ; L is an optionally Substituted (C-Cs)alkylene, optionally V is an optionally Substituted (C-Cs) alkylene, optionally Substituted bridged (C-C)cycloalkylene, optionally Substi Substituted bridged (C-C)cycloalkylene, optionally substi tuted (C-C)cycloalkylene, optionally substituted bridged tuted (C-C)cycloalkylene, optionally Substituted bridged (Cs-Co)cycloalkenylene, optionally Substituted (C-C)cy (C-C)cycloalkenylene, optionally substituted (C-C)cy cloalkenylene, optionally substituted (C-C)arylene, 55 cloalkenylene, optionally substituted (C-Co)arylene, optionally Substituted (C-C)heteroarylene, optionally Sub optionally substituted (C-C)heteroarylene, optionally sub stituted bridged (C-C)heterocyclylene or an optionally stituted bridged (C-C)heterocyclylene or an optionally Substituted (C-Co)heterocyclylene; Substituted (C-Co)heterocyclylene; M is a bond, - R -, -R C(O)- R - R C(O)C W is a bond, - R - R C(O)- R - R C(O)C (O) R. , R. C(O)O R. , R. C(O)C(O)N 60 (O) R. , R. C(O)O R. , R. C(O)C(O)N (R) R - R N(R) C(O)C(O) R - R O - (R) R - R N(R) C(O)C(O) R - R O R , -R S(O), R -, -R S(O) R , -R R , -R S(O), R -, -R S(O) R , -R S R , R. N(R) R. , R N(R)C(O) R. , S R , R. N(R) R. , R N(R)C(O) R. ,

65 US 9,365,579 B2 15 16 W is In a fifth embodiment the invention provides a compound of Formula (Ia)

3 Formula (Ia) R Ns N- Ra; / -( R2 I N N Ré 10 Sa R5 N N where in all cases, W is linked to either a carbon or a V nitrogen atom in V: H Z is independently hydrogen, deuterium, N(R)(R'), 15 pharmaceutically acceptable salts, pro-drugs, biologically halogen, -OR. - SR", S(O)R", S(O),R, NO, active metabolites, Stereoisomers and isomers thereof C(O)CR, CN, C(O)N(R)(R), N(R)C(O)R, wherein N(R)C(O)OR, N(R)C(O)N(R), C(O R)(R), R", R and R are each independently hydrogen, deute —C(O)R", —CF, OCF, N(R)S(O).R., -S(O)N rium, N(R)(R), halogen, —OR", -SR", -S(O)R", (R)(R), S(O)N(R)C(O)R’, an optionally substituted - S(O),R, NO,-C(O)OR, CN, C(O)N(R)(R), —(C-C)alkyl, an optionally substituted —(C-C)alkenyl, N(R)C(O)(R), C(O)R.", N(R)S(O), , S(O)N an optionally Substituted —(C-C)alkynyl, an optionally (R')— —CF, —OCF, optionally Substituted —(C-C) Substituted —(C-C)cycloalkyl, an optionally Substituted 25 alkyl, optionally Substituted —(C-C)alkenyl, optionally —(C-C)heteroaryl, an optionally substituted —(C-C) Substituted —(C-C)alkynyl, optionally Substituted —(C- heterocyclyl, an optionally substituted (C-C)aryl, an Co)cycloalkyl, optionally Substituted —(C-Co)heteroaryl, optionally substituted —(C-C)alkylene-(C-C)cy optionally substituted —(C-C) heterocyclyl, or optionally cloalkyl, an optionally substituted —(C-C)alkylene-(C- 30 Substituted —(C-C)aryl; Co)aryl, an optionally substituted —(C-C)alkylene-(C- wherein in a moiety containing N(R)(R), the nitrogen, Co)heteroaryl, or an optionally substituted —(C-C) R" and R may form a ring such that N(R)(R') rep alkylene-(C-C)heterocyclyl: resents an optionally substituted (C-Co)heterocyclyl linked through a nitrogen; wherein in a moiety containing N(R)(R'), the nitrogen, 35 R" and R may form a ring such that N(R)(R') rep R is an optionally substituted bridged (Cs-C)cycloalkyl resents an optionally substituted (C-Co)heterocyclyl group, optionally Substituted bridged (C-C)heterocyclyl group, optionally Substituted adamantyl, optionally Substi or an optionally Substituted (C-C) heteroaryl linked tuted (C-Cs)alkyl, optionally substituted (C-C)cy through a nitrogen; 40 cloalkyl, optionally substituted (C-C)cycloalkenyl, option R" and Rare each independently hydrogen, deuterium, an ally substituted (C-C)aryl, optionally substituted (C-C) optionally Substituted —(C-C)alkyl, an optionally Substi heteroaryl or optionally Substituted (C-C)heterocyclyl; or tuted —(C-Co.)alkenyl, an optionally substituted —(C- Ris-A-D-E-G, wherein: Co)alkynyl, an optionally substituted —(C-Co.)alkylene 45 A is a bond, —C(O)—, optionally Substituted (C-C) O—(C-Co.)alkyl, an optionally Substituted —(C-Co) alkylene, optionally Substituted (C-C)alkenylene, option cycloalkyl, an optionally substituted —(C-Co)aryl, an ally substituted (C-C)alkynylene, optionally substituted optionally Substituted —(C-C)heteroaryl, an optionally (C-C)cycloalkylene, optionally Substituted (C-C)het 50 erocyclylene, -C(O)N(R)-R -, - N(R)C(O) R , Substituted—(C-C)heterocyclyl, an optionally Substituted O R , – N(R) R , – S R -, -C(O-R) —(C-C)alkylene-(C-Co)cycloalkyl, an optionally Substi (R)-R , S(O)N(R)-R -, -N(R)S(O), R - tuted —(C-C)alkylene-(C-C)aryl, an optionally Substi tuted —(C-C)alkylene-(C-C)heteroaryl, or an option D is an optionally substituted (C-C)alkylene, optionally ally Substituted —(C-C)alkylene-(C-C)heterocyclyl; 55 Substituted bridged (C-C)cycloalkylene, optionally substi and tuted (C-C)cycloalkylene, optionally Substituted (C-Co) arylene, optionally substituted (C-C)heteroarylene, R is each independently a bond, an optionally substituted optionally substituted bridged (C-C)heterocyclylene oran (C-Co.)alkylene, an optionally Substituted (C-C)alk 60 optionally substituted (C-Co)heterocyclylene; enylene, an optionally substituted (C-C)alkynylene, an optionally Substituted (C-C)alkylene-O-(C-C)alky lene group, an optionally Substituted (C-Co)cycloalkylene, an optionally Substituted (C-Co)arylene, an optionally Sub 65 stituted (C-Co)heteroarylene, or an optionally Substituted (C-Co)heterocyclylene. US 9,365,579 B2 17 18 E is In a seventh embodiment the invention provides a com pound according to the first embodiment wherein T is N, U is N, X is CR. Y is N and forms a compound of Formula (Ia)

Formula (Ia) R3 N I Ns Re -( R2 10 N-N where in all cases, E is linked to either a carbon or a DCO)-S. N nitrogen atom in D; R5 N V G is independently hydrogen, deuterium, N(R)(R), H halogen, -OR. - SR", S(O)R", S(O),R, NO, 15 C(O)CR, CN, C(O)N(R)(R), N(R)C(O)R, In an eighth embodiment the invention provides a com N(R)C(O)OR, N(R)C(O)N(R), C(O R)(R), pound according to the first embodiment wherein T is CR, U —C(O)R", —CF, OCF, N(R)S(O).R., -S(O)N is N, X is CR and Y is N and forms a compound of Formula (R)(R), S(O)N(R)C(O)R, an optionally substituted (Ib) (C-C)alkyl, an optionally substituted (C-C)alkenyl, an optionally Substituted (C-C)alkynyl, an optionally Substi Formula (Ib) tuted (C-C)cycloalkyl, an optionally substituted (C-C) R3 heteroaryl, an optionally substituted (C-C) heterocyclyl, 25 an optionally Substituted (C-C)aryl, an optionally Substi 6 N-( R2 tuted —(C-C)alkylene-(C-C)cycloalkyl, an optionally R" SN Substituted —(C-C)alkylene-(C-C)aryl, an optionally Y-R Substituted —(C-C)alkylene-(C-Co)heteroaryl, or an N optionally substituted —(C-C)alkylene-(C-C)heterocy R5 N NV clyl; 30 H wherein in a moiety containing N(R)(R'), the nitrogen, R" and R may form a ring such that N(R)(R') rep In a ninth embodiment the invention provides a compound resents an optionally substituted (C-Co)heterocyclyl according to the first embodiment wherein T is N, U is CR, linked through a nitrogen; 35 X is CR, and Y is N and forms a compound of Formula (Ic) R" and R are independently hydrogen, deuterium, an optionally Substituted (C-Co.)alkyl, an optionally Substi Formula (Ic) tuted (C-Co.)alkenyl, an optionally Substituted (C-Co) R4 R3 alkynyl, an optionally substituted —(C-C)alkylene-O- (C-Co.)alkyl, an optionally Substituted (C-Co)cycloalkyl, 40 an optionally Substituted (C-C)aryl, an optionally Substi N)-( N , tuted (C-C)heteroaryl, an optionally Substituted (C-Co) N RI heterocyclyl, an optionally substituted —(C-C)alkyl-(C- 5 Sa N Co)cycloalkyl, an optionally Substituted—(C-C)alkylene 45 R N V (Co-Co.)aryl, an optionally substituted —(C-C)alkylene H (C-Co)heteroaryl, or an optionally Substituted —(C-C) alkylene-(C-C)heterocyclyl; and In a tenth embodiment the invention provides a compound R is each independently a bond, optionally substituted according to the first embodiment wherein T is CR, U is CR, (C-Co.)alkylene, an optionally Substituted (C-C)alk 50 X is CR and Y is N and forms a compound of Formula (Id) enylene, an optionally substituted (C-C)alkynylene, an optionally substituted —(C-C)alkylene-O-(C-C) alkyl-group, an optionally Substituted (C-C)cycloalky Formula (Id) R4 R3 lene, an optionally Substituted (C-C)arylene, an optionally 55 Substituted (C-C)heteroarylene, or an optionally substi 's- R2 6 tuted (C-C)heterocyclylene. R N-N In a sixth embodiment the invention provides a compound Y-R according to the first embodiment wherein R, R and Rare N each independently hydrogen, deuterium, halogen, —OR". 60 R5 N N CN, C(O)N(R)(R), N(R)C(O)(R), CF, V —OCF, an optionally Substituted —(C-C)alkyl, option ally Substituted —(C-C)alkynyl, optionally Substituted In an eleventh embodiment the invention provides a com —(C-C)cycloalkyl, optionally substituted —(C-C)het 65 pound according to the first embodiment wherein T is CR, U eroaryl, —(C-Co)heterocyclyl or optionally Substituted is N, X is NR and Y is C and forms a compound of Formula —(C-Co)aryl. (Ie) US 9,365,579 B2 19 20

Formula (Ie) Formula (II)

10

In a sixteenth embodiment the invention provides com In a twelfth embodiment the invention provides a com pound according to the first embodiment wherein R is hydro pound according to the first embodiment wherein T is O, U is gen, an optionally Substituted bridged (C-C)cycloalkyl N, X is CR and Y is C and forms a compound of Formula (If) 15 group, optionally substituted bridged (C-C)heterocyclyl group, optionally Substituted (C-C)alkyl, optionally Substi tuted (C-C)cycloalkyl, optionally Substituted (C-Co) aryl, optionally substituted (C-C)heteroaryl or optionally Formula (If) Substituted (C-Co)heterocyclyl. In a seventeenth embodiment the invention provides a compound according to any of the foregoing embodiments wherein R is hydrogen, optionally substituted cyclopropyl. optionally substituted cyclobutyl, optionally substituted 25 cyclopentyl, optionally substituted cyclohexyl, optionally Substituted phenyl, optionally Substituted adamantanyl. optionally substituted azetidinyl, optionally substituted bicy clo2.1.1 hexyl, optionally substituted bicyclo[2.2.1]heptyl, optionally substituted bicyclo[2.2.2]octyl, optionally substi In a thirteenth embodiment the invention provides a com 30 tuted bicyclo[3.2.1]octyl, optionally substituted bicyclo pound according to the first embodiment wherein T is NR, U 4.3.1 decyl, optionally substituted bicyclo[3.3.1nonyl, is N, X is CR, and Y is C and forms a compound of Formula optionally substituted bornyl, optionally substituted borne (Ig) nyl, optionally substituted norbornyl, optionally substituted norbornenyl, optionally substituted bicyclo[3.1.1 heptyl, 35 optionally substituted tricyclobutyl, optionally substituted azanorbornyl, optionally Substituted quinuclidinyl, option Formula (Ig) ally substituted isoquinuclidinyl, optionally substituted tro panyl, optionally Substituted azabicyclo3.2.1]octanyl. optionally Substituted azabicyclo2.2.1]heptanyl, optionally 40 substituted 2-azabicyclo[3.2.1]octanyl, optionally substi tuted azabicyclo3.2.1]octanyl, optionally substituted azabi cyclo3.2.2 nonanyl, optionally Substituted azabicyclo 3.3.0nonanyl, optionally substituted azabicyclo[3.3.1 nonanyl, optionally Substituted bicyclo2.2.1]hept-2-enyl, 45 optionally Substituted piperidinyl, optionally Substituted pyr rolidinyl or optionally substituted tetrahydrofuranyl. In a fourteenth embodiment the invention provides a com In an eighteenth embodiment the invention provides a pound according to the first embodiment wherein T is CR, U compound according to any of the foregoing embodiments is CR, X is NR, and Y is C and forms a compound of wherein R is optionally substituted cyclopropyl, optionally 50 substituted cyclobutyl, optionally substituted cyclopentyl, Formula (Ih) optionally substituted cyclohexyl, optionally substituted phe nyl, optionally Substituted adamantanyl, optionally Substi tuted azetidinyl, optionally substituted bicyclo[2.1.1 hexyl, Formula (Ih) optionally substituted bicyclo2.2.1]heptyl, optionally sub 55 stituted bicyclo[2.2.2]octyl, optionally substituted bicyclo 3.2.1]octyl, optionally substituted bicyclo[3.1.1 heptyl, optionally Substituted azabicyclo3.2.1]octanyl, optionally substituted azabicyclo[2.2.1]heptanyl, optionally substituted 2-azabicyclo[3.2.1]octanyl, optionally Substituted azabicy 60 clo3.2.2]nonanyl, optionally substituted bicyclo[2.2.1]hept 2-enyl, optionally substituted piperidinyl, optionally Substi tuted pyrrolidinyl or optionally substituted tetrahydrofuranyl. In a nineteenth embodiment the invention provides a com pound according to any of the foregoing embodiments In a fifteenth embodiment the invention provides com 65 wherein R is A-D-E-G. pound according to the first embodiment wherein T is S. U is In a twentieth embodiment the invention provides a com N, X is CR andY is C and forms a compound of Formula (li) pound according to any of the foregoing embodiments US 9,365,579 B2 21 22 wherein A is a bond, —C(O)—, optionally substituted (C- In a twenty-eighth embodiment the invention provides a C.)alkylene, -C(O)N(R)-R -, - N(R)C(O) R. , compound according to any of the foregoing embodiments O R , – N(R)-R -, - S R , —C(O-R) wherein A is a bond or optionally substituted (C-C)alky (R)-R , -S(O)N(R)-R -, -N(R)S(O), R - lene. or N(R)C(O)N(R) R. In a twenty-ninth embodiment the invention provides a In a twenty-first embodiment the invention provides a com compound according to any of the foregoing embodiments pound according to any of the foregoing embodiments wherein D is an optionally substituted cyclobutylene, option wherein D is optionally substituted azetidinyl, optionally sub ally Substituted cyclopentylene, optionally Substituted cyclo stituted bridged (C-C)cycloalkylene, optionally Substi hexylene, optionally Substituted aZetidinyl, optionally Substi tuted (C-C)cycloalkylene, optionally Substituted bridged 10 tuted bicyclo[2.2.1]heptylene, optionally substituted bicyclo (C-C)cycloalkenylene, optionally substituted (Cs-Co)cy 2.1.1 hexylene, bicyclo2.2.2]octanylene, optionally cloalkenylene, optionally Substituted (C-Co)arylene, substituted piperidine, or optionally substituted pyrrolidine: optionally Substituted (C-C)heteroarylene, optionally Sub E is R. C(O) R - R N(R) R - R N stituted bridged (C-C)heterocyclylene, or an optionally 15 (R)S(O), R , -R S(O), R , or - R S(O)N Substituted (C-Co)heterocyclylene. (R) R, wherein R for each occurrence is independently a In a twenty-second embodiment the invention provides a bond, an optionally Substituted (C-C)alkylene oran option compound according to any of the foregoing embodiments ally substituted (C-C)cycloalkylene; and wherein E is a bond, —R —R C(O)—R —R G is —CN, optionally substituted (C-C)alkyl, optionally O-R , R S(O), R , R N(R) R. , substituted cyclopropyl, optionally substituted cyclobutyl, R N(R)C(O) R. , - Re C(O)N(R) R. , optionally Substituted cyclopentyl, optionally Substituted - RN(R)S(O), R - R N(R)C(O)N(R) R. , phenyl, optionally substituted pyrazinyl, optionally Substi or - R S(O)N(R) R -. tuted pyridazinyl, optionally Substituted pyrimidinyl, option In a twenty-third embodiment the invention provides a ally substituted pyrazolyl, optionally substituted pyridinyl, compound according to any of the foregoing embodiments 25 optionally substituted thiazolidinyl or optionally substituted wherein G is OR, CN, N(R)S(O).R. —S(O)N(R) triazolyl. (R), optionally substituted (C-C)alkyl, optionally substi In a thirtieth embodiment the invention provides a com tuted (C-C)cycloalkyl, optionally Substituted (C-Co)het pound according to any of the foregoing embodiments eroaryl, optionally substituted (C-C)heterocyclyl or wherein D is an optionally substituted cyclobutylene, option 30 ally Substituted cyclopentylene, optionally Substituted cyclo optionally substituted phenyl. hexylene, optionally Substituted aZetidinyl, optionally Substi In a twenty-fourth embodiment the invention provides a tuted piperidine, optionally substituted bicyclo2.2.1 compound according to any of the foregoing embodiments heptylene, or bicyclo2.2.2]octanylene. wherein R is A-D-E-G and A is a bond, C(O) , optionally In a thirty-first embodiment the invention provides a com substituted (C-C)alkylene. —C(O)N(R')—R —N(R) 35 pound according to any of the foregoing embodiments wherein G is —CN, optionally substituted (C-C)alkyl, optionally Substituted cyclopropyl, optionally Substituted cyclobutyl, optionally Substituted cyclopentyl or optionally In a twenty-fifth embodiment the invention provides a Substituted phenyl, optionally Substituted pyrazinyl, option compound according to any of the foregoing embodiments 40 ally substituted pyridaZinyl, optionally Substituted pyrazolyl, wherein D is an optionally substituted azetidinyl, optionally or optionally substituted pyridinyl. substituted bicyclo[2.2.2]octanylene, optionally substituted In a thirty-second embodiment the invention provides a bicyclo[2.2.1]heptylene, optionally substituted bicyclo compound according to any of the foregoing embodiments 2.1.1 hexylene, optionally substituted cyclobutylene, wherein A is a bond, D is optionally substituted cyclopenty optionally Substituted cyclopentylene, optionally Substituted 45 lene, optionally Substituted bicyclo2.2.2]octanyl, optionally cyclohexylene, optionally substituted bicyclo[2.2.1]hept-2- substituted azetidinyl, or optionally substituted piperidine: enylene, optionally Substituted piperidine, or optionally Sub E is - R C(O)- R - R N(R)-R – R S stituted pyrrolidine. (O)N(R) R, R S(O), R , or R. N(R)S In a twenty-sixth embodiment the invention provides a (O), R : compound according to any of the foregoing embodiments 50 wherein R for each occurrence is independently a bond or an optionally Substituted (C-C)alkylene; and wherein E is - R C(O) R. , R O R, R S G is —CN, optionally substituted cyclopropyl, optionally (O), R - R N(R) R, R N(R)C(O) R. , substituted cyclobutyl, optionally substituted cyclopentyl, -R C(O)N(R)R , R. N(R)S(O), R , or optionally Substituted phenyl, optionally Substituted pyra R S(O)N(R)R -. 55 Zine, optionally Substituted pyridazine, optionally Substituted In a twenty-seventh embodiment the invention provides a pyrazole, or optionally Substituted pyridine. compound according to any of the foregoing embodiments In a thirty-third embodiment the invention provides a com wherein G is OR, CN, N(R)S(O).R. —S(O)N(R) pound according to any of the foregoing embodiments (R), optionally substituted (C-C)alkyl, optionally substi wherein G is —CN, optionally substituted cyclopropyl or tuted cyclopropyl, optionally Substituted cyclobutyl, option 60 optionally substituted cyclopentyl. ally Substituted cyclopentyl, optionally Substituted phenyl, In a thirty-fourth embodiment the invention provides a optionally Substituted pyridazine, optionally Substituted compound according to any of the foregoing embodiments pyrazine, optionally Substituted pyrimidine, optionally Sub wherein R', R. R. RandR when present are each inde stituted pyrazole, optionally Substituted pyrrolidine, option pendently hydrogen or an optionally Substituted —(C-C) ally Substituted quinazoline, optionally Substituted pyridinel, 65 alkyl. optionally substituted thiazolidinel or optionally substituted In a thirty-fifth embodiment the invention provides a com triazole. pound according to the first, second, fourth, fifth, seventh and US 9,365,579 B2 23 24 sixteenth through thirty-third embodiments wherein the com In a fortieth embodiment the invention provides a com pound is a compound of Formula (Ia) pound according to the first, fourth, fifth and sixteenth through thirty-third embodiments wherein the compound is Formula (Ia) R3 5 CH3 Ns / -( R2 H LY N N () NYs Y RI. s % NO 10 Ns-S C NV W H s

In a thirty-sixth embodiment the invention provides a com 15 pound according to the first, fourth, eighth, and sixteenth through thirty-third embodiments wherein the compound is a compound of Formula (Ib) In a forty-first embodiment the invention provides a com Formula (Ib) pound according to any of the foregoing embodiments R3 wherein the compound is

6 N-( R2 H R" SN I N R1 25 N N-4 R5 N NV H NS / \, N N N 30 In a thirty-seventh embodiment the invention provides a CyN compound according to the first, fourth, ninth and sixteenth N N through thirty-third embodiments wherein the compound is a H compound of Formula (Ic) 35 In a forty-second embodiment the invention provides a Formula (Ic) compound according to the first through fortieth embodi ments wherein A is a bond, D is optionally substituted cyclo pentylene or optionally substituted piperidine, E is —R N 40 (R) R. , R. S(O), N(R) R, R C(O) R, —R S(O) R, or R N(R')S(O), R ; and G is —CN, optionally substituted phenyl, optionally substituted pyrazine, optionally Substituted pyridazine, optionally Sub stituted pyrazole, or optionally substituted pyridine. 45 In a forty-third embodiment the invention provides a com In a thirty-eighth embodiment the invention provides a pound according to any of the foregoing embodiments compound according to any of the foregoing embodiments wherein T is CR. wherein T is N, U is N, X is CR and Y is N. In a forty-fourth embodiment the invention provides a In a thirty-ninth embodiment the invention provides a com 50 compound according to any of the foregoing embodiments pound according to the first, fourth, fifth and sixteenth wherein U is N. through thirty-third embodiments wherein the compound is In a forty-fifth embodiment the invention provides a com pound according to any of the foregoing embodiments 55 wherein X is CR. HC A In a forty-sixth embodiment the invention provides a com 3 v. ()s %Ys N. pound according to any of the foregoing embodiments wherein Y is N. 60 N s In a forty-seventh embodiment the invention provides a N-N compound according to any of the foregoing embodiments wherein T is CR, U is N, X is CR and Y is N. N Y N N In a forty-eighth embodiment the invention provides a H 65 compound according to the first, fourth, eighth, sixteenth through thirty-third, thirty-sixth and forty-second through forty-seventh embodiments wherein the compound is US 9,365,579 B2 25 26 Zole, optionally Substituted imidazoxazole, optionally Substi S tuted imidazopyrazine, optionally Substituted imidazopyri o-ys dine, optionally substituted indazole, optionally substituted N indole, optionally Substituted isoquinoline, optionally Substi tuted isothiazole, optionally Substituted isoxazole, optionally Substituted oxadiazole, optionally Substituted oxazole, optionally Substituted pyrazole, optionally Substituted pyri dine, optionally Substituted pyrimidine, optionally Substi CH3 tuted pyrazolopyridine, optionally Substituted pyrrole, - 10 optionally Substituted quinoline, optionally Substituted quinazoline, optionally Substituted thiazole, or optionally Cys N substituted thiophene. In a fifty-fourth embodiment the invention provides a com 15 pound according to the first through fifteenth and forty-sev In a forty-ninth embodiment the invention provides a com enth through fifty-second embodiments wherein R is hydro pound according to any of the foregoing embodiments gen, halogen, NH, or N(R)(R). wherein G is optionally substituted phenyl, optionally substi tuted pyrazine, optionally Substituted pyrazole, optionally In a fifty-fifth embodiment the invention provides a com substituted pyridazine or optionally substituted pyridine. pound according to the first through fifteenth and forty-sev In a fiftieth embodiment the invention provides a com enth through fifty-third embodiments wherein T is CH, U is pound according to the first through sixteenth embodiments N, Y is N, and X is CR wherein R is (C-C) optionally wherein R and Rare each independently hydrogen, deute Substituted alkyl, (C-C) optionally substituted cycloalkyl, rium, N(R)(R), halogen, —OR", -SR", -S(O)R", optionally substituted (C-C)aryl, optionally substituted S(O),R, NO, C(O)OR, CN, C(O)N(R)(R), 25 (C-Co)heteroaryl, or optionally Substituted (C-Co)hetero N(R)C(O)(R), C(O)R’, C(OH)RR, N(R)S cyclyl. (O) R', -S(O)N(R)(R), —CF, —OCF, optionally In a fifty-sixth embodiment the invention provides a com Substituted—(C-C)alkyl, optionally substituted —(C-C) pound according to the first through fifteenth and forty-sev cycloalkyl, optionally substituted benzo(b)thienyl, optionally enth through fifty-fourth embodiments wherein R is option substituted benzimidazole, optionally substituted benzofu 30 ally substituted pyrrolidine, optionally substituted piperidine, ran, optionally Substituted benzoxazole, optionally Substi optionally Substituted piperazine, optionally Substituted aze tuted benzothiazole, optionally substituted benzothiadiazole, tidine, optionally Substituted (C-Co.)aryl, or optionally Sub optionally substituted furan, optionally substituted imida stituted (C-Co)heterocyclyl. Zole, optionally substituted indoline, optionally substituted indole, optionally substituted indazole, optionally substituted 35 In a fifty-seventh embodiment the invention provides a isoxazole, optionally substituted isoindoline, optionally Sub compound according to the first through fifteenth and forty stituted morpholine, optionally Substituted oxadiazole, seventh through fifty-fourth embodiments wherein T is CH, U optionally Substituted phenyl, optionally Substituted pipera is N, Y is C and X is NR wherein R is (C-C) optionally Zine, optionally Substituted piperidine, optionally Substituted Substituted alkyl, (C-C) optionally substituted cycloalkyl, pyran, optionally Substituted pyrazole, optionally substituted 40 optionally substituted (C-C)aryl, optionally substitute pyrazolo 3,4-dipyrimidine, optionally Substituted pyridine, (C-Co)heteroaryl, or optionally Substituted (C-Co)hetero optionally substituted pyrimidine, optionally Substituted pyr cyclyl. rolidinel, optionally substituted pyrrole, optionally substi In a fifty-eighth embodiment the invention provides a com tuted optionally pyrrolo2,3-dipyrimidine, Substituted quino pound according to the first through fifteenth and forty-eighth line, optionally Substituted thiomorpholine, optionally 45 substituted tetrahydropyran, optionally substituted tetrahy through fifty-seventh embodiments wherein R is optionally drofuran, optionally substituted tetrahydroindol, optionally substituted pyrrolidine, optionally substituted piperidine, substituted thiazole, or optionally substituted thienyl. optionally Substituted piperazine, optionally Substituted aze In a fifty-first embodiment the invention provides a com tidine, optionally Substituted (C-Co.)aryl, or optionally Sub pound according to the first through sixteenth and forty 50 stituted (C-Co)heterocyclyl. seventh embodiments wherein R' is optionally substituted In a fifty-ninth embodiment the invention provides a com (Co-Co.)aryl or optionally Substituted (C-Co)heteroaryl. pound according to the first through fifteenth and forty-eighth In a fifty-second embodiment the invention provides a through fifty-eighth embodiments wherein T is N.U is N.Y is compound according to the first through sixteenth, forty N and X is CR wherein R is (C-C) optionally substituted seventh and fiftieth embodiments wherein R is hydrogen, 55 halogen, —CN, C(O)NR'R''. —CF, optionally substi alkyl, (C-C) optionally substituted cycloalkyl, optionally tuted (C-C)alkyl, optionally substituted (C-C)cy Substituted (C-Co)aryl, optionally substituted (C-C)het cloalkyl, optionally substituted (C-Co.)aryl, optionally Sub eroaryl, or optionally Substituted (C-Co)heterocyclyl. stituted (C-C)heteroaryl or optionally Substituted (C-Co) In a sixtieth embodiment the invention provides a com heterocyclyl. 60 pound according to the first through fifteenth and forty-eighth In a fifty-third embodiment the invention provides a com through fifty-nineth embodiments wherein R is optionally pound according to the first through fifteenth, forty-seventh substituted pyrrolidine, optionally substituted piperidine, and forty-ninth embodiments wherein R' is optionally sub optionally Substituted piperazine, optionally Substituted aze stituted azaindole, optionally Substituted benzofuran, option tidine, optionally Substituted (C-Co.)aryl, or optionally Sub ally substituted benzothiazole, optionally substituted benzox 65 stituted (C-Co)heterocyclyl. azole, optionally substituted dihydropyrroloimidazole, In a sixty-first embodiment the invention provides the use optionally substituted furan, optionally substituted imida of a compound of Formula 2: US 9,365,579 B2 27 28 ally Substituted (C-C)alkynylene, optionally Substituted Formula 2 (C-C)cycloalkylene, optionally substituted (C-C)het ". . HN N

DC 2 N RI R5 N N D is an optionally Substituted (C-Cs)alkylene, optionally Ye Substituted bridged (C-C)cycloalkylene, optionally substi tuted (C-C)cycloalkylene, optionally Substituted bridged 10 (Cs-Co)cycloalkenylene, optionally Substituted (C-C)cy to form a compound of Formula (Ia) cloalkenylene, optionally substituted (C-C)arylene, optionally Substituted (C-C)heteroarylene, optionally Sub stituted bridged (C-C)heterocyclylene or an optionally Formula (Ia) R3 Substituted (C-Co)heterocyclylene; 15 E is a bond, -R , -R C(O) R , -R C(O)C (O) R , R C(O)O R. , R C(O)C(O)N N N (R) R - R N(R) C(O)C(O) R - R O R , -R S(O), R , R. S(O) R. , -R S R , R. N(R) R. , R. N(R)C(O) R. , N N R5 N RC(O)N(R)R , R OC(O)N(R)- R - R - V N(R)C(O)CR , R OC(O) R, R N(R)C(O) H N(R)-R , -R N(R)S(O), R , or - R S(O), N(R) R ; or pharmaceutically acceptable salts, pro-drugs, biologically E is active metabolites, stereoisomers and isomers thereof 25 wherein R is a hydrogen, SON(CH), —SO,(2,4,6-trimeth ylphenyl), —SO phenyl, -SO(4-butylphenyl). —SO(4- methylphenyl), —SO(4-methoxyphenyl), - C(O) OCHCC1, C(O)CCHCHSi(CH), C(O)OC(CH), N- Ra; —C(O)OC(CH)(CCI), —C(O)C-1-adamantyl, I -CH=CH, CHCHCl, -CH(OCHCH)CH, Re —CHCH-2-pyridyl, —CHCH-4-pyridyl, Si(C(CH)) (CH), —Si(CH(CH)), —CH2phenyl, —CH2(4-CHO 35 where in all cases, E is linked to either a carbon or a phenyl), —CH2(3,4-di-methoxyphenyl), —CH2(2-nitrophe nitrogen atom in D; nyl), -(2,4-dinitrophenyl), —CHC(O)phenyl, -C(phenyl). G is hydrogen, deuterium, N(R)(R'), halogen, OR", —CH(phenyl), —C(phenyl)-(4-pyridyl), —N(CH), - SR", S(O)R', S(O).R, NO, C(O)OR, CN, —CH-OH, —CHOCH, —CH(OCHCH), C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, —CHOCHCHCl, -CHOCHCH-Si(CH), —CHOC 40 OC(O)N(R), N(R)C(O)N(R), C(O-R)(R), (CH), —CHOC(O)C(CH), —CHOCHphenyl, -(2-tet —C(O)R", -CF, OCF, N(R)S(O).R., -S(O)N rahydropyranyl), —C(O)H, or—P(S)(phenyl); (R)(R), S(O)N(R)C(O)R, an optionally substituted R", R and Rare each independently hydrogen, deute —(C-C)alkyl, an optionally Substituted —(C-C)alkenyl, rium, N(R)(R), halogen, —OR", -SR", -S(O)R", an optionally Substituted —(C-C)alkynyl, an optionally S(O),R, NO, C(O)OR, CN, C(O)N(R)(R), 45 Substituted —(C-Co)cycloalkyl, an optionally Substituted N(R)C(O)(R), C(O)R", C(OH)RR, N(R)S —(C-Co)heteroaryl, an optionally Substituted —(C-Co) (O) R, -S(O)N(R)(R), CF, OCF, optionally heterocyclyl, an optionally substituted —(C-C)aryl, an Substituted (C-C)alkyl, optionally Substituted (C-C)alk optionally substituted —(C-C)alkyl-(C-C)cycloalkyl, enyl, optionally substituted (C-C)alkynyl, optionally sub an optionally substituted—(C-C)alkylene-(C-C)aryl, an stituted (C-Co)cycloalkyl, optionally Substituted (C-Co) 50 optionally substituted —(C-C)alkylene-(C-Co)het heteroaryl, optionally Substituted (C-C) heterocyclyl, or eroaryl, or an optionally Substituted —(C-C)alkylene-(C- optionally substituted (C-Co)aryl; Co)heterocyclyl, wherein in a moiety containing N(R)(R'), the nitrogen, wherein in a moiety containing N(R)(R), the nitrogen, R" and R may form a ring such that N(R)(R') rep R" and R may form a ring such that N(R)(R) represents resents an optionally substituted (C-Co)heterocyclyl 55 an optionally Substituted (C-Co)heterocyclyl or an option or optionally substituted (C-C)heteroaryl linked ally substituted (C-C) heteroaryl linked through a nitro through a nitrogen; gen, R is hydrogen, an optionally substituted bridged (C5-C12) R" and Rare each independently hydrogen, deuterium, an cycloalkyl, optionally substituted bridged (C-C)heterocy optionally Substituted (C-Co.)alkyl, an optionally Substi clyl, optionally substituted (C-Cs)alkyl, optionally Substi 60 tuted (C-Co.)alkenyl, an optionally Substituted (C-Co) tuted (C-C)cycloalkyl, optionally substituted (C-C) alkynyl, an optionally substituted —(C-C)alkylene-O- cycloalkenyl, optionally Substituted (C-Co.)aryl, optionally (C-Co.)alkyl, an optionally Substituted (C-Co)cycloalkyl, Substituted (C-C)heteroaryl, optionally substituted (C- an optionally substituted (C-C)aryl, an optionally substi Co)heterocyclyl; or tuted (C-C)heteroaryl, an optionally Substituted (C-Co) Ris-A-D-E-G, wherein: 65 heterocyclyl, an optionally Substituted —(C-C)alkylene A is a bond, —C(O)—, optionally substituted (C-C) (C-Co)cycloalkyl, an optionally Substituted —(C-C) alkylene, optionally Substituted (C-C)alkenylene, option alkylene-(C-C)aryl, an optionally Substituted —(C-C) US 9,365,579 B2 29 30 alkylene-(C-Co)heteroaryl, or an optionally Substituted resents an optionally substituted (C-Co)heterocyclyl —(C-C)alkylene-(C-C)heterocyclyl; and or optionally substituted (C-C)heteroaryl linked R for each occurrence is independently a bond, an option through a nitrogen; ally Substituted (C-Co.)alkylene, an optionally Substituted R is hydrogen, an optionally substituted bridged (C5-C12) (C-C)alkenylene, an optionally substituted (C-C)alky 5 cycloalkyl, optionally substituted bridged (C-C)heterocy nylene, an optionally substituted —(C-Co.)alkylene-O- clyl, optionally substituted (C-Cs)alkyl, optionally Substi (C-Co.)alkylene group, an optionally substituted (C-Co) tuted (C-C)cycloalkyl, optionally Substituted (C-Cs) cycloalkylene, an optionally substituted (C-C)arylene, an cycloalkenyl, optionally substituted (C-C)aryl, optionally optionally Substituted (C-C)heteroarylene, or an option Substituted (C-C)heteroaryl, optionally Substituted (C- ally Substituted (C-Co)heterocyclylene. 10 Co)heterocyclyl; or In a sixty-second embodiment the invention provides the R is -A-D-E-G, wherein: use of a compound of Formula 3: A is a bond, —C(O)—, optionally Substituted (C-C) alkylene, optionally substituted (C-C)alkenylene, option ally Substituted (C-C)alkynylene, optionally Substituted 15 Formula 3 (C-C)cycloalkylene, optionally substituted (C-C)het NH2 R2 R6 N n N R1 2 D is an optionally substituted (C-C)alkylene, optionally R 5 N NV Substituted bridged (C-C)cycloalkylene, optionally substi RP tuted (C-C)cycloalkylene, optionally Substituted bridged (Cs-Co)cycloalkenylene, optionally Substituted (C-C)cy to form a compound of Formula (Ib) cloalkenylene, optionally substituted (C-Co)arylene, 25 optionally substituted (C-C)heteroarylene, optionally sub stituted bridged (C-C)heterocyclylene or an optionally Formula (Ib) Substituted (C-C)heterocyclylene; R3 E is a bond, -R , -R C(O) R , -R C(O)C Ns (O) R , R C(O)O R. , R C(O)C(O)N S( 30 (R) R - R N(R) C(O)C(O) R - R O R6 N N R , -R S(O), R -, -R S(O) R , -R Y-R, S R , R. N(R) R. , R N(R)C(O) R. , N RC(O)N(R)R , R OC(O)N(R) R - R - R5 N NV N(R)C(O)CR , R OC(O) R, R N(R)C(O) H 35 N(R)-R , -R N(R)S(O), R , or - R S(O), N(R) R ; or pharmaceutically acceptable salts, pro-drugs, biologically E is active metabolites, stereoisomers and isomers thereof wherein 40 R is a hydrogen, SON(CH), —SO,(2,4,6-trimeth ylphenyl), —SO phenyl, -SO(4-butylphenyl). —SO(4- methylphenyl), —SO(4-methoxyphenyl), - C(O) OCHCC1, C(O)CCHCHSi(CH), C(O)OC(CH), N- Ra; —C(O)OC(CH)(CC1), —C(O)C)-1-adamantyl, 45 I -CH=CH, CHCHCl, -CH(OCHCH)CH, Re —CHCH-2-pyridyl, —CHCH-4-pyridyl, Si(C(CH)) (CH), —Si(CH(CH)), —CH2phenyl, —CH2(4-CHO where in all cases, E is linked to either a carbon or a phenyl), —CH2(3,4-di-methoxyphenyl), —CH2(2-nitrophe nitrogen atom in D; nyl), -(2,4-dinitrophenyl), —CHC(O)phenyl, —C(phenyl), 50 G is hydrogen, deuterium, N(R)(R'), halogen, OR", —CH(phenyl), —C(phenyl)-(4-pyridyl), —N(CH), - SR", S(O)R', S(O).R, NO, C(O)OR, CN, —CH-OH, —CHOCH, —CH(OCHCH), C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, —CHOCHCHCl, -CHOCH2CHSiCCH), —CHOC OC(O)N(R), N(R)C(O)N(R), C(O-R)(R), (CH), —CHOC(O)C(CH), CHOCH phenyl, -(2-tet —C(O)R’, -CF, OCF, N(R)S(O).R. —S(O)N rahydropyranyl), —C(O)H, or—P(S)(phenyl); 55 (R)(R), -S(O)N(R)C(O)R’, an optionally substituted R", R and R are each independently hydrogen, deute —(C-C)alkyl, an optionally substituted —(C-C)alkenyl, rium, N(R)(R), halogen, —OR", -SR", -S(O)R", an optionally Substituted —(C-C)alkynyl, an optionally S(O),R, NO, C(O)OR, CN, C(O)N(R)(R), Substituted —(C-Co)cycloalkyl, an optionally Substituted N(R)C(O)(R), C(O)R’, C(OH)RR, N(R)S —(C-Co)heteroaryl, an optionally Substituted —(C-Co) (O) R', -S(O)N(R)(R), —CF, —OCF, optionally 60 heterocyclyl, an optionally substituted —(C-Co.)aryl, an Substituted (C-C)alkyl, optionally substituted (C-C)alk optionally substituted —(C-C)alkylene-(C-C)cy enyl, optionally Substituted (C-C)alkynyl, optionally Sub cloalkyl, an optionally substituted —(C-C)alkylene-(C- stituted (C-C)cycloalkyl, optionally substituted (C-C) Co)aryl, an optionally substituted —(C-C)alkylene-(C- heteroaryl, optionally Substituted (C-C) heterocyclyl, or Co)heteroaryl, or an optionally Substituted —(C-C) optionally substituted (C-Co)aryl; 65 alkylene-(C-Co)heterocyclyl; wherein in a moiety containing N(R)(R), the nitrogen, wherein in a moiety containing N(R)(R), the nitrogen, R" and R may form a ring such that N(R)(R') rep R" and R may form a ring such that N(R)(R') rep US 9,365,579 B2 31 32 resents an optionally substituted (C-Co)heterocyclyl resents an optionally substituted (C-Co)heterocyclyl or an optionally substituted (C-C) heteroaryl linked or an optionally substituted (C-C) heteroaryl linked through a nitrogen; through a nitrogen; R is a hydrogen, halogen, deuterium, an optionally sub R" and Rare each independently hydrogen, deuterium, an stituted bridged (C-C)cycloalkyl group, optionally substi 5 optionally substituted (C-C)alkyl, an optionally substi tuted bridged (C-C)heterocyclyl group, optionally Substi tuted (C-Co.)alkenyl, an optionally Substituted (C-Co) tuted (C-Cs)alkyl, optionally Substituted (C-Co) alkynyl, an optionally substituted —(C-Co.)alkylene-O- cycloalkyl, optionally substituted (C-C)cycloalkenyl, (C-C)alkyl, an optionally substituted (C-C)cycloalkyl, optionally Substituted (C-Co)aryl, optionally Substituted an optionally Substituted (C-C)aryl, an optionally Substi (C-Co)heteroaryl, optionally Substituted (C-C)heterocy 10 tuted (C-C)heteroaryl, an optionally Substituted (C-Co) clyl or -J.-L-M-Q; heterocyclyl, an optionally substituted —(C-C)alkylene wherein: (C-Co)cycloalkyl, an optionally Substituted —(C-C) J is a bond, —C(O)—, optionally substituted (C-C)alky alkylene-(C-C)aryl, an optionally Substituted —(C-C) lene, optionally Substituted (C-C)alkenylene, optionally alkylene-(C-C)heteroaryl, or an optionally substituted Substituted (C-C)alkynylene, optionally substituted (C- 15 —(C-C)alkylene-(C-Co)heterocyclyl; and C)cycloalkylene, optionally Substituted (C-C)heterocy R for each occurrence is independently a bond, an option ally substituted (C-C)alkylene, an optionally substituted (C-Co.)alkenylene, an optionally Substituted (C-C)alky R - N(R)S(O), R or N(R)C(O)N(R) Re . nylene, an optionally substituted —(C-Co.)alkylene-O- L is a bond, an optionally Substituted (C-Cs)alkylene, (C-C)alkylene group, an optionally substituted (C-C) optionally Substituted bridged (C-C)cycloalkylene, cycloalkylene, an optionally substituted (C-Co)arylene, an optionally Substituted (C-C)cycloalkylene, optionally optionally Substituted (C-C)heteroarylene, or an option Substituted bridged (Cs-Co)cycloalkenylene, optionally Sub ally substituted (C-C)heterocyclylene. stituted (C-C)cycloalkenylene, optionally substituted (C- 25 In a sixty-third embodiment the invention provides use of a Co)arylene, optionally substituted (C-Co)heteroarylene, compound of Formula 4: optionally substituted bridged (C-C)heterocyclylene oran optionally substituted (C-Co)heterocyclylene; Formula 4 Rp2 30 R2 HN 4.

N N 35 R5 N V RPI

to form a compound of Formula (Ic)

40 Formula (Ic) R4 R3 )-( R2 45 N N-N

N R5 N N where in all cases, M is linked to either a carbon or a H nitrogen atom in L.; 50 Q is hydrogen, deuterium, N(R)(R), halogen, OR", - SR", S(O)R, S(O).R, NO, C(O)OR, CN, or pharmaceutically acceptable salts, pro-drugs, biologically C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, active metabolites, stereoisomers and isomers thereof N(R)C(O)N(R), C(O-R)(R), C(O)R, CF, wherein OCF, N(R)S(O).R., S(O)N(R)(R), S(O)N 55 R" is hydrogen, -SO.N(CH), -SO,(2,4,6-trimeth (R)C(O)R’, an optionally substituted (C-C)alkyl, an ylphenyl). —SO phenyl, -SO(4-butylphenyl). —SO(4- optionally Substituted (C-C)alkenyl, an optionally Substi methylphenyl), —SO(4-methoxyphenyl), - C(O) tuted (C-C)alkynyl, an optionally Substituted (C-C)cy OCHCC1, C(O)OCHCHSiCCH), C(O)OC(CH), cloalkyl, an optionally Substituted (C-Co)heteroaryl, an - C(O)OC(CH)(CC1), —C(O)C)-1-adamantyl, optionally substituted (C-Co)heterocyclyl, an optionally 60 -CH=CH, CHCHCl, -CH(OCHCH)CH, Substituted (C-C)aryl, an optionally substituted—(C-C) —CHCH-2-pyridyl, —CHCH-4-pyridyl, Si(C(CH)) alkylene-(C-C)cycloalkyl, an optionally Substituted (CH), —Si(CH(CH)), —CH2phenyl, —CH2(4-CHO —(C-C)alkylene-(C-C)aryl, an optionally substituted phenyl), —CH2(3,4-di-methoxyphenyl), —CH2(2-nitrophe —(C-C)alkylene-(C-Co)heteroaryl, or an optionally Sub nyl), -(2,4-dinitrophenyl), —CHC(O)phenyl, —C(phenyl), stituted —(C-C)alkylene-(C-C)heterocyclyl; 65 —CH(phenyl), —C(phenyl)-(4-pyridyl), —N(CH), wherein in a moiety containing N(R)(R), the nitrogen, - CH-OH, —CHOCH, —CH(OCHCH), R" and R may form a ring such that N(R)(R') rep —CHOCHCHCl, -CHOCH2CHSiCCH), —CHOC US 9,365,579 B2 33 34 (CH), —CHOC(O)C(CH), CHOCH-phenyl, -(2-tet G is hydrogen, deuterium, N(R)(R'), halogen, OR", rahydropyranyl), —C(O)H, or—P(S)(phenyl); - SR", S(O)R', S(O),R, NO,-C(O)OR, CN, R’ is hydrogen, C(O)C C(CH), —C(O)OCH-phe C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, nyl, —C(O)O-fluoren-9-yl, —C(O)CH, —C(O)CF, OC(O)N(R), N(R)C(O)N(R), C(O-R)(R), —C(O)—CH(CH), —CH2-phenyl, —CH-(4-methox —C(O)R’, -CF, OCF, N(R)S(O).R. —S(O)N yphenyl), —S(O)-phenyl or —S(O)-(4-methylphenyl); (R)(R), -S(O)N(R)C(O)R’, an optionally substituted R", R and Rare each independently hydrogen, deute —(C-C)alkyl, an optionally Substituted —(C-C)alkenyl, rium, N(R)(R), halogen, OR, - SR", -S(O)R", an optionally substituted —(C-C)alkynyl, an optionally S(O),R, NO, C(O)OR, CN, C(O)N(R)(R), Substituted —(C-Co)cycloalkyl, an optionally Substituted N(R)C(O)(R), C(O)R", C(OH)RR, N(R)S 10 —(C-Co)heteroaryl, an optionally Substituted —(C-Co) (O) R, -S(O)N(R)(R), CF, OCF, optionally heterocyclyl, an optionally substituted —(C-C)aryl, an Substituted (C-C)alkyl, optionally Substituted (C-C)alk optionally Substituted —(C-C)alkylene-(C-C)cy enyl, optionally substituted (C-C)alkynyl, optionally sub cloalkyl, an optionally substituted —(C-C)alkylene-(C- stituted (C-Co)cycloalkyl, optionally Substituted (C-Co) Co)aryl, an optionally substituted —(C-C)alkylene-(C- heteroaryl, optionally substituted (C-C) heterocyclyl, or 15 Co)heteroaryl, or an optionally Substituted —(C-C) optionally substituted (C-Co)aryl; wherein in a moiety containing N(R)(R'), the nitrogen, alkylene-(C-Co)heterocyclyl; R" and R may form a ring such that N(R)(R') rep wherein in a moiety containing N(R)(R), the nitrogen, resents an optionally substituted (C-Co)heterocyclyl R" and R may form a ring such that N(R)(R') rep or optionally substituted (C-C)heteroaryl linked resents an optionally substituted (C-Co)heterocyclyl through a nitrogen; or an optionally substituted (C-C) heteroaryl linked R is hydrogen, an optionally substituted bridged (C5-C12) through a nitrogen; cycloalkyl, optionally substituted bridged (C-C)heterocy R" is a hydrogen, halogen, deuterium, an optionally Sub clyl, optionally substituted (C-Cs)alkyl, optionally Substi stituted bridged (Cs-C)cycloalkyl group, optionally substi tuted (C-C)cycloalkyl, optionally substituted (C-C) 25 tuted bridged (C-C)heterocyclyl group, optionally Substi cycloalkenyl, optionally Substituted (C-Co.)aryl, optionally tuted (C-Cs)alkyl, optionally Substituted (C-Co) Substituted (C-C)heteroaryl, optionally substituted (C- cycloalkyl, optionally Substituted (C-Cs)cycloalkenyl, Co)heterocyclyl; or optionally substituted (C-C)aryl, optionally substituted Ris-A-D-E-G, wherein: (C-Co)heteroaryl, optionally substituted (C-C)heterocy A is a bond, —C(O)—, optionally substituted (C-C) 30 clyl or -J.-L-M-Q; alkylene, optionally Substituted (C-C)alkenylene, option wherein: J is a bond, —C(O)—, optionally substituted (C-C)alky ally substituted (C-C)alkynylene, optionally substituted lene, optionally substituted (C-C)alkenylene, optionally (C-C)cycloalkylene, optionally Substituted (C-C)het Substituted (C-C)alkynylene, optionally Substituted (C- 35 C)cycloalkylene, optionally substituted (C-C)heterocy

D is an optionally Substituted (C-Cs)alkylene, optionally Substituted bridged (C-C)cycloalkylene, optionally substi R - N(R)S(O), R or N(R)C(O)N(R) R. ; tuted (C-C)cycloalkylene, optionally Substituted bridged 40 L is a bond, an optionally substituted (C-C)alkylene, (C-C)cycloalkenylene, optionally substituted (C-C)cy optionally Substituted bridged (C-C)cycloalkylene, cloalkenylene, optionally Substituted (C-Co)arylene, optionally substituted (C-C)cycloalkylene, optionally optionally Substituted (C-C)heteroarylene, optionally Sub Substituted bridged (Cs-Co)cycloalkenylene, optionally Sub stituted bridged (C-C)heterocyclylene or an optionally stituted (C-Co)cycloalkenylene, optionally Substituted (C- Substituted (C-Co)heterocyclylene; 45 Co)arylene, optionally substituted (C-Co)heteroarylene, E is a bond, - R -, -R C(O) R , -R C(O)C optionally substituted bridged (C-C)heterocyclylene oran (O) R , R C(O)O R. , R C(O)C(O)N optionally substituted (C-C)heterocyclylene; (R) R - R N(R) C(O)C(O) R - R O M is a bond, - R -, -R C(O) R , -R C(O)C R , -R S(O), R -, -R S(O) R , -R (O) R-, -R C(O)O R , R OC(O) R, S R , R. N(R) R. , R. N(R)C(O) R. , 50 R. C(O)C(O)N(R) R. , R. N(R) C(O)C RC(O)N(R)R , R OC(O)N(R)- R - R - (O) R - R O R - R S(O), R -, -R N(R)C(O)CR , R OC(O) R, R N(R)C(O) S(O) R. , R. S. R. , R N(R) R. , N(R)-R , -R N(R)S(O), R , or -R S(O), R N(R)C(O) R - R C(O)N(R)R - R - N(R) R ; or OC(O)N(R) R R N(R)C(O)OR R N E is 55 (R)C(O)N(R) R. , R. N(R)S(O), R , or

60

N- Ra;

Re 65 where in all cases, E is linked to either a carbon or a nitrogen atom in D; US 9,365,579 B2 35 36 where in all cases, Mis linked to either a carbon or a nitrogen to form a compound of Formula (Id) atom in L.; Q is hydrogen, deuterium, N(R)(R), halogen, —OR", Formula (Id) - SR", S(O)R', S(O),R, NO,-C(O)OR, CN, R4 R3

C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, r R2 N(R)C(O)N(R), C(O R)(R), C(O)R, CF, 6 OCF, N(R)S(O).R., S(O)N(R)(R), S(O)N R N-N (R)C(O)R’, an optionally substituted (C-C)alkyl, an 10 N Y-R, optionally Substituted (C-C)alkenyl, an optionally Substi R5 N NV tuted (C-C)alkynyl, an optionally Substituted (C-C)cy H cloalkyl, an optionally Substituted (C-Co)heteroaryl, an optionally Substituted (C-C) heterocyclyl, an optionally 15 pharmaceutically acceptable salts, pro-drugs, biologically Substituted (C-C)aryl, an optionally Substituted —(C-C) active metabolites, stereoisomers and isomers thereof wherein alkylene-(C-C)cycloalkyl, an optionally Substituted RP is hydrogen, —SON(CH), —SO(2,4,6-trimeth —(C-C)alkylene-(Co-Co)aryl, an optionally substituted ylphenyl), —SO phenyl, -SO(4-butylphenyl). —SO(4- —(C-C)alkylene-(C-Co)heteroaryl, or an optionally Sub methylphenyl), —SO(4-methoxyphenyl), - C(O) stituted —(C-C)alkylene-(C-C)heterocyclyl; OCHCC1, C(O)OCHCHSi(CH), —C(O)OC(CH), - C(O)OC(CH)(CC1), —C(O)C)-1-adamantyl, wherein in a moiety containing N(R)(R), the nitrogen, -CH=CH, CHCHCl, -CH(OCHCH)CH, R" and R may form a ring such that N(R)(R') rep —CHCH-2-pyridyl, —CHCH-4-pyridyl, Si(C(CH)) 25 (CH), —Si(CH(CH)2), —CH2-phenyl, —CH2(4-CHO resents an optionally substituted (C-Co)heterocyclyl phenyl), —CH2(3,4-di-methoxyphenyl), —CH2(2-nitrophe or an optionally Substituted (C-C) heteroaryl linked nyl), -(2,4-dinitrophenyl), —CHC(O)phenyl, —C(phenyl), through a nitrogen; —CH(phenyl), —C(phenyl)-(4-pyridyl). —N(CH), R" and Rare each independently hydrogen, deuterium, an - CH-OH, —CHOCH, —CH(OCHCH), 30 —CHOCHCHCl, -CHOCHCH-Si(CH), —CHOC optionally Substituted (C-Co.)alkyl, an optionally Substi (CH), —CHOC(O)C(CH), CHOCH-phenyl, -(2-tet tuted (C-C)alkenyl, an optionally Substituted (C-C) rahydropyranyl), —C(O)H, or—P(S)(phenyl): alkynyl, an optionally substituted —(C-C)alkylene-O- R", R and R are each independently hydrogen, deute (C-C)alkyl, an optionally substituted (C-C)cycloalkyl, rium, N(R)(R), halogen, —OR", -SR", -S(O)R", 35 - S(O),R, NO,-C(O)OR, CN, C(O)N(R)(R), an optionally substituted (C-C)aryl, an optionally substi N(R)C(O)(R), C(O)R", C(OH)RR, N(R)S tuted (C-C)heteroaryl, an optionally substituted (C-C) (O) R', -S(O)N(R)(R), —CF, —OCF, optionally heterocyclyl, an optionally substituted —(C-C)alkylene Substituted (C-C)alkyl, optionally Substituted (C-C)alk (C-C)cycloalkyl, an optionally substituted —(C-C) enyl, optionally Substituted (C-C)alkynyl, optionally Sub 40 stituted (C-C)cycloalkyl, optionally substituted (C-C) alkylene-(C-C)aryl, an optionally Substituted —(C-C) heteroaryl, optionally Substituted (C-C) heterocyclyl, or alkylene-(C-Co)heteroaryl, or an optionally Substituted optionally substituted (C-C)aryl; —(C-C)alkylene-(C-Co)heterocyclyl; and wherein in a moiety containing N(R)(R), the nitrogen, R for each occurrence is independently a bond, an option R" and R may form a ring such that N(R)(R') represents 45 an optionally Substituted (C-C)heterocyclyl or optionally ally substituted (C-C)alkylene, an optionally substituted Substituted (C-Co)heteroaryl linked through a nitrogen; (C-Co.)alkenylene, an optionally Substituted (C-Co.)alky R is hydrogen, an optionally substituted bridged (Cs-C) nylene, an optionally substituted —(C-Co.)alkylene-O- cycloalkyl, optionally Substituted bridged (C-C)heterocy (C-Co.)alkylene group, an optionally substituted (C-Co) clyl, optionally substituted (C-C)alkyl, optionally substi 50 tuted (C-C)cycloalkyl, optionally Substituted (C-Cs) cycloalkylene, an optionally Substituted (C-Co.)arylene, an cycloalkenyl, optionally Substituted (C-Co.)aryl, optionally optionally Substituted (C-C)heteroarylene, or an option Substituted (C-C)heteroaryl, optionally Substituted (C- ally Substituted (C-Co)heterocyclylene. Co)heterocyclyl; or In a sixty-fourth embodiment the invention provides the R is -A-D-E-G, wherein: 55 A is a bond, —C(O)—, optionally Substituted (C-C) use of a compound of Formula 5 alkylene, optionally substituted (C-C)alkenylene, option ally Substituted (C-C)alkynylene, optionally Substituted (C-C)cycloalkylene, optionally Substituted (C-C)het Formula 5 O 2 R 60 N R6 21

S. Y-R D is an optionally substituted (C-C)alkylene, optionally R5 N N Substituted bridged (C-C)cycloalkylene, optionally substi V RP 65 tuted (C-C)cycloalkylene, optionally Substituted bridged (Cs-Co)cycloalkenylene, optionally Substituted (C-C)cy cloalkenylene, optionally substituted (C-Co)arylene, US 9,365,579 B2 37 38 optionally Substituted (C-C)heteroarylene, optionally Sub stituted (C-Co)cycloalkenylene, optionally Substituted (C- stituted bridged (C-C)heterocyclylene or an optionally Co)arylene, optionally substituted (C-C)heteroarylene, Substituted (C-Co)heterocyclylene; optionally substituted bridged (C-C)heterocyclylene oran E is a bond, - R -, -R C(O) R , -R C(O)C optionally substituted (C-Co)heterocyclylene; (O) R. , R C(O)O Re . Re C(O)C(O)N 5 M is a bond, - R -, -R C(O) R , -R C(O)C (R) R - R N(R) C(O)C(O) R - R O (O) R , R C(O)O R. , R OC(O) R, R , -R S(O), R -, -R S(O) R , -R R C(O)C(O)N(R) R. , R N(R) C(O)C S R , R. N(R) R. , R N(R)C(O) R. , (O) R-, -R-O-R-, -R S(O), R -, -R RC(O)N(R)R , R OC(O)N(R) R - R - S(O)—R , R S R , R N(R) R. , N(R)C(O)OR , R OC(O) R, R N(R)C(O) ' R N(R)C(O) R - R C(O)N(R)R - R - N(R)-R , R N(R)S(O), R , or R S(O), OC(O)N(R) Re Re N(R)C(O)OR Re N N(R) R ; or (R)C(O)N(R) R. , R. N(R)S(O), R , or E is

15

N-R's I N-R Re I Re where in all cases, E is linked to either a carbon or a nitrogen atom in D; 25 where in all cases, Mis linked to either a carbon or a nitrogen G is hydrogen, deuterium, N(R)(R), halogen, —OR", atom in L. - SR", S(O)R', S(O).R, NO, C(O)OR, CN, C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, Q is hydrogen, deuterium, N(R)(R), halogen, OR", OC(O)N(R), N(R)C(O)N(R), C(O R)(R), - SR", S(O)R', S(O).R, NO, C(O)OR, CN, —C(O)R, CF, OCF, N(R)S(O),R, S(O)N 30 C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, (R)(R), S(O)N(R)C(O)R’, an optionally substituted N(R)C(O)N(R), C(O-R)(R), C(O)R, CF, —(C-C)alkyl, an optionally substituted —(C-C)alkenyl, OCF, N(R)S(O),R, S(O)N(R)(R), S(O)N an optionally substituted —(C-C)alkynyl, an optionally (R)C(O)R’, an optionally substituted (C-C)alkyl, an Substituted —(C-C)cycloalkyl, an optionally Substituted optionally substituted (C-C)alkenyl, an optionally substi —(C-C)heteroaryl, an optionally substituted —(C-C) 35 tuted (C-C)alkynyl, an optionally Substituted (C-C)cy heterocyclyl, an optionally substituted —(C-Co)aryl, an cloalkyl, an optionally Substituted (C-Co)heteroaryl, an optionally Substituted —(C-C)alkylene-(C-C)cy optionally substituted (C-C)heterocyclyl, an optionally cloalkyl, an optionally Substituted —(C-C)alkylene-(C- Substituted (C-C)aryl, an optionally Substituted —(C-C) Co)aryl, an optionally Substituted —(C-C)alkylene-(C- alkylene-(C-C)cycloalkyl, an optionally Substituted Co)heteroaryl, or an optionally substituted —(C-C)alkyl 40 —(C-C)alkyl-(C-C)aryl, an optionally substituted (C-Co)heterocyclyl; —(C-C)alkylene-(C-Co)heteroaryl, or an optionally Sub wherein in a moiety containing N(R)(R), the nitrogen, stituted —(C-C)alkylene-(C-C)heterocyclyl: R" and R may form a ring such that N(R)(R') rep wherein in a moiety containing N(R)(R), the nitrogen, resents an optionally substituted (C-Co)heterocyclyl R" and R may form a ring such that N(R)(R') rep or an optionally Substituted (C-C) heteroaryl linked 45 resents an optionally substituted (C-Co)heterocyclyl through a nitrogen; or an optionally Substituted (C-C) heteroaryl linked R" and Rare each independently a hydrogen, halogen, through a nitrogen; deuterium, an optionally substituted bridged (C-C)cy R" and Rare each independently hydrogen, deuterium, an cloalkyl group, optionally substituted bridged (C-C)het optionally substituted (C-C)alkyl, an optionally substi erocyclyl group, optionally Substituted (C-Cs)alkyl, option 50 tuted (C-Co.)alkenyl, an optionally Substituted (C-Co) ally Substituted (C-Co)cycloalkyl, optionally Substituted alkynyl, an optionally substituted —(C-Co.)alkylene-O- (C-C)cycloalkenyl, optionally substituted (C-Co)aryl, (C-Co.)alkyl, an optionally Substituted (C-Co)cycloalkyl, optionally substituted (C-Co)heteroaryl, optionally Substi an optionally Substituted (C-C)aryl, an optionally Substi tuted (C-C)heterocyclyl or -J.-L-M-Q; tuted (C-C)heteroaryl, an optionally substituted (C-C) wherein: 55 heterocyclyl, an optionally Substituted —(C-C)alkylene J is a bond, —C(O)—, optionally substituted (C-C)alky (C-C)cycloalkyl, an optionally substituted —(C-C) lene, optionally Substituted (C-C)alkenylene, optionally alkylene-(C-C)aryl, an optionally Substituted —(C-C) Substituted (C-C)alkynylene, optionally Substituted (C- alkylene-(C-Co)heteroaryl, or an optionally Substituted C)cycloalkylene, optionally Substituted (C-C)heterocy —(C-C)alkylene-(C-Co)heterocyclyl; and 60 R for each occurrence is independently a bond, an option ally substituted (C-C)alkylene, an optionally substituted (C-Co.)alkenylene, an optionally Substituted (C-C)alky nylene, an optionally substituted —(C-C)alkylene-O- L is a bond, an optionally Substituted (C-Cs)alkylene, (C-Co.)alkylene group, an optionally substituted (C-Co) optionally Substituted bridged (C-C)cycloalkylene, 65 cycloalkylene, an optionally substituted (C-Co)arylene, an optionally Substituted (C-C)cycloalkylene, optionally optionally Substituted (C-C)heteroarylene, or an option Substituted bridged (Cs-Co)cycloalkenylene, optionally Sub ally Substituted (C-Co)heterocyclylene

US 9,365,579 B2 41 42 —(C-C)alkyl, an optionally substituted —(C-C)alkenyl, (R)C(O)R’, an optionally substituted (C-C)alkyl, an an optionally substituted —(C-C)alkynyl, an optionally optionally substituted (C-C)alkenyl, an optionally substi Substituted —(C-C)cycloalkyl, an optionally Substituted tuted (C-C)alkynyl, an optionally Substituted (C-C)cy —(C-Co)heteroaryl, an optionally Substituted —(C-Co) cloalkyl, an optionally Substituted (C-Co)heteroaryl, an heterocyclyl, an optionally substituted —(C-C)aryl, an optionally Substituted (C-C) heterocyclyl, an optionally optionally Substituted —(C-C)alkylene-(C-C)cy Substituted (C-C)aryl, an optionally Substituted —(C-C) cloalkyl, an optionally Substituted —(C-C)alkylene-(C- alkylene-(C-C)cycloalkyl, an optionally substituted Co)aryl, an optionally substituted —(C-C)alkylene-(C- —(C-C)alkylene-(Co-Co.)aryl, an optionally substituted Co)heteroaryl, or an optionally Substituted —(C-C) —(C-C)alkylene-(C-C)heteroaryl, or an optionally sub alkylene-(C-Co)heterocyclyl; 10 stituted —(C-C)alkylene-(C-C)heterocyclyl; wherein in a moiety containing N(R)(R), the nitrogen, wherein in a moiety containing N(R)(R), the nitrogen, R" and R may form a ring such that N(R)(R') rep R" and R may form a ring such that N(R)(R') rep resents an optionally substituted (C-C)heterocyclyl resents an optionally substituted (C-Co)heterocyclyl or an optionally Substituted (C-C) heteroaryl linked or an optionally substituted (C-C) heteroaryl linked through a nitrogen; 15 through a nitrogen; R is a hydrogen, halogen, deuterium, an optionally sub R" and Rare each independently hydrogen, deuterium, an stituted bridged (Cs-C)cycloalkyl group, optionally Substi optionally Substituted (C-Co.)alkyl, an optionally Substi tuted bridged (C-C)heterocyclyl group, optionally substi tuted (C-Co.)alkenyl, an optionally Substituted (C-Co) tuted (C-Cs)alkyl, optionally Substituted (C-Co) alkynyl, an optionally Substituted (C-Co.)alkyl-O-(C- cycloalkyl, optionally substituted (C-C)cycloalkenyl, Co)alkyl, an optionally substituted (C-C)cycloalkyl, an optionally Substituted (C-Co)aryl, optionally Substituted optionally substituted (C-C)aryl, an optionally substituted (C-Co)heteroaryl, optionally Substituted (C-C)heterocy (C-Co)heteroaryl, an optionally Substituted (C-C)het clyl or -J.-L-M-Q; erocyclyl, an optionally substituted —(C-C)alkylene-(C- wherein: Co)cycloalkyl, an optionally Substituted—(C-C)alkylene J is a bond, —C(O)—, optionally substituted (C-C)alky 25 (Co-Co)aryl, an optionally substituted —(C-C)alkylene lene, optionally Substituted (C-C)alkenylene, optionally (C-Co)heteroaryl, or an optionally Substituted —(C-C) Substituted (C-C)alkynylene, optionally substituted (C- alkylene-(C-Co)heterocyclyl; and C)cycloalkylene, optionally Substituted (C-C)heterocy R for each occurrence is independently a bond, an option ally Substituted (C-Co.)alkylene, an optionally Substituted 30 (C-C)alkenylene, an optionally substituted (C-C)alky nylene, an optionally substituted —(C-Co.)alkylene-O- R - N(R)S(O), R or N(R)C(O)N(R) Re . (C-Co.)alkylene group, an optionally substituted (C-Co) L is a bond, an optionally Substituted (C-Cs)alkylene, cycloalkylene, an optionally substituted (C-C)arylene, an optionally substituted bridged (C-C)cycloalkylene, optionally Substituted (C-C)heteroarylene, or an option optionally Substituted (C-C)cycloalkylene, optionally 35 ally substituted (C-C)heterocyclylene. Substituted bridged (Cs-Co)cycloalkenylene, optionally Sub In a sixty-sixth embodiment the invention provides a phar stituted (C-Co)cycloalkenylene, optionally Substituted (C- maceutical composition comprising a compound of Formula Co)arylene, optionally substituted (C-Co)heteroarylene, (I) as defined in claim 1 optionally substituted bridged (C-C)heterocyclylene oran optionally substituted (C-Co)heterocyclylene; 40 Formula (I) 'O.UsX 45 IC) Y-R,

50 a pharmaceutically acceptable carrier and excipient and a second therapeutic agent selected from the group consisting of cytokine Suppressive anti-inflammatory drugs, antibodies to orantagonists of other human cytokines or growth factors, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-12, IL-15, 55 IL-16, IL-21, IL-23, interferons, EMAP-II, GM-CSF, FGF, PDGF, CTLA or their ligands including CD154, HUMIRATM, REMICADETM, SIMPONITM (golimumab), CIMZIATM, ACTEMRATM, CDP 571, soluble p55 or p75 TNF receptors, ENBRELTM, Lenercept, TNFC converting 60 enzyme inhibitors, IL-1 inhibitors, Interleukin 11, IL-18 where in all cases, Mis linked to either a carbon or a nitrogen antagonists, IL-12 antagonists, IL-12 antibodies, soluble atom in L.; IL-12 receptors, IL-12 binding proteins, non-depleting anti Q is hydrogen, deuterium, N(R)(R), halogen, —OR", CD4 inhibitors FK506, rapamycin, mycophenolate mofetil, - SR", S(O)R', S(O).R, NO, C(O)OR, CN, leflunomide, NSAIDs, ibuprofen, corticosteroids, phos C(O)N(R)(R), N(R)C(O)R, N(R)C(O)OR, 65 phodiesterase inhibitors, adensosine agonists, antithrombotic N(R)C(O)N(R), C(O R)(R), C(O)R, CF, agents, complement inhibitors, adrenergic agents, IL-1B con OCF, N(R)S(O).R., S(O)N(R)(R), S(O)N Verting enzyme inhibitors, T-cell signalling kinase inhibitors, US 9,365,579 B2 43 44 metalloproteinase inhibitors, Sulfasalazine, 6-mercaptopu triene, cyclosporine, diclofenac sodium/misoprostol, fluoci rines, derivatives p75TNFRIgG, sIL-1RI, SIL-1RII, sIL-6R, nonide, glucosamine Sulfate, gold sodium thiomalate, hydro celecoxib, hydroxychloroquine sulfate, rofecoxib, inflix codone bitartratefapap, risedronate Sodium, Sulfadiazine, imab, naproxen, Valdecoxib, SulfaSalazine, meloxicam, thioguanine, Valdecoxib, alefacept, and efalizumab, acetate, gold sodium thiomalate, aspirin, triamcinolone diclofenac, naproxen, ibuprofen, piroXicam, indomethacin, acetonide, propoxyphene napsylate/apap, folate, nabume COX2 inhibitors, rofecoxib, Valdecoxib, hydroxychloro tone, diclofenac, piroXicam, etodolac, diclofenac sodium, quine, steroids, prednisolone, budenoside, dexamethasone, oxaprozin, oxycodone HCl, hydrocodone bitartratefapap, cytotoxics, azathioprine, cyclophosphamide, mycophenolate diclofenac sodium/misoprostol, fentanyl, anakinra, tramadol mofetil, inhibitors of PDE4, purine synthesis inhibitor, sul HCl, Salsalate, Sulindac, cyanocobalamin/fa/pyridoxine, 10 fasalazine, 5-aminosalicylic acid, olsalazine, Imuran R. acetaminophen, alendronate Sodium, morphine Sulfate, CTLA-4-IgG, anti-B7 family antibodies, anti-PD-1 family lidocaine hydrochloride, indomethacin, glucosamine Sulf antibodies, anti-cytokine antibodies, fonotolizumab, anti chondroitin, amitriptyline HCl, Sulfadiazine, oxycodone IFNg antibody, anti-receptor receptor antibodies, anti-IL-6 HCl/acetaminophen, olopatadine HCl misoprostol, naproxen receptor antibody, antibodies to B-cell surface molecules, UP Sodium, omeprazole, cyclophosphamide, rituximab, IL-1 15 394, Rituximab, anti-CD20 antibody and lymphostat-B. TRAP, MRA, CTLA4-IG, IL-18 BP, anti-IL-12, anti-IL15, VX-740, Roflumilast, IC-485, CDC-801, S1P1 agonists, DETAILED DESCRIPTION OF THE INVENTION FTY720, PKC family inhibitors, Ruboxistaurin, AEB-071, Mesopram, methotrexate, leflunomide, corticosteroids, bude Protein kinases are a broad and diverse class, of over 500 noside, dexamethasone, Sulfasalazine, 5-aminosalicylic acid, enzymes, that include oncogenes, growth factors receptors, olsalazine, IL-1B converting enzyme inhibitors, IL-1 ra, T cell signal transduction intermediates, apoptosis related kinases signaling inhibitors, tyrosine kinase inhibitors, 6-mercap and cyclin dependent kinases. They are responsible for the topurines, IL-11, mesalamine, prednisone, azathioprine, mer transfer of a phosphate group to specific tyrosine, serine or captopurine, infliximab, methylprednisolone sodium Succi threonine amino acid residues, and are broadly classified as nate, diphenoxylate/atrop Sulfate, loperamide hydrochloride, 25 tyrosine and serine/threonine kinases as a result of their Sub omeprazole, folate, ciprofloxacin/dextrose-water, hydroc strate specificity. odone, bitartratefapap, tetracycline hydrochloride, fluocino The Jak family kinases (Jak1, Jak2, Jak3 and Tyk2) are nide, metronidazole, thimerosal/boric acid, cholestyramine? cytoplasmic tyrosine kinases that associate with membrane Sucrose, ciprofloxacin hydrochloride, hyoscyamine Sulfate, bound cytokine receptors. Cytokine binding to their receptor meperidine hydrochloride, midazolam hydrochloride, oxyc 30 initiates Jakkinase activation via trans and autophosphoryla odone HCl/acetaminophen, promethazine hydrochloride, tion processes. The activated Jakkinases phosphorylate resi sodium phosphate, sulfamethoxazoleftrimethoprim, polycar dues on the cytokine receptors creating phosphotyrosine bophil, propoxyphene napsylate, hydrocortisone, multivita binding sites for SH2 domain containing proteins such as mins, balsalazide disodium, codeine phosphatefapap, Signal Transduction Activators of Transcript (STAT) factors colesevelam HCl, cyanocobalamin, folic acid, levofloxacin, 35 and other signal regulators transduction Such as SOCS pro natalizumab, interferon-gamma, methylprednisolone, aza teins and SHIP phosphatases. Activation of STAT factors via thioprine, cyclophosphamide, cyclosporine, methotrexate, this process leads to their dimerization, nuclear translocation 4-aminopyridine, tizanidine, interferon-B1a, AVONEXR, and new mRNA transcription resulting in expression of interferon-B1b, BETASERONR, interferon C-n3, interferon immunocyte proliferation and Survival factors as well as addi C., interferon B1A-IF, Peginterferon C. 2b. Copolymer 1, 40 tional cytokines, chemokines and molecules that facilitate COPAXONE(R), hyperbaric oxygen, intravenous immunoglo cellular trafficking (see Journal of Immunology, 2007, 178, p. bulin, cladribine, cyclosporine, FK506, mycophenolate 2623). Jakkinases transduce signals for many different cytok mofetil, leflunomide, NSAIDs, corticosteroids, predniso ine families and hence potentially play roles in diseases with lone, phosphodiesterase inhibitors, adensosine agonists, anti widely different pathologies including but not limited to the thrombotic agents, complement inhibitors, adrenergic agents, 45 following examples. Both Jak1 and Jak3 control signaling of antiinflammatory cytokines, interferon-?3. IFNB1a, IFNB1b, the so-called common gamma chain cytokines (IL2, IL4, IL7. copaxone, corticosteroids, caspase inhibitors, inhibitors of IL9, IL 15 and IL21), hence simultaneous inhibition of either caspase-1, antibodies to CD40 ligand and CD80, alemtu Jak1 or Jak3 could be predicted to impact Th1 mediated Zumab, dronabinol, daclizumab, mitoxantrone, Xaliproden diseases such as rheumatoid arthritis viablockade of IL2, IL7 hydrochloride, fampridine, glatiramer acetate, natalizumab, 50 and IL15 signaling. On the other hand, IL2 signaling has sinnabidol, C-immunokine NNSO3, ABR-215062, recently been shown to be essential for development and AnergiX.MS, chemokine receptor antagonists, BBR-2778, homeostasis of T-regulatory cells (MalekT Ret al., Immunity, calagualine, CPI-1189, liposome encapsulated mitoxantrone, 2002, 17(2), p. 167-78). Thus, based on genetic data, block THC.CBD, cannabinoid agonists, MBP-8298, mesopram, ade of IL2 signaling alone is predicted to result in autoimmu MNA-715, anti-IL-6 receptor antibody, neurovax, pirfeni 55 nity (Yamanouchi Jet al., Nat Genet., 2007, 39(3), p. 329-37, done allotrap 1258 (RDP-1258), STNF-R1, talampanel, teri and Willerford D M et al., Immunity, 1995, 3(4), p. 521-30). flunomide, TGF-beta2, tiplimotide, VLA-4 antagonists, Th2 mediated diseases such as asthma oratopic dermatitis via interferon gamma antagonists, IL-4 agonists, diclofenac, IL4 and IL9 signaling blockade. Jak1 and Tyk2 mediate sig misoprostol, naproxen, meloxicam, indomethacin, naling of IL13 (see Int. Immunity, 2000, 12, p. 1499). Hence, diclofenac, methotrexate, azathioprine, minocyclin, pred 60 blockade of these may also be predicted to have a therapeutic nisone, etanercept, rofecoxib, SulfaSalazine, naproxen, effect in asthma. These two kinases are also thought to medi leflunomide, methylprednisolone acetate, indomethacin, ate Type I interferon signaling; their blockade could therefore hydroxychloroquine Sulfate, prednisone, Sulindac, be predicted to reduce the severity of systemic lupus erythe betamethasone diprop augmented, infliximab, methotrexate, matosus (SLE). Tyk2 and Jak2 mediate signaling of IL12 and folate, triamcinolone acetonide, diclofenac, dimethylsulfox 65 IL23. In fact, blockade of these cytokines using monoclonal ide, piroXicam, diclofenac sodium, ketoprofen, meloxicam, antibodies has been effective in treating psoriasis. Therefore methylprednisolone, nabumetone, tolmetin Sodium, calcipo blockade of this pathway using inhibitors of these kinases US 9,365,579 B2 45 46 could be predicted to be effective in psoriasis as well. In etal muscle (Mischak, H. et al., FEBS Lett., 1993,326, p. 51), Summary, this invention describes Small-molecule com with Some expression reported in mast cells (Liu, Y. et al., J. pounds that inhibit, regulate and/or modulate Jak family Leukoc. Biol., 2001, 69, p. 831) and endothelial cells (Mattila, kinase activity that is pivotal to several mechanisms thought P. et al., Life Sci., 1994, 55, p. 1253). critical to the progression of autoimmune diseases including, Upon T cell activation, a Supramolecular activation com but not limited to, rheumatoid arthritis (RA), systemic lupus plex (SMAC) forms at the site of contact between the T cell erythematosus (SLE), multiple sclerosis (MS), Crohn's dis and the antigen presenting cell (APC). PKCtheta is the only ease, psoriasis and asthma. PKC isoform found to localize at the SMAC (Monks, C. et al., Several pathologically significant cytokines signal via Nature, 1997, 385, 83), placing it in proximity with other Jak1 alone (Guschin D, et al., EMBO.J. 1995 April 3: 14(7): 10 1421-9; Parganas E, et al., Cell. 1998 May 1; 93(3):385-95; signaling enzymes that mediate T cell activation processes. Rodig S.J., et al., Cell. 1998 May 1:93(3):373-83). Blockade In another study (Baier-Bitterlich, G. et al., Mol. Cell. of one of these, IL6, using an IL6R neutralizing antibody, has Biol., 1996, 16, 842) the role of PKCtheta in the activation of been shown to significantly improve disease scores in human AP-1, a transcription factor important in the activation of the rheumatoid arthritis patients (Nishimoto N. et al., Ann Rheum 15 IL-2 gene, was confirmed. In unstimulated T cells, constitu Dis., 2007, 66(9), p. 1162-7). Similarly, blockaded of GCSF tively active PKCtheta stimulated AP-1 activity while in cells signaling, which is also mediated by Jak1 alone, using neu with dominant negative PKCtheta, AP-1 activity was not tralizing monoclonal antibodies or target gene deletion pro induced upon activation by PMA. tects mice from experimental arthritis (Lawlor K. E. et al., Other studies showed that PKCtheta, via activation of IKB Proc Natl Acad Sci U.S.A., 2004, 101 (31), p. 11398-403). kinase beta, mediates activation of NF-kB induced by T cell Accordingly, the identification of Small-molecule com receptor/CD28 co-stimulation (N. Coudronniere et al., Proc. pounds that inhibit, regulate and/or modulate the signal trans Nat. Acad. Sci. U.S.A., 2000,97, p. 3394; and Lin, X. et al., duction of kinases, such as Jakl, is a desirable means to Mol. Cell. Biol., 2000, 20, p. 2933). prevent or treat autoimmune diseases or other diseases related Proliferation of peripheral T cells from PKCtheta knockout to abberant Jak1 function. 25 mice, in response to T cell receptor (TCR)/CD28 stimulation Jak2 is also activated in a wide variety of human cancers was greatly diminished compared to T cells from wild type Such as prostate, colon, ovarian and breast cancers, mela mice. In addition, the amount of IL-2 released from the T cells noma, leukemia and other haematopoietic malignancies. In was also greatly reduced (Sun, Z. et al., Nature, 2000, 404, p. addition, Somatic point mutation of the Jak2 gene has been 402). It has also been shown that PKCtheta-deficient mice identified to be highly associated with classic myeloprolif 30 show impaired pulmonary inflammation and airway hyperre erative disorders (MPD) and infrequently in other myeloid disorders. Constitutive activation of Jak2 activity is also sponsiveness (AHR) in a Th2-dependent murine asthma caused by chromosomal translocation in hematopoeitic model, with no defects in viral clearance and Th1-dependent malignancies. It has also been shown that inhibition of the cytotoxic T cell function (Berg-Brown, N. N. et al., J. Exp. Jak/STAT pathway, and in particular inhibition of Jak2 activ 35 Med., 2004, 199, p. 743; Marsland, B.J. et al., J. Exp. Med., ity, results in anti-proliferative and pro-apoptotic effects 2004, 200, p. 181). The impaired Th2 cell response results in largely due to inhibition of phosphorylation of STAT. Further reduced levels of IL-4 and immunoglobulin E (IgE), contrib more, pharmacological modulation or inhibition of Jak2 uting to the AHR and inflammatory pathophysiology. Other activity could effectively block tumor growth and induce wise, the PKCtheta knockout mice seemed normal and fertile. apoptosis by reducing the STAT phosphorylation in cell cul 40 Evidence also exists that PKCtheta participates in the IgE ture and human tumor Xenografts in vivo. Accordingly, the receptor (FceRI)-mediated response of mast cells (Liu, Y. et identification of small-molecule compounds that inhibit, al., J. Leukoc. Biol., 2001, 69, p. 831). In human-cultured regulate and/or modulate the signal transduction of kinases, mast cells (HCMC), it has been demonstrated that PKC particularly Jak2, is desirable as a means to treat or prevent kinase activity rapidly localizes to the membrane following diseases and conditions associated with cancers. 45 FceRI cross-linking (Kimata, M. et al., Biochem. Biophys. Jakkinases also transmit signals regulating essential physi Res. Commun., 1999, 257(3), p. 895). A recent study exam ological processes whose inhibition could be undesirable. For ining in vitro activity of bone marrow mast cells (BMMC) example Jak2 mediates the signaling of Erythropoetin (Epo) derived from wild-type and PKCtheta-deficient mice shows and Granulocyte/Monocyte-Colony Stimulating Factor. Indi that upon FceRI cross linking, BMMCs from PKCtheta-de viduals with genetic, congenital or acquired defects in these 50 signaling pathways can develop potentially life-threatening ficient mice reduced levels of IL-6, tumor necrosis factor complications such as anemia and neutrophil dysfunction. alpha (TNFC) and IL-13 in comparison with BMMCs from Accordingly, one non-limiting aspect of this invention also wild-type mice, Suggesting a potential role for PKCtheta in relates to a method to identify compounds that may have a mast cell cytokine production in addition to T cell activation favorable safety profile as a result of them selectively avoid 55 (Ciarletta, A. B. et al., poster presentation at the 2005 Ameri ing inhibition of Jak2. can Thoracic Society International Conference). The protein kinase C family is a group of serine/threonine The studies cited above and others studies confirm the kinases that comprises twelve related isoenzymes. Its mem critical role of PKCtheta in T cells activation and in mast cell bers are encoded by different genes and are sub-classified (MC) signaling. Thus an inhibitor of PKCtheta would be of according to their requirements for activation. The classical 60 therapeutic benefit in treating immunological disorders and enzymes (cPKC) require diacylglycerol (DAG), phosphati other diseases mediated by the inappropriate activation of T dylserine (PS) and calcium for activation. The novel PKC's cells and MC signaling. (nPKC) require DAG and PS but are calcium independent. Many of the kinases, whether a receptor or non-receptor The atypical PKC's (aPKC) do not require calcium or DAG. tyrosine kinase or a S/Tkinase have been found to be involved PKCtheta is a member of the nPKC sub-family (Baier, G., 65 in cellular signaling pathways involved in numerous patho et al., J. Biol. Chem., 1993, 268, 4997). It has a restricted genic conditions, including immunomodulation, inflamma expression pattern, found predominantly in T cells and skel tion, or proliferative disorders such as cancer. US 9,365,579 B2 47 48 Many autoimmune diseases and disease associated with gardt’s disease, Eales disease, retinopathy, macular degenera chronic inflammation, as well as acute responses, have been tion, restenosis, ischemia/reperfusion injury, ischemic stroke, linked to excessive or unregulated production or activity of vascular occlusion, carotid obstructive disease, ulcerative one or more cytokines. colitis, inflammatory bowel disease, diabetes, diabetes mel The compounds of the invention are also useful in the litus, insulin dependent diabetes mellitus, allergic diseases, treatment of cardiovascular disorders, such as acute myocar dermatitis Scleroderma, graft versus host disease, organ trans dial infarction, acute coronary syndrome, chronic heart fail plant rejection (including but not limited to bone marrow and ure, myocardial infarction, atherosclerosis, viral myocarditis, Solid organ rejection), acute or chronic immune disease asso cardiac allograft rejection, and sepsis-associated cardiac dys ciated with organ transplantation, sarcoidosis, disseminated function. Furthermore, the compounds of the present inven 10 intravascular coagulation, Kawasaki's disease, nephrotic tion are also useful for the treatment of central nervous system syndrome, chronic fatigue syndrome, Wegener's granuloma disorders such as meningococcal meningitis, Alzheimer's tosis, Henoch-Schoenlein purpurea, microscopic vasculitis of disease and Parkinson's disease. the kidneys, chronic active hepatitis, septic shock, toxic shock The compounds of the invention are also useful in the syndrome, sepsis syndrome, cachexia, infectious diseases, treatment of an ocular condition, a cancer, a Solid tumor, a 15 parasitic diseases, acquired immunodeficiency syndrome, sarcoma, fibrosarcoma, osteoma, melanoma, retinoblastoma, acute transverse myelitis, Huntington's chorea, stroke, pri a rhabdomyosarcoma, glioblastoma, neuroblastoma, terato mary biliary cirrhosis, hemolytic anemia, malignancies, carcinoma, hypersensitivity reactions, hyperkinetic move Addison's disease, idiopathic Addison's disease, sporadic, ment disorders, hypersensitivity pneumonitis, hypertension, polyglandular deficiency type I and polyglandular deficiency hypokinetic movement disorders, aordic and peripheral aneu type II, Schmidt’s syndrome, adult (acute) respiratory dis ryisms, hypothalamic-pituitary-adrenal axis evaluation, aor tress syndrome, alopecia, alopecia areata, seronegative arth tic dissection, arterial hypertension, arteriosclerosis, arterio opathy, arthropathy, Reiter's disease, psoriatic arthropathy, venous fistula, ataxia, spinocerebellar degenerations, ulcerative colitic arthropathy, enteropathic synovitis, streptococcal myositis, structural lesions of the cerebellum, chlamydia, yersinia and salmonella associated arthropathy, Subacute Sclerosing panencephalitis, Syncope, syphilis of the 25 atheromatous disease/arteriosclerosis, atopicallergy, autoim cardiovascular system, systemic anaphalaxis, systemic mune bullous disease, pemphigus Vulgaris, pemphigus folia inflammatory response syndrome, systemic onset juvenile ceus, pemphigoid, linear IgA disease, autoimmune rheumatoid arthritis, T-cell or FAB ALL, Telangiectasia, haemolytic anaemia, Coombs positive haemolytic anaemia, thromboangitis obliterans, transplants, trauma?hemorrhage, acquired pernicious anaemia, juvenile pernicious anaemia, type III hypersensitivity reactions, type IV hypersensitivity, 30 peripheral vascular disorders, peritonitis, pernicious anemia, unstable angina, uremia, urosepsis, urticaria, Valvular heart myalgic encephalitis/Royal Free Disease, chronic mucocuta diseases, varicose veins, vasculitis, venous diseases, venous neous candidiasis, giant cell arteritis, primary sclerosing thrombosis, ventricular fibrillation, viral and fungal infec hepatitis, cryptogenic autoimmune hepatitis, Acquired tions, vital encephalitisfaseptic meningitis, Vital-associated Immunodeficiency Disease Syndrome, Acquired Immunode hemaphagocytic syndrome, Wernicke-Korsakoff syndrome, 35 ficiency Related Diseases, Hepatitis A, Hepatitis B. Hepatitis Wilson's disease, Xenograft rejection of any organ or tissue, C. His bundle arrythmias, HIV infection/HIV neuropathy, heart transplant rejection, hemachromatosis, hemodialysis, common varied immunodeficiency (common variable hemolytic uremic syndrome/thrombolytic thrombocytopenic hypogammaglobulinaemia), dilated cardiomyopathy, female purpura, hemorrhage, idiopathic pulmonary fibrosis, anti infertility, ovarian failure, premature ovarian failure, fibrotic body mediated cytotoxicity, Asthenia, infantile spinal mus 40 lung disease, chronic wound healing, cryptogenic fibrosing cularatrophy, inflammation of the aorta, influenza A, ionizing alveolitis, post-inflammatory interstitial lung disease, inter radiation exposure, iridocyclitis/uveitis/optic neuritis, juve Stitial pneumonitis, pneumocystis carinii pneumonia, pneu nile spinal muscular atrophy, lymphoma, myeloma, leu monia, connective tissue disease associated interstitial lung kaemia, malignant ascites, hematopoietic cancers, a diabetic disease, mixed connective tissue disease, associated lung dis condition Such as insulin-dependent diabetes mellitus glau 45 ease, systemic Sclerosis associated interstitial lung disease, coma, diabetic retinopathy or microangiopathy, sickle cell rheumatoid arthritis associated interstitial lung disease, sys anaemia, chronic inflammation, glomerulonephritis, graft temic lupus erythematosus associated lung disease, dermato rejection, Lyme disease, Von Hippel Lindau disease, pem myositis/polymyositis associatedlung disease. Sjögren's dis phigoid, Paget’s disease, fibrosis, sarcoidosis, cirrhosis, thy ease associated lung disease, ankylosing spondylitis roiditis, hyperviscosity syndrome, Osler-Weber-Rendu dis 50 associated lung disease, vasculitic diffuse lung disease, hae ease, chronic occlusive pulmonary disease, asthma or edema mosiderosis associated lung disease, drug-induced interstitial following burns, trauma, radiation, stroke, hypoxia, ischemia, lung disease, radiation fibrosis, bronchiolitis obliterans, ovarian hyperstimulation syndrome, post perfusion syn chronic eosinophilic pneumonia, lymphocytic infiltrative drome, post pump syndrome, post-MI cardiotomy syndrome, lung disease, postinfectious interstitial lung disease, gouty preeclampsia, menometrorrhagia, endometriosis, pulmonary 55 arthritis, autoimmune hepatitis, type-1 autoimmune hepatitis hypertension, infantile hemangioma, or infection by Herpes (classical autoimmune or lupoid hepatitis), type-2 autoim simplex, Herpes Zoster, human immunodeficiency virus, mune hepatitis (anti-LKM antibody hepatitis), autoimmune parapoxvirus, protozoa or toxoplasmosis, progressive Supra mediated hypoglycaemia, type B insulin resistance with nucleo palsy, primary pulmonary hypertension, radiation acanthosis nigricans, hypoparathyroidism, acute immune therapy, Raynaud's phenomenon, Raynaud's disease, Ref 60 disease associated with organ transplantation, chronic Sum's disease, regular narrow QRS tachycardia, renovascular immune disease associated with organ transplantation, hypertension, restrictive cardiomyopathy, sarcoma, senile osteoarthritis, primary Sclerosing cholangitis, psoriasis type chorea, senile dementia of Lewy body type, shock, skin 1, psoriasis type 2, idiopathic leucopaenia, autoimmune neu allograft, skin changes syndrome, ocular or macular edema, tropaenia, renal disease NOS, glomerulonephritides, micro ocular neovascular disease, Scleritis, radial keratotomy, uvei 65 scopic vaSulitis of the kidneys, Lyme disease, discoid lupus tis, vitritis, myopia, optic pits, chronic retinal detachment, erythematosus, male infertility idiopathic or NOS, sperm post-laser treatment complications, conjunctivitis, Star autoimmunity, multiple Sclerosis (all Subtypes), sympathetic US 9,365,579 B2 49 50 ophthalmia, pulmonary hypertension secondary to connec and PDGF. Compounds of the invention can be combined tive tissue disease, acute and chronic pain (different forms of with antibodies to cell surface molecules such as CD2, CD3, pain), Goodpasture's syndrome, pulmonary manifestation of CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD80 polyarteritis nodosa, acute rheumatic fever, rheumatoid (B7.1), CD86 (B7.2), CD90, CTLA ortheir ligands including spondylitis, Still's disease, systemic Sclerosis, Sjögren's Syn 5 CD154 (gp39 or CD40L). drome, Takayasu's disease/arteritis, autoimmune thrombocy Preferred combinations of therapeutic agents may interfere topaenia, toxicity, transplants, and diseases involving inap at different points in the autoimmune and Subsequent inflam propriate vascularization for example diabetic retinopathy, matory cascade; preferred examples include TNFantagonists retinopathy of prematurity, choroidal neovascularization due like chimeric, humanized or human TNF antibodies, D2E7 to age-related macular degeneration, and infantile hemangio 10 (U.S. Pat. No. 6,090,382, HUMIRATM), CA2 (REMI mas in human beings. In addition, such compounds may be CADETM), SIMPONITM (golimumab), CIMZIATM, useful in the treatment of disorders such as ascites, effusions, ACTEMRATM, CDP 571, and soluble p55 or p75 TNF recep and exudates, including for example macular edema, cerebral tors, derivatives, thereof, (p75TNFR1 gG (ENBRELTM) or edema, acute lung injury, adult respiratory distress syndrome p55TNFR1 gC (Lenercept), and also TNFC. converting (ARDS), proliferative disorders such as restenosis, fibrotic 15 enzyme (TACE) inhibitors; similarly IL-1 inhibitors (Inter disorders such as hepatic cirrhosis and atherosclerosis, leukin-1-converting enzyme inhibitors, IL-1RA etc.) may be mesangial cell proliferative disorders such as diabetic neph effective for the same reason. Other preferred combinations ropathy, malignant nephrosclerosis, thrombotic microangi include Interleukin 11. Yet other preferred combinations are opathy syndromes, and glomerulopathies, myocardial angio the other key players of the autoimmune response which may genesis, coronary and cerebral collaterals, ischemic limb act parallel to, dependent on or in concert with IL-18 func angiogenesis, ischemia/reperfusion injury, peptic ulcer Heli tion; especially preferred are IL-12 antagonists including cobacter related diseases, virally-induced angiogenic disor IL-12 antibodies or soluble IL-12 receptors, or IL-12 binding ders, preeclampsia, menometrorrhagia, cat scratch fever, proteins. It has been shown that IL-12 and IL-18 have over rubeosis, neovascular glaucoma and retinopathies such as lapping but distinct functions and a combination of antago those associated with diabetic retinopathy, retinopathy of pre 25 nists to both may be most effective. Yet another preferred maturity, or age-related macular degeneration. In addition, combination is non-depleting anti-CD4 inhibitors. Yet other these compounds can be used as active agents against hyper preferred combinations include antagonists of the co-stimu proliferative disorders such as thyroid hyperplasia (especially latory pathway CD80 (B7.1) or CD86 (B7.2) including anti Grave's disease), and cysts (such as hypervascularity of ova bodies, soluble receptors or antagonistic ligands. rian Stroma characteristic of polycystic ovarian syndrome 30 A compound of Formula (I) of the invention may also be (Stein-Leventhal Syndrome) and polycystic kidney disease combined with agents, such as methotrexate, 6-MP. azathio since such diseases require a proliferation of blood vessel prine sulphasalazine, mesalazine, olsalazine chloroquinine/ cells for growth and/or metastasis. hydroxychloroquine, pencillamine, aurothiomalate (intra Compounds of Formula (I) of the invention can be used muscular and oral), azathioprine, cochicine, corticosteroids alone or in combination with an additional agent, e.g., a 35 (oral, inhaled and local injection), beta-2 adrenoreceptorago therapeutic agent, said additional agent being selected by the nists (salbutamol, terbutaline, Salmeteral), Xanthines (theo skilled artisan for its intended purpose. For example, the phylline, aminophylline), cromoglycate, nedocromil, keto additional agent can be a therapeutic agent art-recognized as tifen, ipratropium and oxitropium, cyclosporin, FK506, being useful to treat the disease or condition being treated by rapamycin, mycophenolate mofetil, leflunomide, NSAIDs, the compound of the present invention. The additional agent 40 for example, ibuprofen, corticosteroids Such as prednisolone, also can be an agent that imparts a beneficial attribute to the phosphodiesterase inhibitors, adensosine agonists, anti therapeutic composition e.g., an agent that affects the Viscos thrombotic agents, complement inhibitors, adrenergic agents, ity of the composition. agents which interfere with signalling by proinflammatory It should further be understood that the combinations cytokines such as TNFC. or IL-1 (e.g., NIK, IKK, p38 or MAP which are to be included within this invention are those com 45 kinase inhibitors), IL-1B converting enzyme inhibitors, T-cell binations useful for their intended purpose. The agents set signalling inhibitors such as kinase inhibitors, metallopro forth below are illustrative for purposes and not intended to be teinase inhibitors, SulfaSalazine, 6-mercaptopurines, angio limited. The combinations, which are part of this invention, tensin converting enzyme inhibitors, soluble cytokine recep can be the compounds of the present invention and at least one tors and derivatives thereof (e.g. soluble p55 or p75 TNF additional agent selected from the lists below. The combina 50 receptors and the derivatives p75TNFRIgG (EnbrelTM) and tion can also include more than one additional agent, e.g., two p55TNFRIgG (Lenercept), sIL-1RI, SIL-1RII, SIL-6R), anti or three additional agents if the combination is such that the inflammatory cytokines (e.g. IL-4, IL-10, IL-11, IL-13 and formed composition can perform its intended function. TGFB), celecoxib, folic acid, hydroxychloroquine sulfate, Preferred combinations are non-steroidal anti-inflamma rofecoxib, etanercept, infliximab, naproxen, Valdecoxib, Sul tory drug(s) also referred to as NSAIDS which include drugs 55 fasalazine, methylprednisolone, meloxicam, methylpred like ibuprofen. Other preferred combinations are corticoster nisolone acetate, gold sodium thiomalate, aspirin, triamcino oids including prednisolone; the well known side-effects of lone acetonide, propoxyphene napsylate/apap, folate, steroid use can be reduced or even eliminated by tapering the nabumetone, diclofenac, piroXicam, etodolac, diclofenac steroid dose required when treating patients in combination Sodium, oxaprozin, oxycodone HCl, hydrocodone bitartrate/ with the compounds of this invention. Non-limiting examples 60 apap, diclofenac sodium/misoprostol, fentanyl, anakinra, tra of therapeutic agents for rheumatoid arthritis with which a madol HCl, salsalate, Sulindac, cyanocobalamin/fa/pyridox compound of Formula (I) of the invention can be combined ine, acetaminophen, alendronate Sodium, prednisolone, include the following: cytokine Suppressive anti-inflamma morphine Sulfate, lidocaine hydrochloride, indomethacin, tory drug(s) (CSAIDs); antibodies to or antagonists of other glucosamine Sulf chondroitin, amitriptyline HCl, Sulfadiaz human cytokines or growth factors, for example, TNF, LT, 65 ine, oxycodone HCl/acetaminophen, olopatadine HCl miso IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-12, IL-15, prostol, naproxen Sodium, omeprazole, cyclophosphamide, IL-16, IL-21, IL-23, interferons, EMAP-II, GM-CSF, FGF, rituximab, IL-1 TRAP, MRA, CTLA4-IG, IL-18 BP, anti-IL US 9,365,579 B2 51 52 12, Anti-IL15, BIRB-796, SCIO-469, VX-702, AMG-548, Non-limiting examples of therapeutic agents for multiple VX-740, Roflumilast, IC-485, CDC-801, S1P1 agonists sclerosis with which a compound of Formula (I) can be com (such as FTY720), PKC family inhibitors (such as Ruboxis bined include the following: corticosteroids; prednisolone: taurin or AEB-071) and Mesopram. Preferred combinations methylprednisolone; azathioprine; cyclophosphamide; include methotrexate or leflunomide and in moderate or cyclosporine; methotrexate, 4-aminopyridine; tizanidine; severe rheumatoid arthritis cases, cyclosporine and anti-TNF interferon-?31a (AVONEX(R); Biogen); interferon-B1b (BE antibodies as noted above. TASERONR); Chiron/Berlex); interferon C-n3) (Interferon Non-limiting examples of therapeutic agents for inflam Sciences/Fujimoto), interferon-C. (Alfa Wassermann/J&J), matory bowel disease with which a compound of Formula (I) interferon B1A-IF (Serono/Inhale Therapeutics), Peginter 10 feron C.2b (Enzon/Schering-Plough), Copolymer 1 (Cop-1: of the invention can be combined include the following: bude COPAXONE(R); Teva Pharmaceutical Industries, Inc.); noside; epidermal growth factor, corticosteroids; hyperbaric oxygen; intravenous immunoglobulin; cladribine; cyclosporin, SulfaSalazine; aminosalicylates; 6-mercaptopu antibodies to or antagonists of other human cytokines or rine, azathioprine; metronidazole; lipoxygenase inhibitors; growth factors and their receptors, for example, TNF, LT, mesalamine; olsalazine; balsalazide, antioxidants; throm 15 IL-1, IL-2, IL-6, IL-7, IL-8, IL-12, IL-23, IL-15, IL-16, boxane inhibitors; IL-1 receptor antagonists; anti-IL-1B EMAP-II, GM-CSF, FGF, and PDGF. A compound of For monoclonal antibodies; anti-IL-6 monoclonal antibodies; mula (I) can be combined with antibodies to cell surface growth factors; elastase inhibitors; pyridinyl-imidazole com molecules such as CD2, CD3, CD4, CD8, CD19, CD20, pounds; antibodies to or antagonists of other human cytok CD25, CD28, CD30, CD40, CD45, CD69, CD80, CD86, ines or growth factors, for example, TNF, LT, IL-1, IL-2, IL-6, CD90 or their ligands. A compound of Formula (I) may also IL-7, IL-8, IL-12, IL-15, IL-16, IL-23, EMAP-II, GM-CSF, be combined with agents such as methotrexate, cyclosporine, FGF, and PDGF; cell surface molecules such as CD2, CD3, FK506, rapamycin, mycophenolate mofetil, leflunomide, an CD4, CD8, CD25, CD28, CD30, CD40, CD45, CD69, CD90 S1P1 agonist, NSAIDs, for example, ibuprofen, corticoster or their ligands; methotrexate; cyclosporine; FK506; rapamy oids Such as prednisolone, phosphodiesterase inhibitors, cin; mycophenolate mofetil: leflunomide: NSAIDs, for 25 adensosine agonists, antithrombotic agents, complement example, ibuprofen; corticosteroids such as prednisolone; inhibitors, adrenergic agents, agents which interfere with sig phosphodiesterase inhibitors; adenosine agonists; antithrom nalling by proinflammatory cytokines such as TNFC. or IL-1 botic agents; complement inhibitors; adrenergic agents; (e.g., NIK, IKK, p38 or MAP kinase inhibitors), IL-1B con agents which interfere with signalling by proinflammatory verting enzyme inhibitors, TACE inhibitors, T-cell signaling cytokines such as TNFC. or IL-1 (e.g. NIK, IKK, or MAP 30 inhibitors such as kinase inhibitors, metalloproteinase inhibi kinase inhibitors); IL-13 converting enzyme inhibitors; tors, Sulfasalazine, azathioprine, 6-mercaptopurines, angio TNFC. converting enzyme inhibitors; T-cell signalling inhibi tensin converting enzyme inhibitors, soluble cytokine recep tors such as kinase inhibitors; metalloproteinase inhibitors; tors and derivatives thereof (e.g. soluble p55 or p75 TNF Sulfasalazine; azathioprine; 6-mercaptopurines; angiotensin receptors, sIL-1RI, SIL-1RII, SIL-6R) and antiinflammatory converting enzyme inhibitors; Soluble cytokine receptors and 35 cytokines (e.g. IL-4, IL-10, IL-13 and TGFB). derivatives thereof (e.g. soluble p55 or p75 TNF receptors, Preferred examples of therapeutic agents for multiple scle sIL-1RI, SIL-1RII, SIL-6R) and antiinflammatory cytokines rosis in which a compound of Formula (I) can be combined to (e.g. IL-4, IL-10, IL-11, IL-13 and TGFB). Preferred include interferon-?3, for example, IFNB1a and IFNB1b: examples of therapeutic agents for Crohn's disease with copaxone, corticosteroids, caspase inhibitors, for example which a compound of Formula (I) can be combined include 40 inhibitors of caspase-1, IL-1 inhibitors, TNF inhibitors, and the following: TNF antagonists, for example, anti-TNF anti antibodies to CD40 ligand and CD80. bodies, D2E7 (U.S. Pat. No. 6,090,382, HUMIRATM), CA2 A compound of Formula (I) may also be combined with (REMICADETM), CDP 571, TNFR-Ig constructs, agents, such as alemtuzumab, dronabinol, daclizumab, (p75TNFRIgG (ENBRELTM) and p55TNFRIgG (LENER mitoxantrone, Xaliproden hydrochloride, fampridine, glati CEPTTM) inhibitors and PDE4 inhibitors. A compound of 45 ramer acetate, natalizumab, sinnabidol, C.-immunokine Formula (I) can be combined with corticosteroids, for NNSO3, ABR-215062, AnergiX.MS, chemokine receptor example, budenoside and dexamethasone; Sulfasalazine, antagonists, BBR-2778, calagualine, CPI-1189, LEM (lipo 5-aminosalicylic acid; olsalazine; and agents which interfere some encapsulated mitoxantrone), THC.CBD (cannabinoid with synthesis or action of proinflammatory cytokines Such as agonist), MBP-8298, mesopram (PDE4 inhibitor), MNA IL-1, for example, IL-1B converting enzyme inhibitors and 50 715, anti-IL-6 receptor antibody, neurovax, pirfenidone allot IL-1 rC.: T cell signaling inhibitors, for example, tyrosine rap 1258 (RDP-1258), STNF-R1, talampanel, teriflunomide, kinase inhibitors 6-mercaptopurines; IL-11; mesalamine; TGF-beta2, tiplimotide, VLA-4 antagonists (for example, prednisone; azathioprine; mercaptopurine; infliximab, meth TR-14035, VLA4 Ultrahaler, Antegran-ELAN/Biogen), ylprednisolone sodium Succinate; diphenoxylate/atrop Sul interferon gamma antagonists and IL-4 agonists. fate; loperamide hydrochloride; methotrexate; omeprazole; 55 Non-limiting examples of therapeutic agents for ankylos folate; ciprofloxacin/dextrose-water; hydrocodone bitartrate/ ing spondylitis with which a compound of Formula (I) can be apap; tetracycline hydrochloride; fluocinonide; metronida combined include the following: ibuprofen, diclofenac, miso Zole; thimerosal/boric acid; cholestyramine/Sucrose; ciprof prostol, naproxen, meloxicam, indomethacin, diclofenac, loxacin hydrochloride; hyoscyamine Sulfate; meperidine celecoxib, rofecoxib, SulfaSalazine, methotrexate, azathio hydrochloride; midazolam hydrochloride; oxycodone HC1/ 60 prine, minocyclin, prednisone, and anti-TNF antibodies, acetaminophen; promethazine hydrochloride; sodium phos D2E7 (U.S. Pat. No. 6,090,382: HUMIRATM), CA2 (REMI phate; sulfamethoxazole/trimethoprim; celecoxib; polycar CADETM), CDP 571, TNFR-Ig constructs, (p75TNFRIgG bophil; propoxyphene napsylate; hydrocortisone; (ENBRELTM) and p55TNFRIgG (LENERCEPTTM) multivitamins; balsalazide disodium; codeine phosphate/ Non-limiting examples of therapeutic agents for asthma apap; colesevelam HCl, cyanocobalamin; folic acid; levof 65 with which a compound of Formula (I) can be combined loxacin; methylprednisolone; natalizumab and interferon include the following: albuterol, salmeterol/fluticasone, mon gamma. telukast Sodium, fluticasone propionate, budesonide, pred US 9,365,579 B2 53 54 nisone, salmeterol Xinafoate, levalbuterol HCl, albuterol sul bumetanide, alteplase, enalaprilat, amiodarone hydrochlo fate?ipratropium, prednisolone sodium phosphate, ride, tirofiban HCl m-hydrate, diltiazem hydrochloride, cap triamcinolone acetonide, beclomethasone dipropionate, ipra topril, irbesartan, Valsartan, propranolol hydrochloride, fosi tropium bromide, azithromycin, pirbuterol acetate, predniso nopril sodium, lidocaine hydrochloride, eptifibatide, lone, theophylline anhydrous, methylprednisolone sodium cefazolin Sodium, atropine Sulfate, aminocaproic acid, Succinate, clarithromycin, Zafirlukast, formoterol fumarate, spironolactone, interferon, Sotalol hydrochloride, potassium influenza virus vaccine, amoxicillin trihydrate, flunisolide, chloride, docusate Sodium, dobutamine HCl, alprazolam, allergy injection, cromolyn Sodium, hydrochlo pravastatin Sodium, atorvastatin calcium, midazolam hydro ride, flunisolide/menthol, amoxicillin/clavulanate, levofloxa chloride, meperidine hydrochloride, isosorbide dinitrate, epi cin, inhaler assist device, guaifenesin, dexamethasone 10 Sodium phosphate, moxifloxacin HCl, doxycycline hyclate, nephrine, dopamine hydrochloride, bivalirudin, rosuvastatin, guaifenesin/d-methorphan, p-ephedrine/cod/chlorphenir, eZetimibe/simvastatin, avasimibe, and cariporide. gatifloxacin, hydrochloride, mometasone furoate, Non-limiting examples of therapeutic agents for psoriasis salmeterol Xinafoate, benzonatate, cephalexin, pe/hydroc with which a compound of Formula (I) can be combined odone/chlorphenir, cetirizine HCl/pseudoephed, phenyleph 15 include the following: calcipotriene, clobetasol propionate, rine/cod/promethazine, codeine/promethazine, cefprozil, triamcinolone acetonide, halobetasol propionate, tazarotene, dexamethasone, guaifenesin/pseudoephedrine, chlorphe methotrexate, fluocinonide, betamethasone diprop aug niramine/hydrocodone, nedocromil Sodium, terbutaline Sul mented, fluocinolone acetonide, acitretin, tar shampoo, fate, epinephrine, methylprednisolone, anti-IL-13 antibody, betamethasone Valerate, mometaSone furoate, ketoconazole, and metaproterenol Sulfate. pramoxine/fluocinolone, hydrocortisone Valerate, flurandre Non-limiting examples of therapeutic agents for COPD nolide, urea, betamethasone, clobetasol propionate/emoll. with which a compound of Formula (I) can be combined fluticasone propionate, azithromycin, hydrocortisone, mois include the following: albuterol sulfate/ipratropium, ipratro turizing formula, folic acid, desonide, pimecrolimus, coal tar, pium bromide, salmeterol/fluticasone, albuterol, salmeterol diflorasone diacetate, etanercept folate, lactic acid, methox Xinafoate, fluticasone propionate, prednisone, theophylline 25 salen, he/bismuth Subgal/ZnOX/resor, methylprednisolone anhydrous, methylprednisolone sodium Succinate, mon acetate, prednisone, Sunscreen, halcinonide, salicylic acid, telukast sodium, budesonide, formoterol fumarate, triamci anthralin, clocortolone pivalate, coal extract, coal tar/salicylic nolone acetonide, levofloxacin, guaifenesin, azithromycin, acid, coal tarisalicylic acid/sulfur, desoximetasone, diaz beclomethasone dipropionate, levalbuterol HCl, flunisolide, epam, emollient, fluocinonide/emollient, mineral oil/castor ceftriaxone sodium, amoxicillin trihydrate, gatifloxacin, 30 oil/na lact, mineral oil/peanut oil, petroleum/isopropyl Zafirlukast, amoxicillin/clavulanate, flunisolide?menthol, myristate, psoralen, Salicylic acid, Soap/tribromsalan, thime chlorpheniramine/hydrocodone, metaproterenol sulfate, rosal/boric acid, celecoxib, infliximab, cyclosporine, ale methylprednisolone, mometaSone furoate, p-ephedrine/cod/ facept, efalizumab, tacrolimus, pimecrolimus, PUVA, UVB, chlorphenir, pirbuterol acetate, p-ephedrine/loratadine, terb sulfasalazine, ABT-874 and ustekinamab. utaline sulfate, tiotropium bromide, (R,R)-formoterol, 35 Non-limiting examples of therapeutic agents for psoriatic Tg AAT. cilomilast and roflumilast. arthritis with which a compound of Formula (I) can be com Non-limiting examples of therapeutic agents for HCV with bined include the following: methotrexate, etanercept, rofe which a compound of Formula (I) can be combined include coxib, celecoxib, folic acid, SulfaSalazine, naproxen, lefluno the following: Interferon-alpha-2C, Interferon-alpha-2B, mide, methylprednisolone acetate, indomethacin, Interferon-alpha con1, Interferon-alpha-nl, pegylated inter 40 hydroxychloroquine Sulfate, prednisone, Sulindac, feron-alpha-2C, pegylated interferon-alpha-2B, ribavirin, betamethasone diprop augmented, infliximab, methotrexate, peginterferon alfa-2b--ribavirin, urSodeoxycholic acid, gly folate, triamcinolone acetonide, diclofenac, dimethylsulfox cyrrhizic acid, thymalfasin, Maxamine, VX-497 and any ide, piroXicam, diclofenac sodium, ketoprofen, meloxicam, compounds that are used to treat HCV through intervention methylprednisolone, nabumetone, tolmetin Sodium, calcipo with the following targets: HCV polymerase, HCV protease, 45 triene, cyclosporine, diclofenac sodium/misoprostol, fluoci HCV helicase, and HCV IRES (internal ribosome entry site). nonide, glucosamine Sulfate, gold sodium thiomalate, hydro Non-limiting examples of therapeutic agents for Idiopathic codone bitartrate/apap, ibuprofen, risedronate Sodium, Pulmonary Fibrosis with which a compound of Formula (I) sulfadiazine, thioguanine, Valdecoxib, alefacept, D2E7 (U.S. can be combined include the following: prednisone, azathio Pat. No. 6,090,382, HUMIRATM), and efalizumab. prine, albuterol, colchicine, albuterol Sulfate, digoxin, 50 Non-limiting examples of therapeutic agents for restenosis gamma interferon, methylprednisolone sodium Succinate, with which a compound of Formula (I) can be combined lorazepam, furosemide, lisinopril, nitroglycerin, spironolac include the following: sirolimus, paclitaxel, everolimus, tac tone, cyclophosphamide, ipratropium bromide, actinomycin rolimus, ABT-578, and acetaminophen. d, alteplase, fluticasone propionate, levofloxacin, metaprot Non-limiting examples of therapeutic agents for Sciatica erenol sulfate, morphine Sulfate, oxycodone HCl, potassium 55 with which a compound of Formula (I) can be combined chloride, triamcinolone acetonide, tacrolimus anhydrous, include the following: hydrocodone bitartrate/apap, rofe calcium, interferon-alpha, methotrexate, mycophenolate coxib, cyclobenzaprine HCl, methylprednisolone, naproxen, mofetil and interferon-gamma-1B. ibuprofen, oxycodone HCl/acetaminophen, celecoxib, Valde Non-limiting examples of therapeutic agents for myocar coxib, methylprednisolone acetate, prednisone, codeine dial infarction with which a compound of Formula (I) can be 60 phosphatefapap, tramadol hel/acetaminophen, metaxalone, combined include the following: aspirin, nitroglycerin, meto meloxicam, methocarbamol, lidocaine hydrochloride, prolol tartrate, enoxaparin Sodium, heparin Sodium, clopi diclofenac sodium, gabapentin, dexamethasone, carisopro dogrel bisulfate, carvedilol, atenolol, morphine Sulfate, meto dol, ketorolac tromethamine, indomethacin, acetaminophen, prolol Succinate, warfarin Sodium, lisinopril, isosorbide diazepam, nabumetone, oxycodone HCl, tizanidine HCl, mononitrate, digoxin, furosemide, simvastatin, ramipril, 65 diclofenac sodium/misoprostol, propoxyphene n-pap, asa/ tenecteplase, enalapril maleate, torsemide, retavase, losartan oxycod/oxycodone ter, ibuprofen/hydrocodone bit, tramadol potassium, quinapril hydrochloride/magnesium carbonate, HCl, etodolac, propoxyphene HCl, amitriptyline HCl, cari US 9,365,579 B2 55 56 Soprodol/codeine phos/asa, morphine Sulfate, multivitamins, Certain compounds of Formula (I) and their salts may exist naproxen Sodium, orphenadrine citrate, and temazepam. in more than one crystal form and the present invention Preferred examples of therapeutic agents for SLE (Lupus) includes each crystal form and mixtures thereof with which a compound of Formula (I) can be combined Certain compounds of Formula (I) and their salts may also include the following: NSAIDS, for example, diclofenac, 5 exist in the form of solvates, for example hydrates, and the naproxen, ibuprofen, piroXicam, indomethacin; COX2 present invention includes each Solvate and mixtures thereof. inhibitors, for example, celecoxib, rofecoxib, Valdecoxib; Certain compounds of Formula (I) may contain one or anti-malarials, for example, hydroxychloroquine; steroids, more chiral centers, and exist in different optically active for example, prednisone, prednisolone, budenoside, dexam forms. When compounds of Formula (I) contain one chiral ethasone; cytotoxics, for example, azathioprine, cyclophos 10 center, the compounds exist in two enantiomeric forms and the present invention includes both enantiomers and mixtures phamide, mycophenolate mofetil, methotrexate; inhibitors of of enantiomers, such as racemic mixtures. The enantiomers PDE4 or purine synthesis inhibitor, for example Cellcept(R). A may be resolved by methods known to those skilled in the art, compound of Formula (I) may also be combined with agents for example by formation of diastereoisomeric salts which Such as SulfaSalazine, 5-aminosalicylic acid, olsalazine, Imu 15 may be separated, for example, by crystallization; formation ran Randagents which interfere with synthesis, production or of diastereoisomeric derivatives or complexes which may be action of proinflammatory cytokines Such as IL-1, for separated, for example, by crystallization, gas-liquid or liquid example, caspase inhibitors like IL-1B converting enzyme chromatography; selective reaction of one enantiomer with inhibitors and IL-1 ra. A compound of Formula (I) may also be an enantiomer-specific reagent, for example enzymatic used with T cell signaling inhibitors, for example, tyrosine esterification; or gas-liquid or liquid chromatography in a kinase inhibitors; or molecules that target T cell activation chiral environment, for example on a chiral Support for molecules, for example, CTLA-4-IgG or anti-B7 family anti example silica with a bound chiral ligand or in the presence of bodies, anti-PD-1 family antibodies. A compound of Formula a chiral solvent. It will be appreciated that where the desired (I) can be combined with IL-11 or anti-cytokine antibodies, enantiomer is converted into another chemical entity by one for example, fonotolizumab (anti-IFNg antibody), or anti 25 of the separation procedures described above, a further step is receptor receptor antibodies, for example, anti-IL-6 receptor required to liberate the desired enantiomeric form. Alterna antibody and antibodies to B-cell surface molecules. A com tively, specific enantiomers may be synthesized by asymmet pound of Formula (I) may also be used with LJP 394 (abeti ric synthesis using optically active reagents, Substrates, cata mus), agents that deplete or inactivate B-cells, for example, lysts or solvents, or by converting one enantiomer into the Rituximab (anti-CD20 antibody), lymphostat-B (anti-BlyS 30 other by asymmetric transformation. antibody), TNF antagonists, for example, anti-TNF antibod When a compound of Formula (I) contains more than one chiral center, it may exist in diastereoisomeric forms. The ies, D2E7 (U.S. Pat. No. 6,090,382: HUMIRATM), CA2 diastereoisomeric compounds may be separated by methods (REMICADETM), CDP 571, TNFR-Ig constructs, known to those skilled in the art, for example chromatography (p75TNFRIgG (ENBRELTM) and p55TNFRIgG (LENER 35 or crystallization and the individual enantiomers may be CEPTTM). separated as described above. The present invention includes In this invention, the following definitions are applicable: each diastereoisomer of compounds of Formula (I), and mix A “therapeutically effective amount” is an amount of a tures thereof. compound of Formula (I) or a combination of two or more Certain compounds of Formula (I) may exist in different Such compounds, which inhibits, totally or partially, the pro 40 tautomeric forms or as different geometric isomers, and the gression of the condition oralleviates, at least partially, one or present invention includes each tautomer and/or geometric more symptoms of the condition. A therapeutically effective isomer of compounds of Formula (I) and mixtures thereof. amount can also be an amount which is prophylactically Certain compounds of Formula (I) may exist in different effective. The amount which is therapeutically effective will stable conformational forms which may be separable. Tor depend upon the patient's size and gender, the condition to be 45 sional asymmetry due to restricted rotation about an asym treated, the severity of the condition and the result sought. For metric single bond, for example because of steric hindrance a given patient, a therapeutically effective amount can be or ring strain, may permit separation of different conformers. determined by methods known to those of skill in the art. The present invention includes each conformational isomer “Pharmaceutically acceptable salts' refers to those salts of compounds of Formula (I) and mixtures thereof. which retain the biological effectiveness and properties of the 50 Certain compounds of Formula (I) may exist in Zwitteri free bases and which are obtained by reaction with inorganic onic form and the present invention includes each Zwitteri acids, for example, hydrochloric acid, hydrobromic acid, Sul onic form of compounds of Formula (I) and mixtures thereof. furic acid, nitric acid, and phosphoric acid or organic acids As used herein the term “pro-drug” refers to an agent which Such as Sulfonic acid, carboxylic acid, organic phosphoric is converted into the parent drug in vivo by Some physiologi acid, methanesulfonic acid, ethanesulfonic acid, p-toluene 55 cal chemical process (e.g., a prodrug on being brought to the Sulfonic acid, citric acid, fumaric acid, maleic acid. Succinic physiological pH is converted to the desired drug form). acid, benzoic acid, Salicylic acid, lactic acid, tartaric acid (e.g. Pro-drugs are often useful because, in Some situations, they (+) or (-)-tartaric acid or mixtures thereof), amino acids (e.g. may be easier to administer than the parent drug. They may, (+) or (-)-amino acids or mixtures thereof), and the like. for instance, be bioavailable by oral administration whereas These salts can be prepared by methods known to those 60 the parent drug is not. The pro-drug may also have improved skilled in the art. solubility in pharmacological compositions over the parent Certain compounds of Formula (I) which have acidic sub drug. An example, without limitation, of a pro-drug would be stituents may exist as salts with pharmaceutically acceptable a compound of the present invention wherein it is adminis bases. The present invention includes such salts. Examples of tered as an ester (the “pro-drug') to facilitate transmittal Such salts include Sodium salts, potassium salts, lysine salts 65 across a cell membrane where water solubility is not benefi and arginine salts. These salts may be prepared by methods cial, but then it is metabolically hydrolyzed to the carboxylic known to those skilled in the art. acid once inside the cell where water solubility is beneficial. US 9,365,579 B2 57 58 Pro-drugs have many useful properties. For example, a rolidinyl, quinucludinyl, thiomorpholinyl, tetrahydropyra pro-drug may be more water Soluble than the ultimate drug, nyl, tetrahydrofuranyl, tetrahydroindolyl, thiomorpholinyl thereby facilitating intravenous administration of the drug. A and tropanyl. pro-drug may also have a higher level of oral bioavailability The term "heteroaryl' or "heteroarylene' as used herein, than the ultimate drug. After administration, the prodrug is include aromatic ring systems, including, but not limited to, enzymatically or chemically cleaved to deliver the ultimate monocyclic, bicyclic and tricyclic rings, and have 5 to 12 drug in the blood or tissue. atoms including at least one heteroatom, Such as nitrogen, Exemplary pro-drugs upon cleavage release the corre oxygen, or Sulfur. For purposes of exemplification, which sponding free acid, and Such hydrolyzable ester-forming resi should not be construed as limiting the scope of this inven 10 tion: azaindolyl, benzo(b)thienyl, benzimidazolyl, benzo dues of the compounds of this invention include but are not furanyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, limited to carboxylic acid substituents wherein the free benzoxadiazolyl, furanyl, imidazolyl, imidazopyridinyl, hydrogen is replaced by (C-C)alkyl, (C-C)alkanoy indolyl, indazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, loxymethyl, (C-C)1-(alkanoyloxy)ethyl, 1-methyl-1-(al oxazolyl, purinyl, pyranyl, pyrazinyl, pyrazolyl pyridinyl, kanoyloxy)-ethyl having from 5 to 10 carbon atoms, alkoxy 15 pyrimidinyl, pyrrolyl pyrrolo2,3-dipyrimidinyl, pyrazolo carbonyloxymethyl having from 3 to 6 carbon atoms, 3,4-dipyrimidinyl, quinolinyl, quinazolinyl, triazolyl, thiaz 1-(alkoxycarbonyloxy)ethyl having from 4 to 7 carbon olyl, thiophenyl, tetrazolyl, thiadiazolyl, or thienyl. atoms, 1-methyl-1-(alkoxycarbonyloxy)ethyl having from 5 An "heterocycloalkyl group, as used herein, is a hetero to 8 carbon atoms, N-(alkoxycarbonyl)aminomethyl having cyclic group that is linked to a compound by an aliphatic from 3 to 9 carbonatoms, 1-(N-(alkoxycarbonyl)amino)ethyl group having from one to about eight carbon atoms. For having from 4 to 10 carbon atoms, 3-phthalidyl, 4-crotono example, a heterocycloalkyl group is a morpholinomethyl lactonyl, gamma-butyrolacton-4-yl, di-N,N-(C-C)alky group. lamino(C-C)alkyl (such as f-dimethylaminoethyl), car As used herein, “alkyl”, “alkylene' or notations such as bamoyl-(C-C)alkyl, N,N-di(C-C)-alkylcarbamoyl-(C- “(C-C)” include straight chained or branched hydrocarbons C.)alkyl and piperidino-, pyrrolidino- or morpholino(C-C) 25 which are completely saturated. Examples of alkyls are alkyl. methyl, ethyl, propyl, isopropyl, butyl, pentyl, hexyl and iso Other exemplary pro-drugs release an alcohol of Formula mers thereof. As used herein, “alkenyl', 'alkenylene'. “alky (I) wherein the free hydrogen of the hydroxyl substituent nylene' and “alkynyl means C-C and includes straight (e.g., R' contains hydroxyl) is replaced by (C-C)alkanoy chained or branched hydrocarbons which contain one or more loxymethyl, 1-((C-C)alkanoyloxy)ethyl, 1-methyl-1-((C- 30 units of unsaturation, one or more double bonds for alkenyl C.)alkanoyloxy)ethyl, (C-C)alkoxycarbonyloxymethyl, and one or more triple bonds for alkynyl. N (C-C)alkoxycarbonylamino-methyl, succinoyl, (C- As used herein, “aromatic' groups (or “aryl” or “arylene' C.)alkanoyl, C.-amino(C-C)alkanoyl, arylactyl and C-ami groups) include aromatic carbocyclic ring systems (e.g. phe noacyl, or C.-aminoacyl-C-aminoacyl wherein said C.-ami nyl) and fused polycyclic aromatic ring systems (e.g. naph noacyl moieties are independently any of the naturally 35 thyl, biphenyl and 1,2,3,4-tetrahydronaphthyl). occurring L-amino acids found in proteins, P(O)(OH), As used herein, “cycloalkyl or “cycloalkylene' means —P(O)(O(C-C)alkyl) or glycosyl (the radical resulting C-C monocyclic or multicyclic (e.g., bicyclic, tricyclic, from detachment of the hydroxyl of the hemiacetal of a car spirocyclic, etc.) hydrocarbons that is completely saturated or bohydrate). has one or more unsaturated bonds but does not amount to an As used herein, the term “bridged (C-C) cycloalkyl 40 aromatic group. Examples of a cycloalkyl group are cyclo group” means a saturated or unsaturated, bicyclic or polycy propyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl clic bridged hydrocarbon group having two or three C-Co and cyclohexenyl. cycloalkyl rings. Non bridged cycloalkyls are excluded. As used herein, many moieties or Substituents are termed Bridged cyclic hydrocarbon may include, such as bicyclo as being either “substituted or “optionally substituted”. 2.1.1 hexyl, bicyclo[2.2.1]heptyl, bicyclo[2.2.2]octyl, bicy 45 When a moiety is modified by one of these terms, unless clo3.2.1]octyl, bicyclo4.3.1 decyl, bicyclo[3.3.1nonyl, otherwise noted, it denotes that any portion of the moiety that bornyl, bornenyl, norbornyl, norbornenyl, 6,6-dimethylbicy is known to one skilled in the art as being available for clo3.1.1 heptyl, tricyclobutyl, and adamantyl. substitution can be substituted, which includes one or more As used herein the term “bridged (C-C) heterocyclyl” substituents, where if more than one substituent then each means bicyclic or polycyclic aza-bridged hydrocarbon 50 substituent is independently selected. Such means for substi groups and may include azanorbornyl, quinuclidinyl, isoqui tution are well-known in the art and/or taught by the instant nuclidinyl, tropanyl, azabicyclo3.2.1]octanyl, azabicyclo disclosure. For purposes of exemplification, which should not 2.2.1]heptanyl, 2-azabicyclo3.2.1]octanyl, azabicyclo be construed as limiting the scope of this invention, some 3.2.1]octanyl, azabicyclo3.2.2 nonanyl, azabicyclo[3.3.0 examples of groups that are substituents are: (C-C)alkyl nonanyl, and azabicyclo3.3.1 nonanyl. 55 groups, (C-Cs)alkenyl groups, (C-Cs)alkynyl groups, (C- The term “heterocyclic”, “heterocyclyl or “heterocy Co)cycloalkyl groups, halogen (F, Cl, Br or I), halogenated clylene', as used herein, include non-aromatic, ring systems, (C-Cs)alkyl groups (for example but not limited to CFs). including, but not limited to, monocyclic, bicyclic, tricyclic —O—(C-Cs)alkyl groups, —OH, -S-(C-Cs)alkyl and spirocyclic rings, which can be completely saturated or groups, - SH, -NH(C-Cs)alkyl groups, —N((C-Cs) which can contain one or more units of unsaturation, for the 60 alkyl) groups, NH, —C(O)NH2. —C(O)NH(C-C) avoidance of doubt, the degree of unsaturation does not result alkyl groups, —C(O)N(C-C)alkyl). - NHC(O)H, in an aromatic ring system) and have 5 to 12 atoms including —NHC(O) (C-C)alkyl groups, —NHC(O) (C-C)cy at least one heteroatom, such as nitrogen, oxygen, or Sulfur. cloalkyl groups, N((C-C)alkyl)C(O)H, N((C-C) For purposes of exemplification, which should not be con alkyl)C(O)(C-C)alkyl groups, NHC(O)NH2. —NHC(O) Strued as limiting the scope of this invention, the following are 65 NH(C-C)alkyl groups, N((C-C)alkyl)C(O)NH examples of heterocyclic rings: azepinyl, aZetidinyl, indoli groups, —NHC(O)N(C-Cs)alkyl) groups, —N((C-Cs) nyl, isoindolinyl, morpholinyl, piperazinyl, piperidinyl, pyr alkyl)C(O)N(C-C)alkyl) groups, N(C-Cs)alkyl)C US 9,365,579 B2 59 60 (O)NH((C-C)alkyl), —C(O)H, —C(O)(C-C)alkyl istration, penetrants appropriate to the barrier to be permeated groups, —CN. —NO. —S(O)(C-C)alkyl groups, —S(O) are used in the formulation. Such penetrants are generally (C-Cs)alkyl groups, —S(O)N(C-Cs)alkyl) groups, known in the art. —S(O)NH(C-C)alkyl groups, S(O)NH(C-C)cy For oral administration, the compounds can be formulated cloalkyl groups, —S(O)NH groups, —NHS(O) (C-C) readily by combining the active compounds with pharmaceu alkyl groups, —N((C-Cs)alkyl)S(O) (C-Cs)alkyl groups, tically acceptable carriers well known in the art. Such carriers —(C-Cs)alkyl-O-(C-Cs)alkyl groups, —O—(C-Cs) enable the compounds of the invention to be formulated as alkyl-O (C-C)alkyl groups, —C(O)OH, —C(O)O(C- tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, Cs)alkyl groups, NHOH, NHO(C-C)alkyl groups, —O- Suspensions and the like, for oral ingestion by a patient to be halogenated (C-Cs)alkyl groups (for example but not limited 10 treated. Pharmaceutical preparations for oral use can be to —OCF). —S(O)-halogenated (C-C)alkyl groups (for obtained by combining the active compound with a solid example but not limited to S(O)CF), —S-halogenated (C- excipient, optionally grinding a resulting mixture, and pro Cs)alkyl groups (for example but not limited to SCF), —(C- cessing the mixture of granules, after adding Suitable auxil C) heterocycle (for example but not limited to pyrrolidine, iaries, if desired, to obtain tablets or dragee cores. Suitable tetrahydrofuran, pyran or morpholine), —(C-C) heteroaryl 15 excipients are, in particular, fillers such as Sugars, including (for example but not limited to tetrazole, imidazole, furan, lactose, Sucrose, mannitol, or Sorbitol; cellulose preparations pyrazine or pyrazole), -phenyl, - NHC(O)C)—(C-C)alkyl Such as, for example, maize starch, wheat starch, rice starch, groups, —N(C-C)alkyl)C(O)C)—(C-C)alkyl groups, potato starch, gelatin, gum tragacanth, methyl cellulose, —C(=NH)—(C-C)alkyl groups, C(=NOH)—(C-C) hydroxypropylmethyl-cellulose, sodium carboxymethylcel alkyl groups, or —C(=N-O-(C-C)alkyl)-(C-C)alkyl lulose, and/or polyvinylpyrrolidone (PVP). If desired, disin groups. tegrating agents may be added, such as the cross-linked poly “O'” in Formula (I) represents an aromatic ring. vinyl pyrrolidone, agar, oralginic acid or a salt thereofsuch as One or more compounds of this invention can be adminis Sodium alginate. tered to a human patient by themselves or in pharmaceutical 25 Dragee cores are provided with suitable coatings. For this compositions where they are mixed with biologically suitable purpose, concentrated Sugar Solutions may be used, which carriers or excipient(s)at doses to treat orameliorate a disease may optionally contain gum arabic, talc, polyvinyl pyrroli or condition as described herein. Mixtures of these com done, carbopol gel, polyethylene glycol, and/or titanium pounds can also be administered to the patient as a simple dioxide, lacquer Solutions, and Suitable organic solvents or mixture or in Suitable formulated pharmaceutical composi 30 solvent mixtures. Dyestuffs or pigments may be added to the tions. Atherapeutically effective dose refers to that amount of tablets or dragee coatings for identification or to characterize the compound or compounds sufficient to result in the pre different combinations of active compound doses. vention or attenuation of a disease or condition as described Pharmaceutical preparations that can be used orally herein. Techniques for formulation and administration of the include push-fit capsules made of gelatin, as well as Soft, compounds of the instant application may be found in refer 35 sealed capsules made of gelatin and a plasticizer, Such as ences well known to one of ordinary skill in the art, such as glycerol or Sorbitol. The push-fit capsules can contain the “Remington’s Pharmaceutical Sciences.” Mack Publishing active ingredients in admixture with filler Such as lactose, Co., Easton, Pa., latest edition. binders such as starches, and/or lubricants such as talc or Suitable routes of administration may, for example, magnesium Stearate and, optionally, stabilizers. In soft cap include oral, eyedrop, rectal, transmucosal, topical, or intes 40 Sules, the active compounds may be dissolved or Suspended in tinal administration; parenteral delivery, including intramus Suitable liquids, such as fatty oils, liquid paraffin, or liquid cular, Subcutaneous, intramedullary injections, as well as polyethylene glycols. In addition, stabilizers may be added. intrathecal, direct intraventricular, intravenous, intraperito All formulations for oral administration should be in dosages neal, intranasal, or intraocular injections. Suitable for Such administration. Alternatively, one may administer the compound in a local 45 For buccal administration, the compositions may take the rather than a systemic manner, for example, via injection of form of tablets or lozenges formulated in conventional man the compound directly into an edematous site, often in a depot . or Sustained release formulation. For administration by inhalation, the compounds for use Furthermore, one may administer the drug in a targeted according to the present invention are conveniently delivered drug delivery system, for example, in a liposome coated with 50 in the form of an aerosol spray presentation from pressurized endothelial cell-specific antibody. packs or a nebuliser, with the use of a Suitable propellant, e.g., The pharmaceutical compositions of the present invention dichlorodifluoromethane, trichlorofluoromethane, dichlo may be manufactured in a manner that is itself known, e.g., by rotetrafluoroethane, carbon dioxide or other suitable gas. In means of conventional mixing, dissolving, granulating, dra the case of pressurized aerosol the dosage unit may be deter gee-making, levigating, emulsifying, encapsulating, entrap 55 mined by providing a valve to deliver a metered amount. ping or lyophilizing processes. Capsules and cartridges of e.g. gelatin for use in an inhaler or Pharmaceutical compositions for use in accordance with insufflator may be formulated containing a powder mix of the the present invention thus may be formulated in a conven compound and a suitable powder base such as lactose or tional manner using one or more physiologically acceptable starch. carriers comprising excipients and auxiliaries which facilitate 60 The compounds can be formulated for parenteral adminis processing of the active compounds into preparations which tration by injection, e.g. bolus injection or continuous infu can be used pharmaceutically. Proper formulation is depen Sion. Formulations for injection may be presented in unit dent upon the route of administration chosen. dosage form, e.g. in ampoules or in multi-dose containers, For injection, the agents of the invention may be formu with an added preservative. The compositions may take Such lated in aqueous solutions, preferably in physiologically 65 forms as Suspensions, solutions or emulsions in oily or aque compatible buffers such as Hanks solution, Ringer's solu ous vehicles, and may contain formulatory agents such as tion, or physiological saline buffer. For transmucosal admin Suspending, stabilizing and/or dispersing agents. US 9,365,579 B2 61 62 Pharmaceutical formulations for parenteral administration Such carriers or excipients include but are not limited to include aqueous solutions of the active compounds in water calcium carbonate, calcium phosphate, various Sugars, soluble form. Additionally, Suspensions of the active com starches, cellulose derivatives, gelatin, and polymers such as pounds may be prepared as appropriate oily injection Suspen polyethylene glycols. sions. Suitable lipophilic solvents or vehicles include fatty Many of the compounds of the invention may be provided oils such as sesame oil, or synthetic fatty acid esters. Such as as salts with pharmaceutically compatible counterions. Phar ethyl oleate or triglycerides, or liposomes. Aqueous injection maceutically compatible salts may be formed with many Suspensions may contain Substances which increase the vis acids, including but not limited to hydrochloric, Sulfuric, cosity of the Suspension, such as Sodium carboxymethyl cel acetic, lactic, tartaric, malic, Succinic, etc. Salts tend to be lulose, Sorbitol, or dextran. Optionally, the Suspension may 10 more soluble in aqueous or other protonic Solvents than are also contain Suitable stabilizers or agents which increase the the corresponding free base forms. solubility of the compounds to allow for the preparation of Pharmaceutical compositions suitable for use in the highly concentrated Solutions. present invention include compositions wherein the active Alternatively, the active ingredient may be in powder form ingredients are contained in an effective amount to achieve its for constitution with a Suitable vehicle, e.g., Sterile pyrogen 15 intended purpose. More specifically, a therapeutically effec free water, before use. tive amount means an amount effective to prevent develop The compounds may also be formulated in rectal compo ment of or to alleviate the existing symptoms of the Subject sitions such as Suppositories or retention enemas, e.g., con being treated. Determination of the effective amounts is well taining conventional Suppository bases such as cocoa butter within the capability of those skilled in the art. or other glycerides. For any compound used in a method of the present inven In addition to the formulations described previously, the tion, the therapeutically effective dose can be estimated ini compounds may also be formulated as a depot preparation. tially from cellular assays. For example, a dose can be for Such long acting formulations may be administered by mulated in cellular and animal models to achieve a circulating implantation (for example Subcutaneously or intramuscularly concentration range that includes the ICso as determined in or by intramuscular injection). Thus, for example, the com 25 cellular assays (i.e., the concentration of the test compound pounds may be formulated with suitable polymeric or hydro which achieves a half-maximal inhibition of a given protein phobic materials (for example as an emulsion in an acceptable kinase activity). In some cases it is appropriate to determine oil) or ion exchange resins, or as sparingly soluble deriva the ICs in the presence of 3 to 5% serum albumin since such tives, for example, as a sparingly soluble salt. a determination approximates the binding effects of plasma An example of a pharmaceutical carrier for the hydropho 30 protein on the compound. Such information can be used to bic compounds of the invention is a cosolvent system com more accurately determine useful doses in humans. Further, prising benzyl alcohol, a nonpolar surfactant, a water-mis the most preferred compounds for systemic administration cible organic polymer, and an aqueous phase. The cosolvent effectively inhibit protein kinase signaling in intact cells at system may be the VPD co-solvent system. VPD is a solution levels that are safely achievable in plasma. of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant 35 Atherapeutically effective dose refers to that amount of the polysorbate 80, and 65% w/v polyethylene glycol 300, made compound that results in amelioration of symptoms in a up to volume in absolute ethanol. The VPD co-solvent system patient. Toxicity and therapeutic efficacy of such compounds (VPD:5W) consists of VPD diluted 1:1 with a 5% dextrose in can be determined by standard pharmaceutical procedures in water solution. This co-solvent system dissolves hydrophobic cell cultures or experimental animals, e.g., for determining compounds well, and itself produces low toxicity upon sys 40 the maximum tolerated dose (MTD) and the EDs (effective temic administration. Naturally, the proportions of a co-sol dose for 50% maximal response). The dose ratio between vent system may be varied considerably without destroying toxic and therapeutic effects is the therapeutic index and it can its solubility and toxicity characteristics. Furthermore, the be expressed as the ratio between MTD and EDso. Com identity of the co-solvent components may be varied: for pounds which exhibit high therapeutic indices are preferred. example, other low-toxicity nonpolar Surfactants may be used 45 The data obtained from these cell culture assays and animal instead of polysorbate 80; the fraction size of polyethylene studies can be used informulating a range of dosage for use in glycol may be varied; other biocompatible polymers may humans. The dosage of Such compounds lies preferably replace polyethylene glycol, e.g. polyvinyl pyrrolidone; and within a range of circulating concentrations that include the other Sugars or polysaccharides may substitute for dextrose. EDs with little or no toxicity. The dosage may vary within Alternatively, other delivery systems for hydrophobic 50 this range depending upon the dosage form employed and the pharmaceutical compounds may be employed. Liposomes route of administration utilized. The exact formulation, route and emulsions are well known examples of delivery vehicles of administration and dosage can be chosen by the individual or carriers for hydrophobic drugs. Certain organic solvents physician in view of the patient’s condition (see e.g. Finglet Such as dimethysulfoxide also may be employed, although al., 1975, in “The Pharmacological Basis of Therapeutics'. usually at the cost of greater toxicity. Additionally, the com 55 Ch. 1 p. 1). In the treatment of crises, the administration of an pounds may be delivered using a Sustained-release system, acute bolus or an infusion approaching the MTD may be Such as semipermeable matrices of Solid hydrophobic poly required to obtain a rapid response. mers containing the therapeutic agent. Various Sustained-re Dosage amount and interval may be adjusted individually lease materials have been established and are well known by to provide plasma levels of the active moiety which are suf those skilled in the art. Sustained-release capsules may, 60 ficient to maintain the kinase modulating effects, or minimal depending on their chemical nature, release the compounds effective concentration (MEC). The MEC will vary for each for a few weeks up to over 100 days. Depending on the compound but can be estimated from in vitro data; e.g. the chemical nature and the biological stability of the therapeutic concentration necessary to achieve 50-90% inhibition of pro reagent, additional strategies for protein stabilization may be tein kinase using the assays described herein. Dosages nec employed. 65 essary to achieve the MEC will depend on individual charac The pharmaceutical compositions also may comprise Suit teristics and route of administration. However, HPLC assays able Solid or gel phase carriers or excipients. Examples of or bioassays can be used to determine plasma concentrations. US 9,365,579 B2 63 64 Dosage intervals can also be determined using the MEC In the compositions of the present invention the active value. Compounds should be administered using a regimen compound may, if desired, be associated with other compat which maintains plasma levels above the MEC for 10-90% of ible pharmacologically active ingredients. For example, the the time, preferably between 30-90% and most preferably compounds of this invention can be administered in combi between 50-90% until the desired amelioration of symptoms 5 nation with another therapeutic agent that is known to treat a is achieved. In cases of local administration or selective disease or condition described herein. For example, with one uptake, the effective local concentration of the drug may not or more additional pharmaceutical agents that inhibit or pre be related to plasma concentration. vent the production of VEGF or angiopoietins, attenuate The amount of composition administered will, of course, intracellular responses to VEGF orangiopoietins, block intra be dependent on the subject being treated, on the subjects 10 cellular signal transduction, inhibit vascular hyperpermeabil weight, the severity of the affliction, the manner of adminis ity, reduce inflammation, or inhibit or prevent the formation tration and the judgment of the prescribing physician. ofedema or neovascularization. The compounds of the inven The compositions may, if desired, be presented in a pack or tion can be administered prior to, Subsequent to or simulta dispenser device which may contain one or more unit dosage neously with the additional pharmaceutical agent, whichever forms containing the active ingredient. The pack may for 15 course of administration is appropriate. The additional phar example comprise metal or plastic foil. Such as ablisterpack. maceutical agents include, but are not limited to, anti-edemic The pack or dispenser device may be accompanied by instruc steroids, NSAIDS, ras inhibitors, anti-TNF agents, anti-IL1 tions for administration. Compositions comprising a com agents, antihistamines, PAF-antagonists, COX-1 inhibitors, pound of the invention formulated in a compatible pharma COX-2 inhibitors, NO synthase inhibitors, Akt/PTB inhibi ceutical carrier may also be prepared, placed in an appropriate tors, IGF-1R inhibitors, PKC inhibitors, PI3 kinase inhibi container, and labelled for treatment of an indicated condi tors, calcineurin inhibitors and immunosuppressants. The tion. compounds of the invention and the additional pharmaceuti In some formulations it may be beneficial to use the com cal agents act either additively or synergistically. Thus, the pounds of the present invention in the form of particles of very administration of Such a combination of Substances that Small size, for example as obtained by fluid energy milling. 25 inhibit angiogenesis, vascular hyperpermeability and/or The use of compounds of the present invention in the inhibit the formation of edema can provide greater relief from manufacture of pharmaceutical compositions is illustrated by the deleterious effects of a hyperproliferative disorder, angio the following description. In this description the term “active genesis, vascular hyperpermeability or edema than the compound denotes any compound of the invention but par administration of either substance alone. In the treatment of ticularly any compound which is the final product of one of 30 malignant disorders combinations with antiproliferative or the following Examples. cytotoxic chemotherapies or radiation are included in the a) Capsules scope of the present invention. In the preparation of capsules, 10 parts by weight of active The present invention also comprises the use of a com compound and 240 parts by weight of lactose can be de pound of Formula (I) as a medicament. aggregated and blended. The mixture can be filled into hard 35 A further aspect of the present invention provides the use of gelatin capsules, each capsule containing a unit dose or part of a compound of Formula (I) or a salt thereof in the manufacture a unit dose of active compound. of a medicament for treating vascular hyperpermeability, b) Tablets angiogenesis-dependent disorders, proliferative diseases Tablets can be prepared, for example, from the following and/or disorders of the immune system in mammals, particu ingredients. 40 larly human beings. The present invention also provides a method of treating vascular hyperpermeability, inappropriate neovasculariza Parts by weight tion, proliferative diseases and/or disorders of the immune Active compound 10 system which comprises the administration of a therapeuti Lactose 190 45 cally effective amount of a compound of Formula (I) to a Maize starch 22 mammal, particularly a human being, in need thereof Polyvinylpyrrollidone 10 Abbreviations Magnesium Stearate 3 aa Amino acids AcOH Glacial The active compound, the lactose and some of the starch 50 ATP Adenosine triphosphate can be de-aggregated, blended and the resulting mixture can Boc t-Butoxycarbonyl be granulated with a solution of the polyvinylpyrrolidone in t-BuOH tert-Butanol ethanol. The dry granulate can be blended with the magne BOP-Cl Bis(2-oxo-3-oxazolidinyl)phosphinic chloride sium stearate and the rest of the starch. The mixture is then BSA Bovine serum albumin compressed in a tabletting machine to give tablets each con 55 BuOH Butanol taining a unit dose or a part of a unit dose of active compound. Cbz Carboxybenzyl c) Enteric Coated Tablets CDI 1.1'-Carbonyldiimidazole Tablets can be prepared by the method described in (b) CT Computed tomography above. The tablets can be enteric coated in a conventional CyPFt-Bu 1-Dicyclohexylphosphino-2-di-tert-butylphos manner using a solution of 20% cellulose acetate phthalate 60 phinoethylferrocene and 3% diethyl phthalate in ethanol:dichloromethane (1:1). d Doublet d) Suppositories dba Dibenzylideneacetone In the preparation of Suppositories, for example, 100 parts DCC Dicyclohexylcarbodiimide by weight of active compound can be incorporated in 1300 DCE Dichloroethane parts by weight of triglyceride Suppository base and the mix 65 DCM Dichloromethane (methylene chloride) ture formed into Suppositories each containing a therapeuti dd Doublet of doublets cally effective amount of active ingredient. DIBAL-H Diisobutylaluminium hydride US 9,365,579 B2 65 66 DIEAN,N-Diisopropylethylamine SLMStandard liters per minute DMA Dimethylacetamide t Triplet DMAP N,N-Dimethylaminopyridine t-Tertiary DME 1,2-Dimethoxyethane TBAFTetra-n-Butylammonium fluoride DMEMDulbecco's Modified Eagle Medium TEA Triethylamine DMF N,N-Dimethylformamide tert-Tertiary DMSO Dimethylsulfoxide TFA Trifluoroacetate DNP-HSA Dinitrophenyl-human serum albumin TFAA Trifluoracetic anhydride DTT Dithiothreitol THF Tetrahydrofuran dppf 1,1'-Bis(diphenylphosphino) ferrocene 10 EDC.HCl N-(3-Dimethylaminopropyl)-N'-ethylcarbodiim TIPS Triisopropylsilyl ide hydrochloride TLC Thin layer chromatography EDTA Ethylene diamine tetraacetic acid TMS Trimethylsilyl equiv Equivalent(s) USP United States Pharmacopeia UV Ultraviolet EtNH Diethylamine 15 EtOAc Ethyl acetate wt % Weight percent EtO Diethyl ether Assays EtOH Ethanol In Vitro Jak1 Kinase Activity Measured by Homogenous FBS Fetal bovine serum Time-Resolved Fluorescence (HTRF) FLAG DYKDDDDK peptide sequence Purified Jak1 enzyme (aa845-1142: expressed in SF9 cells g Gram(s) as a GST fusion and purified by glutathione affinity chroma GST Glutathione S-transferase tography) was mixed with 2 uM peptide substrate (biotin h Hour(s) TYR2, Sequence: Biotin-(Ahx)-AEEEYFFLFA-) at HATU O-(7-AZabenzotriazol-1-yl)-N,N,N',N'-tetramethylu varying concentrations of inhibitor in reaction buffer: 50 mM ronium hexafluorophosphate 25 MOPSO pH 6.5, 10 mM MgCl2 mM MnC1 2.5 mM DTT, HEPES N-2-Hydroxyethylpiperazine-N-2-ethanesulfonic 0.01% BSA, 0.1 mM NaVO and 0.001 mM ATP. After acid about 60 min incubation at room temperature, the reaction HOBt Hydroxybenzotriazole was quenched by addition of EDTA (final concentration: 100 HPLC High-pressure liquid chromatography mM) and developed by addition of revelation reagents (final IBCF Isobutylchloroformate 30 approximate concentrations: 30 mM HEPES pH 7.0, 0.06% i.d. Intradermal BSA, 0.006% Tween-20, 0.24 M KF, 80 ng/mL PT66K (eu IFA Incomplete Freunds Adjuvant ropium labeled anti-phosphotyrosine antibody cat IPA. Isopropyl alcohol #61T66KLB Cisbio, Bedford, Mass.) and 3.12 ug/mL SAXL LC/MS Liquid chromatography/mass spectrometry (Phycolink streptavidin-allophycocyanin acceptor, cat LDA Lithium diisopropylamide 35 iPJ52S, Prozyme, San Leandro, Calif.). The developed reac LHMDS Lithium bis(trimethylsilyl)amide tion was incubated in the dark either at about 4°C. for about m Multiplet 14 hor for about 60 min at room temperature, then read via a M Molar time-resolved fluorescence detector (Rubystar, BMG) using a MeCN Acetonitrile 337 nm laser for excitation and emission wavelengths of 620 MeOH Methyl alcohol 40 nm and 665 nm. Within the linear range of the assay, the ratio min Minute(s) of observed signal at 620 nm and 665 nm is directly related to mmol Millimole phosphorylated product and used to calculate the ICs values. MOPS3-(N-morpholino)-2-hydroxypropanesulfonic acid Other kinase assays were performed using a similar proto MOPSO 3-(N-morpholino)-propanesulfonic acid col. Additional purified enzymes Tyk2 (aa 880-1185 with an MS Mass spectrometry 45 N-terminal histidine-tag and C-terminal FLAG tag; purified n- Normal (nonbranched) in-house by immobilized metalion affinity chromatography), N Normal RET (aa 711-1072 with an N-terminal histidine-tag; purified NaOt-Bu Sodium tert-butoxide by immobilized metalion affinity chromatography) and KDR NHOAc Ammonium acetate (aa 792-1354 with an N-terminal histidine-tag; purified in NMMN-Methylmorpholine 50 house by immobilized metal ion affinity and ion-exchange NMP N-methylpyrrolidinone chromatography) were expressed in SF9 cells and Aurora 1/B NMR Nuclear magnetic resonance (aa1-344 with a N-terminal histidine-tag and purified by OD Optical density immobilized metal ion affinity chromatography) was Or Optical rotation expressed in E. coli. Other enzymes used are available from PBS Phosphate buffered saline 55 commercial sources. Enzymes were mixed with biotinylated pH -log H' substrates at varying concentrations of inhibitor in different pNAG Nitrophenyl-N-acetyl-B-D-glucosaminide reaction buffers (see Table 1). After about 60 min incubation ppm Parts per million at room temperature, the reaction was quenched by addition PrOH Propanol of EDTA and developed by addition of revelation reagents psi Pounds per square inch 60 (final approximate concentrations: 30 mM HEPES pH 7.0, rcf Relative centrifugal force 0.06% BSA, 0.006% Tween-20, 0.24M KF, varying amounts RP-HPLC Reverse-phase high-pressure liquid chromatogra of donor europium labeled antibodies and acceptor Streptavi phy din labeled allophycocyanin (SAXL)). The developed reac R. Retention time tions were incubated in the dark at about 4°C. for about 14h rt Room temperature 65 or for about 60 min at room temperature, then read in a S Singlet time-resolved fluorescence detector (Rubystar, BMG SEM 2-(Trimethylsilyl)ethoxymethyl Labtech) as described above. US 9,365,579 B2 67 68 TABLE 1

Specific conditions er 40 enzyme reaction) for the various enzymes are detailed below:

Enzyme ATP DMSO Reaction Assay Conc. Substrate Conc. Conc. Time Detection Enzyme Construct Substrate Buffer Conc. (mM) (%) (min) condition Jak1 aa. 845-1142 Bio in MOPSO 2 uM 5 60 TY

Jak2 Millipore Bioti ill MOPSO 2.5 uM 60 catti 14-640 TY

Jak3 Millipore Bioti ill MOPSO uM 60 catti 14-629 TY

Tyk2 aa880-1185 Bioti ill MOPSO uM 60 TY

Aurora aa1-344 KinASE MOPS uM O.1 60 1 B

KDR aaf89-1354 Biotin HEPES 10 uM O.1 60 TYR2

JNK1 Millipore MOPS 10 uM 60 cathi 14-327

JNK2 Millipore MOPS uM 60 cathi 14-329

RET aaf11-1072 HEPES 10 60

P70S6 Millipore MOPS O.25 uM 60 Kinase catti 14-486

Invitrogen KinASE MOPS 0.7 uM 5 60 catfi PV3879

Syk Millipore Biotin MOPSO 3.8 uM 60 cat #14-314 TYR1

CDK2. Millipore Biotin MOPS 50 uM O.1 60 Cyclin A catti 14-448 MBP US 9,365,579 B2 69 70 Reaction Buffers: (Gibco 15140-122), and 10% heat inactivated FBS (Gibco MOPSO buffer contains: 50 mM MOPSO pH 6.5, 10 mM 10438026). Other materials used in the assay: DMSO (Sigma MgCl2 mMMnCl2.5 mMDTT, 0.01% BSA, and 0.1 mM D2650), 96-well dilution plates (polypropylene) (Corning NaVO 3365), 96-well assay plates (white, 1/2 area, 96 well) (Corn HEPES buffer contains: 50 mM HEPES pH 7.1, 2.5 mM ing 3642), D-PBS (Gibco 14040133), IL-2 (R&D 202-IL-10 DTT, 10 mMMgCl2 mMMnCl, 0.01% BSA, and 0.1 mM (10 ug)), Alphascreen pSTAT5 kit (Perkin Elmer NaVO TGRS5S10K) and Alphascreen protein A kit (Perkin Elmer MOPS buffer contains: 20 mM MOPS pH 7.2, 10 mM 6760617M) MgCl, 5 mM EGTA, 5 mM Beta-phosphoglycerol, 1 mM Methods: NaVO, 0.01% Triton-X-100 and 1 mM DTT 10 T-Blasts were thawed and cultured for about 24 h without Substrates: IL-2 prior to assay. Test compounds or controls are dissolved Biotin-ATF2-peptide Sequence: Biotin-(Ahx)- and serially diluted in 100% DMSO. DMSO stocks are sub AGAGDQTPTPTRFLKRPR-amide sequently diluted 1:50 in cell culture media to create the 4x Biotin-TYR1-peptide sequence: Biotin-(Ahx)-GAEEE compound stocks (containing 2% DMSO). Using a Corning IYAAFFA-COOH 15 white 96 well, /2 area plate, cells are plated at 2x10/10 Biotin-TYR2-peptide sequence: Biotin-(Ahx)-AEEEYF ul/well in 10 uL media followed by addition of 5ull of 4x test FLFA-amide compound in duplicate. Cells are incubated with compound Biotin-MBP-peptide sequence: Biotin-(Ahx)-VHFFKNIVT for about 0.5 h at about 37° C. Next, 5 uL of IL-2 stock is PRTPPPSQGKGAEGQR-amide added at 20 ng/mL final concentration. IL-2 is stored as a 4 Biotin-polyGluTyr peptide was purchased from Cisbio (cat ug/mL stock Solution, as specified by the manufacturer, at #61GTOBLA, Bedford, Mass.) about -20°C. in aliquots and diluted 1:50 with assay media KinEASE S2 and S3 peptides were purchased from Cisbio (to 80 ng/mL) just prior to use. The contents of the wells are (cat #62STOPEB, Bedford, Mass.) mixed by carefully tapping sides of plate(s) several times Detection Reagents: followed by incubation at about 37°C. for about 15 min. The Anti-pATF2-Eu was custom-labeled by Cisbio (Bedford, 25 assay is terminated by adding 5 uL of 5x AlphaScreen lysis Mass.) buffer and shaking on an orbital shaker for about 10 min at Anti-pMBP-Eu was custom-labeled by Cisbio (Bedford, room temperature. AlphaScreen acceptor bead mix is recon Mass.) stituted following Perkin Elmer's protocol. 30 uL/well of PT66K was purchased from Cisbio (cat #61T66KLB, Bed reconstituted AlphaScreen acceptor bead mix was added, cov ford, Mass.) 30 ered with foil then shaken on orbital shaker for about 2 minon SAXL was purchased from Prozyme (cat #PJ25S, San Lean high then about 2 h on low. Donor bead mix is reconstituted dro, Calif.) following PerkinElmer's AlphaScreen protocol; 12 uL?well In Vitro Syk Kinase Activity Measured by Homogenous are added, covered with foil then shaken for about 2 min on Time-Resolved Fluorescence (HTRF) high, and about 2 h on low. Plates are read on an EnVision 1 nM purified full-length Syk enzyme (purchased from 35 reader following Perkin Elmer's AlphaScreen protocol Millipore, Billerica, Mass., Cat #14-314) was mixed with 0.1 instructions. uM peptide substrate (biotin-TYR1, Sequence: Biotin TF-1 IL-6 pSTAT3 Cellular Assay (Ahx)-GAEEEIYAAFFA-COOH) at varying concentrations Materials: of inhibitor in reaction buffer: 50 mM MOPSO pH 6.5, 10 TF-1 cells (ATCC #CRL-2003). Culture medium: DMEM mM MgCl, 2 mM MnC1, 2.5 mM DTT, 0.01% BSA, 0.1 40 medium (Gibco 1 1960-044) with 2 mM L-glutamine (Gibco mM NaVO and 0.01 mM ATP. After about 60 min incuba 25030-081), 10 mM HEPES (Gibco 15630-080), 100 g/mL tion at room temperature, the reaction was quenched by addi Pen/Strep (Gibco 15140-122), 1.5 g/L sodium bicarbonate tion of EDTA (final concentration: 100 mM) and developed (Gibco 25080-094), 1 mM sodium pyruvate (Gibco 11360 by addition of revelation reagents (final approximate concen 070), 10% heat inactivated FBS (Gibco 10437-028), and 2 trations: 30 mM HEPES pH 7.0, 0.06% BSA, 0.006%Tween 45 ng/mL GM-CSF (R&D 215-GM-010). Other materials used 20, 0.24 M KF, 90 ng/mL PT66K (europium labeled anti in this assay: DMSO (Sigma D2650), 96-well dilution plates phosphotyrosine antibody cat #61T66KLB Cisbio, Bedford, (polypropylene) (Corning 3365), 96-well assay plates (white, Mass.) and 0.6 g/mL SAXL (Phycolink streptavidin-allo !/2 area, 96 well) (Corning 3642), D-PBS (Gibco 14040133), phycocyanin acceptor, cat #PJ52S, Prozyme, San Leandro, IL-6 (R&D 206-IL/CF-050 (50 ug)), AlphascreenpSTAT3 kit Calif.). The developed reaction was incubated in the dark 50 (Perkin Elmer TGRS3S10K) and Alphascreen protein A kit either at about 4°C. for about 14 hor for about 60 minatroom (Perkin Elmer 6760617M). temperature, then read via a time-resolved fluorescence Methods: detector (Rubystar, BMG) using a 337 nm laser for excitation Prior to the assay, cells are cultured for about 18 h in the and emission wavelengths of 620 nm and 665 nm. Within the culture medium without GM-CSF. Test compounds or con linear range of the assay, the ratio of observed signal at 620 55 trols are dissolved and serially diluted in 100% DMSO. nm and 665 nm is directly related to phosphorylated product DMSO stocks are subsequently diluted 1:50 in cell culture and used to calculate the ICso values. media to create the 4x compound stocks (containing 2% Human T-Blasts IL-2 pSTAT5 Cellular Assay DMSO). Using a Corning white 96 well, /2 area plate, cells Materials: are plated at 2x107/10 uL/well in 10 uL media followed by Phytohemaglutinin T-blasts were prepared from Leuko 60 addition of 5ul of the 4x test compound stock in duplicate. packs purchased from Biological Specialty Corporation, Col Cells are incubated with compound for about 0.5 h at about mar, Pa. 18915, and cryopreserved in 5% DMSO/media prior 37° C. followed by addition of 5uL of 400 ng/mL IL-6. IL-6 to assay. is stored in 10 ug/mL aliquots using endotoxin free D-PBS For this assay the cells were thawed in assay medium with (0.1% BSA) at about -20° C. Prior to assay IL-6 is diluted to the following composition: RPMI 1640 medium (Gibco 65 400 ng/mL in culture media and applied (5 LL/well) to all 11875093) with 2 mM L-glutamine (Gibco 25030-081), 10 wells, except to negative control wells where 5 u/well of mM HEPES (Gibco 15630-080), 100 g/mL Pen/Strep media is added. The contents of the wells are mixed carefully US 9,365,579 B2 71 72 by tapping the side of the plate several times. Plates are centration of 0.1 lug/mL. 50 L of cells are added to each well incubated at about 37°C. for about 30 min. Cells are lysed by of a 96 well flat bottom plate (50x10 cells/well) and incu adding 5 uL of 5x AlphaScreen cell lysis buffer to all wells, bated overnight at about 37° C. in 5% CO. The next day, shaken for about 10 min at room temperature then assayed. compounds are prepared in 100% DMSO at 10 mM. Each Alternatively, assay plates may be frozen at about-80°C. and compound is then serially diluted 1:4 six times in 100% thawed later at room temperature. Using the pSTAT3 Sure DMSO. Each compound dilution is then diluted 1:20 and then Fire Assay kit (Perkin Elmer #TGRS3S10K) acceptor bead 1:25, both dilutions in Tyrode's buffer. Media is aspirated mix is reconstituted following Perkin Elmer's AlphaScreen from the cell plates and the cells are rinsed twice with 100 uL protocol instructions. 30 uL are added per well then the plate of Tyrode's buffer (prewarmed to about 37° C.). 50 uL of is covered with foil and shaken on an orbital shaker for about 10 compounds diluted in Tyrode's buffer are added to each well 2 min on high, then about 2 h on low at room temperature. and the plates are incubated for about 15 min at about 37°C. Donor bead mix is reconstituted following Perkin Elmer's in 5% CO. 50 uL of 0.2 pg/mL DNP-HSA in Tyrode's buffer AlphaScreen protocol instructions. 12 uL are added per well, is then added to each well and the plates are incubated for then covered with foil and shaken on orbital shaker for about about 30 min at about 37°C. in 5% CO. The final concen 2 min on high, then about 2h on low at about 37°C. Plates are 15 tration of the various components in the incubation mix are read on an EnVision reader following PerkinElmer's AlphaS 0.002-10 uM compounds, 0.1% DMSO, and 0.1 ug/mL DNP creen protocol instructions at room temperature. HSA. As one control, 0.2% DMSO (no compound) in UT7/EPO pSTAT5 Cellular Assay Tyrode's buffer is added to a set of wells to determine maxi Materials: mum stimulated release. As a second control, Tyrode's buffer UT7/EPO cells are passaged with erythropoietin (EPO), without DNP-HSA is added to a set of wells with containing split twice per week and fresh culture medium is thawed and 0.2% DMSO without compounds to determine unstimulated added at time of split. Culture Medium: DMEM medium release. Each condition (compounds and controls) is set up in (Gibco 11960-044) with 2 mM L-glutamine (Gibco 25030 triplicate wells. At the end of the 30 min incubation, 50 uL of 081), 10 mM HEPES (Gibco 15630-080), 100 U/mL Pen/ supernate is transferred to a new 96 well plate. The remaining Strep (Gibco 15140-122), 10% heat inactivated FBS (Gibco 25 supernate in the cell plates is aspirated and replaced with 50 10437-028), EPO (5 uL/mL=7.1 uL of a 7 g/mL stock per uL of 0.1% Triton X-100 in Tyrode's buffer to lyse the cells. mL of medium). Assay media: DMEM, 2 mM L-glutamine, 50 uL of freshly prepared 1.8 mM 4-Nitrophenyl N-acetyl 5% FBS, 10 mM HEPES. Other materials used in the assay: B-D-glucosaminide (pNAG) is then added to each well of DMSO (Sigma D2650), 96-well dilution plates (polypropy Supernate and cell lysate and the plates are incubated for lene) (Corning 3365), 96-well assay plates (white, /2 area, 96 30 about 60 minat about 37° C. in 5% CO. 100 uL of 7.5 mg/mL well) (Corning 3642), D-PBS (Gibco 14040133), IL-2 (R&D sodium bicarbonate is added to each well to stop the reaction. 202-IL-10 (10 ug)), Alphascreen pSTAT5 kit (Perkin Elmer The plates are then read at 405 nm on a Molecular Devices TGRS5S10K) and Alphascreen protein A kit (Perkin Elmer SpectraMax 250 plate reader. 6760617M) Calculation of Results Methods: 35 1) The plate background ODaos obtained from wells contain Culture cells for about 16 h without EPO prior to running ing Tyrode's buffer and pNAG (no supernate or lysate) is assay. Test compounds or controls are dissolved and serially Subtracted from the ODaos reading for each well containing diluted in 100% DMSO. DMSO stocks are subsequently Supernate or lysate. diluted 1:50 in cell culture media to create the 4x compound 2) The release for each well is expressed as the percentage of stocks (containing 2% DMSO). Using a Corning white 96 40 the total release for that well, where the total release is well, /2 area plate, cells are plated at 2x10/10 uL/well in 10 twice the release in the supernate plus the release in the cell uL media followed by addition of 5 uL of 4x test compound lysate. This calculation corrects for variable cell number in stock in duplicate. Cells are incubated with compound for each well. about 0.5 h at about 37° C. After incubation, 5 uL of EPO is 3) The maximum response is the mean response of wells added to afford a final concentration of 1 nM EPO. The 45 containing DNP-HSA but no compound. contents of the wells are mixed by carefully tapping sides of 4) The minimum response is the mean response of wells the plate several times followed by incubation at about 37°C. containing no DNP-HSA and no compound. for about 20 min. 5 uL of 5x AlphaScreen lysis buffer are 5) The response in each compound well is calculated as a added followed by shaking on an orbital shaker for about 10 percentage of the maximum response (expressed as % min at room temperature. 30 uL/well of acceptor beads are 50 control) where the maximum response is 100% and the added after reconstitution following PerkinElmer's AlphaS minimum response is 0%. creen protocol, covered with foil and shaken on orbital shaker 6) A dose response curve is generated for each compound and for about 2 min on high, then about 2 hon low. Donor beads the ICs of the curve is calculated using Prism GraphPad are reconstituted following PerkinElmer's AlphaScreen pro Software and nonlinear least squares regression analysis. tocol instructions followed by addition of 12 uL/well, covered 55 Acute In Vivo Measurement of JAK Inhibition by Com with foil and shaken on an orbital shaker for about 2 min on pounds is Measured Using the: high, about 2 h on low. Plates are read on an EnVision reader Concanavalin a (Con A)-Induced Cytokine Production in following PerkinElmer's AlphaScreen protocol instructions. Lewis Rats Antigen-Induced Degranulation of RBL-2H3 Cells: The test compound is formulated in an inert vehicle (for RBL-2H3 cells are maintained in T75 flasks at about 37°C. 60 example but not limited to 0.5% hydroxypropylmethyl cellu and 5% CO, and passaged every 3-4 days. To harvest cells, lose (Sigma, cat # H3785)/0.02% Tween 80 (Sigma, cat 20 mL of PBS is used to rinse the flask once, and then 3 mL #4780) in water) at the desired concentration to achieve doses of Trypsin-EDTA is added and incubated at about 37°C. for in the range of 0.01-100 mg/kg. Six-week-old male Lewis rats about 2 min. Cells are transferred to a tube with 20 mL (125 g-150 g) (Charles River Laboratories) are dosed with the medium, spun down at 1000 RPM at room temperature for 65 compound orally, at time Zero (0 min). After about 30 min the about 5 min and resuspended at 1x10 cells/mL. Cells are rats are injected intravenously (i.v.) with 10 mg/kg. Con sensitized by adding DNP-specific mouse IgE to a final con canavalin A (ConA, AmershamBioscience, cat #17-0450-01) US 9,365,579 B2 73 74 dissolved in PBS (Invitrogen, cat #14190). About 4h later, the immunization, all rats are exsanguinated by cardiac puncture rats are cardiac bled and their plasma is analyzed for levels of under isofluraneanesthesia, and the left hind paw is collected IL-2 (ELISA kit: R&D Systems cat #R2000) and IFN-y to assess the impact on bone erosion using micro-CT scans (ELISA kit: R&D Systems cat #RIF00). (SCANCO Medical, Southeastern, PA, Model # uCT40) at a Acute InVivo Measurement of Fcy Receptor Signaling Inhi 5 voxel size of 18 lum, a threshold of 400, sigma-gauss 0.8. bition of the Compounds is Measured Using the: Support-gauss 1.0. Bone Volume and density is determined Reverse Passive Arthus Model for a 360 um (200 slice) vertical section encompassing the On day 0 OVA was made up at a concentration of 17.5 tarsal section of the paw. The 360 um section is analyzed from mg/mL, in PBS by rocking gently until a solution was formed. the base of the metatarsals to the top of the tibia, with the 2% Evans Blue solution (Sigma Aldrich, catfi E2129) was 10 then added to double the volume for a final concentration of lower reference point fixed at the tibiotalar junction. Drug 8.75 mg/mL of OVA and 1% Evans Blue dye. Anti-OVA exposure is determined in the plasma using LC/MS. or the: antibody (Abazyme), Stock concentration 10 mg/mL, was Collagen Induced Arthritis (CIA) in a Lewis Rat thawed and a 400 ug/100 uL solution was made with PBS. On day -1 Collagen Type II (CII), soluble from bovine Compounds were made up by adding the vehicle, 0.5% 15 nasal septum (Elastin Products, cathi CN276) was weighed HPMC with 0.02% Tween80, and vortexing for about 15 out for a dose of 600 ug?rat, 0.01 Macetic acid (150 uL HOAc seconds followed by homogenizing for a minimum of about 2 USP grade.J. T. Baker, orderi9522-03, and 250 mL Milli Q min at 28,000 rpm until there was a fine particulate suspen Water) was added for a concentration of 4 mg/mL. The vial sion with no clumps of compound. Rats were weighed and was covered with aluminum foil and placed on a rocker at dosed with compound at a pre-determined t-max based on about 4°C. overnight. On day 0 collagen stock solution was pharmacokinetic studies. Animals were then placed under diluted 1:1 with Incomplete Freunds adjuvant (IFA) (Difco general anesthesia with a 5% isoflourane and oxygen mixture labs, catfi263910) using a glass Hamilton luer lock syringe and shaved. Using a /2 mL insulin Syringe two sites were (SGE Syringe Perfection VWRicatio07230), final concentra injected i.d., 1 site with 100 uL of 400 g/100LL of anti-OVA tion 2 mg/mL. Female Lewis rats (Charles River Laborato antibody, and 1 site with 100 uL of sterile PBS. Each site was 25 ries) acclimated for 7 days at the time of immunization weigh then circled with permanent marker for explant later. Right ing approximately 150 g were anesthetized in an anesthesia after i.d. injections animals were injected with 200 uL of the chamber using isoflurane (5%) and oxygen. Once the rats OVA (10 mg/kg)/Evans Blue mixture i.v., using a /2 mL were completely anesthetized, they were transferred to a nose insulin Syringe. About four hours post injection animals were cone to maintain anesthesia during the injections. Rats were euthanized, bled via cardiac puncture and blood was collected 30 using a plasma separating tube. Blood samples were stored on shaved at the base of the tail, 300 uL of collagen was injected ice until centrifugation (within about 2 h of collection). Each i.d. on the rump of the rat, n=9 per group. 100 uL at three sites injection site was removed with a disposable biopsy punch with a 500LL, leurlock syringe and a 27g needle. IFA control (Acuderm Acu-Punch Disposable 12 mm), cut into four rats are injected in the same manner (n-6). The IFA is a 1:1 pieces and placed in a pre-labeled 2 mLeppendorf tube. One 35 emulsion with the 0.01 Macetic acid. Boost was done on day mL of DMF was added to each biopsy tube and placed in a 6 of the study. Shaving was not done on this day and injections heat block for about 24 h at about 50° C. About 24 h after were done in the same manner as the immunization. The incubation 100 uL of each sample was added to a 96 well flat inflammation appears in both hind paws 10 days after the bottom plate. The samples were read at 620 nm on a plate initial immunization. 10 days post immunization, the com reader using the Softmax Software. Background was removed 40 pound was formulated in an inert vehicle (for example but not by subtracting the OD from the PBS injected site from the OD limited to 0.5% hydroxypropylmethyl cellulose (Sigma, cat it of the anti-OVA injected site for each individual animal. H3785)/0.02% Tween 80 (Sigma, cat #4780) in water) and Plasma samples were spun down in a microcentrifuge for dosed orally once or twice a day for at least 9 days. Baseline about 5 min at 16.1 rcf. 200 uL of plasma was placed in a 1.7 paw Volume was taken on day 7 using a water displacement mL eppendorf tube for drug level measurement and tubes 45 pleythsmograph (Vgo Basile North America Inc. PA 19473, were stored at -80° C. until evaluation. Model #7140). Rats were lightly anesthetized with an inhal Chronic In Vivo Effects of the Compounds on Anc Arthritis ant anesthetic (isoflurane) and both hind paws were dipped Disease Model is Measured Using the: into the plethysmograph and the paw Volume was recorded. Adjuvant Induced Arthritis (AIA) in a Lewis Rat The rats were scored 2 to 3 times a week up to day 18 after Female Lewis rats, (6 weeks of age, 125 g-150 g in weight 50 immunization. On day 18 after immunization, all rats were from Charles River Laboratories) are immunized intrader exsanguinated by cardiac puncture under isoflurane anesthe mally (i.d.) in the right hind-footpad with 100 uL of a sus sia, and the hind paws were collected to assess the impact on pension of mineral oil (Sigma, cat if M5905) and containing bone erosion using micro-CT scans (SCANCO Medical, 200 ug M. tuberculosis, H37RA (Difco, cat #231 141). The Southeastern, PA, Model it LCT 40) at a voxel size of 18 um, inflammation appears in the contra-lateral (left) hind paw 55 a threshold of 400, sigma-gauss 0.8, Support-gauss 1.0. Bone seven days after the initial immunization. Seven days post volume and density was determined for a 360 Lum (200 slice) immunization, the compound is formulated in an inert vehicle Vertical section encompassing the tarsal section of the paw. (for example but not limited to 0.5% hydroxypropylmethyl The 360 um section was analyzed from the base of the meta cellulose (Sigma, cat # H3785)/0.02% Tween 80 (Sigma, cat tarsals to the top of the tibia, with the lower reference point #4780) in water) and dosed orally once or twice a day for at 60 fixed at the tibiotalarjunction. Drug exposure was determined least 10 days. Baseline paw Volume is taken on day 0 using a from plasma using LC/MS. water displacement pleythsmograph (Vgo Basile North The teachings of all references, including journal articles, America Inc. PA 19473, Model #7140). Rats are lightly anes patents and published patent applications, are incorporated thetized with an inhalantanesthetic (isoflurane) and the con herein by reference in their entirety. tra-lateral (left) hind paw is dipped into the plethysmograph 65 The following examples are for illustrative purposes and and the paw volume is recorded. The rats are scored every are not to be construed as limiting the scope of the present other day up to day 17 after immunization. On day 17 after invention. US 9,365,579 B2 75 76 General Synthetic Schemes natively, reaction of pyrrolopyrazines 2 with hydrazine under Compounds of the invention may be prepared using the Buchwald-Hartwig amination conditions as described above synthetic transformations illustrated in Schemes I-XII. Start may give hydrazinylpyrrolopyrazines 4 directly. The forma ing materials are commercially available, may be prepared by tion of hydrazides 5 from hydrazinylpyrrolopyrazines 4 (Scheme I, step d) may be accomplished by a variety of the procedures described herein, by literature procedures, or 5 methods known to one skilled in the art including in situ by procedures that would be well known to one skilled in the conditions such as those described in Example #1, General art of organic chemistry. Methods for preparing pyrrolo2.3- Procedure A, or standard peptide coupling methods such as e1.2.4 triazolo 4.3-alpyrazine compounds of the invention those found in Larock, R. C. referenced above. The are illustrated in Scheme I. In Scheme 1, step a, commercially hydrazides 5 may be cyclized to pyrrolotriazolopyrazines 6 available 2-bromo-5H-pyrrolo2,3-bipyrazine (also called 10 using conditions such as those described in Example #1, 5-bromo-4,7-diazaindole from Ark Pharm, Inc) is protected General Procedure C, or by methods known to one skilled in as a Sulfonamide using conditions such as those described in the art (for example, Bioorganic & Medicinal Chemistry Let Preparation #1 or by methods known to one skilled in the art ters 2007, 17(12),3373-3377 or Journal of Medicinal Chem (for example, Larock, R. C. “Comprehensive Organic Trans istry 1990,33(9), 2326-34). Further functionalization of pyr formations: A Guide to Functional Group Preparations, 2" 15 rolotriazolopyrazines 6 can be performed, if desired, using edition”, 1999, Wiley-VCH or Greene, T. W. and Wuts, P. G. reactions known to one skilled in the art (for example, Larock, M. “Protective Groups in Organic Synthesis, 3" Edition”, R. C. referenced above). For example, formation of , 1999, Wiley-Interscience). Alternatively, protected pyrrolo ureas, Sulfonamides, arylamines, or heteroarylamines can be 2,3-bipyrazine 2 can be prepared from commercially avail prepared from pyrrolotriazolopyrazines 6 containing a pri able 3.5-dibromopyrazin-2-amine via a Sonogashira cross mary or secondary amine (for example, Examples #3 and #4 coupling (Scheme 1, step g) to give alkyne 9 which can be or General Procedures L., M, N or O). Also, deprotection of cyclized (Scheme 1, Steph) to provide pyrrolopyrazines 2 pyrrolotriazolopyrazines 6 can be performed using condi using methods known to one skilled in the art (for example tions such as those described in Greene, T.W. and Wuts, P. G. Preparation #7, Method B). In Scheme I, step b, a substituted M. referenced above or in General Procedures I or J. For hydrazine is introduced by reaction with pyrrolopyrazines 2 25 example, a protecting group Such as a benzyloxycarbonyl under Buchwald-Hartwigamination conditions (for example, group can be removed from a protected amine to yield the Preparation #2 or Advanced Synthesis & Catalysis 2004,346, unprotected amine (for example, Example #2) and the depro 1599-1626) to give pyrrolopyrazines 3. If R" is such that tected compounds 6 may then be reacted further as described pyrrolopyrazines 3 contain a hydrazide (R"= C(O)R") or above. Removal of the Sulfonamide protecting group of pyr hydrazone, the material may be directly cyclized to pyrrolo 30 rolotriazolopyrazines 6 may be accomplished using condi triazolopyrazines 6 using conditions such as those described tions such as those described in Example #1, General Proce in General Procedure C, the initial step of Example #1, Gen dure H, or by methods known to one skilled in the art (for eral Procedure Gorby methods known to one skilled in the art example, the books from Larock, R. C. or Greene, T. W. and (for example, Bioorganic & Medicinal Chemistry Letters Wuts, P. G. M. referenced above) to give pyrrolotriazolopy 2007, 17(12), 3373-3377 or Journal of Medicinal Chemistry 35 razines 7 (Scheme I, step f). Further functionalization of the 1990, 33(9), 2326-34). In some cases, pyrrolotriazolopyra R" group in pyrrolotriazolopyrazines 7 can be performed, if Zines 6 may be reacted in situ to give pyrrolotriazolopyrazines desired, using reactions known to one skilled in the art (for 7 (for example, Example #1 or General Procedures B and E). example, Larock, R. C. referenced above). For example, for Additional reactions may also occur without isolation of ini mation of amides, ureas, Sulfonamides, aryl amines, or het tial pyrrolotriazolopyrazines 6 or 7 as seen in General Proce 40 eroaryl amines can be prepared from pyrrolotriazolopyra dures D and F. If R" is a protecting group, deprotection of Zines 7 with an R" containing a primary or secondary amine compounds 3 to yield hydrazinylpyrrolopyrazines 4 can be (for example, Examples #3 and #4 or General Procedures L. performed using conditions such as those described in Gen M, N or O). Also, deprotection of the R" group in pyrrolot eral Procedure I, General Procedure J, or Greene, T. W. and riazolopyrazines 7 to yield an unprotected compound can be Wuts, P. G. M. “Protective Groups in Organic Synthesis, 3" 45 performed using conditions such as those described in Edition', 1999, Wiley-Interscience. For example, a protect Greene, T. W. and Wuts, P. G. M. referenced above or in ing group Such as a t-butoxycarbonyl group can be removed General Procedures I or J. For example, a protecting group with acid using conditions such as those described in Prepa Such as a benzyloxycarbonyl group can be removed from a ration #3, General Procedure I or by methods known to one protected amine to yield the unprotected amine (for example, skilled in the art (for example, the books from Larock, R. C. 50 Example #2 or General Procedure J) and the deprotected or Greene, T. W. and Wuts, P. G. M. referenced above). Alter compounds 7 may then be reacted further as described above.

Scheme I:

Si|- n Br N Br Br N 2 n n

2 - 9. - || 2 N NH2 N NH2 8 9 US 9,365,579 B2 77 78 -continued y t Br s Br s HN N HN s CO)4NN - O4N - C)4N - C)4N H Y. 29 Y. 29 Y. 29 cav 2\ 2\ R R R 1 2 3 4

R' R' R' 1. Ns Ns O NH NN -( N N.N -( N HN N e- -e- N

N N 2 N N N vY2 -o N vY2 -o o2V o2V R R 7 6 5

The formation of hydrazones 10 from hydrazinylpyrrol- Methods for preparing imidazol-2-alpyrrolo2,3-epyra opyrazines 4 (Scheme II, step a) may be accomplished by a zines compounds of the invention are illustrated in Scheme variety of methods known to one skilled in the art including in III. In step a, a carbamate is introduced by reacting pyrrol situ conditions such as those described in General Procedure opyrazines 2 with tert-butyl carbamate under Buchwald G. The hydrazones 10 may be cyclized to pyrrolotriazolopy- Hartwig amination conditions (for example, Example #8, razines 6 using conditions such as those described in General 35 Step A: Preparation #2, or Advanced Synthesis & Catalysis Procedure G or by methods known to one skilled in the art. 2004, 346, 1599-1626) to give pyrrolopyrazin-2-ylcarbam Further functionalization of pyrrolotriazolopyrazines 6 can ates 11. Deprotection of compounds 11 to yield 2-aminopy be performed, if desired, using reactions known to one skilled rrolopyrazine Sulfonamides 12 can be performed using con in the art (for example, Larock, R. C. referenced above). ditions such as those described in Example #8, Step B; Further functionalization of pyrrolotriazolopyrazines 6 40 General Procedure I, or Greene, T. W. and Wuts, P. G. M. including Sulfonamide hydrolysis to give pyrrolotriazolopy- “Protective Groups in Organic Synthesis, 3" Edition”, 1999, razines 7 (Scheme I, step f) are described above. Wiley-Interscience. The formation of imidazopyrrolopyra zines 13 substituted in the 7-position can be achieved by reacting 2-aminopyrrolopyrazine Sulfonamides 12 with Scheme II: 45 appropriately substituted 2-halomethyl ketones by methods known to one skilled in the art (for example, Journal of t Medicinal Chemistry, 1987, 30(11), 2031-2046 or Example HN N #8, Step C). Further functionalization of imidazopyrrolopy n N razines 13 can be performed, if desired, using reactions 50 known to one skilled in the art (for example, Larock, R. C. 4. N 8. referenced above). For example, formation of amides, ureas, Y2 Sulfonamides, arylamines or heteroarylamines can be pre o2 \ pared from imidazopyrrolopyrazines 13 containing a primary R R or secondary amine (for example, Examples #3 and #4 or l 4 R' 55 General Procedures L., M, N or O). Also, deprotection of NN N imidazopyrrolopyrazines 13 can be performed using condi - tions such as those described in Greene, T.W. and Wuts, P. G. HN N N N M. referenced above or in General Procedures I or J and the n N - - N deprotected compounds 13 may then be reacted further as 60 described above. Removal of the sulfonamide protecting 4. N O s N group of imidazopyrrolopyrazines 13 may be accomplished Y2 Ye using conditions such as those described in Example #8, Step o21\ oav D; General Procedure H, or by methods known to one skilled R R in the art (for example, the books from Larock, R. C. or 10 6 65 Greene, T. W. and Wuts, P. G. M. referenced above) to give imidazopyrrolopyrazines 14. Alternatively, alkylation of pyr rolopyrazin-2-ylcarbamates 11 with appropriately substi US 9,365,579 B2 79 80 tuted 2-halomethyl ketones by methods known to one skilled densation with hydroxyl amine followed by reduction with in the art (for example, Example #9, Step A: Tetrahedron Zinc, providing amines 21 (for example, Example #10, Step Letters, 2006, 47(34), 6113-6115; or Journal of Medicinal C). Alternatively amines 21 can be prepared by reduction of Chemistry, 2005, 48(14), 4535-4546) yields pyrrolopyra aldehydes 19 to the corresponding alcohols (for example, zines 15. Cyclization of pyrrolopyrazines 15 into imidazopy Example #13, Step D), conversion of the alcohol to the chlo rrolopyrazines 16 can be accomplished by methods known to ride and displacement with azide to provide the azides 20 (for one skilled in the art (for example, Example #9, Step B; example, Example #13, Step E). Reduction of the azides European Journal of Medicinal Chemistry, 2001, 36(3), 255 provide amines 21 (for example, Example #13, Step F). Alter 264; or Bioorganic and Medicinal Chemistry Letters, 2007, natively amines 21 can be prepared by conversion of bro 17(5), 1233-1237). Further functionalization of the R" group 10 mides 2 to the corresponding nitriles 25 (for example, Prepa in imidazopyrrolopyrazines 16 can be performed, if desired, ration #28), followed by reduction to amines 21 (for example, using reactions known to one skilled in the art (for example, Preparation #28). Coupling of amines 21 with acids provides Larock, R. C. referenced above). For example, formation of amides 22 (for example. Example #10, Step C). Cyclization amides, ureas, Sulfonamides, aryl amines, or heteroaryl of amides 22 can be accomplished by conversion to the thioa amines can be prepared from imidazopyrrolopyrazines 16 15 mide followed by treatment with an activating agent (such as with an R' group containing a primary or secondary amine a mercury salt, a silver salt or a copper salt) providing the (for example, Examples #3 and #4 or General Procedures L. imidazo 1.5-alpyrrolo2,3-epyrazines 23 (for example, M, N or O). Also, deprotection of the R" group in imidazopy Example #10, Step D). Deprotection of compounds 23 to rrolopyrazines 16 to yield an unprotected compound 17 can yield imidazo 1.5-alpyrrolo2,3-epyrazines 24 can be per be performed using conditions such as those described in formed using conditions such as those described in Greene, T. Greene, T. W. and Wuts, P. G. M. referenced above or in W. and Wuts, P. G. M. “Protective Groups in Organic Syn General Procedures I or J and the deprotected compounds 17 thesis, 3" Edition”, 1999, Wiley-Interscience, General Pro may then be reacted further as described above. Removal of cedure H, or Example #10, Step E. Further functionalization the Sulfonamide protecting group of imidazopyrrolopyra of the R" group in imidazo 1.5-alpyrrolo2,3-epyrazines 23 Zines 16 may be accomplished using conditions such as those 25 or imidazo 1.5-alpyrrolo2,3-epyrazines 24 can be per described in Example #9, Step C; General Procedure H, or by formed, if desired, using reactions known to one skilled in the methods known to one skilled in the art (for example, the art (for example, Larock, R. C. referenced above). For books from Larock, R. C. or Greene, T.W. and Wuts, P. G. M. example, formation of amides, ureas, Sulfonamides, aryl referenced above) to give imidazopyrrolopyrazines 17. amines, or heteroaryl amines can be prepared from com

Scheme III NY R'')- R')- Brn-NsCy On-Nn-NsCy H2N-N-NsCy NaN y NaN y e N Hs O 3 N He- a N -e- S. N -e- s N N \eo N \49 b N Yeo N \49 d N of Y c/Y of Y c/Y 2 11 12 13 14

R'' R' R' O /-( /-( O N N N N N N

N N N N O) 4° H o21\ R 15 16 17

Methods for preparing imidazo 1.5-alpyrrolo2,3-epyra 60 pounds 23 or 24 with an R" group containing a primary or zines compounds of the invention are illustrated in Scheme secondary amine (for example, General Procedures L., M. N IV. In step a, a vinyl group is introduced by reacting pyrrol or O). Also, deprotection of the R" group in compounds 23 or opyrazines 2 with a boronic acid under Suzuki cross coupling 24 to yield an unprotected compound can be performed using conditions (for example, Example #10, Step A). Oxidative conditions such as those described in Greene, T.W. and Wuts, cleavage of the alkenes, 18, provides aldehydes 19 (for 65 P. G. M. referenced above or in General Procedures I or J and example, Example #10, Step B). Conversion to the corre the deprotected compounds may then be reacted further as sponding primary amines can be accomplished by first con described above. US 9,365,579 B2 81 82

Scheme IV

N Ys 20 N Ys 2O N Ys 2O N Ys 2O a V a V a V a V O R O R O R O R 2 18 19 N 2O i

N Šs N N

Cy2 Troy2 N N N M2O2 N \2O2 Oa \R. O4\ R. 25 21

R' O y -rryR' l N N NY. 29 4NYe / Y. % Y. 24 23 22

Methods for preparing 3H-dipyrrolo 1,2-a:2',3'-elpyra above or in General Procedures I or J and the deprotected zines compounds of the invention are illustrated in Scheme V. compounds may then be reacted further as described above. In step a, aldehyde 19 is reacted under Horner-Emmons con ditions to provide C.B-unsaturated ketones 26 (for example, 45 Example #11, Step A). Reduction of the double bond provides Scheme V the saturated ketones 27 (for example, Example #11, Step B). N Cyclization to the tricycles 28 can be accomplished by treat 21 N ment of 27 with an activating agent by methods known to one skilled in the art (for example, Example #11, Step C). Depro- 9 Ory - 4. N 8. tection of compounds 28 to yield 3H-dipyrrolo 1,2-a:2',3'-e \29 pyrazines 29 can be performed using conditions such as those a\ described in Greene, T. W. and Wuts, P. G. M. “Protective O R. Groups in Organic Synthesis, 3rd Edition', 1999, Wiley- 55 19 Interscience; General Procedure H, or Example #11, Step D. Further functionalization of the R' group in 3H-dipyrrolo1, 2-a:2',3'-elpyrazines 28 or 29 can be performed, if desired, O using reactions known to one skilled in the art (for example, N Larock, R. C. referenced above). For example, formation of go R' 21 n amides, ureas, Sulfonamides, aryl amines, or heteroaryl amines can be prepared from compounds 28 or 29 with an R" CO)4. N -- group containing a primary or secondary amine (for example, \29 General Procedures L., M, N or O). Also, deprotection of the zS R" group in compounds 28 or 29 to yield an unprotected 65 O R. compound can be performed using conditions such as those 26 described in Greene, T. W. and Wuts, P. G. M. referenced US 9,365,579 B2 83 84 -continued Procedure BB). Decarboxylation of compounds 32 to give O C.f3-unsaturated ketones 33 is accomplished by standard N methods known to one skilled in the art (for example, General R'' N N 5 Procedure CC). As shown in step c, hydrogenation of C. B 2 unsaturated ketones 33 provides the saturated ketones 34 (for N N O example, General Procedure DD). Reductive amination of Yé ketones 34 with dibenzylamine yields compounds 35 using O R. conditions such as those described in General Procedure EE. 27 The debenzylation of compounds 35 may be accomplished via hydrogenation as described in General Procedure FF to give amines 36. Alternate conditions may be used to access R' R' amines 36 from ketones 34, for example, as described in 15 Larock, R. C. “Comprehensive Organic Transformations: A Guide to Functional Group Preparations, 2" edition”, 1999, N N N N N N Wiley-VCH. Amines 36 may undergo further functionaliza d tion using reactions known to one skilled in the art (for N N s N o 20 example, Larock, R. C. referenced above). For example, for S 2 mation of amides, ureas, Sulfonamides, aryl amines, or het % Y. eroarylamines can be prepared from amines 36 (for example, 29 28 General Procedures L., M. Nor O) to give compounds 37. The 2s ester of compounds 37 may be hydrolyzed under aqueous base oracid conditions to give the desired carboxylic acids 38 Methods for preparing substituted cyclopentyl carboxylic (for example, General Procedure GG or Larock, R. C. refer acids 38 for use in the preparation of compounds of the enced above). If desired, chiral separation of compounds 33, invention are illustrated in Scheme VI. In step a, B-ketoesters 30 34, 35,36, 37, or 38 may be done using methods known to one 31 may be condensed with methyl 4-chloroacetoacetate 30 to skilled in the art such as chiral preparative HPLC (for give cyclic B-ketoester enolate salts 32 (for example, General example, General Procedure II).

Scheme VI

O O O O O O-R 31

-- -- O -e- -e- 8. ONR b O C O 1. C O R R 1. O O 32 33 30

O O-R

O R 34

R"NNH R"NNH H Bn

38 37 36 35 US 9,365,579 B2 85 86 Methods for preparing 4-substituted piperidine-3-car Methods for preparing dihydropyrazolo 4,3-dipyrrolo2, boxylic acid compounds of the invention are illustrated in 3-bipyridine compounds of the invention are illustrated in Scheme VII. In stepa, 4-substituted or unsubstituted nicotinic Scheme VIII. In step a, reaction of aldehyde 42 with a Grig nard reagent provides alcohols 43 using methods known to acids 39 may be fully saturated using methods that are known one skilled in the art (for example. Example #23, Step A). to one skilled in the art (for example, Example #13, Step G). Preparation of ketones 44 (step b) can be accomplished by The resulting piperidine carboxylic acid 40 may be protected treatment of alcohols 43 with an oxidizing agent by methods with a suitable amine protecting group Such as those known to one skilled in the art (for example, Example #23, described in Greene, T. W. and Wuts, P. G. M. “Protective Step B). Alternatively, ketones 44 can be prepared by reaction Groups in Organic Synthesis, 3rd Edition', 1999, Wiley of heteroaryl iodide 45 with an aldehyde (step c) to provide Interscience; Larock, R. C. “Comprehensive Organic Trans 10 alcohols 43 (for example, Example #24, Step A) followed by formations: A Guide to Functional Group Preparations, 2" oxidation as described previously. Preparation of ketones 44 edition”, 1999, Wiley-VCH; or Example #13, Step G to give can be accomplished directly by reaction of heteroaryl iodide piperidine carboxylic acids 41. 45 with an appropriately substituted acid chloride by methods known to one skilled in the art (such as Heterocycles, 2003, Scheme VII 59(1), 369-385). Ketones 44 can then be converted to hydra 15 Zones 46 through reaction with hydrazine using conditions R O R O such as those described in Example #24, Step C. Cyclization of hydrazones 46 to provide dihydropyrazolo 4,3-dipyrrolo N OH OH 2,3-bipyridines 47 can be accomplished via an intramolecu -e- -e- 8. b lar Buchwald-Hartwig cyclization (for example, Example 2 #24, Step B, or Organic Letters, 2008, 10(18), 4109-41 12). N N H Further functionalization of the R" group in dihydropyrazolo 4.3-dipyrrolo2,3-bipyridines 47 can be performed, if 39 40 desired, using reactions known to one skilled in the art (for R O example, Larock, R. C. referenced above). For example, for 25 mation of amides, ureas, Sulfonamides, aryl amines, or het OH eroarylamines can be prepared from compounds 47 with an R" group containing a primary or secondary amine (for example, General Procedures L., M, N or O). Also, deprotec N tion of the R" group in compounds 47 to yield an unprotected R 30 compound can be performed using conditions such as those described in Greene, T. W. and Wuts, P. G. M. referenced 41 above or in General Procedures I or J and the deprotected compounds may then be reacted further as described above. Scheme VIII

C N N 4. N R 45

O HO F.R' N. O R' n

C C C N N r 2 y-to-ry N N N N N R R 42 43 44

NH2

R'' a

C N N 4. N 47 46 US 9,365,579 B2 87 88 Methods for preparing isoxazolo 4,5-dipyrrolo2,3-bpy yield dihydropyrazolo 3,4-dipyrrolo2,3-bipyridines 54 ridine compounds of the invention are described in Scheme using conditions such as those described in Example #42. IX. Ketones 44 can be reacted with hydroxylamine hydro Step D. Further functionalization of the R' group in dihydro chloride (step a) to provide oximes 48 by methods known to pyrazolo 3,4-dipyrrolo2,3-bipyridines 54 can be performed, one skilled in the art (for example. Example #28, Step A). if desired, using reactions known to one skilled in the art (for Cyclization of oximes 48 to provide the desired isoxazolo4. example, Larock, R. C. referenced above). For example, for 5-dipyrrolo2,3-bipyridines 49 (step b) is accomplished mation of amides, ureas, Sulfonamides, aryl amines, or het using methods known to one skilled in the art (for example, eroarylamines can be prepared from compounds 54 with an Example #28, Step B or Tetrahedron, 2007. 63(12), 2695 R" group containing a primary or secondary amine (for 2711). Further functionalization of the R" group in isoxazolo 10 example, General Procedures L., M, N or O). Also, deprotec 4,5-dipyrrolo2,3-bipyridines 49 can be performed, if tion of the R" group in compounds 54 to yield an unprotected desired, using reactions known to one skilled in the art (for compound can be performed using conditions such as those example, Larock, R. C. referenced above). For example, for described in Greene, T. W. and Wuts, P. G. M. referenced mation of amides, ureas, Sulfonamides, aryl amines, or het above or in General Procedures I or J and the deprotected eroarylamines can be prepared from compounds 49 with an 15 compounds may then be reacted further as described above. R" group containing a primary or secondary amine (for example, General Procedures L., M, N or O). Also, deprotec tion of the R" group in compounds 49 to yield an unprotected Scheme X compound can be performed using conditions such as those described in Greene, T. W. and Wuts, P. G. M. referenced R" above or in General Procedures I or J and the deprotected t C compounds may then be reacted further as described above. N N ry -8. - , y - b - Scheme DX 25 % N % N 50 51 t R' R' O R'' Nan-R" N-N? N-N^ W W C C N y - - N Y - N y - N Y - 4. N 8. 4. N b 2 C 2 V V N N 29 N N 29 R R OES OES 35 V V 44 48 R R

52 53

40 R' N-N? / N 45 Y-R Methods for preparing 1,6-dihydropyrazolo 3,4-dipyrrolo 4. N 2,3-bipyridine compounds of the invention are described in Scheme X. Commercially available 4-chloro-1H-pyrrolo-2, 54 3-bipyridine-5-carbaldehyde 50 is reacted with an appropri 50 ately substituted hydrazine or hydrazine hydrochloride Methods for preparing 1,6-dihydrodipyrrolo2,3-b:2',3'-d (Scheme X, step a) to provide the desired 1,6-dihydropyra pyridine compounds of the invention are described in Scheme Zolo 3,4-dipyrrolo2,3-bipyridines 51 by methods known to XI. As shown in step a, heteroaryl chlorides 55 are reacted one skilled in the art (for example, Example #27). Addition with an appropriately Substituted amine using methods such ally, the 1,6-dihydropyrazolo 3,4-dipyrrolo2,3-bipyridines 55 as those described in Larock, R. C. “Comprehensive Organic 51 can be protected as a Sulfonamide (Scheme X, step b) using Transformations: A Guide to Functional Group Preparations, conditions such as those described in Preparation #1 or by 2" edition”, 1999, Wiley-VCH to give esters 56 with con methods known to one skilled in the art (for example, Larock, comitant deprotection. Esters 56 can be converted to the R. C. “Comprehensive Organic Transformations: A Guide to corresponding aldehydes 57 (step b) and then cyclized to give Functional Group Preparations, 2" edition”, 1999, Wiley 60 the desired 1,6-dihydrodipyrrolo2,3-b:2',3'-dpyridines 58 VCH or Greene, T.W. and Wuts, P. G. M. “Protective Groups using methods known to one skilled in the art (for example, in Organic Synthesis, 3" Edition”, 1999, Wiley-Inter Larock, R. C. referenced above). Further functionalization of science). The protected compounds 52 can be iodinated by the R' group in 1,6-dihydrodipyrrolo2,3-b:2',3'-dpyridines methods known to one skilled in the art (for example, 58 can be performed, if desired, using reactions known to one Example #42, Step C). Halogenated tricycles 53 are reacted 65 skilled in the art (for example, Larock, R. C. referenced with an appropriately substituted boronic acid or ester under above). For example, formation of amides, ureas, Sulfona Suzuki cross coupling conditions followed by deprotection to mides, aryl amines, or heteroaryl amines can be prepared US 9,365,579 B2 89 90 from compounds 58 with an R" group containing a primary Groups in Organic Synthesis, 3" Edition”, 1999, Wiley-In or secondary amine (for example, General Procedures L., M. terscience or Example #20, Step D). Pyrrolo2,3-bipyrazines Nor O). Also, deprotection of the R" group in compounds 58 61 can be converted to the corresponding hydroxymethyl to yield an unprotected compound can be performed using derivatives 62 through introduction of an alkene via a Suzuki conditions such as those described in Greene, T.W. and Wuts, 5 P. G. M. referenced above or in General Procedures I or J and cross coupling followed by oxidative cleavage and reduction the deprotected compounds may then be reacted further as of the intermediate aldehyde using methods known to one described above. skilled in the art (for example, Larock, R. C. “Comprehensive Organic Transformations: A Guide to Functional Group 10 Preparations, 2" edition”, 1999, Wiley-VCH or Example Scheme XI #20. Step E). Methanamines 63 can be prepared from O C hydroxylmethyl compounds 62 (stepe) by conversion to the azide (for example. Example #20, Step F) followed by a RN N N 15 Staudinger reduction using methods known to one skilled in -as the art (for example, Larock, R. C. referenced above or 4. N 8. Example #20, Step G). The methanamines 63 can be con V R Verted to an appropriately functionalized amides 64 using 55 methods known to one skilled in the art (for example, R' Example #20, Step H). Amides 64 can be deprotected using O HN1 methods known to one skilled in the art (for example, Greene, R" T. W. and Wuts referenced above or Example #20, Step I) to No N b e provide functionalized pyrrolo2,3-bipyrazines 65 (step g). In Scheme XII, steph, cyclization of amides 65 can be accom 2 25 N N plished by conversion to the thioamide followed by treatment 56 with an activating agent providing the imidazo 1.5-alpyrrolo 2,3-epyrazines 66 (for example, Example #20, Step J). t HN1 R' Alternatively, cyclization of amides 64 can be accomplished using the conditions described above (Scheme XII, step i) (for N example, Example #22, Step B) followed by deprotection of N e imidazo 1.5-alpyrrolo2,3-epyrazines 67 (Scheme XII, step 2 C j) using methods known to one skilled in the art (for example, N N Greene, T. W. and Wuts referenced above or Example #22, 57 Step C). Further functionalization of the R" group in imidazo 1.5-alpyrrolo2,3-epyrazines 66 or 67 can be performed, if Methods for preparing imidazo 1.5-alpyrrolo2,3-epyra desired, using reactions known to one skilled in the art (for Zines 66 of the invention are illustrated in Scheme XII. example, Larock, R. C. referenced above). For example, for 5-Bromo-3-((trimethylsilyl)ethynyl)pyrazine-2-amine 9 can 40 mation of amides, ureas, Sulfonamides, aryl amines, or het be reacted with an appropriately functionalized halide to give eroaryl amines can be prepared from compounds 66 or 67 substituted alkynes 59 (Scheme XII, step a) by methods with an R" group containing a primary or secondary amine known to one skilled in the art (for example, Example #20, (for example, General Procedures L., M. Nor O). Also, depro Step B). Alkynes 59 can be reacted under basic conditions to tection of the R" group in compounds 66 or 67 to yield an give pyrrolo2,3-bipyrazines 60 (as in Example #20, Step C). 45 unprotected compound can be performed using conditions The pyrrolo2,3-bipyrazines 60 can be functionalized with an such as those described in Greene, T. W. and Wuts, P. G. M. appropriate protecting group, Such as (2-(trimethylsilyl) referenced above or in General Procedures I or J and the ethoxy)methyl, by methods known to one skilled in the art deprotected compounds may then be reacted further as (for example, Greene, T. W. and Wuts, P. G. M. “Protective described above.

Scheme XII "Cry N S R B. N. af \ Br N. 4 Brn-Ns s -es -e-C N d ( 3 8. C a Y-R N. NH N1, NH, NYN / 9 59 60 61 US 9,365,579 B2

-continued R ols NH NH2 Ns Ns R' 1. COY-RN1 N COY-RN1 N Nss O NH No No NN a- Ns Cy-R h Cy-R -- N 7 - - N / s N ea N 9. Si Si N H N H ors s 66 65 64 63 N Ns, R' 1. NN OY-R No N / Sis 67

25 General Procedures and Examples The general synthetic schemes that were utilized to con- -continued struct the majority of compounds disclosed in this application R R are described below in Schemes 1-39. These schemes are 30 /N -( /N -( provided for illustrative purposes only and are not to be con- N N N N-N Strued as limiting the scope of the invention. N -e- N N N 35 N N N N \ leO Scheme 1. Formation of a hydrazide from a carboxylic acid o (General Procedure A) R O O 40 Y n R' ulsOH - R'' ulN R H Schene 3. Cyclization of a hydrazide (General Procedure C) 45 O Scheme 2. Formation of a hydrazide from an acid chloride followed by ul cyclization and Sulfonamide hydrolysis R' NH (General Procedure B) HN 2 O 50 N ls N N R' C N \-0e

55 o R O R''

R' us NH (Ns N-N HN N 60 N

Her S. N Sn N N N V O \ O eS e S2W 65 O k R US 9,365,579 B2 93 94 -continued Scheme 4. Cyclization of a hydrazide followed by sulfonamide hydrolysis R R and Boc-deprotection Ns Ns / 5 N -( (General Procedure D) N N N N-N O C) os-N-Sr. s N-NN O HN O2 s 2 N -- R s N 15 o Scheme 6. Cyclization of a hydrazide with loss of Boc-protecting group R followed by Sulfonamide hydrolysis (General Procedure F) R-NH O N Y os-N-NR t N-N( N x O HN N 25 21 N Ho s N o s N R 25'\ 30 R

R-NH N Y 35 N-N( N x N RNNH, N RNNH, N N-N N-N N N y -- Y 40 N N N N N -29 N RNNH,

N-K N 45 N N N N Scheme 7. Formation of a hydrazone followed by cyclization and Sulfonamide hydrolysis 50 O (General Procedure G)

Scheme 5. Cyclization of a hydrazide followed by sulfonamide hydrolysis R' us H (General Procedure E) 55 O

1N - N 60 HN "Ne N Hess s

o 65 R US 9,365,579 B2 95 96 -continued R' R'' Scheme 12. Formation of an amide from a carboxylic acid and an amine Ns Ns / W (General Procedure L) N N N N N N 5 O R O Y -e- y S. N OH N N N N O2 \20i 10 R

Scheme 13. Formation of a urea from an amine and a carbamoyl chloride 15 (General Procedure M) O -R i R' -- HN -- Scheme 8. Hydrolysis of a sulfonamide C R" (General Procedure H) 20 R R" R" O R- N R N. N Rs.1 - \ e-o H O2 i 25 s R R R"

30 Scheme 14. Formation of a Sulfonamide from an amine (General Procedure N) O O Scheme 9. Acidic cleavage of a Boc-protected amine V/ -R \/ (General Procedure I) \/ HN R1n -R 35 R1 No R" O R"

R a ls O -k -e- R YNH

R R 40 Scheme 15. Displacement of an aryl or heteroaryl halide with an amine (General Procedures O and O.1) -R Arn -R

Scheme 10. Deprotection of a Cbz-protected amine 45 A-X + " - (General Procedure J) O R l R 50 Scheme 16. Boc-protection of an amine (General Procedure P) O YNH O R R R - - O -k R 55 R

Scheme 11. Formation of an amide from an activated acid and an amine Scheme 17. Cbz-protection of an amine (General Procedure Q) (General Procedure K) 60 O O i O Y R R -- HN R NH 1 He- s O R X R" R R 65 US 9,365,579 B2

-continued Scheme 18. Reduction of a pyridine (General Procedure R) R (R) (R) N-R XS X 5 Ns -e- M 2 N N-N N N N N N 10 N V O2 s2O\ Scheme 19. Reduction of an ester to an alcohol (General Procedure S) R O us -- 1 No 15 R OR

Scheme 23. Formation of an acid chloride (General Procedure W) 2O O O Scheme 20. Oxidation of an alcohol to an aldehyde (General Procedure T) us e us O R OH R C 1No —- ul R H 25

Scheme 24. Formation of a urea using CDI (General Procedure X) Scheme 21. Formation of a semicarbazide (General Procedure U) O NH2 30 R'' NNH HN N / N N N —- 2 -\ N2- sN R Sa N 35 O 2S leO R'' ls 1. R O V NN N N HN R R R" R NS. R" n N Y N 1. 40 O O NH R' l R n N N1

"Ne R R" N 45 s N V O2 sleO R 50 Scheme 25. Formation of an ester from a carboxylic acid (General Procedure Y) O O Scheme 22. Cyclization of a semicarbazide (General Procedure V) us us t 55 R OH R OR R YaY R N 1. t "NeN 60 Schemecheme 26. N-Alkylation usiusing an alkylIkwl halidhalide or N C-haloketone (General Procedure Z) N R" - in 2O - I -, -- N o- 65 R R 1 n R US 9,365,579 B2 99 100

Scheme 27: Cyclization of an amide using a dithiaphosphetane reagent Scheme 30: Hydrogenation of an alkene (General Procedure DD) (General Procedure AA) O 5 l NH

21N N -e- 10 Sa - in o 2O R 15 Scheme 31: Reductive amination of a ketone or aldehyde R (General Procedure EE)

N-( R" R' N N O NY N 2O N 1. - H R R(H) o-29 R 25

Scheme 32: Debenzylation of an amine (General Procedure FF)

R 30 RN N YNH -e- Scheme 28: Knoevenagel condensation to form a substituted R R cyclopentadiene (General Procedure BB) O O O O He- 35 R NO ulus O1 R. R. No lulux Na" O O Scheme 33: Hydrolysis of an ester to a carboxylic acid N t R (General Procedure GG) O 40 R O O

R ul OR R ul OH O O V 45 R

Scheme 34: Dehydration of a amide to a nitrile (General Procedure HH) 50

O Scheme 29: Decarboxylation of a B-ketoester enolate 2 (General Procedure CC) R NH2 -2 55

Na"8. O O N R" O O Scheme 35: Chiral preparative HPLC separation of stereoisomers R -e- R 60 (General Procedure II)

R O O H H wH O \ O V 65 -e- and . US 9,365,579 B2 101 102 General Procedure M Formation of aurea from an amine and Scheme 36: Acidic hydrolysis of an acetyl protected amine a carbamoyl chloride (General Procedure JJ) General Procedure N Formation of a sulfonamide from an amine General Procedure O Displacement of an aryl or heteroaryl 's-NN - - - YNH halide with an amine General Procedure P Boc-protection of an amine R R 10 General Procedure Q Cbz-protection of an amine General Procedure R Reduction of a pyridine General Procedure S Reduction of an ester to an alcohol Scheme 37: Cyclopropanation using chloroiodomethane (General Procedure KK) General Procedure TOxidation of an alcohol to an aldehyde 15 General Procedure UFormation of a semicarbazide General Procedure V Cyclization of a semicarbazide General Procedure W Formation of an acid chloride r), - . General Procedure X Formation of a urea using CDI R R General Procedure Y Formation of an ester from a carboxylic acid General Procedure Z N-Alkylation using an alkyl halide or C-haloketone Scheme 38: Formation of a bromomethyl ketone from an acid chloride 25 (General Procedure LL) General Procedure AA Cyclization of an amide using a dithiaphosphetane reagent O O General Procedure BB Knoevenagel condensation to form a -e- 30 Substituted cyclopentadiene R us C R -- Br General Procedure CCDecarboxylation of a f-ketoester eno late General Procedure DD Hydrogenation of an alkene 35 General Procedure EE Reductive amination of a ketone or Scheme 39: Reduction of an C.B-unsaturated ketone to an allylic alcohol (General Procedure MM) aldehyde General Procedure FF Debenzylation of an amine General Procedure GG Hydrolysis of an ester to a carboxylic OH 40 acid R R General Procedure HH Dehydration of an amide to a nitrile R R General Procedure II Chiral preparative HPLC separation of Stereoisomers List of General Procedures 45 General Procedure JJ Acidic hydrolysis of an acetyl protected General Procedure A Formation of a hydrazide from a car amine boxylic acid General Procedure B Formation of a hydrazide from an acid General Procedure KK Cyclopropanation using chlor chloride followed by cyclization and sulfonamide hydroly oiodomethane S1S 50 General Procedure LL Formation of a bromomethyl ketone General Procedure C Cyclization of a hydrazide from an acid chloride General Procedure D Cyclization of a hydrazide followed by General Procedure MM Reduction of an O.B-unsaturated sulfonamide hydrolysis and Boc-deprotection ketone to an allylic alcohol General Procedure E Cyclization of a hydrazide followed by The following examples are ordered according to the final sulfonamide hydrolysis 55 General Procedure F Cyclization of a hydrazide with loss of general procedure used in their preparation. The synthetic Boc-protecting group followed by Sulfonamide hydrolysis routes to any novel intermediates are detailed by sequentially General Procedure G Formation of a hydrazone followed by listing the general procedure (letter codes) in parentheses cyclization and Sulfonamide hydrolysis after their name with additional reactants or reagents as General Procedure H Hydrolysis of a sulfonamide 60 appropriate. A worked example of this protocol is given General Procedure I Acidic cleavage of a Boc-protected below using Example #H.1.1 as a non-limiting illustration. amine Example #H.1.1 is N-(4-(6H-pyrrolo2,3-e12.4 triazolo4. General Procedure J Deprotection of a Cbz-protected amine 3-alpyrazin-1-yl)bicyclo[2.2.2]octan-1-yl)-3-chlorobenze General Procedure K Formation of an amide from an acti nesulfonamide, which was prepared from 3-chloro-N-(4-(6- Vated acid and an amine 65 tosyl-6H-pyrrolo2,3-e 1.2.4 triazolo 4,3-alpyrazin-1-yl) General Procedure L Formation of an amide from a carboxy bicyclo2.2.2]octan-1-yl)benzenesulfonamide using General lic acid and an amine Procedure H as represented in Scheme A. US 9,365,579 B2 103 104

Scheme A Scheme B C t 5 HN e O Cy W s N -- si Y. 29 NH 10 o2

N General Procedure H Preparation #9

r H N. 15 N O

N-N N K HerGen. Proc. A O N N O Oesa 2O O OH Commercially available from Prime Organics Precusor to Example #H.1.1 C 25 O "Y- O K HN \ NH OES 30 HN 2N Gen. Proc. C CySa re N Ye /Ns 35 o? O

rs N 40 O O Ns M Example #H.1.1 N N N Gen. Proc. I N -e- The precursor to Example iH.1. 1.3-chloro-N-(4-(6-tosyl- 45 Dy 6H-pyrrolo2,3-e12.4 triazolo 4,3-alpyrazin-1-yl)bicyclo N V20 2.2.2]octan-1-yl)benzene Sulfonamide, was prepared as 2 S shown in Scheme B. 2-Hydrazinyl-5-tosyl-5H-pyrrolo2,3- O bipyrazine (Preparation #9) and 4-(tert-butoxycarbony lamino)bicyclo-2.2.2]octane-1-carboxylic acid are reacted 50 NH following the conditions given in General Procedure A to give tert-butyl 4-(2-(5-tosyl-5H-pyrrolo2,3-bipyrazin-2-yl)hy drazinecarbonyl)bicyclo[2.2.2]octan-1-ylcarbamate. This Ns hydrazide is cyclized using the conditions given in General ( N Gen.Proc. N Procedure C to afford tert-butyl 4-(6-tosyl-6H-pyrrolo2,3-e 55 N Hs 1.2.4 triazolo 4,3-alpyrazin-1-yl)bicyclo2.2.2]octan-1-yl N C carbamate. This carbamate is deprotected using General Pro- N N cedure I to yield 4-(6-tosyl-6H-pyrrolo2,3-e 1.2.4 triazolo N V 29 4,3-alpyrazin-1-yl)bicyclo[2.2.2]octan-1-amine. This so 21 S O amine is Sulfonylated using the conditions described in Gen- oSs eral Procedure N to give the precursor to Example #H.1.1. V The reaction sequence detailed above is translated in the C preparations and examples section to “using. A from Prepara- Commercially tion #9 and 4-(tert-butoxycarbonylamino)bicyclo-2.2.2]oc- 65 available from tane-1-carboxylic acid Prime Organics, C with TEA, I, N Sigma-Aldrich from 3-chlorobenzenesulfonyl chloride'. US 9,365,579 B2 105 106 -continued Analytical Methods C

5 Analytical data is included within the procedures below, in O OSs the illustrations of the general procedures, or in the tables of t examples. Unless otherwise stated, all "H NMR data were to collected on a Varian Mercury Plus 400 MHz or a Varian Inova 600 MHz instrument and chemical shifts are quoted in NS parts per million (ppm). LC/MS and HPLC data is referenced N N to the table of LC/MS and HPLC conditions using the lower N case method letter provided in Table 2. N N V2O 2.

Precusor to Example #H.1.1

TABLE 2

LCMS and HPLC methods

Method Conditions 8. LCMS: The gradient was 5-60% B in 1.5 min then 60-95% B to 2.5 min with a hold at 95% B for 1.2 min (1.3 mL/min flow rate). Mobile phase A was 10 mM NHOAc, mobile phase B was HPLC grade MeCN. The column used for the chromatography is a 4.6 x 50 mm MAC-MOD Halo C8 column (2.7 m particles). Detection methods are diode array (DAD) and evaporative light scattering (ELSD) detection as well as positive/negative electrospray ionization. b HPLC: The gradient was 10-60% B over 40 min (25 mL/min flow rate). Mobile phase A was 50 mM NHOAc (pH 4.5) and mobile phase B was HPLC grade MeCN. The column used for the chromatography was a 21.2 x 250 mm Hypersil C18HS column (8 m particles). Detection method is UV, , = 254 nm. C HPLC: The gradient was 10-100% B over 40 min, hold 5 min at 100% B, 2 min back to 10% B, 4 minhold at 10% B (21 mL/min flow rate). Mobile phase A was 50 mMNHOAc (pH 4.5) and mobile phase B was HPLC grade MeCN. The column used for the chromatography was a 21.2 x 250 mm Hypersil C18 HS column (8 um particles). Detection method is UV, , = 344 nm. d LC/MS: The gradient was 5-60% B in 0.75 min then 60-95% B to 1.15 min with a hold at 95% B for 0.75 min (1.3 mL/min flow rate). Mobile phase A was 10 mM NHOAc, mobile phase B was HPLC grade MeCN. The column used for the chromatography is a 4.6 x 50 mm MAC-MOD Halo C8 column (2.7 m particles). Detection methods are diode array (DAD) and evaporative light scattering (ELSD) detection as well as positive/negative electrospray ionization. e HPLC: The gradient was 5-95% B over 20 min (21 mL/min flow rate). Mobile phase A was 50 mM NHOAc (pH 4.5) and mobile phase B was HPLC grade MeCN. The column used for the chromatography was a 21.2 x 250 mm Hypersil C18HS column (8 m particles). Detection method is UV, , = 254 nm. f HPLC: The gradient was 0-30% B over 20 min (21 mL/min flow rate). Mobile phase A was 50 mM NHOAc (pH 4.5) and mobile phase B was HPLC grade MeCN. The column used for the chromatography was a 21.2 x 250 mm Hypersil C18HS column (8 m particles). Detection method is UV, , = 254 nm. 9. HPLC: The gradient was 0-50% B over 20 min (21 mL/min flow rate). Mobile phase A was 50 mM NHOAc (pH 4.5) and mobile phase B was HPLC grade MeCN. The column used for the chromatography was a 21.2 x 250 mm Hypersil C18HS column (8 m particles). Detection method is UV, , = 254 nm. h HPLC: The gradient was 20-60% B over 40 min (81 mL/min flow rate), mobile phase A was 50 mM NHOAc (pH 4.5) and mobile phase B was HPLC grade M eCN, the column used for the chromatography was a 25 x 250 mm Hypersil C18 HS column (10 um particles), detection method is UV, W = 315 nm. i HPLC: The gradient was 10-80% B over 9 min then 80-100% B over 0.10 min with a hold at 100% B for 1.50 min (22.5 mL/min flow rate). Mobile phase A was 50 mMNHOAc (pH 4.5) and mobile phase B was HPLC grade MeCN, the column used for the chromatography was a 19 x 50 mm Waters Atlantis T3 OBDC18 column (5 um particles), detection methods are Photodiode array DAD and Waters ZQ 2000 mass spectrometer. US 9,365,579 B2 107 108 TABLE 2-continued

LCMS and HPLC methods

Method Conditions HPLC: The gradient was 0-40% B over 30 min (21 mL/min flow rate). Mobile phase A was 50 mM NHOAc (pH 4.5) and mobile phase B was HPLC grade MeCN. The column used for the chromatography was a 21.2 x 250 mm Hypersil C18 HS column (8 m particles). Detection method is UV, , = 254 nm. HPLC: The gradient was 25-100% B over 25 min (21 mL/min flow rate). Mobile phase A was 50 mM NHOAc (pH 4.5) and mobile phase B was HPLC grade MeCN. The column used for chromatography was a 21.2 x 250 mm Hypersil HS C18 column (8 um particles). Detection method is UV, W = 380 nm. LCMS: The gradient was 0.1 min at 10% B, 10-100% B over 2.5 min with a hold at 100% B for 0.3 min, then to 10% B over 0.1 min. Mobile phase A was 0.1% TFA in water and mobile phase B was HPLC grade MeCN. The column used for the chromatography was a 2.1 mm x 30 mm Phenomenex Luna Combi-HTS C8(2) (5 M particles). Detection methods are Waters 996 diode-array detector and Sedere Sedex-75 ELSD. The ZMD mass spectrometer was operated under positive APCI ionization conditions. HPLC: The gradient was 10-100% B over 50 min (21 mL/min flow rate). Mobile phase A was 50 mM NHOAc (pH 4.5) and mobile phase B was HPLC grade MeCN. The column used for the c hromatography was a 21.2 x 250 mm Hypersil C18 HS column (8 m particles). Detection method is UV, W = 341 nm. LCMS: The gradient was 30-60% B in 1.50 min then 60-95% B to 2.5 min with a hold at 95% B for 1.2 min (1.3 mL/min flow rate). Mobile phase A was 10 mM ammonium acetate, mobile phase B was HPLC grade MeCN. The column used for he chromatography is a 4.6 x 50 mm MAC-MOD Halo C8 column (2.7 m particles). Detection methods are iode array (DAD) and evaporative light scattering (ELSD) detection as well as positive/negative electrospray ionization. LCMS: The gradient was 5-60% B in 1.5 min then 60-95% B to 2.5 min with a hold at 95% B for 1.2 min (1.3 mL/min flow rate). Mobile phase A was 10 mM ammonium acetate, mobile phase B was HPLC grade MeCN. The column used for he chromatography is a 4.6x30mm Vydac Genesis C8 column (4m particles). Detection methods are diode array (DAD) as well as positive/negative electrospray ionization and MS’ data dependen scanning on the positive ion scan (45 eV collision energy). HPLC: The column used for the chromatography was a 21.2 x 250 mm Hypersil C18 HS column (8 m particles). TThe gradient was 5-95% B over 50 min (21 mL/min flow rate). Mobile phase A was 0.05Naqueous ammonium acetate buffer (pH 4.5) and mobile phase B was HPLC grade MeCN. Detection method is UV, , = 254 nm. HPLC: The gradient was 10% to 5 0% B in 40 min (81 mL/min flow rate). Mobile phase A was 50 mMammonium acetate in water, mobile phase B was HPLC grade MeCN. The column used for the c hromatography was a Microsorb C18, 100 A, 5 Im, 46 x 250 mm column. Detection method is UV, , = 310 nm. HPLC: The gradient was 30% to 70% B in 40 min (81 mL/min flow rate). Mobile phase A was 50 mMammonium acetate in water, mobile phase B was HPLC grade MeCN. The column used for the c hromatography was a Microsorb C18, 100 A, 5 Im, 46 x 250 mm column. Detection method is UV, , = 254 nm. HPLC: The gradient was 10-40% B over 50 min, 40-100% over 3 min, hold 5 min at 00% B, 2 min back to 10% B, 3 min hold at 10% B (21 mL/min flow rate). Mobile phase A was 50 mM NHOAc (pH 4.5) and mobile phase B was HPLC grade MeCN. The column used for the c hromatography was a 21.2 x 250 mm Hypersil C18 HS column (8 m particles). Detection method is UV, W = 326 mm. HPLC: The column used for the chromatography is a 19 x 50 mm Waters Atlantis T-3 column(5um particles). The gradient was 20-25% B in 3.0 min then 25-95% B to 9.00 min with a hold at 95% B for 0.10 min (25 mL/min flow rate). Mobile phase A was 50 mM ammonium acetate, m obile phase B was HPLC grade acetonitrile. Detection methods are Waters 2996 PDA and Mass Spec is a Waters ZQ 2000. Mass spec detection uses both pos?neg Switching under APCI ionization. HPLC: The gradient was 5-100% B over 20 min (21 mL/min flow rate). Mobile phase A was 50 mM NHOAc (pH 4.5) and mobile phase B was HPLC grade MeCN. The column used for the chromatography was a 21.2 x 250 mm Hypersil C18 HS column (8 m particles). Detection method is UV, , = 254 nm.

TABLE 3

Chiral HPLC methods

Method Conditions The gradient was 5-60% A in 19 min with a hold at 60%. A for 2 min (20 mL/min flow rate). Mobile phase A was ethanol (200 proof), mobile phase B was HPLC grade heptane with 0.1% diethylamine added. The column used for the chromatography was a Daicel IC, 20 x 250 mm column (5 Imparticles). Detection methods were evaporative light scattering (ELSD) detection as well as optical rotation. US 9,365,579 B2 109 110 TABLE 3-continued

Chiral HPLC methods

Method Conditions 2 The gradient was 30-58% A in 12 min (20 mL/min flow rate). Mobile phase A was HPLC grade isopropanol, mobile phase B is HPLC grade heptane. The column used for the chromatography is a Daicel LA, 20 x 250 mm column (5 Imparticles). Detection methods were UV, , = 280 nm, evaporative light scattering (ELSD) detection as well as optical rotation. 3 Isocratic 30% A for 25 min (20 mL/min flow rate). Mobile phase A was ethanol (200 proof), mobile phase B was HPLC grade heptane with 0.1% diethylamine added. The column used for the chromatography was a Daicel IA, 20 x 250 mm column (5 Imparticles). Detection methods were evaporative light scattering (ELSD) detection as well as optical rotation. 4 Isocratic 20%. A for 40 min (20 mL/min flow rate). Mobile phase A was ethanol (200 proof), mobile phase B was HPLC grade heptane with 0.1% diethylamine added. The column used for the chromatography was a Daicel IA, 20 x 250 mm column (5 Imparticles). Detection methods were evaporative light scattering (ELSD) detection as well as optical rotation. 5 The gradient was 30-65% A in 18 min (20 mL/min flow rate). Mobile phase A was HPLC grade isopropanol, mobile phase B was HPLC grade heptane with 0.1% diethylamine added. The column used for the chromatography was a Daicel IA, 20 x 250 mm column (5 Im particles). Detection methods were UV, , = 280 nm, evaporative light scattering (ELSD) detection as well as optical rotation. 6 The gradient was 10-55% A in 19 min with a hold at 55% for 0.5 min (20 mL/min flow rate). Mobile phase A was a 50:50 mixture of HPLC grade methanol and ethanol (200 proof), mobile phase B was HPLC grade heptane with 0.1% diethylamine added. The column used for the chromatography was a Daicel IA, 20 x 250 mm column (5 Im particles). Detection methods were evaporative light scattering (ELSD) detection as well as optical rotation. 7 The gradient was 30-70% A in 18 min (20 mL/min flow rate). Mobile phase A was ethanol (200 proof), mobile phase B was HPLC grade heptane with 0.1% diethylamine added. The column used for the chromatography was a Daicel IC, 20 x 250 mm column (5 Im particles). Detection methods were UV, , = 280 nm, evaporative light scattering (ELSD) detection as well as optical rotation. 8 Isocratic 20% A for 30 min (20 mL/min flow rate). Mobile phase A was HPLC grade isopropanol, mobile phase B was HPLC grade heptane with 0.1% diethylamine added. The column used for the chromatography was a Daicel IA, 20 x 250 mm column (5 Im particles). Detection methods were evaporative light scattering (ELSD) detection as well as optical rotation. 9 Isocratic 50%. A for 25 min (20 mL/min flow rate). Mobile phase A was a 50:50 mixture of HPLC grade methanol and ethanol (200 proof), mobile phase B was HPLC grade heptane with 0.1% diethylamine added. The column used for the chromatography was a Daicel IA, 20 x 250 mm column (5 Imparticles). Detection methods were evaporative light scattering (ELSD) detection as well as optical rotation. 10 Isocratic 70%. A for 25 min (20 mL/min flow rate). Mobile phase A was ethanol (200 proof), mobile phase B was HPLC grade heptane with 0.1% diethylamine added. The column used for the chromatography was a Daicel IA, 20 x 250 mm column (5 Imparticles). Detection methods were evaporative light scattering (ELSD) detection as well as optical rotation.

PREPARATIONS AND EXAMPLES 50 Preparation #1 2-Bromo-5-(4-tert-butylphenylsulfonyl)-5H-pyrrolo The general synthetic methods used in each General Pro- 2,3-bipyrazine cedure follow and include an illustration of a compound that was synthesized using the designated General Procedure. 55 Br N None of the specific conditions and reagents noted herein are t y to be construed as limiting the scope of the invention and are 4 N provided for illustrative purposes only. All starting materials \ leO are commercially available from Sigma-Aldrich (including Fluka and Discovery CPR) unless otherwise noted after the 60 chemical name. Reagent/reactant names given are as named on the commercial bottle or as generated by IUPAC conven tions, CambridgeSoft(R) Chemdraw Ultra 9.0.7 or AutoNom 2000. Compounds designated as salts (e.g. hydrochloride, as acetate) may contain more than one molar equivalent of the salt. US 9,365,579 B2 111 112 A solution of 2-bromo-5H-pyrrolo2,3-bipyrazine (5.00 g, pyrazine (21.8 g. 55.3 mmol, Preparation #1), tert-butyl 25.2 mmol, Ark Pharm) in DMF (150 mL) was cooled in an hydrazinecarboxylate (36.5 g. 276 mmol), and NaOt-Bu ice bath to about 0° C. and then NaH (60% dispersion in (7.97g, 83 mmol) were added. After an additional vacuum/ mineral oil, 1.21 g, 30.3 mmol) was added. After about 15 nitrogen purge, the reaction was heated at about 80° C. for min, 4-tert-butylbenzene-1-sulfonyl chloride (6.46g, 27.8 about 5.5 h. The reaction was cooled to ambient temperature mmol) was added. The reaction was maintained between and filtered through Celite R, while washing with EtOAc (500 about 0-10°C. for about 2 h. Then, the reaction was diluted mL). The filtrate was washed with saturated aqueous NHCl with water (200 mL) to give a yellow suspension. The solid (3x500 mL), saturated aqueous NaHCO, (500 mL) and brine was collected by vacuum filtration, while washing with addi 10 (500 mL), dried over anhydrous NaSO, filtered, and then tional water (100 mL), and dried in a vacuum oven at about concentrated under reduced pressure to give about 55 g of a 70° C. to give 2-bromo-5-(4-tert-butylphenylsulfonyl)-5H crude brown oil. The brown oil was adsorbed onto silica and pyrrolo[2,3-bipyrazine (9.05 g, 91%). LC/MS (Table 2, purified by silica gel chromatography eluting with a gradient Method a) R, 3.05 min: MS m/z: 394/396 (M+H)". 15 of 10-50% EtOAc in heptane to give tert-butyl 2-(5-(4-tert butylphenylsulfonyl)-5H-pyrrolo2,3-bipyrazin-2-yl)hydra Preparation #2 Zinecarboxylate (4.51 g, 18% yield) and 4.68 g of a mixture of tert-butyl 2-(5-(4-tert-butylphenylsulfonyl)-5H-pyrrolo2,3- tert-Butyl 2-(5-(4-tert-butylphenylsulfonyl)-5H-pyr bipyrazin-2-yl)hydrazinecarboxylate major regioisomer rolo2,3-bipyrazin-2-yl)hydrazinecarboxylate and tert-butyl 1-(5-(4-tert-butylphenylsulfonyl)-5H-pyr and tert-butyl 1-(5-(4-tert-butylphenylsulfonyl)-5H-pyrrolo rolo2,3-bipyrazin-2-yl)hydrazinecarboxylate 2,3-bipyrazin-2-yl)hydrazinecarboxylate minor regioiso mer: LC/MS (Table 2, Methoda) R-2.68 min: MS m/z. 446 (M+H)" major regioisomer: R2.77 min; MS m/z: 446 25 (M+H) minor regioisomer. Preparation #3 5-(4-tert-Butylphenylsulfonyl)-2-hydrazinyl-5H 30 pyrrolo2,3-bipyrazine

H -N N N 35 22' 2NN N \-0 oaS 40

t 45 O N s > r y To a mixture of tert-butyl 2-(5-(4-tert-butylphenylsulfo 4. N nyl)-5H-pyrrolo2,3-bipyrazin-2-yl)hydrazinecarboxylate 50 and tert-butyl 1-(5-(4-tert-butylphenylsulfonyl)-5H-pyrrolo -2' 2,3-bipyrazin-2-yl)hydrazinecarboxylate (11.24 g, 25.2 mmol, Preparation #2) in 1,4-dioxane (125 mL) was added HCl (4M in 1,4-dioxane, 125 mL, 500 mmol). The reaction mixture was heated at about 60° C. for about 1 hand then the reaction mixture was cooled to ambient temperature. The mixture was filtered, while washing with EtO (150 mL), and the solid was partitioned between EtOAc (500 mL) and satu rated aqueous NaHCO (500 mL). The layers were separated To a flask was added Pd(dba) (5.06g, 5.53 mmol), di-tert 60 and the organic layer was washed with Saturated aqueous butyl-(2,4,6'-triisopropyl-biphenyl-2-yl)-phosphane (4.70 NaHCO and brine (200 mL each), dried over anhydrous g, 11.06 mmol), and 1,4-dioxane (350 mL). The catalyst Na2SO, filtered, concentrated under reduced pressure, and ligand mixture was degassed via Vacuum/nitrogen purge (3 dried in a vacuum oven at about 70° C. to give 5-(4-tert times) and heated at about 80° C. for about 10 min. The 65 butylphenylsulfonyl)-2-hydrazinyl-5H-pyrrolo2,3-bipyra reaction mixture is briefly removed from the oil bath then zine as a tan solid (7.54g, 87%); LC/MS (Table 2, Methoda) 2-bromo-5-(4-tert-butylphenylsulfonyl)-5H-pyrrolo2,3-b R=2.20 min: MS m/z: 346 (M+H)". US 9,365,579 B2 113 114 Preparation #4 solution was quenched with NHCl and was then extracted with EtOAc (2x100 mL). The combined organic extracts 2-Methylcyclohexanecarbonyl chloride were dried over anhydrous NaSO and concentrated under reduced pressure to give 1-(benzyloxycarbonyl)piperidine-4- carboxylic acid as a white solid (4.56 g. 22%); LC/MS (Table 2, Method a) R-1.93 min: MS m/z. 262 (M-H). Step B: Benzyl 10 4-(chlorocarbonyl)piperidine-1-carboxylate

OH C

To a solution of 2-methylcyclohexanecarboxylic acid (6.00 mL, 42.6 mmol, mixture of cis and trans) in DCM (60 mL) 15 was added oxalyl chloride (4.80 mL, 55.3 mmol) followed by DMF (0.03 mL, 0.4 mmol). The reaction mixture was stirred at ambient temperature for about 4 h before it was concen trated under reduced pressure to constant weight to afford cro -e- 2-methylcyclohexanecarbonyl chloride (mixture of diastere omers) as a yellow oil (7.0 g, 97%): "H NMR (400 MHz, CDC1) & 2.98-2.94 (m. 1H), 2.39-2.35 (m, 1H), 1.91-1.82 (m. 1H), 1.79-1.72 (m, 1H), 1.69-1.60 (m, 2H), 1.57-1.47 (m, 2H), 1.42-1.36 (m, 1H), 1.34-1.26 (m, 1H), 1.04-0.96 (m, 3H). 25 Preparation #5 soro Benzyl 4-(chlorocarbonyl)piperidine-1-carboxylate 30 To a solution of 1-(benzyloxycarbonyl)piperidine-4-car boxylic acid (4.50 g, 17.1 mmol. Preparation #5, Step A) in DCM (40 mL) at ambient temperature was added oxalyl O chloride (3.00 mL, 34.2 mmol) followed by DMF (0.10 mL, 1.3 mmol). After about 3 h, the reaction was concentrated 35 under reduced pressure to constant weight to afford benzyl ls 4-(chlorocarbonyl)piperidine-1-carboxylate as a yellow oil (3.88g, 81%): 'H NMR (CDC1) & 7.44-7.35 (m,5H), 5.16 (s, 2H), 4.20-4.10 (m, 2H), 3.03-2.89 (m, 3H), 2.15-2.05 (m, 2H), 1.81-1.76 (m, 2H). C 40 Preparation #6 Step A: 1-(Benzyloxycarbonyl)piperidine-4-carboxylic acid Perfluorophenyl 2-cyanoacetate 45

50 y O 4. O F OH y/ - HO 55 F F

OH 60 To a solution of 2,3,4,5,6-pentafluorophenol (1.08 g, 5.88 mmol) and 2- (0.50g, 5.9 mmol) in DCM (20 To a solution of piperidine-4-carboxylic acid (10.0 g, 77.4 mL) was added DCC (1.21 g, 5.88 mmol). After stirring for mmol) and NaCOs (8.21 g, 77.4 mmol) in water (100 mL) about 4 h at ambient temperature, the reaction was concen was added a solution of benzyl 2,5-dioxopyrrolidin-1-yl car trated under reduced pressure and then purified over silica gel bonate (19.3 g, 77.4 mmol) in MeCN (100 mL). The reaction 65 (20 g) using DCM as the eluent to afford perfluorophenyl was stirred at ambient temperature for about 16 hand then 2-cyanoacetate as a white solid (1.39 g, 94%): 'H NMR (400 concentrated under reduced pressure. The resulting aqueous MHz, CDC1) & 3.85 (s. 2H). US 9,365,579 B2 115 116 Preparation #7 (PPh)(1.11 g, 1.58 mmol). The reaction mixture was cooled at about 0°C. and a solution of (trimethylsilyl)acetylene (20.8 2-Bromo-5-tosyl-5H-pyrrolo2,3-bipyrazine mL, 150 mmol) in THF (146 mL) was added drop-wise. The (Method A) reaction mixture was stirred at about 0-10° C. for about 7 h and then concentrated under reduced pressure. The dark brown residue was dissolved in DCM (600 mL) and filtered Br N through a Celite(R) pad (3 cm in heightx9 cm in diameter) while eluting with DCM (300 mL). The filtrate was washed Cy2 Br N N N with water (2x500 mL) and brine (500 mL), dried over anhy drous MgSO, filtered through a Florisil R. pad (1 cm in height City - or by 9 cm in diameter) while washing with DCM/MeOH (9:1, % N 15 200 mL), and concentrated under reduced pressure to give a brown solid. The solid was triturated and sonicated with warm petroleum ether (b.p. 30-60° C., 250 mL), cooled and collected, washing with petroleum ether (b.p. 30-60° C.; 2x100 mL), and dried in a vacuum oven at about 70° C. to give A solution of 2-bromo-5H-pyrrolo2,3-bipyrazine (78.0 g, 5-bromo-3-((trimethylsilyl)ethynyl)pyrazin-2-amine (34.6 g. 394 mmol, Ark Pharm) in anhydrous DMF (272 mL) was 70%): LC/MS (Table 2, Methodd) R-1.59 min; MS m/z: 272 added drop-wise over about 60 minto a stirred suspension of (M+H)". NaH (12.8 g. 532 mmol) in anhydrous DMF (543 mL) at 25 about 0-5° C. The brown reaction solution was stirred for Step B: 2-Bromo-5-tosyl-5H-pyrrolo2,3-bipyrazine about 30 min at about 0-5°C. then a solution of p-toluene sulfonyl chloride (94.0g, 492 mmol) in anhydrous DMF (272 mL) was added drop-wise over about 60 min at about 0-5°C. 30 The reaction mixture was stirred at about 0-5°C. for about 1 h then allowed to warm to ambient temperature and stirred for l Br N 2^ about 18h at ambient temperature. The reaction mixture was N poured slowly into ice water (6 L), followed by the addition of 35 aqueous 2.5 N NaOH (50.0 mL, 125 mmol). The precipitate r N NH2 was collected by filtration and stirred with cold water (3x200 mL). The solid was collected by filtration and dried to con stant weight in a vacuum oven at about 55° C. to yield 2-bromo-5-tosyl-5H-pyrrolo2,3-bipyrazine (134.6 g. 97%) 40 as a pale beige solid: LC/MS (Table 2, Method d) R-1.58 min: MS m/z. 352/354 (M+H)".

Preparation #7 45 2-Bromo-5-tosyl-5H-pyrrolo2,3-bipyrazine (Method B) 50 Step A: To a solution of 5-bromo-3-((trimethylsilyl)ethynyl) 5-Bromo-3-((trimethylsilyl)ethynyl)pyrazin-2-amine pyrazin-2-amine (3.00 g, 11.1 mmol) in DMF (60 mL) at about 0°C. was added NaH (60% dispersion in mineral oil,

55 0.577 g. 14.4 mmol) in three portions. After about 15 min, |- p-toluenesulfonyl chloride (2.75 g, 14.4 mmol) was added Br N Br Sin and the reaction was allowed to warm slowly to ambient -- Br N 2 temperature. After about 16 h, the reaction mixture was 2 n 60 poured onto ice-cold water (120 mL) and the precipitate was N NH2 2 collected by vacuum filtration. The crude solid was dissolved N NH2 in DCM (15 mL) and purified by silica gel chromatography eluting with DCM. The product-containing fractions were

To a solution of 3,5-dibromopyrazin-2-amine (40.0 g, 158 65 concentrated under reduced pressure to give 2-bromo-5-to mmol), TEA (66.1 mL, 475 mmol), and copper(I) iodide syl-5H-pyrrolo2,3-bipyrazine (2.16 g. 52%): LC/MS (Table (0.301 g, 1.58 mmol) in THF (1172 ml) was added PdCl 2, Method d) R-1.58 min: MS m/z. 352/354 (M+H)". US 9,365,579 B2 117 118 Preparation #8 boxylate major regioisomer and tert-butyl 1-(5-tosyl-5H pyrrolo2,3-bipyrazin-2-yl)hydrazinecarboxylate minor tert-Butyl 2-(5-tosyl-5H-pyrrolo2,3-bipyrazin-2-yl) regioisomer (18.8 g. 50%): LC/MS (Table 2, Method d) hydrazinecarboxylate and tert-butyl 1-(5-tosyl-5H R=1.47 min: MS m/z. 404 (M+H)". pyrrolo2,3-bipyrazin-2-yl)hydrazinecarboxylate Preparation #9 2-Hydrazinyl-5-tosyl-5H-pyrrolo2,3-bipyrazine

10

Y N

15 try4. -- -2' 4Ny l

25 To a mixture of tert-butyl 2-(5-tosyl-5H-pyrrolo2,3-b pyrazin-2-yl)hydrazinecarboxylate and tert-butyl 1-(5-tosyl t 5H-pyrrolo2,3-bipyrazin-2-yl)hydrazinecarboxylate (18.8 O N N g, 46.6 mmol, Preparation #8) in 1,4-dioxane (239 mL) was added HCl (4 M in 1,4-dioxane, 86 mL, 345 mmol). The 30 reaction was heated at about 60° C. for about 1 h and then O 4. N cooled to about 15-20°C. The solid was collected by vacuum filtration, washed with cold 1,4-dioxane (2x20 mL), and then 25 stirred with a solution of saturated NaHCO and water (1:1, 150 mL). After about 1 h, the effervescence had subsided and the solid was collected by vacuum filtration, washed with ice cold water (3x20 mL), and dried in a vacuum oven to a constant weight to afford 2-hydrazinyl-5-tosyl-5H-pyrrolo2, 3-bipyrazine as a light yellowish brown solid (8.01 g, 50%): LC/MS (Table 2, Method d) R-1.28 min: MS m/z: 304 40 To a flask was added Pd(dba), (3.90 g, 4.26 mmol), di-tert (M+H)". butyl-(2,4,6'-triisopropylbiphenyl-2-yl)phosphane (3.62 g, Preparation #10 8.52 mmol), and anhydrous 1,4-dioxane (453 mL). The cata lyst-ligand mixture was degassed via vacuum/nitrogen purge (R)-tert-Butyl (3 times) and heated at about 80° C. for about 10 min. The 45 1-(chlorocarbonyl)pyrrolidin-3-ylcarbamate reaction mixture is briefly removed from the oil bath then 2-bromo-5-tosyl-5H-pyrrolo2,3-bipyrazine (30.0 g, 85 mmol, Preparation #7), tert-butyl hydrazinecarboxylate (16.9 g, 128 mmol), and NaOt-Bu (12.28 g. 128 mmol) were added. O After an additional vacuum/nitrogen purge, the reaction was 50 heated at about 80° C. After about 50 min, the reaction mix ture was cooled to ambient temperature and filtered through a pad of silica gel (6 cm in heightx6 cm in diameter), topped with Celite(R) (1 cm in heightx6 cm in diameter), while wash ing with EtOAc (3x150 mL). Water (300 mL) was added to 55 . . the filtrate and the organic layer was separated. The aqueous N layer was extracted with additional EtOAc (3x200 mL). The 9 C combined organic extracts were washed with Saturated aque O ous NHCl, saturated aqueous NaHCO, and brine (400 mL each), dried over anhydrous MgSO4, filtered, and concen 60 A flask was charged with (R)-tert-butylpyrrolidin-3-ylcar trated under reduced pressure to give a dark brown oil (45 g). bamate (1.0 g, 5.4 mmol, Lancaster) in DCM (15 mL) to give The brown oil was dissolved in DCM (250 mL), silica gel a colorless solution. Pyridine (0.89 mL, 10.8 mmol) was (200 g) was added, and the mixture was concentrated under added and the solution was cooled to about 0°C., followed by reduced pressure. The resulting silica mixture was purified the addition of triphosgene (0.64g, 2.1 mmol). The mixture using silica gel chromatography eluting with a gradient of 65 was stirred for about 1 h while slowly warming to ambient 25-65% EtOAc in heptane to give a mixture of tert-butyl temperature. To the reaction solution was added DCM (50 2-(5-tosyl-5H-pyrrolo2,3-bipyrazin-2-yl)hydrazinecar mL) and the solution was washed with water (20 mL) and HCl

US 9,365,579 B2 121 122 mmol) was added and reaction mixture was stirred for about Preparation #12 5 h. Concentrated NH-OH (5 mL) was added and the result (1R,2R,4S)-4-(tert-butoxycarbonylamino)-2-ethyl-1- ing mixture was stirred for about 5 min. The resulting Sus methylcyclopentanecarboxylic acid and (1S,2S.4R)- pension was filtered and the filter cake was washed with 5 4-(tert-butoxycarbonylamino)-2-ethyl-1-methylcy EtOAc (60 mL). The filtrate was partitioned and the aqueous clopentanecarboxylic acid layer was extracted with EtOAc (30 mL). The combined organic layer was washed with brine, dried over anhydrous O MgSO filtered, and concentrated to yield (1R,2S,4R,5S)- 10 ethyl 4-amino-1-methylbicyclo[3.1.0 hexane-2-carboxylate and (1S,2R,4S.5R)-ethyl 4-amino-1-methylbicyclo[3.1.0 HO hexane-2-carboxylate (0.21 g, 69%). This amine (0.212 g, NH 1.16 mmol) was reacted with cyclopropanesulfonyl chloride 15 (0.244 g, 1.74 mmol) using General Procedure N to give (1R,2S.4R,5S)-ethyl 4-(cyclopropanesulfonamido)-1-meth ylbicyclo[3.1.0 hexane-2-carboxylate and (1S,2R,4S.5R)- ethyl 4-(cyclopropanesulfonamido)-1-methylbicyclo[3.1.0 hexane-2-carboxylate (0.11 g, 33%): LC/MS (Table 2, Step A: (2R,4S)-Ethyl 4-(dibenzylamino)-2-ethyl-1- Method a) R, 2.06 min; MS m/z: 286 (M-H). methylcyclopentanecarboxylate and (2S,4R)-ethyl 4-(dilbenzylamino)-2-ethyl-1-methylcyclopentanecar boxylate Step D: (1R,2S,4R,5S)-4-(Cyclopropanesulfona 25 mido)-1-methylbicyclo[3.1.0 hexane-2-carboxylic acid and (1S,2R,4S.5R)-4-(cyclopropanesulfona mido)-1-methylbicyclo[3.1.0 hexane-2-carboxylic acid 30 /No 35 C 40 --, -) 45 C C

50 To a solution of LDA (1.8M in THF, 3.04 mL, 5.47 mmol) and THF (40 mL) at about -78°C. was added ethyl 4-(diben Zylamino)-2-ethylcyclopentanecarboxylate (1.0g, 2.7 mmol. Preparation #EE. 1) in THF (4 mL). The reaction mixture was stirred at about -78° C. for about 1 h. Mel (2.57 mL, 41.0 mmol) was added and reaction mixture was stirred at about A mixture of (1R,2S.4R,5S)-ethyl 4-(cyclopropane 55 -78°C. for about 1 h and was then warmed to about -40°C. DCM (150 mL) was added followed by saturated aqueous sulfonamido)-1-methylbicyclo[3.1.0 hexane-2-carboxylate NHCl solution (50 mL). The layers were separated and the and (1S,2R,4S.5R)-ethyl 4-(cyclopropanesulfonamido)-1- aqueous layer was extracted with DCM (2x30 mL). The methyl-bicyclo[3.1.0 hexane-2-carboxylate (0.109 g, 0.379 combined organic layers were washed with brine, dried over 60 anhydrous MgSO4, filtered, and concentrated to dryness mmol) was hydrolyzed using General Procedure GG to give under reduced pressure. The residue was purified by flash (1R,2S.4R,5S)-4-(cyclopropanesulfonamido)-1-methyl-bi silica gel chromatography eluting with a gradient of 0-10% cyclo[3.1.0 hexane-2-carboxylic acid and (1S,2R,4S.5R)-4- EtOAc in DCM to give (2R,4S)-ethyl 4-(dibenzylamino)-2- (cyclopropanesulfonamido)-1-methylbicyclo3.1.0 hexane ethyl-1-methylcyclopentanecarboxylate and (2S,4R)-ethyl 65 4-(dilbenzylamino)-2-ethyl-1-methylcyclopentanecarboxy 2-carboxylic acid (0.113g, 100%): LC/MS (Table 2, Method late (0.864g, 84%). LC/MS (Table 2, Methoda) R-2.25 min: a) R-1.57 min: MS m/z: 258 (M-H). MS m/z. 380 (M+H)".

US 9,365,579 B2 125 126 tanecarboxylic acid (0.256g, 86%): LC/MS (Table 2, Method Step A: (1R,4S)-tert-Butyl 3-oxo-2-azabicyclo[2.2.1 a) R, 2.22 min; MS m/z: 270 (M-H). hept-5-ene-2-carboxylate Preparation #13 O (1S,2R,4S) and (1R,2S,4R)-4-(tert-butoxycarbony

lamino)-2-ethyl-1-methylcyclopentanecarboxylic -e- acid Ol O ()'sO To a solution of (1R,4S)-2-azabicyclo[2.2.1]hept-5-en-3- one (1.50 g, 13.7 mmol) in THF (100 mL) was added TEA 1 15 (1.90 mL, 13.7 mmol) and DMAP (0.27g, 2.2 mmol). The O s O mixture was stirred for about 5 minat about 0°C. followed by the addition of di-tert-butyl dicarbonate (3.40 mL, 14.4 /No s' /No mmol) in THF (15 mL). The reaction was stirred at ambient NHBOC NHBOC temperature for about 24 h. The solvent was removed under 1:1 reduced pressure and the crude residue was taken up in DCM (50 mL) and washed with water (25 mL) and brine (25 mL). The organic layer was dried over anhydrous MgSO, filtered, and concentrated under reduced pressure. The crude material 25 was purified by silica gel chromatography eluting with a gradient of 0-30% EtOAc/heptane to afford (1R,4S)-tert-bu tyl 3-oxo-2-azabicyclo[2.2.1]hept-5-ene-2-carboxylate (2.7 g, 93%) as a white solid: "H NMR (400 MHz, DMSO-d) & 7.26-6.86 (dd. 1H), 6.86-6.64 (m. 1H), 5.08-4.78 (d. 1H), 30 3.52-3.21 (dd. 1H), 2.32-2.24 (d. 1H), 2.09-2.02 (d. 1H), NHBOC 105-1.36 (s, 9H). Step B: (1S,2R,4R,5R)-7-Oxo-3-trimethylsilanyl-6- aza-tricyclo[3.2.1.0(2.4)octane-6-carboxylic acid A mixture of (1S,2R,4S) and (1R,2S,4R)-ethyl 4-(tert-bu 35 tert-butyl ester toxycarbonylamino)-2-ethyl-1-methylcyclopentanecar boxylate (0.180g, 0.600 mmol) was hydrolyzed according to

General procedure GG to give (1S,2R,4S)-4-(tert-butoxycar bonylamino)-2-ethyl-1-methylcyclo-pentanecarboxylic acid 40 and (1S,2R,4S)-4-(tert-butoxycarbonylamino)-2-ethyl-1- methyl-cyclopentanecarboxylic acid (0.083 g, 51%): LC/MS (Table 2, Method a) R, 2.23 min: MS m/z: 270 (M-H). 45 Preparation #14

(1R,2S.4R,5R)-4-(tert-Butoxycarbonylamino)-6- (trimethylsilyl)bicyclo[3.1.0 hexane-2-carboxylic 50 acid To a solution of (1R,4S)-tert-butyl 3-oxo-2-azabicyclo 2.2.1]hept-5-ene-2-carboxylate (1.3 g. 6.2 mmol) and palla dium(II) acetate (0.070 g., 0.31 mmol) in Et2O (62 mL) was 55 added trimethylsilyldiazomethane (2M in hexanes, 3.00 mL. 11.5 mmol) drop-wise at ambient temperature over about 1 h. The mixture was stirred at ambient temperature for about 18 hand filtered through Celite(R). The CeliteR pad was washed 60 with EtO (50 mL) and the filtrate was concentrated under reduced pressure. The crude material was purified by silica gel chromatography eluting with a gradient of 0-30% EtOAc/ heptane to afford (1S,2R,4R,5R)-7-oxo-3-trimethylsilanyl 6-aza-tricyclo[3.2.1.0(2.4)octane-6-carboxylic acid tert-bu 65 tylester (1.7g.92%). "H NMR (400 MHz, DMSO-d) & 4.37 (s, 1H), 2.70 (s, 1H), 1.45 (m. 10H), 1.23 (t, 1H), 0.76 (t, 1H), 0.10 (s. 2H), -0.03 (s, 9H). US 9,365,579 B2 127 128

Step C: (1R,2S,4R,5R)-4-(tert -continued Butoxycarbonylamino)-6-(trimethylsilyl)bicyclo 3.1.0 hexane-2-carboxylic acid

To a solution of tert-butyl (1R,2R,4S.5R)-4-(6-tosyl-6H pyrrolo2,3-e12.4 triazolo 4.3-alpyrazin-1-yl)-6-(trim A mixture of (1S,2R,4R,5R)-7-oxo-3-trimethylsilanyl-6- 25 ethylsilyl)bicyclo[3.1.0 hexan-2-ylcarbamate (0.780 g, 1.34 aza-tricyclo[3.2.1.0(2.4)octane-6-carboxylic acid tert-butyl ester (1.7 g., 5.7 mmol) and potassium fluoride on alumina mmol, prepared using. A from Preparation #9 and Preparation (2.10 g, 14.1 mmol) in THF (38 mL) was heated to about 60° #14 with HATU, C with TEA) in DCM (20 mL) was added C. for about 18 h. The mixture was cooled to ambient tem trifluoromethanesulfonic acid (0.48 mL, 5.4 mmol). After perature and filtered through Celite(R). The Celite(R) pad was 30 stirring at ambient temperature for about 18 h, additional rinsed with EtOAc (50 mL) and the filtrate was concentrated trifluoromethanesulfonic acid (0.48 mL, 5.4 mmol) was under reduced pressure to afford (1R,2S,4R,5R)-4-(tert-bu toxycarbonylamino)-6-(trimethylsilyl)bicyclo[3.1.0 hex added and the mixture was stirred for about an additional 18 h. The reaction mixture was diluted with DCM (40 mL) and ane-2-carboxylic acid (1.82 g, 100%): LC/MS (Table 2, 35 Method a) R-2.62 min: MS m/z. 312 (M-H). slowly poured into a vigorously stirred slurry of ice water (30 mL). After about 5 min the reaction mixture was neutralized Preparation #15 with saturated aqueous NaHCO. The layers were separated and the aqueous layer was extracted with DCM (40 mL). The 40 (1R,2R,4S,5S)-4-(6-Tosyl-6H-pyrrolo2,3-e12.4 combined organic layers were washed with brine, dried over triazolo 4,3-alpyrazin-1-yl)bicyclo[3.1.0 hexan-2- anhydrous MgSO filtered, and concentrated under reduced amine pressure to afford to afford (1R,2R,4S,5S)-4-(6-tosyl-6H pyrrolo2,3-e12.4 triazolo 4.3-alpyrazin-1-yl)bicyclo 45 3.1.0 hexan-2-amine as a light brown solid (0.55 g, 87%): LC/MS (Table 2, Method a) R-1.75 min: MS m/z: 409 (M+H)".

50 Preparation #16

55 Lithium (R)-4-(tert-butoxycarbonyl)-1-methylpipera Zine-2-carboxylate

60

65 US 9,365,579 B2 129 130 Step A: (R)-1-tert-Butyl 3-methyl To a solution of (R)-1-tert-butyl 3-methyl 4-methylpipera 4-methylpiperazine-1,3-dicarboxylate Zine-1,3-dicarboxylate (1.2g, 4.6 mmol) in 1,4-dioxane (18 mL) and water (18 mL) was added LiOH.H2O (0.290 g. 6.91 mmol). After heating at about 80°C. for about 1 h, the reac tion mixture was cooled to room temperature and the Solvent was removed under reduced pressure. The solid was dried in a vacuum ovenat about 65°C. for about 18h to afford lithium

10 (R)-4-(tert-butoxycarbonyl)-1-methylpiperazine-2-carboxy late (1.46 g, quantitative): LC/MS (Table 2, Method a) R=1.17 min: MS m/z. 245 (M+H)". Sr.Sysk 15 O Preparation #17

To (R)-1-tert-butyl 3-methylpiperazine-1,3-dicarboxylate (1S,4R)-4-(tert-Butoxycarbonylamino)cyclopent-2- (1.2g, 4.9 mmol, ASW Med Chem Inc) in MeCN and MeOH enecarboxylic acid (1:1, 100 mL) was added formaldehyde (37% aqueous, 13.2 25 mL, 177 mmol), followed by the addition of sodium triac etoxyborohydride (5.20 g, 24.5 mmol). The mixture was stirred for about 15 min at ambient temperature. AcOH (5.6 mL, 98 mmol) was added drop-wise and the mixture was 30 stirred for about 1 h. The solvent was removed under reduced pressure and the residue was dissolved in DCM (100 mL) and N neutralized using aqueous 2 N NaOH. Saturated aqueous NaHCO (50 mL) was added and the layers were separated. 35 The organic layer was washed with brine (50 mL), dried over Olx anhydrous MgSO filtered, and concentrated under reduced pressure. The crude material was purified by silica gel chro matography eluting with a gradient of 20-80% EtOAc/hep 40 tane to afford (R)-1-tert-butyl 3-methyl 4-methylpiperazine 1,3-dicarboxylate (1.1 g, 85%): LC/MS (Table 2, Methoda) To a solution of (1R,4S)-2-azabicyclo[2.2.1]hept-5-en-3- R=1.91 min: MS m/z: 259 (M+H)". one (5.0 g, 46 mmol) in water (30.5 mL) was added aqueous 45 HCl (2 M, 23.0 mL, 46.0 mmol). After heating at about 80° C. Step B: Lithium (R)-4-(tert-butoxycarbonyl)-1-meth for about 2 h, the reaction mixture was cooled to ambient ylpiperazine-2-carboxylate temperature and the solvent was removed under reduced pres sure. The solid was dried in a vacuum ovenat about 70° C. and used without further purification. To a solution of (1S,4R)-4- 50 aminocyclopent-2-enecarboxylic acid hydrochloride (9.20g, 45.8 mmol) in 1,4-dioxane (15 mL) and water (18.3 mL) at about 0°C. was added DIEA (32.0 mL, 183 mmol). After

55 stirring for about 5 min, a solution of di-tert-butyl dicarbonate (11.7 mL, 50.4 mmol) in 1,4-dioxane (5 mL) was added. The reaction mixture was warmed to ambient temperature and stirred for about 18 h. Solvent was removed under reduced

60 pressure and the crude oil was dried in a vacuum oven at about 65° C. for about 3 h. The crude product was purified by silica gel chromatography eluting with a gradient of 80-100% EtOAc/heptane to afford (1S,4R)-4-(tert-butoxycarbony lamino)cyclopent-2-enecarboxylic acid (5.2 g, 50% over 2 steps): LC/MS (Table 2, Method a) R-1.81 min: MS m/z: 226 (M-H). US 9,365,579 B2 131 132 Preparation #18 Preparation #19 (9H-Fluoren-9-yl)methyl 4-methyl-3-(3-tosyl-3H (1S,2R,4S.5R)-4-(6-Tosyl-6H-pyrrolo2,3-e12.4 imidazol-2-alpyrrolo2,3-epyrazin-8-yl)piperidine triazolo 4,3-alpyrazin-1-yl)bicyclo[3.1.0 hexan-2- 1-carboxylate amine - H -, O Sr. 10 Ns, 15 ( Cy -- N N n s N Cy N N 29 22' 4.

25

30

35

40

To a solution of (9H-fluoren-9-yl)methyl 3-(2-(tert-bu 45 toxycarbonyl(5-tosyl-5H-pyrrolo2,3-bipyrazin-2-yl) To a solution of ethyl (1S,2R,4S.5R)-4-(6-tosyl-6H-pyr amino)acetyl)-4-methylpiperidine-1-carboxylate (0.627 g. 0.836 mmol, prepared using W from Preparation #20, LL, Z rolo2,3-e 1.2.4 triazolo 4,3-alpyrazin-1-yl)bicyclo[3.1.0 from Example #8, Step A) in DCM (10 mL) was added TFA hexan-2-ylcarbamate (0.16 g., 0.34 mmol, prepared using GG (1.50 mL, 19.5 mmol) and the resulting mixture was stirred at from Preparation #KK.1, A from Preparation #9 with HATU 50 ambient temperature under nitrogen for about 1 h. The solu and TEA, C with TEA) in DCM (2.3 mL) was added trimeth tion was concentrated and the residue was partitioned ylsilyl iodide (0.11 mL, 0.75 mmol). After stirring at ambient between saturated aqueous NaHCO (25 mL) and EtOAc (25 temperature for about 24 h. additional trimethylsilyl iodide mL). The organic phase was washed with brine (20 mL), dried (0.11 mL, 0.75 mmol) was added and the reaction mixture over anhydrous MgSO, filtered, and concentrated to yield was heated to about 40° C. for about 4 days. The reaction 55 crude (9H-fluoren-9-yl)methyl 4-methyl-3-(2-(5-tosyl-5H mixture was cooled to ambient temperature, followed by the pyrrolo2,3-bipyrazin-2-ylamino)acetyl)-piperidine-1-car boxylate as an amorphous brown solid. The crude material addition of saturated aqueous NaHCO (20 mL). The mixture was added to 1,4-dioxane (5 mL), Lawesson’s reagent (0.203 was stirred for about 5 min and the layers were separated. The g, 0.502 mmol) was added, and the resulting Suspension was aqueous layer was further extracted with DCM (20 mL). The 60 heated at about 80° C. for about 20 min. The solvent was combined organic layers were washed with brine (20 mL), removed under reduced pressure and the residue was purified dried over anhydrous MgSO filtered, and concentrated by silica gel chromatography eluting with a gradient of 0 to under reduced pressure to afford (1S,2R,4S.5R)-4-(6-tosyl 1.5% MeOH/DCM to give (9H-fluoren-9-yl)methyl 4-me 6H-pyrrolo2,3-e12.4 triazolo 4,3-alpyrazin-1-yl)bicyclo thyl-3-(3-tosyl-3H-imidazo 1,2-alpyrrolo2,3-epyrazin-8- 3.1.0 hexan-2-amine that contained 1 molar equiv DCM 65 yl)piperidine-1-carboxylate as an off-white solid (0.21 g, (0.17g, 100%): LC/MS (Table 2, Methoda) R-1.76 min: MS 40%): LC/MS (Table 2, Methoda) R, 2.68 min: MS m/z: 632 m/z: 409 (M+H)". (M+H)". US 9,365,579 B2 133 134 Preparation #20 1-(((9H-Fluoren-9-yl)methoxy)carbonyl)-4-meth ylpiperidine-3-carboxylic acid

C (

To a solution of 1-(tert-butoxycarbonyl)-4-methylpiperi -continued N dine-3-carboxylic acid (1.50 g. 6.17 mmol. Example #13, 25 a Step G) in 1,4-dioxane (10 mL) was added aqueous HCl (4N N in 1,4-dioxane (4.62 mL, 18.5 mmol). The reaction mixture was heated at about 60° C. for about 16 h before being Q || 2 allowed to cool to ambient temperature. To the mixture was in S N added NaHCO (2.07 g. 24.7 mmol) and water (10.0 mL) \, followed by (9H-fluoren-9-yl)methyl 2,5-dioxopyrrolidin-1- 30 yl carbonate (4.16 g, 12.3 mmol). The reaction was stirred at about 25° C. for about 16 h. The reaction was acidified to Ns about pH 1 with aqueous 1N HCl and was extracted with { EtOAc (75 mL). The organic layer was washed with brine (50 N N mL), dried over anhydrous NaSO, filtered, and concen 35 Cy trated under reduced pressure. The product was purified by N N silica gel chromatography (40 g column) eluting with a gra N V 29 dient of 1-5% MeOH in DCM to give 1-(((9H-fluoren-9-yl) S methoxy)carbonyl)-4-methylpiperidine-3-carboxylic acid ca (0.72 g, 31%) as a clear oil: LC/MS (Table 2, Method a) 40 R=2.44 min: MS m/z. 366 (M+H)". Preparation #21 5-Cyano-N-((1R,3S)-2,2-dimethyl-3-(6-tosyl-6H pyrrolo2,3-e12.4 triazolo 4.3-alpyrazin-1-yl)cy To a solution of 5-bromo-N-((1R,3S)-2,2-dimethyl-3-(6- clobutyl)pyridine-2-sulfonamide tosyl-6H-pyrrolo2,3-e 1.2.4 triazolo 4,3-alpyrazin-1-yl) cyclobutyl)pyridine-2-sulfonamide (0.69 g, 1.1 mmol, pre pared using. A from (1S,3R)-3-acetamido-2,2- Q || 2 50 dimethylcyclobutanecarboxylic acid Tetrahedron. HN-y OrN V Asymmetry 2008, 19,302-308 and Preparation #9 with EDC, O C with DIEA, JJ, N from 5-bromopyridine-2-sulfonyl chlo ride Chem Impex) in degassed DMF (1.5 mL) was added dicyanozinc (0.321 g, 2.74 mmol) followed by Pd(PhP) Ns 55 (0.063 g, 0.055 mmol, Strem). The reaction was heated at { about 80°C. for about 16 hunder a nitrogen atmosphere. The N N reaction mixture was allowed to cool to ambient temperature Cy Hs before it was diluted with aqueous NaOH (1N, 10 mL) and Sa extracted with EtOAc (25 mL). The organic layer was washed N N 29 60 with brine (20 mL), dried over anhydrous NaSO filtered, and concentrated under reduced pressure. The product was 4. purified by silica gel chromatography (12 g) eluting with a gradient of 1-10% MeOH in DCM to give 5-cyano-N-((1R, 3S)-2,2-dimethyl-3-(6-tosyl-6H-pyrrolo2,3-e 1.2.4 tria 65 Zolo 4.3-alpyrazin-1-yl)cyclobutyl)pyridine-2-sulfonamide (0.09 g, 14%) as a tan solid: LC/MS (Table 2, Method a) R=2.14 min: MS m/z. 577 (M+H)". US 9,365,579 B2 135 136 Preparation #22 anhydrous NaSO4, filtered, and concentrated to dryness under reduced pressure to give 6-fluoro-4-methylnicotina 2-Acetylamino-5-carboxyadamantane mide (0.69 g, 61%) as white solid: LC/MS (Table 2, Method d) R-1.03 min: MS m/z 153 (M-H). Preparation #24 1-(5-Tosyl-5H-pyrrolo2,3-bipyrazin-2-yl)etha HCI namine hydrochloride 10

NH2 oHCI

O Y 15 O HO

To E-2-amino-5-carboxyadamantane methyl ester hydro chloride (1.0 g, 4.1 mmol, as prepared in Org. Process Res. Dev, 2008, 12 (6), 1114-1118) and DIEA (2.13 mL, 12.2 mmol) in 1,4-dioxane (15 mL) was added AcO (0.576 mL, 6.10 mmol). The reaction was stirred at about 25°C. for about 3 h before the addition of aqueous NaOH (2N, 8.14 mL, 16.3 25 mmol). The reaction was stirred at about 25°C. for about 16 h before it was partitioned between EtOAc (100 mL) and aqueous 1 NHCl (50 mL). The organic layer was washed with Step A: brine (50 mL), dried over anhydrous NaSO filtered, and 1-(5-Tosyl-5H-pyrrolo2,3-bipyrazin-2-yl)ethanol concentrated under reduced pressure to give 2-acetylamino 30 5-carboxyadamantane (0.47g, 4.9%) as a white solid: LC/MS (Table 2, Method a) R-1.43 min: MS m/z: 236 (M-H). Preparation #23 35 6-Fluoro-4-methylnicotinamide

40

OH 45 OH

C 50

55 A round bottom flask was charged with 6-fluoro-4-meth ylnicotinic acid (1.13 g, 7.28 mmol. Frontier) and DCM (73 mL) to give a clear solution. Thionyl chloride (5.32 mL, 72.8 mmol) was added drop-wise and the mixture was stirred at room temperature overnight. The reaction mixture was con 60 To a solution of methylmagnesium chloride (0.232 mL, centrated to dryness under reduced pressure and the residue 0.697 mmol) in THF (10 mL) at about -78°C. was added a was dissolved in EtOAc (10 mL) and added drop-wise to a solution of 5-tosyl-5H-pyrrolo2,3-bipyrazine-2-carbalde rapidly stirred mixture of EtOAc (40 mL) and concentrated hyde (0.210 g, 0.697 mmol. Example #10, Step B) in DCM aqueous NH-OH (36.9 ml, 947 mmol). The mixture was (10.0 mL). After about 10 min saturated aqueous NHCl was stirred for about 1 h, and the layers were separated. The 65 added to the reaction mixture. After warming to room tem aqueous layer was further extracted with EtOAc (50 mL) and perature, EtOAc (30 mL) was added to the reaction mixture the combined extracts were washed with brine, dried over and the organic layer was separated, dried over anhydrous US 9,365,579 B2 137 138 Na2SO, filtered, and concentrated under reduced pressure. Step C: 1-(5-Tosyl-5H-pyrrolo2,3-bipyrazin-2-yl) The crude material was purified by silica gel chromatography ethanamine hydrochloride eluting with 20-80% EtOAc/heptane to provide 1-(5-tosyl 5H-pyrrolo2,3-bipyrazin-2-yl)ethanol (0.050 g, 23%) as a 5 yellow oil. LC/MS (Table 2, Methoda) R-2.04 min: MS m/z. 318 (M+H)". 2N eN N-\ Step B: 10 N2 2NN 2-(1-Azidoethyl)-5-tosyl-5H-pyrrolo2,3-bipyrazine N O2A 20 --

15 tryN 4. N N 2$1\o -- HCI 4. N

25 -2'

30

N & S N To a solution of 2-(1-azidoethyl)-5-tosyl-5H-pyrrolo2,3- bipyrazine (0.65 g, 1.9 mmol) in THF (10 mL) and water (5 y 35 mL) was added triphenylphosphine (0.598 g, 2.28 mmol). 4. The reaction mixture was heated to about 45° C. and after about 12h the reaction mixture was cooled to room tempera 22' ture and concentrated under reduced pressure. The residue was dissolved in EtOAc (40 mL) and HCl gas was passed 40 through the solution until pH of 1. EtO (40 mL) was slowly added and the solvent was decanted away from the resulting solid. The solid was dried under vacuum to provide 1-(5- tosyl-5H-pyrrolo2,3-bipyrazin-2-yl)ethanamine hydrochlo ride (0.65 g, 97%) as a tan solid: LC/MS (Table 2, Methoda) 45 R–1.56 min; MS m/z. 317 (M+H)". To a solution of 1-(5-tosyl-5H-pyrrolo2,3-bipyrazin-2-yl) Preparation #25 ethanol (0.600 g, 1.89 mmol) in DCM (10 mL) was added 2,2-Dimethyl-4-OXocyclopentanecarboxylic acid SOCl (0.690 mL, 9.45 mmol) at ambient temperature. After 50 about 4 h the reaction mixture was diluted with EtOAc (50 mL) and saturated aqueous NaHCO (50 mL) was added to the reaction mixture. After gas evolution ceased, the organic layer was separated, dried over anhydrous NaSO filtered, 55 and concentrated under reduced pressure. The residue was dissolved in DMF (10 mL) and sodium azide (0.615 g, 9.45 mmol) was added to the reaction mixture. After about 15 h. OH EtOAc (50 mL) and water (50 mL) were added to the reaction 60 mixture. The organic layer was separated, concentrated under reduced pressure, and purified by silica gel chromatography To a solution of 4.4-dimethylcyclopent-2-enone (2.0 g, 18 eluting with 20-80% EtOAc/heptane to provide 2-(1-azido mmol) in EtOH (50 mL), water (7.5 mL) and AcOH (1.5 mL) ethyl)-5-tosyl-5H-pyrrolo2,3-bipyrazine (0.65g, 100%) as a was added potassium cyanide (2.36 g., 36.3 mmol). The reac 65 tion mixture was heated to about 40° C. and after about 15 h colorless solid: LC/MS (Table 2, Methoda) R-2.67 min: MS the reaction mixture was concentrated under reduced pres m/Z. 343 (M+H)". sure. The residue was diluted with EtOAc (50 mL) and US 9,365,579 B2 139 140 washed with brine. The organic layer was separated, dried -continued over anhydrous NaSO, filtered, and concentrated. The resi O due was dissolved in aqueous HCl (6N, 50 mL) and heated to reflux. After about 3 days the reaction mixture was cooled to room temperature and concentrated under reduced pressure - O)-6- X- O to provide 2,2-dimethyl-4-oxocyclopentanecarboxylic acid (3.7g, 90%, -70% purity by "H NMR) that was carried on without additional purification: LC/MS (Table 2, Method a) ( X R=1.30 min: MS m/z: 155 (M-H). 10 To a solution of 4-(methoxycarbonyl)bicyclo[2.2.1]hep Preparation #26 tane-1-carboxylic acid (2.01 g, 10.1 mmol) in toluene (30 mL) was added diphenyl phosphoryl azide (2.20 mL, 10.2 mmol) and TEA (1.60 mL, 11.5 mmol). The mixture was 4-(tert-Butoxycarbonylamino)bicyclo[2.2.1]heptane stirred at room temperature for about 1 h followed by heating 1-carboxylic acid 15 at about 50° C. for about 3 hand further heating at about 70° C. for about 2h. The reaction was cooled to room temperature and concentrated to dryness under reduced pressure. The residue was diluted in tert-butanol (10.0 mL. 105 mmol) and the mixture was heated at about 80° C. for about 16 h. The reaction mixture was cooled to room temperature and dis )-6- solved in EtO (50 mL). The organic layer was washed with water, aqueous 1 M NaOH, water, and brine (25 mL each). The organic layer was dried over anhydrous NaSO, filtered, X 25 and concentrated to provide methyl 4-(tert-butoxycarbony lamino)bicyclo[2.2.1-heptane-1-carboxylate as an off-white solid (2.22 g, 81%): "H NMR (400 MHz, DMSO-d) & 7.03 Step A: 4-(Methoxycarbonyl)bicyclo[2.2.1]heptane (s, 1H), 3.59 (s.3H), 1.95-1.74 (m, 6H), 1.60 (s, 4H), 1.37 (s, 1-carboxylic acid 9H). 30 Step C. 4-(tert-Butoxycarbonylamino)bicyclo[2.2.1 heptane-1-carboxylic acid

O

- O)-6- X- He 40 O To a solution of dimethylbicyclo[2.2.1]heptane-1,4-dicar boxylate (2.00 g, 9.44 mmol, as prepared in Aust. J. Chem., 1985,38, 1705-18) in MeOH (47 mL) was added KOH (0.475 g, 8.46 mmol) and water (2.5 mL). The reaction was stirred at 45 reflux for about 16 hand then cooled to room temperature and se concentrated to dryness under reduced pressure. Water (25 HO ) mL) was added to the remaining residue and the mixture was extracted with EtO (2x25 mL). The aqueous layer was acidi fied to about pH 4 using aqueous 6 NHCl and was extracted 50 with DCM (3x20 mL). The combined DCM extracts were To a solution of methyl 4-(tert-butoxycarbonylamino)bi dried over anhydrous MgSO, filtered, and concentrated to cyclo2.2.1]heptane-1-carboxylate (2.21 g, 8.20 mmol) in provide 4-(methoxycarbonyl)bicyclo[2.2.1]heptane-1-car THF (27 mL) and MeOH (14 mL) was added aqueous NaOH boxylic acid as an off-white solid (1.19 g, 71%): 'H NMR (1 N. 20.0 mL. 20.0 mmol). The mixture was stirred at room (400 MHz, DMSO-d) & 12.19 (s, 1H), 3.61 (s.3H), 1.92 (d. 55 temperature for about 16 hand concentrated to dryness under J=6.6 Hz, 4H), 1.76 (s. 2H), 1.65-1.54 (m, 4H). reduced pressure. Water (25 mL) was added to the remaining residue and the mixture was extracted with EtO (2x25 mL) Step B: Methyl 4-(tert-butoxycarbonylamino)bicyclo and the EtO extracts were discarded. The aqueous layer was 2.2.1]heptane-1-carboxylate 60 acidified to about pH 4 using aqueous 6 NHCl and extracted with EtO (3x10 mL). The combined organic layers were dried over anhydrous MgSO filtered, and concentrated to dryness under reduced pressure to provide 4-(tert-butoxycar O O bonylamino)bicyclo2.2.1]heptane-1-carboxylic acid as an 65 off-white solid (1.69 g, 81%): 'H NMR (400 MHz, DMSO - O OH d) & 12.07 (s, 1H), 7.00 (s, 1H), 2.00-1.69 (m, 6H), 1.67-1.45 (m, 4H), 1.37 (s, 9H). US 9,365,579 B2 141 142 Preparation #27 -continued N 6-Chloro-4-(trifluoromethyl)nicotinamide 4N \29 HCI 2S

10

OH A 5-L reactor was charged with 2-bromo-5-tosyl-5H-pyr 15 rolo2,3-bipyrazine (98.8 g. 281 mmol, Preparation #7), Zinc dust (3.50 g, 53.3 mmol), palladium(II) trifluroacetate (4.0g, 12 mmol), and racemic-2-(di-t-butylphophino)-1,1'-bi napthyl (9.8 g. 24.7 mmol). The flask was equipped with a powder addition device into which zinc cyanide (10.0 g, 157 mmol) was placed to be added at a later step. The vessel was 6-Chloro-4-(trifluoromethyl)nicotinic acid (1.0 g, 4.4 purged with argon for no longer than about 30 min and then argon sparged DMA (2 L) was added to the reactor. The mmol, Oakwood) was dissolved in DCM (44 mL) to give a mixture was stirred and heated to about 50° C. while main clear solution. SOCl (3.2 mL, 44 mmol) was added drop taining an argon sparge. The resulting dark brown Solution wise and the reaction mixture was stirred at room temperature 25 was further heated to about 95°C. while adding the zinc overnight and then at reflux for about 16 h. The mixture was cyanide, from the powder addition device, portion-wise over concentrated under reduced pressure to give a yellow oil that about 15 min. Upon reaching about 95°C., the brown mixture was dissolved into EtOAc (10 mL). The solution was added is stirred for about an additional 16 h. The reaction mixture drop-wise to a rapidly stirred mixture of EtOAc (20 mL) and was cooled to room temperature, resulting in the precipitation concentrated aqueous NH-OH (22 mL, 580 mmol). The 30 of salts. The mixture was filtered through a Buchner funnel resulting cloudy mixture was stirred for about 2 hand sepa containing filter-aid and the filter cake was washed with rated. The aqueous layer was further extracted with EtOAC DMA (20 mL). A solution of the crude product in DMA was (30 mL). The combined organic extracts were washed with added to cold (<10°C.) water (16 L) and stirred for about 30 brine, dried over anhydrous NaSO, filtered, and concen min. The resulting Suspension was filtered and the filter cake trated under reduced pressure to give 6-chloro-4-(trifluorom 35 was rinsed again with water (1 L). The resulting wet cake was ethyl)nicotinamide (0.85g. 85%) as off-white solid: LC/MS dried in a vacuum oven at about 50° C. The crude solid was (Table 2, Method a) R-1.62 min: MS m/z. 223 (M+H)". dissolved in DCM (1.5 L) and further dried over anhydrous MgSO4. After filtration, the solution was passed through a Preparation #28 pad of silica (140 g), washing with additional solvent until 40 only predominantly impurities were detected eluting off the (5-Tosyl-5H-pyrrolo2,3-bipyrazin-2-yl)methanamine pad. The solvent was removed and the crude solid was tritu hydrochloride rated with MeOH/DCM (4:1, 10 volumes of solvent per gram of crude solid) at ambient temperature for about 5h. The solid was filtered and washed with MeOH (300 mL). The product 45 was dried in a vacuum oven to provide 5-tosyl-5H-pyrrolo2, Br N 3-bipyrazine-2-carbonitrile (58.8 g. 70%) as a colorless solid: "H NMR (400 MHz, CDC1,) 88.67 (s, 1H),8.21 (d.J–4.2 Hz, Cy 1H), 8.07 (d. J=8.4 Hz, 2H), 7.34 (d. J=8.1 Hz, 2H), 6.89 (d. 2NN J=4.2 Hz, 1H), 2.42 (s.3 H). A 2-L316-stainless steel pressure N V 4' s 50 reactor was charged with 5% Pd/C (15.4 g of 63.6 wt % water wet material, 5.6 g dry basis, Johnson Matthey A503032-5), 5-tosyl-5H-pyrrolo2,3-bipyrazine-2-carbonitrile (55 g, 184 mmol), THF (1.1 L), deionized water (165 mL), aqueous HCl, (37 wt %, 30 mL,369 mmol) and quinoline (1.1 mL, 9.0 55 mmol). The vessel was purged, pressurized, and maintained at 40 psi with hydrogen Supplied from a high pressure reser NC N voir. The mixture was vigorously agitated at about 25° C. After about 5 h the reactor was vented and purged with 2NN nitrogen to remove most of the dissolved hydrogen, and the N Ye -- 60 reaction mixture was filtered to remove the catalyst. The o2 reactor and catalyst cake were rinsed with THF:HO (1:1, 2x40 mL). The combined filtrate and rinses were concen trated and EtOH (500 mL) was added. After two additional solvent switches with EtOH (2x500 mL), the crude residue 65 was concentrated to give a residue (76 g) that was suspended in EtOH (550 mL) and stirred at ambient temperature for about 4 h. The solid was collected by filtration and washed US 9,365,579 B2 143 144 with cold EtOH (50 mL). The wet cake was dried in a vacuum -continued oven to provide (5-tosyl-5H-pyrrolo2,3-bipyrazin-2-yl) O methanamine hydrochloride (51.2 g, 82%) as a colorless solid: LC/MS (Table 2, Methoda) R-1.44 min: MS m/z:303 (M+H)". General Procedure A: Formation of a Hydrazide from a Car boxylic Acid To mixture of a 2-hydrazinylpyrrolo2,3-bipyrazine (pref erably 1 equiv) and a carboxylic acid (1-2 equiv, preferably 10 1.1-1.3 equiv) in a solvent such as DCM or THF, preferably DCM, is added a coupling agent such as EDC.HCl or HATU (1.0-2.0 equiv, preferably 1.2-1.6 equiv) with or without an organic base such as TEA or DIEA (2-5 equiv, preferably 3-4 15 equiv). After about 1-72 h (preferably 2-6 h) at about 20-60° C. (preferably about room temperature), the reaction is worked up using one of the following methods. Method 1: To mixture of 2-hydrazinyl-5-tosyl-5H-pyrrolo2,3-b Water is added and the layers are separated. Optionally, the pyrazine (2.50 g, 8.24 mmol. Preparation #9) and (1R,3S)-3- mixture may be filtered through Celite(R) prior to the separa (tert-butoxycarbonylamino)cyclopentanecarboxylic acid (2.08 g., 9.07 mmol, Peptech) in DCM (30 mL) was added tion of the layers. The aqueous layer is then extracted with an EDC.HCl (1.90 g, 9.89 mmol). After about 4.5 h at ambient organic solvent such as EtOAc or DCM. The combined temperature, water (30 mL) was added and the layers were organic layers are optionally washed with brine, dried over 25 separated. The aqueous layer was then extracted with EtOAc anhydrous NaSO or MgSO, filtered or decanted, and con (15 mL). The combined organic layers were washed with centrated under reduced pressure. Method 2: The reaction is brine, dried over anhydrous MgSO, filtered, and concen diluted with an organic solvent such as EtOAc or DCM and is trated under reduced pressure. The crude material was dis solved in DCM (15 mL) and purified by silica gel chroma washed with either water or brine or both. The aqueous layer 30 tography eluting with a gradient of 40-100% EtOAc in is optionally further extracted with an organic solvent Such as heptane to give tert-butyl (1S,3R)-3-(2-(5-tosyl-5H-pyrrolo EtOAc or DCM. Then the organic layer or combined organic 2,3-bipyrazin-2-yl)hydrazinecarbonyl)cyclopentylcarbam layers are optionally washed with brine, dried over anhydrous ate (4.20 g, 97%): LC/MS (Table 2, Methoda) R-2.27 min: NaSO or MgSO filtered or decanted, and concentrated 35 MS m/z. 515 (M+H)". under reduced pressure. Method 3: The reaction is diluted General Procedure B: Formation of a Hydrazide from an Acid with an organic solvent such as EtOAc or DCM and water is Chloride Followed by Cyclization and Sulfonamide Hydroly S1S added. The layers are separated and the organic layer is To a solution of 5-sulfonyl-2-hydrazinyl-5H-pyrrolo2,3- directly purified by chromatography. In all cases, the crude 40 bipyrazine (preferably 1 equiv) and TEA or DIEA (1-10 material is optionally purified by precipitation, crystalliza equiv, preferably 4 equiv) in 1,4-dioxane at about 0-25°C. tion, and/or trituration from an appropriate solvent or solvents (preferably ambient temperature) is added an acid chloride and/or by chromatography to give the target compound. (1-1.5 equiv, preferably 1 equiv). After the complete addition, Illustration of General Procedure A the reaction is allowed to warm to ambient temperature if 45 cooled initially. After about 0.5-2 h (preferably about 1 h), Preparation #A.1 SOC1 (1-10 equiv, preferably 3 equiv) is added and the reac tion is heated at about 60-100° C. (preferably about 80-90° tert-Butyl (1S,3R)-3-(2-(5-tosyl-5H-pyrrolo2,3-b C.) for about 0.25-8 h (preferably about 1 h). The reaction is pyrazin-2-yl)hydrazinecarbonyl)cyclopentylcarbam allowed to cool to ambient temperature and thenaqueous base ate 50 (such as aqueous NaCO or aqueous NaOH, preferably aqueous NaOH) is added followed by the optional, but not preferable, addition of MeOH (5-50% of the reaction volume, preferably 50%). The reaction is heated at about 50-90° C. for about 1-96 h (preferably about 3 hat about 60° C. if using t 55 HN N aqueous NaOH or about 3 days at about 90° C. if using aqueous NaCO). The reaction is concentrated under Cy reduced pressure and then is partitioned between an organic 4. N solvent (such as EtOAc or DCM, preferably EtOAc) and 60 water, saturated aqueous NaHCO, and/or brine, preferably ca saturated aqueous NaHCO. The organic layer is separated and optionally washed with water and/or brine, dried over anhydrous NaSO or MgSO, filtered or decanted, and con centrated under reduced pressure. The crude material is 65 optionally purified by precipitation, crystallization, and/or trituration from an appropriate solvent or solvents and/or by chromatography to give the target compound. US 9,365,579 B2 145 146 Illustration of General Procedure B crude material is optionally purified by precipitation, crystal lization, and/or trituration from an appropriate solvent or Example iB.1.1 Solvents and/or by chromatography to give the target com pound. 1-(2-Methylcyclohexyl)-6H-pyrrolo2,3-e12.4 5 Illustration of General Procedure C triazolo 4.3-alpyrazine Preparation #C.1 H N N tert-Butyl-(1S,3R)-3-(6-tosyl-6H-pyrrolo2,3-e 1.2, roy 10 4triazolo 4,3-alpyrazin-1-yl)cyclopentylcarbamate 2 N N OS SSo

15

To a solution of 5-(4-tert-butylphenylsulfonyl)-2-hydrazi nyl-5H-pyrrolo2,3-bipyrazine (0.40 g, 1.2 mmol, Prepara tion #3) and DIEA (0.20 mL, 1.2 mmol) in 1,4-dioxane (12 mL) at about 0°C. was added 2-methylcyclohexanecarbonyl chloride (0.19 g, 1.2 mmol, Preparation #4). After the com 25 plete addition, the ice bath was removed and the reaction was allowed to warm to ambient temperature. After about 1 h, SOCl (0.42 mL, 5.8 mmol) was added and the reaction was heated at about 90° C. for about 1 h. The reaction was allowed to cool to ambient temperature and then 2 Maqueous NaCO 30 (2N, 11.6 mL, 23.2 mmol) and MeOH (12 mL) were added. The reaction was heated at about 90° C. for about 3 days. The reaction was concentrated under reduced pressure and then partitioned between EtOAc (50 mL) and saturated aqueous NaHCOs (40 mL). The organic layer was separated and dried 35 over anhydrous NaSO filtered and the solvent was concen trated under reduced pressure. The residue was purified over silica gel (12 g) using EtOAC as the eluent and then further purified by RP-HPLC (Table 2, Method b). The combined product-containing fractions were concentrated under reduced pressure to remove the MeCN and the resulting pre 40 cipitate was collected by vacuum filtration to afford 1-(2- methylcyclohexyl)-6H-pyrrolo2,3-e12.4 triazolo4.3-a pyrazine as a white solid (0.10 g, 35%). LC/MS (Table 2, To a solution of tert-butyl (1S,3R)-3-(2-(5-tosyl-5H-pyr Method a) R-1.84 min: MS m/z: 256 (M+H)". rolo2,3-bipyrazin-2-yl)hydrazinecarbonyl)cyclopentylcar General Procedure C: Cyclization of a Hydrazide 45 bamate (9.30 g. 18.1 mmol. Preparation if A.1) in 1,4-dioxane To a solution of a 2-hydrazinyl-5H-pyrrolo2,3-bipyrazine (100 mL) was added TEA (10.0 mL, 72.3 mmol) and SOCl, (preferably 1 equiv) in an organic solvent (for example 1,4- (2.11 mL, 28.9 mmol). The mixture was heated at about 80° dioxane) is added a base such as TEA or DIEA (1-5 equiv, C. for about 1.5 h. The reaction mixture was cooled to ambi preferably 2-4 equiv) and SOCl (1-5 equiv, preferably 1-2 ent temperature, EtOAc and water (200 mL each) were added, equiv). The mixture is heated at about 60-100°C. (preferably 50 and the layers were separated. The aqueous Solution was about 80° C.) for about 1-16 h (preferably about 1-2 h). The extracted with EtOAc (2x100 mL) and the combined organic reaction mixture is cooled to ambient temperature and layers were washed with saturated aqueous NaHCO, and worked up using one of the following methods. Method 1: An brine (100 mL each). The organic extracts were dried over organic solvent (such as EtOAc or DCM) and water are anhydrous NaSO filtered, and concentrated under reduced added. The layers are separated and the aqueous layer is pressure. The crude material was purified by silica gel chro optionally extracted with additional organic solvent. The 55 matography eluting with a gradient of 25-100% EtOAc in combined organic layers may be optionally washed with DCM to give tert-Butyl-(1S,3R)-3-(6-tosyl-6H-pyrrolo2,3- aqueous base (such as NaHCO) and/or brine, dried over e1.2.4 triazolo 4.3-alpyrazin-1-yl)cyclopentylcarbamate anhydrous NaSO or MgSO, then decanted or filtered prior (7.65g. 85%): LC/MS (Table 2, Methoda) R-2.37 min: MS to concentrating under reduced pressure. Method 2: An m/z: 497 (M+H)". organic solvent (such as EtOAc or DCM) is added and the 60 General Procedure D: Cyclization of a Hydrazide Followed organic layer is optionally washed with brine or water, dried by Sulfonamide Hydrolysis and Boc-Deprotection over anhydrous MgSO or NaSO filtered or decanted, and A round-bottomed flask is charged with a 5-sulfonyl-2- concentrated under reduced pressure. Method 3: The reaction hydrazinyl-5H-pyrrolo2,3-bipyrazine (preferably 1 equiv), mixture is partitioned between an organic solvent (Such as an organic solvent (such as 1,4-dioxane or THF, preferably EtOAc or DCM) and saturated aqueous NaHCO or brine, 65 1,4-dioxane), SOCl (2-5 equiv, preferably 2 equiv) and an dried over anhydrous NaSO or MgSO, then decanted or organic base such as DIEA or TEA (0-5 equiv, preferably 3 filtered prior to concentrating under reduced pressure. The equiv). The resulting mixture is stirred at about 25-120° C. US 9,365,579 B2 147 148 (preferably about 90° C.) for about 0.25-5h (preferably about -continued 1 h) and then allowed to cool to ambient temperature. To the reaction mixture is added an aqueous base (such as aqueous NaCO or aqueous NaOH, 1-30 equiv, preferably 1-2 equiv oHCI for aqueous NaOH, preferably 15-20 equiv for aqueous s NaCO) and the resulting mixture is heated at about 60-120° C. (preferably about 90° C.) for about 1-10 h (preferably about 5 h) then allowed to cool to ambient temperature. MeOH (5-50% of the reaction volume, preferably 20-30%) is added to the reaction mixture and the resulting solution is heated at about 60-120° C. (preferably about 90° C.) for about 10 5-24 h (preferably about 16 h) and then allowed to cool to ambient temperature. The layers are separated and the organic solvent is concentrated under reduced pressure. To the residue is added an organic solvent (Such as 1,4-dioxane or THF, preferably 1,4-dioxane) followed by a solution of 15 HCl, such as 4M HCl in 1,4-dioxane (20-40 equiv, preferably A round-bottomed flask was charged with cis-tert-butyl-4- 25 equiv). The resulting suspension is stirred at about 20-80° (2-(5-tosyl-5H-pyrrolo2,3-bipyrazin-2-yl)hydrazinecarbo C. (preferably about 60° C.) for about 1-16 h (preferably nyl)cyclohexylcarbamate (0.415 g, 0.785 mmol, prepared about 1 h) and then allowed to cool to ambient temperature. using. A from cis-4-(tert-butoxycarbonylamino)cyclohexan The solid is collected by vacuum filtration, washed with organic solvent (such as 1,4-dioxane, EtOAc and/or EtO. ecarboxylic acid AMRI and Preparation #9), 1,4-dioxane (9 preferably 1,4-dioxane followed by EtO) to yield the crude mL) and SOC1 (0.115 mL, 1.57 mmol). The resulting mix product as an HCl salt. The crude material is optionally puri ture was heated at about 90° C. for about 1 hand then allowed fied by precipitation, crystallization, or trituration from an to cool to ambient temperature. To the reaction mixture was appropriate solvent or solvents or by chromatography to give 25 the target compound. added aqueous NaCO (5 N, 7.85 mL, 15.7 mmol) and the Illustration of General Procedure D reaction mixture was heated at about 90° C. for about 5 h. Example iD.1.1 MeOH (5 mL) was added to the reaction mixture and the

cis-4-(6H-pyrrolo2,3-e12.4 triazolo 4.3-a 30 resulting mixture was heated at about 90° C. for about 16 h pyrazin-1-yl)cyclohexanamine hydrochloride and and then allowed to cool to ambient temperature. The layers cis-4-(6H-pyrrolo2,3-e 1.2.4 triazolo 4.3-a pyrazin-1-yl)cyclohexanamine were separated and the organic layer was concentrated under O reduced pressure. To the residue was added 1,4-dioxane (10 mL) followed by HCl (4M in 1,4-dioxane, 5 mL, 20.0 mmol). 35 The resulting suspension was heated at about 60°C. for about HNt N 1 hand then allowed to cool to ambient temperature. The solid was collected by vacuum filtration, washed first with 1,4- HN Cy2 dioxane (1 mL) then EtO (50 mL) to yield the crude product cis-4-(6H-pyrrolo2,3-e 1.2.4 triazolo 4,3-alpyrazin-1-yl) 9 so as22' . . cyclohexanamine hydrochloride (0.42 g, 98%, 84% purity). A portion of the crude HCl salt (0.075 g) was further purified by RP-HPLC (Table 2, Method g) to give cis-4-(6H-pyrrolo 45 2,3-e12.4 triazolo 4.3-alpyrazin-1-yl)cyclohexanamine (0.044 g) with 3 equiv NHOAc as an excipient. LC/MS (Table 2, Method a) R, 0.92 min: MS m/z: 257 (M+H)". TABLE D1 Examples prepared using General Procedure D: Rmin (Table 2, m/z ESI+ Hydrazide Product Example # Method) (M+H)" (1R,3S)-3-(6H- D.1.2 0.47 (d) 243 Pyrrolo[2,3- yl)hydrazinecarbonyl)cyclopentylcarbamate e1,2,4-triazolo4,3- (prepared using A from (1S,3R)-3- apyrazin-1-yl)cyclo (tert pentanamine butoxycarbonylamino)cyclopentanecarboxylic hydrochloride acid PepTech and Preparation tert-Butyl trans-4-(2-(5-tosyl-5H trans-4-(6H- D.13 0.44 (d) 257 pyrrolo[2,3-bipyrazin-2- Pyrrolo[2,3- yl)hydrazinecarbonyl)cyclohexylcarbamate e1,2,4-triazolo4,3- (prepared using A from trans-4- apyrazin-1-yl)cyclo (tert hexanamine US 9,365,579 B2 149 150 TABLE D.1-continued Examples prepared using General Procedure D. R. min (Table 2, m/z ESI+ Hydrazide Product Example # Method) (M+H)" butoxycarbonylamino)cyclohexanecarboxylic hydrochloride acid AMRI and Preparation #9) tert-Butyl (1R,3R)-3-(2-(5-(4-tert D.1.4 0.46 (d) 243 butylphenylsulfonyl)-5H-pyrrolo[2,3- Pyrrolo[2,3- bipyrazin-2- e1,2,4-triazolo4,3- yl)hydrazinecarbonyl)cyclopentylcarbamate a pyrazin-1-yl)cyclo (prepared using A from (1S,3S)-3- pentanamine (tert- hydrochloride butoxycarbonylamino)cyclopentanecarboxylic acid Acros and Preparation #3)

General Procedure E: Cyclization of a Hydrazide Followed Illustration of General Procedure E by Sulfonamide Hydrolysis To a solution of a 5-sulfonyl-2-hydrazinyl-5H-pyrrolo2, Example #E.1 3-bipyrazine (preferably 1 equiv) in a solvent Such as 1,4- dioxane is added SOC1 (1-5 equiv, preferably 1-2 equiv). tert-Butyl (1S,3R)-3-(6H-pyrrolo2,3-e 1.2.4 tria Optionally, an organic base, such as TEA or DIEA, (1-5 equiv, preferably 2-4 equiv) is added before SOCl, particu Zolo 4.3-alpyrazin-1-yl)cyclopentylcarbamate larly for Boc-protected substrates. The reaction is heated at 25 about 60-100° C. (preferably about 80° C.). After about 0.5-6 h (preferably about 1-2 h), an aqueous base (such as aqueous NaCO or aqueous NaOH, 1-90 equiv, preferably 15-20 equiv for aqueous Na2CO or 1-2 equiv for aqueous NaOH), is added and heating is resumed at about 60-90° C. (prefer ably about 80°C.) for about 1-72 h (preferably about 1-16h). 30 Optionally, but not preferably, the reaction is cooled to ambi N s ent temperature for a period of time (5 min-72 h), during 22' which time MeOH and/or additional aqueous base (such as saturated NaCO or 1 N NaOH) may be added, and heating is optionally resumed at about 60-90° C. (preferably about 35 80°C.) for about 1-72 h (preferably about 1-16 h). This cycle of optionally cooling to ambient temperature and adding base may occur up to four times. The reaction is worked up using one of the following methods. Method 1: An organic solvent such as EtOAc or DCM is added with the optional addition of 40 water, brine, or saturated aqueous NHCl (preferably water) Y and the layers are separated. The aqueous layer is then option ally extracted with additional organic solvent such as EtOAc or DCM. The combined organic layers are optionally washed NS with brine or water, dried over anhydrous MgSO or NaSO, 45 ps, filtered or decanted, and concentrated under reduced pres N N -e- sure. Method 2: The reaction mixture is decanted and the insoluble material is washed with an organic solvent Such as EtOAc. The combined organic layers are concentrated under reduced pressure. Method 3: The reaction mixture is concen trated under reduced pressure to remove solvent. Water is 50 O) added and the aqueous layer is extracted with an organic 22' solvent such as EtOAc or DCM. The combined organic layers are optionally washed with brine or water, dried over anhy drous MgSO or NaSO filtered or decanted, and concen trated under reduced pressure. Method 4: A reaction mixture 55 containing a precipitate may be filtered to collect the target compound, while optionally washing with water. The filtrate may be optionally concentrated and purified to yield addi tional target compound. Method 5: The reaction mixture is adjusted to neutral pH with the addition of a suitable aqueous acid (such as aqueous HCl) prior to extraction with an organic 60 solvent such as EtOAc or DCM. The combined organic layers are optionally washed with brine or water, dried over anhy drous MgSO or NaSO filtered or decanted, and concen trated under reduced pressure. In all cases, the crude material is optionally purified by precipitation, crystallization, and/or 65 trituration from an appropriate solvent or solvents and/or by chromatography to give the target compound. US 9,365,579 B2 151 152 To a solution of tert-butyl (1S,3R)-3-(2-(5-tosyl-5H-pyr General Procedure F: Cyclization of a Hydrazide with Loss of rolo2,3-bipyrazin-2-yl)hydrazinecarbonyl)cyclopentylcar Boc-Protecting Group Followed by Sulfonamide Hydrolysis bamate (4.73 g, 9.19 mmol. Preparation if A.1) in 1,4-dioxane To a solution of a 5-sulfonyl-2-hydrazinyl-5H-pyrrolo2, (50 mL) was added TEA (5.10 mL, 36.8 mmol) and SOCl, 3-bipyrazine (preferably 1 equiv) and TEA or DIEA (0-6 (1.34 mL, 18.4 mmol). The reaction mixture was heated at equiv, preferably 1 equiv) in 1,4-dioxane is added SOCl. about 80° C. After about 1.5 h, saturated aqueous NaCO (2.0-6.0 equiv, preferably 3 equiv). The reaction is heated at (100 mL) was added and heating was resumed at about 80°C. about 60-120° C. (preferably about 80-90° C.) for about 1-8h for about 6h. The reaction was cooled to ambient temperature 10 (preferably about 1-4 h). The reaction is allowed to cool to for about 3 days and then heated at about 80°C. for about 16 ambient temperature then is optionally, but not preferably, h. Water and EtOAc (100 mL each) were added and the layers diluted with a cosolvent (such as MeCH or EtOH, preferably were separated. The aqueous layer was then extracted with MeOH) by 5-50% of the reaction volume (preferably 50%). additional EtOAc (2x100 mL). The combined organic layers 15 An aqueous base (such as aqueous NaCO or aqueous were washed with brine, dried over anhydrous NaSO fil NaOH, 1-30 equiv, preferably 1-2 equiv for aqueous NaOH, tered, and concentrated under reduced pressure. The crude preferably 15-20 equiv for aqueous NaCO) is added and the solid was triturated with petroleum ether (b.p. 30-60° C.; 30 reaction is heated at about 40-90° C. (preferably about 60°C.) mL) and collected by vacuum filtration, while washing with for about 1-24 h (preferably about 2 h) before it is concen additional petroleum ether (b.p. 30-60° C.; 20 mL), to give trated under reduced pressure. The crude material is option tert-butyl (1S,3R)-3-(6H-pyrrolo2,3-e 1.2.4 triazolo 4.3- ally purified by precipitation, precipitation by Salt formation, apyrazin-1-yl)cyclopentylcarbamate as a light brown Solid crystallization, and/or trituration from an appropriate solvent 25 (2.86g, 86%): LC/MS (Table 2, Methoda) R-1.75 min: MS or solvents and/or by chromatography to give the target com m/Z. 343 (M+H)". pound. TABLE E.1

Examples prepared using General Procedure E

Rmin (Table 2, m/z ESI+ Hydrazide Product Exii Method) (M+H)"

Adamantane-2-carboxylic acid N- 1-Adamantan-2-yl- E.1.1 2.09 (a) 294 5-(4-tert-butyl-benzenesulfonyl)- 6H-pyrrolo[2,3- 5 H-pyrrolo[2,3-bipyrazin-2-yl)- e1,2,4-triazolo4,3- hydrazide (prepared using A from apyrazine Preparation #3 and adamantane-2- carboxylic acid Enamine) Adamantane-1-carboxylic acid N- 1-Adamantan-1-yl- E.1.2 2.01 (a) 294 5-(4-tert-butyl-benzenesulfonyl)- 6H-pyrrolo[2,3- 5 H-pyrrolo[2,3-bipyrazin-2-yl)- e1,2,4-triazolo4,3- hydrazide (prepared using A from apyrazine Preparation #3 and adamantane-1- carboxylic acid, EDCHCl, and TEA) Benzyl (1S,3S)-3-(2-(5-tosyl-5H- Benzyl (1S,3S)-3- E.13 1.85 (a) 363 pyrrolo[2,3-bipyrazin-2- (6H-pyrrolo[2,3- yl)hydrazinecarbonyl)cyclobutylcarbamate, e124triazolo4,3- (prepared using Q from 3- apyrazin-1- aminocyclobutanecarboxylic acid yl)cyclobutylcarbamate hydrochloride (Enamine) and A from Preparation #9) 4-Methoxy-N'-(5-tosyl-5H- 1-(4- E.1.4 1.56 (a) 272 pyrrolo[2,3-bipyrazin-2- Methoxycyclohexyl)- yl)cyclohexanecarbohydrazide, 6H-pyrrolo[2,3- (prepared using A from 4 e1,2,4-triazolo4,3- methoxycyclohexanecarboxylic acid apyrazine and Preparation #9) US 9,365,579 B2 153 154 Illustration of General Procedure F To a solution of tert-butyl (1R,3R)-3-(2-(5-tosyl-5H-pyr rolo2,3-bipyrazin-2-yl)hydrazinecarbonyl)cyclopentyl)me Example #F.1.1 thylcarbamate (0.60 g, 1.1 mmol, prepared using. A from 5 (1R,3R)-3-((tert-butoxycarbonylamino)methyl)cyclopen ((1R,3R)-3-(6H-Pyrrolo2,3-e 1.2.4 triazolo 4.3-a tanecarboxylic acid (AFID) and Preparation #9) and DIEA pyrazin-1-yl)cyclopentyl)methanamine hydrochlo- (0.79 mL, 4.5 mmol) in 1,4-dioxane (5 mL) was added SOCl. ride (0.166 mL, 2.27 mmol). The reaction mixture was heated at 10 about 80° C. for about 1 h before it was allowed to cool to ambient temperature. Aqueous NaOH (2 N, 4 mL, 8 mmol) O was added to the reaction mixture and heated at about 60° C. O ls N for about 2 h. The reaction mixture was allowed to cool to X- NO N y 15 ambient temperature before it was concentrated under O NH 4. N reduced pressure. To the residue was added HCl (4N in 1,4- X- O lso Her dioxane (20 mL). The organic solution was decanted away from the resulting precipitate to afford ((1R,3R)-3-(6H-pyr 20 rolo2,3-e 1.2.4 triazolo 4,3-alpyrazin-1-yl)cyclopentyl) methanamine hydrochloride as a yellow solid (0.11 g, 33%): LC/MS (Table 2, Method a) R-1.01 min: MS m/z: 257 (M+H)". TABLE F.1

Examples prepared using General Procedure F

Rmin (Table 2, m/z ESI+ Hydrazide Product Exit Method) (M + H)

tert-Butyl-trans-3-(2-(5-(4-tert- trans-3-(6H- F.1.2 1.07 (a) 257 butylphenylsulfonyl)-5H-pyrrolo[2,3- Pyrrolo[2,3- bipyrazin-2- e1,2,4-triazolo yl)hydrazinecarbonyl)cyclohexylcarbamate 4,3-alpyrazin-1- (prepared using A from Preparation yl)cyclohexanamine #3 and trans-3-(tert-butoxycarbonyl- acetate amino)cyclohexanecarboxylic acid AMRI), EDCHCl, and TEA) tert-Butyl-cis-3-(2-(5-(4-tert- cis-3-(6H- F.1.3 1.18 (a) 257 butylphenylsulfonyl)-5H-pyrrolo[2,3- Pyrrolo[2,3- bipyrazin-2- e1,2,4-triazolo yl)hydrazinecarbonyl)cyclohexylcarbamate 4,3-alpyrazin-1- (prepared using A from Preparation yl)cyclohexanamine #3 and cis-3-(tert-butoxycarbonyl hydrochloride amino)cyclohexanecarboxylic acid AMRI), EDCHCl, and TEA)

General Procedure G: Formation of a Hydrazone Followed by -continued 55 Cyclization and Sulfonamide Hydrolysis To a solution of a 2-hydrazinyl-5-sulfonyl-5H-pyrrolo2, 3-bipyrazine (preferably 1 equiv) in an organic solvent or ()- solvents such as MeOH or MeOH/DCM (preferably MeOH) S HCI s 60 is added a solution of an aldehyde (1.0-1.3 equiv, preferably s 1.0 equiv) in an organic solvent such as DCM. The reaction N N mixture is stirred at about 15-30° C. (preferably ambient N temperature) for about 1-8 h (preferably about 2 h) before N iodobenzene diacetate (1-3 equiv, preferably 1 equiv) is N1 N 65 added. The reaction is stirred at about 15-30°C. (preferably ambient temperature) for about 15-60 min (preferably about 30 min) before it is concentrated to constant weight. To the US 9,365,579 B2 155 156 residue is added an organic Solvent Such as 1,4-dioxane, THF. -continued MeOH or EtOH (preferably 1,4-dioxane) followed by aque ous base such as aqueous NaCO or NaOH (2-50 equiv), preferably NaOH (2 equiv). The reaction was heated at about 40-80° C. (preferably about 60° C.) for about 1-24 h (prefer ably about 2 h). The crude product is optionally purified by precipitation, crystallization, and/or trituration from an appropriate solvent or solvents and/or by chromatography to give the target compound. Illustration of General Procedure G 10 N N H Example iG.1.1 To a solution of 2-hydrazinyl-5-tosyl-5H-pyrrolo2,3-b 1-(Tetrahydro-2H-pyran-4-yl)-6H-pyrrolo2,3-e12. pyrazine (0.100 g, 0.330 mmol, Preparation #9) in MeOH (2 4triazolo 4,3-alpyrazine 15 mL) was added tetrahydro-2H-pyran-4-carbaldehyde (0.038 g, 0.330 mmol, J&W PharmLab) in DCM (1 mL). The reac tion mixture was stirred at ambient temperature for about 2h before iodobenzene diacetate (0.106 g., 0.330 mmol) was added. The reaction mixture was stirred at ambient tempera N N ture for about 15 min before it was concentrated to constant weight. To the residue was added MeOH (2 mL) followed by 2NN aqueous NaOH (2 N, 0.330 mL, 0.659 mmol). The reaction N Vsa O mixture was heated at about 60° C. for about 1 h. The crude 2 reaction mixture was purified by RP-HPLC (Table 2, Method 25 f). The combined product-containing fractions were concen trated under reduced pressure to remove Mecn and then lyophilized to afford 1-(tetrahydro-2H-pyran-4-yl)-6H-pyr rolo2,3-e12.4 triazolo 4,3-alpyrazine as a white solid (0.028g,35%). LC/MS (Table 2, Methoda) R-1.25 min: MS m/z: 244 (M+H)". TABLE G.1 Examples prepared from 2-hydrazinyl-5-tosyl-5H-pyrrolo2,3-bipyrazine Preparation #9) using General Procedure Gi Rmin (Table 2, Aldehyde Product Exii Method) (M+H)" 2,6-Dimethylcyclohex-2- 1-(2,6-Dimethylcyclohex-2-enyl)- G.1.2 1.97 (a) 268 enecarbaldehyde 6H-pyrrolo[2,3- e1,2,4-triazolo4,3-alpyrazine 4-(4-Hydroxy-4- 5-(4-(6H-Pyrrolo[2,3- G.1.3 1.82 (a) 340 methylpentyl)cyclohex-3 e1,2,4-triazolo4,3-alpyrazin-1- enecarbaldehyde yl)cyclohex-1-enyl)-2- methylpentan-2-ol Bicyclo[2.2.1]hept-5-ene 1-(Bicyclo[2.2.1]hept-5-en-2-yl)- G.1.4 1.72 (a) 252 2-carbaldehyde 6H-pyrrolo[2.3 e1,2,4-triazolo4,3-alpyrazine Cyclooctanecarbaldehyde 1-Cyclooctyl-6H-pyrrolo[2,3- G.1.5 2.02 (a) 270 (Oakwood) e1,2,4-triazolo4,3-alpyrazine 4-o-Tolyltetrahydro-2H 1-(3-o-Tolyltetrahydro-2H-pyran- G.1.6 .84 (a) 334 pyran-4-carbaldehyde 4-yl)-6H-pyrrolo2,3- (ASDI) e1,2,4-triazolo4,3-alpyrazine Benzaldehyde 1-Phenyl-6H-pyrrolo2,3- G.1.7 .83 (a) 236 e1,2,4-triazolo4,3-alpyrazine 6-Me hylcyclohex-3- 1-(6-Methylcyclohex-3-enyl)-6H- G.1.8 .83 (a) 254 enecarbaldehyde (ASDI) pyrrolo2,3-e1,2,4-triazolo 4,3- apyrazine 4-(Thiophen-2- 1-(4-(Thiophen-2-yl)tetrahydro- G.1.9 .31 (a) 326 yl)tetrahydro-2H-pyran-4- 2H-pyran-4-yl)-6H-pyrrolo[2,3- carbaldehyde (ASDI) e1,2,4-triazolo4,3-alpyrazine 2-(Pyridin-4- 1-(2-(Pyridin-4-yl)cyclopropyl)- G.1.10 1.04 (d) 277 yl)cyclopropanecarbaldehyde 6H-pyrrolo[2.3 (ASDI) e1,2,4-triazolo4,3-alpyrazine p-Tolualdehyde 1-p-Tolyl-6H-pyrrolo[2,3- G.1.11 1.28 (d) 250 e1,2,4-triazolo4,3-alpyrazine trifluoroacetate Cycloheptanecarbaldehyde 1-Cycloheptyl-6H-pyrrolo[2,3- G.1.12 1.32 (d) 256 e1,2,4-triazolo4,3-alpyrazine trifluoroacetate 2-Cycloproylacetaldehyde 1-(Cyclopropylmethyl)-6H G.1.13 1.19 (d) 214 pyrrolo2,3-e1,2,4-triazolo 4,3- apyrazine trifluoroacetate

US 9,365,579 B2 159 160 TABLE G.1-continued

Examples prepared from 2-hydrazinyl-5-tosyl-5H-pyrrolo[2,3-bipyrazine (Preparation #9) using General Procedure G

R. min (Table 2, m/z ESI+ Aldehyde Product Exhi Method) (M+H)*

3- 1-(3-(Trifluoromethyl)phenyl)-6H- G.1.35 .33 (d) 304 (Trifluoromethyl)benzaldehyde pyrrolo2,3-e124triazolo4,3- apyrazine trifluoroacetate 3-Methylthiophene-2- 1-(3-Methylthiophen-2-yl)-6H- G.1.36 .24 (d) 256 carboxaldehyde pyrrolo2,3-e124triazolo4,3- apyrazine trifluoroacetate Cyclopropylcarboxaldehyde 1-Cyclopropyl-6H-pyrrolo[2,3- G.1.37 .17 (d) 200 e1,2,4-triazolo4,3-alpyrazin trifluoroacetate 3,3- 1-Neopentyl-6H-pyrrolo[2,3- G.1.38 .26 (1) 230 Dimethylbutyraldehyde e1,2,4-triazolo4,3-alpyrazin trifluoroacetate 2,3-Dimethylbenzaldehyde 1-(2,3-Dimethylphenyl)-6H- G.1.39 .29 (d) 264 pyrrolo2,3-e124triazolo4,3- apyrazine trifluoroacetate

General Procedure H: Hydrolysis of a Sulfonamide Illustration of General Procedure H To a flask containing a sulfonamide, for example, a Sulfo Example #H.1.1 nyl-protected pyrrole, (preferably 1 equiv) in an organic Sol 30 N-(4-(6H-Pyrrolo2,3-e 1,2,4-triazolo 4.3-a vent (such as 1,4-dioxane, MeOH, or THF/MeOH, preferably pyrazin-1-yl)bicyclo[2.2.2]octan-1-yl)-3-chloroben 1,4-dioxane) is added an aqueous base (such as aqueous Zenesulfonamide NaCO or aqueous NaOH, 1-30 equiv, preferably 1-2 equiv for aqueous NaOH, preferably 15-20 equiv for aqueous 35 NaCO). The mixture is stirred at about 25-100° C. (prefer C ably about 60°C.) for about 1-72 h (preferably about 1-16h). In cases where the reaction does not proceed to completion as O monitored by TLC, LC/MS, or HPLC, additional aqueous 40 base (such as aqueous NaCO, 10-20 equiv, preferably 10 O s equiv or aqueous NaOH, 1-5 equiv, preferably 1-2 equiv) is NH added and the reaction is continued at about 25-100° C. (preferably about 60°C.) for about 0.25-3h (preferably about 45 Ns 1-2 h). The reaction is worked up using one of the following A methods. Method 1. The organic solvent is optionally N N removed under reduced pressure and the aqueous Solution is neutralized with the addition of a Suitable aqueous acid (Such as aqueous HCl). A Suitable organic solvent (such as EtOAC 50 CO)N or DCM) and water are added, the layers are separated, and \2' the organic Solution is dried over anhydrous Na2SO4 or / C MgSO filtered, and concentrated to dryness under reduced pressure to give the target compound. Method 2. The organic 55 Solvent is optionally removed under reduced pressure a Suit 6s able organic solvent (such as EtOAc or DCM) and water are NH added, the layers are separated, and the organic solution is dried over anhydrous NaSO or MgSO, filtered, and con centrated to dryness under reduced pressure to give the target 60 Ns compound. Method 3. The reaction mixture is concentrated / and directly purified by one of the subsequent methods. The N N crude material obtained from any of the preceding methods is optionally purified by precipitation, crystallization, and/or 65 trituration from an appropriate solvent or solvents and/or by a N1 N chromatography to give the target compound.

US 9,365,579 B2 189 190 TABLE H.1-continued Examples prepared using General Procedure H R. min (Table 2, m/z ESI+ Sulfonamide Product Ex. # Method) (M+H)" fluorobenzenesulfonyl chloride and DIEA) 4-Fluoro-2-methyl-N-(1S,3R)-3-(6- N-(1S,3R)-3-(6H H.1.107 1.29 (d) 415 tosyl-6H-pyrrolo[2,3- Pyrrolo[2,3- e1,2,4-triazolo4,3-alpyrazin-1- e1,2,4-triazolo4,3- yl)cyclopentyl)benzenesulfonamide apyrazin-1- (prepared using I from Preparation yl)cyclopentyl)-4-fluoro-2- #C.1, N with 4-fluoro-2- methylbenzenesulfonamide methylbenzenesulfonyl chloride and DIEA) 2-Fluoro-5-methyl-N-(1S,3R)-3-(6- N-(1S,3R)-3-(6H H.1.108 1.28 (d) 415 tosyl-6H-pyrrolo[2,3- Pyrrolo[2,3- e1,2,4-triazolo4,3-alpyrazin-1- e1,2,4-triazolo4,3- yl)cyclopentyl)benzenesulfonamide apyrazin-1- (prepared using I from Preparation yl)cyclopentyl)-2-fluoro-5- #C.1, N with 2-fluoro-5- methylbenzenesulfonamide methylbenzenesulfonyl chloride and DIEA) 2,5-Dichloro-N-((1S,3R)-3-(6-tosyl N-(1S,3R)-3-(6H H.1.109 1.34 (d) 457 6H-pyrrolo[2,3- Pyrrolo[2,3- e1,2,4-triazolo4,3-alpyrazin-1- e1,2,4-triazolo4,3- yl)cyclopentyl)thiophene-3- apyrazin-1- Sulfonamide (prepared using I from yl)cyclopentyl)-2,5- Preparation #C.1, N with 2,5- dichlorothiophene-3- dichlorothiophene-3-sulfonyl Sulfonamide chloride and DIEA)

30 General Procedure I: Acidic Cleavage of a Boc-Protected NaSO or MgSO4, then decanted or filtered, prior to concen Amine trating under reduced pressure to give the target compound. To a solution of a Boc-protected amine (preferably 1 equiv) Optionally, the crude material is purified by chromatography, in an organic solvent (such as DCM, 1,4-dioxane, or MeCH) trituration with an appropriate solvent, or crystallization from is added TFA or HCl (preferably 4 N HCl in 1,4-dioxane 35 one or more solvents to give the target compound. solution, 2-35 equiv, preferably 2-15 equiv). The reaction is stirred at about 20-100° C. (preferably ambient temperature Example #I.1.1 to about 60° C.) for about 1-24 h (preferably about 1-6 h). Optionally additional TFA or HCl (preferably 4 N HCl in (R)-1-(Piperidin-3-yl)-6H-pyrrolo2,3-e12.4 tria 1,4-dioxane solution, 2-35 equiv, preferably 2-15 equiv) may 40 Zolo 4,3-alpyrazine hydrochloride be added to the reaction mixture in cases where the reaction does not proceed to completion as monitored by TLC, LC/MS, or HPLC. The reaction is then continued at ambient temperature or optionally heated up to about 100° C. (pref erably heated at about 60° C.) for about 1-24 h (preferably 45 about 1-6 h). If a solid is present in the reaction mixture, the W reaction mixture may be filtered and the solid washed with an Ns s C-A organic solvent such as 1,4-dioxane or EtO. The resulting s solid is then optionally dried under reduced pressure. Alter N N natively, the filtered material may be partitioned between an 50 organic solvent (such as EtOAc, DCM or 1,4-dioxane) and an N y aqueous base (such as saturated aqueous NaHCO or Satu N1 N rated aqueous NaCOs, preferably Saturated aqueous NaHCO). The mixture is stirred for about 1-5 h (preferably about 1 h). Any insoluble material is collected by filtration 55 NH and may be washed with a suitable solvent (such as coldwater and/or EtO) then may be optionally dried under reduced pressure. The organic layer may optionally be washed with brine, dried over anhydrous NaSO or MgSO4, then decanted or filtered, prior to concentrating under reduced 60 pressure to give the target compound. Alternatively, the reac tion is partitioned between a basic aqueous solution (such as NaCO, NaHCO, or NaOH, preferably NaOH) and an organic solvent (such as EtOAc or DCM). The aqueous layer A round bottom flask was charged with (R)-tert-butyl is then optionally extracted with additional organic solvent 65 3-(6H-pyrrolo2,3-e 1.2.4 triazolo 4,3-alpyrazin-1-yl)pip such as EtOAc or DCM. The combined organic layers may eridine-1-carboxylate (0.92 g, 2.68 mmol; prepared using A optionally be washed with brine, dried over anhydrous from Preparation #9, (R)-1-(tert-butoxycarbonyl)piperidine

US 9,365,579 B2 193 194 General Procedure J: Deprotection of a Cbz-Protected Amine -continued A mixture of an O-benzylcarbamate (preferably 1 equiv) and 10% Pd on carbon (0.05-0.30 equiv, preferably 0.10 NH equiv) in a protic solvent (such as MeCH, EtOH, AcOH, preferably EtOH) is shaken or stirred under hydrogenat about 5 15-100 psi (preferably about 60 psi) for about 4-48 h (pref- N erably about 4-16 h) at ambient temperature. The reaction is - filtered through Celite(R) and concentrated to dryness under N N reduced pressure. The crude material is optionally purified by N precipitation, crystallization, and/or trituration from an 10 appropriate solvent or solvents and/or by chromatography to s N give the target compound. H Illustration of General Procedure J Example #J.1.1 15 1-(Piperidin-4-yl)-6H-pyrrolo2,3-e12.4 triazolo 4.3-alpyrazine Benzyl 4-(6H-pyrrolo2,3-e 1.2.4 triazolo 4.3-a O 2O pyrazin-1-yl)piperidine-1-carboxylate (0.34g, 0.90 mmol. X-/O Example #2, Step A) and 10% Pd on carbon (0.10 g, 0.09 N mmol) in MeCH (30 mL) was shaken under hydrogen at 25 about 60 psi for about 5 hat ambient temperature. The reac Ns tion was filtered through Celite(R) and concentrated under N N reduced pressure to constant weight to afford 1-(piperidin-4- N 30 yl)-6H-pyrrolo2,3-e12.4 triazolo 4.3-alpyrazine as a yel

s NH low solid (0.18g, 77%). LC/MS (Table 2, Methoda) R, 0.70 min: MS m/z. 243 (M+H)". TABLE J1

Examples prepared using General Procedure J

R, min (Table 2, m/z ESI+ Cbz-protected Amine Product Example # Method) (M+H)"

Benzyl 4-methyl-3-(6H- 1-(4-Methylpiperidin-3- J.1.2 1.03 (a) 257 pyrrolo 2,3-e1,2,4-triazolo4.3-yl)-6H-pyrrolo[2,3- apyrazin-1-yl)piperidine-1- e1,2,4-triazolo 4,3- carboxylate (prepared using R apyrazine major rom 4-methylnicotinic acid, Q, product W and B from Preparation #3) Benzyl 4-methyl-3-(6H- 1-(1,3- J.13 0.71 (a) 271 pyrrolo 2,3-e1,2,4-triazolo4.3- Dimethylpiperidin-4- apyrazin-1-yl)piperidine-1- yl)-6H-pyrrolo[2,3- carboxylate (prepared using R e1,2,4-triazolo4,3- rom 4-methylnicotinic acid, Q, apyrazine acetate W and B from Preparation #3) minor product Benzyl cis-3-(6H-pyrrolo[2,3- cis-3-(6H-Pyrrolo[2,3- J.1.4 0.56 (a) 229 e1,2,4-triazolo4,3-alpyrazin-1- e1,2,4-triazolo4,3- yl)cyclobutylcarbamate apyrazin-1- (prepared using Q from 3- yl)cyclobutanamine aminocyclobutanecarboxylic acid hydrochloride Enamine. A from Preparation #9, E) US 9,365,579 B2 195 196 General Procedure K: Formation of an Amide from an Acti an excipient (0.025 g, 22%). LC/MS (Table 2, Method a) vated Acid and an Amine R–1.33 min; MS m/z. 324 (M+H)". To around-bottomed flask containing anamine oranamine salt (preferably 1 equiv) in an organic solvent (Such as DCM. TABLE K1 5 DMF, or 1,4-dioxane, preferably DCM or DMF) is added an Examples prepared from perfluorophenyl 2-cyanoacetate organic base such as DIEA or TEA (0-5 equiv, preferably 3 Preparation #6) using General Procedure K. equiv). The reaction mixture is optionally made homoge neous by heating or Sonicating (preferably by Sonicating). To Rmin mz (Table 2, ESI the reaction mixture is added an activated acid (such as a Amine Product Ex. # Method) (M+H)" perfluorophenyl ester derivative or an acid chloride). The 10 (1R,3S)-3-(6H-Pyrrolo N-((1R,3S)-3-(6H- K.1.2 1.27 (a) 310 resulting mixture is stirred at ambient temperature for about 2,3-e1,2,4-triazolo Pyrrolo2,3- 1-24 h (preferably about 16 h). The reaction mixture may be 4,3-alpyrazin-1- e1,2,4-triazolo directly purified by chromatography. Alternatively, the sol yl)cyclopentanamine 4,3-alpyrazin-1- hydrochloride yl)cyclopentyl)-2- vent is concentrated under reduced pressure or a Suitable 15 (Example #D.1.2) cyanoacetamide organic solvent (such as EtOAc or DCM) is added and the trans-4-(6H-Pyrrolo N-(trans-4-(6H- K.1.3 1.35 (a) 324 solution is washed with water or brine. The layers are sepa 2,3-e1,2,4-triazolo Pyrrolo2,3- rated and the organic solution is optionally dried over anhy 4,3-alpyrazin-1-yl) e1,2,4-triazolo cyclohexanamine 4,3-alpyrazin-1- drous NaSO or MgSO filtered or decanted, and concen hydrochloride yl)cyclohexyl)-2- trated to dryness under reduced pressure. The crude material (Example #D.1.3) cyanoacetamide is optionally purified by precipitation, crystallization, and/or (1R,3R)-3-(6H-Pyrrolo N-((1R,3R)-3-(6H- K.14 1.39 (a) 310 trituration from an appropriate solvent or solvents and/or by 2,3-e1,2,4-triazolo Pyrrolo2,3- chromatography to give the target compound. 4,3-alpyrazin-1- e1,2,4-triazolo yl)cyclopentanamine 4,3-alpyrazin-1- Illustration of General Procedure K hydrochloride yl)cyclopentyl)-2- 25 (Example #D. 1.4) cyanoacetamide Example #K.1.1 ((1S,3R)-3-(6H-Pyrrolo N-(1S,3R)-3-(6H- K.1.5 1.38 (a) 310 2,3-e1,2,4-triazolo Pyrrolo2,3- 4,3-alpyrazin-1-yl) e1,2,4-triazolo N-(cis-4-(6H-Pyrrolo2,3-e 1,2,4-triazolo 4.3-a cyclopentanamine 4,3-alpyrazin-1- pyrazin-1-yl)cyclohexyl)-2-cyanoacetamide hydrochloride yl)cyclopentyl)-2- (Example #6, Step C) cyanoacetamide 30 (1S,3S)-3-(6H-Pyrrolo N-(1S,3S)-3-(6H- K.1.6 1.05 (d) 310 2,3-e1,2,4-triazolo Pyrrolo2,3- 4,3-alpyrazin-1- e1,2,4-triazolo yl)cyclopentanamine 4,3-alpyrazin-1- hydrochloride yl)cyclopentyl)-2- (Example # I.1.2) cyanoacetamide

General Procedure L: Formation of an Amide from a Car boxylic Acid and an Amine To a solution or Suspension of a carboxylic acid (1-5 equiv, preferably 1.5 equiv) and an amine (1-5 equiv, preferably 1 equiv) in an organic solvent (such as DCM, DCE, THF, or 1,4-dioxane, preferably DCM) is added a peptide coupling reagent (such as BOP-C1, IBCF, HATU, or EDC.HCl, pref erably EDC.HCl, 1-10 equiv, preferably 1-10 equiv), a base (such as TEA, DIEA, or pyridine, preferably TEA, 0-20 equiv, preferably 2 equiv) and HOBt (0-5 equiv, preferably 0-1 equiv when EDC.HCl is used). The reaction mixture is then stirred at ambient temperature for about 15 minto 24h (preferably about 16 h). The reaction mixture is then worked up using one of the following methods. Method 1: The reac tion mixture is diluted with water or Saturated aqueous NaHCO. The layers are separated. The aqueous layer is optionally extracted with additional organic solvent Such as To a Suspension of cis-4-(6H-pyrrolo2,3-e12.4 triazolo 55 EtOAc or DCM. The organic layeris (or combined layers are) 4,3-alpyrazin-1-yl)cyclohexanamine hydrochloride (0.106 optionally washed with water, saturated aqueous NaHCO g, 0.206 mmol. Example iD.1.1) in DCM (4 mL) was added and/or brine, dried over anhydrous MgSO or NaSO, fil TEA (0.086 mL, 0.62 mmol). The reaction mixture was soni tered or decanted, and concentrated under reduced pressure. cated until the reaction was homogeneous. To the reaction Method 2: The crude reaction mixture is filtered through a pad solution was added perfluorophenyl 2-cyanoacetate (0.078 g. 60 of silica gel, washing with a suitable solvent (such as EtOAc, 0.31 mmol, Preparation #6). The resulting solution was MeOH, or DCM, preferably MeOH), and concentrated under stirred at ambient temperature for about 16 h. The crude reduced pressure. Method 3: The crude reaction mixture is reaction mixture was purified by silica gel chromatography directly purified by chromatography without a work up. In all (40 g) eluting with a gradient of 0-20% EtOAc in DCM and cases, the crude material is optionally further purified by then further purified by RP-HPLC (Table 2, Methode) to give 65 precipitation, crystallization, and/or trituration from an N-(cis-4-(6H-pyrrolo2,3-e 1.2.4 triazolo 4,3-alpyrazin-1- appropriate solvent or solvents and/or by chromatography to yl)cyclohexyl)-2-cyanoacetamide with 3 equiv NHOAc as give the target compound.