Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ARYL AND HETEROARYL UREA CHK1 INHIBITORS FOR USE AS RADIOSENSITIZERS AND CHAMOSENSITIZERS
Document Type and Number:
WIPO Patent Application WO/2002/070494
Kind Code:
A1
Abstract:
Aryl- and heteroaryl substituted urea compounds useful in the treatment of diseases and conditions related to DNA damage or lesions in DNA replication are disclosed. Methods of making the compounds, and their use as therapeutic agents, for example, in treating cancer and other diseases characterized by defects in DNA replication, chromosome segregation, or cell division also are disclosed. Wherein W' is a six-membered aromatic ring containing at least 2 introgen atoms and optionally substitutedas defined in the claims, Z' is a five- or six membered aromatic or heteraromatic ring as defined in the claims, Y' is O or S.

Inventors:
KEEGAN KATHLEEN S
KESICKI EDWARD A
GAUDINO JOHN JOSEPH
COOK ADAM WADE
COWEN SCOTT DOUGLAS
BURGESS LAURENCE EDWARD
Application Number:
PCT/US2002/006452
Publication Date:
September 12, 2002
Filing Date:
March 01, 2002
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ICOS CORP (US)
International Classes:
A61K31/4965; A61K31/497; A61K31/498; A61K31/5377; A61K45/06; A61P35/00; A61P43/00; C07D213/30; C07D213/40; C07D213/75; C07D213/77; C07D215/34; C07D231/40; C07D239/42; C07D239/46; C07D241/12; C07D241/20; C07D241/30; C07D241/44; C07D241/54; C07D251/22; C07D253/06; C07D253/07; C07D257/06; C07D277/48; C07D401/12; C07D401/14; C07D403/06; C07D403/12; C07D405/12; C07D409/12; C07D409/14; C07D453/02; C07D521/00; (IPC1-7): C07D241/20; C07D401/12; C07D403/12; C07D241/54; A61K31/4965; A61K31/498; A61P35/00; C07D405/12; C07D251/22; C07D231/40; C07D239/46; C07D253/06; C07D239/42; C07D257/06; C07D213/75; C07D277/48
Domestic Patent References:
WO2000026203A12000-05-11
WO2000056725A12000-09-28
WO1994026715A11994-11-24
WO2000056725A12000-09-28
Foreign References:
US5215738A1993-06-01
US5041653A1991-08-20
US2762743A1956-09-11
US6051218A2000-04-18
US5215738A1993-06-01
US5041653A1991-08-20
Other References:
DUTTA ET AL.: "Synthesis and properties of N-carbamoyl derivatives of cytosine, cytidine, uracil and thymine.", J. CARBOHYDR. NUCLEOS. NUCLEOT., vol. 7, no. 4, 1980, pages 217 - 40, XP001087721
HEINISCH ET AL: "Synthesis of N-aryl-N'-heteroaryl-substituted urea and thiourea derivatives and evaluation of their anticonvulsant activity", ARCHIV DER PHARMAZIE, VCH VERLAGSGESELLSCHAFT MBH, WEINHEIM, DE, vol. 330, no. 7, 1 July 1997 (1997-07-01), pages 207 - 210, XP002087695, ISSN: 0365-6233
FERNERY ET AL., SCIENCE, vol. 277, no. 5331, 5 September 1997 (1997-09-05), pages 1495 - 7
BLASINA ET AL., CURR. BIOL., vol. 9, no. 1, 14 January 1999 (1999-01-14), pages 1 - 10
Z. MA ET AL., TETRAHEDRON: ASYMMETRY, vol. 8, no. 6, 1997, pages 883 - 888
BRADLEY ET AL., CLIN. CANCER RES., vol. 7, 2001, pages 3229
Attorney, Agent or Firm:
Napoli, James J. (Gerstein & Borun 6300 Sears Tower 233 South Wacker Drive Chicago, IL, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:
1. A method of inhibiting checkpoint kinase 1 in a cell comprising a step of contacting the cell with an effective amount of a compound of formula wherein X1 is null,O,S,CH2, or N (Rl) ; X2 isO,S, orN (R1); Y is O or S; or =Y represents two hydrogen atoms attached to a common carbon atom; W is selected from the group consisting of heteroaryl, aryl, heterocycloalkyl, cycloalkyl, and C13 alkyl substituted with a heteroaryl or aryl group; Z is selected from the group consisting of hydro, aryl, and heteroaryl; wherein said aryl groups of W and Z are optionally substituted with one to four substituents represented by R2, said heteroaryl groups of W and Z are optionally substituted with one to four substit uents represented by R5, and said heterocycloalkyl and cycloalkyl groups of W are optionally substi tuted with one to two substituents represented by R6 ; RI is selected from the group consisting of hydro, C16alkyl, C25alkenyl, C26alkynyl, and aryl; R2 is selected from the group consisting of halo, optionally substituted C16alkyl, C26alkenyl, OCF3, NO2, CN, NC, N (R3) z, OR3, CO2R3, C (0) N (R3)2, C (O) R3, N (R1) COR3, N (R1) C (O) OR3, N (R3) C (O) OR3, N (R3) C (0) C13alkyleneC(O)R3, N (R3) C (O) C13alkyleneC(O)OR3, N (R3) C (O) C1 3alkyleneOR3, N (R3) C (O) C1 3alkyleneNHC (O) OR3, N (R3) C (O) C13alkyleneSO2NR3, C13alkyleneOR3, and SR3; R3 is selected from the group consisting of hydro, C16alkyl, C26alkenyl, cycloalkyl, aryl, heter oaryl, SO2R4, C16alkyl substituted with one or more of halo, hydroxy, aryl, heteroaryl, heterocyclo alkyl, N (R4) 2, and SO2R4, C13alkylenearyl, C13alkyl eneheteroaryl, C13alkyleneC38heterocycloalkyl, C13 alkyleneSO2aryl, optionally substituted Cl3alkylene N(R4)2, OCF3, C13alkyleneN(R4)3+, C38heterocycloalkyl, and CH (C1 3alkyleneN (R4) 2) 2, or two R3 groups are taken together to form an optionally substituted 3to 6 membered aliphatic ring; R4 is selected from the group consisting of hydro, C16alkyl, cycloalkyl, aryl, heteroaryl, C13 alkylenearyl, and SO2C16alkyl, or two R4 groups are taken together to form an optionally substituted 3 to 6membered ring; R5 is selected from the group consisting of Cl6alkyl, aryl, N (R3) z, OR3, halo, N3, CN, C13alkyl enearyl, C13alkyleneN (R3) 2, C (0) R3, and 0 C1 3aSkY1eneN W I 0 R6 is selected from the group consisting of halo and C,, alkyl ; and pharmaceutically acceptable salts, prodrugs, or solvates thereof.
2. The method of claim 1 wherein Xl and X2 areN (H) ; Y is O or S; W is heteroaryl containing at least two heteroatoms selected from the group consisting of N, O, and S, said ring is optionally substituted with from one to four substituents selected from the group consisting of Cl6alkyl, aryl, N (R3) 2, OR3, and halo; Z is selected from the group consisting of wherein Q is selected from the group consisting of hydro, OR3, SR3, and N (R3) 2; J is selected from the group consisting of R NR20, 0, and S; K is selected from the group consisting of CR21,NR21, O, and S; L is selected from the group consisting of CR22,NR22, O, and S; M is selected from the group consisting of CR23,NR23, 0, and S; wherein: R20, R21, and R22 are each independently selected from the group consisting of null, hydro, halo, optionally substituted C16alkyl, C26alkenyl, OCF3, NO2, CN, NC, N (R25)2, OR25, CO2R25, C (O) N (R25) 2/ C (0) R25, N (R24)COR25, N (R24) C (0) Ores, N (R 25) C (O) OR25, N (R25) C (O) Cl3alkyleneC (O) R25 N (R25) C (O) Cl3alkylene C (0) OR25, N (R25)C(O)C13alkyleneOR25, N (R25) C (O) C13alkyl eneNHC (O) OR25, N (R25)C(O)C13alkyleneSO2NR25, CF3, C13 alkyleneN (R25)SO2aryl, C13alkyleneN(R25)SO2heteroaryl, C13alkyleneOC13alkylenearyl, C13alkyleneN(R25)C13 alkylenearyl, C13alkyleneN(R25)C13alkyleneheteroaryl, C13alkyleneN(R25)C(O)R7, C13alkyleneN(R25) C (O) Cl3 alkyleneOR25, C13alkyleneN(R25) C (O) aryl, C13alkylene N (R25) C (O) Cl3alkyleneN (R25)2, C13alkyleneN(R25) C (O) het eroaryl, C13alkyleneOR25, and SR25 ; R23 is selected from the group consisting of null, hydro, optionally substituted Cl 6alkyl, and halo; R24 is selected from the group consisting of hydro, C16alkyl, C2 6alkenyl, C26alkynyl, and aryl; R25 is selected from the group consisting of hydro, C16alkyl, C26alkenyl, cycloalkyl, hetero cycle, aryl, heteroaryl, SO2R26, and C16alkyl substi tuted with halo, hydroxy, aryl, heteroaryl, heterocycloalkyl, N (R26) 2, or SO2R26 ; and R26 is selected from the group consisting of hydro, C16alkyl, cycloalkyl, aryl, and SO2C16 alkyl, or two R4 groups are taken together to form an optionally substituted 3to 6membered ring.
3. The method of claim 2 wherein W is selected from the group consisting of pyridazinyl, pyrimidinyl, pyrazinyl, and triazinyl, optionally substituted with from one to four substituents selected from the group consisting of optionally substituted Cl6alkyl, aryl, N (R3) 2, OR3, and halo.
4. The method of claim 2 wherein W is selected from the group consisting of.
5. The method of claim 2 wherein J is selected from the group consisting of CR20 and NR, wherein R2° is null, hydro, optionally substituted C16alkyl, and halo; K is selected from the group consisting of CR21 and NR21 ; L is selected from the group consisting of CR22 and NR22 ; and one of R21 and R22 is hydro and the other is a substituent selected from the group consisting of CO2R25, C (O) N (R) 2 C (O) R25, N (R24)COR25, N (R24) C (O) OR25, N (R25) C (O) OR25, N (R25) C (O) C13alkyleneC(O)R25, N (R25) C (Oj C13alkyleneC(O)OR25, N (R25)C(O)C13alkyleneOR25, N (R25)C(O)C13alkyleneNHC(O)OR25, N (R25)C(O)C13alkylene SO2NR25, CF3, C13alkyleneN(R25)SO2aryl, C13alkylene N(R25)SO2heteroaryl, C13alkyleneOC13alkylenearyl, C13 alkyleneN (R25)C13alkylenearyls, C13alkyleneN(R25)C13 alkyleneheteroaryl, C13alkyleneN (R25) C (O) R7, C13alkyl eneN (R25) C (O) C13alkyleneOR2, C13alkyleneN(R25) C (O) aryl, C13alkyleneN (R25) C (O) Cl3alkyleneN (R25)2, C13alkylene N (R25) C (O) heteroaryl, C13alkyleneOR25, and SR25.
6. The method of claim 2 wherein W is pyrazinyl.
7. The method of claim 1 wherein X1 is null, X2 isN (H), Y is O, and Z is hydro.
8. A method of sensitizing cells in an individual undergoing a chemotherapeutic or radio therapeutic treatment for a medical condition, com prising administering a therapeutically effective amount of a compound of formula (I) in combination with a chemotherapeutic agent, a radiotherapeutic agent, or a mixture thereof to the individual, said compound of formula (I) having a structure wherein X1 is null,O,S,CH2, or N (R1); X2 isO,S, orN (R1); Y is O or S; or =Y represents two hydrogen atoms attached to a common carbon atom; W is selected from the group consisting of heteroaryl, aryl, heterocycloalkyl, cycloalkyl, and C13 alkyl substituted with a heteroaryl or aryl group; Z is selected from the group consisting of hydro, aryl, and heteroaryl; wherein said aryl groups of W and Z are optionally substituted with one to four substituents represented by R, said heteroaryl groups of W and Z are optionally substituted with one to four substit uents represented by R5, and said heterocycloalkyl and cycloalkyl groups of W are optionally substi tuted with one to two substituents represented by R6 ; RI is selected from the group consisting of hydro, C16akyl, C26alkenyl, C26alkynyl, and aryl; R is selected from the group consisting of halo, optionally substituted C16alkyl, C26alkenyl, OCF3, NO2, CN, NC, N (R3)2, OR3, CO2R3, C (O) N (R3) 2, C (0) R3, N (Rl) COR3, N (Rl) C (0) OR3, N (R3) C (O) OR3, N (R3) C (O) C13alkyleneC (O) R3, N (R3) C (O) Cl3alkyleneC (O) OR3, N (R3) C (O) C13alkyleneOR3, N (R3) C (O) Cl3alkyleneNHC (O) OR3, N (R3) C (O) Cl3alkyleneSO2NR3, Cl3alkyleneOR3, and SR3; R3 is selected from the group consisting of hydro, C16alkyl, C26alkenyl, cycloalkyl, aryl, heter oaryl, SO2R4, C16alkyl substituted with one or more of halo, hydroxy, aryl, heteroaryl, heterocycloalk yl, N (R4)2, and SO2R4, C13alkylenearyl, C13alkylene heteroaryl, C13alkyleneC38heterocycloalkyl, C13alkyl eneSO2aryl, optionally substituted C13alkyleneN(R4)2, OCF3, C13alkyleneN(R4)3+, C38heterocycloalkyl, and CH (Cl3alkyleneN (R4) 2) 2, or two R3 groups are taken together to form an optionally substituted 3to 6 membered aliphatic ring; R4 is selected from the group consisting of hydro, C16alkyl, cycloalkyl, aryl, heteroaryl, Cl_3 alkylenearyl, and SO2C16alkyl, or two R4 groups are taken together to form an optionally substituted 3 to 6membered ring; RS is selected from the group consisting of C16alkyl, aryl, N (R3)2, OR3, halo, N3, CN, C13alkyl enearyl, C13alkyleneN (R3)2, C (O) R3, and 0 C13 alkyl eneNX3 \ 0 R6 is selected from the group consisting of halo and Cl 6alkyl ; and pharmaceutically acceptable salts, prodrugs, or solvates thereof.
9. The method of claim 8 further com prising administering one or more cytokine, lympho kine, growth factor, or other hematopoietic factor.
10. The method of claim 8 wherein: XI and X2 are N(H); Y is O or S; W is heteroaryl containing at least two heteroatoms selected from the group consisting of N, O, and S, said ring is optionally substituted with from one to four substituents selected from the group consisting of C16alkyl, aryl, N (R3) 2, OR3, and halo; Z is selected from the group consisting of: wherein Q is selected from the group consisting of hydro, OR3, SR3, and N (R'), ; J is selected from the group consisting of CR20, NR20, O, and S; K is selected from the group consisting of CR21 NR21, O, and S ; L is selected from the group consisting of CR22,NR22, O, and S ; M is selected from the group consisting of CR23,NR23, 0, and S ; wherein: R20, R21, and R22 are each independently selected from the group consisting of null, hydro, halo, optionally substituted C16alkyl, C2 6alkenyl, OCF3, NO2, CN, NC, N (R25)2, OR25, CO2R25, C (O) N (R25)2, C (O) R25, N (R24) COR25, N (R24) C (0) OR25, N (R25) C (0) OR 5, N (R25) C (O)C13alkyleneC(O)R25, N (R25) C (O) C13alkylene C (0) OR25, N (R25) C(O0C13alkyleneOR25, N(R25)C(O)C13alkyl eneNHC (O) OR25, N (R25) C(O0C13alkyleneSO2nR25, CF3, C13 alkyleneN (R25)SO2aryl, C13alkyleneN(R25)SO2heteroaryl, C13alkyleneOC13alkylenearyl, C13alkyleneN(R25)C13 alkylenearyl, C13alkyleneN (R25) C13alkyleneheteroaryl, C13alkyleneN (R25) C (O) R', C13alkyleneN (R25) C (0) C13 alkyleneOR25, C13alkylenen(R25) C (O) aryl, C13alkylene N (R25)C(O)C13alkyleneN(R25)2, C13alkyleneN(R25) C (O) het eroaryl, C13alkyleneOR25, and SR25 ; R23 is selected from the group consisting of null, hydro, optionally substituted Cl6alkyl, and halo; R24 is selected from the group consisting of hydro, C16alkyl, C26alkenyl, C26alkynyl, and aryl; R25 is selected from the group consisting of hydro, C16alkyl, C26alkenyl, cycloalkyl, hetero cycle, aryl, heteroaryl, SO2R26, and Cl6alkyl substi tuted with halo, hydroxy, aryl, heteroaryl, heterocycloalkyl, N (R26) 2, or SO2R26 ; and R26 is selected from the group consisting of hydro, C16alkyl, cycloalkyl, aryl, and SO2C16 alkyl, or two R4 groups are taken together to form an optionally substituted 3to 6membered ring.
11. The method of claim 10 wherein W is selected from the group consisting of pyridazinyl, pyrimidinyl, pyrazinyl, and triazinyl, optionally substituted with from one to four substituents selected from the group consisting of optionally substituted Cl 6alkyl, aryl, N (R3)2, OR3, C13alkylene aryl, C13alkyleneheteroaryl, C13alkyleneC38hetero cycloalkyl, C13alkyleneSO2aryl, optionally substi tuted Cl3alkyleneN (R4)2, OCF3, C13alkylenen(R4)3+, C38heterocycloalkyl, CH (Cl3alkyleneN (R4) 2) 2, and halo.
12. The method of claim 10 wherein J is selected from the group consisting of CR20 and NR2°, wherein R is null, hydro, optionally substituted Cl 6alkyl, and halo; K is selected from the group consisting of CR21 and NR21; L is selected from the group consisting of CR22 and NR22 ; and one of R21 and R22 is hydro and the other is a substituent selected from the group consisting of CO2R25, C (0) N (R25)2, C (O) R25, N (R24)COR25, N (R24) C (O) oR N (R25) C (O) OR25, N (R25) C(O) C13alkyleneC(O)R25, N (R25) C (0) C13alkyleneC(O)OR25, N (R25) C (O) C13alkyleneOR25, N (R25) C (O) Cl3alkyleneNHC (O) OR25, N (R25) C(O0 C13alkylene SO2NR25, C13alkyleneOR25, CF3, C13alkyleneN(R25)SO2aryl, C13alkyleneN(R25)SO2heteroaryl, C13alkyleneOC13alkyl enearyl, C13alkylenen(R25) C13alkylenearyl, C13alkyl eneN (R25)C13alkyleneheteroaryl, C13alkyleneN(R25) C (0) R3, C13alkyleneN (R25) C (O) C13alkyleneOR3, C13alkyl eneN (R25) C (O) aryl, C13alkyleneN (R25) C (O) Cl3alkylene N (R25)2, C13alkyleneN(R25) C (O) heteroaryl, and SR25.
13. The method of claim 10 wherein W is pyrazinyl.
14. The method of claim 8 wherein the chemotherapeutic agent is selected from the group consisting of an alkylating agent, an antimetabo lite, a hormone or antagonist thereof, a radio isotope, an antibody, and mixtures thereof.
15. The method of claim 8 wherein the radiotherapeutic agent is selected from the group consisting of gammaradiation, Xray radiation, ultraviolet light, visible light, infrared radia tion, and microwave radiation.
16. The method of claim 8 wherein the condition is a cancer selected from the group con sisting of a colorectal cancer, a head and neck cancer, a pancreatic cancer, a breast cancer, a gastric cancer, a bladder cancer, a vulvar cancer, a leukemia, a lymphoma, a melanoma, a renal cell carcinoma, an ovarian cancer, a brain tumor, an osteosarcoma, and a lung carcinoma.
17. The method of claim 8 wherein the condition is a cancer selected from the group con sisting of myxoid and round cell carcinoma, a locally advanced tumor, metastatic cancer, Ewing's sarcoma, a cancer metastase, a lymphatic metastase, squamous cell carcinoma, esophageal squamous cell carcinoma, oral carcinoma, multiple myeloma, acute lymphocytic leukemia, acute nonlymphocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, hairy cell leukemia, effusion lymphomas (body cavity based lymphomas), thymic lymphoma lung cancer, small cell carcinoma, cutaneous T cell lym phoma, Hodgkin's lymphoma, nonHodgkin's lymphoma, cancer of the adrenal cortex, ACTHproducing tumors, nonsmall cell cancers, breast cancer, small cell carcinoma, ductal carcinoma, stomach cancer, colon cancer, colorectal cancer, polyps associated with colorectal neoplasia, pancreatic cancer, liver cancer, bladder cancer, primary superficial bladder tumors, invasive transitional cell carcinoma of the bladder, muscleinvasive bladder cancer, prostate cancer, ovarian carcinoma, primary peritoneal epi thelial neoplasms, cervical carcinoma, uterine endometrial cancers, vaginal cancer, cancer of the vulva, uterine cancer and solid tumors in the ovarian follicle, testicular cancer, penile cancer, renal cell carcinoma, intrinsic brain tumors, neuroblastoma, astrocytic brain tumors, gliomas, metastatic tumor cell invasion in the central nervous system, osteomas and osteosarcomas, malig nant melanoma, tumor progression of human skin keratinocytes, squamous cell cancer, thyroid cancer, retinoblastoma, neuroblastoma, peritoneal effusion, malignant pleural effusion, mesothelioma, Wilms's tumors, gall bladder cancer, trophoblastic neo plasms, hemangiopericytoma, and Kapos's sarcoma.
18. The method of claim 8 wherein the treatment is administered for an inflammatory con dition selected from the group consisting of rheumatoid arthritis, psoriasis, vitiligo, Wegener's granulomatosis, and systemic lupus erythematosus.
19. A compound having a formula wherein Y' is O or S; W' is selected from the group consisting of optionally substituted with from one to four sub stituents selected from the group consisting of Cl6alkyl, aryl, N (R7)2, OR7, N3, CN, C (O) R7, C13alkyl enearyl, C13alkyleneN (R12)2, po C1 3a1kY1eneN X 0 and halo; Z'is selected from the group consisting of: wherein: Q'is selected from the group consisting of hydro, OR, SR, and N (R) 2, with the proviso that Q'is hydro only when at least one of J', K', L', and M'is N, O, or S; J'is selected from the group consisting of CR8, NR8, O, and S; K'is selected from the group consisting of CR9, NR9, O, and S; L'is selected from the group consisting of CRl°, NR10, O, and S; M'is selected from the group consisting of CR11, NR11, O, and S, with the proviso that Z is different from a pyridone; wherein: R', independently, is selected from the group consisting of hydro, C16alkyl, C26alkenyl, cycloalkyl, aryl, heteroaryl, SO2Rl2, C16alkyl sub stituted with one or more of halo, hydroxy, aryl, heteroaryl, heterocycloalkyl, N (R12) 2, and SO2R12, C13alkylenearyl, C13alkyleneheteroaryl, C13alkyl eneC38heterocycloalkyl, C13alkyleneSO2aryl, option ally substituted Cl3alkyleneN (R12)2, OCF3, C13alkyl eneN (R12)3+, C38heterocycloalkyl, and CH (C13alkylene N (R12)2)2, or two R7 groups are taken together to form an optionally substituted 3to 6membered aliphatic ring; Re, R9, and Rl° are each independently selected from the group consisting of null, hydro, halo, optionally substituted C16alkyl, C26alkenyl, OCF3, NO2, CN, NC, N (R) 2, OR, CO2R7, C (0) N (R) 21 C (O) R7, N (R13)COR7, N (R13) C (O) OR 7, N (R7) C (O) OR7, N (R7) C (O) C13alkyleneC(O)R7, N (R') C (0) Cialkylene C (O) oR7, N (R7) C (O) C13alkyleneOR7, N (R7) C (O) Cl 3alkyl eneNHC (O) OR7, N (R7) C (O) C13alkyleneSO2NR7, CF3, C13 alkyleneN (R12)SO2aryl, C13alkyleneN(R12)SO2heteroaryl, C13alkyleneOc13alkylenearyl, C13alkyleneN(R12)C13 alkylenearyl, C13alkyleneN(R12)C13alkyleneheteroaryl, C13alkylenen(R12) C (O) R7, C13alkyleneN(R12) C (O) C13 alkyleneoR2, C13alkyleneN (R12) C (O) aryl, C13alkylene N (R12) C (O) Cl3alkyleneN (R12)2, C13alkyleneN(R12)C(O) heteroaryl, C13alkyleneoR7, and SR, wherein R is as defined above; Rll is selected from the group consisting of null, hydro, optionally substituted C16alkyl, and halo; R12 is selected from the group consisting of hydro, C16alkyl, cycloalkyl, aryl, heteroaryl, C13alkylenearyl, and SO2Cl6alkyl, or two Rl2 groups are taken together to form an optionally substituted 3to 6membered ring; and R"is selected from the group consisting of hydro, Cl6alkylt C26alkenyl, C26alkynyl, and aryl; provided that when Q'is hydro or OCH3, at least one of R8, R9, and R10 is different from hydro, CH3, OCH3, and halo, and pharmaceutically acceptable salts, prodrugs, or solvates thereof.
20. The compound of claim 19 wherein W' is selected from the group consisting of.
21. The compound of claim 20 wherein W' is substituted with one to four substituents selected from the group consisting of methyl, CF3, optionally substituted aryl, N3, benzyl, C (0) R', Cl_3alkyleneN (R12) 2, OR7, N (R 2, halo, and 0 C1 3alkyleneN 0.
22. The compound of claim 19 wherein Q' is OR\.
23. The compound of claim 22 wherein Q' is OCH3.
24. The compound of claim 19 wherein R13 is hydro.
25. The compound of claim 19 wherein J'is selected from the group consisting of CR8 and NR8, wherein R8 is null, hydro, C16alkyl, and halo; K'is selected from the group consisting of CR9 and NR9 ; L'is selected from the group consisting of CRl° and NR'° ; and one of RI and R10 is hydro and the other is a substituent selected from the group consisting of Cozy C (O) N (R7)2, C (O) R7, N (R13) COR7 N (R13) C (O) OR7, N (R7) C (O) OR', N (R7) C (0) C13alkyleneC(O)R7, N (R7) C (O) Cl3alkyleneC (O) OR7, N (R') C (O) C13alkyleneOR7, N (R7) C (O) Cl3alkyleneNHC (O) OR7, N (R7) C (O) C13alkyleneSO2NR7, C13alkyleneOR7, CF3, C13alkyleneN(R12)SO2aryl, C13 alkyleneN (R12)SO2heteroaryl, C13alkyleneOC13alkylene aryl, C13alkyleneN(R12)C13alkylenearyl, C13alkylene N (R12)C13alkyleneheteroaryl, C13alkyleneN(R12) C (O) R', C13alkyleneN (R12) C (O) C13alkyleneOR2, C13alkyleneN(R12) C (O) aryl, C13alkyleneN (R12) C (O) C13alkyleneN (R12)2 C13 alkyleneN (R12) C (O) heteroaryl, and SR7.
26. A method of inhibiting checkpoint kinase 1 (Chkl) in a cell comprising the step of contacting the cell with an effective amount of a compound of claim 19.
27. A method of sensitizing cells in an individual undergoing a chemotherapeutic or radio therapeutic treatment for a medical condition, com prising administering a therapeutically effective amount of a compound of claim 19 in combination with a chemotherapeutic agent, a radiotherapeutic agent, or a mixture thereof to the individual.
28. A compound having a structure wherein R27 and R28 are R27 R28 H 0 H NH H nu I H i NH0X\ t CH3 H NH NU N H R27 R28 H nu 0 H ° tV3 A H _"o H N O ni 0 NH 0 or wherein R29 is _ R29 fez I wiz.
29. A compound selected from the group consisting of: N(2dimethylamino1phenylehtyl)3methoxy4[3 (5methylpyrazin2yl)ureido]benzamine ; N(1azabicyclo [2.2.2] oct3yl)3methoxy4 [3 (5 methylpyrazin2yl)ureido]benzamide ; N(3R1cylcohexylmethylpyrrolidin3yl)3meth oxy4 [3 (5methylpyrazin2yl)ureido]benzamide ; 1[2(2dimethylaminoethoxy)5methylphenyl]3 pyrazin2ylurea; <BR> <BR> 1 [2 (3dimethylaminopropoxy)5methylphenyl]3 (5methylpyrazin2yl)urea ; <BR> <BR> 1 (5methylpyrazin2yl)3 [5methyl2 (pyridin3 ylmethoxy)phenyl]urea ; <BR> <BR> 1 [2 (2dimethylamino1dimethylaminomethylethoxy) 5methylphenyl]3 (5methylpyrazin2yl)urea ; <BR> <BR> 1 [5methyl2 (2S1methylpyrrolidin2ylmethoxy) phenyl]3 (5methylpyrazin2yl)urea ; 1{5methyl2[2(1methylpyrrolidin2yl)ethoxy] phenyl}3 (5methylpyrazin2yl)urea ; 1{5methyl2(1methylpiperidin4yloxy)phenyl] 3 (5methylpyrazin2yl)urea ; 1 [5methyl2 (3 (S)lmethylpiperidin3ylmeth oxy)phenyl]3 (5methylpyrazin2yl)urea ; 1 [5methyl2 (3 (R)lmethylpiperidin3ylmeth oxy)phenyl]3 (5methylpyrazin2yl)urea ; 1 [5methyl2 (lmethylpiperidin2ylmethoxy) phenyl]3 (5methylpyrazin2yl)urea ; 1{5methyl2(1methylpiperidin3yloxy)phenyl] 3 (5methylpyrazin2yl)urea ; 1 [5methyl2 (lmethylpiperidin3ylmethoxy) phenyl]3quinoxalin2ylurea ; <BR> <BR> 1 [5methyl2 (piperidin3ylmethoxy)phenyl]3 (5 methylpyrazin2yl)urea ; 1 [5fluoro2 (lmethylpiperidin3ylmethoxy) phenyl]3 (5methylpyrazin2yl)urea ; 1[5fluoro2(1methylpiperidin4yloxy)phenyl] 3 (5methylpyrazin2yl)urea ; 1[4fluoro2(1methylpiperidin4yloxy)phenyl] 3 (5methylpyrazin2yl)urea ; 1 (2methoxy4methylaminomethylphenyl)3 (5 methylpyrazin2yl)urea; 1[4{[(furan3ylmethyl)amino]methyl}2methoxy phenyl)3 (5methylpyrazin2yl)urea ; and 1 {2methoxy4 [ (4methoxybenzylamino)methyl] phenyl}3 (5methylpyrazin2yl)urea.
30. A composition comprising a compound of formula (II) and a pharmaceutically acceptable carrier, said compound of formula (II) having a formula wherein Y'is 0 or S; W'is selected from the group consisting of optionally substituted with from one to four sub stituents selected from the group consisting of C16alkyl, aryl, N (R7)2, OR7, N3, CN, C (0) R7, C13alky enearyl, C13alkyleneN(R12)2, 0 CialkyleneN \ 0 and halo; Z is selected from the group consisting of: wherein: Q' is selected from the group consisting of hydro, OR', SR', and N (R') 2, with the proviso that Q'is hydro only when at least one of J', K', L', and M'is N, O, or S; J' is selected from the group consisting of CR8, NR8, O, and S; K'is selected from the group consisting of CR9, NR9, O, and S; L' is selected from the group consisting of CR10, NR10, O, and S; M'is selected from the group consisting of CR11, NRll, O, and S, with the proviso that Z is different from a pyridone ; wherein: R7, independently, is selected from the group consisting of hydro, C16alkyl, C26alkenyl, cycloalkyl, aryl, heteroaryl, SO2R12, C16alkyl sub stituted with one or more of halo, hydroxy, aryl, heteroaryl, heterocycloalkyl, N (R) 2, and SO2R12, C13alkylenearyl, C13alkyleneheteroaryl, C13alkylene C38heterocycloalkyl, C13alkyleneSO2aryl, optionally substituted C13alkyleneN(R12)2, OCF3, C13alkylene N (R12)3+, C38heterocycloalkyl, and CH (C13alkylene N (R12)2)2, or two R7 groups are taken together to form an optionally substituted 3to 6membered aliphatic ring; R8, R9, and R10 are each independently selected from the group consisting of null, hydro, halo, optionally substituted C16alkyl, C26alkenyl, OCF3, NO2, CN, NC, N (R7)2, OR7, CO2R7, C (O) N (R7) 2' C (O) R7 N (R13)COR7, N (R13) C (O) OR', N (R7) C (O) OR 7, N (R7) C (O) Cl3alkyleneC (0) R7, N (R') C (O) C13alkylene C (O) oR7, N (R7) C (O) C13alkyleneOR7, N (R7) C (O) Cl 3alkyl eneNHC (O0 OR7, N (R7) C (O) C13alkyleneSO2NR7, C13alkyl eneOR7, and SR, wherein R is as defined above; Rll is selected from the group consisting of null, hydro, optionally substituted Cl 6alkyl, and halo; R12 is selected from the group consisting of hydro, Cl 6alkyl, cycloalkyl, aryl, heteroaryl, C13alkylenearyl, and SO2C16alkyl, or two R12 groups are taken together to form an optionally substituted 3to 6membered ring; and R13 is selected from the group consisting of hydro, C16alkyl, C26alkenyl, C26alkynyl, and aryl; provided that when Q'is hydro or OCH3, at least one of R8, R9, and R10 is different from hydro, CH3, OCH3, and halo, and pharmaceutically acceptable salts, prodrugs, or solvates thereof.
Description:
ARYL AND HETEROARYL UREA CHK1 INHIBITORS FOR USE AS RADIOSENSITIZERS AND CHAMOSENSITIZERS CROSS REFERENCE TO RELATED APPLICATIONS This application claims the benefit of U. S. provisional application Serial No. 60/273,124, filed March 2,2001.

TECHNICAL FIELD OF THE INVENTION The present invention relates to compounds useful for inhibiting enzymes that maintain and re- pair the integrity of genetic material. More par- ticularly, the present invention relates to a series of aryl-and heteroaryl-substituted urea compounds, methods of making the compounds, and their use as therapeutic agents, for example, in treating cancer and other diseases characterized by defects in de- oxyribonucleic acid (DNA) replication, chromosome segregation, or cell division.

BACKGROUND OF THE INVENTION An important and significant goal in healthcare is to discover and make available safer and more effective drugs for the treatment of cancer. Most chemotherapeutic agents act by dis- rupting DNA metabolism, DNA synthesis, DNA tran- scription, or microtubule spindle function, or by perturbing chromosomal structural integrity by introducing DNA lesions. These processes affect both normal and tumor cells. The maintenance of DNA integrity is essential to cell viability in normal cells, therefore, anticancer drugs have the lowest therapeutic index of any drug class.

An individual cell creates an exact copy of its chromosomes, and then segregates each copy into two cells by a process called mitosis. The mitotic cycle can be divided into three major events: DNA replication, chromosome segregation, and cell division. Cells have sensing mechanisms to maintain the order of these steps with respect to one another and to ensure that each step is executed with high fidelity. The sensing mechanisms for these processes are referred to as"checkpoints"in L. H. Hartwell et al., Science, Nov. 3,1989, 246 (4930) : 629-34.

Cell cycle checkpoints have been reported to comprise at least three distinct classes of poly- peptides. Each class of polypeptides acts sequen- tially in response to cell cycle signals or defects in chromosomal mechanisms (Carr, (1996) Science, 271: 314-315). One family of proteins detects or senses DNA damage or abnormalities in the cell cycle. These sensors include Ataxia-Telangiectasia Mutated (Atm) and Ataxia-Telangiectasia Rad-related (Atr) (Keegan et al., (1996) Genes Dev., 10: 2423- 2437). Another class of polypeptides amplify and transmit the signal detected by the detector and is exemplified by Rad53 (Allen et al. (1994) Genes Dev., 8: 2416-2488) and Chkl. In addition, cell cycle effectors, such as p53, mediate a cellular response, including, for example, arrest of mitosis and/or meiosis and apoptosis.

DNA damage can be induced by drugs, radi- ation, or can be spontaneously generated during the course of normal metabolism. DNA damage checkpoints ensure that cells with unrepaired DNA lesions do not progress into the DNA synthesis phase or mitosis until chromosomal lesions have been removed. Cell

cycle arrest can enhance the opportunity for DNA repair and increase the fidelity of cell division.

DNA damage can be recognized throughout the cell cycle. Checkpoints ensure that the growth of cells is arrested at multiple cell cycle phases. As a result, multiple cell cycle signaling pathways may result during sensitization of cells to DNA damaging agents.

Much of the current understanding of the function of cell cycle checkpoints has been derived from the study of tumor-derived cell lines. In many cases, tumor cells have lost key cell cycle check- points (Hartwell et al., Science, Dec. 16,1994; 266 (5192): 1821-8). It has been reported that a key step in the evolution of cells to a neoplastic state is the acquisition of mutations that inactivate cell cycle checkpoint pathways, such as p53. (Weinberg, R. A. (1995) Cell 81: 323-330; Levine, A. J. (1997) Cell 88: 3234-331). Loss of these cell cycle check- points results in the inappropriate cycling of tumor cells in response to DNA damaging agents. When faced with cellular stresses, such as DNA damage, and cell cycle events with decreased fidelity, tumor cells have difficulty altering the kinetics of cell cycle progression. Therefore, inhibition and dis- ruption of additional DNA damage checkpoint pathways may further sensitize tumor cells to anticancer treatments, such as radiation and chemotherapy.

Noncancerous tissue, which has intact cell cycle checkpoints, typically is insulated from temporary disruption of a single checkpoint pathway.

Tumor cells, however, have defects in pathways con- trolling cell cycle progression such that the per- turbation of additional checkpoints, for example, the DNA damage checkpoint, renders them particularly

sensitive to DNA damaging agents. For example, tumor cells that contain mutant p53 are defective both in the G1 DNA damage checkpoint and in the ability to maintain the G2 DNA damage checkpoint.

(Bunz et al., Science, Nov. 20,1998; 282 (5393): 1497-501; Levine). Checkpoint inhibitors that target initiation of the G2 checkpoint or the S phase checkpoint are expected to further cripple the ability of these tumor cells to repair DNA damage and, therefore, selectively kill them over normal cells. Therefore, checkpoint inhibitors are expected to enhance the therapeutic index, which is a measure of the probability of tumor control relative to the probability of toxicity to normal tissue, of both radiation and systemic chemotherapy.

The ability of checkpoint inhibitors to enhance the therapeutic index may be dependent upon tumor type. Tumors with cell cycle defects comple- mentary to the DNA damage checkpoint pathways may be most sensitive to inhibitor drug treatment. In contrast, DNA-PK inhibitors, another distinct class of potential therapeutic agents, are expected to sensitize tumors independently of cell type. A systematic approach of applying checkpoint inhibi- tors and DNA-PK inhibitors also may be effective in the treatment of metastatic diseases that radiation therapy cannot target.

The checkpoint proteins Atm and Atr are hypothesized to initiate a signal transduction path- way leading to cell cycle arrest in the presence of DNA damage or any block to DNA replication. Atm has been shown to play a role in a DNA damage checkpoint in response to ionizing radiation (IR). Patients lacking functional Atm develop the disease Ataxia- Telangiectasia (A-T). Symptoms of A-T include ex-

treme sensitivity to ionizing radiation (IR), cer- ebellar degeneration, oculotaneous telangiectasias, gonadal deficiencies, immunodeficiencies and in- creased risk of cancer (Shiloh, Eur. J. Hum. Genet 1995; 3 (2): 116-38). Fibroblasts derived from these patients are thought to have defects in G1, S, and G2 checkpoints and are defective in their response to IR (Kastan et al., Cell, Nov. 13,1992; 71 (4): 587-97; Scott et al., Int. J. Radiat. Biol., Dec, 1994; 66 (6 Suppl) : S157-63 ; and Beamish et al., J.

Biol. Chem. Aug. 26,1993; 271 (34): 20486-93).

Therefore, Atm may sense double-strand DNA damage caused by IR and radiomimetic drugs, and signal the cell cycle to arrest, such that damage can be re- paired.

Atr is a checkpoint protein stimulated by agents that cause double strand DNA breaks, single strand DNA breaks, and agents that block DNA radi- ation. Overexpression of Atr in muscle cells on iso-chromosome 3q results in a block to differenti- ation, abnormal centrosome numbers, chromosome in- stability, and abolishes the G1 arrest in response to IR (Smith et al., Nat. Genet., May 1998; 19 (1): 39-46). Overexpression of a kinase inactive, dominant negative mutant of Atr sensitizes cells to IR, ultraviolet light (W), MMS, and cisplatin (Cliby et al., EMBO J. Jan. 2,1998,17 (1): 159-69 and Wright et al., Proc. Na t'1 Acad. Sci. U. S. A., June 23,1998; 95 (13): 7445-50). Cells containing overexpressed, mutant strain Atr also fail to arrest in G2 in response to IR. In addition, Atr is associated with chromosomes in meiotic cells where DNA breaks and abnormal DNA structures persist as a result of the process of meiotic recombination (Keegan et al., Genes Dev. October 1,1996; 10 (19) :

433-37). Atr, like Atm, also senses DNA damage and agents that block DNA replication, as well as initiates a cell cycle arrest at G2 and S for DNA repair.

Chkl is hypothesized to lie downstream from protein kinases Atm and/or Atr in the DNA damage checkpoint signal transduction pathway.

(See, Sanchez et al., Science, 1997; 277: 1497-1501; U. S. Patent No. 6,218,109.) In mammalian cells, Chkl is phosphorylated in response to agents that cause DNA damage including IR, UV, and hydroxyurea (Sanchez et al., 1997; Lui et al., Genes Dev. 2000 ; 14: 1448-1459). The phosphorylation and activation of Chkl in mammalian cells is dependent on Atm (Chen et al., 1999) and Atr (Lui et al., 2000). In the yeast S. pombe, Chkl also appears to be involved in the response to IR and blocks to replication (Boddy et al., 1998; Walworth et al., 1993). Furthermore, Chkl has been shown to phosphorylate both weel (O'Connell et al., EMBO J. 1997; 16: 545-554) and Pdsl (Sanchez et al., Science 1999; 286: 1166-1171) gene products known to be important in cell cycle control. These studies demonstrate that mammalian Chkl plays a role in both the Atm-dependent DNA damage checkpoint leading to arrest at S phase.

However, a role for Chkl in the S phase replication checkpoint in mammalian cells has yet to be eluci- dated. Interestingly, Chkl knockout mice are embryonically lethal, thereby suggesting a role for Chkl in a developing organism in addition to its role in DNA damage checkpoints.

Chkl may invoke a G2 arrest by phosphor- ylating and inactivating Cdc25C, the dual speci- ficity phosphatase that normally dephosphorylates

cyclin B/cdc2 as cells progress into mitosis (Fernery et al., Science, Sep. 5,1997; 277 (5331): 1495-7; Sanchez et al. ; Matsuoka et al. ; and Blasina et al., Curr. Biol., Jan. 14,1999; 9 (1) : 1-10).

This mechanism of regulation of Cdc2 activity stim- ulates cell cycle arrest to prevent cells from entering mitosis in the presence of DNA damage or unreplicated DNA.

SUMMARY OF THE INVENTION The present invention is directed to potent and selective chemosensitizing agents useful in the treatment of diseases and conditions related to DNA damage or lesions in DNA replication. The present compounds are inhibitors of the checkpoint kinase Chkl. In particular, aryl-and heteroaryl- substituted urea compounds have demonstrated sig- nificant activity for inhibiting Chkl.

In one aspect, the present invention is directed to a method of inhibiting checkpoint kinase Chkl comprising the step of administering a compound of formula (I), or a composition containing the same, to an individual. Compounds of formula (I) have a structural formula: wherein:

XI is null,-0-,-S-,-CH2-, or-N (R1)-; X2 is-O-,-S-, or-N (R1)-; Y is O or S; or =Y represents two hydrogen atoms attached to a common carbon atom; W is selected from the group consisting of heteroaryl, aryl, heterocycloalkyl, cycloalkyl, and Ci3 alkyl substituted with a heteroaryl or aryl group; and Z is selected from the group consisting of hydro, aryl, and heteroaryl; wherein said aryl groups of W and Z are optionally substituted with one to four substituents represented by R2, said heteroaryl groups of W and Z are optionally substituted with one to four substit- uents represented by R5, and said heterocycloalkyl and cycloalkyl groups of W are optionally substi- tuted with one to two substituents represented by R6 ; R1 is selected from the group consisting of hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, and aryl; R2 is selected from the group consisting of halogen, C1-6alkyl, C2-6alkenyl, OCF3, NO2, CN, NC, N (R3)2, OR3, CO2R3, C(O) N (R3) 2, C (0) R3, N (Rl) COR N (R1) C (0) OR3, N (R3) C (O) OR3, N (R3) C (O)-C1-3alkylene- C (O) R3, N (R3) C (O)-C1-3alkylene-C(O)OR3, N (R3) C (O)- C1-3alkylene-OR3, N (R3) C (O)-C1-3alkylene-NHC (O) OR3, N (R3) C (O)-C1-3alkylene-SO2NR3, C1-3alkylene-OR3, and SR3; R3 is selected from the group consisting of hydro, C1-6alkyl, C2-6alkenyl, cycloalkyl, aryl, heter- oaryl, SO2R4, C1-6alkyl substituted with one or more of halo, hydroxy, aryl, heteroaryl, heterocycloalk- yl, N (R4)2, and SO2R4, C1-3alkylenearyl, C1-3alkylene- heteroaryl, C1-3alkyleneC3-8heterocycloalkyl, C1-3alk- yleneSO2aryl, optionally substituted C13alkylene- N(R4)2, OCF3, C1-3alkyleneN (R4)3+, C3-8heterocycloalkyl, and CH (cl-3alkyleneN (R4) 2) 2, or two R3 groups are taken

together to form an optionally substituted 3-to 6- membered aliphatic ring; R4 is selected from the group consisting of hydro, C1-6alkyl, cycloalkyl, aryl, heteroaryl, C1-3- alkylenearyl, and SO2C1-6alkyl, or two R4 groups are taken together to form an optionally substituted 3- to 6-membered ring; Rus vis selected from the group consisting of C1-6alkyl, aryl, N (RI) 2, OR3, halo, N3, CN, C1-3alkyl- enearyl, C3alkyleneN (R3) 2, C (O) R3, and O 0 C1_3alkylene-N 0 0 R6 is selected from the group consisting of halo and C16alkyl, and pharmaceutically acceptable salts or solvates thereof.

In another aspect, the present invention is directed to aryl-and heteroaryl-disubstituted urea compounds having a structural formula (II) wherein Y'is O or S; W' is selected from the group consisting of

optionally substituted with from one to four sub- stituents selected from the group consisting of C1-6alkyl, aryl, N (R7) 2' OR7, N3, CN, C (O) R7, C1-3alk- ylenearyl, Cl-3alkyleneN (R12)2, 0 C1_3alkylene-N > \ 0 Z'is selected from the group consisting of:

wherein: Q'is selected from the group consisting of hydrogen, OR', SR', and N (R 2; J'is selected from the group consisting of C-R8, N-R8, O, and S; K'is selected from the group consisting of C-R9, N-R9, O, and S; L'is selected from the group consisting of C-R10, N-R10, O, and S; M'is selected from the group consisting of C-R11, N-R11, O, and S; wherein: R', independently, is selected from the group consisting of hydro, C1-6alkyl, C2-6alkenyl, cycloalkyl, aryl, heteroaryl, SO2R12, C1-6alkyl sub- stituted with one or more of halo, hydroxy, aryl, heteroaryl, heterocycloalkyl, N (R12) 2, and SO2R12, C1-3alkylenearyl, C1-3alkyleneheteroaryl, C1-3alkylene- C3-8heterocycloalkyl, C1-3alkyleneSO2aryl, optionally substituted C1-3alkylenen (R12)2, OCF3, C1-3alkylene- N (R12) 3+, C38heterocycloalkyl, and CH (C1-3alkylene- N (R12)2)2, or two R7 groups are taken together to form an optionally substituted 3-to 6-membered aliphatic ring; R8, R9, and R10 are each independently selected from the group consisting of null, hydro, halo, optionally substituted C1-6alkyl, C2-6alkenyl, OCF3, NO2, CN, NC, N (R) 2, OR, CO2R7, C (O) N (R7) 2, C (O) R7, N (R13) COR7, N (R13) C (O) OR7, N (R7) C (O) OR 7, N (R7) C (O) C1-3alkyleneC (O) R7, N (R7) C (O) C1-3alkylene- C (O) oR7, N (R7) C (O) C1-3alkyleneOR7, N (R7) C (O) C1-3alkyl- eneNHC (O) OR7, N (R7) C (O) C1-3alkyleneSO2NR7, CF3, C1-3- alkyleneN (R12) SO2aryl, C1-3alkyleneN (R12) SO2heteroaryl, C1-3alkyleneOC1-3alkylenearyl, C1-3alkyleneN (R12) C1-3- alkylenearyl, C1-3alkyleneN (R12) C1-3alkyleneheteroaryl,

Cl3alkyleneN (Rl2) C (O) R', C1-3alkyleneN(R12) C (O) C1-3- alkyleneOR2, C1-3alkyleneN (Rl2) C (O) aryl, C1-3alkylene- N (R12) C (O) C1-3alkyleneN(R12)2, C1-3alkylenN(R12) C (0)- heteroaryl, C1-3alkyleneOR7, and SR', wherein R'is as defined above; Rll is selected from the group consisting of null, hydro, C1-6alkyl, and halo; Rl2 is selected from the group consisting of hydro, C1-6alkyl, Ccycloalkyl, aryl, and SO2C1-6alk- yl, or two Rl2 groups are taken together to form a 3- to 6-membered ring; and R13 is selected from the group consisting of hydrogen, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, and aryl; provided that when Q'is hydrogen or OCH3, at least one of R8, R9, and Rl° is not selected from hydrogen, CH3, OCH3, or halo, and pharmaceutically acceptable salts or solvates thereof.

Another aspect of the present invention relates to carbamido-substituted heteroaryl groups having the structural formula (III) wherein W"'is selected from the group consisting of heteroaryl, aryl, heterocycloalkyl, cycloalkyl, and Cl3 alkyl substituted with a hetero- aryl or aryl group;

wherein said aryl groups are optionally substituted with one to four substituents repre- sented by Rl4, said heteroaryl groups are optionally substituted with one to four substituents repre- sented by R18, and said heterocycloalkyl and cyclo- alkyl groups are optionally substituted with one to two substituents represented by Rl9 ; R14 is selected from the group consisting of halo, C1-6alkyl, C2-6alkenyl, OCF3, NO2, CN, NC, N (R16) 2, OR16, CO2R16, C (O) N (R16) 2, C (O) R, N (Rl5) COR N (Rl5) C (O) OR16, N (R16) C (O) OR16, N (R16) C (0) C1-3alkylene- C (O) R16, N (R16) C (O) C1-3 alkyleneC(O) OR16, N (R16) C (O) C1-3- alkyleneOR16, N (R16) C (O) Cl3alkyleneNHC (O) OR16, N (R16) C (O) C1-3alkyleneSO2NR16, C1-3alkyleneOR16, and SR16; Rl5 is selected from the group consisting of hydro, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, and aryl; R16 is selected from the group consisting of hydro, C1-6alkyl, C2-6alkenyl, cycloalkyl, aryl, heteroaryl, SO2R17, Cl-, alkyl substituted with one or more of halo, hydroxy, aryl, heteroaryl, hetero- cycloalkyl, N (R17) 2, and SO2R17, C1-3alkylenearyl, C1-3alkyleneheteroaryl, C1-3alkyleneC3-8heterocyclo- alkyl, C1-3alkyleneSO2aryl, optionally substituted C1-3alkyleneN (R17)2, OCF3, C1-3alkyleneN (R17)3+, C3-8het- erocycloalkyl, CH (C13alkyleneN (Rl') 2) 2 ; or two R16 groups are taken together to form an optionally substituted 3-to 6-membered aliphatic ring.

R17 is selected from the group consisting of hydro, C1-6alkyl, cycloalkyl, aryl, and SO2C1-6- alkyl, or two R17 groups are taken together to form an optionally substituted 3-to 6-membered ring ; R18 is selected from the group consisting of Cl6alkyl, aryl, N (R15)2, OR15, and halo; and

R19 is selected from the group consisting of halo and C,-, alkyl.

The present invention also is directed to pharmaceutical compositions containing one or more compounds of structural formula (II), to use of the compounds and compositions containing the compounds in therapeutic treatment of a disease or disorder, and to methods of preparing the compounds and intermediates involved in the synthesis of the compounds of structural formula (II).

DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS Radiation and most chemotherapeutic agents are therapeutically beneficial because they take advantage of inappropriate tumor cell proliferation.

Cellular processes, such as DNA damage repair and cell cycle checkpoints, protect tumor cells from the toxic effects of physical and chemical agents.

Treatments that modulate the underlying molecular mechanisms of cell cycle progression and resistance to DNA damage can potentiate tumor cell killing and enhance the therapeutic index of existing therapies.

Most chemotherapeutic agents act by dis- rupting DNA metabolism. Because these processes are shared by both normal and tumor cells, and because the maintenance of DNA integrity is essential to cell viability, anticancer drugs have the lowest therapeutic index of any drug class. By identifying and inhibiting cellular processes that tumor cells rely upon, the effectiveness of radiation and chemo- therapy treatment regimens can be enhanced.

The interruption of the DNA damage check- point protein function provides a novel means of killing tumor cells relative to normal cells. For

example, Chkl ensures that cells with unrepaired DNA lesions caused by certain drugs or radiation do not progress through DNA synthesis phase or mitosis un- til chromosomal lesions have been removed. Accord- ingly, a tumor cell treated with a Chkl inhibitor in combination with a DNA damaging agent can kill using lower amounts of DNA damaging agent than tumor cells treated with the DNA damaging agent alone.

Failure of cell cycle checkpoints in normal cells predisposes an individual to, or di- rectly causes, many disease states, such as cancer, ataxia telangiectasia, embryo abnormalities, and various immunological defects associated with aber- rant B and T cell development. The latter are associated with the pathological states of lupus, arthritis, and autoimmune diseases. Intense re- search efforts, therefore, have focused on identi- fying cell cycle checkpoints and the proteins essential for the function of the checkpoints.

Noncancerous tissue having intact cell checkpoints typically is insulated from temporary disruption of a single checkpoint pathway, such as the Chkl pathway. Tumor cells, however, have multiple defects in pathways controlling cell cycle progression such that perturbation of the DNA damage checkpoint can render cells particularly sensitive to DNA damaging agents. Therefore, checkpoint in- hibitors are expected to enhance the therapeutic index, which is a measure of the probability of tumor control relative to the probability of tox- icity to normal tissue to radiation and systemic chemotherapy. In contrast, other classes of inhib- itors may not be amenable to combination chemo- therapy because both normal and tumor tissue may be similarly sensitized.

One aspect of the present invention is directed to a method of inhibiting Chkl, comprising the step of administering a therapeutically effec- tive amount of a compound of formula (I), or a composition containing the same, to an individual.

Compounds of formula (I) have a structural formula wherein Xl is null,-O-,-S-,-CH2-, or- N (R1) ; X2 is-O-,-S-, or-N (R1)-; Y is O or S; or =Y represents two hydrogen atoms attached to a common carbon atom; W is selected from the group consisting of heteroaryl, aryl, heterocycloalkyl, cycloalkyl, and Ci3 alkyl substituted with a heteroaryl or aryl group; and Z is selected from the group consisting of hydrogen, aryl, and heteroaryl ; wherein said aryl groups of W and Z are optionally substituted with one to four substituents represented by R', said heteroaryl groups of W and Z are optionally substituted with one to four substit- uents represented by R5, and said heterocycloalkyl and cycloalkyl groups of W are optionally substi- tuted with one to two substituents represented by RI ; R1 is selected from the group consisting of hydrogen, Cl6alkyl, C26alkenyl, C26alkynyl, and aryl;

R2 is selected from the group consisting of halogen, C1-6alkyl, C2-6alkenyl, OCF3, NO2, CN, NC, N (R3) 2, OR3, CO2R3, C (O) N (R3) 2, C (0) R3, N (R1) COR3, N (R1) C (O) OR3, N (R3) C (O) OR3, N (R3) C (O) C1-3alkyleneC (0) R3, N (R3) C (O) Cl_3alkylene-C (O) OR3, N (R3) C (O) C1-3alkyleneOR3, N (R3) C (O) C1-3alkyleneNHC(O)OR3, N (R3) C (O) C1-3alkylene- So2NR3, C13alkyleneOR3, and SR3 ; R3 is selected from the group consisting of hydro, C1-6alkyl, C2-6alkenyl, cycloalkyl, aryl, heter- oaryl, SO2R4, C1-6alkyl substituted with one or more of halo, hydroxy, aryl, heteroaryl, heterocyclo- alkyl, N (R4)2, and SO2R4, C1-3alkylenearyl, C1-3alkyl- eneheteroaryl, C1-3alkyleneC3-8heterocycloalkyl, C1-3- alkyleneSO2aryl, optionally substituted C13alkylene- N(R4)2, OCF3, C1-3alkyleneN(R4)3+, C3-8heterocycloalkyl, and CH (Cl-3alkyleneN (R4) 2) 2, or two R3 groups are taken together to form an optionally substituted 3-to 6- membered aliphatic ring; R4 is selected from the group consisting of hydro, C1-6alkyl, cycloalkyl, aryl, heteroaryl, C1-3- alkylenearyl, and SO2C1-6alkyl, or two R4 groups are taken together to form an optionally substituted 3- to 6-membered ring; R5 is selected from the group consisting of C1-6alkyl, aryl, N (R3) 2, OR3, halo, N3, CN, C1-3alkyl- enearyl, Cl-3alkyleneN (R3)2, C (O) R3, and 0 Cialkylene-N \ 0

RI is selected from zhe group consisting of halo and C1-6alkyl, and pharmaceutically acceptable salts or solvates thereof.

Preferred compounds used in the method are those wherein X1 and X2 are -N(H)-; Y is O or S; W is heteroaryl containing at least two heteroatoms selected from the group consisting of N, O, and S, said ring is optionally substituted with from one to four substituents selected from the group consisting of C1-6alkyl, aryl, N (R3) 2' OR3, and halo, wherein R3 is as previously defined; Z is selected from the group consisting of: wherein: Q is selected from the group consisting of hydrogen, OR3, SR3, and N (R3) 2 ; J is selected from the group consisting of CR2°, NR 0, and S; K is selected from the group consisting of CR NR21, 0, and S; L is selected from the group consisting of CR22,NR22 O, and S; M is selected from the group consisting of CR23,NR23, 0, and S; wherein:

R20, R21, and R22 are each independently se- lected from the group consisting of null, hydro, halo, C1-6alkyl, C2-6alkenyl, OCF3, NO2, CN, NC, N (R25)2, OR25, Co2R25, C (O) N (R25) 2, C (O) R25, N (R24) COR25, N(R24)- C (O) OR25, N (R25) C (O) OR25, N (R25) C(O) C1-3alkyleneC(O)R25, N (R25) C(O) C1-3alkyleneC(O)OR25, N (R250 C (O) C1-3alkylene- OR25, N (R25) C (O) C1-3alkyleneNHC (O) OR25, N (R25) C (O) C1-3- alkyleneSO2NR25, CF3, Cl3alkyleneN (R25) SO2aryl, C1 3- alkyleneN (R25) SO2heteroaryl, C1-2alkyleneOC1-3alkylene- aryl, C1-3alkyleneN (R25) C1-3alkylenearyl, C1-3alkylene- N (R25) C1-3alkyleneheteroaryl, C1-3alkyleneN (R25) C (O) R7, C1-3alkyleneN (R25) C(O)C1-3alkyleneOR2, C1-3alkylene- N (R25) C (O) aryl, C1-3alkyleneN (R25) C (O) Cl3alkylene- N (R25)2, c1-3alkyleneN (R25) C (O) heteroaryl, C1-3alkyl- eneOR25, and SR25 ; R23 is selected from the group consisting of null, hydro, C1-6alkyl, and halo; R24 is selected from the group consisting of hydro, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, and aryl; R25 is selected from the group consisting of hydro, C1-6alkyl, C2-6alkenyl, cycloal. kyl, aryl, heteroaryl, SO2R26, and C16alkyl substituted with halo, hydroxy, aryl, heteroaryl, heterocycloalkyl, N (R26) 2, or SO2R26 ; and R26 is selected from the group consisting of hydro, C1-6alkyl, cycloalkyl, aryl, and SO2C1-6- alkyl, or two R26 groups are taken together to form an optionally substituted 3-to 6-membered ring.

More preferred compounds of the method are those of structural formula (I) wherein W is se- lected from the group consisting of pyridazinyl, pyrimidinyl, pyrazinyl, and triazinyl, optionally substituted with from one to four substituents selected from the group consisting of C1-6alkyl, aryl, N (R3) 2, OR3, N3, CN, C (0) R7, C1-3alkylenearyl, C1-3alkyleneN (R4),

0 o o o and halo, wherein R3, Xl, X2, Y, and Z are as defined for formula (I).

Additional preferred compounds of formula (I) are those wherein W is selected from the group consisting of pyridazine, pyrimidine, pyrazine, and triazine, optionally substituted with from one to four substituents selected from the group consisting of optionally substituted C16alkyl, aryl, N (R3) 2, OR3, <BR> <BR> <BR> <BR> Cl3alkylenearyl, Cl3alkyleneheteroaryl, Cl3alkylene- C3-8heterocycloalkyl, C1-3alkylenesO2aryl, optionally substituted C13alkyleneN (R4)2, OCF3, C1-3alkylene- N(R4)3+, C3-8heterocycloalkyl, CH (C1-3alkyleneN (R4)2)2, and halo; Xl and X2 are-N (H)- ; Y is O or S; and Z is selected from the group consisting of: wherein R3, Q, J, K, L, and M are as previously defined.

Compounds preferred for use in the method also include those of formula (I) wherein J is se-

lected from the group consisting of CRO and NR20, wherein R20 is null, hydro, C1-6alkyl, and halo; K is selected from the group consisting of CR21 and NR21 ; L is selected from the group consisting of CR22 and NR22 ; and one of R21 and R22 is hydro and the other is a substituent selected from the group consisting of CO2R25, C (O) N (R25)2, C (O) R25, N (R24) COR25, N (R24) C (O) OR 2-5, N (R25) C (O) OR25, N (R25) C (O) C1-3alkyleneC (O) R25, N (R25)- C (O) C13alkyleneC (O) OR25, N (R25) C (O) C1-3alkyleneOR25, N (R25) C (O) C1-3alkyleneNHC (O) OR25, N (R25) C (O) c1-3alkyl- eneSO2NR25, C1-3alkyleneOR25, and SR25, wherein R, R W, X1, X2, Y and M are as previously defined.

Compounds useful in the method also in- clude structures of formula (I) wherein X1 is null, X2 is-N (H)-, Y is O, Z is hydro, and W is as previ- ously defined.

The method of inhibiting Chkl also can be used to sensitize a tumor cell to a chemotherapeutic agent. As such, the present invention also is directed to a method of sensitizing a tumor cell to a chemotherapeutic agent comprising administering a compound of formula (I), or a salt, solvate, or derivative thereof, or a composition comprising the same, to an individual.

In another aspect, the present invention is directed to aryl-and heteroaryl-disubstituted urea compounds having a structural formula (II)

wherein Y' is O or S; W' is selected from the group consisting of

optionally substituted with from one to four sub- stituents selected from the group consisting of C1-6alkyl, aryl, N (R') 2, OR7, N3, CN, C (O) R7, C1-3alk- ylenearyl, C1-3alkyleneN (R12)2,

0 po Cl-3alkylene-N I // o Z'is selected from the group consisting of: wherein: Q'is selected from the group consisting of hydrogen, OR', SR', and N (R7)2; J'is selected from the group consisting of C-R8, N-R8, O, and S; K'is selected from the group consisting of C-R9, N-R9, O, and S; L'is selected from the group consisting of C-R10, N-R10, O, and S ; M'is selected from the group consisting of C-R11, N-R11, O, and S ; wherein: R', independently, is selected from the group consisting of hydro, C1-6alkyl, C2-6alkenyl, cycloalkyl, aryl, heteroaryl, SO2R12, C1-6alkyl substituted with one or more of halo, hydroxy, aryl, heteroaryl, heterocycloalkyl, N (R) 2, and SO2R12, C1-3alkylenearyl, C1-3alkyleneheteroaryl, C1-3alkylene-

C3-8heterocycloalkyl, C1-3alkyleneSo2aryl, optionally substituted Cl3alkyleneN (Rl2) 2, OCF3, C1-3alkylene- N (R12)3+, C3-8heterocycloalkyl, and CH (C1-3alkylene- N (R12)2)2, or two R7 groups are taken together to form an optionally substituted 3-to 6-membered aliphatic ring; R8, R9, and and are each independently selected from the group consisting of null, hydro, halo, optionally substituted C1-6alkyl, C2-6alkenyl, OCF3, NO2, CN, NC, N (R7)2, OR7, CO2R7, C (0) N (R') 2, C (O) R', N (R13) COR7, N (R13) C (O) OR', N (R') C (O) OR', N (R') C (O) C1-3alkyleneC (O) R7, N (R') C (O) C1-3alkYLENE- C (O) OR7, N (R7) C (O) C1-3alkyleneOR7, N (R') C (O) C1-3alkyl- eneNHC (O) OR7, N(R7) C (O) C1-3alkyleneSO2NR7, cF3, C1-3- alkyleneN (R12) SO2aryl, C1-3alkyleneN (R12) SO2heteroaryl, C1-3alkyleneOC1-3alkylenearyl, C1-3alkyleneN (R12) C1-3- alkylenearyl, C1-3alkyleneN (R12) C1-3alkyleneheteroaryl, C1-3alkyleneN (R12) C (O) R', C1-3alkyleneN (R12) C (O) C1-3alk- yleneOR2, C1-3alkyleneN (R12) C(O) aryl, C1-3alkyleneN (R12)- C (O) C1-3alkyleneN (R12)2, C1-3alkyleneN (R12) C (O) hetero- aryl, C1-3alkyleneOR7, and SR, wherein R'is as defined above; R1l is selected from the group consisting of null, hydro, optionally substituted C1-6alkyl, and halo; R12 is selected from the group consisting of hydro, C1-6alkyl, cycloalkyl, aryl, heteroaryl, C1-3alkylenearyl, and SO2C1-6alkyl, or two R12 groups are taken together to form an optionally substituted 3-to 6-membered ring; and R13 is selected from the group consisting of hydro, C1-6alkyl, C2-6alkenyl, C2-6alkynyl, and aryl; provided that when Q'is hydro or OCH3, at least one of R8, R9, and R10 is different from hydro, CH3, OCH2, and halo,

and pharmaceutically acceptable salts or solvates thereof.

Preferred compounds of structural formula (II) are those wherein W' is selected from the group consisting of optionally substituted with one to four substituents selected from the group consisting of C16alkyl, optionally substituted aryl, N (R') 2, CF3, C (0) R7, N3, CN, C1-3alkylenearyl, C1-3alkyleneN (R12)2, OR7, halo, 0 Cl-3alkylene-N 1 \ o wherein R7, Y and Z are as previously defined.

More preferred compounds of formula (II) are those wherein:

J'is selected from the group consisting of CR8 and NR8, wherein RI is null, hydro, C1-6alkyl, and halo; K'is selected from the group consisting of CR9 and NR9 ; L'is selected from the group consisting of CR10 and NR10 ; and one of RI and R10 is hydro and the other is a substituent selected from the group consisting of Cozy C (O) N (R7)2, C (0) R7, N (R13) COR7, N (R13) C (0) OR7, N (R7) C (O) oR7, N (R') C (O) C1-3alkyleneC (O) R7, N (R7)C- (O)C1-3alkyleneC(O)OR7, N (R7) C (O) C1-3alkyleneOR7, N (R7) C (O) C13alkyleneNHC (O) OR7, N (R7) C (O) C13alkylene- SO2NR7, C1-3alkyleneOR7, CF3, C1-3alkyleneN (R12) SO2aryl, C1-3alkyleneN (R12) SO2heteroaryl, C1-3alkyleneOC1-3alkyl- enearyl, C1-3alkyleneN (R12) C1-3alkylenearyl, C1-3alk- yleneN (R12) C1-3alkyleneheteroaryl, c1-3alkyleneN (R12)- C (0) R7, C1-3alkyleneN (R12) C (0) C1-3alkyleneOR2, C1-3alk- yleneN (R12) C (O) aryl, C1-3alkyleneN (R12) C (O) C1-3alkylene- N (R12) 2, C1 3alkyleneN (R12) C (O) heteroaryl, and SR7, wherein R7, R13, W', Y', and M'are as previously defined.

Yet another aspect of the invention re- lates to compounds, and compositions containing compounds, of structural formula (III)

wherein W'is selected from the group consisting of heteroaryl, aryl, heterocycloalkyl, cycloalkyl, and C13 alkyl substituted with a hetero- aryl or aryl group; wherein said aryl groups are optionally substituted with one to four substituents repre- sented by R14, said heteroaryl groups are optionally substituted with one to four substituents repre- sented by R", and said heterocycloalkyl and cyclo- alkyl groups are optionally substituted with one to two substituents represented by R19 ; R14 is selected from the group consisting of halo, C1-6alkyl, C2-6alkenyl, OCF3, NO2, CN, NC, N (R16) 2, OR16, CO2R16, C (O) N (R16) 2, C (O) R16, N (R15) COR16, N (R15) C (0) OR'6, N (R16) C (O) OR16, N (R16) C (O) cl3alkyler C (0) R16, N (R16) C (O) C1-3alkyleneC(O) OR16, N (R16) C (0) C1-3alkyleneOR16, N (R) C (0) C13alkyleneNHC (O) OR16, N (R16) C (O) C1-3alkyleneSO2NR16, C1-3alkyleneOR16, and SR16 ; R15 is selected from the group consisting of hydro, C1-6alkyl, C2-6alkenyl, c2-6alkynyl, and aryl; R16 is selected from the group consisting of hydro, C1-6alkyl, C2-6alkenyl, cycloalkyl, aryl, heteroaryl, SO2R1', and C16alkyl substituted with one or more of halo, hydroxy, aryl, heteroaryl, hetero- cycloalkyl, N (R) 2, and SO2R17, C1-3alkylenearyl, C1-3- alkyleneheteroaryl, C1-3alkyleneC3-8heterocycloalkyl, C1-3alkyleneSO2aryl, optionally substituted C1-3alkyl- eneN (R17)2, OC3, C1-3alkyleneN (R1703+, C3-8heterocyclo- alkyl, CH (Cl3alkyleneN (R17)2)2, or two R16 groups are taken together to form a 3-to 6-membered aliphatic ring.

R17 is selected from the group consisting of hydro, C1-6alkyl, cycloalkyl, aryl, and SO2C1-6- alkyl, or two R17 groups are taken together to form an optionally substituted 3-to 6-membered ring;

R18 is selected from the group consisting of C1-6alkyl, aryl, N (Rl5) 21 OR", and halo; and R"is selected from the group consisting of halo and Cl6alkyl.

As used herein, the term"alkyl"includes straight chained and branched hydrocarbon groups containing the indicated number of carbon atoms, typically methyl, ethyl, and straight-chain and branched propyl and butyl groups. Unless otherwise indicated, the hydrocarbon group can contain up to 20 carbon atoms. The term"alkyl"includes"bridged alkyl,"i. e., a C6-C16 bicyclic or polycyclic hydro- carbon group, for example, norbornyl, adamantyl, bicyclo [2.2.2] octyl, bicyclo [2.2.1] heptyl, bicyclo- [3.2.1] octyl, or deca hydronaphthyl. Alkyl groups can be substituted, for example, with hydroxy (OH), halo, aryl, heteroaryl, cycloalkyl, heterocyclo- alkyl, amino (N (Ra)2), and sulfonyl (SO2Ra), wherein Ra is selected from the group consisting of hydro, C16alkyl, cycloalkyl, aryl, and SO2C16alkyl, or two Ra groups are taken together to form an optionally substituted 3-to 6-membered ring.

The term"cycloalkyl"is defined as a cyclic C3-shydrocarbon group, e. g., cyclopropyl, cyclobutyl, cyclohexyl, and cyclopentyl."Hetero- cycloalkyl"is defined similarly as cycloalkyl, and includes bicyclic and polycyclic groups, except the ring contains one to three heteroatoms selected from the group consisting of oxygen, nitrogen, and sulfur. Cycloalkyl and heterocycloalkyl groups can be saturated or partially unsaturated ring systems substituted with, for example, one to three groups, independently selected from C14alkyl, C13alkyleneOH, C (=O) NH2, NH2, oxo (=O), aryl, trifluoroethanoyl, and OH. Heterocycloalkyl groups optionally are further

N-substituted with Cl3alkylenearyl or C1-3alkylene- heteroaryl.

The term"alkenyl"is defined identically as"alkyl,"except the substituent contains a carbon-carbon double bond.

The term"alkynyl"is defined identically as"alkyl,"except the substituent contains a carbon-carbon triple bond.

The term"alkylene"refers to an alkyl group having a substituent. For example, the term "C1-3alkyleneC (O) OR" refers to an alkyl group con- taining one to three carbon atoms substituted with a -C (O) OR group. The alkylene group is optionally substituted with one or more of aryl, heteroaryl, and OR', wherein R7 is defined hereafter.

The term"halo"or"halogen"is defined herein to include fluorine, bromine, chlorine, and iodine.

The term"aryl,"alone or in combination, is defined herein as a monocyclic or polycyclic aromatic group, preferably a monocyclic or bicyclic aromatic group, e. g., phenyl or naphthyl. Unless otherwise indicated, an"aryl"group can be unsub- stituted or substituted, for example, with one or more, and in particular one to four, halo, C1-6alkyl, C2-6alkenyl, OCF3, NO2, CN, NC, N (Ra) 2, ORb, CO2Rb, C (O) N (Rb) 2, C (O) Rb, N (Ra) CORb, N (Ra) C (O) ORb, N (Ra) C (0)- OR, N (Ra) C (O) Cl3alkyleneC (O) Rb, N (Rb). C (O) C13alkylene- C (O) ORb, N (Rb) C (O) C1-3alkyleneORb, N (Rb) C (O) C1-3alkyl- eneNHC (O) ORb, N (Rb) C (O) C1-3alkyleneSO2NRb, C1-3alkyl- eneORb, and SRb, wherein Rb is selected from the group consisting of hydro, C1-6alkyl, C2-6alkenyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, S02Ra, and C1-6alkyl substituted with halo, hydroxy, aryl, heteroaryl, heterocycloalkyl, N (Ra)2, or SO2Ra,

and Ra, as previously defined. Exemplary aryl groups include phenyl, naphthyl, tetrahydronaphthyl, 2- chlorophenyl, 3-chlorophenyl, 4-chlorophenyl, 2- methylphenyl, 4-methoxyphenyl, 3-trifluoromethyl phenyl, 4-nitrophenyl, 2-methoxyphenyl, 2,4-methoxy- chlorophenyl, and the like. The terms''arylCl3- alkyi"and"heteroarylCialky !" are defined as an aryl or heteroaryl group having a C13alkyl substitu- ent.

The term"heteroaryl"is defined herein as a monocyclic or bicyclic ring system containing one or two aromatic rings and containing at least one nitrogen, oxygen, or sulfur atom in an aromatic ring, and that can be unsubstituted or substituted, for example, with one or more, and in particular one to four, substituents, for example, hydrogen, Cl_6- alkyl, C16alkoxy, aryl, N (Ra) 2, oRb, and halo, wherein Ra and Rb are as previously defined. Examples of heteroaryl groups include, but are not limited to, thienyl, furyl, pyridyl, oxazolyl, quinolyl, iso- quinolyl, indolyl, triazolyl, isothiazolyl, isoxazolyl, imidizolyl, benzothiazolyl, pyrazinyl, pyrimidinyl, thiazolyl, and thiadiazolyl.

The term"hydroxy"is defined as-OH.

The term"3-to 6-membered ring"as used herein refers to carbocyclic and heterocyclic aliphatic or aromatic groups, including, but not limited to, morpholinyl, piperldinyl, phenyl, thio- phenyl, furyl, pyrrolyl, imidazolyl, pyrimidinyl, and pyridinyl, optionally substituted with one or more, and in particular one to three, groups exemplified above for"aryl"groups.

The carbon atom content of hydrocarbon- containing moieties is indicated by a subscript designating the minimum and maximum number of carbon

atoms in the moiety, e. g., "C1-6alkyl" refers to an alkyl group having one to six carbon atoms, inclu- sive.

In the structures herein, for a bond lack- ing a substituent, the substituent is methyl, for example, When no substituent is indicated as attached to a carbon atom on a ring, it is under- stood that the carbon atom contains the appropriate number of hydrogen atoms. In addition, when no substituent is indicated as attached to a carbonyl group or a nitrogen atom, for example, the sub- stituent is understood to be hydrogen, e. g., The abbreviation"Me"is methyl. The abbreviation CO and C (O) is carbonyl (C= (O)).

The notation N (RX) 2, wherein x represents an alpha or numeric character, such as for example Ra, Rb, R4, R, and the like, is used to denote two Rx groups attached to a common nitrogen atom. When used in such notation, the Rx group can be the same

or different, and is selected from the group as defined by the R'group.

The present invention also is directed to pharmaceutical compositions containing one or more compounds of structural formula (II) and (III), to use of the compounds and compositions containing the compounds in therapeutic treatment of a disease or disorder, and to methods of preparing the compounds and intermediates involved in the synthesis of the compounds of structural formula (II) and (III).

Compounds useful for the method of the present invention have demonstrated activity in inhibiting Chkl in vitro. Compounds of the present invention have demonstrated selectivity for Chkl as against other protein kinases including Cdc2, Chk2, Atr, DNA-PK, PKA, and CaM KII.

Compounds of the present invention can be used to potentiate the therapeutic effects of radi- ation and/or chemotherapeutics used in the treatment of cancers and other cell proliferation disorders in humans or animals. For example, compounds of the invention can be used to enhance treatment of tumors that are customarily treated with an antimetabolite, e. g., methotrexate or 5-fluorouracil (5-FU). The method of the present invention comprises adminis- tration of a Chkl inhibitor compound in combination with a chemotherapeutic agent that can effect single-or double-strand DNA breaks or that can block DNA replication or cell proliferation.

Alternatively, the method of the present invention comprises administration of a Chkl inhibitor com- pound in combination with therapies that include use of an antibody, e. g., herceptin, that has activity in inhibiting the proliferation of cancer cells.

Accordingly, cancers such as colorectal cancers,

head and neck cancers, pancreatic cancers, breast cancers, gastric cancers, bladder cancers, vulvar cancers, leukemias, lymphomas, melanomas, renal cell carcinomas, ovarian cancers, brain tumors, osteo- sarcomas, and lung carcinomas, are susceptible to enhanced treatment in combination with the Chkl inhibitors of the invention.

Tumors or neoplasms include growths of tissue cells wherein multiplication of cells is uncontrolled and progressive. Some such growths are benign, but others are termed"malignant,"and can lead to death of the organism. Malignant neoplasms, or"cancers,"are distinguished from benign growths in that, in addition to exhibiting aggressive cellular proliferation, can invade surrounding tissues and metastasize. Moreover, malignant neo- plasms are characterized by showing a greater loss of differentiation (greater"dedifferentiation") and organization relative to one another and surrounding tissues. This property is called"anaplasia." Neoplasms treatable by the present inven- tion also include solid tumors, i. e., carcinomas and sarcomas. Carcinomas include malignant neoplasms derived from epithelial cells which infiltrate (i. e., invade) surrounding tissues and give rise to metastases. Adenocarcinomas are carcinomas derived from glandular tissue, or from tissues that form recognizable glandular structures. Another broad category of cancers includes sarcomas, which are tumors whose cells are embedded in a fibrillar or homogeneous substance, like embryonic connective tissue. The invention also enables treatment of cancers of the myeloid or lymphoid systems, including leukemias, lymphomas, and other cancers that typically are not present as a tumor mass, but

are distributed in the vascular or lymphoreticular systems.

Chkl activity is associated with various forms of cancer in, for example, adult and pediatric oncology, growth of solid tumors/malignancies, myxoid and round cell carcinoma, locally advanced tumors, metastatic cancer, human soft tissue sar- comas, including Ewing's sarcoma, cancer metastases, including lymphatic metastases, squamous cell carcinoma, particularly of the head and neck, esoph- ageal squamous cell carcinoma, oral carcinoma, blood cell malignancies, including multiple myeloma, leukemias, including acute lymphocytic leukemia, acute nonlymphocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, and hairy cell leukemia, effusion lymphomas (body cavity based lymphomas), thymic lymphoma lung cancer (including small cell carcinoma, cutaneous T-cell lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, cancer of the adrenal cortex, ACTH-producing tumors, nonsmall cell cancers, breast cancer, including small cell carcinoma and ductal carcinoma), gastro- intestinal cancers (including stomach cancer, colon cancer, colorectal cancer, and polyps associated with colorectal neoplasia), pancreatic cancer, liver cancer, urological cancers (including bladder can- cer, such as primary superficial bladder tumors, invasive transitional cell carcinoma of the bladder, and muscle-invasive bladder cancer', prostate can- cer, malignancies of the female genital tract (including ovarian carcinoma, primary peritoneal epithelial neoplasms, cervical carcinoma, uterine endometrial cancers, vaginal cancer, cancer of the vulva, uterine cancer and solid tumors in the ovarian follicle), malignancies of the male genital

tract (including testicular cancer and penile can- cer), kidney cancer (including renal cell carcinoma, brain cancer (including intrinsic brain tumors, neuroblastoma, astrocytic brain tumors, gliomas, and metastatic tumor cell invasion in the central ner- vous system), bone cancers (including osteomas and osteosarcomas), skin cancers (including malignant melanoma, tumor progression of human skin keratino- cytes, and squamous cell cancer), thyroid cancer, retinoblastoma, neuroblastoma, peritoneal effusion, malignant pleural effusion, mesothelioma, Wilms's tumors, gall bladder cancer, trophoblastic neo- plasms, hemangiopericytoma, and Kapos's sarcoma.

Compounds of the present invention also can potentiate the efficacy of drugs in the treat- ment of inflammatory diseases. Examples of diseases that can benefit from combination therapy with com- pounds suitable for the method of the present inven- tion are rheumatoid arthritis, psoriasis, vitiligo, Wegener's granulomatosis, and systemic lupus erythematosus (SLE). Treatment of arthritis, Wegener's granulomatosis, and SLE often involves the use of immunosuppressive therapies, such as ionizing radiation, methotrexate, and cyclophosphamide. Such treatments typically induce, either directly or indirectly, DNA damage. Inhibition of Chkl activity within the offending immune cells render the cells more sensitive to control by these standard treat- ments. Psoriasis and vitiligo commonly are treated with ultraviolet radiation (UV) in combination with psoralen. The present DNA damaging agents induce the killing effect of W and psoralen, and increase the therapeutic index of this treatment regimen. In general, compounds useful in methods of the present invention potentiate control of inflammatory disease

cells when in combination with currently used immunosuppressive drugs.

The present invention includes all possible stereoisomers and geometric isomers of compounds of the present method and of structural formulae (I), (II), and (III). The present inven- tion includes not only racemic compounds, but optically active isomers as well. When a compound of structural formulae (I), (II), or (III) is de- sired as a single enantiomer, it can be obtained either by resolution of the final product or by stereospecific synthesis from either isomerically pure starting material or use of a chiral auxiliary reagent, for example, see Z. Ma et al., Tetrahedron : Asymmetry, 8 (6), pages 883-888 (1997). Resolution of the final product, an intermediate, or a starting material can be achieved by any suitable method known in the art. Additionally, in situations where tautomers of the compounds of structural formulae (I), (II), and (III) are possible, the present invention is intended to include all tautomeric forms of the compounds. As demonstrated hereafter, specific stereoisomers can exhibit an exceptional ability to inhibit Chkl in combination with chemo- or radiotherapy with diminshed adverse effects typically associated with chemotherapeutic or radio- therapeutic treatments.

Prodrugs of compositions of structural formulae (I), (II), and (III) also can be used as the compound and in the method of the present inven- tion. It is well established that a prodrug ap- proach, wherein a compound is derivatized into a form suitable for formulation and/or administration, and then is released as a drug in vivo, has been successfully employed to transiently (e. g., biore-

versibly) alter the physicochemical properties of the compound (see, H. Bundgaard, Ed., Design of Prodrugs, Elsevier, Amsterdam, (1985); R. B. Silver- man, The Organic Chemistry of Drug Design and Drug Action, Academic Press, San Diego, chapter 8, (1992); K. M. Hillgren et al., Med. Res. Rev., 15,83 (1995)).

Compounds of the present invention can contain several functional groups. The introduced functional groups, if desired or necessary, then can be modified to provide a prodrug for dose of formu- lation and/or administration. Suitable prodrugs include, for example, acid derivatives, like amides, esters, and the like. It also is appreciated by those skilled in the art that N-oxides can be used as a prodrug.

As used herein, the term pharmaceutically acceptable salts refers compounds of structural formula (I), (II), and (III) which contain acidic moieties and form salts with suitable cations.

Suitable pharmaceutically acceptable cations include alkali metal (e. g., sodium or potassium) and alkaline earth metal (e. g., calcium or magnesium) cations. The pharmaceutically acceptable salts of the compounds of structural formula (I), (II), and (III), which contain a basic center, are acid addi- tion salts formed with pharmaceutically acceptable acids. Examples include the hydrochloride, hydro- bromide, sulfate or bisulfate, phosphate or hydrogen phosphate, acetate, benzoate, succinate, fumarate, maleate, lactate, citrate, tartrate, gluconate, methanesulfonate, benzene sulphonate, and p-toluene- sulphonate salts. In light of the foregoing, any reference to compounds of the present invention appearing herein is intended to include compounds of

structural formula (I), (II), and (III), as well as pharmaceutically acceptable salts and solvates thereof.

The compounds of the present invention can be therapeutically administered as the neat chemi- cal, but it is preferable to administer compounds of structural formula (I), (II), and (III) as a pharma- ceutical composition or formulation. Accordingly, the present invention further provides pharmaceuti- cal formulations comprising a compound of structural formula (I), (II), and/or (III), or pharmaceutically acceptable salts thereof, together with one or more pharmaceutically acceptable carriers and, option- ally, other therapeutic and/or prophylactic ingredi- ents. The carriers are"acceptable"in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.

Inhibition of the checkpoint kinase typi- cally is measured using a dose-response assay in which a sensitive assay system is contacted with a compound of interest over a range of concentrations, including concentrations at which no or minimal effect is observed, through higher concentrations at which partial effect is observed, to saturating con- centrations at which a maximum effect is observed.

Theoretically, such assays of the dose-response effect of inhibitor compounds can be described as a sigmoidal curve expressing a degree of inhibition as a function of concentration. The curve also theo- retically passes through a point at which the con- centration is sufficient to reduce activity of the checkpoint enzyme to a level that is 50% that of the difference between minimal and maximal enzyme activ- ity in the assay. This concentration is defined as

the Inhibitory Concentration (50%) or IC, o value.

Determination of IC50 values preferably are made using conventional biochemical (acellular) assay techniques or cell-based assay techniques.

Comparisons of the efficacy of inhibitors often are provided with reference to comparative IC., values, wherein a higher IC., indicates that the test compound is less potent, and a lower IC50 indicates that the compound is more potent, than a reference compound. Compounds useful in the method of the present invention demonstrate an IC51) value of at least 0.1 nM when measured using the dose-response assay. Preferred compounds demonstrate an IC. 50 value of less than 10 uM. More preferred compounds demon- strate an IC50 value of less than 500 nM. Still more preferred compounds of the. present invention demon- strate an IC50 value of less than 250 nM, less than 100 nM, or less than 50 nM.

Compounds and pharmaceutical compositions suitable for use in the present invention include those wherein the active ingredient is administered in an effective amount to achieve its intended purpose. More specifically, a"therapeutically effective amount"means an amount effective to inhibit development of, or to alleviate the existing symptoms of, the subject being treated. Determina- tion of the effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.

A"therapeutically effective dose"refers to that amount of the compound that results in achieving the desired effect. Toxicity and thera- peutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cul- tures or experimental animals, e. g., for determining

the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index, which is expressed as the ratio of LD50 to ED50. Compounds that exhibit high therapeutic indices (i. e., a toxic dose that is substantially higher than the effective dose) are preferred. The data obtained can be used in formulating a dosage range for use in humans. The dosage of such com- pounds preferably lies within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage can vary within this range depending upon the dosage form employed, and the route of administration utilized.

The exact formulation, route of adminis- tration, and dosage is chosen by the individual physician in view of the patient's condition. Dos- age amount and interval can be adjusted individually to provide plasma levels of the active compound that are sufficient to maintain desired therapeutic effects.

Pharmaceutical compositions of the inven- tion can be formulated to include one or more cyto- kines, lymphokines, growth factors, or other hema- topoietic factors which can reduce negative side effects that may arise from, or be associated with, administration of the pharmaceutical composition alone. Cytokines, lymphokines, growth factors, or other hematopoietic factors particularly useful in pharmaceutical compositions of the invention in- clude, but are not limited to, M-CSF, GM-CSF, TNF, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL- 9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IFN, TNF, G-CSF, Meg-CSF, GM-CSF,

thrombopoietin, stem cell factor, erythropoietin, angiopoietins, including Ang-1, Ang-2, Ang-4, Ang-Y, and/or the human angiopoietin-like polypeptide, vascular endothelial growth factor (VEGF), angio- genin, bone morphogenic protein-1 (BMP-1), BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, BMP-7, BMP-8, BMP-9, BMP-10, BMP-11, BMP-12, BMP-13, BMP-14, BMP-15, BMP receptor IA, BMP receptor IB, brain derived neuro- trophic factor, ciliary neutrophic factor, ciliary neutrophic factor receptor cytokine-induced neutro- phil chemotactic factor 1, cytokine-induced neutro- phil chemotactic factor 2, cytokine-induced neutro- phil chemotactic factor 2, endothelial cell growth factor, endothelin 1, epidermal growth factor, epithelial-derived neutrophil attractant, fibroblast growth factor (FGF) 4, FGF 5, FGF 6, FGF 7, FGF 8, FGF 8b, FGF 8c, FGF 9, FGF 10, FGF acidic, FGF basic, glial cell line-derived neutrophic factor receptor 1, glial cell line-derived neutrophic factor receptor 2, growth. related protein, growth related protein, growth related protein, growth related protein, heparin binding epidermal growth factor, hepatocyte growth factor, hepatocyte growth factor receptor, insulin-like growth factor I, insulin-like growth factor receptor, insulin-like growth factor II, insulin-like growth factor binding protein, keratinocyte growth factor, leukemia inhib- itory factor, leukemia inhibitory factor receptor, nerve growth factor nerve growth factor receptor, neurotrophin-3, neurotrophin-4, placenta growth factor, placenta growth factor 2, platelet-derived endothelial cell growth factor, platelet derived growth factor, platelet derived growth factor A chain, platelet derived growth factor AA, platelet derived growth factor AB, platelet derived growth

factor B chain, platelet derived growth factor BB, platelet derived growth factor receptor, platelet derived growth factor receptor, pre-B cell growth stimulating factor, stem cell factor, stem cell factor receptor, transforming growth factor (TGF), TGF, TGF 1, TGF 1.2, TGF 2, TGF 3, TGF 5, latent TGF 1, TGF, binding protein I, TGF binding protein II, TGF binding protein III, tumor necrosis factor receptor type I, tumor necrosis factor receptor type II, urokinase-type plasminogen activator receptor, vascular endothelial growth factor, and chimeric proteins and biologically or immunologically active fragments thereof.

The compounds useful according to the in- vention may be conjugated or linked to auxiliary moieties that promote any property of the compounds that may be beneficial in methods of therapeutic use. Such conjugates can enhance delivery of the compounds to a particular anatomical site or region of interest (e. g., a tumor), enable sustained thera- peutic concentrations of the compounds in target cells, alter pharmacokinetic and pharmacodynamic properties of the compounds, and/or improve the therapeutic index or safety profile of the com- pounds. Suitable auxiliary moieties include, for example, amino acids, aligopeptides, or polypep- tides, e. g., antibodies such as monoclonal antibod- ies and other engineered antibodies; and natural or synthetic ligands to receptors in target cells or tissues. Other suitable auxiliaries include fatty acid or lipid moieties, to promote biodistribution or uptake of the compound by target cells (see, e. g., Bradley et al., Clin. Cancer Res. (2001) 7: 3229.

The therapeutic index of compositions com- prising one or more compounds of the invention can be enhanced by conjugation of the compound (s) with antitumor antibodies as previously described (for example, Pietersz and McKinzie, Immunol. Rev. (1992) 129: 57; Trail et al., Science (1993) 261: 212; Rowlinson-Busza and Epenetos, Curr. Opin. Oncol.

1992; 4: 1142). Tumor directed delivery of compounds of the invention would enhance the therapeutic bene- fit by minimizing potential nonspecific toxicities which can result from radiation treatment or chemo- therapy. In another aspect, Chkl inhibitors and radioisotopes or chemotherapeutic agents can be conjugated to the same antibody molecule. Alterna- tively, Chkl inhibitor-conjugated tumor specific antibodies can be administered before, during, or after administration of chemotherapeutic-conjugated antitumor antibody or radioimmunotherapy.

Compounds of the present invention can enhance the therapeutic benefit of radiation and chemotherapy treatment, including induction chemo- therapy, primary (neoadjuvant) chemotherapy, and both adjuvant radiation therapy and adjuvant chemo- therapy. In addition, radiation and chemotherapy are frequently indicated as adjuvants to surgery in the treatment of cancer. The goal of radiation and chemotherapy in the adjuvant setting is to reduce the risk of recurrence and enhance disease-free survival when the primary tumor has been controlled.

Chemotherapy is utilized as a treatment adjuvant for colon, lung, and breast cancer, frequently when the disease is metastatic. Adjuvant radiation therapy is indicated in several diseases including colon, lung, and breast cancers as described above. For example, radiation frequently is used both pre-and

post-surgery as components of the treatment strategy for rectal carcinoma. Compounds for the present invention are therefore particularly useful follow- ing surgery in the treatment of cancer in combina- tion with radio-and/or chemotherapy.

A compound of the present invention also can radiosensitize a cell. The term"radiosensi- tize,"as used herein, is defined as a molecule, preferably a low molecular weight molecule, admin- istered to human or other animal in a therapeuti- cally effective amount to increase the sensitivity of the cells to be radiosensitized to electromag- netic radiation and/or to promote the treatment of diseases that are treatable with electromagnetic radiation. Diseases that are treatable with electromagnetic radiation include neoplastic di- seases, benign and malignant tumors, and cancerous cells.

Electromagnetic radiation treatment of other diseases not listed herein also is con- templated by the present invention. The terms "electromagnetic radiation"and"radiation"as used herein include, but are not limited to, radiation having the wavelength of 10-2° to 100 meters. Pre- ferred embodiments of the present invention employ the electromagnetic radiation of: gamma-radiation (10-2° to 10-l3 m), X-ray radiation (10-12 to 10-9 m), ultraviolet light (10 nm to 400 nm), visible light (400 nm to 700 nm), infrared radiation (700 nm to 1.0 mm), and microwave radiation (1 mm to 30 cm).

Many cancer treatment protocols currently employ radiosensitizers activated by electromagnetic radiation, e. g., X-rays. Examples of X-ray-acti- vated radiosensitizers include, but are not limited to, the following: metronidazole, misonidazole,

desmethylmisonidazole, pimonidazole, etanidazole, nimorazole, mitomycin C, RSU 1069, SR 4233, E09, RB 6145, nicotinamide, 5-bromodeoxyuridine (BUdR), 5- iododeoxyuridine (IUdR), bromodeoxycytidine, fluoro- deoxyuridine (FUdR), hydroxyurea, cisplatin, and therapeutically effective analogs and derivatives of the same.

Photodynamic therapy (PDT) of cancers employs visible light as the radiation activator of the sensitizing agent. Examples of photodynamic radiosensitizers include the following, but are not limited to: hematoporphyrin derivatives, PHOTO- FRIN@, benzoporphyrin derivatives, NPe6, tin etio- porphyrin (SnET2), pheoborbide-a, bacteriochloro- phyll-a, naphthalocyanines, phthalocyanines, zinc phthalocyanine, and therapeutically effective analogs and derivatives of the same.

Radiosensitizers can be administered in conjunction with a therapeutically effective amount of one or more compounds in addition to the Chkl inhibitor, such compounds including, but not limited to, compounds that promote the incorporation of radiosensitizers to the target cells, compounds that control the flow of therapeutics, nutrients, and/or oxygen to the target cells, chemotherapeutic agents that act on the tumor with or without additional radiation, or other therapeutically effective com- pounds for treating cancer or other disease. Exam- ples of additional therapeutic agents that can be used in conjunction with radiosensitizers include, but are not limited to, 5-fluorouracil (5-FU), leucovorin, oxygen, carbogen, red cell transfusions, perfluorocarbons (e. g., FLUOSOLW@-DA), 2,3-DPG, BW12C, calcium channel blockers, pentoxifylline, antiangiogenesis compounds, hydralazine, and L-BSO.

Chemotherapeutic agents that can be used include, but are not limited to, alkylating agents, antimetabolites, hormones and antagonists thereof, radioisotopes, antibodies, as well as natural prod- ucts, and combinations thereof. For example, an inhibitor compound of the present invention can be administered with antibiotics, such as doxorubicin and other anthracycline analogs, nitrogen mustards, such as cyclophosphamide, pyrimidine analogs such as 5-fluorouracil, cisplatin, hydroxyurea, taxol and its natural and synthetic derivatives, and the like.

As another example, in the case of mixed tumors, such as adenocarcinoma of the breast, where the tumors include gonadotropin-dependent and gonado- tropin-independent cells, the compound can be admin- istered in conjunction with leuprolide or goserelin (synthetic peptide analogs of LH-RH. Other anti- neoplastic protocols include the use of an inhibitor compound with another treatment modality, e. g., surgery or radiation, also referred to herein as "adjunct anti-neoplastic modalities."Examples of chemotherapeutic agents useful for the method of the present invention are listed in the following table.

Alkvlatinc aqents Epipodophylotoxins Hormones and antagonists Nitrogen mustards etoposide Adrenocorticosteroids/ mechlorethamine teniposide antagonists cyclophosphamide Antibiotics prednisone and equiv- ifosfamide actimomycin D alents melphalan daunomycin (rubido-dexamethasone chlorambucil mycin) ainoglutethimide Nitrosoureas doxorubicin (adria-Proqestins carmustine (BCNU) mycin) hydroxyprogesterone lomustine (CCNU) mitoxantroneidarubicin caproate semustine (methyl-CCNU) bleomycinsplicamycin medroxyprogesterone Ethvlenimine/Methvl- (mithramycin) acetate melamine mitomycinC megestrol acetate thriethylenemelamine dactinomycin Oestrogens (TEM) Enzymes diethylstilbestrol triethylene L-asparaginase ethynyl estradiol/ thiophosphoramide Biological response equivalents (thiotepa) modifiers Antiestroqen hexamethylmelamine interferon-alpha tamoxifen (HMM, altretamine) IL-2 Androqens Alkyl sulfonates G-CSF testosterone propionate busulfan GM-CSn fluoxymesterone/equiv- Triazines Differentiation Agents alents dacarbazine (DTIC) retinoic acid deriva-Antiandroqens Antimetabolites tives flutamide Folic Acid analogs Radiosensitizers gonadotropin-releasing methotrexate metronidazole hormone analogs trimetrexate misonidazole. leuprolide Pyrimidine analogs desmethylmisonidazole Nonsteroidal 5-fluorouracil pimonidazole antiandroqens fluorodeoxyuridine etanidazole flutamide gemcitabine nimorazol. e Photosensitizers cytosine arabinoside RSU 1069 hematoporphyrin (AraC, cytarabine) E09 derivatives 5-azacytidine RB 6145 Photofrins 2, 2'-difluorodeoxy-SR4233 benzoporphyrin cytidine nicotinamide derivatives Purine analogs 5-bromodeozyuridine Npe6 6-mercaptopurine 5-iododeoxyuridine tin eti. oporphyrin (SnET2) 6-thioguanine bromodeoxycytidine pheoboride-a azathioprine Miscellaneous agents bacteriochlorophyll-a 2'-deoxycoformycin Platinium coordination naphthalocyanines (pentostatin) complexes phthalocyanines erythrohydroxynonyl-cisplatin zinc phthalocyanines adenine (EHNA) carboplatin fludarabine phosphate Anthracenedione 2-chlorodeoxyadenosine mitoxantrone (cladribine, 2-CdA) Substituted urea Type I Topoisomerase hydroxyurea Inhibitors Methvlhvdrazine deriva- camptothecin tives topotecan N-methylhydrazine (MIH) irinotecan procarbazine Natural products Adrenocortical suppres- Antimitotic drugs sant paclitaxel mitotane (o, p'-DDD) Vinca alkaloids ainoglutethimide vinblastine (VLB) Cvtokines vincristine interferon (a, y) vinorelbine interleukin-2 Taxoteree (docetaxel) estramustine estramustine phosphate Examples of chemotherapeutic agents that are particularly useful in conjunction with radio-

sensitizers include, for example, adriamycin, camptothecin, carboplatin, cisplatin, daunorubicin, doxorubicin, interferon (alpha, beta, gamma), inter- leukin 2, irinotecan, docetaxel, paclitaxel, topotecan, and therapeutically effective analogs and derivatives of the same.

As appreciated by persons skilled in the art, reference herein to treatment extends to prophylaxis, as well as to treatment of established diseases or symptoms. It is further appreciated that the amount of a compound of the invention re- quired for use in treatment varies with the nature of the condition being treated, and with the age and the condition of the patient, and is ultimately determined by the attendant physician or veterinar- ian. In general, however, doses employed for adult human treatment typically are in the range of 0.001 mg/kg to about 100 mg/kg per day. The desired dose can be conveniently administered in a single dose, or as multiple doses administered at appropriate . intervals, for example as two, three, four or more subdoses per day. In practice, the physician determines the actual dosing regimen most suitable for an individual patient, and the dosage varies with the age, weight, and response of the particular patient. The above dosages are exemplary of the average case, but there can be individual instances in which higher or lower dosages are merited, and such are within the scope of the present invention.

Formulations of the present invention can be administered in a standard manner for the treat- ment of the indicated diseases, such as orally, parenterally, transmucosally (e. g., sublingually or via buccal administration), topically, transdermal- ly, rectally, via inhalation (e. g., nasal or deep

lung inhalation). Parenteral administration in- cludes, but is not limited to intravenous, intra- arterial, intraperitoneal, subcutaneous, intramuscu- lar, intrathecal, and intraarticular. Parenteral administration also can be accomplished using a high pressure technique, like POWDERJECT.

For oral administration, including buccal administration, the composition can be in the form of tablets or lozenges formulated in conventional manner. For example, tablets and capsules for oral administration can contain conventional excipients such as binding agents (tor example, syrup, acacia, gelatin, sorbitol, tragacanth, mucilage of starch, or polyvinylpyrrolidone), fillers (for example, lactose, sugar, microcrystalline cellulose, maize- starch, calcium phosphate, or sorbitol), lubricants (for example, magnesium stearate, stearic acid, talc, polyethylene glycol or silica), disintegrants (for example, potato starch or sodium starch glycolate), or wetting agents (for example, sodium lauryl sulfate). The tablets can be coated accord- ing to methods well known in the art.

Alternatively, the compounds of the pres- ent invention can be incorporated into oral liquid preparations such as aqueous or oily suspensions, solutions, emulsions, syrups, or elixirs, for exam- ple. Moreover, formulations containing these com- pounds can be presented as a dry product for con- stitution with water or other suitable vehicle before use. Such liquid preparations can contain conventional additives, for example suspending agents, such as sorbitol syrup, methyl cellulose, glucose/sugar syrup, gelatin, hydroxyethylcellulose,' hydroxypropylmethylcellulose, carboxymethylcellu- lose, aluminum stearate gel, and hydrogenated edible

fats; emulsifying agents, such as lecithin, sorbitan monooleate, or acacia; nonaqueous vehicles (which can include edible oils), such as almond oil, frac- tionated coconut oil, oily esters, propylene glycol, and ethyl alcohol; and preservatives, such as methyl or propyl p-hydroxybenzoate and sorbic acid.

Such preparations also can be formulated as suppositories, e. g., containing conventional suppository bases, such as cocoa butter or other glycerides. Compositions for inhalation typically can be provided in the form of a solution, suspen- sion, or emulsion that can be administered as a dry powder or in the form of an aerosol using a conven- tional propellant, such as dichlorodifluoromethane or trichlorofluoromethane. Typical topical and transdermal formulations comprise conventional aqueous or nonaqueous vehicles, such as eye drops, creams, ointments, lotions, and pastes, or are in the form of a medicated plaster, patch, or membrane.

Additionally, compositions of the present invention can be formulated for parenteral adminis- tration by injection or continuous infusion. Form- ulations for injection can be in the form of suspen- sions, solutions, or emulsions in oily or aqueous vehicles, and can contain formulation agents, such as suspending, stabilizing, and/or dispersing agents. Alternatively, the active ingredient can be in powder form for constitution with a suitable vehicle (e. g., sterile, pyrogen-free water) before use.

A composition in accordance with the pres- ent invention also can be formulated as a depot preparation. Such long acting formulations can be administered by implantation (for example, subcutan- eously or intramuscularly) or by intramuscular in-

jection. Accordingly, the compounds of the inven- tion can be formulated with suitable polymeric or hydrophobic materials (e. g., an emulsion in an acceptable oil), ion exchange resins, or as spar- ingly soluble derivatives (e. g., a sparingly soluble salt).

For veterinary use, a compound of formula (I), (II), or (III), or a nontoxic salt thereof, is administered as a suitably acceptable formulation in accordance with normal veterinary practice. The veterinarian can readily determine the dosing regi- men and route of administration that is most ap- propriate for a particular animal.

Thus, the invention provides a pharmaceu- tical composition comprising a compound of the formula (I), (II), or (III), together with a pharm- aceutically acceptable diluent or carrier therefor.

Also provided is a. process of preparing a pharmaceu- tical composition comprising a compound of formula (I), (II), or (III) comprising mixing a compound of formula (I), (II), or (III), together with a pharma- ceutically acceptable diluent or carrier therefor.

Specific, nonlimiting examples of com- pounds of structural formula (I), (II), and (III) are provided below, the synthesis of which were performed in accordance with the procedures set forth below.

For ease of understanding, a compound having a particular structure is identified by the corresponding compound number provided in the following tables summarizing some of the compounds useful in the method. For example, the structure identified as Compound 1 is a compound of structural formula (IV), wherein R21 is hydrogen and R28 is -C (O) NH (CH2) 2 (2-N-methylpyrrolidyl).

Compounds suitable in the method include, but are not limited to:

Compoun R27 _-R2fl d No. 1 H hr O , N-. 2 H o \NH _NH 3 H I 6 iH 4 H NU N 5 H 5 H NHN 6 H 6 H NH v _N

Compoun R27 R2fl d No. e H i> H X NH2 i NH < Cl 9 H $ NH < Nk 7 H 0 8 H 0 8 H zu Y 11 H 1-1, "Y 0 11 H 0 11 H O NH 12 CH3 13 H HN. Y"-NH2 o 14 NH2 H 15 H

Compoun R27 R2j3 d No. 16 H 16 H HN sx<NH .. I I 0 17 du H 18 H HNy' 0 18 H "OH o 19 H 50H 19 H OH 20 H 20 X H H Nu 0 0 21 Cl rot 22 H o 0 N 23 ci H 23 Cl H 24 H 0 LO O

9 Compoun R27 R28 d No. 25 H CL 0 0 2No. 26 r H 27 H 27 H nu 28 H 'NH 28 H, hr--i N o 29 H Cl 30 H un HOH OH 0 31 H L) 32 H NH2 33 H 32 H NH2 34 H 34 H o O

Compoun R27 R2a d No. 35 H 11- WNH uN oSsa N Hz 0 36 H N Ou F F 37 H 0 37 I 0 38 H H 39 H O 'NH 0 40 H 0 40 H zou --y OH 0 41 H O zou NH 42 H /0 \\0 43 H NI N

Compoun R27 R28 d No. 1 H W N 44 H nez 0 45 H HN 46 o 46 OH 0 zozo NH 0 0 47 H 0 NU 48 H xi OH 49 H 4 9,"" H HAN 0 50 H 50 H nu S 0 51 H O OANH~NH"O

Compoun R27 R28 d No. 52 H g\o/ 53 H N O o o 54 H M vv OOH 55 H han O 56 H 14N 0 s . w gNH+OH 58 OU 0 58 H 58 H nui 59 H X ON

Compoun R27 R28 d No. 60 H Ho" i- 61 H 0 5 ;., J 62 . H I 62 H 64 H 6 4 I 65 H /O- . 14 0 1

Compoun R27 R28 d No. 66 H Nu 0 67 H NU 0 68 H 68 H NH y 69 H 67 O 0xNHX 6 8 §<NH m S j % 0 X 70 H oc O S O 0 71 H nu fuzz O 72 H NH N 72 H ly 0

Compoun R27 R28 d No. 73 H r NI 0 v 0 74 H t H o y 75 H NH NH_ N '\/ O 76 H nu ou 77 H 0 77 H 78 H 78 H O NH 79 H O NH \

Compoun R 21 R28 d No. 80 H ° NH 81 H 82 H 0 NH 82 H O NH \ O N ^, \ / O NIi N 83 H Y "1 ONHN S4 , H 84 H ^~ < N O NU 85 H nu 86 NH nu H N,, 86 N H 0 nu OH 87 H NH N Compoun R 27 R28 d No. 88 nid H N b

Hetero Ring Substitutions:

D Compound No. Ras 89 N 5 90 , wNov 90 / 91 N 1 zu 1 N KA )

Compound No. R29 93 N NON 94 N 5 HN 95 0 N NU N 96 Y 7 tN N 97 N 98 99 N /' NEZ 100 100 / 100 N z Br Br t 100 Br/tXBr >

Compound No. R29 101 FIV \ V O 102 N Ber bu 103 N 104 X fY" 105 N 106 J Compound No. R29 107 zu Cof

Thiourea Compounds:

Compound No. R30 R3 108 H I / c I I nez 110 H CON) I ill H N zu Compound No. R30 R31 112 Ci 113 H N lez N 114 Ci N N

Miscellaneous Class: Compound R32 Arl Ar2 No. 115-N (H)- fi F 0 N/ 116-N (H)- OUZO w N N

Compound R32 Arl Ar2 No. 117-N (H)- N N 0 O 118-N (CH3)- N C N \ 119-N (H)- . 1V I N 120-N (H)- N w N 121-N (H)- N), A N N 122-N (H)- N r ce N 123-N (H)- Zu \

CompoundR"Arl Arl No. 124-N (H)- N N) N 125-N (H)- N N y N 0 O 126-N (H)- %/ VT I 127-N (H)- N C N I N bu 128-O- N 5 129-N (H)- Ci 130-N (H)- N i w ci

Compound R32 Arl Ar2 No. 131-N (H)- ci Cl 132 null 0 / N ci 133-N (H)- N cri '--' ci zu ce O 135-0- 0 \ O

Compound R32 Arl Ar2 No. 136-N (H)- F 2/ F I O 137-N (H)- ci Cl 138-N (H)- Fuzz F 139-N (H)- HO cri xi cri Cl / nazi I 141-O- I J

Compound R32 Arl Ar2 No. 142-N (H)- Cl C1 143-0- 144-N (H)- a ci F C1 145-N (H)- O o o 146-N (H)- cri Cl"CI

CompoundR32 Ar-Ar2 No. 147-N (H)- F ci a F C1 148-N (H)- 0 po 149-0- 0 (H)--/ ci C1 150-N (H)-- /O//O 151-N (H)- N/ C N I 152-N (H)- If Con F

Compound R32 Arl Ar2 No. 153-N (H)- %/ 'F N F 154-N (H)- N ci a ci C1 155-N (H)- N ci i N C1 156-N (H)- o/\ N N 157-N (H)- N N 158-N (H)- N N

No w R3 2 Arl Ar2 No. 159-N (H)- N 1X 160-N (H)- O (N \ N N c N 162-N (H)- N N 0 N O cri C1 164-N (H)- Nu2 N CompoundR32 Arl Ar2 No. 165-N (H)- zu oc_, N I

Some preferred compounds include Compound Nos. 2,4,6,12,72,76,83,84,88,89, and 90.

Generally, compounds of structural form- ulae (I), (II), and (III), including those of form- ulae (IV), (V), (VI), and (VII), can be prepared according to the following synthetic scheme. In the scheme described below, it is understood in the art that protecting groups can be employed where neces- sary in accordance with general principles of syn- thetic chemistry. These protecting groups are re- moved in the final steps of the synthesis under basic, acidic, or hydrogenolytic conditions which are readily apparent to those skilled in the art.

By employing appropriate manipulation and protection of any chemical functionalities, synthesis of com- pounds of structural formulae (I), (II), and (III) not specifically set forth herein can be accom- plished by methods analogous to the schemes set forth below.

Unless otherwise noted, all starting materials were obtained from commercial suppliers and used without further purification. All reac- tions and chromatography fractions were analyzed by thin-layer chromatography on 250-mm silica gel plates, visualized with W (ultraviolet) light and I2

(iodine) stain. Flash column chromatography was carried out using Biotage 40M silica gel (230-400 mesh). Products and intermediates were purified by flash chromatography or reverse-phase HPLC.

As illustrated below, the compounds of general structural formulae (I) and (II) can be prepared by the following general synthetic schemes.

General Scheme 1 In general, an aryl amine represented by the formula Ar-NH2 is reacted with about 0.75-1.25 molar equivalent of 4-nitrophenyl chloroformate.

The reaction preferably is performed under an inert atmosphere, for example, nitrogen (N2), and typically is maintained at low temperature (about 0°C). The resulting product is treated with about 0.75-1.25 molar equivalent of a heteroaryl amine represented by the formula HetAr-NH2, preferably under an inert atmosphere at room temperature (about 25°C), to afford a crude aryl pyrazine-disubstituted urea compound.

A more particular illustration for the preparation compounds of standard formulae (I) and (II) can include, for. example, the following General Scheme 2.

Step (1) : TMS Diazomethane Esterification To a cooled (about 0°C), stirred solution of 4-amino-3-methoxybenzoic acid (5.0 g; 30 mmol) in dry methanol (150 mL) was added trimethylsilyl diazomethane (60 mL of 2.0 M solution in hexanes, 120 mmol) slowly over 1 hour. After stirring for 4 hours, the reaction was concentrated at reduced pressure, dissolved in ethyl acetate (200 mL), washed with 10% aqueous sodium carbonate and brine, then dried (MgSO4), filtered, and concentrated in General Scheme 2 OCO 1 O nu H N H2N \ TMSCHN2 I O N/\ H2Nt st P ? I I step l) CO CH aN CO CH C02H 2 3 2 3 step (3) /N ; i 0 N O N 0 step (14) 0 Y"ir"ri-YV N CO2H N C02CH3 R-NH2 step (5) O/ N-NH. NH \ W N CONHR

vacuo to provide the desired ester as an off-white solid (94% yield).

Step (2): p-Nitrophenyl Carbamate Procedure To a stirred, cooled (about 0°C) solution of methyl-3-amino-4-methoxy benzoate (5.0 g; 27.6 mmol) in dry dichloromethane (175 mL) was added pyridine (2.34 mL; 29 mmol) followed by 4-nitro- phenyl chloroformate (5.8 g ; 29 mmol3 under a nitro- gen (N2) atmosphere. After stirring for 8 hours, the reaction was washed with 2N aqueous hydrochloric acid (2 X 200 mL), saturated aqueous sodium bicar- bonate (2 X 200 mL), and brine (200 mL), then dried (MgSO4), and filtered. The filtered solution was diluted with ethyl acetate and hexanes (about 800 mL) until a precipitate formed. The solid was collected on a Buchner funnel with suction, and air dried to provide the desired carbamate as a white solid (70% yield).

Step (3): Carbamate Coupling Procedure To a stirred solution of 4-methoxy-3- (4- nitro-phenoxycarbonylamino)-benzoic acid methyl ester (30 g; 8.7 mmol) in dry N-methyl pyrrolidine (50 mL) was added the amino pyrazine (0.84 g; 8.8 mmol) under a N2 atmosphere at room temperature. The reaction mixture was heated to 80°C for 6 hours, then allowed to cool to room temperature. Dilution with ethyl acetate (200 mL) and water (200 mL) pro- vided the desired urea as a white solid (54% yield).

Step (4): Lithium Hydroxide Hydrolysis Procedure To a stirred solution of 4-methoxy-4- (3- pyrazin-2-yl-ureido)-benzoic acid methyl ester (1.0 g; 3.3 mmol) in methanol (35 mL) was added aqueous lithium hydroxide (5 mL of a 2N solution; 10 mmol) at room temperature. The reaction was heated to 67°C for 15 hours, then allowed to cool to room temperature. The reaction then was diluted with water (100 mL), and washed with ethyl acetate (2 X 100 mL). The pH of the aqueous layer was adjusted to pH 5.2 with 2N aqueous hydrochloric acid, and the resulting precipitate was collected on a Buchner funnel with suction and air-dried to provide the desired acid as a white solid.

Step (5): HBTU Couplinq Procedure To a stirred solution of the acid (30 mg; 0.11 mmol) in dry N-methyl pyrrolidinone (2 mL) was added O-benzotrazol-l-yl-N, N, N', N'-tetramethyl- uronium hexafluorophosphate (HBTU; 45 mg; 0.12 mmol), 4- (2-aminioethyl)-morpholine (15.7 L; 0.12 mmol) and diisopropyl ethyl amine (34 L, 0.2 mmol) at room temperature under a nitrogen atmosphere.

The resulting solution was stirred 5 hours, then diluted with ethyl acetate (30 mL) and 10% aqueous sodium carbonate (30 mL). After stirring vigorously at room temperature for 15 minutes, the precipitate was collected on a Buchner funnel with suction and air-dried to provide the desired amide as a white solid (59% yield).

The following compounds were prepared using the general procedure described accompanying General Scheme 2, but substituting the R group below for the R group shown in General Scheme 2:

Compound No. R group Characterization 22 H NMR (400 MHz, D, DMSO) d ) 9 10. 15 (brs, 1H), 8.90 (s, 1H), 8.69 (d, 1H). 8.53 (t, 1H), 8.27 (t, 1H). 8.18 (d, 1H), 7.53 (dd, 1H), 3.96 (br s, 2H), 3.93 (s, 3H), 3.67-- 3.53 (m, 6H), 3.29 (t, 2H), 3.10 (t, 2H). LRMS (esi, positive) m/e 401.2 (M+1). 11 1H NMR (400 MHz, D6 DMSO) d 10. 14 (br s, 1H), 8.91 (s, e</1H), 8.66 (d, 1H), 8.27 (dd, $ N 1H), 8. 18 (d, 1H), 7.51 (dd, I 1H), 7.03 (d, 1H), 3.91 (s, 3H), 3.53 (br s, 1H), 3.33 (d, 2H), 3.21 (br s, 1H), 3.02 (br s, 1H), 2.78 (d, 2H), 2.48 (s, 3H), 2.31 (br s, 1H), 2.13 (br s, 1H), 1.99-1.82 (m, 2H). LRMS (esi, positive) m/e 399.2 (M+1). 25 1H NMR (400 MHz, D6 DMSO) d 10. 20 (br s, 1H), 8.87 (s, 1H), 8.31-8.26 (M, 2H), 8.21 ci (br s, 1H), 7.52-7.44 (m, 2H), 3.95 (s, 3H), 3.72 (t, 2H), 3.56 (t, 3H). 28 lit NMR (400 MHz, D6 DMSO) d 10.22 (br s, 1H), 8.90 (br s, 1H), 8.64 (t, 1H), 8.31-8.28 N (m, 2H), 8. 19 (d, 1H), 7. 51- 00 7. 47 (m, 2H), 3.97 (br s, 2H), 3. 93 (s, 3H), 3.69-3.52 (m, 6H), 3.30 (t, 2H), 3.11 (t, 2H). LRMS (esi, positive) m/e 401.1 (M+1).

Compound No. R group Characterization l 1H NMR (400 MHz, D6 DMSO) d 10. 20 (br s, 1H), 8.88 (br s, 1H), 8.40 (t, 1H), 8.32 (t, 1H), 8.23 (dd, 2H), 7.47 (d, 1H), 7.43 (dd, 1H), 3.94 (s, 3H), 2.91 (c, 2H), 2.19 (s, 3H), 2.05-1.98 (m, 2H), 1.93- 1.80 (m, 2H), 1.60 (c, 1H), 1.43 (c, 1H). LRMS (esi, positive) m/e 399.0 (M+1). 35 O lH NMR (400 MHz, D6 DMSO) d 10. 21 (br s, 1H), 8.88 (br s, 1H), 8.48 (br s, 1H), 8.32 (t, 1H), 8.26 (s, 1H), 8.23 O (d, 1H), 7. 50 (br s, 1H), 7.46 (dd, 1H), 3.95 (s, 3H), 3.36 (c, 2H), 3.10 (t, 2H), 2.83 (s, 3H). LRMS (esi, positive) m/e 409.0 (M+1). 44 lH NMR (400 MHz, D DMSO) d 11. 47 (br s, 1H), 8.48 (br s, 1H), 8.41 (s, 1H), 8.35 (d, 1H), 8.25-8.23 (M, 2H), 7.06- 7. C2 (m, 2H), 3.98 (s, 3H), 3.38 (br s, 2H), 3.04 (br s, 3H), 1.21 (br s, 3H). LRMS (esi, positive) m/e 330.0 (M+1).

General Scheme 3

Isocyanate Procedure: To a stirred solution of 2-methoxy-5-meth- yl-phenylisocyanate (43 mL; 0.3 mmol) in dry di- chloroethane (0.4 mL) was added 2-aminoquinoxaline (43.5 mg; 0.3 mmol) in a reaction vial under a nitrogen atmosphere. The vial was capped and heated to 80°C overnight (14 hours). The reaction mixture then was filtered, and the residue washed with di- chloromethane to provide the desired urea as a white solid (91% yield).

The following compounds were prepared using the procedure described accompanying General Scheme 3, but substituting the Ar group in the table below for the Ar group in General Scheme 3: Compound No. Ar group Characterization 90 1H NMR (400 MHz, D6 DMSO) d N 11. 65 (br s, 1H), 10.58 (br s, 1H), 10.58 (br s, 1H), 8. 80 (s, 1H), 8.15 (d, 1H), 7. 98 (d 1H), 7.84-7.80 (m, N 2H), 7.65 (c, 1H), 6.98 (d, 1H), 6.98 (d, 1H), 6.84 (dd, 1H), 3.99 (s, 3H), 2.25 (s, 3H). LRMS (esi, positive) m/e 309.1 (M+1). 98 lu NMR (400 MHz, D6 DMSO) d N 9. 61 (s, 1H), 8. 06 (d, 1H), 7. 35 (d, 1H), 6.88 (dd, 1H), 8. 87 (d, 1H), 5.42 (d, 1H), 3. 89 (s, 3H),'2. 33 (s, 3H). LRMS (esi, positive) m/e 259.0 (M+1). 97'H NMR (4v00 MHz, D6 DMSO) d N oS 11. 29 (s, 1H), 10.64 (s, 1H), Nz 9 S 9. 05 (d, 1H), 8.70 (d, 1H), 8. 05 (d, 1H), 6.92 (d, 1H), 6. 81 (c, 1H), 3.85 (s, 3H), 2.24 (s, 3H). LRMS (esi, positive) m/e 259.0 (M+1). Compound No. Ar group Characterization 91 1H NMR (400 MHz, D CDC13) d 11. 12 (br s, 1H), 8.61 (br s, 1H), 8.28 (s, 1H), 8 24 (s, /N 1H), 8.14 (d, 1H), 8.04-8.01 (m, 2H), 7.55-7.50 (m, 3H), 6. 86 (dd, 1H), 6.76 (d, 1H), N 3. 52 (s, 3H), 2.35 (s, 3H). LRMS (esi, positive) m/e 335.2 (M+1). 12'H NMR (300 Mhz, d6-DMSO) 6 : 10. 10 (s, 1H), 10. 00 (br s, N 1H), 8.90 (s, 1H), 8.32 (s, 1H), 8.23 (s. 1H), 8.04 (s, X »-1H), 7.93 (d, 1H), 6.81 (d, N 1H), 3.87 (s, 3H), 2.28 (s, 3H). 13C NMR (75 Mhz, d6-DMSO) 5 : 151.5,149.3,146.0,140.9, 137.2,135.2,129.2,127.7, 122.7,119.5,110.8,55.9, 20.5

EXAMPLES Example 1 Preparation of Compound 115 1- [2- (1, 1-difluoromethoxy) -phenyl]-3-pyrazin-2-yl-urea 2-(Difluoromethoxy) phenylisocyanate (1.0 g, 5.4 mmol) and aminopyrazine (0.51g, 5.4 mmol) were reacted for 6 hours in refluxing dimethoxy- ethane (20 mL). The reaction mixture was cooled to room temperature to precipitate the product, which was collected by filtration, washed with ethyl

acetate, and dried in vacuo (765 mg, 50%). 1H NMR (300 Mhz, d6-DMSO) b : 10.49 (br s, 1H), 10.26 (s, 1H), 8.83 (s, 1 H), 8.35-8.24 (m, 3H), 7.53-7.00 (m, 4H).

Example 2 Preparation of Compound 165 1- (2-methylsulfanylphenyl)-3-pyrazin-2-yl urea 2-(Methylthiophenyl)-isocyanate (1.0 g, 6.1 mmol) and aminopyrazine (0.58 g, 6.1 mmol) were reacted for 16 hours in refluxing dimethoxyethane (40 mL). The product precipitated from the cooled reaction mixture and was collected by filtration, washed with dimethoxyethane, and dried in vacuo (715 mg, 45%). 1H NMR (300 MHz, d6-DMSO) 5 : 10. 35 (br s, 1H), 10.29 (s, 1H), 8.84 (s, 1H), 8. 33 (s, 1H), 8.27 (s, 1H), 8.09 (d, 1H), 7.45 (d, 1H), 7.29, (t, 1H), 7.10 (t, 1H), 2.43 (s, 3H). 13C NMR (75 Mhz, d6- DMSO) 5 : 151.8,149.2,140.5,137. 5, 137. 3,135.2, 130.1,127.1,126.9,123.7,121.4,16.5.

Example 3 Preparation of Compound 159

1-(2-methoxy-5-nitrophenyl)-3-pyrazin-2-yl-urea A mixture of 2-methoxy-5-nitrophenyl iso- cyanate (5.0 g, 25 mmol) and aminopyrazine (2.5 g, 26 mmol) in tetrahydrofuran (THF, 250 mL) was stirred at reflux for 24 hours. The product was precipitated from the cooled reaction mixture and was collected by filtration, washed with ethyl acetate, and dried in vacuo (4.3 g, 57%). 1H NMR (300 MHz, d6-DMSO) (mixture of rotamers) & : 10.38 (br, s, 1H), 10.27 (s, 1H), 9.39,8.88 (2 singlets, 1H), 9.10 (d, 1H), 8.33 (s, 1H), 8.26 (d, 1H), 7.98- 8.25 (m, 1H), 7.97-7.84 (m, 1H), 4.05,4.03 (2 singlets, 77: 28 ratio, 3H).

Example 4 Preparation of Compound 14

1- (5-amino-2-methoxyphenyl)-3-pyrazin-2-yl-urea A solution of (2-methoxy-5-nitrophenyl)-3- pyrazin-2-yl-urea (Compound 159, Example 3) (16.9 g, 55 mmol) in dimethylformamide (DMF, 320 mL) was shaken under H2 in the presence of palladium on carbon (Pd/C) catalyst (1.6 g, 30% Pd) at 80°C for 12 h. A second portion of catalyst was added (1.6 g) and shaking was continued for an additional 8 h at the same temperature. The solution was filtered through a pad of celite using an additional 200 mL of DMF. The filtrate was concentrated in vacuo and the residue was triturated with methanol (100 mL).

The solid was collected, stirred in boiling meth- anol, and solids present (1.8 g) were filtered off and discarded. The filtrate was cooled at 4°C overnight. Solids (1.4 g) were removed by filtra- tion and the filtrate was concentrated in vacuo to a tan solid (2.6 g). The crude solid product was triturated with THF (200 mL), collected by filtra- tion, and dried in vacuo to afford the product as a tan solid (1.85 g, 13%). IH NMR (300 Mhz, d6 DMSO) 5 : 10.10 (s, 1H), 9.94 (br s, 1H), 8.89 (s, 1H), 8.32 (s, 1H), 8.22 (s, 1H), 7.58 (s, 1H), 7.75 (d, 1H), 6.21 (d, 1 H), 4.70 (s, 2H), 3.76 (s, 3H). 13C

NMR (75 Mhz, d6-DMSO) 5 : 151.4,149.4,142.6, 140.9,139.8,137.2,135.2,128.7,112.8,107.7, 106.0,56.8.

Example 5 Preparation of Compound 48 N- [4-methoxy-3- (3-pyrazin-2-yl-ureido) phenyl]-succinamic acid A solution of 1-(5-amino-2-methoxyphez 3-pyrazin-2-yl-urea (Compound 14, Example 4) (260 mg, 1 mmol) and succinic anhydride (131 mg, 1. 3 mmol) in dry pyridine (10 mL) was stirred 16 h at room temperature. The resulting solid was collected by filtration and triturated with chloroform, and dried in vacuo to afford the off-white product (175 mg, 50%). 1H NMR (300 Mhz, d6-DMSO) b : 10.15 (s, 1H), 1C. 05 (s, 1H), 9.87 (s, 1H), 8.90 (s, 1H), 8.33 (s, 2H), 8.24 (s, 1H), 7.42 (dd, J = 8.8,2.2 Hz, 1H), 6.96 (d, J = 8.8 Hz, 1H), 3.87 (s, 3H), 2.54 (br s, 4H).

Example 6 Preparation of Compound 36

(S)-l- (2, 2,2-trifluoroethanoyl) pyrrolidine-2- carboxylic acid [4-methoxy-3- (3-pyrazin-2-yl- ureido) pheriyl]-amide A solution of 1- (5-amino-methoxyphenyl)-3- pyrazin-2-yl-urea (Compound 14, Example 4) (105 mg, 0.4 mmol) in dry pyridine (2 mL) at 0°C was treated with a solution of N-trifluoroacetyl- (S)-prolyl , chloride (0.1 M in dichloromethane, 4.5 mL, 0.45 mmol) and stirred 2 h at room temperature. The reaction was quenched with 1 N HCl (50 mL) and extracted with ethyl acetate (3 X 50 mL). The combined organic layers were washed with 1 N HCl (2 X 20 mL), water (20 mL), brine (20 mL), dried over sodium sulfate, and concentrated in vacuo to a beige solid (60 mg). Recrystallization from acetonitrile yielded the final solid product (30 mg, 17t H NMR (300 Mhz, d6-DMSO) 5 : (10.16-10.06, m, 3H), 8.90 (s, 1H), 8.36-8.30 (m, 2H), 8.25 (d, J = 2.6 Hz, 1H), 7.42 (dd, J = 8.8,2.6 Hz, 1H), 6.98 (d, J = 8.8 Hz, 1H), 4.57 (dd, J = 8.5,4.4 Hz, 1 H), 3.88 (s, 3H), 3.73 (t, J = 6.5 Hz, 1H), 2.27-2.21 (m,

1H), 2.06-1.90 (m, 3H). LRMS (ESI, positive) m/e 453.1 (M + 1).

Example 7 Preparation of Compound 16 (S)-pyrrolidine-2-carboxylic acid [4-methoxy-3- (3- pyrazin-2-yl-ureido)-phenyl]-amide A suspension of (S)-1-(2,2,2-trifluoro- ethanoyl)-pyrrolidine-2-carboxylic acid [4-methoxy- 3- (3-pyrazin-2-yl-ureido)-phenyl]-amide (Compound 36, Example 6) (22 mg, 0.05 mmol) in a mixture of methanol (MeOH, 5 mL) and water (about 0.25 mL) was treated with KOH (100 mg, large excess). Within 10 minutes, all ingredients were in solution. The reaction mixture was treated with water (20 mL) and extracted with ethyl acetate (2 x 20 mL). The organic layers were combined and washed with water (10 mL) and brine (10 mL), dried with sodium sul- fate, and concentrated to a tan solid (13 mg, 75%).

'H NMR (300 MHz, d6-DMSO) b : 10.13 (s, 1H), 10.03 (s, 1H), 9.85 (s, 1H), 8.90 (s, 1H), 8.38-8.28 (m, 2H), 8.25 (d, J = 2.6 Hz, 1H), 7.41 (dd, J = 8.8, 2.6 Hz), 6.98 (d, J = 8.8 Hz, 1H), 3.88 (s, 3H, 3.69 (dd, J = 8.7,5.6 Hz, 1H), 2.94-2.87 (m, 2H), 2.10- 1.97 (m, 1H), 1.81-1.60 (m, 3H). LRMS (ESI, posi- tive) m/e 357.1 (M + 1).

Example 8 Preparation of Compound 42

N- [4-methoxy-3- (3-pyrazin-2-yl-ureido) phenyl]- methanesulfonamide A solution of 1- (5-amino-2-methoxyphenyl)- 3-pyrazin-2-yl-urea (Compound 14, Example 4) (260 mg, 1 mmol) in dry pyridine (15 mL) was treated with methanesulfonyl chloride (0.08 mL, 1 mmol) and stirred 16 h at room temperature. The reaction mixture was concentrated in vacuo and the solid residue was triturated with ethanol, collected by filtration, and dried in vacuo to afford the product (205 mg, 61%). 1H NMR (300 Mhz, d6-DMSO) 5 : 10.16 (s, 1H), 10.07 (s, 1H), 9.40 (s, 1H), 8.92 (s, 1H), 8.35 (s, 1 H), 8.27 (s, 1H), 8.19 (d, J = 2. 2 Hz, 1H), 7.04 (d, J = 8.8 Hz, 1H), 6.91 (dd, J = 8.7, 2.4 Hz, 1H), 3.91 (s, 3H), 2.91 (s, 3H).

Example 9 Preparation of Compound 65

1-(2-methoxy-4-nitrophenyl)-3-pyrazin-2-yl-urea A mixture of 2-methoxy-4-nitrophenyl iso- cyanate (15.0 g, 77 mmol) and aminopyrazine (7.35 g, 77 mmol) in THF (600 mL) was stirred at reflux for 24 hours. The product precipitated from the cooled reaction mixture and was collected by filtration, washed with ethyl acetate, triturated with hot ethanol, and dried in vacuo (16.3 g, 73%). 1H NMR (300 Mhz, d6DMSO) 5 : 10.50 (br s, 1H), 10.42 (s, 1H), 8. 94 (s, 1H), 8.48 (d, 1H), 8.39 (s, 1H), 8.32 (d, 1H), 7.95 (dd, J = 9.1,2.4 Hz, 1H), 7. 84 (d, J = 2.4 Hz, 1 H), 4.08 (s, 3H).

Example 10 Preparation of Compound 32

1- (4-amino-2-methoxyphenyl)-3-pyrazin-2-yl-urea A solution of (2-methoxy-4-nitrophenyl)-3- pyrazin-2-yl-urea (Compound 65, Example 9) (7.9 g, 27 mmol) in DMF (300 mL) was shaken under H2 in the presence of Pd/C catalyst (1.6 g, 10% Pd) at 110°C for 4 h. The mixture was filtered through a pad of celite using an additional 200 mL of DMF. The filtrate was concentrated in vacuo and the residue was recrystallized from ethanol (with a hot filtra- tion step) to yield the light gray product (2.9 g, 41%). 1H NMR (300 MHz, d6-DMSO) 5 : 9.83 (s, 1H), 9.50 (s, 1H), 8.86 (s, 1H), 8.28 (s, 1H), 8.19 (d, J = 2.5 Hz, 1H), 7.64 (d, J = 8. 5 Hz, 1H), 6.31 (d, J = 2.0 Hz, 1H), 6.13 (dd, J = 8. 5,2.0 Hz, 1H), 4.92 (s, 2H), 3.79 (s, 3H). 13C NMR (75 Mhz, d5-DMSO) o : 151.6,150.0,149.6,145.2,140.9,136.9,135.1, 121.6,116.8,105.5,98.0,55.4.

Example 11 Preparation of Compound 3

C-dimethylamino-N- [3-methoxy-4- (3-pyrazin- 2-yl-ureido) phenyl]-acetamide A solution of N, N-dimethylglycine (124 mg, 1.2 mmol) and triethylamine (0. 33 mL, 2.4 mmol) in dry acetonitrile (5 mL) at 0°C was treated dropwise with isobutyl chloroformate (0.16 mL, 1.2 mmol) and stirred 15 min. This mixture was treated dropwise with a solution of 1- (4-amino-2-methoxyphenyl)-3- pyrazin-2-yl-urea (Compound 32, Example 10) (100 mg, 0.4 mmol) in dimethyl sulfoxide (DMSO, 1 mL). The reaction mixture was stirred at room temperature for 3 h, quenched with water (20 mL), and extracted with ethyl acetate (2 x 15 mL). The combined organic layers were washed with water (10 mL) and brine (10 mL), dried over sodium sulfate, and concentrated in vacuo. The residue was dissolved in DMSO (1 mL) and purified by HPLC (YMC 20 x 50 mm C18 CombiPrep column, 20 mL/min, 2-50% CH3CN/water in 6 min, all solvents contained 0.05% trifluoroacetic acid (TFA), 0. 35 mL injections, detector at 254 nm, detector path length 0.2 mm). Fractions containing the product were concentrated in vacuo to afford the product as the trifluoroacetate (TFA) salt (24 mg, 17%).

Example 12 Preparation of Compound 8

3-chloro-N- [3-methoxy-4- (3-pyrazin-2-yl- ureido) phenyl]-propionamide A solution of 1- (4-amino-2-methoxyphenyl)- 3-pyrazin-2-yl-urea (Compound 32, Example 10) (259 mg, 1 mmol) in pyridine (3 mL) at 0°C was treated with chloroacetyl chloride (0. 29 mL, 3 mmol). The suspension was warmed at 80°C until most solids dissolved, the reaction. mixture was cooled to room temperature and the product was precipitated with ether (10 mL). This crude product was used without purification for further reactions, but a portion (30 mg) was purified by HPLC (Luna 10 x 250 mm C18 column, 4.7 mL/min, 2-80% CH3CN/water in 15 min, all solvents contained 0.05% TFA, 0.25 mL injections, detector at 254 nm, detector path length 0.3 mm).

Fractions containing the product were concentrated in vacuo to afford the product. 1H NMR (300 Mhz, d6- DMSO) 5 : 10.00 (s, 2H), 9.94 (s, 1H), 8.87 (s, 1H), 8.32 (dd, J = 2.5,1.5 Hz, 1H), 8.23 (d, J = 2. 6 Hz, 1H), 8.05 (d, J = 8. 7 Hz, 1H), 7.49 (d, J = 2. 1 Hz, 1H), 7.07 (dd, J = 8. 7,2.1 Hz, 1H), 3.90-3.80 (m, 5H), 2.80 (t, J= 6.2 Hz, 2H). LRMS (ESI, positive)- m/e 350,352 (M + 1).

Example 13 Preparation of Compound 4

(cyclohexyl-methyl-amino)-N- [3-methoxy-4-<BR> <BR> (3-pyrazin-2-yl-ureido) phenyl]-propionamide A mixture of 3-chloro-N- [3-methoxy-4- (3- pyrazin-2-yl-ureido) phenyl]-propionamide (Compound 8, Example 12) and N-cyclohexyl-methylamine (0.5 mL, large excess) was warmed at 80°C for 1 h and cooled to room temperature. Crude product was precipitated from ether (10 mL), collected by filtration, and dissolved in DMSO (0. 5 mL). Aliquots (about 0.25 mL) were purified by HPLC (Luna 10 x 250 mm C18 column, 4.7 mL/min, 2-80% CH3CN/water in 15 min, all solvents contained 0.05% trifluoroacetic acid, detector at 254 nm, detector path length 0.3 mm).

Fractions containing the product were concentrated in vacuo to afford the product as the TFA salt (4.7 mg, 11%). 1H NMR (300 Mhz, d6-DMSO) 5 : 10.18 (s, 1H), 10.07 (s, 1H), 9.98 (s, 1H), 9.04 (br s, 1H), 8.88 (s, 1H), 8.33 (dd, J = 2.6,1.5 Hz, 1H), 8.24 (d, J = 2.6 Hz, 1H), 8.08 (d, J = 8.7 Hz, 1H), 7.43 (d, J = 2.1 Hz, 1H), 7.09 (dd, J = 8.7 ; 2.1 Hz, 1H), 3.90-3.83 (m, 1H), 3.30-3.16 (m, 2H), 2.81 (t, J = 6.7 Hz, 1H), 2.74 (d, J= 5.0 Hz, 2H), 2.03-1.89 (m, 2H), 1.89-1.75 (m, 2H), 1.70-1.53 (m, 1H), 1.46- 1.10 (m, 5H). LRMS (ESU, positive) m/e 427.2 (M + 1).

Example 14 Preparation of Compound 2

3-cyclopentylamino-N- [3-methoxy-4- (3-pyrazi. n - 2-yl-ureido) phenyl]-propionamide A mixture of 3-chloro-N- [3-methoxy-4- (3- pyrazin-2-yl-ureido) phenyl]-propionamide (Compound 8, Example 12) and cyclopentylamine (0.5 mL, large excess) was warmed at 80°C for 1 h and cooled to room temperature. The product was precipitated from ether (10 mL), collected by filtration, washed with ether, and dried in vacuo (24 mg, 60%). 1H NMR (300 Mhz, d6-DMSO) 5 : 10. 14 (s, 1H), 10.03 (s, 1H), 9.93 (s, 1H), 8.87 (d, J = l. lHz, 1H), 8.31 (dd, J = 2.6, 1.5 Hz. 1H), 8.23 (d, J = 2. 7 Hz, 1H), 8.03 (d, J = 8.7 Hz, 1H), 7.47 (d, J = 2. 1 Hz, 1H), 7.04 (dd, J = 8.7,2.1 Hz, 1H), 3.87 (s, 3H), 3.05 (quintet, J = 6.3 Hz, 1H), 2.80 (t, J = 6. 6 Hz, 2H), 2.44 (t, J = 6.6 Hz, 2H), 1.80-1.67 (m, 2H), 1.65-1.56 (m, 2H), 1.53-142 (m, 2H), 1.37-1.27 (m, 2H). LRMS (ESI, positive) m/e 399.1 (M + 1).

Compound 166: 3-Methoxy-4- (3-pyrazin-2-yl-ureido)-benzoic acid Step 1: Methyl-3-amino-4-methoxy benzoate. To a cooled (about 0 °C), stirred solution of 4-amino-3- methoxybenzoic acid (5.0 g, 30 mmol) in dry methanol (150 mL) was added trimethylsilyldiazomethane (60 mL of 2M solution in hexanes, 120 mmol) slowly over 1 hour. After stirring for 4 hours, the reaction was concentrated at reduced pressure, dissolved in ethyl acetate (200 mL), washed with 10% aqueous sodium carbonate and brine, then dried (MgS04), filtered, and concentrated in vacuo to provide the desired ester as an off-white solid (94% yield).

Step 2: 4-Methoxy-3- (4-nitro-phenoxycarbonylamino)-benzoic acid methyl ester. To a stirred, cooled (about 0 °C) solution of methyl-3-amino-4-methoxy benzoate (5.0 g, 27.6 mmol) in dry dichloromethane (90 mL) was added pyridine (2.34 mL, 29 mmol) followed by 4-nitophenyl chloroformate (5.8 g, 29 mmol) under a nitrogen atmosphere. After stirring for 1 hour, the reaction was diluted to 200 mL with dichloromethane and washed with 2N aqueous hydrochloric acid (2 x 200 mL), saturated aqueous sodium bicarbonate (2 x 200 mL), and brine (200 mL), then dried (MgS04), and filtered. The filtered solution was concentrated to a white solid corresponding to the desired carbamate (98% yield).

Step 3: 3-Methoxy-4- (3-pyrazin-2-yl-ureido)-benzoic acid methyl ester. To a stirred solution of 4- methoxy-3- (4-nitro-phenoxycarbonylamino)-benzoic acid methyl ester (10.64 g, 30.7 mmol) in dry N- methyl pyrrolidinone (31 mL) at room temperature under nitrogen was added arninopyrazine (2.92 g, 30.7 mmol) and the reaction was warmed to 85°C. After 6 hours, the reaction was cooled to room temperature and triturated with ethyl acetate (200 mL). The precipitate formed was filtered off, rinsed with ethyl acetate and dried to give the urea as a tan solid (66 % yield).

Step 4: 3-Methoxy-4- (3-pyrazin-2-yl-ureido)-benzoic acid. To a stirred suspension of 3-methoxy-4- (3-pyrazin-2-yl-ureido)-benzoic acid methyl ester (6.07 g, 20 mmol) in 200 mL 3: 1 MeOH: H20 at room temperature under nitrogen was added lithium hydroxide monohydrate (8.4 g, 200 mmol) and the reaction heated to 65'C overnight. The reaction was then cooled to room temperature and most of the methanol removed by rotary evaporation. The remaining suspension was neutralized to pH about 4 with concentrated HC1. The formed precipitate was isolated by filtration and rinsing with H2O and then drying under high vacuum to give the desired acid as a white solid (5.34 g, 93%).

'H-NMR (400 MHz, d6-DMSO) 8 8.89 (br s, 1H), 8.34 (s, 1H), 8.24 (s, 1H), 8.16 (d, 1H), 7.56 (s, 1H), 7.50 (d, 1H), 3.91 (s, 3H) Compound 167: N-Butyl-3-methoxy-4- (3-pyrazin-2-yl-ureido)-benzamide To a stirred suspension of Compound lxx (32 mg, 0.11 mmol) in 1 mL of NMP at room temperature in a capped reaction vial was added HBTU (0.4 M in NMP, 300 liL, 0.12 mmol) and the suspension stirred for 15 minutes. N-Butyl amine (0.4 M in NMP, 300 uL, 0.12 mmol) was then added followed by DIEA (38 pL, 0.22 mmol). The reaction was stirred at room temperature overnight and was then diluted with EtOAc (20 mi) and 10% Na, C03 (20 mL) and stirred rapidly for 5 minutes. A precipitate formed which was isolated by filtration and rinsing with HO and EtOAc. After air drying, the amide was isolated as an off-white solid (12.2 mg, 32 %).

'H-NMR (400 MHz, d6-DMSO) 8 8.92 (br s, 1H), 8. 37 (br s, 2H), 8.23 (d, 1H), 8.21 (s, 1H), 7.53 (s, 1H), 7.47 (d, 1H), 3.97 (s. 3H), 3.23 (q, 2H), 1.52 (m, 2H), 1.35 (m, 2H), 0.92 (t, 3H) LRMS (apci, positive) m/e 344.1 (M+1) Compound 168: N-Benzyl-3-methoxy-4-(3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using benzyl amine (39% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.96 (t, 1H), S. 91 (s, 1H), 8.56 (s, 1H), 8.50 (s, 1H), 8.44 (m, 1H), 7.58 (s, 1H), 7.56 (d, 1H), 7. 35 (m, 4H), 7.23 (m, 1H), 4.46 (d, 2H), 3.97, (s, 3H) LRMS (apci, positive) m/e 378.1 (M+1) Compound169: 3-Methoxy-N-phenethyl-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using phenethyl amine (49% yield).

H-NMR (400 MHz, d6-DMSO) 8 8.92 (br s, 1H), 8.51 (t, 1H), 8. 36 (br s, 1H), 8. 25 (s, 1H), 8.22 (d, 1H), 7.52 (s, 1H), 7.45 (d, 1H), 7.36-7.20 (m, 5H), 3.98 (s, 3H), 3.46 (m, 2H), 2.84 (dd, 2H) LRMS (apci, positive) m/e 392.1 (M+l) Compound 170: 3-Methoxy-N- (3-phenyl-propyl)-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using phenpropyl amine (71% yield).

H-NMR (400 MHz, d6-DMSO) 8 8.92 (br s, 1H), 8.40 (t, 1H), 8. 36 (br s, 1H), 8.25 (d, 1H), 8.23 (s, 1H), 7.52 (s, 1H), 7.46 (d, 1H), 7. 32-7. 18 (m, 5H), 3.97 (s, 3H), 3.25 (m, 2H), 2.61 (m, 2H), 1.82 (m, 2H) LRMS (apci, positive) m/e 406.1 (M+1) Compound 171: N- (2-Benzenesulfonyl-ethyl)-3-methoxy-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using 2-benzenesulfonyl-ethylamine (57% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.90 (br s, 1H), 8.43 (br m, 1H), 8. 35 (br s, 1H), 8.25 (s, 1H), 8.22 (d, 1H), 7.96 (s, 1H), 7.93 (s, 1H), 7.72 (m, 1H), 7.63 (m, 2H), 7. 38 (s, 1H), 7. 33 (d, 1H), 3.95 (s, 3H), 3.59 (m, 2H), 3.55 (m, 2H) LRMS (apci, positive) m/e 456.0 (M+1) Compound 172 : N- (4-Iodo-benzyl)-3-methoxy-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using 4-iodo benzyl amine (66% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.97 (t, 1H), 8.90 (s, 1H), 8.36 (s, 1H), 8.26 (d, 1H), 8.24 (s, 1H), 7.69 (d, 2H), 7.56 (m, 2H), 7.16 (d, 2H), 4.41 (d, 2H), 3.97 (s, 3H) LRMS (apci, positive) m/e 504.0 (M+1) Compound 173 : 3-Methoxy-4- (3-pyrazin-2-yl-ureido)-N- (2-pyridin-2-yl-ethyl)-benzamide Prepared according to the procedure of Compound 167 using 2-pyridin-2-yl-ethylamine (57% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.90 (br s, 1H), 8.51 (br m, 2H), 8. 36 (s, 1H), 8.22 (m, 2H), 7.71 (t, 1H), 7.48 (s, 1H), 7.43 (d, 1H), 7.26 (d, 1H), 7.21 (m, 1H), 3.97 (s, 3H), 3.60 (m, 2H), 3.00 (dd, 2H) LRMS (esi, positive) m/e 393.3 (M+1) Compound 174 : 3-Methoxy-4- (3-pyrazin-2-yl-ureido)-N- (2-pyridin-4-yl-ethyl)-benzamide Prepared according to the procedure of Compound 167 using 2-pyridin-4-yl-ethylamine (45% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.93 (s, 1H), 8.63 (d, 2H), 8.51 (t,] H), 8. 37 (s, 1H), 8.27 (s, 1H), 8.24 (d, 1H), 7.60 (d, 2H), 7.43 (m, 2H), 3.97 (s, 3H), 3.58 (m, 2H), 3.01 (m, 2H) LRMS (esi, positive) m/e 393.1 (M+1) Compound 175 : N- (lH-Benzoimidazol-2-ylmethyl)-3-methoxy-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using C- (lH-benzoimidazol-2-yl)-methylamine (53% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8. 90 (s, 1H), 8.35 (s, 1H), 8. 32 (d, 1H), 8.22 (s, 1H), 7.61 (m, 3H), 7.47 (m, 2H), 7.12 (m, 2H), 4.66 (s, 2H), 3.98 (s, 3H) LRMS (esi, positive) m/e 418.2 (M+1) Compound 176: N- [2- (lH-Indol-3-yl)-ethyl]-3-methoxy-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using tryptamine (74% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.92 (s, 1H), 8.55 (br t, 1H), 8.26 (s, 1H), 8.24 (d, 1H), 8.23 (s, 1H), 7.60 (d, 1H), 7.53 (s, 1H), 7.50 (d, 1H), 7.26 (d, 1H), 7.19 (s, 1H), 7.05 (dd, 1H), 6.98 (dd, 1H), 3.97 (s, 3H), 3.56 (m, 2H), 2.96 (m, 2H) LRMS (esi, positive) m/e 431.2 (M+1) Compound 177: 3-Methoxy-N- [3- (methyl-phenyl-amino)-propyl]-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using N1-methyl-Nl-phenyl-propane-1, 3- diamine (68% yield). 'H-NMR (400 MHz, DMSO) 8 8. 89 (s, 1H), 8.41 (br s, 1H), 8. 36 (s, 1H), 8.23 (d, 1H), 8.22 (s, 1H), 7.52 (s, 1H), 7.49 (d, 1H), 7.16 (m, 2H), 6.70 (d, 2H), 6.59 (dd, 1H), 3.96 (s, 3H), 3.38 (m, 2H), 3.30 (m, 2H), 2.87 (s, 3H), 1.77 (m, 2H) LRMS (esi, positive) m/e 435.2 (M+1) Compound 178: N- (I-Benzyl-pyrrolidin-3-yl)-3-methoxy-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using 3-amino-l-benzyl pyrrolidine (62% yield).

H-NMR (400 MHz, d6-DMSO) 8 8. 88 (br s, 1H), 8.36 (br m, 2H), 8.24 (d, 1H), 8.21 (s, 1H), 7.51 (s, 1H), 7.50 (d, 1H), 7.32 (m, 4H), 7.22 (m, 1H), 4.39 (br m, 1H), 3.96 (s, 3H), 3.59 (s, 2H), 2.79 (m, 1H), 2.62 (m, 1H), 2.40 (m, 1H), 2.16 (m, 1H), 1.81 (m, 2H) LRMS (esi, positive) m/e 447.2 (M+1) Compound 179 : N- (3- (R)-1-Benzyl-pyrrolidin-3-yl)-3-methoxy-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using 3-(R)-amino-l-benzyl pyrrolidine (57% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.83 (br s, 1H), 8.36-8. 24 (m, 3H), 8.15 (m, 1H), 7.48 (m, 2H), 7.32 (m, 4H), 7.22 (m, 1H), 4. 37 (m, 1H), 3.96 (s, 3H), 3.59 (s, 2H), 2.78 (m, 1H), 2.63 (m, 1H). 2.41 (m, 1H), 2.17 (m, 1H), 1.80 (m, 2H) LRMS (esi, positive) m/e 447.1 (M+1) Compound 180: N-(3-(S)-1-Benzyl-pyrrolidin-3-yl)-3-methoxy-4-(3-pyrazin-2- yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using 3-(S)-amino-l-benzyl pyrrolidine (57% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.85 (s, 1H), 8. 34 (br s, 1H), 8. 30 (s, 1H), 8.25 (d, 1H), 8.19 (s, 1H), 7.50 (m, 2H), 7.32 (m, 4H), 7.22 (m, 1H), 4. 39 (m, 1H), 3.97 (s, 3H), 3.59 (s, 2H), 2.79 (m, 1H), 2.62 (m, 1H), 2.41 (m, 1H), 2.16 (m, 1H), 1.81 (m, 2H) LRMS (esi, positive) m/e 447.1 (M+1) Compound 181: N- (2-Dimethylamino-ethyl)-3-metlioxy-N-methyl-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using N, N, N'-trimethyl-ethane-1, 2-diamine (57% yield).

'H-NMR (400 MHz, D2O) 8 8.17 (s, 1H), 8.05 (s, 1H), 7.94 (s, 1H), 7.76 (d, 1H), 6.92 (m, 2H), 3.79 (m, 2H), 3.75 (s, 3H), 3.36 (m, 2H), 2.97 (s, 3H), 2.88 (s, 6H) LRMS (esi, positive) m/e 373.2 (M+1) Compound 182 : 3-Methoxy-N- (3-methylamino-propyl)-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using Nl-methyl-propane-1, 3-diamine (25% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.93 (s, 1H), 8. 38-8. 25 (m, 4H), 7.59 (d, 1H), 7.52 (m, 1H), 3.98 (s, 3H), 3.92 (m, 2H), 2.92, (m, 2H), 2.50 (s, 3H), 1.82 (m, 2H) LRMS (esi, positive) m/e 359.1 (M+1) Compound 183: N- (3-Dimethylamino-propyl)-3-methoxy-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using N, N-dimethyl propyldiamine (81 % yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.93 (s, 1H), 8.56 (t, 1H), 8.37 (s, 1H), 8.25 (d, 1H), 8.23 (s, 1H), 7.52 (s, 1H), 7.50 (d, 1H), 4.10 (m, 2H), 3.97 (s, 3H), 3.35 (s, 6H), 3.05 (m, 2H), 1.84 (m, 2H) LRMS (esi, positive) m/e 373.1 (M+1) Compound 184 : N- (3-Dimethylamino-propyl)-3-methoxy-N-methyl-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using N, N, N'-trimethyl propyldiamine (88% yield).

'H-NMR (400 MHz, CDC13/CD30D) 8 8. 60 (s, 1H), 8. 33 (d, 1H), 8.23 (s, 1H), 8.19 (s, 1H), 7.01 (m, 2H), 3.99 (s, 3H), 3.83 (s, 3H), 3.59 (m, 2H), 2.78 (m, 2H), 2.59 (s, 6H), 2.22 (m, 2H) LRMS (esi, positive) m/e 387.1 (M+1) Compound 185: 3-Methoxy-N- (3-morpholin-4-yl-propyl)-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using 3-morpholin-4-yl-propylamine (53% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.92 (s, 1H), 8.57 (t, 1H), 8. 36 (s, 1H), 8.28 (d, 1H), 8.24 (s, 1H), 7.52 (s, 1H), 7.50 (d, 1H), 3.96 (s, 3H), 3.61 (m, 2H), 3.42 (m, 2H), 3.32 (m, 4H), 3.10 (m, 4H), 1.90 (m, 2H) LRMS (esi, positive) m/e 415.1 (M+1) Compound 186: 3-Methoxy-N- [3- (4-methyl-piperazin-1-yl)-propyl]-4- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 167 using 3- (4-methyl-piperazin-1-yl)-propylamine (63% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.92 (s, 1H), 8.41 (m, 1H), 8.36 (s, 1H), 8.24 (m, 2H), 7.52 (s, 1H), 7.49 (d, 1H), 3.97 (s, 3H), 3.31 (m, 11H), 2.70 (m, 2H), 2.41 (m, 2H), 1.72 (m, 2H) LRMS (esi, positive) m/e 428.1 (M+i) Compound 187: {2- [3-Methoxy-4- (3-pyrazin-2-yl-ureido)-benzoylamino]-ethyl}-trimethyl-am. nonium chloride Prepared according to the procedure of Compound 167 using 2-N, N, N-trimethylammonium ethylamin (46% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.92 (s, 1H), 8.77 (m, 1H), 8.36 (s, 1H), 8. 30 (d, 1H), 8. 24 (s, 1H), . 7. 53 (s, 1H), 7.51 (d, 1H), 3.97 (s, 3H), 3.70 (m, 2H), 3.50 (m, 2H), 3.15 (s, 9H) LRMS (esi, positive) m/e 373.1 (M+) Compound 188 : 4-Methoxy-3- (3-pyrazin-2-yl-ureido)-benzoic acid Step 1: 4-Methoxy-3- (4-nitro-phenoxycarbonylamino)-benzoic acid methyl ester. To a stirred, cooled (0°C) solution of methyl-3-amino-4-methoxy benzoate (5.0 g; 27.6 mmol) in methylene chloride (100 mL) was added pyridine (2. 34 mL; 29 mmol) followed by 4 nitrophenyl chloroformate (5.8 g; 29 mmol). After stirring for 8 hours, the reaction was diluted with methylene chloride (100 mL), washed with IN hydrochloric acid (2 x 125 mL), 10% aqueous sodium carbonate (2 x 125 mL), brine (1 x 125 mL), then dried (MgS04), and filtered. The filtered material was concentrated under reduced pressure.

The residue was taken up in ethyl acetate (100 mL) followed by hexanes (700 mL). A precipitate formed which was filtered to yield an off white solid (80% yield).

Step 2: 4-Methoxy-3- (3-pyrazin-2-yl-ureido)-benzoic acid methyl ester. To a stirred solution of the carbamate piece (1.0 g; 2.9 mmol) in N-methyl pyrrolidinone (5 mL) was added amino pyrazine (285 mg; 3.0 mmol). The reaction was heated to 85 OC and stirred for 12 hours. The reaction was allowed to cool to room temperature, then diluted with ethyl acetate (50 mL) and water (50 mL). A precipitate formed which was filtered and dried under reduced pressure to yield an off white solid (55% yield).

Step 3: 4-Methoxy-3- (3-pyrazin-2-yl-ureido)-benzoic acid. To a stirred solution of 4-methoxy-3- (3- pyrazin-2-yl-ureido)-benzoic acid methyl ester (1.0 g; 3.3 mmol) in methanol (25 mL) was added lithium hydroxide (5 mL of a 2M aqueous solution). The reaction was heated to 60 °C and stirred for 12 hours. The reaction was allowed to cool to room temperature and the pH was adjusted to 5.5 with hydrochloric acid (1N). A precipitate formed which was filtered and dried under reduced pressure to yield an off white solid (58% yield).

Compound 189: N-Butyl-4-methoxy-3- (3-pyrazin-2-yl-ureido)-benzamide To a stirred solution of 4-Methoxy-3- (3-pyrazin-2-yl-ureido)-benzoic acid (32 mg; 0.11 mmol) in N- methyl pyrrolidinone (1 mL) was added O-benzotrazol-1-yl-N, N, N', N'-tetramethyl-uronium hexafluorophosphate (HBTU ; 45 mg; 0.12 mmol), butyl amine (12 uL, 0. 12 mmol), and diisopropylethylamine (35 gL. ; 0.20 mmol). The reaction was stirred at room temperature for 12 hours.

The reaction was diluted with ethyl acetate (20 mL) and 10% aqueous sodium carbonate (20 mL) and stirred for 5 minutes. A precipitate formed which was filtered and dried under reduced pressure to yield an off white solid (49% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.90 (s, 1H), 8.63 (s, 1H), 8. 35 (s, 1H), 8.23 (m, 1H), 8.21 (s, 1H), 7.52 (d, 1H), 7.06 (d, 1H), 3.95 (s, 3H), 3.22 (m, 2H), 1.50 (m, 2H), 1.33 (m, 2H), 0.90 (t, 3H) LRMS (apci, positive) m/e 344.1 (M+1) Compound 190: N-Benzyl-4-methoxy-3- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 189 using benzyl amine (70% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.92 (s, 1H), 8. 90 (br s, 1H), 8.73 (s, 1H), 8. 33 (s, 1H), 8.23 (s, 1H), 7.60 (d, 1H), 7.33 (m, 4H), 7.23 (m, 1H), 7.12 (d, 1H), 4.44 (s, 2H), 3.96 (s, 3H) LRMS (apci positive) m/e 378.1 (M+1) Compound 121.

4-Methoxy-N-phenethyl-3- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 189 using phenethyl amine (68% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8. 92 (s, 1H), 8.64 (s, 1H), 8.40 (m, 1H), 8.35 (s, IH), 8. 23 (s, 1H), 7.52 (d, 1H), 7.33-7.18 (m, 5H), 7.08 (d, 1H), 3.96 (s, 3H), 3.44 (m, 2H), 2.82 (m, 2H) LRMS (apci positive) m/e 392.1 (M+1) Compound 192: 4-Methoxy-N- (3-phenyl-propyl)-3- (3-pyrazin-2-yl-ureido)-benzarride Prepared according to the procedure of Compound 189 using phenpropyl amine (65% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8. 92 (s, 1H), 8.64 (s, 1H), 8.36 (s, 1H), 8.23 (s, 1H), 7.56 (d, 1H), 7. 32-7. 22 (m, 5H), 7.18 (m, 1H), 7.10 (d, 1H), 3.97 (s, 3H), 3.24 (m, 2H), 2.62 (dd, 2H), 1.82 (m, 2H) LRMS (apci positive) m/e 406.1 (tvI+1) Compound193: N- (2-Benzenesulfonyl-ethyl)-4-methoxy-3- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 189 using 2-benzenesulfonyl-ethylamine (42% yield).

'H-NMR (400 MHz, d6-DMSO) S 8.90 (s, 1H), 8.58 (s, 1H), 8. 38 (br s, 1H), 8.32 (s, 1H), 8.23 (s, 1H), 7.94 (d, 2H), 7.74 (m, 1H), 7.65 (d, 2H), 7.38 (d, 1H), 7.05 (d, 1H), 3.95 (s, 3H), 3.57 (m, 2H), 3.51 (m, 2H) LRMS (apci positive) m/e 456.0 (M+1) Compound 194: 4-Methoxy-3- (3-pyrazin-2-yl-ureido)-N- (2-pyridin-2-yl-ethyl)-benzamide Prepared according to the procedure of Compound 189 using 2-pyridin-2-yl-ethylamine (16% yield) 'H-NMR (400 MHz, d6-DMSO) 8 8.90 (s, 1H), 8.64 (s, 1H), 8.52 (d, 1H), 8.41 (m, 1H), 8.32 (s, 1H), 8.21 (s, 1H), 7.72 (m, 1H), 7.50 (d, 1H), 7.28 (d, 1H), 7.22 (m, 1H), 7.08 (d, 1H), 3.96 (s, 3H), 3.59 (m, 2H), 2.98 (m, 2H) LRMS (esi positive) m/e 415.2 (M+1) Compound 195: 4-Methoxy-3- (3-pyrazin-2-yl-ureido)-N- (2-pyridin-4-yl-ethyl)-benzamide Prepared according to the procedure of Compound 189 using 2-pyridin-4-yl-ethylamine (41 % yield) 'H-NMR (400 MHz, d6-DMSO) 8 8.91 (s, 1H), 8. 63 (s, 1H), 8.46 (d, 2H), 8.40 (m, 1H), 8.35 (s, 1H), 8.24 (s, 1H), 7.47 (d, 1H), 7.26 (d, 2H), 7.08 (d, 1H), 3.96 (s, 3H), 3.50 (m, 2H), 2.84 (m, 2H) LRMS (esi positive) m/e 415.2 (M+1) Compound 196: N-(1H-Benzoimidazol-2-ylmethyl)-4-methoxy-3-(3-pyrazin-2-yl- ureido)-benzamide Prepared according to the procedure of Compound 189 using C- (1H-benzoimudazol-2-yl)-methylamine (26% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.99 (t, 1H), 8.91 (s, 1H), 8.77 (s, 1H), 8.36 (s, 1H), 8.23 (s, 1H), 7.66 (d, 1H), 7.56 (d, 1H), 7.44 (d, 1H), 7. 15 (m, 3H), 4.65 (d, 2H), 3.97 (s, 3H) LRMS (esipositive) m/e 418 1 (M+1) Compound 197: N-[2-(1H-Indol-3-yl)-ethyl]-4-methoxy-3-(3-pyrazin-2-yl-urei do)-benzamide Prepared according to the procedure of Compound 189 using tryptamine (51% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.92 (s, 1H), 8.69 (s, 1H), 8.43 (t, 1H), 8. 33 (s, 1H), 8.24 (s, 1H), 7.60 (d, 1H), 7.56 (d, 1H), 7. 35 (d, 1H), 7. 17 (s, 1H), 7.11 (d, 1H), 7.07 (m, 1H), 6.97 (m, 1H), 3.96 (s, 3H), 3.54 (m, 2H), 2.95 (m, 2H).

LRMS (esi positive) m/e 431. 1 (M+1) Compound 198: 4-Methoxy-N- [3- (methyl-phenyl-amino)-propyl]-3- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 189 using N-methyl-N-phenyl propyldiamine (81% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.92 (s, 1H), 8.64 (s, 1H), 8.37 (m, 1H), 8.35 (s, 1H), 8.23 (s, 1H), 7.55 (d, 1H), 7.14 (m, 3H), 6.70 (d, 2H), 6.58 (t, 1H), 3.96 (s, 3H), 3.37 (m, 2H), 3.25 (m, 2H), 2.86 (s, 3H), 1.77 (m, 2H).

LRMS (esi positive) m/e 435.2 (M+1) Compound 199: N- (l-Benzyl-pyrrolidin-3-yl)-4-methoxy-3- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 189 using 3-amino-1-benzyl pyrrolidine (48% yield). 'H-NMR (400 MHz, d6-DMSO) 5 8.90 (s, 1H), 8.62 (s, 1H), 8.36 (s, 1H), 8.30 (d, 1H), 8.22 (s, 1H), 7.56 (d, 1H), 7.32 (m, 4H), 7.22 (m, 1H), 7.15 (d, 1H), 4.37 (m, 1H), 3.96 (s, 3H), 3.59 (s, 2H), 2.79 (m, 1H), 2.60 (m, 1H), 2.39 (m, 1H), 2.14 (m, 1H), 1.80 (m, 2H).

LRMS (esi positive) m/e 447.2 (M+1) Compound200: N- (2-Dimethylamino-ethyl)-4-methoxy-N-methyl-3- (3-pyrazin-2-yl-ureido)-benzamide, Prepared according to the procedure of Compound 189 using N, N, N'-trimethyl ethyldiamine (93% yield).

'H-NMR (400 MHz, d6-DMSO) 8 9. 39 (br s, 1H), 8.91 (s, 1H), 8. 35 (s, 1H), 8.33 (d, 1H), 8.23 (s, 1H), 7.16-7.09 (m, 2H), 3.96 (s, 3H), 3.76 (m, 2H), 3. 37 (m, 2H), 2.99 (s, 3H), 2.85 (br s, 6H).

LRMS (esi, positive) m/e 373.2 (M+l) Compound 201: 4-Methoxy-N- (3-methylamino-propyl)-3- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 189 using N-methyl propyldiamine (15% yield).

'H-NMR (400 MHz, D2O) 8 7.98 (m, 2H), 7.91 (s, 1H), 7.82 (s, 1H), 7.17 (d, 1H), 6.73 (d, 1H), 3.73 (s, 3H), 3. 29 (m, 2H), 2.98 (m, 2H), 2.61 (s, 3H), 1.88 (m, 2H).

LRMS (esi, positive) m/e 359.2 (M+1) Compound 202: N- (3-Dimethylamino-propyl)-4-methoxy-3- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 189 using N, N-dimethyl propyldiamine (51% yield).

'H-NMR (400 MHz, D2O) 8 7.98 (s, 1H), 7.96 (s, 2H), 7.81 (s, 1H), 7.16 (d, 1H), 6.72 (d, 1H), 3.73 (s, 3H), 3.29 (m, 2H), 3.09 (m, 2H), 2.80 (s, 6H), 1.93 (m, 2H). LRMS (esi, positive) m/e 373.2 (M+1) Compound 203 : N- (3-Dimethylamino-propyl)-4-methoxy-N-methyl-3- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 189 using N, N, N'-trimethyl propyldiamine (60% yield).

'H-NMR (400 MHz, D2O) 8 8. 37 (s, 1H), 8.19 (s, 1H), 8.04 (s, 1H), 7.80 (d, 1H), 7.13 (m, 1H), 7.01 (m, 1H), 3.82 (s, 3H), 3.51 (m, 2H), 3.11 (m, 2H), 2.96 (s, 3H), 2.80 (s, 6H), 2.01 (m, 2H).

LRMS (esi, positive) m/e 387.1 (M+1) Compound 204: 4-Methoxy-N-[3-(4-methyl-piperazin-1-yl)-propyl]-3-(3-pyrazi n-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 189 using 3- (4-methyl-piperazin-1-yl)-propylamine (57% yield).

H-NMR (400 MHz, D2O) 8 8.18 (s, 1H), 8.06 (s, 1H), 8.04 (s, 1H), 7. 96 (s, 1H), 7.32 (d, 1H), 6.85 (d.

1H), 3.79 (s, 3H), 3.48 (br s, 8H), 3.36 (m, 2H), 3.17 (m, 2H), 2.83 (s, 3H), 1.96 (m, 2H) LRMS (esi, positive) m/e 428.2 (M+1) Compound 205 : {2- [4-Methoxy-3- (3-pyrazin-2-yl-ureido)-benzoylamino]-ethyl}-trimethyl-ammon ium chloride Prepared according to the procedure of Compound 189 using 2-trimethylammonium ethyl amine (63% yield).

H-NMR (400 MHz, D20) 5 8.17 (s, 1H), 8.05 (s, 1H), 8.03 (s, 1H), 7.93 (s, 1H), 7.32 (d, 1H), 6.84 (d, 1H), 3.80 (s, 3H), 3.76 (m, 2H), 3.44 (m, 2H), 3.11 (s, 9H).

LRMS (esi, positive) m/e 373.0 (M+) Compound 206: 4-Methoxy-N- (3-morpholin-4-yl-propyl)-3- (3-pyrazin-2-yl-ureido)-benzamide Prepared according to the procedure of Compound 189 using 3-morpholin-4-yl-propylamine (69% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.91 (s, 1H), 8.63 (s, 1H), 8.36 (s, 1H), 8.35 (m, 1H), 8. 23 (s, 1H), 7.54 (d, 1H), 7.10 (d, 1H), 3.96 (s, 3H), 3.57 (m, 4H), 3.26 (m, 2H), 2.34 (m, 4H), 2. 32 (m, 2H). 1.66 (m, 2H).

LRMS (esi, positive) m/e 415. 2 (M+l) Compound 207: 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzoic acid Step 1: (5-Methyl-pyrazin-2-yl)-carbamic acid tert-butyl ester. To a stirred solution of 5-methyl pyrazine carboxylic acid (13.8 g, 100 mmol) in 300 ml. of toluene at room temperature under nitrogen was added triethyl amine (14 mL, 100 mmol) followed by diphenyl phosphoryl azide (21.6 mL, 100 mmol). After 30 min. at room temperature, 2-methyl-2-propanol (19 mL, 200 mmol) was added and the solution immersed in a 90 °C oil bath. After 2 hours, the reaction was cooled to RT, diluted to 600 mL with EtOAc, and washed 3 x 60 mL with 10% Na2CO3 and 1 x 600 mL with saturated NaCI. The organics were dried (MgS04), filtered and concentrated to a yellow solid (17.5 g, 83%).'H-NMR (400 MHz, CDC13) 8 9.16 (s, 1H), 8.05 (s, 1H), 7.56 (br s, 1H), 2.50 (s, 3H), 1.55 (s, 9H).

Step 2: 5-Methyl-2-aminopyrazine. To a stirred solution of (5-methyl-pyrazin-2-yl)-carbamic acid tert-butyl ester (2.1 g, 10 mmol) in 30 mL CH2C12 at 0 °C under nitrogen was added trifluoroacetic acid (30 mL). The solution was allowed to warm to RT overnight. The solution was rotary evaporated to remove TFA and the residue was redissolved in 200 mL CH2CI, and stirred with 100 mL 10% Na2CO3.

The organics were isolated and the aqueous solution extracted 3 x 100 mL with CH2CI2. The organics were combined, dried (MgS04), filtered and concentrated to an orange solid (1 g, 92%).'H-NMR (400 MHz, CDC13) 8 8.46 (s, 1H), 7.70 (s, 1H), 2.49 (s, 3H). Step 3: 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzoic acid methyl ester. To a stirred solution of 3-methoxy-4- (4-nitro-phenoxycarbonylamino)-benzoic acid methyl ester (11. 7 g, 33.8 mmol) in 34 mL NMP at room temperature under nitrogen was added 5-methyl-2-aminopyrazine (3.69 g, 33.8 mmol) and the reaction was immersed in an 85 °C oil bath. After 6 hours the reaction was allowed to cool to room temperature and a precipitate formed. EtOAc (200 mL) was added and the precipitate was isolated by filtration (4.7 g, 44%).'H-NMR (400 MHz, d6-DMSO) 8 8. 79 (br s, 1H), 8. 36 (d, 1H), 8. 23 (s, 1H), 7.60 (d, 1H), 7. 52 (s, 1H), 3.98 (s, 3H), 3.81 (s, 3H), 2.42 (s, 3H).

Step 4: 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzoic acid. To a stirred suspension of 3- methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzoic acid methyl ester (7. 15 g, 22.6 mmol) in 3: 1 MeOH: H20 (226 mL) at RT under N2 was added lithium hydroxide monohydrate (9.5 g, 226 mmol) as a solid and the mixture heated to 65 °C. After reaching temperature, the suspension gradually became a bright yellow solution. After about 4 hours a precipitate formed but the reaction was continued overnight. After cooling to RT, MeOH was removed by rotovap and the aqueous suspension diluted with 100 mL H2O and neutralized to pH=5 with concentrated HCI. As pH=5 was approached, the suspension turned from yellow to white. The suspension was then filtered through paper on a large ceramic funnel. The filtration went very slowly, taking several hours. The filter cake was washed twice with When most of the HO was removed, the residue was dried under high vacuum in a dessicator overnight to give the free acid as a white solid (6 g, 88%). 1H-NMR (400 MHz, d6-DMSO) 8 8. 79 (br s, 1H), 8.36 (d, 1H), 8.22 (s, 1H), 7.57 (d, 1H), 7.51 (s, 1H), 3.97 (s, 3H), 2.42 (s, 3H).

Compound 208: 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-N- (2-pyridin-2-yl-ethyl)-benzamide To a stirred solution of 3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzoic acid (30 mg, 0.1 mmol) in 1 mL NMP at RT in a capped reaction vial was added HBTU (42 mg, 0.11 mmol). The suspension was stirred for 15 minutes and then treated with 2-ethylaminopyridine (13.2 pL, 0.11 mmol) followed by Hunigs Base (35 µL, 0.2 mmol). After stirring overnight, NMP was removed by bulb to bulb transfer at 70 °C under high vacuum and the residue stirred with a mixture of CHiC12 (10 mL) and 10% Na2CO3 (10 mL) until complete dissolution occurred. The organics were isolated, dried (MgS04), filtered and concentrated. The residue was triturated with EtOAc to produce a solid which was isolated by filtration (71% yield). 'H-NMR (400 MHz, d6-DMSO) 8 8.90 (s, 1H), 8.58 (d, 1H), 8. 50 (t, 1H), 8.23 (d, 1H), 8. 21 (s, 1H), 7.82 (t, 1H), 7.48-7.31 (m, 4H), 3.97 (s, 3H), 3.62 (m, 2H), 3.04 (m, 2H), 2.42 (s, 3H).

LRMS (esi, positive) m/e 407.1 (M+1) Compound 209 : N- (1-Benzyl-piperidin-4-yl)-3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzamide Prepared according to the method of Compound 208 using 4-amino-1-benzyl piperidine except the crude product was purified by chromatography on a Biotage 12S column eluting with 92.5/7.5 CHzCl2/MeOH. (61% yield).

'H-NMR (400 MHz, CDC13/CD30D) 8 8.44 (br s, 1H), 8. 32 (d, 1H), 8. 09 (s, 1H), 7.47 (s, 1H), 7.38- 7.28 (m, 6H), 7.09 (d, 1H), 4.00 (m, 1H), 3.99 (s, 3H), 3.60 (s, 2H), 2.98 (m, 2H), 2.52 (s, 3H), 2.25 (m, 2H), 2.00 (m, 2H), 1.64 (m, 2H) LRMS (esi, positive) m/e 475.2 (M+l) Compound 210 : N- (3-Dimethylamino-propyl)-3-methoxy-4- [3- 5-methyl-pyrazin-2-yl)-ureido]-benzamide Prepared according to the method of Compound 208 using N, N-dimethyl propyldiamine (70% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.90 (br s, 1H) 8.55 (t, 1H) 8.26 (d, IH), 8. 22 (s, 1H), 7.52 (s, 1H), 7.50 (d, 1H), 3.98 (s, 3H), 3. 32 (m, 2H), 3.07 (m, 2H), 2.77 (s, 6H), 2.41 (s, 3H), 1.89 (m, 2H).

LRMS (esi, positive) m/e 387.1 (M+1) Compound 211 : 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-N- (3-morpholin-4-yl-propyl)-benzamide Prepared according to the method of Compound 208 using 3-morpholin-4-yl-propylamine (79% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.90 (br s, 1H), 8.57 (t, 1H), 8.25 (d, 1H), 8.21 (s, 1H), 7.51 (s, 1H), 7. 50 (d, 1H), 3.97 (s, 3H), 3.62 (m, 2H), 3.31 (m, 8H), 3.12 (m, 2H), 2.41 (s. 3H), 1.92 (m, 2H) LRMS (esi, positive) m/e 429.1 (M+1) Compound212: N- (2-Dimethylamino-2-phenyl-ethyl)-3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzamide Prepared according to the method of Compound 208 using N- (2-dimethylamino)-2-phenyl ethyl amine except the crude product was purified by chromatography on a Biotage 12S column eluting with 92.5/7.5 CH2CIz/MeOH. (12% yield).

'H-NMR (400 MHz, d6-DMSO) 8 10. 05 (s, 1H), 8.88 (s, 1H), 8.20 (m, 3H), 7.41-7.19 (m, 7H), 3.96 (s, 3H), 3.78 (m, 1H), 3.70 (m, 1H), 3.57 (m, 1H), 3.38 (m, 1H), 2. 50 (s, 6H), 2.40 (s, 3H) LRMS (esi, positive) m/e 448.9 (M+1) Compound 213: N- (2-Dimethylaniino- I-phenyl-ethyl)-3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzande Prepared according to the method of Compound 208 using N- (2-dimethylamino)-l-phenyl ethyl amine except the crude product was purified by chromatography on a Biotage 12S column eluting with 92.5/7.5 CH2Cl2/MeOH. (27% yield).

'H-NMR (400 MHz, d6-DMSO) 8 11.61 (br s, 1H), 9.71 (br s, 1H), 8. 42 (d, 1H), 8. 36 (s, 1H), 8.09 (s, 1H), 7.52-7.21 (m, 7H), 5.05 (br s, 1H), 3.93 (s, 3H), 2.80 (m, 1H), 2.52 (s, 3H), 2.37 (s, 6H), 1.79 (br s, 2H) LRMS (esi, positive) m/e 449.0 (M+1) Compound 214 : N-(1-Aza-bicyclo [2.2.2] oct-3-yl)-3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzamide Prepared according to the method of Compound 208 using 1-aza-bicyclo [22. 2] oct-3-ylamine (27% yield).

H-NMR (400 MHz, d6-DMSO) 8 10.17 (br s, 1H), 8.90 (br s, 1H), 8. 24 (d, 111), 8.23 (s, 1H), 8.17 (d, 1H), 7.53 (d, 1H), 7.50 (s, 1H), 3.98 (s, 3H), 3.95 (m, 1H), 3.10 (m, 1H), 2.90 (m, 1H), 2.64 (m, 4H), 2.42 (s, 3H), 1.87 (m, 1H), 1.80 (m, 1H), 1.59 (m, 2H), 1.31 (m, 1H).

LRMS (esi, positive) m/e 411. 0 (M+1) Compound215: 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-N- (3-R.-1-pyridin-2-ylmethyl-pyrrolidin-3-. yl)- benzamide Step 1: (3-R-1-Pyridin-2-ylmethyl-pyrrolidin-3-yl)-carbamic acid tert-butyl ester. To a stirred solution of (R)-boc-3-aminopyrrolidine (372.5 mg, 2 mmol) in dichloroethane (6 mL) at room temperature under nitrogen was added pyridine-2-carboxaldehyde (190 iL, 2 mmol) followed by sodium triacetoxyborohydride (593 mg, 2.8 mmol). The reaction was stirred at room temperature overnight and was then quenched by addition of saturated NaHC03 (6 mL) with stirring for 15 minutes. The reaction was then partitioned between CH2C12 (25 mL) and 10% Na2CO3 (25 mL). The organics were isolated, dried (MgS04), filtered and concentrated to the pure product (526 mg, 95%).

Step 2: 3-R-1-Pyridin-2-ylmethyl-pyrrolidin-3-ylamine dihydrochloride. A stirred solution of (3-R-1- pyridin-2-ylmethyl-pyrrolidin-3-yl)-carbamic acid tert-butyl ester (277 mg, 1 mmol) in 10 mL. 4N HCl in dioxane at room temperature in a capped flask was reacted overnight. The reaction was concentrated by rotary evaporation and high vacuum to give the di HCl salt (250 mg, quantitative).

Step 3: Prepared according to the procedure of Compound 208 except 3-R-1-pyridin-2-ylmethyl- pyrrolidin-3-ylamine dihydrochloride salt was mixed with excess DIEA (70 pL, 0.4 mmol) in 500 gL NMP to form a solution which was added to the acid/HBTU mixture. The crude product was purified by chromatography on a Biotage 12S column eluting with 9/1 CH2ClZ/MeOH (60% yield).

'H-NMR (400 MHz, CDC13) 8 8.57 (s, 1H), 8.51 (d, 1H), 8.39 (d, 1H), 8.17 (s, 1H), 8.09 (s, 1H), 7.92 (s, 1H), 7.64 (d, 1H), 7.50 (s, 1H), 7.30 (d, 1H), 6.70 (d, 1H), 4.69 (m, 1H), 4.00 (s, 3H), 3.66 (dd, 2H), 2.98 (m, 1H), 2.80 (m, 1H), 2. 70 (m, 1H), 2.53 (s, 3H), 2.39 (m, 3H), 1.76. (m, 1H). LRMS (esi, positive) m/e 462.3 (M+1) Compound 216: 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-N- (3-R-1-methyl-pyrrolidin-3-yl)-benzamide Prepared according to the procedure of Compound 215 using 3-(R)-amino-1-methyl pyrrolidine (29% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.78 (br s, 1H), 8.36 (d, 1H), 8.22 (d, 1H), 8.20 (s, 1H), 7.52 (s, 1H), 7.50 (d, 1H), 4.39 (m, 1H), 3.96 (s, 3H), 2.64 (m, 1H), 2.61 (m, 1H), 2.43, (s, 3H), 2. 39 (m, 2H), 2. 24 (s, 3H), 2.17 (m, 1H), 1.76 (m, 1H). LRMS (esi, positive) m/e 385. 3 (M+1) Compound 217: N- (3-R-1-Benzyl-pyrrolidin-3-yl)-3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzanide Prepared according to the method of Compound 215 using 3- (R)-amino-1-benzyl pyrrolidine except the crude product was purified by chromatography on a Biotage 12S column eluting with 92.5/7.5 CH2Cl21MeOH. (54% yield).

'H-NMR (400 MHz, CDC13) 8 9.09 (br s, 1H), 8.43 (d, 1H), 8.09 (s, 1H), 8. 02 (s, 1H), 7.57 (m, 1H), 7.53 (s, 1H), 7.46 (d, 1H), 7.33-7.22 (m, 4H), 4.79 (m, 1H), 4.00 (s, 3H), 3.78 (dd, 2H), 3.13 (m, 2H), 2.76 (m, 1H), 2.55 (s, 3H), 2.44 (m, 1H), 1.79 (m, 1H). LRMS (esi, positive) m/e 461.1 (M+1) Compound 218 : 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-N- (3-R-1-pyridin-4-ylmethyl-pyrrolidin-3-yl)- benzamide Prepared according to the procedure of Compound 215 using 1- (R)-Pyridin-4-ylmethyl-pyrrolidin-3- ylamine (60% yield).

'H-NMR (400 MHz, CDCl3) # 8.56 (d, 2H), 8.41 (d, 1H), 8.21 (d, 1H), 8.20 (s, 1H), 8.08 (s, 1H), 7.52 (s, 1H), 7.32 (d, 1H), 7.25 (d, 2H), 6.71 (d, 1H), 4.73 (m, 1H), 4.00 (s, 3H), 3.66 (dd, 2H), 2.97 (m, 1H), 2.82 (m, 1H), 2.71 (m, 1H), 2.54 (s, 3H), 2.39 (m, 2H), 1.78 (m, 1H).

LRMS (esi, positive) m/e 462.3 (M+1) Compound 219: 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-N- (3-R-1-thiophen-2-ylmethyl-pyrrolidin-3-yl)- benzamide Prepared according to the procedure of Compound 215 using (R)-l-thiophen-2-ylmethyl-pyrrolidin-3- ylamine (60% yield).

'H-NMR (400 MHz, CDCl3) 6 8.96 (br s, 1H), 8.43 (d, 1H), 8. 09 (d, 2H), 7.57 (s, 1H), 7.50 (d, IH), 7.10 (d, 1H), 6.91 (m, 2H), 4.80 (m, 1H), 4.00 (s, 3H), 3.96 (dd, 2H), 3.17 (m, 1H), 3.08 (m, 1H) 2. 71 (m, 1H), 2.54 (s, 3H), 2.42 (m, 2H), 1.80 (m, 1H). LRMS (esi, positive) m/e 467, 2 (M+1) Compound 220: N- (3-R-1-Cyclohexylmethyl-pyrrolidin-3-yl)-3-methoxy-4- 3- (5-methyl-pyrazin-2-yl)-ureido]- benzamide Prepared according to the procedure of Compound 215 using 1-cyclohexylmethyl-pyrrolidin-3-R- ylamine (71% yield).

'H-NMR (400 MHz, CDC13) 8 8.89 (br s, 1H), 8.32 (d, 1H), 8.23 (d, 1H), 8.21 (s, 1H), 7.51 (s, 1H), 7.49 (d, 1H), 4.38 (m, 1H), 3.96 (s, 3H), 2.76 (m, 1H), 2.56 (m, 1H), 2.42 (s, 3H), 2. 38 (m, 1H), 2.25- 2.06 (m, 3H), 1.77 (m, 3H), 1.61 (m, 3H), 1.40 (m, 1H), 1.19 (m, 3H), 0.83 (m, 2H) LRMS (esi, positive) m/e 467.3 (M+1) Compound 221 : 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureide]-N- (3-R-1-methyl-pyrrolidin-3-yl)-benzamide Prepared according to the procedure of Compound 215 using 1-methyl-pyrrolidin-3-R-ylamine (51% yield).

IH-NMR (400 MHz, d6-DMSO) 8 8.78 (br s, 1H), 8. 35 (d, 1H), 8.22 (d, 1H), 8.21 (s, 1H), 7.52 (s, 1H), 7.49 (d, 1H), 4.39 (m, 1H), 3.96 (s, 3H), 2.66 (m, 1H), 2.61 (m, 1H), 2.43 (s, 3H), 2.39 (m, 2H), 2.24 (s, 3H), 2. 17 (m, 1H), 1. 75 (m, 1H) LRMS (esi, positive) m/e 385.4 (M+1) Compound 222 : N- (3-S-1-Benzyl-pyrrolidin-3-yl)-3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzamide Prepared according to the method of Compound 215 using 1-Benzyl-pyrrolidin-3-S-ylamine except the crude product was purified by chromatography on a Biotage 12S column eluting with 92.5/7.5 CH2ClJMeOH. (54% yield).

'H-NMR (400 MHz, CDCl3) 8 8.81 (br s, 1H), 8.43 (d, 1H), 8.06 (d, 2H), 7.53 (s, 1H), 7.42 (d, 1H), 7. 35-7. 20 (m, 5H), 4.78 (m, 1H), 3.99 (s, 3H), 3.78 (dd, 2H), 3.15 (m, 1H), 3.04 (m, 1H), 2.77 (m, 1H), 2.54 (s, 3H), 2.44 (m, 1H), 1.79 (m, 2H). - LRMS (esi, positive) m/e 461.1 (M+1) Compound 223: 3-Methoxy-4-[3-(5-methyl-pyrazin-2-yl)-ureido]-N-(3-S-1-pyri din-2-ylmethyl-pyrrolidin-3-yl)- benzamide Prepared according to the procedure of Compound 215 using 1-pyridin-2-ylmethyl-pyrrolidin-3-S- ylamine (60% yield).

'H-NMR (400 MHz, CDCl3) 8 8.56 (d, 1H), 8.38 (s, 1H), 8.32 (d, 1H), 8.06 (s, 1H), 7.68 (t, 1H), 7.51 (s, 1H), 7.40 (d, 1H), 7.35 (d, 1H), 7.21 (m, 1H), 4. 72 (m, 1H), 4.00 (s, 3H), 3.79 (dd, 2H), 3.40 (m, 1H), 3.03 (m, 1H), 2.86 (m, 1H), 2.64 (m, 1H), 2.53 (s, 3H), 2.35 (m, 1H), 1.78 (m, 2H). LRMS (esi, positive) m/e 462 1 (M+1) Compound 224 : 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-N- (3-S-1-pyridin-3-ylmethyl-pyrrolidin-3-yl)- benzamide Prepared according to the procedure of Compound 215 using 1-pyridin-3-ylmethyl-pyrrolidin-3-S- ylamine (60% yield).

'H-NMR (400 MHz, CDC13) 5 8. 56 (s, 1H), 8.51 (m, 1H), 8.39 (d, 1H), 8.18 (s, 1H), 8.08 (s, 2H), 7.64 (d, 1H), 7.50 (s, 1H), 7. 33 (d, 1H), 6.77 (d, 1H), 4.72 (m, 1H), 4.00 (s, 3H), 3.66 (dd, 2H), 2.97 (m, 1H), 2.82 (m, 1H), 2.71 (m, 1H), 2.54 (s, 3H), 2.40 (m, 2H), 1.76 (m, 2H).

LRMS (esi, positive) m/e 462. 3 (M+1) Compound 225 : 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-N- (3-S-1-pyridin-4-ylmethyl-pyrrolidin-3-yl)- benzamide Prepared according to the procedure of Compound 215 using 1-pyridin-4-ylmethyl-pyrrolidin-3-S- ylamine (60% yield).

'H-NMR (400 MHz, CDCl3) 8 8. 56 (d, 2H), 8.41 (d, 1H), 8.24 (s, 1H), 8.20 (s, 1H), 8.08 (s, 1H), 7.53 (s, 1H), 7. 32 (d, 1H), 7.24 (d, 2H), 6.75 (d, 1H), 4.72 (m, 1H), 4.00 (s, 3H), 3.67 (dd, 2H), 2.98 (m, 1H), 2.82 (m, 1H), 2.71 (m, 1H), 2.54 (s, 3H), 2.40 (m, 2H), 1.79 (m, 2H).

LRMS (esi, positive) m/e 462.3 (M+1) Compound 226 : 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-N- (=-S-1-thiophen-2-ylmethyl-pyrrolidin-3-yl)- benzamide Prepared according to the procedure of Compound 215 using 1-thiophen-2-ylmethyl-pyrrolidin-3-S- ylamine (55% yield).

'H-NMR (400 MHz, CDCl3) 8 8.71 (br s. 1H), 8.42 (d, 1H), 8.11 (d, 2H), 7.55 (s, 1H), 7.44 (d, 1H), 7.39 (d, 1H), 7.20 (d, 1H), 6.91 (m, 2H), 4.78 (m, 1H), 4.00 (s, 3H), 3.94 (dd, 2H), 3.17 (m, 1H), 3.05 (m, 1H), 2.69 (m, 1H), 2.53 (s, 3H), 2,42 (m, 2H), 1.80 (m, 2H). LRMS (esi, positive) m/e 467.2 (M+1) Compound 227 : N- (3-S-1-Cyclohexylmethyl-pyrrolidin-3-yl)-3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]- benzamide Prepared according to the procedure of Compound 215 using 1-cyclohexylmethyl-pyrrolidin-3-S- ylamine (60% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.78 (s, 1H), 8.31 (d, 1H), 8.23 (d, 1H), 8.20 (s, 1H), 7.51 (s, 1H), 7.48 (d, 1H), 4.37 (m, 1H), 3.96 (s, 3H), 2.76 (m, 1H), 2.56 (m, 1H), 2.42 (s, 3H), 2.38 (m, 1H), 2.25- 2.06 (m, 3H), 1.77 (m, 3H), 1.61 (m, 3H), 1.40 (m, 1H), 1.19 (m, 3H), 0.83 (m, 2H). LRMS (esi, positive) m/e 467.3 (M+1) Compound 228: N- (3-S-1-Benzyl-pyrrolidin-3-yl)-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-3-trifluoromethoxy-benzamide Step 1: 4-Amino-3-trifluoromethoxy-benzoic acid methyl ester. To a stirred solution of 4-amino-3- trifluoromethoxy-benzoic acid (1.2 g, 5.4 mmol) in 16 mL of 4: 1 THF : MeOH at 0 °C was added TMS- diazomethane (2M solution in hexane, 6 mL, 12 mmol) dropwise and conversion was monitored by TLC in 2/3 EtOAc/hexane. When complete, the reaction was concentrated to a white solid corresponding to the methyl ester (1.27 g, quantitative).

Step 2: 4- (4-Nitro-phenoxycarbonylamino)-3-trifluoromethoxy-benzoic acid methyl ester. To a stirred solution of 4-amino-3-trifluoromethoxy-benzoic acid methyl ester (1 38 gm, 5. 9 mmol) in 18 mL CH2C12 at 0 °C under nitrogen was added pyridine (521 ut, 6.4 mmol) followed by p- nitrophenylchloroformate (1.18 gm, 5.9 mmol). After 4 hours at 0 °C the reaction was diluted to 60 mL with CH2Cl2 and washed 2 x 60 mL with 2N HCI, 1 x 60 mL with H20 and I x 60 mI, with saturated NaCI. The organics were dried (MgS04), filtered and concentrated to a white solid corresponding to the carbamate (2.1 gm, 89 %).

Step 3: 4- [3- (5-Methyl-pyrazin-2-yl)-ureido]-3-trifluoromethoxy-benzoic acid methyl ester. To a stirred solution of 4- (4-nitro-phenoxycarbonylamino)-3-trifluorornethoxy-benzoic acid methyl ester (400 mg, 1 mmol) in 1 mL of NMP in a capped reaction vial at room temperature was added 2-amino- 5-methyl-pyrazine (109 mg, 1 mmol) and the solution heated to 90 °C for 6 hours. After cooling to room temperature, the solution was diluted to 30 mL with EtOAc and washed 4 x 30 mL with 10% NaEICO3 to remove the phenol by-product and 1 x 30 mL with saturated NaCI. The organics were dried (MgS04), filtered and concentrated. Trituration and filtration with EtOAc gave a beige solid corresponding to the urea ester (136 mg, 37%).

Step 4: 4- [3- (5-Methyl-pyrazin-2-yl)-ureido]-3-trifluoromethoxy-benzoic acid. To a stirred suspension of 4- [3- (5-methyl-pyrazin-2-yl)-ureido]-3-trifluoromethoxy-benzoic acid methyl ester (136 mg, 0.37 mmol) in 4 mL 3: 1 MeOH : H20 under nitrogen was added lithium hydroxide monohydrate (154 mg, 3.7 mmol), and the reaction heated to 65 °C. The reaction turned yellow/green and eventually became a solution. After stirring overnight, the reaction was cooled to RT and partially concentrated by rotovap to remove most of the MeOH. The residual suspension was neutratized with 2N HCI until pH=6. The reaction formed a floculent precipitate which was filtered off with H2O and dried overnight under high vacuum to give the acid as a white solid (102 mg, 78 %).

Step 5-To a stirred suspension of 4- [3- (5-methyl-pyrazin-2-yl)-ureido]-3-trifluoromethoxy-benzoic acid (102 mg, 0.29 mmol) in 2.9 mL NMP in a capped reaction vial at room temperature was added HBTU (109 mg, 0. 29 mmol). After 15 minutes 3-S-1-Benzyl-pyrrolidin-3-ylamine dihydrochloride (prepared analogously to l-Pyridin-2-ylmethyl-pyrrolidin-3-ylamme used in the synthesis of Compound 215) (73 mg, 0.29 mmol) was added followed by DIEA (200 pL, 1.2 mmol). The reaction was stirred overnight and NMP was then removed under high vacuum at 70 °C. The residue was partitioned between 30 mL CH2CI2 and 30 mL 10% Na2CO3. The organics were isolated and washed 1 x 30 mL with saturated NaCI, dried (MgS04), filtered and concentrated to a yellow foam corresponding to the desired amide (103 mg, 70%).

'H-NMR (400 MHz, CDC13/CD30D) 6 8.56 (d, 1H), 8.37 (br s, 1H), 8. 08 (s, 1H), 7.84 (s, 1H), 7.66 (d, 1H), 7.40-7.28 (m, 6H), 4.73 (m, 1H), 3.68 (dd, 2H), 2. 77 (dd, 2H), 2.53 (s, 3H), 2.43 (m, 1H), 2.37 (dd, 2H), 1.78 (m, 1H) LRMS (esi, positive) m/e 514.9 (M+1) Compound229: N- (1-Benzyl-piperidin-4-ylmethyl)-3-methoxy-4- [3- (5-methyl-pyrazin-2-yl j-meido]-benzamide Step 1: N-benzylisoripecotamide. To a suspension of isonipecotamide (12.3 g, 96 mmol) in 200 mL of dichloromethane, was added, benzaldehyde (10.6 g, 100 mmol) and sodium triacetoxyborohydride (29.7 gm, 140 mmol) and the mixture was stirred at room temperature for 5 days. The thick white mixture was diluted with 100 mL water and extracted with EtOAc (2 x 20 mL). The aqueous phase was basified with IN NaOH to pH > 12. The resulting white precipitate was collected by suction filtration. The white solid was subsequently taken up in 50 mL of EtOAc, and was washed with 20 mL of brine, then dried (MgSO4), filtered and concentrated to give 10.84 g (50 %) of the desired product.

'H-NMR (400 MHz, CDCl3) 8 7.85 (m, SH), 5.45 (s, 1H), 5. 34 (s, 1H), 3.5 (s, 2H), 2.93 (d, J = 10.96 Hz, 2H), 2.16 (t, J= 12.13 Hz, 1H). 2.01 (t, J= 11. 74 Hz, 2H), 1.87 (d, J= 12.52 Hz, 2H), 1.76 (q, J= 12.52 Hz, 2H).

Step 2 : 4-Aminomethyl-l-benzyl piperdine. To a solution of N-benzylisonipecotamide (7. 34g, 34 mmol) in 60 mL of anhydrous THF, was added, LiAlH4 (1. 9 g, 51 mmol) and the mixture was stirred at room temperature for 10 minutes followed by heating to reflux for 3 h. The reaction was quenched by addition of 100 mL sat'd. sodium potassium tartrate, and was extracted with EtOAc (3 x 50 mL). The combined extracts were washed with 20 mL water and 20 mL brine then dried over MgSO4, filtered and concentrated to give the desired product.'H-NMR (400 MHz, CDCl3) 8 7.13 (m, 5H), 3.42 (s, 2H), 2.91 (m, 2H), 2.59 (m, 2H), 1.97 (m, 2H), 1.62 (m, 2H), 2.21 (m, 5H).

Step 3: N- (l-Benzyl-piperidin-4-ylmethyl)-3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]- benzamide. Prepared from 4-aminomethyl-l-benzyl piperdine, according to the procedure of Compound208: 'H-NMR (400 MHz, d6-DMSO) 8 10.1 (s, 2H), 8.81 (s, 1H), 8.48 (s, 1H), 8.26 (m, 2H), 7.52 (m, 7H), 3.97 (s, 5H), 3.33 (s, 3H), 3.18 (s, 3H), 2.43 (s, 3H), 1.75 (s, 3H). 1.44 (s, 2H). MS APCI-pos, M+1 = 489.1.

Compound230: N- [3-S-1- (4-Fluoro-benzyl)-pyrrolidin-3-yl]-3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]- benzamide Step 1: (3S)-3- (tert-Butoxycarbonylamino)-1- (4-fluoro-benzyl)-pyrrolidine. To a solution of (3S)- (+)- 3- (tert-butoxycarbonylamino) pyrrolidine (410 mg, 2.20 mmol), in 22 mL EtOH, was added, (288 uL, 2. 31 mmol) of 4-fluorobenzyl bromide, and (788 mg, 2.42 mmol) of finely powdered cesium carbonate. The stirred reaction mixture was heated at 80 °C, under nitrogen for 3h, after which time, TLC indicated the reaction was complete. The reaction was then concentrated in vacuo to about 5 mL, and was then diluted with 30 mL of EtOAc, and washed with 20 mL of 5% NH40H. The aqueous fraction was extracted with diethyl ether (3 x 20 mi.). The combined organics were washed with 20 mL of brine, dried (MgS04), filtered and concentrated. The crude white solid was triturated with 1: 1 ether-hexane to give 501 mg (77%) of the desired product as a white solid.'H-NMR (400 MHz, CDC13) S 7.2-7. 3 (m, 2H), 6.9-7.1 (m, 2H), 4.85 (br. s, 1H, exchanges), 4.18 (br. s, 1H), 3.55 (s, 2H), 2.65 (br. m, 1H), 2.58 (m, 1H), 2.50 (m, 1H), 2.2-2.4 (m, 2H), 1.5-1.7, (m, 1H), 1.4 (s, 9H).

Step 2: (3S)-3-Amino-1- (4-fluoro-benzyl)-pyrrolidine. A solution of (3S)-3-(tert- butoxycarbonylamino)-1-(4-fluoro-benzyl)-pyrrolidine (400 mg, 1.36 mmol) was stirred in 15 mL of formic acid at room temperature. After 3h, the clear colorless solution was concentrated to dryness and the residue was taken-up in 20 mL of EtOAc and washed with 10 mL of 5% NH40H, followed by 10 mL. of brine. The solution was then dried over MgS04, filtered and concentrated to give 240 mg (91%) of product, as a yellow oil.'H-NMR (400 MHz, CDC13) 8 7.28 (m, 2H), 6.99 (m, 2H), 3.56 (d, J= 6. 2 Hz, 2H), 3.47-3.53 (m, 1H), 2.67-2.71 (m, 2H), 2.42-2.48 (m, 1H), 2.28-2.31 (m, 1H), 2.21-2.28 (m, 1H), 1.59 (s, 2H, NH2), 1.43-1.52 (m, 1H).

Step 3: N [ (3S)-1- (4-Fluoro-benzyl)-pyrrolidin-3-yl]-3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]- benzamide. Prepared from (3S)-3-amino-1- (4-fluoro-benzyl)-pyrrolidine, according to the procedure of Compound 208.'H-NMR (400 MHz, CDC13) 8 10.8 (s, 2H), 8.8 (s, 1H), 8. 38 (s, 1H), 8. 27 (s, 1H), 8.24 (s, 2H), 7.51 (s, 1H), 7.49 (s, 1H), 7. 37 (s, 2H), 7.15 (m, 2H), 4.4 (br. s, 1H), 3.98 (s, 3H), 3.6 (br. s, 2H), 3.06 (br. s, 1H), 2.65 (br. s, 1H), 2.81 (br. s, 1H), 2.43 (s, 3H), 2.16 (s, 1H), 1.83 (s, 1H). MS apc :- pos M+1 = 479.2.

Compound 231 : 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzoic acid 3-S-1-benzyl-pyrrolidin-3-yl ester Step 1: 3-Methoxy-4-nitro-benzoyl chloride. Thionyl chloride 3.7 mL, (50 mmol) was added drop wise, at room temperature, to a stirred solution of 3-inethoxy-4-nitro-benzoic acid (1.0 g, 5.07 mmol) in 15 mL of dioxane under a nitrogen atmosphere. After the addition was complete, the reaction was allowed to stir at room temperature for 1 hr. The reaction flask was then fitted with a distillation head, and excess thionyl chloride and about l/z of the solvent was removed by distillation in a 120 °C oil bath.

The remaining solvent was removed by rotary evaporation, and the residue was taken up in 20 mL toluene and again about'4 of the solvent was removed by distillation. The remaining solution was then cooled to 0 °C, and the white solid which precipitated, was collected by suction filtration, and rinsed with 1-1 Et2O-hexane.'H-NMR (400 MHz, CDCl3) 8 7.87 (d, J= 7.83 Hz, 1H), 7.82 (d, J= 7.83 Hz, 1H), 7.77 (s, 1H), 4.04 (s, 3H).

Step 2: (3S)-l-Benzyl-pyrrolidin-3-ol. A solution of (3S)-3-hydroxypyrrolidine (2.0 g, 25 mmol) in 50 mL CH, C12, under a nitrogen atmosphere, was cooled to 0 °C and benzaldehyde (2.92 g, 27.5 mmol) was added, followed by lg of powdered 4A molecular sieves. Sodium triacetoxyborohydride (7.4 gm, 35 mmol) was added in several portions over 30 min, and the reaction was allowed to stir at room temperature for 18h. The reaction was again cooled to 0 °C and quenched by addition of 10 mL methanol, followed by 5 mL of 1N HC1. The molecular sieve solids were removed by filtration through a glass fiber paper, and the filtrate was extracted with 20 mL diethyl ether. The organic phase was discarded, and the aqueous phase was first basified by addition of conc. ammonium hydroxide, and then extracted with EtOAc (3 x 20 mL). The combined extracts were washed with 20 mL brine, dried (MgS04), filtered, and concentrated in vacuo to furnish 3.2 g (73%) of clear yellow oil, which required no further purification.'H-NMR (400 MHz, CDC13) 8 7.2-7.4 (m, SH), 4.33 (m, 1H), 3.63, (s, 2H), 2.83-3.89 (m, 1H), 2.67 (d, J= 9.9 Hz, 1H), 2.53-2.55 (m, 1H), 2.23-2.34 (m, 1H), 2.14-2.20 (m, 2H), 1.70-1.77 (m, 1H).

Step 3: 3-Methoxy-4-nitro-benzoic acid (3S)-l-benzyl-pyrrolidin-3-yl ester. A solution of 3-methoxy- 4-nitro-benzoyl chloride (608 mg, 2.82 mmol) in10 mL of CH2CI2 was added to stirred solution of 500 mg (2.82 mmol) of (3S)-l-benzyl-pyrrolidin-3-ol and 1 mL of pyridine in 15 mL CH, C12 at room temperature. After stirring for 18 h, the reaction mixture was diluted with 20 mL of saturated aqueous NaHC03, and extracted with CH2CI2 (2 x 10 mL). The combined extracts were washed with 20 mL of brine, dried over MgS04, filtered, and concentrated in vacuo to furnish 780 mg (71%) of the desired ester as a yellow oil, which solidified after drying under high vacuum. The resulting solid was further purified by trituration with 1: 1 ether-hexane.'H-NMR (400 MHz, CDCl3) 8 7.83 (d, J = 7. 83 Hz, 1H), 7.74 (s, 1H), 7.68 (d, J= 7.83 Hz, 1H), 7.2-7.4 (m, 5H), 5.44 (br. m, 1H), 4.02 (s, 2H), 3.69 (dd, J= 30 Hz, J= 13 Hz, 1H), 2.8-3.0 (m, 2H), 2.5-2.6 (m, 1H), 2.3-2.45 (m, 1H), 2.0-2.1 (m, 1H) Step 4: 4-Amino-3-methoxy-benzoic acid (3S)-l-benzyl-pyrrolidin-3-yl ester. The aniline was prepared by nickel boride reduction of 3-methoxy-4-nitro-benzoic acid (3S)-l-benzyl-pyrrolidin-3-yl ester analogously to the preparation of 4-[(Benzyl-methyl-arnino)-methyl]-2-methoxy-phenylamine used in the synthesis of Compound 340.'H-NMR (400 MHz, CDC13) 8 7.54 (d, J= 8.21 Hz, 1H), 7.44 (s, 1H), 7.2-7.4 (m, 5H) 6.65 (d, J= 8.21 Hz, 1H), 5.38 (br. m, 1H), 4.22 (s, 1H), 3.90 (s, 3H), 3.69 (q, J = 24.63 Hz, 1H), 3.0 (m, 1H), 2.7-2.9 (m, 2H), 2.5-2.6 (m, 1H), 2. 3-2. 4 (m, 1H), 1.9-2.1 (m, 1H). MS apci-pos M+1=327. 2.

Step 5 : 3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzoic acid (3S)-l-benzyl-pyrrolidin-3-yl ester. Prepared from 4-amino-3-methoxy-benzoic acid (3S)-l-benzyl-pyrrolidin-3-yl ester according to the procedure of Compound 208 :'H-NMR (400 MHz, CDCI3) 6 10.15 (s, 2H), 8.8 (s, 1H), 8.35 (d, J= 8.61 Hz, 1H), 8.25 (s, 1H), 7.58 (d, J= 8.61 Hz, 1H), 7.5 (s, 1H), 7.30-7. 35 (m, 4H), 7.26-7.24 (m, 1H), 5.28 (s, 1H), 3.99 (s, 3H), 3.62 (s, 2H), 2.82 (m, 1H), 2.74 (m, 1H), 2.67 (m, 1H), 2.44 (s, 3H), 2.28 (m, 1H), 1.91 (m, 1H). MS apci-pos M+1 = 462.2.

Compound232: N- (3-S-1-Benzyl-pyrrolidin-3-yl)-3-methoxy-4- [3- (5-trifluoromethyl-pyrazin-2-yl)-ureido]-benzamide Step 1: 3-Methoxy-4- [3- (5-trifluoromethyl-pyrazin-2-yl)-ureido]-benzoic acid methyl ester. To a stirred solution of 5-trifluoromethylaminopyrazine (326 mg, 2 mmol) at room temperature in NMP (2 mL) in a capped reaction vial was added 4-methoxy-3- (4-nitrophenylcarbonylamino)-benzoic acid methyl ester (692 mg, 2 mmol) and the solution was heated to 85° C for 6 hours. The reaction was cooled to RT and triturated with EtOAc (5 mL) and the tan solid that formed was isolated by filtration and rinsing with EtOAc (213 mg, 28%).

'H-NMR (400 MHz, d6-DMSO) 5 10. 78 (s, 1H), 10.16 (br s, 1H), 9.07 (s, 1H), 8.84 (s, 1H), 8.37 (d, 1H), 7.63 (d, 1H), 7.56 (s, 1H), 4.01 (s, 3H), 3.83 (s, 3H) Step 2: 3-Methoxy-4- [3- (5-trifluoromethyl-pyrazin-2-yl)-ureido]-benzoic acid. To a stirred solution of 3-methoxy-4- [3- (5-trifluoromethyl-pyrazin-2-yl)-ureido]-benzoic acid methyl ester (213 mg, 0.575 mmol) in 5.75 mL 3: 1 MeOH:H2O at room temperature under nitrogen was added lithium hydroxide monohydrate (240 mg, 5.8 mmol) and the reaction warmed to 65 °C. After reaching temperature, the suspension gradually became a bright yellow solution. After about 4 hours a precipitate formed but the reaction was continued overnight After cooling to RT, MeOH was removed by rotovap and the aqueous suspension neutralized to pH=5 with concentrated HCI. As pH=5 was approached, the suspension turned from yellow to white. The suspension was then filtered through paper on a ceramic funnel. When most of the H20 was removed the residue was dryed under high vacuum in a dessicator overnight (133 mg, 55%).

Step 3: To a stirred solution of 3-methoxy-4- [3- (5-trifluoromethyl-pyrazin-2-yl)-ureido]-benzoic acid (35 mg, 0.1 mmol) in 0.5 mL NMP in a capped reaction vial at RT was added HBTU (41 mg, 0.11 mmol) and the activation stirred for 15 min. 1-Benzyl-pyrrolidin-3-ylamine dihydrochloride (prepared analogously to l-Pyridin-2-ylmethyl-pyrrolidin-3-ylamine used in the synthesis of Compound 215) (crude 0.1 mmol) was then added as a solution in 0.5 mL NMP with DIEA (69 L, 0.4 mmol). After stirring overnight at RT, NMP was removed by distillation under high vacuum at 70° C. The residue was dissolved in 25 mL CHUCK with a small amount of MeOH and washed 2 x 25 mL with 10% Na2CO3. The organics were dried (MgS04), filtered and concentrated to a residue that was chromatographed on a biotage 12S column with 5/95 MeOH/CH2CI2. This material was concentrated to dryness and triturated/filtered with Et2O to give pure product as a white solid (9.5 mg, 19%).

'H-NMR (400 MHz, CDC13) b 11.53 (s, 1H), 9.86 (br s, 1H), 8.57 (s, 1H), 8.40 (d, 1H), 8. 12 (s, 1H), 7.56 (s, 1H), 7.46 (m, 2H), 7.25 (m, 4H), 4.79 (m, 1H), 4.01 (s, 3H), 3.73 (dd, 2H), 3.22 (m, 2H), 2.76 (m, 1H), 2.47 (m, 2H), 1.76 (m, 1H) LRMS (esi, positive) m/e 515.. 1 (M+1) Compound 233: 5-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-N- (2-pyridin-2-yl-ethyl)-2-tifluoromethyl-benzamide Step 1: Mixture of 5-Fluoro-4-nitro-2-trifluoromethyl-benzoic acid and 5-Fluoro-3-nitro-2- trifluoromethyl-benzoic acid. To a stirred solution of 3-fluoro-6-trifuoromethylbenzoic acid (3.58 gm, 17.2 mmol) in 17 mL concentrated H2SO4 at 0° C was carefully added 70% HN03 (17 mL) dropwise. The reaction was heated to 100° C overnight and was then cooled to room temperature and poured into 250 mL of H2O/ice with stirring. EtOAc (250 mL) was added and the mixture stirred. The layers were separated and the organics washed 1 x 250 mL with H2O and 1 x 250 mL with saturated Nazi. The organics were dried (MgS04), filtered and concentrated to an oil which solidified upon standing. The solid was a 1: 1 mixture of regioisomers and was carried on crude.

Step 2: Mixture of 5-Fluoro-4-nitro-2-trifluorometliyl-benzoic acid methyl ester and 5-Fluoro-3-nitro- 2-trifluoromethyl-benzoic acid methyl ester. To a stirred solution of the nitro acids (crude, 17.2 mmol) in 60 mL of 4: 1 THF: MeOH at 0 °C was added 2M TMS-diazomethane in hexane dropwise until a yellow color persisted. After 30 minutes the reaction was concentrated to a crude oil and used directly in the next step.

Step 3: Mixture of 5-Methoxy-4-nitro-2-trifluoromethyl-benzoic acid methyl ester and 5-Methoxy-3- nitro-2-trifluoromethyl-benzoic acid methyl ester. To a stirred solution of the fluoro nitro esters (crude, 17.2 mmol) in 22 mL MeOH at room temperature was added 150 mL of 0.5 M sodium methoxide in MeOH. The red solution was stirred for 30 minutes and then partitioned between CH., C12 (500 mL) and H2O (500 mL). The organics were washed 2 x 500 mL with HO, dried (MgS04), filtered and concentrated to a white solid.

Step 4: 5-Methoxy-4-amino-2-trifluoromethyl-benzoic acid methyl ester. To a stirred solution of the methoxy nitro esters (crude, 17.2 mmol) in 172 mL MeOH at room temperature purged with nitrogen was carefully added 10% Pd on C (1 gm). The suspension was put through a vacuum/purge cycle three times with hydrogen gas and then held under 1 atmosphere of hydrogen. After stirring overnight the suspension was filtered through GF/F filter paper and concentrated to a clear oil. This material was loaded directly onto a Biotage 40M column with CH2Cl2 and eluted first with 9/1 hexane/EtOAc to elute the undesired hi Rf regioisomer and then with 85/15 hexane/EtOAc to elute the desired lower Rf regioisomer. After concentration, the lower Rf isomer solidified to a clear solid (1.75 gm, 41%).'H- NMR (400 MHz, CDCI3) 8 7. 33 (s, 1H), 6.98 (s, 1H), 3.92 (s, 3H), 3.85 (s, 3H) Step 5: 5-Methoxy-4- (4-nitro-phenoxycarbonylamino)-2-trifluoromethyl-benzoic acid methyl ester.

To a stirred solution of 5-methoxy-4-amino-2-trifluoromethyl-benzoic acid methyl ester (552 mg, 2.22 mmol) in 6.6 mL CH, Cl-, at 0° C under nitrogen was added pyridine (180 PL, 2.22 mmol) followed by p-nitrophenyl chloroformate (448 mg, 2. 22 mmol) as a solid. After stirring for 1 hour, the reaction was diluted to 30 mL with CH2CI2 and washed 2 x 30 mL with 2N HCl and 1 x 30 mL with H, O. The organics were dried (MgSO4), filtered and concentrated to the p-nitrophenyl carbamate which was isolated as a white foam (878 mg, 96%).'H-NMR (400 MHz, CDCl3) # 8.57 (br s, 1H), 8.30 (d, 2H), 7.77 (br s, 1H), 7.40 (d, 2H), 7. 37 (s, 1H), 4.03 (s, 3H), 3.94 (s, 3H) Step 6: 5-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-2-trifluoromethyl-benzoic acid methyl ester.

To a stirred solution of 5-methoxy-4- (4-nitro-phenoxycarbonylamino)-2-trifluoromethyl-benzoic acid methyl ester (878 mg, 2.1 mmol) in 4.2 mL NMP at room temperature under nitrogen was added 2- amino-5-methylpyrazine (232 mg, 2.1 mmol) and the reaction heated to 85 °C. After six hours the reaction was cooled to room temperature and a precipitate formed. The precipitate was triturated with EtOAc (25 mL) and the urea isolated by filtration as a tan solid (470 mg, 58%).

Step 7: 5-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-2-trifluoromethyl-benzoic acid. To a stirred suspension of 5-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-2-trifluoromethyl-benzoic acid methyl ester (257 mg, 0.67 mmol) in 6.7 mL 3: 1 MeOH: H20 at room temperature under nitrogen was added lithium hydroxide monohydrate (281 mg, 6.7 mmol) and the mixture was heated to 85 °C. After stirring overnight the reaction was cooled to room temperature, MeOH was removed by rotovap and the residual suspension was neutralized with concentrated HCl to pH of approximately 5. The suspension was filtered and rinsed with H20 and the filter cake dried under high vacuum to give the acid as a white solid (161 mg, 61%).'H.. NMR (400 MHz, d6-DMSO) 8 8.90 (br s, 1H), 8.63 (s, 1H), 8.22 (s, 1H), 7.38 (s, 1H), 4.00 (s, 3H), 2.42 (s, 3H). LRMS (esi, negative) m/e 369.0 (M-1).

Step 8: 5-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-N- (2-pyridin-2-yl-ethyl)-2-trifluoromethyl- benzamide. To a stirred solution of 5-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-2-tnfluoromethyl- benzoic acid (37 mg, 0.1 mmol) in 1 mL NMP at room temperature in a capped reaction vial was added HBTU (42 mg, 0.11 mmol) and the suspension stirred for 15 minutes. 2-aminoethylpyridine (13 uL, 0.11 mmol) was added followed by DIEA (35 gL, 0.2 mmol) and the reaction was stirred overnight.

NMP was then removed by bulb to bulb transfer under high vacuum at 70 °C and the residue triturated and filtered with EtOAc to give the desired amide as a tan solid (32 mg, 68%).

'H-NMR (400 MHz, d6-DMSO) 8 8.90 (br s, 1H), 8.66 (d, 1H), 8.61 (s, 1H), 8.52 (t, 1H), 8.23 (s, 1H), 8.10 (br m, 1H), 7.61 (br d, 1H), 7.57 (br m, 1H), 7.07 (s, 1H), 4.00 (s, 3H), 3.62 (m, 2H), 3.11 (m, 2H), 2.42 (s, 3H) LRMS (esi, positive) m/e 475.1 (M+1) Compound 234: 3- (3-Dimethylamino-propoxy)-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzoic acid Step 1: 3- (3-Dimethylamino-propoxy)-4-nitro-benzoic acid methyl ester. To a stirred, cooled (about 0°C) solution of 3-hydroxy-4-nitro benzoic acid methyl ester. (394 mg, 2.0 mmol), triphenyl phosphine (525 mg, 2.0 mmol), and 3- (dimethylamino)-propanol (237 1L, 2.0 mmol) in dry tertrahydrofuran (5 mL) was added diisopropyl azodicarboxylate (394 1L, 2. 0 mmol in 1 mL of tetrahydrofuran). After stirring for 12 hours, the reaction was diluted with hydrochloric acid (30 mL of a 1M solution) and extracted with ethyl acetate (2x 50mL). The aqueous solution was basified with 10% aqueous sodium carbonate (50 mL) and the product was extracted with ethyl acetate (3 x 100 mL). The ethyl acetate was washed with brine (1 x 30 mL), then dried (MgS04), and filtered. The filtered solution was concentrated under reduced pressure to provide the desired crude product (86 % yield).'H-NMR (400 MHz, CDC13) 8 7.81 (d, 1H), 7.78 (s, 1H), 7.65 (d, 1H), 4.23 (t, 2H), 3.91 (s, 3H), 2.44 (t, 2H), 2.22 (s, 6H), 2.05 (m, 2H).

Step 2: 4-Amino-3- (3-dimethylamino-propoxy)-benzoic acid methyl ester. To a stirred, cooled (about 0°C) solution of3- (3-dimethylamino-propoxy)-4-nitro-benzoic acid methyl ester (282 mg, 1.0 mmol) in methanol (2 mL) and saturated aqueous ammonium chloride (lnil) was added zinc dust (2.0 mmol).

After stirring for 12 hours, the reaction was diluted with 30 mL of ethyl acetate and washed with 30 mL of 10% aqueous sodium carbonate (2 x 30. mL), brine (1 x 30 mL), then dried (MgS04), and filtered.

The filtered solution was concentrated under reduced pressure to yield the desired product as a tan solid (88% yield).'H-NMR (400 MHz, CDC13) 8 7.52 (d, 1H), 7.48 (s, 1H), 7.25 (s, 1H), 6.65 (d, 1H), 4.29 (br s, 1H), 4.09 (t, 2H), 3.85 (s, 3H), 2.49 (t, 2H), 2. 25 (s, 6H), 2.00 (m, 2H).

Step 3: 3- (3-Dimethylamino-propoxy)-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzoic acid methyl ester.

To a stirred, cooled (about 0°C) solution of 4-amino-3- (3-dimethylamino-propoxy)-benzoic acid methyl ester (252 mg, 1.0 mmol) in toluene (3.0 mL) was added triethylamine (139 uL, 1.0 mmol) and triphosgene (98 mg, 0. 33 mmol). After stirring for 30 minutes, 5-methyl-2-amino pyrazine (109 mg, 1.0 mmol) was added and the reaction was heated to 65 degrees C. The reaction was allowed to cool to room temperature, then diluted with ethyl acetate (50 mL) and water (50 mL). A precipitate formed which was filtered and dried under reduced pressure to yield the desired material as a white solid (40% yield).'H-NMR (400 MHz, d6-DMSO) 8 8.62 (br s, 1H), 8.41 (d, 1H), 8.19 (s, 1H), 7.59 (d, 1H) 7. 49 (s, 1H), 4.15 (t, 2H), 3.81 (s, 3H), 2.42 (m, 5H), 2.18 (s, 6H), 2.01 (m, 2H).

Step 4: 3- (3-Dimethylamino-propoxy)-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzoic acid. To a stirred solution of 3- (3-dimethylamino-propoxy)-4- [3- (5-nethyl-pyrazin-2-yl)-ureido]-benzoic acid methyl ester (1.0 gm; 3.3 mmol) in methanol (25 mL) was added lithium hydroxide (5 mL of a 2M aqueous solution). The reaction was heated to 60 degrees and stirred for 12 hours. The reaction was allowed to cool to room temperature and the pH was adjusted to 5.5 with IN hydrochloric acid. A precipitate formed which was filtered and dried under reduced pressure to yield the desired product as a tan solid (52% yield).'H-NMR (400 MHz, d6-DMSO) 6 8. 62 (br s, 1H), 8. 39 (d, 1H), 821 (s. 1H), 7.59 (d, 1H), 7.43 (s, 1H), 4.15 (t, 2H), 2.42 (m, 5H), 2.18 (s, 6H), 2.01 (m, 2H).

Compound 23 5 : 4- [3- (5-Methyl-pyrazin-2-yl)-ureido]-3- (pyridin-3-ylmethoxy)-benzoic acid Step 1 : 4-Nitro-3- (pyridin-3-ylmethoxy)-benzoic acid methyl ester. To a stirred, cooled (about 0°C) solution of 3-hydroxy-4-nitro benzoic acid methyl ester (394 mg, 2.0 mmol), triphenyl phosphine (525 mg, 2.0 mmol), and 3-pyridylcarbinol (194 L, 2.0 mmol) in dry tertrahydrofuran (5 mL) was added diisopropyl azodicarboxylate (394 L, 2.0 mmol in 1 mL of tetrahydrofuran). After stirring for 12 hours, the reaction was diluted with hydrochloric acid (30 mL of a 1M solution) and extracted with ethyl acetate (2x 50mL). The aqueous solution was basified with 10% aqueous sodium carbonate (50 mL) and the product was extracted with ethyl acetate (3 x 100 mL). The ethyl acetate was washed with brine (1 x 30 mL), then dried (MgS04), and filtered. The filtered solution was concentrated under reduced pressure to provide the desired crude product as an off white solid (76 % yield).'H-NMR (400 MHz, CDC13) 8 8.71 (s, 1H), 8.62 (d, 1H), 7.88 (m, 3H), 7.79 (d, 1H), 7.39 (M, 1H), 5.31 (s, 2H), 3.98 (s, 3H).

Step 2: 4-Amino-3-(pyridin-3-ylmethoxy)-benzoic acid methyl ester. To a stirred, cooled (about 0°C) solution of 4-nitro-3- (pyridin-3-ylmethoxy)-benzoic acid methyl ester (288 mg, 1.0 mmol) in methanol (2 mL) and saturated aqueous ammonium chloride (ImL) was added zinc dust (131 mg, 2.0 mmol).

After stirring for 12 hours, the reaction was diluted with 30 mL of ethyl acetate and washed with 30 mL of 10% aqueous sodium carbonate (2 x 30 mL), brine (1 x 30 mL), then dried (MgS04), and filtered.

The filtered solution was concentrated under reduced pressure to yield the desired product as a yellow solid (97% yield).'H-NMR (400 MHz, CDC) 8 8. 78 (s, 1H), 8.61 (d, 1H), 7.79 (d, 1H), 7.59 (m.

2H), 7. 39 (m, 1H), 6.71 (d, 1H), 5.18 (s, 2H), 4.28 (br s, 2H), 3.85 (s, 3H).

Step 3: 4-[3-(5-Methyl-pyrazin-2-yl)-ureido]-3-(pyridin-3-ylmethoxy) -benzoic acid methyl ester. To a stirred, cooled (about 0°C) solution of 4-amino-3-(pyridin-3-ylmethoxy)-benzoic acid methyl ester (258 mg, 1.0 mmol) in toluene (3.0 mL) was added triethylamine (139 uL, 1.0 mmol) and triphosgene (98 mg, 0.33 mmol). After stirring for 30 minutes, 5-methyl-2-amino pyrazine (109 mg, 1.0 mmol) was added and the reaction was heated to 65 degrees C. The reaction was allowed to cool to room temperature, then diluted with ethyl acetate (50 mL) and water (50 mL). A precipitate formed which was filtered and dried under reduced pressure to yield the desired material as a white solid (47% yield).

'H-NMR (400 MHz, d6-DMSO) 8 10.29 (s, 1H), 8.79 (s, 1H), 8.68 (d, 1H), 8. 59 (br s, 1H), 8. 48 (d, 1H), 7.70 (s, 1H), 7.62 (d, 1H), 7.51 (m, 1H), 5.32 (s, 2H). 3.88 (s, 3H), 2.32 (s, 3H).

Step 4: 4- [3- (5-Methyl-pyrazin-2-yl)-ureido]-3- (pyridin-3-y ? methoxy)-benzoic acid. To a stirred solution of 4- [3- (5-methyl-pyrazin-2-yl)-ureido]-3- (pyridin-3-ylmethoxy)-benzoic acid methyl ester (1.3 gm; 3.3 mmol) in methanol (25 mL) was added lithium hydroxide (5 mL of a 2M aqueous solution). The reaction was heated to 60 degrees and stirred for 12 hours. The reaction was allowed to cool to room temperature and the pH was adjusted to 5.5 with 1N hydrochloric acid. A precipitate formed which was filtered and dried under reduced pressure to yield the desired product as a tan solid (90% yield). IH-NMR (400 MHz, d6-DMSO) õ 10. 29 (s, 1H), 8.79 (s, 1H), 8. 68 (d, 1H), 8.59 (br s, 1H), 8.48 (d, 1H), 7.70 (s, 1H), 7.62 (d, 1H), 7. 51 (m 1H), 5.32 (s, 2H), 2. 32 (s, 3H).

Compound 236: 3- (3-Dimethylamino-propoxy)-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzamide Prepared according to the procedure of Compound 208 using ammonia (33% yield) 'H-NMR (400 MHz, d6-DMSO) 8 10.18 (s, 1H), 8.56 (br s, 1H), 8. 20 (d, 1H), 8.11 (s. 1H), 7.43 (d, 1H), 7.40 (s, 1H), 4.03 (m, 2H), 2.40 (m, 2H), 2.33 (s, 3H), 2.05 (s, 6H), 1.91 (m, 2H) LRLCMS (esi, positive) m/e 374.2 (M+1) Compound 237 : 3- (3-Dimethylamino-propoxy)-N, N-dimethyl-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzamide Prepared according to the procedure of Compound 208 using dimethylamine (97% yield) 'H-NMR (400 MHz, CDCl3) 8 11. 30 (br s, 1H), 8.39 (d, 1H), 8. 29 (s. 1H), 8.20 (s, 1H), 7.78 (br s, 1H), 7.07 (s, 1H), 7.04 (d, 1H), 4.16 (t, 2H), 2.55 (s, 3H), 2.53 (m, 2H), 2.26 (s, 6H), 2.10 (m, 2. H) LRMS (esi, positive) m/e 401.1 (M+1) Compound 238: N-Benzyl-3-(3-dimethylamino-propoxy)-4-[3-(5-methyl-pyrazin- 2-yl)-ureido]-benzamide Prepared according to the procedure of Compound 208 using benzylamine (65% yield) 'H-NMR (400 MHz, CDCl3) 8 8.43 (d, 1H), 8.26 (s, 1H), 8.20 (s, 1H), 7.91 (br s, 1H), 7.56 (s, 1H), 7.38 (m, 4H), 7.31 (m, 1H), 6.41 (m, 1H), 4.64 (d, 2H), 4.20 (t, 2H), 2.53 (s, 3H), 2.52 (m, 2H), 2.24 (s, 6H), 2.14 (m, 2H) LRMS (esi, positive) m/e 463.2 (M+1) Compound239: N-Benzyl-3- (3-dimethylamino-propoxy)-N-methyl-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzanide Prepared according to the procedure of Compound 208 using N-methyl benzylamine (68% yield).

'H-NMR (400 MHz, CDC13) 8 8. 37 (br s, iH), 8.25 (s, 1H), 8.18 (s, 1H), 7.37 (m, 4H), 7.31 (m, 1H), 7.18 (m, 2H), 4.67 (br m, 2H), 4.07 (br m, 2H), 2.99 (br s, 3H), 2.53 (s, 3H), 2.50 (br m, 2H), 2.24 (s, 6H), 2.06 (br m, 2H) LRMS (esi, positive) m/e 477.2 (M+1) Compound 240: 3- (3-Dimethylamino-propoxy)-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-N- (2-morpholin-4-yl-ethyl)- benzamide Prepared according to the procedure of Compound 208 using 2-morpholin-4-yl-ethylaamine (69% yield).

'H-NMR (400 MHz. CDC13) 8 8. 44 (d, 1H), 8. 31 (s, 1H), 8.21 (s, 1H), 7.90 (br s, 1H), 7.55 (s, 1H), 7.24 (d, 1H), 6.76 (m, 1H), 4.20 (t, 2H), 3.76 (m, 4H), 3.56 (m, 2H), 2.61 (m, 2H), 2.53 (m, 7H), 2.24 (s, 6H), 2.13 (m, 2H) LRMS (esi, positive) m/e 486.2 (M+1) Compound 241: 3-(3-Dimethylamino-propoxy)-4-[3-(5-methyl-pyrazin-2-yl)-ure ido]-N-[2-(1-methyl-pyrrolidin-2-yl)-e thyl]-benzamide Prepared according to the procedure of Compound 208 using 2- (l-methyl-pyrrolidin-2-ylamine (68% yield).

'H-NMR (400 MHz, CDC13) 8 1 i. 35 (br s, 1H), 8.42 (s, 1H), 8.40 (s, 1H), 8.24 (s, 1H), 8.20 (s, 1H), 7.57 (s, 1H), 7.46 (br s, 1H), 7.20 (m, 1H), 4.19 (m, 2H), 3.76 (m, 1H), 3.44 (m, 1H), 3.15 (m, 1H), 2.54 (s, 3H), 2.42 (m, 1H), 2.37 (s, 3H), 2.25 (s, 6H), 2.22 (m, 1H), 2.13 (m, 2H), 1. 90 (m, 2H), 1.77 (m, 2H) LRMS (esi, positive) m/e 484.3 (M+1) Compound 242: N- (2-Dimethylamino-ethyl)-3- (3-dimethylamino-propoxy)-4- [3- (5-methyl-pyrazin-2-yl)-ureido]- benzamide Prepared according to the procedure of Compound 208 using 2-dimethylamino-ethylamine (37% yield).

'H-NMR (400 MHz, CDC13) 8 11.38 (br s, 1H), 8.42 (d, 1H), 8. 30 (s, 1H), 8. 20 (s, 1H), 7.96 (br s, 1H), 7.53 (s, 1H), 7.27 (d, 1H), 6.79 (m, 1H), 4.20 (m, 2H), 3.53 (m, 2H), 2.52 (m, 2H), 2.52 (s, 3H), 2.26 (m, 2H), 2.24 (s, 6H), 2.22 (s, 6H), 2.12 (m, 2H) LRMS (esi, positive) m/e 444.2 (M+1) Compound 243 : N- (3-S-1-Benzyl-pyrrolidin-3-yl)-3- (3-dimethylamino-propoxy)-4- [3-5-methyl-pyrazin-2-yl)-ureido]- benzamide Prepared according to the procedure of Compound 215 using 1-benzyl-pyrrolidin-3-S-ylamine (20% yield) 'H-NMR (400 MHz, CDC13) 5 8.61 (br s, 1H), 8.45 (d, 1H), 8. 20 (s, 1H), 8.16 (m, 1H), 7, 51 (s, 1H), 7.39 (d, 1H), 7. 31 (m, 4H), 7.16 (m, 1H), 4. 72 (m, 1H), 4 18 (m, 2H), 3.72 (dd, 2H), 3.04 (m, 1H), 2.96 (m, 1H), 2.69 (m, 1H), 2.53 (m, 2H), 2.51 (s, 3H), 2.40 (m, 2H), 2.25 (s, 6H), 2. 11 (m, 2H), 1.76 (m, 1H) LRMS (esi, positive) m/e 532.2 (M+1) Compound244: 4- [3- (5-Methyl-pyrazin-2-yl)-ureido]-3- (pyridin-3-ylmethoxy)-benzamide Prepared according to the procedure of Compound 208 using ammonia (99% yield).

'H-NMR (400 MHz, d6-DMSO) 8 10.27 (s, 1H), 8.82 (s, 1H), 8.66 (d, 1H), 8.60 (br s, 1H), 8. 29 (d, 1H), 8.03 (m, 1H), 8.00 (s, 1H), 7.76 (s, 1H), 7.56 (d, 1H), 7.50 (m, 1H), 7. 37 (br m, 1H), 7.26 (br s, 1H), 5.32 (s, 2H), 2.32 (s, 3H) LRMS (apci, positive) m/e 379.1 (M+i) Compound245: N-Methyl-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-3- (pyridin-3-ylmethoxy)-benzamide Prepared according to the procedure of Compound 208 using methylamine (99% yield).

'H-NMR (400 MHz, d6-DMSO) 8 10.25 (s, 1H), 8.80 (s, 1H), 8.60 (m, 3H), 8. 26 (d, 1H), 8.03 (d, 1H), 7.78 (s, 1H), 7.52 (m, 2H), 7.37 (m, 1H), 5.34 (s, 2H), 2.79 (d, 3H), 2. 36 (s, 3H) LRMS (apci, positive) m/e 393.2 (M+1) Compound 246: N,N-Dimethyl-4-[3-(5-methyl-pyrazin-2-yl)-ureido]-3-(pyridin -3-ylmethoxy)-benzamide Prepared according to the procedure of Compound 208 using dimethylamine (88% yield).

'H-NMR (400 MHz, d6-DMSO) 8 10.16 (s, 1H), 8.80 (s, 1H), 8.66 (d, 1H), 8.58 (s, 1H), 8.28 (d, 1H), 7.96 (d, 1H), 7.50 (m, 1H), 7. 37 (br s, 1H), 7.25 (s, 1H), 7.03 (d, 1H), 5.30 (s, 2H), 2.95 (s, 6H), 2.33 (s, 3H) LRMS (apci, positive) m/e 407.4 (M+1) Compound 247: N-Benzyl-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-3- (pyridin-3-ylmethoxy)-benzamide Prepared according to the procedure of Compound 208 using benzylamine (41% yield).

'H-NMR (400 MHz, d6-DMSO) 8 10. 23 (s, 1H), 9.05 (t, 1H), 8.82 (s, 1H), 8.67 (d, 1H), 8. 58 (br s, 1H), 8. 33 (d, 1H), 8.00 (d, 1H), 7.78 (s, 1H), 7.59 (d, 1H), 7.50 (m, 1H), 7.33 (m, 4H), 7.25 (m, 1H), 5.32 (s, 2H), 4.50 (d, 2H), 2.32 (s, 3H) LRMS (apci, positive) m/e 469.1 (M+1) Compound 248: N-Benzyl-N-methyl-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-3- (pyridin-3-ylmethoxy)-benzamide Prepared according to the procedure of Compound 208 using N-methyl benzylamine (71% yield).

'H-NMR (400 MHz, d6-DMSO) 8 10. 16 (s, 1H), 8.76 (br s, 1H), 8. 64 (d, 1H), 8.57 (m, 1H), 8.28 (br m, 1H), 7.95 (br m, 1H), 7.48 (m, 1H), 7.40-7.23 (br m, 7H), 7.06 (m, 1 H), 5.23 (br m, 2H), 4.62 (br ni, 2H), 2. 88 (s, 3H), 2.33 (s, 3H) LRMS (apci, positive) m/e 483. 3 (M+l) Compound249: 4-[3-(5-Methyl-pyrazin-2-yl)-ureido]-N-(2-morpholin-4-yl-eth yl)-3-(pyridin-3-ylmethoxy)-benzamide Prepared according to the procedure of Compound 208 using 2-morpholin-4-yl-ethylamine (99% yield).

'H-NMR (400 MHz, d6-DMSO) 8 10.27 (s, 1H), 8.81 (s, 1H), 8.66 (d, 1H), 8.61 (br s, 1H), 8.52 (t, 1H), 8.29 (d, 1H), 8.03 (d, 1H), 7.76 (s, 1H), 7.52 (m, 2H), 7.36 (m, 1H), 5. 33 (s, 2H), 3.56 (m, 4H), 3.40 (m, 2H), 2.48 (m, 2H), 2.43 (m, 4H), 2.34 (s, 3H) LRMS (apci, positive) m/e 492.4 (M+l) Compound250: 4- [3- (5-Methyl-pyrazin-2-yl)-ureido]-N- [2- (I-methyl-pyrrolidin-2-yl)-ethyl]-<BR> 3- (pyridin-3-ylmethoxy)-benzamide Prepared according to the procedure of Compound 208 using 2- (l-methyl-pyrrolidin-2-yl)-ethylamine (99% yield).

H-NMR (400 MHz, d6-DMSO) 5 10. 21 (s, 1H), 8.81 (s, 1H), 8.64 (d, 1H), 8.58 (br s, 1H), 8.49 (m, 1H), 8.33 (d, 1H), 8.00 (d, 1H), 7.70 (s, 1H), 7.52 (m, 2H), 7.34 (m, 1H), 5.33 (s, 2H), 3.28 (m, 2H), 2.95 (m, 1H), 2.36 (s, 3H), 2.21 (s, 3H), 2.04 (m, 2H), 1.90 (m, 2H), 1.62 (m, 2H), 1.44 (m, 2H) LRMS (apci, positive) m/e 490.3 (M+l) Compound 251 : N-(2-Dimethylamino-ethyl)-4-[3-(5-methyl-pyrazin-2-yl)-ureid o]-3-(pyridin-3-ylmethoxy)-benzamide Prepared according to the procedure of Compound 208 using 2-dimethylamino-ethylamine (99% yield).

'H-NMR (400 MHz, d6-DMSO) 8 10.24 (s, 1H), 8.82 (s, 1H), 8.66 (d, 1H), 8.60 (br s, 1H), 8.43 (t, 1H), 8. 30 (d, 1H), 8. 02 (d, 1H), 7.74 (s, 1H), 7.52 (m, 2H), 7.35 (br m, 1H), 5.32 (s, 2H), 3.36 (m, 2H), 2.39 (m, 2H), 2.33 (s, 3H), 2.18 (s, 6H) LRMS (apci, positive) m/e 450.2 (M+l) Compound 252 : N- (3-S-1-Benzyl-pyrrolidin-3-yl)-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-3- (pyridin-3-ylmethoxy)- benzamide Prepared according to the procedure of Compound 215 using 1-benzyl-pyrrolidin-3-S-ylamine (99% yield).

H-NMR (400 MHz, d6-DMSO) 8 10.26 (s, 1H), 8.82 (s, 1H), 8. 66 (d, 1H), 8. 60 (br s, 1H), 8.47 (d, 1H), 8.28 (d, 1H), 8. 02 (d, 1H), 7.74 (s, 1H), 7.55 (d, 1H), 7.51 (m, 1H), 7.32 (m, 4H), 7.24 (m, 1H), 5.31 (s, 2H), 4.40 (m, 1H), 3.60 (s, 2H), 2.80 (m, 1H), 2.64 (m, 1H), 2.45 (m, 2H), 2. 34 (s, 3H), 2.15 (m, 1H), 1.85 (m, 1H) LRMS (apci, positive) m/e 538.2 (M+l) Compound 253: 1- [2- (3-Dimethylamino-propoxy)-5-methyl-phenyl]-3-pyrazin-2-yl-ur ea Step 1 : (2-Hydroxy-5-methyl-phenyl)-carbamic acid tert-butyl ester. To a stirred solution of 2-amino- 4-methyl-phenol (6.15 gm; 50 mmol) in dioxane (100 mL) was added carbonic acid di-tert-butyl ester (9.8 gm; 45 mmol) followed by sodium bicarbonate (12.6 gm; 150 mmol in 75 mL of H-, O) After stirring for 8 hours, the reaction was diluted with 100 mL. of ethyl acetate and washed with 1N aqueous hydrochloric acid (2 x 100 mL), saturated aqueous sodium bicarbonate (1 x 100 mL), and brine (100 mL), then dried (MgS04), and filtered. The filtered solution was concentrated under reduced pressure to provide the desired (2-Hydroxy-5-methyl-phenyl)-carbamic acid tert-butyl ester as a brown solid (95% yield).

Step 2: [2- (3-Dimethylamino-propoxy)-5-methyl-phenyl]-carbamic acid tert-butyl ester. To a stirred, cooled (about 0°C) solution of (2-hydroxy-5-methyl-phenyl)-carbamic acid tert-butyl ester (447mg, 2.0 mmol), triphenyl phosphine (525 mg, 2.0 mmol), and 3-(dimethylamino)-1-propanol (237 1L, 2.0 mmol) in dry tetrahydrofuran (5 mL) was added diisopropyl azodicarboxylate (394 uL, 2.0 mmol) in 1 mL of tetrahydrofuran). After stirring for 12 hours, the reaction was diluted with 30 mL of ethyl acetate and washed with 30 mL of 10% aqueous sodium carbonate (2 x 30 mL), brine (1 x 30 mI.), then dried (MgS04), and filtered. The filtered solution was concentrated under reduced pressure to provide the desired crude product. Step 3: 2- (3-Dimethylamino-propoxy)-5-methyl-phenylamine. To a stirred solution of [2- (3- dimethylamino-propoxy)-5-methyl-phenyl]-carbamic acid tert-butyl ester (617 mg, 2.0 mmol) in 5 mL of dioxane was added hydrochloric acid (2 mL; 4N in dioxane). After stirring for 12 hours, the reaction was diluted with 20 mL of 1N hydrochloric acid and washed with ethyl acetate (2 x 30 mL). The aqueous layer was basified with 10% aqueous sodium carbonate (50 mL) and extracted with ethyl acetate (3 x 50 mL). The ethyl acetate was washed with brine (1 x 30 mL), then dried (MgS04), and filtered. The filtered solution was concentrated under reduced to yield the corresponding aniline.

Step 4: 1- [2- (3-Dimethylamino-propoxy)-5-methyl-phenyl]-3-pyrazin-2-yl-ur ea. To a stirred, cooled (about 0°C) solution of 2- (3-dimethylamino-propoxy)-5-methyl-phenylamine (208 mg, 1.0 mmol) in toluene (3.0 mL) was added triethylamine (140 tL, 1.0 mmol) and triphosgene (98 mg, 0.33 mmol).

After stirring for 30 minutes, amino pyrazine (95 mg, 1.0 mmol) was added and the reaction was heated to 65 degrees C. After stirring for 4 hours, the reaction was cooled to room temperature and stirred for 8 hours. The precipitate that formed was filtered, rinsed with toluene (2 x 1 mL), and dried under reduced pressure (35% yield).

'H-NMR (400 MHz, CDC13) 8 8.45 (s, 1H), 8.39 (br s, 1H), 8.22 (s, 1H), 8.21 (d, 1H), 8.19 (d, 1H), 4.09 (t, 2H), 2.55 (t, 2H), 2. 36 (s, 3H), 2.26 (s, 6H), 2.05 (m, 2H).

LRMS (esi, positive) m/e 330.10 (M+1) Compound 254: 1-[2-(2-Dimethylamino-ethoxy)-5-methyl-phenyl]-3-pyrazin-2-y l-urea Prepared according to the method of Compound 253 using N, N-dimethyl ethanolamine (36% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.79 (br s, 1H), 8.25 (s, 1H), 8.22 (s, 1H), 8.05 (s, 1H), 6.95 (d, 1H), 6.80 (d, 1H), 4.15 (t, 2H), 2.55 (s, 3H), 2.31 (t, 2H), 2.26 (s, 6H).

LRMS (esi, positive) m/e 316.21 (M+ 1) Compound 255 : 1- [5-Methyl-2- (pyridin-3-ylmethoxy)-phenyl]-3-pyrazin-2-yl-urea Prepared according to the method of Compound 253 using 3-hydroxymethyl pyridine (10% yield).

'H-NMR (400 MHz, CDC) 8 8.82 (s, 1H), 8.65 (d, 1H), 8.35 (s, 1H), 8.30 (s, 1H), 8.21 (s, 2H), 8.05 (s, 1H), 7.81 (m, 2H), 7.35 (m, 1H), 6.9 (dd, 2H), 5.15 (s, 2H), 2.39 (s, 3H).

LRMS (esi, positive) m/e 336.21 (M+1) Compound 256: 1- {5-Methyl-2- [3- (2-oxo-pyrrolidin-1-yl)-propoxy]-phenyll-3-pyrazin--2-yl-ure a Prepared according to the method of Compound 253 using 3- (2-oxo-pyrrolidin-1-yl)-propanol (10% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.79 (s, 1H), 8. 29 (s, 1H), 8.25 (s, 1H), 8. 05 (s, 1H), 6.92 (d, 1H), 6.79 (d, 1H), 3.99 (t, 2H), 3.38 (m, 4H), 2.22 (s, 3H), 2.20 (t, 2H), 2.00 (t, 2H), 1.91 (t, 2H).

LRMS (esi, positive) m/e 392.2 (M+Na) Compound 257: 1-[5-Methyl-2-(2-morpholin-4-yl-ethoxy)-phenyl]-3-pyrazin-2- yl-urea Prepared according to the method of Compound 253 using 2-morpholin-4-yl-ethanol (39% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.79 (s, 1H), 8.29 (d, 1H), 8.25 (d, 1H), 8.05 (s, 1H), 6.95 (d, 1H), 6.79 (d, 1H), 4.19 (t, 2H), 3.59 (m, 4H), 2. 80 (t, 2H), 2.49 (m, 4H), 2.22 (s, 3H).

LRMS (esi, positive) m/e 358.2 (M b1) Compound258: 1- [5-Methyl-2- (3-morpholin-4-yl-propoxy)-phenyl.]-3- (5-methyl-pyrazin-2-yl)-urea Prepared according to the method of Compound 253 using 3-morpholin4-yl-propanol (8% yield).

'H-NMR (400 MHz, d6-DMSO) 8 9.01 (s, 1H), 8.62 (br s, 1H), 8.22 (s, 1H), 8.19 (s, 1H), 8.05 (s, 1H), 6.91 (d, 1H), 6.79 (d, 1H), 4.05 (t, 2H), 3.59 (m, 4H), 2.48 (s, 3H), 2.45 (t, 2H), 2.35 (m, 4H), 2.21 (s, 3H), 2.00 (t, 2H).

LRMS (esi, positive) m/e 386.31 (M+l) Compound259: 1- [2- (3-Dimethylamino-propoxy)-5-methyl-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Prepared according to the method of Compound 253 3-dimethylamino-propanol (40% yield).

'H-NMR (400 MHz, CDCl3) 8 8.37 (s, 1H), 8.19 (s, 1H), 8. 18 (s, 1H), 8.09 (br s, 1H), 6.80 (dd, 2H), 4.05 (t, 2H), 2.55 (t, 2H), 2.54 (s, 3H), 2. 36 (s, 3H), 2.26 (s, 6H), 2.05 (m, 2H).

LRMS (esi, positive) m/e 344.20 (M+1) Compound 260: 1- [5-Methyl-2- (2-morpholin-4-yl-ethoxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Prepared according to the method of Compound 253 using 2-morpholin-4-yl-ethanol (10% yield). 'H-NMR (400 MHz, CDC13) 8 10.79 (br s, 1H), 8.82 (s, 1H), 8.59 (s, 1H), 8.19 (s, 1H), 8.05 (s, 1H), 6.81 (dd, 2H), 4.20 (t, 2H), 3.75 (m, 4H), 2.91 (t, 2H), 2.61 (m, 4H), 2.55 (s, 3H), 2. 35 (s, 3H).

LRMS (esi, positive) m/e 372.1 (M+1) Compound261: 1- (5-Methyl-pyrazin-2-yl)-3- [5-methyl-2- (pyridin-2-ylmethoxy)-phenyl]-urea Prepared according to the method of Compound 253 using 2-hydroxymethyl pyridine (21% yield).

'H-NMR (400 MHz, CDC13) 8 8.61 (d, 1H), 8.29 (s, 1H), 8.11 (s, 1H), 7.61 (t, 1H), 7.31 (d, 1H), 7.18 (t, 1H), 6.88 (d, 1H), 6.75 (d, 1H), 5.18 (s, 2H), 2. 30 (s, 3H).

LRMS (esi, positive) m/e 372.2 (M+Na) Compound 262: 1-(5-Methyl-pyrazin-2-yl)-3-[5-methyl-2-(pyridin-4-ylmethoxy )-phenyl]-urea Prepared according to the method of Compound 253 using 4-hydroxymethyl pyridine (18% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.84 (s, 1H), 8.55 (d, 2H), 7.91 (s, 1H), 7.47 (d, 2H), 6.88 (d, 1H), 6.72 (d, 1H), 5.28 (s, 2H), 2.22 (s, 3H).

LRMS (esi, positive) m/e 350.21 (M+1) Compound263: 1- (5-Methyl-pyrazin-2-yl)-3- [5-methyl-2- (pyridin-3-ylmethoxy)-phenyl]-urea Prepared according to the method of Compound 253 using 3-hydroxymethyl pyridine (10% yield).

'H-NMR (400 MHz, CDC13) 8 8.82 (s, 1H), 8.68 (m, 2H), 8.25 (s, 1H), 8. 20 (s, 1H), 7.84 (d, 1H), 7.38 (m, 1H), 6.99 (s, 1H), 6.88 (d, 1H), 6.80 (d, 1H), 5.10 (s, 2H), 2. 38 (s, 3H), 2.35 (s, 3H).

LRMS (esi, positive) m/e 350.21 (M+1) Compound 264: 1- [2- (2-Dimethylamino-ethoxy)-5-methyl-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Prepared according to the method of Compound 253 using N, N-dimethyl ethanolamine (11% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.69 (s, 1H), 8. 18 (s, 1H), 8. 03 (s, 1H), 6.95 (d, 1H), 6.79 (d, 1H), 4.11 (t, 2H), 2.72 (t, 2H), 2.43 (s, 3H), 2.25 (s, 3H), 2. 22 (s, 6H).

LRMS (esi, positive) m/e 330.20 (M+1) Compound 265: 1- (5-Methyl-pyrazin-2-yl)-3- [2- (pyridin-3-ylmethoxy)-5-trifluoromethyl-phenyl]-urea Prepared according to the method of Compound 253 3-hydroxymethyl pyridine and 2-hydroxy-5- trifluoromethyl aniline (40% yield).

'H-NMR (400 MHz, d6-DMSO) 8 8.81 (s, 1H), 8.69 (s, 1H), 8.65 (s, 1H), 8.59 (br s, 1H), 8.01 (d, 1H), 7.45 (t, 1H), 7.3 (br s, 1H), 5.39 (s, 2H), 2.35 (s, 3H).

LRMS (esi, positive) m/e 404.10 (M+1) Compound266: 1- [5-Methyl-2- (6-methyl-pyridin-3-ylmethoxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Step 1: (6-Methyl-pyridin-3-yl)-methanol. To a stirred, cooled (0 °C) solution of 6-methyl nicotinic acid (5.0 mmol) in tetrahydrofuran (10 mL) was added lithium aluminum hydride (20 mmol ; 20 mL of a 1M solution in tetrahydrofuran.) dropwise. The reaction was stirred for 4 hours, treated sequentially with 1 mL of H2O, 1 mL of 15% aqueous sodium hydroxide, and 3 mL of H2O. The reaction was filtered and washed with tetrahydrofuran (3 x 50 mL). The filtrate was concentrated under reduced pressure to yield the alcohol as a clear viscous oil.

Steps 2-3: Mitsunobu reaction and aniline deprotection according to the method of Compound 253.

Step 4: To a stirred solution of 5-methyl-pyrazine-2-carboxylic acid (138 mg, 1.0 mmol) in toluene (3.0 mL) was added diphenylphosphoryl azide (216 1L, 1.0 mmol) and triethylamine (140 tL, 1.0 mmol). The reaction was placed under nitrogen and heated to 90 degrees C for 15 minutes. The temperature was reduced to 65 °C and 5-methyl-2- (6-methyl-pyridin-3-ylmethoxy)-phenylamine (228 mg, 1.0 mmol) was added. The reaction was stirred at that temperature for 4 hours and then stirred at room temperature for 8 hours. The precipitate that formed during the reaction was filtered, rinsed with toluene (2 x 1 mL), and dried under reduced pressure (36% yield).

'H-NMR (400 MHz, CDC13) 8 11.45 (br s, 1H), 8.65 (s, 1H), 8.29 (s, 1H), 8. 22 (s, 1H), 8.20 (s, 1H), 7.70 (d, 1H), 7.19 (d, 1H), 6.9 (m, 3H), 5.05 (s, 2H), 2.65 (s, 3H), 2.4 (s, 3H), 2.35 (s, 3H).

LRMS (esi, positive) m/e 364.16 (M+1) Compound 267: 1- (5-Methyl-pyrazin-2-yl)-3- [5-methyl-2- (2-pyridin-2-yl-ethoxy)-phenyl]-urea Steps 1-2: Mitsunobu reaction using 2- (2-pyridyl)-ethanol and aniline deprotection according to the method of Compound 253.

Step 3: Urea formation according to the method of Compound 266 using 5-methyl-2- (2-pyridin-2-yl- ethoxy)-phenylamine (37% yield).

'H-NMR (400 MHz, CDC13) 8 10.70 (br s, 1H), 8.65 (d, 1H), 8.45 (br s, 1H), 8.21 (s, 1H), 7.85 (s, 1H), 7.59 (t, 1H), 7.29 (t, 1H), 6.80 (dd, 2H), 4.49 (t, 2H), 3. 39 (t, 2H), 2.49 (s, 3H), 2.39 (s, 3H).

LRMS (esi, positive) m/e 364.14 (M+1) Compound 268: 1- (5-Methyl-pyrazin-2-yl)-3- [5-methyl-2- (3-pyridin-2-yl-propoxy)-phenyl]-urea Steps 1-2: Mitsunobu reaction using 3- (2-pyridyl)-propanol and aniline deprotection according to the method of Compound 253.

Step 3: Urea formation according to the method of Compound 266 using 5-methyl-2- (3-pyridin-2-yl- propoxy)-phenylamine (5% yield).

'H-NMR (400 MHz, CDCI3) 8 10.89 (br s, 1H), 8.59 (d, 1H), 8.49 (s, 1H), 8.45 (br s, 1H), 8.22 (s, 1H), 8.15 (s, 1H), 7.59 (t, 1H), 7.15 (d, 2H), 6.88 (dd, 2H), 4.05 (t, 2H), 3.10 (t, 2H), 2.45 (s, 3H), 2.40 (t, 2H), 2. 35 (s, 3H).

LRMS (esi, positive) m/e 378.10 (M+1) Compound 269: 1- (5-Methyl-pyrazin-2-yl)-3- [5-methyl-2- (3-pyridin-4-yl-propoxy)-phenyl]-urea Steps 1-2: Mitsunobu reaction using 3- (4-pyridyl)-propanol and aniline deprotection according to the method of Compound 253.

Step 3: Urea formation according to the method of Compound 266 using 5-methyl-2- (4-pyridin-2-yl- propoxy)-phenylamine (28% yield).

'H-NMR (400 MHz, CDC13) 5 11.15 (br s, 1H), 8.51 (d, 2H), 8.25 (s, 1H), 8.20 (s, 1H), 7.79 (s, 1H), 7.75 (s, 1H), 7.15 (d, 2H), 6.82 (d, 1H), 6.75 (d, 1H), 4.05 (t, 2H), 2.91 (t, 2H), 2.40 (s, 3H), 2.36 (s, 3H), 2.25 (m, 2H).

LRMS (esi, positive) m/e 378.16 (M+i) Compound 270: 1- {2- [2- (Benzyl-methyl-amino)-ethoxy]-5-methyl-phenyl}-3- (5-methyl-pyrazin-2-yl)-urea Steps 1-2: Mitsunobu reaction using N-methyl-N-benzyl ethanolamine and aniline deprotection according to the method of Compound 253.

Step 3: Urea formation according to the method of Compound 266 using 2-[2-(benzyl-methyl-amino)- ethoxy]-5-methyl-phenylamine (17% yield).

'H-NMR (400 MHz, CDC13) 8 10.70 (br s, 1H), 8.49 (s, 1H), 8.20 (s, 1H), 7.95 (s, 1H), 7.85 (s, 1H), 7.32 (m, 5H), 6.85 (s, 2H), 4.15 (t, 2H), 3.62 (s, 2H), 2.92 (t, 2H), 2.49 (s, 3H), 2. 33 (s, 3H), 2.29 (s, 3H).

LRMS (esi, positive) m/e 406.01 (M+1) Compound 271: 1- [5-Methyl-2- ( 1-methyl-piperidin-3-ylmethoxy)-phenyl]-3- (5-methyl-pyrazin-2-yi)-urea Steps 1-2: Mitsunobu reaction using 3-hydroxymethyl-1-methyl piperidine and aniline deprotection according to the method of Compound 253.

Step 3: Urea formation according to the method of Compound 266 using 5-methyl-2- (1-methyl- piperidin-3-ylmethoxy)-phenylamine (16% yield).

'H-NMR (400 MHz, CDC13) 8 11.25 (br s, 1H), 8.45 (br s, 1H), 8.35 (s, IH), 8.22 (s, 2H), 6.80 (d, 1H), 6.74 (d, 1H), 3.80 (m, 2H), 3.15 (br d, 1H), 2.80 (br d, 1H), 2.51 (s, 3H), 2. 35 (s, 3H), 2.30 (m, 1H), 2 22 (s, 3H), 1.50-2.00 (m, 6H), 1.00-1.25 (m, 2H).

LRMS (esi, positive) m/e 370.01 (M+1) Compound 272: 1- {2- [2- (4-Dimethylamino-phenyl)-ethoxy]-5-methyl-phenyl}-3- (5-methyl-pyrazin-2-yl)-urea Steps 1-2: Mitsunobu reaction using 2- (4-dimethylamino-phenyl)-ethanol and aniline deprotection according to the method of Compound 253.

Step 3: Urea formation according to the method of Compound 266 using 2- [2- (4-dimethylamino- phenyl)-ethoxy]-5-methyl-phenylamine (10% yield).

'H-NMR (400 MHz, CDCI3) õ 11.15 (br s, 1H), 8. 35 (s, 1H), 8.25 (s, 1H), 7.80 (m, 2H), 7.25 (m, 2H), 6.82 (s, 2H), 6.75 (d, 2H), 4.25 (t, 2H), 3.20 (t, 2H), 2.99 (s, 6H), 2.55 (s, 3H), 2.41 (s, 3H).

LRMS (esi, positive) m/e 405.90 (M+1) Compound 273: 1- (5-Methyl-pyrazin-2-yl)-3- [5-methyl-2- (3-pyridin-3-yl-propoxy)-phenyl]-urea Steps 1-2: Mitsunobu reaction using 3- (3-pyridyl)-propanol and aniline deprotection according to the method of Compound 253.

Step 3: Urea formation according to the method of Compound 266 using 5-methyl-2- (3-pyridin-3-yl- propoxy)-phenylamine (16% yield).

'H-NMR (400 MHz, CDC13) 8 10.95 (br s, 1H), 8.51 (m, 2H), 8.35 (s, 1H), 8.25 (s, 1H), 8.15 (s, 1H), 7.80 (s, 1H), 7.50 (d, 1H), 7.21 (t, 1H), 6.79 (d, 1H), 6.75 (d, 1H), 4.09 (t, 2H), 2.90 (t, 2H), 2.45 (s, 3H), 2.35 (s, 3H), 2.25 (m, 2H).

LRMS (esi, positive) m/e 377.91 (M+1) Compound 274 : 1- [2- (2-Dimethylamino-1-dimethylaminomethyl-ethoxy)-5-methyl-phen yl]-3- (5-methyl-pyrazm-2-yl)- urea Steps 1-2: Mitsunobu reaction using 1, 3-bis-dimethylamino-propan-2-ol and aniline deprotection according to the method of Compound 253.

Step 3: Urea formation according to the method of Compound 266 using 2-(2-amino-4-methyl- phenoxy)-N, N, N', N'-tetramethyl-propane-1, 3-diamine (4% yield).

'H-NMR (400 MHz, CDC13) S 9.69 (br s, 1H), 8.95 (br s, 1H), 8.19 (s, 1H), 8.05 (s, 1H), 6.80 (dd, 2H), 4.19 (m, 1H), 4.09 (m, 1H), 3.05 (m, 1H), 2.65 (m, 2H), 2.50 (s, 3H), 2.45 (s, 6H), 2. 38 (s, 6H), 2.35 (s, 3H).

LRMS (esi, positive) m/e 386.92 (M+1) Compound 275: 1- [5-Methyl-2- (2-S-1-methyl-pyrrolidin-2-ylmethoxy)-phenylj-3- (5-methyl-pyrazin-2-yl)-urea Steps 1-2: Mitsunobu reaction using 1-methyl-pyrrolidin-2-S-ylmethanol and aniline deprotection according to the method of Compound 253.

Step 3: Urea formation according to the method of Compound 266 using 5-methyl-2- (1-methyl- pyrrolidin-2-S-ylmethoxy)-phenylamine (12% yield).

'H-NMR (400 MHz, d6-DMSO) 6 10.10 (s, IH), 9.85 (br s, 1H), 9.65 (brs, 1H), 8. 78 (s, 1H), 8.25 (s, 1H), 6.90 (s, 1H), 7.02 (d, 1H), 6.85 (d, 1H), 4.33 (br s, 2H), 3.88 (m, 1H), 3.59 (m, 1H), 3.19 (m, lH), 2.99 (d, 2H) 2.70-2.85 (m, 2H), 2.45 (s, 3H), 2.29 (s, 3H), 1.80-2.10 (m, 3H).

LRMS (esi, positive) m/e 355 91 (M+1) Compound 276: 1-[2-(2-S-1-Benzyl-pyrrolidin-2-ylmethoxy)-5-methyl-phenyl]- 3-(5-methyl-pyrain-2-yl)-urea Steps 1-2: Mitsunobu reaction using 1-benzyl-pyrrolidin-2-S-ylmethanol and aniline deprotection according to the method of Compound 253.

Step 3: Urea formation according to the method of Compound 266 using 5-methyl-2-(1-benzyl- pyrrolidin-2-S-ylmethoxy)-phenylamine (3% yield).

'H-NMR (400 MHz, d6-DMSO) 8 9.95 (s, 1H), 9.90 (br s, 1H), 9.59 (br s, 1H), 8.69 (s, 1H), 8.10 (s, 1H), 7.89 (s, 1H), 7.25-7.50 (m, 6H), 6.96 (d, 1H), 6.90 (d, 1H), 4.75 (d, 2H), 4.33 (m, 4H), 4.10 (m, 2H), 2.40 (s, 3H), 2.25 (s, 3H), 1.80-2.10 (m, 3H), 1.10-1.30 (m, 2H).

LRMS (esi, positive) m/e 432. 31 (M+l) Compound 277: 1- (5-Methyl-pyrazin-2-yl)-3- [5-methyl-2- (2-pyrrolidin-l.. yl-ethoxy)-phenyl]-urea Prepared according to the procedure for compound 266, using 2-pyrrolidin-1-yl-ethanol. (28% yield).

'H NMR (400 MHz, d6-DMSO) 8 10.01 (s, 1H), 9.85 (br s, 1H), 9.72. (br s, 1H), 8. 75 (s, 1H), 8.25 (s, 1H), 8.01 (s, 1H), 7.01 (d, 1H), 6.80 (d, 1H), 4.40 (t, 2H), 3.62 (m, 4H), 3.21 (m, 2H), 2.40 (s, 3H), 2.25 (s, 3H), 2.00 (m, 2H), 1.88 (m, 2H).

LRMS (ESI, Positive) m/e 356.2 (M+1).

Compound 278: 1- {5-Methyl-2-[2-(1-methyl-pyrrolidin-2-yl)-ethoxy]-phenyl}-3- (5-methyl-pyrazin-2-yl)-urea Prepared according to the procedure for compound 266, using 2- (l-methyl-pyrrolidin-2-yl)-ethanol.

(32% yield).'H NMR (400 MHz, d6-DMSO) 5 10.01 (s, 1H), 9.85 (br s, 1H), 9.72 (br s, 1H), 8.75 (s, 1H), 8. 25 (s, 1H), 8.01 (s, 1H), 7.01 (d, 1H), 6. 80 (d, 1H), 4.20 (m, 3H), 3.00-4.00 (m, 11H), 2.80 (d, 2H), 2.40 (s, 3H), 2.25 (s, 3H).

LRMS (ESI, Positive) m/e 370.2 (M+1).

Compound 279: 1-[2-(3H-Imidazol-4-ylmethoxy)-5-methyl-phenyl]-3-(5-methyl- pyrazin-2-yl)-urea Prepared according to the procedure for compound 266, using (3H-imidazol-4-yl)-methanol. (24% yield).'H NMR (400 MHz, d6-DMSO) 8 8.51 (br s, 1H), 8.01 (s, 1H), 7.69 (s, 1H), 7.20-7.50 (m, 2H), 7.10 (d, 1H), 6. 75 (d, 1H), 4.99 (s, 2H), 2.40 (s, 3H), 2.25 (s, 3H). LRMS (ESI, Positive) rnle 339.1 (M+1).

Compound 280: 1- (5-Methyl-pyrazin-2-yl)-3- [5-methyl-2- (2-pyndin-3-yl-ethoxy)-phenyl]-urea Prepared according to the procedure for compound 266, using 2-pyridin-3-yl-ethanol. (16% yield).'H NMR (400 MHz, CDC13) 8 10.98 (br s, IH), 8 65 (s, 1H), 8.49 (d, 1H), 8.35 (s, 2H) 8.20 (s, 1H), 7.75 (s, 1H), 7.65 (d, 1H), 7.19 (m, 1H), 6.82 (dd, 2H), 4.31 (t, 2H), 3.21 (t, 2H), 2.48 (s, 3H), 2.35 (s, 3H).

LRMS (ESI, Positive) m/e 364.2 (M+1).

Compound 281: 1- [5-Methyl-2- (3-piperidin-1-yl-propoxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Prepared according to the procedure for compound 266, using 3-piperidin-1-yl-propan-1-ol. (33% yield).'H NMR (400 MHz, CDC13) 8 11.21 (br s, 1H), 8.35 (s, 1H), 8.25 (s, 2H), 8. 15 (s, 1H), 6.80 (dd, 2H), 4.15 (t, 2H), 2.53 (t, 2H), 2.52 (s, 3H), 2.45 (m, 4H), 2. 39 (s, 3H), 2.10 (m, 2H), 1.61 (m, 4H), 1.45 (m, 2H). LRMS (ESI, Positive) m/e 384.2 (M+1).

Compound 282: 1- [5-Methyl-2- (l-methyl-piperidin-4-yloxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Prepared according to the procedure for compound 266, using 1-rnethyl-piperidin-4-ol. (4% yield).'H NMR (400 MHz, CDC13) 8 11.22 (br s, 1H), 8.43 (s, 1H), 8.25 (s, 1H), 8.20 (s, 1H), 8.10 (s, 1H), 6.81 (dd, 2H), 4.25 (m, 1H), 2.8 (m, 2H), 2.59 (s, 3H), 2. 39 (s, 3H), 2. 36 (s, 3H), 2.19 (m, 4H), 1.90 (m, 2H). LRMS (ESI, Positive) m/e 355.9 (M+1).

Compound 283: 1- [2- (1-Benzyl-piperidin-4-yloxy)-5-methyl-. phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Prepared according to the procedure for compound 266, using 1-benzyl-piperidin-4-ol. (1% yield).

LRMS (ESI, Positive) m/e 432.0 (M+1).

Compound 284: 1- [5-Methyl-2- (3- (S)-1-methyl-piperidin-3-ylmethoxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Step 1: (1-Methyl-piperidin-3- (S)--yl)-methanol To a stirred, cooled solution of (S)- (+)-N-boc nipecotic acid (5.0 mmol) in tetrahydrofuran (10 mL) lithium aluminum hydride (20 mL, 20 mmol, 1M in tetrahydrofuran.) was added dropwise. The reaction was refluxed for 12 hours and then cooled to 0 °C. The reaction was quenched with 1 mL of H20, 1 ni. L of 15% aqueous sodium hydroxide, 3 mL of H, O. The reaction was filtered and the filter cake was washed with tetrahydrofuran (3 x 50 mI,) The filtrate was concentrated under reduced pressure to yield a clear viscous oil.

Steps 2-3: 5-Methyl-2- (1-methyl-piperidin-3- (S)-ylmethoxy)-phenylamine Mitsunobu reaction and aniline deprotection according to procedure for compound 253.

Step 4: Prepared according to the procedure for compound 266 (33% yield).'H NMR (400 MHz, CDCl3) # 11.25 (br s, 1H), 8.45 (br s, 1H), 8.35 (s, 1H), 8.22 (s, 2H), 6.80 (d, 1H), 6.74 (d, 1H), 3.80 (m, 2H), 3.15 (br d, 1H), 2.80 (br d, 1H), 2.51 (s, 3H), 2. 35 (s, 3H), 2.30 (m, 1H), 2.22 (s, 3H), 1.50- 2.00 (m, 6H), 1.00-1.25 (m, 2H). LRMS (ESI, Positive) m/e 370.0 (M+1).

Compound 285: 1- [5-Methyl-2- (3- (R)-1-methyl-piperidin-3-ylmethoxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Steps 1-3: 5-Methyl-2- (1-methyl-piperidin-3- (R)-ylmethoxy)-phenylamine according to the method for compound 284, using (R)-(+)-N-boc nipecotic acid.

Step 4: 5-Methylpyrazine-2-carboxylic azide (1.2 eq.) dissolved in anhydrous toluene (0. 1 M concentration) was heated to 90 °C. After 20 minutes N2 evolution had subsided, and the caramel colored reaction mixture was cooled to 60 °C before adding the aniline prepared above as a solution in toluene (1 eq.). After stirring for 4 hours at 60 °C, the reaction mixture was cooled to room temperature overnight. A precipitate formed which was isolated by filtration (49% yield). lH NMR (400 MHz, CDCl3) 8 11.25 (br s, 1H), 8.45 (br s, 1H), 8.35 (s, 1H), 8.22 (s, 2H), 6.80 (d, 1H), 6.74 (d, 1H), 3.80 (m, 2H), 3. 15 (br d, 1H), 2.80 (br d, 1H), 2.51 (s, 3H), 2.35 (s, 3H), 2.30 (m, 1H), 2.22 (s, 3H), 1.50-2.00 (m, 6H), 1.00-1.25 (m, 2H). LRMS (ESI, Positive) m/e 370. 0 (M Compound 286 : 1- (5-Methyl-pyrazin-2-yl)-3- [5-methyl-2- ( 1-pyridm-3-yl-ethoxy)-phenyl]-urea Step 1: 1-Pyridin-3-yl-ethanol To a stirred, cooled (-78°C) solution of pyridine-3-carbaldehyde (15 mmol) in tetrahydrofuran (40 mL), methyl magnesium bromide (5 mL, 15 mmol, 3M in diethyl ether) was added. After stirring for 2 hours, the reaction was quenched with saturated aqueous ammonium chloride (5 mL). The pH was adjusted to #5. 0 with aqueous sodium carbonate and the product was extracted with ethyl acetate (3 x 100 mL). The ethyl acetate was washed with brine (1 x 100 mL), dried (MgS04), and filtered. The filtered material was concentrated under reduced pressure to yield a yellow oil.

Steps 2-3: 5-Methyl-2-(1-pyridin-3-yl-ethoxy)-phenylamine Mitsunobu reaction and aniline deprotection according to the method for compound 253.

Step 4: Urea formation was conducted according to the procedure for compound 285. (17% yield).'H NMR (400 MHz, CDC13) 8 11.49 (br s, 1H), 8.81 (s, 1H), 8.73 (s, 1H), 8.52 (d, 1 H), 8. 35 (s, iH), 8.25 (s, 1H), 8.18 (s, 1H), 7.80 (s, 1H), 7.72 (d, 1H), 7.20 (t, 1H), 6.70 (d, 1H), 6.65 (d, 1H), 5.49 (q, 1H), 2.50 (s, 3H), 2.30 (s, 3H), 1.75 (d, 3H). LRMS (ESI, Positive) m/e 363.8 (M+1).

Compound 287-.

1- [5-Methyl-2- (l-methyl-piperidin-2-ylmethoxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Steps 1-2: 5-Methyl-2- (l-methyl-piperidin-2-ylmethoxy)-phenylamine Mitsunobu reaction and aniline deprotection according to the method for compound 253.

Step 3: Urea formation was conducted according to the procedure for compound 285. (11% yield NMR (400 MHz, CDC13) 8 10.90 (br s, 1H), 8.45 (s, 1H), 8.21 (s, 1H), 8.18 (s, 1H), 7.80 (s, 1H), 6.80 (s, 2H), 4.51 (m, 1H), 2.58-3.00 (m, 4H), 2.52 (s, 3H), 2.46 (s, 3H), 2.35 (s, 3H), 1.50-2.25 (m, 7H).

LRMS (ESI, Positive) m/e 369.9 (M+1).

Compound288: 1- (S-Methyl-pyrazin-2-yl)-3- [5-methyl-2- (tetrahydro-furan-2-ylmethoxy)-phenyl]-urea Steps 1-2: 5-Methyl-2- (tetrahydro-furan-2-ylmethoxy)-phenylamine Mitsunobu reaction and aniline deprotection according to the method for compound 253.

Step 3: Urea formation was conducted according to the procedure for compound 285. (12% yield).'H NMR (400 MHz, CDC13) 8 11.25 (br s, 1H), 8.49 (s, 1H), 8.40 (s, 1H), 8.22 (s, 1H), 8. 18 (s, 1H), 6.8 (s, 2H), 4.42 (m, 1H), 3.80-4.10 (m, 4H), 2.52 (s, 3H), 2.35 (s, 3H), 1.20-2.20 (m, 4H). LRMS (ESI, Positive) m/e 342.9 (M+1).

Compound 289: 1- [5-Methyl-2- (l-methyl-piperidin-4-ylmethoxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Step 1: (1-Methyl-piperidin-4-yl)-methanol To a stirred, cooled solution of 1-methyl-piperdine-4- carboxylic acid (5.0 mmol) in tetrahydrofuran (10 mL) lithium aluminum hydride (20 mL, 20 mmol, 1M tetrahydrofuran.) was added dropwise. The reaction was stirred for 4 hours, quenched with 1 mL of H2O, 1 mL of 15% aqueous sodium hydroxide, and 3 mL of H2O. The reaction was filtered and washed with tetrahydrofuran (3 x 50 mL). The filtrate was concentrated under reduced pressure to yield a clear viscous oil.

Steps 2-3: 5-Methyl-2- (1-methyl-piperidin-4-ylmethoxy)-phenylamine Mitsunobu and aniline deprotection according to the procedure for compound 253.

Step 4: Urea formation according to the procedure for compound 285 (54% yield).

'H NMR (400 MHz, CDC13) 8 11. 18 (br s, 1H), 8.62 (br s, 1H), 8.38 (br s, 1H), 8.25 (s, 1H), 8.18 (s, 1H), 6.82 (d, 1H), 6.78 (d, 1H), 3.85 (d, 2H), 2.90 (br d, 2H), 2.51 (s, 3H), 2.35 (m, 6H), 1.50-2.10 (m, 7H).

LRMS (ESI, Positive) m/e 369.2 (M+1).

Compound 290 : 1- [5-Methyl-2- (l-methyl-piperidin-3-yloxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Steps 1-2: 5-Methyl-2- (1-methyl-piperidin-3-yloxy)-phenylamine Mitsunobu reaction and aniline deprotection according to the method for compound 253.

Step 3: Urea formation according to the procedure for compound 285 (3% yield). *H NMR (400 MHz, CDC13) 8 10.75 (br s, 1H), 8.59 (br s, 1H), 8.18 (s, 1H), 8.05 (s, 1H), 7.62 (s, 1H), 6.90 (d, 1H), 6.80 (d, 1H), 4.40 (m, 1H), 2.58 (s, 3H), 2. 39 (s, 6H), 1.60-2.80 (m, 8H).

LRMS (ESI, Positive) m/e 356.1 (M+1).

Compound 291: 1- (5-Methyl-pyrazin-2-yl)-3- [S-methyl-2- ( 1, 2,3,4-tetrahydro-quinolin-3-ylmethoxy)-phenyl]-urea Step 1: Quinoline-3-carboxylic acid methyl ester. To a stirred solution of quinoline-3-carboxylic acid (346 mg, 2 mmol) dissolved in 4: 1 THF: MeOH (6 mL) at 0 °C was added TMS-diazomethane (2M in hexane) portionwise until a diazomethane yellow color persisted. The reaction was concentrated to the give the methyl ester as a tan solid (244 mg, 65%).'H-NMR (400 MHz, CDCI3) 6 9.44 (s, 1H), 8.85 (s, 1H), 8.17 (d, 1H), 7.96 (d, 1H), 7.84 (dd, 1H), 7.62 (dd, 1H), 4.02 (s, 3H) Step 2: 1, 2,3,4-Tetrahydro-quinoline-3-carboxylic acid methyl ester and 1-Ethyl-1, 2,3,4-tetrahydro- quinoline-3-carboxylic acid methyl ester. To a stirred solution of the quinoline-3-carboxylic acid methyl ester (244 mg, 1.3 mmol) in glacial acetic acid (13 mL) at room temperature was added NaBH4 (345 mg, 9.1 mmol) portionwise (vigorous reaction). After complete addition the reaction was dark yellow. After stirring for 3 hours the color had become pale yellow. The reaction mixture was poured into 50 mL of H20 and 50 mL of CH2CI2 and stirred rapidly for 15 min. The layers were separated and the organics concentrated to a yellow oil. TLC in 15/85 EtOAc/hexane showed complete consumption of starting material. and two new lower rf spots. The compound was chromatographed using a Biotage 12M column (loaded with CEbCIz) and eluted with 15/85 EtOAc/hexane. The higher rf spot corresponds the N-ethylated product (123 mg, 43%). The lower rf spot corresponds to the desired tetrahydroquinoline-3-carboxylic acid methyl ester (93 mg, 37%). N-ethyl derivative :'H-NMR (400 MHz, CDC13) b 7.08 (dd, 1H), 6.99 (d, 1H), 6.60 (m, 2H), 3.73 (s, 3H), 3.42 (m, 3H), 3.27 (m, 1H), 2.98 (m, 3H), 1.14 (t, 3H) N-H derivative :'H-NMR (400 MHz, CDC13) 8 6.98 (m, 2H), 6.62 (dd, 1H), 6.47 (d, 1H), 3.71 (s, 3H), 3.52 (m, 1H), 3. 34 (m, 1H), 3.00 (m, 2H), 2.90 (m, 1H) Step 3: (1, 2,3,4-Tetrahydro-quinolin-3-yl)-methanol. To a stirred solution of the 1,2,3,4-Tetrahydro- quinoline-3-carboxylic acid methyl ester (93 mg, 0.49 mmol) in 1.5 mL of Et2O at 0 °C under nitrogen was added LiAlH4 (1M in Et2O) dropwise with vigorous gas evolution and a white precipitate formation. After 30 min., the reaction was carefully quenched with 15% NaOH (3 mL) and 3 mL of Et2O was added and the mixture stirred rapidly at RT for 15 min. The layers were separated and the aqueous layer extracted (1 x 10 mL) with Et2O. The organics were combined, dried (MgSO4), filtered and concentrated to the alcohol (64 mg, 80%).'H-NMR (400 MHz, CDC13) 5 6.97 (m, 2H), 6.62 (dd, 1H), 6.47 (d, 1H), 3.66 (m, 1H), 3.58 (m, 1H), 3.40 (m, 1H), 3.08 (m, 1H), 2.82 (m, 1H), 2.53 (m, 1H), 2.18 (m, lH).

Step 4: [5-Methyl-2- (1, 2,3,4-tetrahydro-quinolin-3-ylmethoxy)-phenyl]-carbamic acid tert-butyl ester.

To a stirred solution of 2-N-Boc-amino-4-methylphenol (88 mg, 0.39 mmol), (1,2,3,4-Tetrahydro- quinolin-3-yl)-methanol (64 mg, 0.39 mmol) and triphenylphosphme (103 mg, 0.39 mmol) in 850 pL of THF at 0 °C under nitrogen was added a solution of DIAD (77 µL, 0.39 mmol) in 850 uL THF. The reaction was allowed to warm to RT overnight, was concentrated and loaded directly onto a Biotage 12M column with CH2CI2 and eluted with 96/4 hexane/EtOAc. The product was isolated as a yellow oil (129 mg, 89%).'H-NMR (400 MHz, CDC13) 8 7.92 (br s, 1H), 7.06 (br s, 1H), 6.99 (m, 2H), 6.81 (m, 1H), 6.73 (s, 2H), 6.65 (dd, 1H), 6.51 (d, 1H), 3.96 (m, 2H), 3. 37 (ddd, 2H), 2.79 (ddd, 2H), 2.56 (m, 1H), 2.28 (s, 3H), 1.54 (s, 9H) Step 5: 3- (2-tert-Butoxycarbonylamino-4-methyl-phenoxymethyl)-3, 4-dihydro-2H-quinoline-l- carboxylic acid benzyl ester. To a stirred solution of [5-Methyl-2- (1, 2,3,4-tetrahydro-quinolin-3- ylmethoxy)-phenyl]-carbamic acid tert-butyl ester. To a stirred solution of 2-N-Boc-amino-4- methylphenol (129 mg, 0.35 mmol) in CH2C12 (1.5 mL) at 0 °C under nitrogen was added DIEA (61 uL, 0. 35 mmol) followed by benzyl chloroformate (50 1L, 0. 35 mmol) and DMAP (4 mg, 0.035 mmol). After 24 hours, the reaction was diluted to 30 mL with CH2CI2 and washed (2 x 30 mL) with 2N HCl and (2 x 30 mL) with saturated NaHCO3. The organics were dried (MgS04), filtered and concentrated to a brown oil, which appeared to be a mixture of product and starting material.

Step 6: 3-(2-Amino-4-methyl-phenoxymethyl)-3, 4-dihydro-2H-quinoline-l-carboxylic acid benzyl ester. A solution of the crude 3- (2-tert-Butoxycarbonylamino-4-methyl-phenoxymethyl)-3, 4-dihydro- 2H-quinoline-l-carboxylic acid benzyl ester (crude, 0.35 mmol) in 4N HCl in dioxane (2 mL) at room temperature was stirred under a drying tube overnight. The suspension was concentrated by rotovap, diluted to 30 mL with CH2CI2 and shaken with 10% Na2CO3 (30 mL). The organics were isolated, dried (MgS04), filtered and concentrated to a brown oil corresponding to the crude aniline, which was used without purification in the urea forming reaction.

Step 7: 3- {4-Methyl-2- [3- (5-methyl-pyrazin-2-yl)-ureido]-phenoxymethyl}-3, 4-dihydro-2H-quinoline- 1-carboxylic acid benzyl ester. A 0.5 M solution of the 5-methyl-pyrazine-2-carboxylic acid azide (204 ttL) was diluted with toluene (408 µL) in a septum capped reaction vial under nitrogen and with stirring, immersed in a 90 °C oil bath. After about 20 minutes nitrogen gas evolution had stopped so the reaction was allowed to cool to RT and was treated with a solution of the crude 3- (2-Amino-4- methyl-phenoxymethyl)-3,4-dihydro-2H-quinoline-l-carboxylic acid benzyl ester (ca. 0.101 mmol) in toluene (6201lu). The mixture was stirred at 65'C for 2 hours. The reaction was cooled to rt overnight and a precipitate formed. The precipitate was filtered off with toluene and appeared to be a mixture of Cbz-protected and deprotected product.

Step 8: 1- (5-Methyl-pyrazin-2-yl)-3- [5-methyl-2- (1, 2,3,4-tetrahydro-quinolin-3-ylmethoxy)-phenyl]- urea. A stirred suspension of the crude 3- {4-Methyl-2- [3- (5-methyl-pyrazin-2-yl)-ureido]- phenoxymethyl}-3, 4-dihydro-2H-quinoline-l-carboxylic acid benzyl ester (6.6 mg, 12 µmol) was heated in 5 mL EtOAc on a heat gun until in solution. The clear solution was cooled to rt and treated with triethylamine (3.4 liL, 24 pmol) followed by Pearlman's catalyst (20% palladium hydroxide on carbon, 9 mg). The mixture was put through a vacuum/purge cycle three times with hydrogen gas and then held under 1 atmosphere hydrogen pressure for 1 hour. The reaction was filtered through GF/F filter paper with EtOAc and concentrated to a white solid which corresponds to the desired product (4.7 mg, 100%).'H-NMR (400 MHz, CDC13/CD30D) 8 8.23 (s, 1H), 8.18 (s, IH), 7.96 (s, 1H), 7.04 (t, 1H), 6.97 (d, 1H), 6.81 (m, 2H), 6.67 (t, 1H), 6.58 (d, 1H), 4.04 (m, 2H), 3.57 (d, 1H), 3.26 (t, 1H), 2.92 (ddd, 2H), 2.64 (m, 1H), 2.34 (s, 3H), 2.31 (s, 3H) LRMS (APCI, Positive) m/e 404.2 (M+1).

Compound 292 : 1- [2- (l-Ethyl-1, 2,3,4-tetrahydro-qumolin-3-yimethoxy)-5-methyl-phenyl]-3-(5- methyl-pyrazin-2-yl)- urea Step 1: (1-Ethyl-1, 2,3,4-tetrahydro-quinolin-3-yl)-methanol. To a stirred solution of 1-Ethyl-1, 2,3,4- tetrahydro-quinoline-3-carboxylic acid methyl ester (123 mg, 0.56 mmol) in 1.5 mL of Et20 at 0 °C under nitrogen. LAH (1M in Et2O) was added dropwise with vigorous gas evolution and a white precipitate formation. After 30 min., TLC in 3/7 EtOAc/hexane showed complete loss of s. m. and appearance of a clean lower rf spot. The reaction was carefully quenched with 15% NaOH (3 mL) and 3 mL of Et20 was added and the mixture stirred rapidly at RT for 15 min. The layers were separated and the aqueous layer extracted 1 x 10 mL with Et2O. The organics were combined, dried (MgS04), filtered and concentrated to a clear oil corresponding to the desired alcohol (105 mg, 95%).

Step 2 : [2- (l-Ethyl-1, 2,3,4-tetrahydro-quinolin-3-ylmethoxy)-5-methyl-phenyl]-carb amic acid tert- butyl ester. To a stirred solution of (l-Ethyl-1, 2,3,4-tetrahydro-quinolin-3-yl)-methanol (105 mg, 0.55 mmol, prepared in step 2, compound 126xx), 2-N-Boc-amino-4-methylphenol (123 mg, 0.55 mmol), and triphenylphosphine (144 mg, 0.55 mmol) in 850 L of THF at 0 °C under nitrogen was added a solution of DIAD (108 L, 0.55 mmol) in 850 iL THF. The reaction was allowed to warm to RT overnight, was concentrated and loaded directly onto a Biotage 12M column with CH2CI2 and eluted with 96/4 hexane/EtOAc to give the desired alkylated phenol as a white foam (40 mg, 18%).'H-NMR (400 MHz, CDC13) 8 7.92 (br s, 1H), 7.08 (dd, 1H), 7.02 (m, 1H), 6.99 (d, 1H), 6.75 (s, 2H), 6.62 (d, 1H), 6.59 (dd, 1H), 3.98 (m, 2H), 3.39 (m, 4H), 3.20 (m, 1H), 2.79 (ddd, 2H), 2.58 (m, 1H), 2.28 (s, 3H), 1.56 (s, 9H), 1.16 (t, 3H).

Step 3: 2- (1-Ethyl-1, 2,3,4-tetrahydro-quinolin-3-ylmethoxy)-5-methyl-phenylamine. A solution of [2- (1-Ethyl-1, 2,3,4-tetrahydro-quinolin-3-ylmethoxy)-5-methyl-phenyl]-carb amic acid tert-butyl ester (40 mg, 0.1 mmol) was stirred in 4N HC1 in dioxane (2 mL) at room temperature under a drying tube overnight. The suspension was concentrated by rotovap, diluted to 30 mL with CH2CI2 and shaken with 10% Na2CO3 (30 mL). The organics were isolated, dried (MgS04), filtered and concentrated to a brown oil, which was used without purification in the following reaction.

Step 4: 1- [2- (l-Ethyl-1, 2,3,4-tetrahydro-quinolin-3-ylmethoxy)-5-methyl-phenyl]-3- (5-methyl- pyrazin-2-yl)-urea. A 0.5 M solution of the acyl azide (182 pL) was diluted with 364 uL toluene in a septum capped reaction vial under nitrogen and with stirring, immersed in a 90 °C oil bath. After about 20 minutes N2 gas evolution had stopped so the reaction was allowed to cool to rt and was treated with a solution of 2- (1-ethyl-1, 2,3,4-tetrahydro-quinolin-3-ylmethoxy)-5-methyl-phenylamine (27 mg, 0.91 mmol) in 550 pL of toluene. The mixture was stirred at 65 °C for 2 hours. The reaction was cooled to rt overnight and a precipitate formed. The precipitate was filtered off with toluene and the desired urea isolated as a tan solid (7 mg, 18 %).'H-NMR (400 MHz, CDC13) 8 11.34 (br s, 1H), 8.25 (s, 1H), 8.21 (s, 1H), 8.11 (br s, 1H), 7.83 (s, 1H), 7.25 (m, 1H), 7.18 (d, 1H), 7.12 (t, 1H), 6.97 (d, 1H), 6.80 (m, 2H), 6.70 (d, 1H), 6.61 (t, 1H), 4.05 (m, 2H), 3.52 (m, 2H), 3. 32 (t, 1H), 3.17 (m, 1H), 2.91 (ddd, 2H), 2.69 (m, 1H), 2. 36 (s, 3H), 2.27 (s, 3H), 1.04 (t, 3H). LRMS (APCI, Positive) m/e 431.9 (M+1).

Compound 293 : 1- [5-Methyl-2- ( 1-methyl-piperidin-3-ylmethoxy)-phenyl]-3-quinoxalin-2-yl-ur ea Step 1: Quinoxaline-2-carbonyl azide. A stirred solution quinoxaline-2-carboxylic acid (348 mg, 2 mmol) in THF (6 mL) at rt under nitrogen was treated with diisopropylethylamine (365 tL, 2.1 mmol) followed by diphenylphosphoryl azide (410 gL, 1.9 mmol). After stirring overnight the reaction was diluted to 60 mL with Et2O and washed 2 x 60 mL with sat. NaCI. There was an insoluble brown oil, which was drained off with the aqueous layer and assumed to be a diphenyl phosphate impurity. The organics were dried (MgS04), filtered and concentrated to a tan solid, which corresponds to the acyl azide (350 mg, 92%).'H-NMR (400 MHz, CDC13) 8 9.58 (s, 1H), 8. 32 (d, 1H), 8.21 (d, 1H), 7.94 (m, 2H) Step 2: 1- [5-Methyl-2- (l-methyl-piperidin-3-ylmethoxy)-phenyl]-3-quinoxalin-2-yl-u rea. A solution of quinoxaline-2-carbonyl azide (66 mg, 0.33 mmol) in toluene (1.7 mL) was stirred under nitrogen and immersed in a 90 °C heating bath. After 20 min. the reaction was cooled to 65 °C and solid 5-methyl- 2- (l-methyl-piperidin-3-ylmethoxy)-phenylamine (70 mg, 0. 3 mmol) was added. The reaction was stirred at 65 °C for 4 hours and then allowed to cool to rt overnight. The resulting precipitate was collected by filtration and washed with toluene (62 mg, 51%). 1H-NMR (400 MHz, CDCl3) # 11.58 (br s, 1H), 9.27 (br s, 1H), 8.63 (s, 1H), 8.22 (s, 1H), 8. 03 (d, 1H), 7.91 (d, 1H), 7.76 (t, 1H), 7.61 (t, 1H), 6.86 (m, 2H), 4.03 (m, 2H), 2.91 (d, 1H), 2.61 (d, 1H), 2.37 (s, 3H), 2.25 (m, 1H), 2.09 (s, 1H), 1.81 (m, 3H), 1.57 (m, 2H), 1.05 (m, 1H). LRMS (APCI, Positive) m/e 405.9 (M+1).

Compound294 : 1- [5-Methyl-2- (pyridin-3-ylmethoxy)-phenyl]-3-quinoxalin-2-yl-urea Step 1: 1- [5-Methyl-2- (pyridin-3-ylmethoxy)-phenyl]-3-quinoxalin-2-yl-urea. A stirred solution of quinoxaline-2-carbonyl azide (92 mg, 0.46 mmol, prepared as above) in 1.5 mL toluene under nitrogen was immersed in a 90 °C heating bath. After 20 min. the reaction was cooled to 65 °C and treated with solid 5-methyl-2- (pyridin-3-ylmethoxy)-phenylamine (90 mg, 0.42 mmol). The reaction was stirred at 65 °C for 4 hours and then allowed to cool to rt overnight. The resulting precipitate was collected by filtration. The crude product was chromatographed on a Biotage 12M column with 2/3 EtOAc/hexane to give pure urea as a tan solid (20 mg, 12%).'H-NMR (400 MHz, CDC3) 8 11.99 (br s, 1H), 9.64 (br s, 1H), 8.76 (s, 1H), 8.61 (s, 1H), 8.48 (d, 1H), 8.24 (s, 1H), 7.98 (d, 1H), 7.79 (d, 1H), 7.55 (t, 1H), 7.42 (t, 1H), 7.17 (d, 1H), 7. 12 (m, 1H), 6.93 (m, 2H), 5.26 (s, 2H), 2.41 (s, 3H). LRMS (APCI, Positive) m/e 385.9 (M+1).

Compound 295 : OH jA OH O'p OzN¢ Step 1 pN Step 2 H2N Step 3 N N N w \ I/N O Compound 130 Step 1 Mitsunobo procedure: 1- [2- (4-Methyl-2-nitro-phenoxy)-ethyl]-aziridine. A solution of 2-nitro-4-methylphenol (505 mg, 3. 3 mmol, 1. 1 eq.) and 2-aziridin-1-yl-ethanol (3.0 mmol, leq.) in 10 mL THF was stirred at 0 °C.

Triphenylphosphine (0.87 g, 3.30 mmol, 1.1 eq.) and diisopropylazodicarboxylate, (0.67 g 3.30 mmol, 1.1 eq.) were added, and the solution was allowed to warm to temperature. After 18h, the reaction mixture was diluted with 100 mL EtOAc, and was washed with water (3 x 20 mL). The organic phase was washed again with 1 N HC1 (3x 20 ml). The aqueous layer was basified with 3 N NaOH to pH > 12 and extracted with EtOAc, (3x 50 mL) to give crude product. The final product was purified by flash chromatography eluting with 5-10 % MeOH in dichloromethane.'H NMR (400 MHz, CDC13) : 8 7.66 (s, 1H), 7.32 (d, J= 8.61 Hz, 1H), 7.01 (d, J= 8.61 Hz, 1H), 4.26 (t, J= 5.09 Hz, 2H), 2.67 (t, J = 5.48 Hz, 2H), 2.34 (s, 3H), 1.79 (m, 2H), 1.34 (m, 2H).

Step 2 Nitro reduction.

2- (2-Aziridin-1-yl-ethoxy)-5-methyl-phenylamine. A solution of 3-nitro-4-alkoxy toluene (1.0 mmol) in 20 mL EtOH was hydrogenated at 2 atm over 300 mg of 10% Pd on carbon for 30 minutes.

The catalyst was removed by filtration through a glass fiber filter and the filtrate was concentrated to give the desired product, which was used directly without further purification.

Step 3 Urea formation 1- [2- (2-Aziridin-1-yl-ethoxy)-5-methyl-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea. A solution of 5- methylpyrazine-2-carboxylic azide (196 mg, 1.2 mmol, 1.2 eq.) in 20 mL anhydrous toluene was heated to 90 °C. After 20 minutes N2 evolution had subsided, and the reaction mixture was cooled to 60 °C before adding aniline (1.0 mmol, 1 eq.) as a solution in 2 mL toluene. After stirring for 4 h at 60 °C.

The reaction mixture was then partitioned between 50 mL EtOAc and sat. NaHCO3. the organic phase was washed with brine, dried over MgS04, filtered and concentrated. The residue was purified by flash chromatography, eluting with 5 % MeOH in dichloromethane.'H NMR (400 MHz, d6-DMSO) : 8 10.80 (s, 1H), 8. 64 (s, 1H), 8.53 (s, 1H), 8.15 (s, 1H), 8.07 (s, 1H), 6.82 (m, 2H), 4.2 (m, 2H), 2.7 (m, 2H), 2.5 (s, 3H), 2. 32 (s, 3H), 1.89 (s, 2H), 1.30 (m, 2H). MS APCI-Pos, M/e 328.0 (M+1) Compound 296 : 1- [2- (3-Dimethylamino-benzyloxy)-5-methyl-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Prepared from (3-dimethylamino-phenyl)-methanol, as described above for compound 295.'H NMR (400 MHz, CDC13) : 8 11.69 (s, 1H), 8.26 (s, 1H), 8.11 (s, 1H), 7.93 (s, 1H), 7.26 (m, 1H), 6.87 (m, 6H), 5.01 (s, 2H), 2.93 (s, 6H), 2. 35 (s, 6H). MS APCI-Pos, M/e 391.9 (M+1) Compound 297: 1- [2- (l-Isopropyl-pyrrolidin-3-yloxy)-5-methyl-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Prepared from 3-hydroxy-1-isopropyl-pyrrolidine, as described above for compound 295.'H NMR (400 MHz, d6-DMSO) : 8 10.08 (s, 2H), 8.65 (s, 1H), 8.14 (s, 1H), 8.03 (s, 1H), 6.81 (d, J= 7.83 Hz, 1H), 6.74 (d, J= 8.61 Hz, 1H), 4.85 (s, 1H), 2.91 (m, 1H), 2.75 (m, 2H), 2.48 (m, 1H), 2.4 (s, 3H), 2.36 (m, 1H), 2.27 (m, 1H), 2.21 (s, 3 H), 1.85 (m, 1H), 1.01 (m, 6H). MS APCI-Pos, M/e 369.9 (M+1) Compound 298: 1- [5-Methyl-2- (l-methyl-pyrrolidin-3-ylmethoxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea.

Prepared from (1-methyl-pyrrolidin-3-yl)-methanol as described above for compound 295.'H NMR (400 MHz, d6-DMSO) : 8 10.13 (s, 2H), 8.67 (s, 1H), 8.15 (s, 1H), 8.06 (s, 1H), 6.81 (d, J= 8.61 Hz, 1H), 6.75 (d, J = 7.83 Hz, 1H), 4.88 (s, 1H), 2.75 (m, 4H), 2.5 (m, 2H), 2.43 (s, 3H), 2.3 (s, 3H), 2.24 (s, 3H). MS APCI-Pos, M/e 341.9 (M+1) Compound 299: rr 0" H step 1 YBoc step 2-4 09, Boc step 5 H H Oye - H H, N N N N\ j N) 0 N) HOz HO) ¢N N N >9 N Compound 134 Step 1: 3-Hydroxymethyl-piperidine-l-carboxylic acid tert-butyl ester. To a stirred solution of 3- hydroxymethyl piperdine (403 mg, 3.5 mmol, 1 eq.) in 20 mL of CH2C12 and 5 mL of sat'd NaHC03, 0 °C was added di-tert-butyl dicarbonate (803 mg, 3.68 mmol, 1.05 eq.) in several portions. After stirring at 0 °C for 2 h, the solution was diluted with 10 mL of water and was extracted with 2 x 20 mL CH2C12. the combined extracts were washed with water, then brine and were dried over MgS04, filtered and concentrated to give the Boc protected amine which was used in the next step.

Steps 2-4: 3- {4-Methyl-2- [3- (5-methyl-pyrazin-2-yl)-ureido]-phenoxymethyl}-piperidine-1- carboxylic acid tert-butyl ester was prepared from 3-hydroxymethyl-piperidine-1-carboxylic acid tert-butyl ester, as described above for compound 295. It was purified by flash chromatography eluting with 5% MeOH in CHzCt.

Step 5: 1- [5-Methyl-2- (piperidin-3-ylmethoxy)-phenyll-3- (5-methyl-pyrazin-2-yl)-urea Removal of the Boc group was accomplished by treatment of a 0'C solution of the protected derivative (180 mg, 0. 395 mmol) in 15 mL CH2CI2 with 2 mL of TFA. After stirring for 18h at room temperature, the reaction was concentrated in vacuo, and the residue was taken up in 20 mL of EtOAc and was washed with 10 mL of NaHC03. The aqueous phase was extracted with EtOAc (2 x 30 mL), and the combined extracts were washed with 20 mL of brine, dried over MgSO4, filtered and concentrated to give 128 mg (91%) of the desired amine.'H NMR (400 MHz, d6-DMSO) : 8 10.16 (s, 1H), 10.09 (s, 1H), 8.53 (s, 1H), 8.11 (s, 1H), 7.97 (s, 1H), 6.8 (d, J= 7.8 Hz, 1H), 6.69 (d, J= 8.6 Hz, 1H), 3.77 (s, 2H), 3.09 (m, 1H), 2.82 (m, 1H), 2. 34 (m, 2H), 2. 3 (s, 3H), 2.27 (m, 1H), 2.15 (s, 3H), 1.92 (m, 1H), 1.75 (m, 1H), 1.53 (m, 1H), 1.37 (m, 1H), 1.11 (m, 1H). MS APCI-Pos, M/e 356.0 (M+1) Compound 300: 1- [5-Fluoro-2- (pyridin-3-ylmethoxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Step 1: 3- (4-Fluoro-2-nitro-phenoxymethyl)-pyridine. To a stirred, cooled (about 0 °C) solution of 1, 4-Difluoro-2-nitro-benzene (3.0 mmol) and 3-pyridylcarbinol (3.1 mmol) in tetrahydrofuran (8mL) was added lithium bis (trimethylsilyl) amide (3.2 mmol ; 3.2 mL of a 1.0 M solution in tetrahydrofuran).

After stirring for 12 hours, the reaction was diluted with 30 mL of ethyl acetate and washed with 30 mL of 10% aqueous sodium carbonate (2 x 30 mL), brine (1 x 30 mL), then dried (MgS04), and filtered.

The filtered solution was concentrated under reduced pressure to provide the desired crude product.

Step 2: 5-Fluoro-2- (pyridin-3-ylmethoxy)-phenylamine. To a stirred, cooled (about 0 °C) solution of a 4-Fluoro-2-nitro-phenoxymethyl)-pyridine (1.0 mmol) in methanol (2 mL) and saturated aqueous ammonium chloride (ImL) was added zinc dust (2.0 mmol). After stirring for 12 hours, the reaction was diluted with 30 mL of ethyl acetate and washed with 30 mL of 10% aqueous sodium carbonate (2 x 30 mL), brine (1 x 30 mL), then dried (MgS04), and filtered. The filtered solution was concentrated under reduced pressure to provide the desired crude product.

Step 3: Urea formation according to method for compound 295 (23% yield NMR (400 MHz, CDC13) 8 11.62 (br s, 1H), 8.80 (s, 1H), 8.75 (d, 1H), 8.43 (br s, 1H), 8.25 (d, 1H), 8.15 (s, 1H), 7.85 (d, 1H), 7.43 (m, 1H), 6.95 (m, 2H), 6.68 (m, 1H), 5.15 (s, 2H), 2.43 (s, 3H). LRMS (ESI, Positive) m/e 354. 10 (M+1).

Compound 301 : 1- [5-Fluoro-2- (l-methyl-piperidin-3-ylmethoxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Steps 1-2: According to procedure for compound 300, using 1, 4-difluoro-2-nitrobenzene and 1- methyl-3-hydroxymethyl piperidine.

Step 3: Urea formation according to method for compound 285 (62% yield).'H NMR (400 MHz, CDC13) 8 8.22 (m, 3H), 7.21 (m, 2H), 6.78 (m, 2H), 3.85 (m, 2H), 3.21 (m, 1H), 2.85 (m, 1H), 2.52 (s, 3H), 2.39 (s, 3H), 1.50-2.30 (m, 8H). LRMS (ESI, Positive) m/e 374.21 (M+1).

Compound 302: 1- [5-Fluoro-2- (l-methyl-piperidin-4-yloxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Steps 1-2: According to procedure for compound 300, using 1, 4-difluoro-2-nitrobenzene and 1- methyl-4-hydroxypiperidine.

Step 3: Urea formation according to method for compound 295 (78% yield).'H NMR (400 MHz, CDC13) 8 11.49 (br s, 1H), 8.89 (br s, 1H), 8.35 (s, 1H), 8.22 (d, 1H), 8.10 (s, 1H), 6.80 (m, 1H), 6.70 (m, 1H), 4.25 (m, 1H), 2.90 (m, 2H), 2.55 (s, 3H), 2.38 (s, 3H), 2.35 (s, 3H), 1.80-2. 30 (m, 6H).

LRMS (ESI, Positive) m/e 359.91 (M+1).

Compound 303 : 1- [4-Fluoro-2- (pyridin-3-ylmethoxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Step 1: 3- (5-Fluoro-2-nitro-phenoxymethyl)-pyridine To a stirred, cooled (about 0 °C) solution of 2-nitro-5-fluoro-phenol (2.0 mmol), triphenyl phosphine (2.0 mmol), and 3-hydroxymethylpyridine (2.0 mmol) in dry tertrahydrofuran (5 mL) was added diisopropyl azodicarboxylate (2.0 mmol in 1 mL of tetrahydrofuran). After stirring for 12 hours, the reaction was diluted with 30 mL of ethyl acetate and washed with 30 mL of 10% aqueous sodium carbonate (2 x 30 mL), brine (1 x 30 mL), then dried (MgS04), and filtered. The filtered solution was concentrated under reduced pressure to provide the desired crude product.

Step 2: 4-Fluoro-2- (pyridin-3-ylmethoxy)-phenylamine Nitro reduction according to the method for compound 300.

Step 3: Urea formation according to method for compound 295 (60% yield).

'H NMR (400 MHz, CDC13) 8 11.41 (br s, 1H), 8.85 (s, 1H), 8.75 (d, 1H), 8.40 (t, 1H), 8.18 (s, 1H), 7.88 (s, 1H), 7.80 (d, 1H), 7.43 (t, 1H), 7.00 (s, 1H), 6.80 (m, 2H), 5.12 (s, 2H), 2.43 (s, 3H). LRMS (ESI, Positive) m/e 354. 21 (M+1).

Compound 304: 1- [4-Fluoro-2- (l-methyl-piperidin-3-ylmethoxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Prepared according to the methods for compound 303, using 2-nitro-5-fluorophenol and 1-methyl-3- hydroxymethylpiperidine.

'H NMR (400 MHz, CDC13) 8 8.50 (br s, 1H), 8.19 (m, 2H), 6.65 (m, 2H), 3.85 (m, 2H), 3.60 (s, 3H), 2.80-3.20 (m, 2H), 2.54 (s, 3H), 2.39 (s, 3H), 1.60-2.10 (m 5H).

LRMS (ESI, Positive) m/e 373.95 (M+1).

Compound 305: 1- [4-Fluoro-2- (I-methyl-piperidin-4-yloxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Prepared according to the methods for compound 303, using 2-nitro-5-fluorophenol and 1-methyl-4- hydroxypiperidine.'H NMR (400 MHz, CDC13) b 11. 35 (br s, 1H), 9.49 (s, 1H), 8.35 (m, 2H), 8.05 (s, 1H), 6.65 (m, 2H), 4. 35 (m, 1H), 2.90 (m, 2H), 2.54 (s, 3H), 2. 35 (s, 3H), 1.80-2.30 (m, 6H).

LRMS (ESI, Positive) m/e 359.93 (M+1).

Compound 306: 1- [3, 4-Difluoro-2- (pyridin-3-ylmethoxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea Step 1: 3- (2, 3-Difluoro-6-nitro-phenoxymethyl)-pyridine To a stirred, cooled (about 0 °C) solution of 2,3-difluoro-6-nitrophenol (2.0 mmol), triphenyl phosphine (2.0 mmol), and 3- hydroxymethylpyridine (2.0 mmol) in dry tertrahydrofuran (5 mL) was added diisopropyl azodicarboxylate (2.0 mmol in 1 mL of tetrahydrofuran). After stirring for 12 hours, the reaction was diluted with 30 mL of ethyl acetate and washed with 30 mL of 10% aqueous sodium carbonate (2 x 30 mL), brine (1 x 30 mL), then dried (MgS04), and filtered. The filtered solution was concentrated under reduced pressure to provide the desired crude product.

Step 2: 3,4-Difluoro-2- (pyridin-3-ylmethoxy)-phenylamine Nitro reduction according to the method for compound 300.

Step 3: Urea formation according to method for compound 295 (20% yield).'H NMR (400 MHz, CDC13) 8 11.49 (br s, 1H), 8.89 (s, 1H), 8.85 (s, 1H), 8.65 (d, 1H), 8.25 (s, 1H), 8.10 (m, 1H), 7.88 (d, 1H), 7.35 (t, 1H), 7.18 (s, 1H), 6.98 (m, 1H), 5.25 (s, 2H), 2.52 (s, 3H). LRMS (ESI, Positive) m/e 372.10 (M+1).

Compound 307: 1- (5-Methyl-pyrazin-2-yl)-3- [5-methyl-2- (pyridin-4-yloxy)-phenyl]-urea Step 1: 5-Methyl-2-(pyridin-4-yloxy)-phenylamine To a stirred solution of 2-amino-4-methyl phenol (616 mg; 5.0 mmol) and 4-chloro pyridine (625 mg; 5.5 mmol) in dimethyl sulfoxide (5 mL) was added sodium hydroxide (600 mg; 15.0 mmol, in 1 mL of water). The reaction was heated to 100 °C and stirred for 12 hours. The reaction was cooled to room temperature, diluted with 50 mL of ethyl acetate and washed with aqueous 10% sodium carbonate (1 x 50 mL), and brine (50 mL), then dried (MgS04), and filtered. The crude product was purified using the biotage 40M cartridge eluting with methylene chloride : methanol: ammonia (90: 8: 2) to yield a light yellow oil (10% yield).

Step 2: Urea formation according to method for compound 295 (36% yield).'H NMR (400 MHz, CDC13) 8 11.41 (br s, 1H), 8.52 (m, 3H), 8.33 (s, 1H), 8.22 (s, 1H), 7.61 (s, 1H), 6.80-7.00 (m, 4H), 2.49 (s, 3H), 2.45 (s, 3H). LRMS (ESI, Positive) m/e 335.91 (M+1).

Compound 308: 1- (5-Methyl-pyrazin-2-yl)-3- [5-methyl-2- (pyridin-3-yloxy)-phenyl]-urea Step 1: 3- (4-Methyl-2-nitro-phenoxy)-pyridine To a stirred solution of 1-chloro-4-methyl-2- nitrobenzene (686 mg; 4.0 mmol) and pyridin-3-ol (418 mg; 4.40 mmol) in dimethylforamide (5 mL) was added potassium carbonate (1.22 g, 8.80 mmol). The reaction was heated to 50 °C and stirred for 12 hours. The reaction was cooled to room temperature, diluted with 50 mL of ethyl acetate and washed with aqueous 10% sodium carbonate (1 x 50 mL), and brine (50 mL), then dried (MgS04), and filtered. The crude product was purified using the biotage 40M cartridge eluting with hexanes and ethyl acetate (1: 1) to yield a light yellow oil (27% yield).

Step 2: 5-Methyl-2-(pyridin-3-yloxy)-phenylamine To a stirred, cooled (about 0 °C) solution of 3- (4- Methyl-2-nitro-phenoxy)-pyridine (1.0 mmol) in methanol (2 mL) and saturated aqueous ammonium chloride (lmL) was added zinc dust (2.0 mmol). After stirring for 12 hours, the reaction was diluted with 30 mL of ethyl acetate and washed with 30 mL of 10% aqueous sodium carbonate (2 x 30 mL), brine (1 x 30 mL), then dried (MgS04), and filtered. The filtered solution was concentrated under reduced pressure to yield a brown oil (95% yield).

Step 3: Urea formation according to method for compound 295 (45% yield).'H NMR (400 MHz, CDC13) 8 11.49 (br s, 1H), 8.55 (s, lH), 8.39 (d, 1H), 8. 35 (s, 1H), 8.15 (s, 1H), 8.05 (br s, 1H), 7.21 (m, 2H), 6.92 (m, 2H), 2.49 (s, 3H), 2.45 (s, 3H).

LRMS (ESI, Positive) m/e 335.91 (M+1).

Compound 309: 1- (5-Methyl-pyrazin-2-yl)-3- [5-methyl-2- (pyridin-2-yloxy)-phenyl]-urea Step 1 : 2- (4-Methyl-2-nitro-phenoxy)-pyridine To a stirred solution of 1-chloro-4-methyl-2- nitrobenzene (686 mg; 4.0 mmol) and pyridin-2-ol (418 mg; 4.40 mmol) in dimethylforamide (5 mL) was added potassium carbonate (1.22 g, 8.80 mmol). The reaction was heated to 50 °C and stirred for 12 hours. The reaction was cooled to room temperature, diluted with 50 mL of ethyl acetate and washed with aqueous 10% sodium carbonate (1 x 50 mL), and brine (50 mL), then dried (MgS04), and filtered. The crude product was purified using the biotage 40M cartridge eluting with hexanes and ethyl acetate (1: 1) to yield a light yellow oil (11% yield).

Step 2: 5-Methyl-2-(pyridin-2-yloxy)-phenylamine To a stirred, cooled (about 0 °C) solution of 2- (4-Methyl-2-nitro-phenoxy)-pyridine (1.0 mmol) in methanol (2 mL) and saturated aqueous ammonium chloride (ImL) was added zinc dust (2.0 mmol). After stirring for 12 hours, the reaction was diluted with 30 mL of ethyl acetate and washed with 30 mL of 10% aqueous sodium carbonate (2 x 30 mL), brine (1 x 30 mL), then dried (MgS04), and filtered. The filtered solution was concentrated under reduced pressure to yield a white foam (77% yield).

Step 3: Urea formation according to method for compound 295 (43% yield).'H NMR (400 MHz, CDC13) 5 8. 51 (br s, 1H), 8.42 (br s, 1H), 8.00 (s, 1H), 7.80 (s, 1H), 7.51 (t, 1H), 7.29 (d, 1H), 7.05 (d, 1H), 6.95 (d, 1H), 6.75 (d, 1H), 6. 35 (t, 1H), 2. 49 (s, 3H), 2.45 (s, 3H). LRMS (ESI, Positive) m/e 335.91 (M+1).

Substituted Aminopvrazine Ureas. General Procedure: To a 0.3 M stirred solution of the aminopyrazine derivative (1 equiv.) in dichloroethane at room temperature under nitrogen was added 2-methoxy-5-methylphenylisocyanate (1 equiv.). The reaction was warmed to 80 °C overnight and then cooled to room temperature. In most cases, the product precipitated and was isolated by filtration. Alternatively the product could be isolated by silica gel chromatography using EtOAc/hexane or CH2CIz/MeOH as eluant.

Compound 310 : 3-(2-Methoxy-5-methyl-phenyl)-1-methyl-1-pyrazin-2-yl-urea Step 1: 2-methylaminopyrazine. To a stirred solution of 2M methylamine in 1 mL of methanol, at room temperature, was added 2-chloropyrazine. The reaction was sealed and heated to 60 °C for 24 hours. The reaction was concentrated to a mixture of starting material and the desired 2- methylaminopyrazine in a 1: 2 ratio. The material was used crude in a urea forming reaction.'H-NMR (400 MHz, CDC13) 8 7.97 (d, 1H), 7.85 (s, 1H), 7.73 (d, 1H), 2.96 (s, 3H) Step 2: To a 0.3 M stirred solution of 2-methylaminopyrazine (1 equiv.) in dichloroethane at room temperature under nitrogen was added 2-methoxy-5-methylphenylisocyanate (1 equiv.). The reaction was warmed to 80 °C overnight and then cooled to room temperature. In most cases, the product precipitated and was isolated by filtration. Alternatively the product could be isolated by silica gel chromatography using EtOAc/hexane or CH2CI2/MeOH as eluant.'H-NMR (400 MHz, CDC13) 8 8.57 (s, 1H), 8.32 (s, 1H), 8. 31 (s, 1H), 8.17 (s, 1H), 6.80 (m, 2H), 3.90 (s, 3H), 3.56 (s, 3H), 2. 32 (s, 3H).

LRMS (ESI, Positive) m/e 273. 2 (M+l).

Compound 311 : 1- (2-Methoxy-5-methyl-phenyl)-3- (5-methyl-pyrazin-2-yl)-urea Prepared according to general procedure described for compound 310, using 2-amino-4- methylpyrazine.'H-NMR (400 MHz, CDC) 8 11.12 (br s, 1H), 8.28 (s, 1H), 8.17 (s, 1H), 8.09 (s, 1H), 6.81 (m, 2H), 3.91 (s, 3H), 2.54 (s, 3H), 2. 35 (s, 3H). LRMS (ESI, Positive) m/e 273.2 (M+1).

Compound 312: 1- (5, 6-Dimethyl-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea Step 1: 2-Amino-5, 6-dimethylpyrazine. Glycine amidine dihydrobromide (620 mg, 2.64 mmol) was stirred in 6 mL of MeOH at-30° C (acetonitrile/CO2 bath) in a capped flask. Butanedione (232 iL, 2.64 mmol) was stirred separately in 6 mL H20 with sodium acetate (700 mg) until homogeneous. The diketone was added to the amidine solution by pipet followed by 2.5 mL of 3.6 M NaOH. The yellow solution was allowed to warm slowly to RT and was then stirred overnight. MeOH was removed by rotovap and the aqueous solution extracted 3 x 30 mL with EtOAc. The combined organic extracts were dried (MgS04), filtered and concentrated to a yellow solid which contained some impurities. The solid was triturated with EtOAc/Et2O and filtered to give pure compound (55 mg, 17%).'H-NMR (400 MHz, CDCI3) 8 7.76 (s, 1H), 4.25 (br s, 2H), 2.40 (s, 3H), 2.37 (s, 3H) Step 2: Prepared according to general procedure described for compound 310, using 2-amino-5, 6- dimethylpyrazine.'H-NMR (400 MHz, CDC13) 8 11.43 (br s, 1H), 8.23 (s, 1H), 8.00 (s, 1H), 7.64 (br s, 1H), 6.81 (m, 2H), 3.95 (s, 3H), 2.59 (s, 3H), 2.52 (s, 3H), 2. 35 (s, 3H). LRMS (APCI, Positive) m/e 287.1 (M+1).

Compound 313: 1- (2-Methoxy-5-methyl-phenyl)-3- (5-trifluoromethyl-pyrazin-2-yl)-urea Step 1: 2-amino-5-trifluoromethylpyrazine. Prepared according to the method of Miesel, J. U. S. patent4,293,552 (1981), Step2: Prepared according to general procedure described for compound 310, using 2-amino-5- trifluoromethylpyrazine.'H-NMR (400 MHz, d6-DMSO) 8 10.59 (s, 1H), 9.78 (br s, 1H), 9.06 (s, 1H), 8.80 (s, 1H), 8.00 (s, 1H), 6.96 (d, 1H), 6.81 (d, 1H), 3.87 (s, 3H), 2.22 (s, 3H). LRMS (ESI, Positive) m/e 327.1 (M+1).

Compound314: 1- (5, 6-Diphenyl-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea Step 1: 2-Hydroxy-5,6-diphenylpyrazine. To a stirred suspension of glycinamide hydrochloride (1.1 gm, 10 mmol) in 20 mL MeOH at 0 °C was added 20% NaOH (10 mL, 50 mmol). A clear solution formed and was treated slowly portionwise with benzil (2.1 gm, 10 mmol) as a solid. The yellow solution was stirred at 0 °C for 2 hours and then neutralized to approximately pH=7 with concentrated HCI. The bright yellow color disappeared and a tan precipitate formed. The material was isolated by filtration with MeOH and triturated with EtOAc to give 2-hydroxy-5,6-diphenylpyrazine (2 gm, 80%).

'H-NMR (400 MHz, CDC13) 8 8.24 (s, 1H), 7.42-7.31 (m, 4H), 7. 39-7. 21 (m, 6H).

Step 2: 2-Chloro-5, 6-diphenylpyrazine. A stirred solution of 2-Hydroxy-5, 6-diphenylpyrazine (430 mg, 1.7 mmol) in 5.2 mL POC13 in a capped reaction vial was heated to 100 °C for 4 hours. The orange solution was cooled to room temperature and stirred rapidly in a mixture of CH2CI2 (100 mL) and ice cold 10% Na2CO3 (100 mL) for 15 minutes. The organic layer was isolated and washed 2 x 100 mL with 10% Na2CO3. The organics were isolated, dried (MgS04), filtered and concentrated to the chloropyrazine, which existed as white solid (450 mg, quantitative).'H-NMR (400 MHz, CDC13) 8 8.59 (s, 1H), 7.45-7.39 (m, 4H), 7.36-7.24 (m, 6H).

Step 3: 2-Azido-5,6-diphenylpyrazine. To a stirred solution of 2-Chloro-5, 6-diphenylpyrazine (45 mg, 0.17 mmol) in 500 gL DMF at room temperature under nitrogen was added sodium azide (11 mg, 0.17 mmol) and the reaction was warmed to 100 °C. After stirring overnight, the reaction was cooled to room temperature, diluted with EtOAc (30 mL) and washed 4 x 30 mL with H20 and 1 x 30 mL with saturated NaCl. The organics were islolated, dried (MgSO4), filtered and concentrated to the 2- azidopyrazine, which exists as a yellow solid (45 mg, quantitative).'H-NMR (400 MHz, CDC13) 8 9.73 (s, 1H), 7. 58-7.42 (m, 6H), 7.36-7.23 (m, 4H).

Step 4: 2-Amino-5,6-diphenylpyrazine. To a stirred solution of 2-Azido-5,6-diphenylpyrazine (45 mg, 0.17 mmol) in 50 mL EtOAc at room temperature was added triethylamine (100 pL) followed by Pearlman's Catalyst (50 mg). The suspension was put through a vacuum/purge cycle three times with hydrogen gas and then held under 1 atmosphere of hydrogen for 2 hours. The suspension was then filtered through GF/F filter paper with EtOAc and concentrated. The crude product was eluted through a Biotage 12S column with 1/1 EtOAc/hexane to give pure product, as a clear oil (25 mg, 59%).'H- NMR (400 MHz, CDC13) S 8.04 (s, 1H), 7.42-7.20 (m, 10H), 4.62 (br s, 2H) Step 5: Prepared according to general procedure described for compound 310, using 2-amino-5,6- diphenylpyrazine.'H-NMR (400 MHz, CDC13) 8 8. 34 (s, 1H), 8.13 (s, 1H), 7.80 (s, 1H), 7.46 (d, 2H), 7. 37-7. 23 (m, 10H), 6.81 (d, 1H), 6.66 (d, 1H), 3.17 (s, 3H), 2. 33 (s, 3H) Compound 315: 1- [3-Benzyl-5- (4-methoxy-phenyl)-pyrazin-2-yl]-3- (2-methoxy-5-methyl-phenyl)-urea Prepared according to general procedure described for compound 310, using 2-amino-3-benzyl-4- (4- methoxyphenyl) pyrazine'H-NMR (400 MHz, CDC13) 5 8.51 (s, 1H), 8.11 (s, 1H), 7.96 (d, 2H), 7. 34 (m, 5H), 7.03 (d, 2H), 6.80 (m, 2H), 4.28 (s, 2H), 3.95 (s, 3H), 3.90 (s, 3H), 2.30 (s, 3H). LRMS (ESI, Positive) m/e 477.2 (M+1).

Compound 316: 1- (6-Azido-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea Step 1 : Tetrazololl, 5-a] pyrazin-5-ylamine. Prepared according to the method of Shaw, J. T.; et al. J.

Heterocyclic Chem. 1980, 17,11.

Step 2: Prepared using p-nitrophenyl carbamate general procedure described for compound 166 (step 2) using Tetrazolo [1, 5-a] pyrazin-5-ylamine.'H-NMR (400 MHz, CDC13) 8 9.72 (br s, 1H), 8.23 (s, 1H), 7.97 (s, 1H), 7.87 (s, 1H), 7.80 (br s, 1H), 6.88 (d, 1H), 6.80 (d, 1H), 3.84 (s, 3H), 2. 36 (s, 3H).

LRMS (ESI, Positive) m/e 300.0 (M+1).

Compound 317: 1-(6-Amino-pyrazin-2-yl)-3-(2-methoxy-5-methyl-phenyl)-urea To a stirred solution of 1-(6-azido-pyrazin-2-yl)-3-(2-methoxy-5-methyl-phenyl)-urea (8 mg, 27 gmol) 95% EtOH (2mL) at room temperature was added concentrated NH40H (10 gL) and 10% Pd on C (25 mg). The suspension was put through a vacuum/purge cycle three times with hydrogen gas and then held under 50 psi of hydrogen pressure and shaken on a Parr Shaker. After 2 hours the vacuum/purge cycle was repeated and the reaction held under hydrogen for another 2 hours. The suspension was then filtered through GF/F filter paper with EtOH and concentrated to a yellow film (3 mg, 41%).'H-NMR (400 MHz, CDC13) 8 8.19 (s, 1H), 7.61 (s, 1H), 7.56 (s, 1H), 6.83 (s, 2H), 3.95 (s, 3H), 2.33 (s, 3H).

LRMS (ESI, Positive) m/e 274.2 (M+1).

Compound 318: 1- (6-Chloro-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea To a stirred solution of 2-amino-6-chloropyrazine (130 mg, 1 mmol) in 3 mL THF at 0 °C under nitrogen was added methyl magnesium iodide (3M in Et2O, 330 pL, 1 mmol) to give a yellow suspension that was stirred at 0 °C for 15 minutes. The suspension was treated with the isocyanate neat (147 zL, 1 mmol) and allowed to warm to room temperature overnight. The reaction was partitioned between EtOAc (30 mL) and 10% Na2CO3 (30 mL). The organics were isolated and washed 1 x 30 mL with 10% Na2CO3 and 1 x 30 mL with saturated NaCI. The organics were dried (MgSO4), filtered and concentrated to a crude residue that was triturated with EtOAc to give, after filtration, the urea product as a white solid (27 mg, 9%).'H-NMR (400 MHz, CDC13) 8 8. 26 (s, 1H), 8.23 (s, 1H), 8.17 (s, 1H), 8.09 (br s, 1H), 6.84 (d, 1H), 6.81 (d, 1H), 3.96 (s, 3H), 2. 35 (s, 3H). LRMS (ESI, Positive) m/e 293.0 (M+1).

Compound 319: 1- (5-Bromo-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea Step 1: 2-amino-5-bromopyrazine. To a stirred, cooled (0°C) solution of amino pyrazine (5.0 g, 52.6 mmol) in methylene chloride (200 mL) was added N-bromosuccinimide (9.39 g, 52.8 mmol). After stirring for 24 hours, the reaction was washed with aqueous 10% sodium carbonate (3 x 50 mL), water (50 mL), then dried (MgS04), and filtered. The filtered material was concentrated under reduced pressure, taken up in minimal ethyl acetate (5 mL) followed by hexanes (200 mL). Yellow crystals formed which were filtered and dried. (56 % yield).

Step 2: Prepared according to general procedure described for compound 310, using 2-amino-4- bromopyrazine.'H-NMR (400 MHz, CDC13/CD30D) 8 8.55 (s, 1H), 8. 32 (s, 1H), 8.03 (s, 1H), 6.81 (m, 2H), 3.92 (s, 3H), 2. 34 (s, 3H).

Compound 320: 1- (3, 5-Dibromo-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea Prepared according to general procedure described for compound 310, using 2-amino-4, 6- dibromopyrazine.'H-NMR (400 MHz, CDC13) 8 7.98 (s, 1H), 7.13 (s, 1H), 6.79 (m, 2H), 3.83 (s, 3H), 2. 32 (s, 3H) Compound 321: 1- [5- (1, 3-Dioxo-1, 3-dihydro-isoindol-2-ylmethyl)-pyrazin-2-yl]-3- (2-methoxy-5-methyl-phenyl)-urea Step 1: (5-Bromomethyl-pyrazin-2-yl)-carbamic acid tert-butyl ester. To a stirred solution of 2- Boc-amino-5-methyl pyrazine (1. 34 gm, 6.4 mmol), in 20 mL CC14 at room temperature under nitrogen was added N-bromosuccinimide (1.14 gm, 6.4 mmol) followed by benzoyl peroxide (125 mg). The solution was irradiated with a 100 watt flood lamp, which caused the reaction to reflux vigorously.

After 2 hours, the reaction was cooled to room temperature, diluted to 125 mL with CH2C12 and washed 1 x 125 mL with 10% sodium bisulfite solution and 1 x 125 mL with saturated NaCI. The organics were dried (MgS04), filtered and concentrated to a brown oil, which was directly loaded onto a Biotage 40S column with CH2C12 and eluted with 15/85 EtOAc/hexane to give the desired benzylic bromide as a yellow solid (954 mg, 51%).'H-NMR (400 MHz, CDC13) 8 9.22 (s, 1H), 8.29 (s, 1H), 7. 37 (br s, 1H), 4.54 (s, 2H), 1.55 (s, 9H).

Step 2: [5- (1, 3-Dioxo-1, 3-dihydro-isoindol-2-ylmethyl)-pyrazin-2-yll-carbamic acid tert-butyl ester. To a stirred solution of phthalimide (971 mg, 6.6 mmol) and powdered K2CO3 (1.37 gm, 9.9 mmol) in acetonitrile (9.9 mL) at room temperature under nitrogen was added the bromide (954 mg, 3.3 mmol) as a solid. The suspension was heated to 65o C for 4 hours. After cooling to room temperature the reaction was partitioned between EtOAc (60 mL) and H2O (60 mL). The organics were isolated and washed 2 x 50 mL with H ? O and 1 x 50 mL with saturated NaCI. The organics were dried (MgS04), filtered and concentrated. The crude product was triturated with CH2CI2 and filtered to remove solid excess phthalimide and the filtrated partially concentrated and loaded directly onto a Biotage 40S column and eluted with 3/7 EtOAc/hexane to give the desired phthalimide as a white solid (495 mg, 42%).'H-NMR (400 MHz, CDC13) 8 9.17 (s, 1H), 8.23 (s, 1H), 7.85 (m, 2H), 7.71 (m, 2H), 7.39 (br s, 1H), 4.97 (s, 2H), 1.52 (s, 9H) Step 3: 2- (5-Amino-pyrazin-2-ylmethyl)-isoindole-1, 3-dione. To a stirred solution of the phthalimide (495 mg, 1.4 mmol) in 7 mL CH2CI2 at room temperature in a capped flask was added trifluoroacetic acid (7 mL). After stirring overnight, the reaction was concentrated to remove excess trifluoroacetic acid and was then dissolved in 200 mL 10/1 CH2Cl2/MeOH, stirred rapidly, and treated with a solution of 10% Na2CO3 (200 mL). The organics were isolated, dried (MgS04), filtered and concentrated to give the free aminopyrazine as a yellow solid (260 mg, 73%).'H-NMR (400 MHz, CDC13) 8 8.25 (s, 1H), 7.85 (m, 2H), 7.77 (s, 1H), 7.74 (m, 2H), 4.83 (s, 2H) Step 4: Prepared according to general procedure described for compound 310 using 2- (5-Amino- pyrazin-2-ylmethyl)-isoindole-1, 3-dione.'H-NMR (400 MHz, CDC13) 8 8.38 (s, 1H), 8.29 (s, 1H), 8.03 (s, 1H), 7.86 (m, 2H), 7.76 (m, 2H), 6.81 (m, 2H), 4.98 (s, 2H), 3.91 (s, 3H), 2. 31 (s, 3H). LRMS (ESI, Positive) m/e 418.1 (M+1).

Compound 322: 1- (5-Aminomethyl-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea To a stirred solution of 1- [5- (1, 3-dioxo-1, 3-dihydro-isoindol-2-ylmethyl)-pyrazin-2-yl]-3- (2-methoxy- 5-methyl-phenyl)-urea (16 mg, 38 gmol) in 380 uL 95% EtOH and 100 uL DMF at room temperature in a capped reaction vial was added hydrazine monohydrate (3.8 pL, 76 tmol). After stirring overnight at room temperature, a white precipitate formed. The precipitate was filtered off, dried and triturated with EtOAc to remove phthalimide based impurities to provide the product as a white solid (7.9 mg, 72%).'H-NMR (400 MHz, d6-DMSO) 5 9.54 (s, 1H), 8.80 (s, 1H), 8.46 (s, 1H), 8.02 (s, 1H), 6.93 (d, 1H), 6.79 (d, 1H), 4.51 (d, 2H), 4.39 (br s, 2H), 3.89 (s, 3H), 2.23 (s, 3H). LRLCMS (ESI, Positive) m/e 288.2 (M+1).

Compound 323: 1- (2-Methoxy-5-methyl-phenyl)-3- (6-methoxy-pyrazin-2-yl)-urea Step 1: 2-anino-6-methoxypyrazine. To a stirred solution of methanol (89 IL ; 2.2 mmol) in dioxane (1 mL) was added sodium hydride (53 mg; 2.2 mmol). After stirring for 30 minutes, 2-amino-6- chloropyrazine (258 mg; 2.0 mmol) was added and the reaction was heated to 90 °C. After stirring for 12 hours, the reaction was cooled to room temperature, diluted with 30 mL of ethyl acetate and washed with aqueous 10% sodium carbonate (1 x 30 mL), and brine (30 mL), then dried (MgS04), and filtered.

The crude product was purified using the Biotage 12i cartridge eluting with hexane and ethyl acetate (3: 1) to yield a white solid (11% yield). Step 2: Prepared according to general procedure described for compound 310 using 2-amino-6- methoxypyrazine. (8% yield).'H NMR (400 MHz, CDC13) 8 8.15 (s, 1H), 8.05 (br s, 1H), 7.92 (s, 1H), 7.82 (s, 1H), 6.89 (d, 1H), 6.80 (d, 1H), 4.05 (s, 3H), 3.81 (s, 3H), 2.38 (s, 3H). LRMS (ESI, Positive) m/e 289.10 (M+1).

Compound324: 1- (6-Benzyloxy-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea Step 1: 2-amino-6-benzyloxypyrazine. To a stirred solution of benzyl alcohol (432 uL ; 4.0 mmol) in dioxane (2 mL) was added sodium hydride (96 mg; 4.0 mmol). After stirring for 30 minutes, 2-amino- 6-chloropyrazine (258 mg; 2.0 mmol) was added and the reaction was heated to 90 °C. After stirring for 12 hours, the reaction was cooled to room temperature, diluted with 30 mL of ethyl acetate and washed with aqueous 10% sodium carbonate (1 x 30 mL), and brine (30 mL), then dried (MgSO4), and filtered. The crude product was purified using the biotage 12i cartridge eluting with hexane and ethyl acetate (3: 1) to yield a white solid (33% yield). Step 2: Prepared according to general procedure described for compound 310 using 2-amino-6- benzyloxypyrazine. (34% yield).'H NMR (400 MHz, d6-DMSO) 8 9.99 (s, 1H), 9.18 (s, 1H), 8.62 (s, 1H), 7.99 (s, 1H), 7.95 (s, 1H), 7.52 (d, 2H), 7.41 (m, 3H), 6.92 (d, 1H), 6.80 (d, 1H), 5.39 (s, 2H), 3.80 (s, 3H), 2.21 (s, 3H). LRMS (ESI, Positive) m/e 365.10 (M+1).

Compound325: 1- (2-Methoxy-5-methyl-phenyl)-3- (5-methoxy-pyrazin-2-yl)-urea To a stirred solution of 1- (5-bromo-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea (47 mg; 0.14 mmol) in N-methyl pyrrolidinone (300 (iL) was added sodium methoxide (0.5 mmol). The reaction was heated to 100 °C. After stirring for 12 hours, the reaction was cooled to room temperature, diluted with 30 mL of ethyl acetate and washed with aqueous 10% sodium carbonate (1 x 30 mL), brine (30 mL), then dried (MgS04), and filtered. The crude product was purified using a 0.5 mm prep plate eluting with hexane and ethyl acetate (1: 1) to yield a yellow solid (13% yield).'H NMR (400 MHz, CDC13) 5 8.12 (s, 1H), 7.99 (s, 1H), 7.91 (s, 1H), 6.80 (dd, 2H), 3.95 (s, 3H), 3.89 (s, 3H), 2.38 (s, 3H).

LRMS (ESI, Positive) m/e 289.10 (M+I).

Compound 326: 1- (5-Ethynyl-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea Stepl : 2-Amino-5-alkynylpyrazine. To a stirred solution of 5-bromo-2-aminopyrazine (432 mg; 2.5 mmol), Pd (Ph3P) 2CI2 (91 mg; 0.13 mmol), Cul (1.2 g, 6.5 mmol) in triethylamine (8 mL) was added TMS-acetylene. The reaction was stirred at 60 °C for 12 hours. The reaction was cooled to room temperature, diluted with 30 mL of ethyl acetate and washed with aqueous 10% sodium carbonate (1 x 30 mL), brine (30 mL), then dried (MgSO4), and filtered. The crude product was diluted in 1 mL of methanol and sodium hydroxide (10 mL of a 1N aqueous solution). After stirring for 12 hours the reaction was diluted with 30 mL of ethyl acetate and washed with aqueous 10% sodium carbonate (1 x 30 mL), brine (30 mL), then dried (MgS04), and filtered. The crude product was purified using a biotage 12L column eluting with methylene chloride and methanol (98: 2) to yield an off white solid (40% yield).

Step 2 : Prepared according to general procedure described for compound 310 using 2-Amino-5- alkynylpyrazine. (20% yield).'H NMR (400 MHz, 6-DMSO) 8 10. 25 (s, 1H), 9.80 (br s, 1H), 8.90 (s, 1H), 8.50 (s, 1H), 8.00 (s, 1H), 6.95 (d, 1H), 6.82 (d, 1H), 4.42 (s, 1H), 3.82 (s, 3H), 2.22 (s, 3H).

LRMS (ESI, Positive) m/e 283.10 (M+1).

Compound 327 : 1- (5-Ethyl-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea Step 1: 2-Amino-5-ethylpyrazine. To a stirred solution of 5-ethynyl-2-aminopyrazine (18 mg; 0.151 mmol), in ethyl acetate (500 pL) was added triethylamine (63 gL ; 0.45 mmol) and Pd (OH) 2 (0. 01 mmol ; 20% wt on carbon). The reaction was placed under a hydrogen atmosphere at 45 psi and shook for 6 hours. The reaction was filtered and concentrated under reduced pressure to yield an off white solid (84% yield).

Step 2 : Prepared according to general procedure described for compound 310 using 2-Amino-5- ethylpyrazine. (27% yield).'H NMR (400 MHz, CDC13) 8 8.65 (s, 1H), 8.38 (s, 1H), 8.18 (s, 1H), 8.08 (s, 1H), 6.80 (dd, 2H), 3.92 (s, 3H), 2.81 (q, 2H), 2.39 (s, 3H), 1.39 (t, 3H). LRMS (ESI, Positive) m/e 287.21 (M+1).

Compound 328: 1- (5-Cyano-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea Step 1: 2-amino-5-cyanopyrazine. To a stirred solution of 5-bromo-2-aminopyrazine (1.0 g, 5.8 mmol), Cul (2.76 g, 14.5 mmol), 18-crown-6 (121 mg; 0.46 mmol), potassium cyanide (943 mg; 14.5 mmol) in dimethylformamide (20 mL) was added Pd (PPh3) 4 (196 mg; 0.17 mmol). After stirring at room temperature for 20 minutes the reaction was placed in an oil bath at 155 °C for 2 hours. The reaction was allowed to cool to room temperature and then poured into chloroform (300 mL). A precipitate formed that was filtered and triturated with hexanes to yield an off white solid (60% yield).

Step 2 : Prepared according to general procedure described for compound 310 using 2-amino-5- cyanopyrazine. (30% yield).'HNMR (400 MHz, d6-DMSO) 8 8.89 (s, 1H), 8.79 (s, 1H), 8.05 (s, 1H), 6.91 (d, 1H), 6.80 (d, 1H), 3.85 (s, 3H), 2.22 (s, 3H). LRMS (ESI, Positive) m/e 283.91 (M+1).

Compound 329: 1- (5-Benzoyl-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea Step 1: 5-Benzoyl-pyrazine-2-carboxylic acid. To a stirred, cooled (0 °C) solution of 2-pyrazine carboxylic acid (3.0 g, 24.2 mmol) and benzaldehyde (7.4 mL; 73 mmol) in a 50% aqueous solution of sulfuric acid (40 mL) and 25 mL of acetic acid was added FeS04 7 H20 (20.3 g, 73 mmol dissolved in 50 mL of water) and t-butyl peroxide (9.2 mL ; 73 mmol) simultaneously. After stirring for 1 hour, the reaction was treated with 200 mL of water. A precipitate formed which was filtered and washed with methylene chloride (3 x 100 mL) to yield a tan solid (36% yield).

Step 2: 1- (5-Benzoyl-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea. To a stirred solution of 5- Benzoyl-pyrazine-2-carboxylic acid (912 mg; 4.0 mmol) and triethylamine (584) iL ; 4.2 mmol) in toluene (12 mL) was added diphenyl phosphoryl azide (860 gL ; 4.0 mmol). The reaction was stirred for 30 minutes followed by the addition of t-butanol (764 pL, 8.0 mmol). The reaction was heated to 90 °C and stirred for 3 hours. The reaction was cooled to room temperature, diluted with 30 mL of ethyl acetate and washed with aqueous 10% sodium carbonate (1 x 30 mL), brine (30 mL), then dried (MgS04), and filtered. The material was purified using a biotage 40M cartridge eluting with hexane and ethyl acetate (1: 1) to yield an off white solid (14% yield).'H NMR (400 MHz, CDC13) 8 9.41 (s, 1H), 9.01 (s, 1H), 8.45 (s, 1H), 8.18 (s, 1H), 8.08 (d, 2H), 7.61 (t, 1H), 7.52 (t, 2H), 6.90 (d, 1H), 6.82 (d, 1H), 3.92 (s, 3H), 2.39 (s, 3H). LRMS (ESI, Positive) m/e 363.21 (M+1).

Compound 330: 1- [5- (Hydroxy-phenyl-methyl)-pyrazin-2-yl]-3- (2-methoxy-5-methyl-phenyl)-urea To a stirred solution of 1- (5-benzoyl-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea (22 mg; 0.061 mmol) in methanol (1 mL) was added sodium borohydride (10 mg; 0.3 mmol). After stirring for 12 hours, the reaction diluted with 30 mL of ethyl acetate and washed with aqueous 10% sodium carbonate (1 x 30 mL), brine (30 mL), then dried (MgS04), and filtered. The filtered material was concentrated under reduced pressure to yield a white solid (91% yield). lH NMR (400 MHz, CDC13) 8 8.48 (br s, 1H), 8.39 (s, 1H), 8.22 (s, 1H), 8.12 (s, 1H), 7.25-7.45 (m, 5H), 6.89 (d, 1H), 6.80 (d, 1H), 5.85 (d, 1H), 3.88 (s, 3H), 2.37 (s, 3H). LRMS (ESI, Positive) m/e 365.24 (M+1).

Compound 331 : Suzuki procedure 1-(2-Methoxy-5-methyl-phenyl)-3-(6-phenyl-pyrazin-2-yl)-urea Step 1: To a stirred solution of 2-amino-6-chloro pyrazine (400 mg; 3.1 mmol) and phenyl boronic acid (415 mg; 3.4 mmol) in dioxane (6 mL) and ethanol (3 mL) was added cesium carbonate (2.28 g, 7.0 mmol in 3 mL of water) followed by Pd (PPh3) 4 (185 mg; 0.16 mmol). The reaction was heated to 75 °C and stirred for 12 hours. The reaction was cooled to room temperature, diluted with 50 mL of ethyl acetate and washed with aqueous 10% sodium carbonate (1 x 50 mL), brine (50 mL), then dried (MgS04), and filtered. The material was purified using a biotage 40M cartridge eluting with ethyl acetate to yield an off white solid (84% yield).

Step 2: Prepared according to general procedure described for compound 310 using 2-amino-6- phenylpyrazine. (33% yield).'H NMR (400 MHz, CDC13) 8 11. 11 (br s, 1H), 8.612 (s, 1H), 8.29 (s, 1H), 8. 24 (s, 1H), 8. 14 (s, 1H), 8.03 (m, 2H), 7.51 (m, 3H), 6.87 (d, 1H), 6.77 (d, 1H). LRMS (ESI, Positive) m/e 355.6 (M+1).

Compound332: 1- (3-Bromo-5-phenyl-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea Step 1: 2-amino-3-bromo-5-phenylpyrazine. To a stirred solution of 3,5-dibromo-2-aminopyrazine (200 mg; 0.79 mmol) and phenyl boronic acid (106 mg; 0.87 mmol) in dioxane (4 mL) and ethanol (2 mL) was added cesium carbonate (571 mg; 1.75 mmol in 2 mL of water) followed by Pd (PPh3) 4 (46 mg; 0.04 mmol). The reaction was heated to 75 °C and stirred for 12 hours. The reaction was cooled to room temperature, diluted with 50 mL of ethyl acetate and washed with aqueous 10% sodium carbonate (1 x 50 mL), brine (50 mL), then dried (MgS04). and filtered. The material was purified using a biotage 12L cartridge eluting with hexanes and ethyl acetate (3: 1) to yield an off white solid (88% yield).

Step 2: Prepared according to general procedure described for compound 310 using 2-amino-3-bromo- 5-phenylpyrazine. (18% yield).'H NMR (400 MHz, CDC13) 8 8. 36 (s, 1H), 8.09 (s, 1H), 7.63 (d, 2H), 7.59 (m, 3H), 6.85 (dd, 2H), 3.92 (s, 3H), 2.39 (s, 3H). LRMS (ESI, Positive) m/e 413. 2 415.2 (M+l).

Compound 333 : 1- (2-Methoxy-5-methyl-phenyl)-3- (5-phenyl-pyrazin-2-yl)-urea Step 1: 2-amino-5-phenylpyrazine. To a stirred solution of 3-bromo-5-phenyl-2-amino pyrazine (80 mg; 0. 32 mmol), in ethyl acetate (1 mL) was added triethylamine (139 pL ; 1.0 mmol) and Pd (OH) 2 (10 mg; 20% wt on carbon). The reaction was placed under a hydrogen atmosphere at 45 psi and shook for 6 hours. The reaction was filtered and concentrated under reduced pressure. The product was purified using a biotage 12L eluting with ethyl acetate to yield an off white solid (75% yield).

Step 2: Prepared according to general procedure described for compound 310 using 2-amino-5- phenylpyrazine. (25% yield).'H NMR (400 MHz, CDC13) 8 8.36 (s, 1H), 8. 22 (s, 1H), 8.18 (s, 1H), 7.63 (m, 2H), 7.59 (m, 3H), 7.28 (br s, 1H), 6.82 (m, 2H), 3.92 (s, 3H), 2. 33 (s, 3H). LRMS (ESI, Positive) m/e 335.21 (M+1).

Compound 334 : 1- (2-Methoxy-5-methyl-phenyl)-3-quinoxalin-2-yl-urea Step 1: To 2-chloroquinoxaline (1.0 g, 6 mmol) was added ammonia in methanol (8 mL of a 2M solution). The reaction was sealed in a vial, heated to 80 °C, and stirred for 12 hours. The reaction was cooled to room temperature and concentrated under reduced pressure. The residue was taken up in methylene chloride and filtered. Hexane was added until a precipitate formed which was filtered and found to be the desired product (5% yield).

Step 2: Prepared according to general procedure described for compound 310 using Quinoxalin-2- ylamine. (26% yield).'H NMR (400 MHz, D6-DMSO) 8 11.63 (br s, 1H), 10.59 (br s, 1H), 8. 80 (s, 1H), 8. 15 (s, 1H), 7.97 (d, 1H), 7.81 (m, 2H), 7.64 (m, 1H), 6.98 (d, 1H), 6.82 (d, 1H), 3.97 (s, 3H), 2.23 (s, 3H). LRMS (ESI, Positive) m/e 309.4 (M+1).

Compound 335 : 1- (3, 6-Dimethyl-pyrazin-2-yl)-3- (2-methoxy-5-methyl-phenyl)-urea Step 1 : 2-Azido-3,6-dimethylpyrazine. To a stirred solution of 2-chloro-3,5-dimethyl pyrazine (1.0 mL; 8.3 mmol) in dimethylformamide (10 mL) was added sodium azide (539 mg; 8. 3 mmol). The reaction was heated to 100 °C and stirred for 12 hours. The reaction was cooled to room temperature, diluted with 50 mL of ethyl acetate and washed with aqueous 10% sodium carbonate (1 x 50 mL), brine (50 mL), then dried (MgS04), and filtered. The material was purified using a biotage 12L cartridge eluting with hexanes and ethyl acetate (3 : 1) to yield an off white solid (42% yield).

Step 2: 2-Amino-3, 6-dimethylpyrazine. To a stirred solution of 3-azido-2, 5-dimethyl pyrazine (100 mg; 0.66 mmol) in methanol (800 L) was added 12N HCI (100 liL) and tin chloride dihydrate (149 mg; 0.66 mmol). The reaction was heated to 60 °C and stirred for 12 hours. The reaction was cooled to room temperature, diluted with 50 mL of ethyl acetate and washed with aqueous 10% sodium carbonate (1 x 50 mL), brine (50 mL), then dried (MgSO4), and filtered. The material was purified using a biotage 12i cartridge eluting with ethyl acetate to yield an off white solid (38% yield).

Step 3: Prepared according to general procedure described for compound 310 using 2-Amino-3,6- dimethylpyrazine. (15% yield).'H NMR (400 MHz, CDC13) 8 8. 22 (br s, 1H), 8.01 (s, 1H), 6.82 (m, 2H), 3.92 (s, 3H), 2.59 (s, 3H), 2.55 (s, 3H), 2.38 (s, 3H). LRMS (ESI, Positive) m/e 287. 20 (M+1).

Compound 336 : OMe OMe OMe H H OMe 02N [Step1 02N Step2 H2Nr Step3 lí NN, N} 1-11 % Br WOMe OMe AN ° 9\°Me N Step 1 : 2-Methoxy-4-methoxymethyl-1-nitro-benzene To a 250 mL round bottomed flask containing 5.4 g (39 mmol) of 3-methoxy-4-nitrobenzyl alcohol in 30 mL THF and 30 mL DMF was added, 38 g (117 mmol, 3 eq.) of finely powdered cesium carbonate followed by 24 mL (390 mmol, 10 eq.) iodomethane. The mixture was stirred at room temperature for 18 h, and was then partitioned between 100 mL water and 100 mL diethyl ether. The aqueous phase was extracted with ether (2 x 100 mL), and the combined organic extracts were washed with brine (2 x 50 mL), dried over MgS04, filtered through a short plug of silica, and concentrated. The residue was purified by flash chromatography, eluting with 1: 1 EtOAc-Hexane, to give 6.76 g (88%) of the methyl ether as a yellow oil.'H NMR (400 MHz, CDC13) : 8 7.84, (d, J=8. 2 Hz, 1H) 7.10 (s, 1H), 6.94 (d, J=9. 1 Hz, 1H), 4.51 (s, 2H), 3.98 (s, 3H), 3.44 (s, 3H).

Step 2: 2-Methoxy-4-methoxymethyl-phenylamine. In a 250 mL Parr apparatus, 2.1 g (10.6 mmol) of 2-methoxy-4-methoxymethyl-1-nitro-benzene, in 40 mL of ethanol was hydrogenated at 2 aim over 300 mg of 10% Pd on carbon for 2.5 h. The catalyst was removed by filtration through a glass fiber filter and the filtrate was concentrated to give 1.61 g (91 %) of product as a light yellow oil.'H NMR (400 MHz, CDC13) : S 6.80 (s, 1H), 6.74 (d, J=7. 8 Hz, 1H), 6.67 (d, J=7. 8 Hz), 4.35 (s, 2H), 3.86 (s, 3H), 3.78 (br d, 2H), 3.35 (s, 3H). MS ESI-pos, IvI+l = 168.1 Step 3: 1- (2-Methoxy4-methoxymethyl-phenyl)-3- (5-methyl-pyrazin-2-yl)-urea.

General diphenylphosphoryl azide coupling method: To a solution of 5-methylpyrazine-2- carboxylic acid (365 mg, 2.64 mmol) in 20 mL of anhydrous toluene, was added, diisopropylethylamine (483 aL, 2.77 mmol) and the mixture was stirred at room temperature until the solid dissolved. Then diphenylphosphoryl azide was added and the solution was heated to 90 °C. After 20 minutes N2 evolution had subsided, and the caramel colored reaction mixture was cooled to 60 °C before adding 2-methoxy-4-methoxymethylaniline as a solution in 4 mL toluene. After stirring for 6 hr at 60 °C, the mixture was cooled to room temperature and diluted with 20 mL of 5% NH40H, and extracted with EtOAc (3 x 50 mL). The combined extracts were washed with 20 mL water and 20 mL brine then dried over MgS04, filtered and concentrated. The brown residue was purified by flash chromatography (eluting with 5% MeOH in CH2Cl2) to give 219 mg (27%) of the desired product.'H NMR (400 MHz, CDC13) : 8 11.36 (s, 1H), 9.47, (s, 1H) 8.40, (s, 1H), 8. 31 (s, 1H), 8. 08 (s, 1H), 6.95 (s, 1H), 6.94 (d, J= 7. 8 Hz, 1H), 4.45 (s, 2H), 3.97 (s, 3H), 3.39 (s, 3H), 2.52 (s, 3H). MS ESI-pos M+1=303. 2 Compound 337 : 1- (4-Benzyloxymethyl-2-methoxy-phenyl)-3- (5-methyl-pyrazin-2-yl)-urea Step 1: 4-Benzyloxymethyl-2-methoxy-1-nitro-benzene. To a stirred suspension of finely powdered cesium carbonate (8.0 g, 24.5 mmol) was added 3-methoxy-4-nitrobenzyl alcohol (1.5 g, 8.18 mmol), followed by benzyl bromide, (2 mL, 16.4 mmol). After stirring at room temperature for 18 h, the suspension diluted with 100 mL diethyl ether, and washed with 3 x 50 mL water, then 50 mL brine.

The organic phase was dried over MgS04, filtered through a plug of silica, and concentrated. The resulting orange oil was purified by flash chromatography, eluting with 2: 1 hexane-EtOAc, to give 1.87 g, (84%) of the benzyl ether.'H NMR (400 MHz, CDC13) : 8 7.85 (d, J= 8.2 Hz, 1H) 7.3-7.4 (m, SH), 7.26 (s, 1H), 6.97 (d, J= 8.2 Hz, 1H), 4.61 (s, 2H), 4.59 (s, 2H), 3.96 (s, 3H).

Step 2 : 4-Benzyloxymethyl-2-methoxy-phenylamine. A solution of 4-nitro-3-methoxybenzylbenzyl ether (2.2 g, 8.1 mmol) and ammonium acetate (2.46 g, 32 mmol, 4 eq.) in 30 mL MeOH was stirred at 0 °C and 1.3 g (20 mmol, 2.5 eq.) of zinc dust was added in several portions. After 1 h, the reaction mixture was partitioned between 40 mL water and 40 mL ethyl acetate. The organic phase was dried over MgSO4 and concentrated. The residue was used directly in next step without further purification.

'H NMR (400 MHz, CDC13) : 8 7.35 (m, 5H), 6.82 (s, 1H), 6.77 (d, J= 6. 3 Hz, 1H), 6.67 (d, J= 7.8 Hz, 1H), 4.51 (s, 2H), 4.45 (s, 2H), 3.85 (s, 3H), 3.79 (s, 2H). MS ESI-pos, M+1 = 244.2.

Step 3: 1- (4-Benzyloxymethyl-2-methoxy-phenyl)-3- (5-methyl-pyrazin-2-yl)-urea. Prepared according to the general diphenylphosphoryl azide coupling method described above for compound 336.'H NMR (400 MHz, CDC13) : 8. 11. 36, (s, 1H), 9.23 (s, 1H), 8.39 (s, 1H), 8.32 (d, J= 7. 8 Hz, 1H), 8. 09 (s, 1H), 7. 38m, 5H), 6.97, (s, 2H), 4.56 (s, 4H), 3.96 (s, 3H), 2.53 (s, 3H).). MS APCI- pos, M+1 = 379. 3 Compound 338 : OMe OMe OMe OMe 02N Step I 02N Step 2 02N -- 1 : , H"--ol OMe H H OMe OMe OMe Step3 H2N Boc n Step5 fNh'NN+ H nl Step 4 Step 6 1 0 N _, Jo 1-{4-[(Benzyl-methyl-arnino)-methyl]-2-methoxy-phenyl}-3-(5- methyl-pyrazin-2-yl)-urea Step 1: 3-Methoxy-4-nitro-benzyl bromide. To a 250 mL round bottomed flask containing 10 g (54.6 mmol) of 4-nitro-3-methoxybenzyl alcohol in 30 mL THF was added, 36 g (109 mmol, 2 eq.) of carbon tetrabromide followed by 15.9 g (60 mmol, 1. 1 eq.) triphenylphosphine at 0 °C. The mixture was stirred at 0 °C for 3 hours. Upon removal of the solvent, the residue was purified by flash chromatography, eluting with 10: 90 EtOAc-Hexane, to give 11 g (82%) of the product as a yellow solid.'H NMR (400 MHz, CDC13) : 8 7.86 (s, 1H), 7.84 (m, 1H), 7.12 (s, 1H), 7.07 (m, 1H), 4.47 (s, 2H), 4.00 (s, 3H).

Step 2: N-Benzyl-N-(3-methoxy-4-nitro-benzyl)-amine. To a 150 mL round bottomed flask containing 1.97 g (8.0 mmol) of 3-methoxy-4-nitro-benzyl bromide in 20 mL THF was added, 2.4 g (24 mmol, 3eq.) of triethylamine followed by 2.5 g (24 mmol, 3 eq.) of benzylamine. The mixture was stirred at room temperature for 2 h, and was then partitioned between 50 mL ethyl acetate and brine.

The organic phase was dried over MgS04 and concentrated. The residue was purified by flash chromatography, eluting with 1-4% MeOH in dichloromethane to give 1.6 g (73%) of the benzyl amine as a yellow oil. IH NMR (400 MHz, CDC13) : 8 7.84 (d, J= 8.61 Hz, 1H), 7.34 (m, 5H), 7.16 (s, 1H), 6.99 (d, J= 8.61 Hz, 1H), 3.97 (s, 3H), 3.86 (s, 2H), 3.81 (s, 2H).

Step 3: Benzyl- (3-methoxy-4-nitro-benzyl)-carbamie acid tert-butyl ester. To a 150 mL round bottomed flask containing 0.92 g (3.4 mmol, 1 eq.) of N-benzyl-N-(3-methoxy-4-nitro-benzyl)-amine in 2 mL dichloromethane was added Boc anhydride (0.74 g, 3.4 mmol, 1 eq.) then stirred at room temperature for 18 h. The reaction mixture was then partitioned between 40 mL water and 40 mL ethyl acetate. The organic phase was dried over MgS04 and concentrated. No further purification was necessary.'H NMR (400 MHz, CDC13) : 8 IH NMR (400 MHz, CDC13) : 8 7.81 (d, J= 8.61 Hz, 1H), 7.2-7. 3 (m, 6H), 6.93 (m, 1H), 6.82 (s, 1H), 4.39 (m, 4H), 3.88 (s, 3H), 1.53 (s, 9H).

Steps 4-6: 1-{4-l (Benzyl-methyl-amino)-methyll-2-methoxy-phenyl}-3-(5-methyl- pyrazin-2-yl)- urea. Benzyl- (3-methoxy-4-nitro-benzyl)-carbamic acid tert-butyl ester was reduced to the corresponding aniline according to the general hydrogenation procedure detailed above for compound 336. The crude aniline was used in the coupling step as follows: A solution of 5-methylpyrazine-2- carboxylic acid (34.5 mg, 0.25 mmol) and added triethylamine (28 mg, 0.275 mmol) in 5 mL of anhydrous toluene was stirred at room temperature until the solid dissolved. Diphenylphosphoryl azide (62 mg, 0.225 mmol) was added and the solution heated to 90 °C for 20 min. The reaction pot was then transferred to a 60 °C oil bath, and the aniline (0.25 mmol) was added as a solution in 2 mL toluene.

After stirring for 4.5 hr at 60 °C, the mixture was cooled to room temperature, diluted with EtOAc, washed with sat'd NaHCO3, then brine. The organic phase was dried over MgS04, filtered and concentrated. The resulting residue was purified by preparative TLC, eluting with 5% MeOH in CH2CI2, to give the desired urea. The Boc group was removed by treatment of the Boc protected amine in 15 mL of CH2CI2 with 3 mL TFA and stirring at room temperature for 3h. The mixture was diluted with EtOAc (50 mL), washed with 20 mL of sat'd NaHCO3 followed by 20 mL of brine. The organic phase was then dried over MgS04, filtered and concentrated to give the free amine'H NMR (400 MHz, d6-DMSO) : 8 11. 35 (s, 1H), 9.68 (s, 1H), 8.41 (s, 1H), 8.28 (d, J= 8.21 Hz, 1H), 8.06 (s, 1H), 7.36 (s, 6H), 6.96 (s, 1H), 6.94 (d, J= 8.21 Hz, 1H), 3.95 (s, 3H), 3.84 (s, 2H), 3.81 (s, 2H), 2.52 (s, 3H), 2.13 (s, 1H). MS APCI-pos, M+1 = 377.9 Compound 339 : 1- (2-Methoxy-4-methylaminomethyl-phenyl)-3- (5-methyl-pyrazin-2-yl)-urea Step 1+2: (3-Methoxy-4-nitro-benzyl)-methyl-carbamic acid tert-butyl ester. In a fashion similar to that described above for the analogous benzyl derivative compound 338,3-methoxy-4-nitro-benzyl bromide was alkylated with methylamine, and the resulting secondary amine was protected as the Boc derivative.'H NMR (400 MHz, CDC13) : 6 7. 84 (d, J= 8. 61 Hz, 1H), 6.98 (s, 1H), 6.87 (d, J= 8. 61 Hz, 1H), 4.46 (s, 2H), 3.95 (s, 7H), 2.85 (s, 3H), 1.53 (s, 9H).

Step 3: (4-Amino-3-methoxy-benzyl)-methyl-carbamic acid tert-butyl ester. In a 250 mL Parr apparatus, 0.98 g (3.5 mmol) of (3-methoxy-4-nitro-benzyl)-methyl-carbamic acid tert-butyl ester in 40 mL of ethanol was hydrogenated at 2 atrn over 300 mg of 10% Pd on carbon for 15 minutes. The catalyst was removed by filtration through a glass fiber filter and the filtrate was concentrated to give a crude product as light yellow oil.'H NMR (400 MHz, CDC13) : 8 6.69 (m, 3H), 3.95 (s, 2H), 3.84 (s, 3H), 2.8 (m, 2H), 2.75 (s, 3H), 1.51 (s, 9H).

Step 4+5: 1- (2-Methoxy-4-methylaminomethyl-phenyl)-3- (5-methyl-pyrazin-2-yl)-urea. A solution of (4-amino-3-methoxy-benzyl)-methyl-carbamic acid ter-butyl ester was converted to the urea according to the general diphenylphosphoryl azide coupling method detailed for compound 336.

The Boc group was removed as described for above for compound 338.'H NMR (400 MHz, d6- DMSO): 5 9.91 (s, 2H), 8.78 (s, 1H), 8.21 (s, 1H), 8.07 (d, J= 8.61 Hz, 1H), 7.01 (s, 1H), 6.85 (d, J= 8.61 Hz, 1H), 3.89 (s, 3H), 3.59 (s, 1H), 2.42 (s, 3H), 2.26 (s, 3H). MS APCI-Pos, M+1 =301.8.

Compound 340: 1- {4-[(Benzyl-methyl-amino)-methyl]-2-methoxy-phenyl}-3-(5-met hyl-pyrazin-2-yl)-urea Step 1: N-Benzyl-N- (3-methoxy-4-nitro-benzyl)-methyl-amine. A solution of N-methyl-benzyl amine was alkylated with 3-methoxy-4-nitro-benzyl bromide as described above for compound 338.

'H NMR (400 MHz, CDC13) : 8 7.83 (d, J= 8.6 Hz, 1H), 7.35 (s, 4H), 7.26 (s, 1H), 7.17 (s, 1H), 7.01 (d, J = 7.04 Hz, 1H), 3.97 (s, 3H), 3.55 (s, 4H), 2.22 (s, 3H).

Step 2: 4-[(Benzyl-methyl-amino)-methyll-2-methoxy-phenylamine.

Nickel boride Reduction General Method: To a stirred solution of nickel chloride hexahydrate (820 mg, 3.45 mmol) in 12 mL EtOH and 3 mL THF, at 0 °C, NaBH4 (130 mg, 3.45 mmol) was added. The resulting black suspension was stirred at 0 °C while N-benzyl- (3-methoxy4-nitro-benzyl)-methyl- amine was added as a solution in 5 mL THF. After several minutes, 260 mg of NaBH4 was added in several portions over 10 minutes, and the reaction mixture was subsequently allowed to warm to room temperature. After 2 h, TLC indicated complete conversion to a new, more polar product. At this point, 1.5 mL of 5% NH40H was added and the reaction was stirred for about 10 min, until the black solids achieved a granular consistency. The reaction was filtered through a glass fiber filter, rinsing with THF. The clear colorless filtrate was concentrated to about'/4 volume, diluted with 30 mL water and extracted with EtOAc (3 x 30 mL). The combined extracts were washed with brine, dried over MgS04 and filtered through a short plug of silica and concentrated in vacuo to afford 575 mg (65%) of the desired product as an off-white solid.'H NMR (400 MHz, CDC13) : 8 7.2-7.4 (m, 5H), 7.85 (s, 1H), 6.85 (s, 1H), 6.74 (d, J= 8.2 Hz, 1H), 6.66 (d, J= 8.2 Hz, 1H), 3.87 (s, 3H), 3.73 (br. s, 2H) 3.49 (s, 2H), 3.45 (s, 2H), 2.18 (s, 3H). MS (APCI-pos) M+l= 256.9 Step 3: 1-{4-[(Benzyl-methyl-amino)-methyl]-2-methoxy-phenyl}-3-(5-m ethyl-pyrazin-2-yl)-urea.

In a 50 mL round bottom flask, 5-methylpyrazine-2-carboxylic acid (250 mg, 1.8 mmol) and diisopropylethylamine (330 pL, 1.9 mmol) in 20 mL toluene, was stirred under a nitrogen atmosphere until the acid dissolved. Diphenylphosphoryl azide (523 mg, 1.9 mmol) was added and the solution was heated to 90 °C. After 20 minutes, nitrogen evolution had subsided, and the solution had darkened to a caramel color. The reaction was cooled to 65 °C, and 4-[(benzyl-methyl-amino)-methyl]-2- methoxy-phenylamine (486 mg, 1.9 mmol) was added as a solution in 5 mL toluene. The reaction was allowed to stir at 65 °C for 6 h, and was then cooled to room temperature, diluted with 30 mL EtOAc, and washed with 15 mL 5% NH40H. The aqueous phase was extracted with EtOAc (2 x 20 mL) and the combined organics were washed with 30 mL brine, dried over MgS04, filtered, and concentrated.

The resulting residue was purified by flash chromatography to give 285 mg (40%) of the desired product, which was further purified by trituration with diethyl ether. Mp=142-143 °C.'H NMR (400 MHz, CDC) : 8 11. 33 (s, 1H), 9.51 (s, 1H), 8.41 (s, 1H), 8.27 (d, J= 8.61 Hz, 1H), 8.08 (s, 1H), 7.2- 7.4 (m, 5H), 6.96 (d, J = 7.83 Hz, 1H) 3.97 (s, 3H), 3.53 (s, 4H), 2.53 (s, 3H), 2.22 (s, 3H). i3C NMR (400 MHz, CDC13) : 8 153.70,149.03,147.48,147.4,145.99,138.73,137.38,134.81,129.1 9,128.42, 127.16,121.89,119.81,111.05,104.49,94.98,87.22,61.81,56.37,4 2.5. MS (APCI-pos) M+1=392. 0.

Compound 341 : 1- (4-Dimethylaminomethyl-2-methoxy-phenyl)-3- (5-methyl-pyrazin-2-yl)-urea Step 1: (3-Methoxy-4-nitro-benzyl)-dimethyl-amine According to the method described above for compound 338,3-methoxy-4-nitro-benzyl bromide was treated with dimethyl amine to give the desired product..'H NMR (400 MHz, CDC13) : 8 7.86 (d, J= 7.8 Hz, 1H), 7.3 (s, 1H), 6.98 (d, J= 8.6 Hz, 1H), 4.01 (s, 3H), 3.5 (s, 2H), 2. 3 (s, 6H).

Step 2: 4-Dimethylaminomethyl-2-methoxy-phenylamine According to the nickel-boride method described for compound 340 above, (3-methoxy-4-nitro-benzyl)-dimethyl-amine, was reduced to the corresponding aniline and was used in the next step without characterization.

Step 3: 1- (4-Dimethylaminomethyl-2-methoxy-phenyl)-3- (5-methyl-pyrazin-2-yl)-urea.

According to the diphenylphosphoryl azide method described above for compound 336,4- dimethylaminomethyl-2-methoxy-phenylamine was converted to the (5-methyl-pyrazin-2-yl)-urea. The crude product was purified by preparative TLC, eluting with 5% MeOH in CH2CI2. IH NMR (400 MHz, d6-DMSO) : 8 11.21 (s, 1H), 8.95 (s, 1H), 8.39 (s, 1H), 8.21 (d, J= 7.83 Hz, 1H), 8.04 (s, 1H), 7.01 (s, 1H), 6.89 (d, J= 7.83 Hz, 1H), 3.98 (s, 3H), 3.92 (s, 2H), 2.26 (s, 3H), 2.24 (s, 6H). MS APCI- Pos, M+1 =316. 0.

Compound 342 : o o o OH N N ON OH O O i H H -H 0 NTy ZON O \ I N NJ II \ I NH2 O 1- (4-Aminomethyl-2-methoxy-phenyl)-3- (5-methyl-pyrazin-2-yl)-urea Step 1: 2- (3-Methoxy-4-nitro-benzyl)-isoindole-1, 3-dione. To a 250 mL round bottomed flask containing 10 g (55 mmol) of 4-nitro-3-methoxybenzyl alcohol in 150 mL THF was added, diethylazodiacarboxylate (8.03 g, 54.6 mmol, 1 eq.) and triphenylphosphine (15.0 g, 57.3 mmol) followed by 11.6 g of phthalimide (57.3 mmol) at 0 °C. Reaction was then allowed to gradually warm to room temperature over night. A white precipitate formed and was collected by suction filtration.

Recrystallization from acetonitrile gave 13.8 g (81%) of the desired product. IH NMR (400 MHz, d6- DMSO): 87. 79 (m, 5H), 7.19 (s, 1H), 7.08 (d, J= 8. 80 Hz, 1H), 4.91 (s, 2H), 3.87 (s, 3H).

Step 2: 2- (4-Amino-3-methoxy-benzyl)-isoindole-1, 3-dione. In a 500 mL Parr vessel, a partially dissolved suspension of 2- (3-methoxy-4-nitro-benzyl)-isoidole-1, 3-dione (1.50 g, 4.80 mmol) in 100 mL EtOH and 30 mL THF was hydrogenated over 250 mg 10 % Pd-C at 2.5 atm for 1 h. The catalyst was removed by filtration through a glass fiber filter, and the clear light yellow filtrate was concentrated in vacuo. The product was washed with 30 mL of diethyl ether and collected by suction filtration to give 1.28 g (95%) of the aniline as fine, light green needles.'H NMR (400 MHz, CDCI3) : 8 7.82 (m, 2H), 7.81 (m, 2H), 6.93 (s, 1H), 6.9 (d, J= 7.8 Hz, 1H), 6.63 (d, J= 7.8 Hz, 1H), 4.74 (s, 2H), 3.84 (s, 3H).

Step 3: Acyl Azide coupling General Method <BR> <BR> 1- [4- (1, 3-Dioxo-1, 3-dihydro-isoindol-2-ylmethyl)-2-methoxy-phenyl]-3- (5-methyl-pyrazin-2-yl)- urea. In a 50 mL round bottom flask, a solution of 5-methyl-pyrazine-2-carbonyl azide (510 mg, 3.15 mmol) in 15 mL of anhydrous toluene was stirred under a nitrogen atmosphere. The reaction flask was immersed in a 90 °C oil bath, and as the internal temperature approached 90 °C, N2 release was evident and the solution started to darken. After 20 min, effervescence had subsided, and the solution had darkened to a caramel color. The reaction flask was moved to a 65 °C bath, and 2- (4-amino-3- methoxy-benzyl)-isoindole-1, 3-dione (884 mg, 3.15 mmol) suspended in toluene (5 mL) was added.

The reaction was stirred at 65 °C for 6 h, then cooled to room temperature. The product precipitated from solution after cooling to room temperature, and was collected by suction filtration.

In cases where the product does not precipitate from the reaction mixture, the following work-up is applied: After cooling to room temperature, the brown solution is diluted with 5% aq. NH40H and the extracted with EtOAc (3x). The combined extracts are washed with brine, dried over MgS04, filtered and concentrated. The residue is then purified by flash chromatography in an appropriate solvent system.

Step 4: 1- (4-Aminomethyl-2-methoxy-phenyl)-3- (5-methyl-pyrazin-2-yl)-urea In a 100 mL round bottom flask, a suspension of 1- [4- (1, 3-dioxo-1, 3-dihydro-isoindol-2-ylmethyl)-2-methoxy-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea (620 mg, 1. 48 mmol) in 22 mL of EtOH, under a nitrogen atmosphere, was warmed, with stirring, to 70 °C. Hydrazine monohydrate (1.4 mL) was added, and the reaction was stirred at 70 °C. After 10 minutes, the reaction had become a completely homogenous caramel colored solution. After several minutes more, product started to precipitate from the solution. After 20 min, the reaction was cooled to room temperature, and the white solid product was collected by suction filtration. The crude product, which contained some phthalhydrazide by-product, was taken up in 80 mL of EtOAc, and washed with water (3 x 20 mL). The washings were back extracted with 30 mL EtOAc, and the combined organics were washed with brine, then dried over MgS04, filtered, and concentrated in vacuo to afford 400 mg, (94%) of the desired amine. Mp=168-169 °C.'H NMR (400 MHz, d6-DMSO) : 8 9.88 (br. s, 2H), 8.78 (s, 1H), 8. 21 (s, 1H), 8.05 (d, J = 8.2 Hz, 1H), 7.05 (s, 1H), 6.85 (d, J= 8.1 Hz, 1H), 3.89 (s, 3H), 3.68 (s, 2H), 2.42 (s, 3H) 1.80 (br. s, 2H, NH2). MS (APCI- pos) M-17 (-NH3) = 270.1 apci-neg M-1=285. 8.

Alkyl Derivatives of Compound 342.

Compound 343: A solution of 1-(4-aminomethyl-2-methoxy-phenyl)-3-(5-methyl-pyrazin-2-yl) -urea (0.25 mmol, 1.0 eq.) and 1 mL trimethylorthoformate in 4 mL MeOH was stirred at room temperature, then thiophene- 2-carboxaldehyde (2.5 mmol, 10 eq.) was added and the mixture was heated at 80 °C. After 18 h the reaction was cooled to room temperature and was concentrated in vacuo. The resulting imine was taken up in 5 mL of anhydrous MeOH and stirred at 0 °C. Sodium borohydride (0.75 mmol, 3 eq.) was added, and the reaction was stirred at 0 °C for 30 minutes and was then diluted with 2 mL of water and partitioned between 50 mL EtOAc and 30 mL sat. NaHC03. The organic phase was washed with brine, dried over MgS04, filtered and concentrated. If needed, the product was purified by flash chromatography, eluting with an appropriate MeOH-CH2CI2 mixture. <BR> <BR> <BR> <BR> <BR> <P>1-(2-Methoxy-4-{ [(thiophen-2-ylmethyl)-aminol-methyl}-phenyl)-3-(5-methyl-py razin-2-yl)-urea 'H NMR (400 MHz, d6-DMSO) : # 10.01 (s, 2H), 8.79 (s, 1H), 8.23 (s, 1H), 8.17 (d, J= 8.61 Hz, 1H), 7.59 (d, J= 6.26 Hz, 1H), 7. 33 (s, 1H), 7.15 (m, 2H), 7.05 (s, 1H), 6.97 (d, J= 8.61 Hz, 1H), 4.02 (s, 2H), 3.91 (s, 3H), 3.88 (s, 1H), 3.78 (s, 1H), 2.43 (s, 3H). MS APCI-Pos no detectable molecular ion.

Compound 344 : Prepared according to the general procedure described for compound 343 using thiophene-3- carboxaldehyde.

1-(2-Methoxy-4-{[(thiophen-3-ylmethyl)-aminol-methyl}-phenyl )-3-(5 methyl-pyrazin-2-yl)-urea 'H NMR (400 MHz, d6-DMSO) : 8 9.92 (s, 2H), 8.78 (s, 1H), 8.21 (s, 1H), 8.07 (d, J= 7.81 Hz, 1H), 7.47 (s, 1H), 7. 3 (s, 1H), 7. 11 (d, J = 4. 88 Hz, 1H), 7.04 (s, 1H), 6.87 (d, J=7. 81 Hz, 1H), 3.9 (s, 3H), 3.67 (s, 2H), 3.65 (s, 2H), 2.42 (s, 3H). MS APCI-Neg, M-1 =382. 0.

Compound 345 : Prepared according to the general procedure described for compound 343 using furfural. <BR> <BR> <BR> <BR> <P>1- (4- { [ (Furan-2-ylmethyl)-amino]-methyl}-2-methoxy-phenyl)-3- (5-methyl-pyrazin-2-yl)-urea'H NMR (400 MHz, MeOD) : # 8.57 (s, 1H), 8.2 (s, 1H), 8.1 (d, J= 8.61 Hz, 1H), 7.46 (s, 1H), 7.03 (s, 1H), 6.88 (d, J= 8.61 Hz, 1H), 6.36 (s, 1H), 6.27 (s, 1H), 3.96 (s, 3H), 3.74 (s, 2H), 3.71 (s, 2H), 2.48 (s, 3H). MS APCI-Neg, M-1 =366.0.

Compound346: Prepared according to the general procedure described for compound 343 using furan-3- carboxaldehyde.

1-(4-{[(Furan-3-ylmethyl)-amino]-methyl}-2-methoxy-phenyl)-3 -(5-methyl-pyrazin-2-yl)-urea IH NMR (400 MHz, MeOD) : 8 8. 58 (s, 1H), 8.21 (s, 1H), 8.13 (d, J= 7.83 Hz, 1H), 7.49 (s, 1H), 7.05 (s, 1H), 6.91 (d, J= 7.04 Hz, 1H), 6.5 (s, 1H), 3.97 (s, 3H), 3.78 (s, 2H), 3.69 (s, 2H), 2.49 (s, 3H). MS APCI-Neg, M-1 =366.0 Compound 347 : Prepared according to the general procedure described for compound 343 using 2- methoxybenzaldehyde.

1-{2-Methoxy-4-[(2-methoxy-benzylamino)-methyl]-phenyl}-3 -(5-methyl-pyrazin-2-yl)-urea 1H NMR (400 MHz, CDC13) : 5 11.3 (s, 1H), 9.97 (s, 1H), 8.44 (s, 1H), 8.22 (d, J= 7.83 Hz, 1H), 7.98 (s, 1H), 7.28 (m, 2H), 7.06 (s, 1H), 6.88 (m, 3H), 4.08 (s, 1H), 3.93 (s, 5H), 3.81 (s, 5H), 2.5 (s, 3H). MS APCI-Pos, M+1 =407. 8.

Compound 348: Prepared according to the general procedure described for compound 343 using 3- methoxybenzaldehyde.

1-{2-Methoxy-4-[(3-methoxy-benzylamino)-methyl]-phenyl}-3 -(5-methyl-pyrazin-2-yl)-urea 1H NMR (400 MHz, d6-DMSO) : õ 8. 57 (s, 1H), 8.20 (s, 1H), 8.11 (d, J= 8. 61 Hz, 1H), 7.22 (t, J = 7. 83 Hz, 1H), 7.04 (s, 1H), 6.91 (m, 3H), 6.83 (d, J= 8.61 Hz, 1H), 3.96 (s, 3H), 3.79 (s, 3H), 3.75 (s, 4H), 2.48 (s, 3H). MS APCI-Neg, M-1 =406. 0 Compound 349: Prepared according to the general procedure described for compound 343 using 4- methoxybenzaldehyde. <BR> <BR> <P>1- {2-Methoxy-4-1 (4-methoxy-benzylamino)-methyl]-phenyl}-3- (5-methyl-pyrazin-2-yl)-urea'H NMR (400 MHz, d6-DMSO) : 8 9.91 (s, 2H), 8.78 (s, 1H), 8.21 (s, 1H), 8.07 (d, J= 7.81 Hz, 1H), 7.25 (d, J= 8.78 Hz, 2H), 7.03 (s, 1H), 6.88 (m, 2H), 3.89 (s, 3H), 3.73 (s, 3H), 3.62 (s, 2H), 3.6 (s, 2H), 2.42 (s, 3H). MS APCI-Pos, M+1 =407.9.

Acyl Derivatives Compound 350 : A solution of 1- (4-aminomethyl-2-methoxy-phenyl)-3- (5-methyl-pyrazin-2-yl)-urea (100 mg, 0.35 mmol) in 30 mL THF and 15 mL aqueous NaHC03, was treated with 3.7 mmol, (1.05 eq.) of acetyl chloride. The biphasic reaction mixture was vigorously stirred at room temperature and after 2h was diluted with 10 mL water and extracted with EtOAc (3 x 20 mL). The combined extracts were washed with brine, dried over MgS04, and filtered through a short plug of silica. The filtrate was concentrated and the resulting residue was triturated with diethyl ether. If required, further purification was accomplished by flash chromatography, eluting with an appropriate methanol-CH2CI2 solvent system.

N- {3-Methoxy-4-l3-(5-methyl-pyrazin-2-yl)-ureido]-benzyl}-acet amide'H NMR (400 MHz, d6- DMSO): # 9. 92 (s, 2H), 8.75 (s, 1H), 8.28 (t, J= 5. 87 Hz, 1H), 8.19 (s, 1H), 8.07 (d, J= 8.61 Hz, 1H), 6.92 (s, 1H), 6.78 (d, J= 8.61 Hz, 1H), 4.19 (s, 1H), 4.18 (s, 1H), 3.87 (s, 3H), 2.4 (s, 3H), 1.85 (s, 3H).

MS ESI-pos, M+1 = 330.2.

Compound 351 : Prepared according to the general procedure described for compound 350 using methoxyacetyl chloride.

2-Methoxy-N- {3-methoxy-4-l3-(5-methyl-pyrazin-2-yl)-ureido]-benzyl}-acet arnide IH NMR (400 MHz, d6-DMSO) : 8 9.51 (s, 2H), 8.34 (s, 1H), 7.89 (t, J= 6.26 Hz, 1H), 7.78 (s, 1H), 7.65 (d, J= 7.83 Hz, 1H), 6.54 (s, 1H), 6. 38 (d, J= 7.83 Hz, 1H), 4.2 (s, 1H), 4.19 (s, 1H), 3.82 (s, 3H), 3.79 (s, 2H), 3.29 (s, 3H), 2. 35 (s, 3H). MS APCI-Pos, M+1 = 359.9.

Compound 352 : Prepared according to the general procedure described for compound 350 using dimethylamino-acetyl chloride. <BR> <BR> <P>2-Dimethylamino-N- {3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureidoJ-benzyl}-acetamide'H NMR (400 MHz, MeOD) : 5 8.56 (s, 1H), 8. 18 (s, 1H), 8.09 (d, J = 7.83 Hz, 1H), 6.97 (s, 1H), 6.86 (d, J= 7.83 Hz, 1H), 4.37 (s, 2H), 3.94 (s, 3H), 3.03 (s, 2H), 2.47 (s, 3H), 2.3 (s, 6H). MS APCI-Neg, M-1 = 370.9.

Compound 353: Prepared according to the general procedure described for compound 350 using 2- (2-pyridyl)-acetyl chloride.

N-{3-Methoxy-4-[3-(5-methyl-pyrazin-2-yl)-ureido]-benzyl} -2-pyridin-2-yl-acetamide 1H NMR (400 MHz, d6-DMSO) : 8 9.94 (s, 2H), 8.77 (s, 1H), 8.63 (t, J= 5.87 Hz, 1H), 8.49 (s, 1H), 8.44 (d, J= 6.26 Hz, 1H), 8.22 (s, 1H), 8. 08 (d, J= 7.83 Hz, 1H), 7.7 (d, J= 10.17 Hz, 1H), 7. 35 (q, J= 5.74 Hz, 1H), 6.87 (s, 1H), 6.79 (d, J = 9.39 Hz, 1H), 4.25 (s, 1H), 4.24 (s, 1H), 3.82 (s, 3H), 3.53 (s, 2H), 2.42 (s, 3H). MS APCI-Pos M+1 = 407.1.

Compound 354 : Prepared according to the general procedure described for compound 350 using 2- (4-methoxyphenyl)- acetyl chloride. <BR> <BR> <BR> <BR> <BR> <P>N-{3-Methoxy-4-13-(5-methyl-pyrazin-2-yl)-ureido]-b enzyl}-2-(4-methoxy-phenyl)-acetamide IH NMR (400 MHz, d6-DMSO) : 8 9.94 (s, 2H), 8.77 (s, 1H), 8.54 (t, J=4. 7 Hz, 1H), 8.21 (s, IH), 8.08 (d, J= 7.83 Hz, 2H), 7.89 (m, 1H), 7.22 (t, J= 7.83 Hz, 1H), 6.89 (s, 3H), 4.25 (s, 1H), 4.23 (s, 1H), 3.8 (s, 3H), 3.73 (s, 2H), 3.45 (s, 2H), 2.42 (s, 3H). MS (APCI-Pos) M+1 = 436. 2.

Compound 355: Prepared according to the general procedure described for compound 350 using benzoyl chloride.

N-{3-Methoxy-4-[3-(5-methyl-pyrazin-2-yl)-ureido]-benzyl} -benzamide 1H NMR (400 MHz, d6- DMSO): 8 9.95 (s, 2H), 9.02 (t, J= 5.48 Hz, 1H), 8.77 (s, 1H), 8. 22 (s, 1H), 8.1 (d, J= 8.61 Hz, 1H), 7.9 (d, J = 7.83 Hz, 2H), 7.48 (m, 3H), 7.04 (s, 1H), 6.88 (d, J= 10.17 Hz, 1H), 4.46 (s, 1H), 4.44 (s, 1H), 3.89 (s, 3H), 2.42 (s, 3H). MS APCI-Pos, M+1 = 391.9.

Compound 356 : Prepared according to the general procedure described for compound 350 using pyridine-2-carbonyl chloride.

Pyridine-2-carboxylic acid 3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzylamide'H NMR (400 MHz, d6-DMSO) : 5 9.94 (s, 2H), 9.29 (t, J= 6.26 Hz, 1H), 8.77 (m, 1H), 8.65 (m, 1H), 8.21 (s, 1H), 8.05 (m, 3H), 7.61 (m, 1H), 7.07 (s, 1H), 6.89 (d, J= 8.61 Hz, 1H), 4.47 (s, 1H), 4.45 (s, 1H), 3.88 (s, 1H), 2.42 (s, 3H). MS APCI-Pos no detectable molecular ion.

Compound 357: Prepared according to the general procedure described for compound 350 using nicotinoyl chloride.

N- {3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzyl}-nicotinamide'H NMR (400 MHz, d6- DMSO): # 10 (s, 2H), 9.26 (t, J= 5. 87 Hz, 1H), 9.11 (s, 1H), 8.82 (s, 1H), 8.77 (d, J= 4.7 Hz, 1H), 8.27 (s, 2H), 8.16 (d, J= 7.83 Hz, 1H), 7.58 (m, 1H), 7.09 (s, 1H), 6.95 (d, J= 8.61 Hz, 1H), 4.53 (s, 1H), 4.51 (s, 1H), 3.95 (s, 3H), 2.47 (s, 3H). MS APCI-Pos, M-1 = 397.9.

Compound 358: Prepared according to the general procedure described for compound 350 using isonicotinoyl chloride.

N- {3-Methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzyl}-isonicotinamide'H NMR (400 MHz, d6-DMSO) : 8 9.92 (s, 2H), 9.27 (t, J= 5.87 Hz, 1H), 8.74 (s, 1H), 8.71 (d, J= 6. 3 Hz, 2H), 8.18 (s, 1H), 8.08 (d, J= 8.6 Hz, 1H), 7.77 (d, J= 6.3 Hz, 2H), 7 (s, 1H), 6.86 (d, J= 8.6 Hz, 1H), 4.44 (s, 1H), 4. 43 (s, 1H), 3.86 (s, 3H), 2.39 (s, 3H). MS APCI-pos no detectable molecular ion.

Compound 359: Prepared according to the general procedure described for compound 350 using thiophene-2-carbonyl chloride.

Thiophene-2-carboxylic acid 3-methoxy4-13- (5-methyl-pyrazin-2-yl)-ureidol-benzylamide'H NMR (400 MHz, d6-DMSO) : 8 9.96 (s, 2H), 9.03 (t, J= 6.26 Hz, 1H), 8.78 (s, 1H), 8.22 (s, 1H), 8.11 (d, J= 7.83 Hz, 1H), 7.82 (d, J= 4.7 Hz, 1H), 7.77 (d, J= 3.91 Hz, 1H), 7.16 (d, J= 3.91 Hz, 1H), 7.03 (s, 1H), 6.88 (d, J= 10.17 Hz, 1H), 4.43 (s, 1H), 4.42 (s, 1H), 3.89 (s, 3H), 2.42 (s, 3H). MS APCI- pos, M-1 = 397.9.

Compound 360 : Prepared according to the general procedure described for compound 350 using 4-dimethylamino-2- carbonyl chloride.

3-Dimethylamino-N-{3-methoxy-4-l3-(5-methyl-pyrazin-2-yl)-ur eidol-benzyl}-benzamide lH NMR (400 MHz, d6-DMSO) : 8 9.94 (s, 2H), 8.9 (t, J= 5.87 Hz, 1H), 8.77 (s, 1H), 8.21 (s, 1H), 8.09 (d, J= 8.61 Hz, 1H), 7.26 (t, J= 7.83 Hz, 1H), 7.2 (s, 2H), 7.02 (s, 1H), 6.87 (d, J= 8.61 Hz, 2H), 4.44 (s, 1H), 4.42 (s, 1H), 3.88 (s, 3H), 2.93 (s, 6H), 2.42 (s, 3H). MS APCI-pos, M+1 = 434.9.

Compound361 : Prepared according to the general procedure described for compound 350 using 1-phenyl-4- trifluoromethyl-lH-pyrazole-3-carbonyl chloride. l-Phenyl-4-trifluoromethyl-lH-pyrazole-3-carboxylic acid 3-methoxy-4-[3-(5-methyl-pyrazin-2- yl)-ureido]-benzylamide 1H NMR (400 MHz, d5-DMSO) : 8 9.96 (s, 2H), 9.09 (t, J= 6.26 Hz, 1H), 8.78 (s, 1H), 8.22 (s, 1H), 8.19 (s, 1H), 8.12 (d, J= 7.83 Hz, 1H), 7.59 (m, 6H), 7.02 (s, 1H), 6.89 (d, J = 8.61 Hz, 1H), 4.43 (s, 1H), 4.42 (s, 1H), 3.9 (s, 3H), 2.42 (s, 3H). MS TIC-pos M+1 = 526.2.

Sulfonylated Derivatives Compound362: A solution of 1- (4-aminomethyl-2-methoxy-phenyl)-3- (5-methyl-pyrazin-2-yl)-urea (0.25 mmol, 1.0 eq.), 4-dimethylaminopyridine (5 mg), diisopropylethylamine (36 mg, 0.275 mmol, l. leq.) in THF (5 mL) was prepared and thiophene-2-sulfonyl chloride (0. 275 mmol, 1.1 eq.) was added. The mixture was stirred at room temperature for 24 h. The reaction mixture was partitioned between EtOAc (75 mL) and sat. NaHC03. After separation of the layers, the organic layer was washed with water, saturated brine, dried over MgS04, filtered and concentrated. The residue was purified by flash chromatography eluting with 5 % MeOH in dichloromethane and trituration with ether to give pure products.

Thiophene-2-sulfonic acid 3-methoxy-4-13-(5-methyi-pyrazin-2-yl)-ureido]-benzylamidelH NMR (400 MHz, d6-DMSO) : 5 9.96 (s, 2H), 8.77 (s, 1H), 8.33 (t, J = 7. 04 Hz, IH), 8.31 (s, 1H), 8.07 (d, J = 7.83 Hz, 1H), 7.93 (d, J= 4. 7 Hz, 1H), 7.59 (s, 1H), 7.18 (t, J= 3. 91 Hz, 1H), 6.9 (s, 1H), 6.8 (t, J= 7.04 Hz, 1H), 4.05 (s, 1H), 4.03 (s, 1H), 3.85 (s, 3H), 2.51 (s, 3H). MS APCI-Pos, M+1 = 433. 9.

Compound 363 : Prepared according to the general procedure described for compound 362 using benzenesulfonyl chloride.

N- {3-Methoxy-4-13-(5-methyl-pyrazin-2-yl)-ureido]-benzyl}-benz enesulfonamide IH NMR (400 MHz, d6-DMSO) : 8 9.95 (s, 2H), 8.77 (s, 1H), 8.21 (s, 1H), 8. 13 (t, J= 6.26 Hz, 1H), 8.05 (d, J= 8.61 Hz, 1H), 7.81 (d, J = 7.04 Hz, 2H), 7.58 (m, 3H), 6.85 (s, 1H), 6.77 (d, 7=8. 61 Hz, lH), 3.96 (s, 1H), 3.95 (s, 1H), 3.81 (s, 3H), 2.42 (s, 3H). MS APCI-Pos, M+l =428. 2.

Compound 364 : Prepared according to the general procedure described for compound 362 using 2- trifluoromethoxybenzenesulfonyl chloride.

N-[3-Methoxy-4-[3-(5-methyl-pyrazin-2-yl)-ureido]-benzyl} -2-trifluoromethoxy- benzenesulfonamide'H NMR (400 MHz, d6-DMSO) : 8 9.94 (s, 2H), 8.77 (s, 1H), 8. 38 (s, 1H), 8.2t (s, 1H), 8.02 (d, J = 8.61 Hz, 1H), 7.89 (m, 1H), 7.72 (t, J= 7.83 Hz, 1H), 7.51 (d, J= 7.83 Hz, 2H), 6.88 (s, 1H), 6.75 (d, J=10. 17 Hz, 1H), 4.11 (s, 2H), 3.82 (s, 3H), 2.42 (s, 3H). MS APCI-Pos, M+1 =512.0.

Compound365: Prepared according to the general procedure described for compound 362 using 3- methoxybenzenesulfonyl chloride. <BR> <BR> <P>3-Methoxy-N-{3-methoxy-4- 3- (5-methyl-pyrazin-2-yl)-ureidol-benzyl}-benzenesulfonamide'H NMR (400 MHz, d6-DMSO) : # 9.92 (s, 2H), 8.73 (s, 1H), 8.17 (s, 1H), 8.08 (s, 1H), 8. 01 (d, J= 7.83 Hz, 1H), 7.45 (t, J= 7.83 Hz, 1H), 7.34 (d, J= 7.83 Hz, 1H), 7.24 (s, 1H), 7.14 (m, 1H), 6.81 (s, 1H), 6.74 (d, J= 7.83 Hz, 1H), 3.93 (s, 2H), 3.77 (s, 3H), 3.76 (s, 3H), 2.38 (s, 3H). MS APCI-Pos, M+1 =458.0.

Compound 366: Prepared according to the general procedure described for compound 362 using 4- methoxybenzenesulfonyl chloride.

4-Methoxy-N-{3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzyl}-benzenesulfonamide'H NMR (400 MHz, d6-DMSO) : 8 9.93 (s, 2H), 8. 74 (s, 1H), 8.18 (s, 1H), 8.02 (d, J= 8.61 Hz, 1H), 7.92 (s, 1H), 7.69 (d, J= 8.61 Hz, 2H), 7.05 (d, J= 9.39 Hz, 2H), 6.81 (s, 1H), 6.74 (d, J= 7.83 Hz, 1H), 3.88 (s, 2H), 3.79 (s, 6H), 2. 39 (s, 3H). MS APCI-Pos, M+1 =458.2.

Compound 367 : Prepared according to the general procedure described for compound 362 using pyridine-2-sulfonyl chloride.

Pyridine-2-sulfonic acid 3-methoxy-4-[3-(5-methyl-pyrazin-2-yl)-ureido]-benzylamide 1H NMR (400 MHz, d6-DMSO) : 8 9.91 (s, 2H), 8.74 (s, 1H), 8.68 (d, J= 4.7 Hz, 1H), 8.17 (s, 1H), 7.99 (m, 2H), 7.86 (d, J = 7.83 Hz, 1H), 7.6 (s, 1H), 6.86 (s, 1H), 6.74 (d, J= 8.61 Hz, 1H), 4.1 (s, 2H), 3.80 (s, 3H), 3.32 (s, 1H), 2.38 (s, 3H). MS APCI-Pos, M+1 =428. 9

Additional preferred compounds of the present invention include <BR> <BR> <BR> N- (2-dimethylamino-1-phenyl-ethyl)-3-methoxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzamine ; N-(1-aza-bicyclo [2.2.2] oct-3-yl)-3-methoxy-4- [3- (5- methyl-pyrazin-2-yl)-ureido]-benzamide ; N-(3-R-1-cyclohexylmethyl-pyrrolidin-3-yl)-3-meth- oxy-4- [3- (5-methyl-pyrazin-2-yl)-ureido]-benzamide ; 1- [2-(2-dimethylamino-ethoxy)-5-methyl-phenyl]-3- pyrazin-2-yl-urea; <BR> <BR> <BR> 1- [2- (3-dimethylamino-propoxy)-5-methyl-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea; <BR> <BR> 1- (5-methyl-pyrazin-2-yl)-3- [5-methyl-2- (pyridin-3- ylmethoxy)-phenyl]-urea ; <BR> <BR> 1- [2- (2-dimethylamino-l-dimethylaminomethyl-ethoxy)- 5-methyl-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea ; <BR> <BR> 1- [5-methyl-2- (2-S-1-methyl-pyrrolidin-2-ylmethoxy)- phenyl]-3- (5-methyl-pyrazin-2-yl)-urea ; 1-{5-methyl-2-[2-(1-methyl-pyrrolidin-2-yl)-ethoxy]- phenyl}-3- (5-methyl-pyrazin-2-yl)-urea ; 1-{5-methyl-2-(1-methyl-piperidin-4-yloxy)-phenyl]- 3- (5-methyl-pyrazin-2-yl)-urea ; 1-[5-methyl-2-(3-(S)-1-methyl-piperidin-3-ylmeth- oxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea ; 1- [5-methyl-2- (3- (R)-l-methyl-piperidin-3-ylmeth- oxy)-phenyl]-3- (5-methyl-pyrazin-2-yl)-urea ; 1-[5-methyl-2-(1-methyl-piperidin-2-ylmethoxy)- phenyl]-3- (5-methyl-pyrazin-2-yl)-urea ; 1-[5-methyl-2-(1-methyl-piperidin-3-yloxy)-phenyl]- 3- (5-methyl-pyrazin-2-yl)-urea ; 1- [5-methyl-2- (l-methyl-piperidin-3-ylmethoxy)- phenyl]-3-quinoxalin-2-yl-urea ; 1-[5-methyl-2-(piperidin-3-ylmethoxy)-phenyl]-3-(5- methyl-pyrazin-2-yl)-urea;

1- [5-fluoro-2- (l-methyl-piperidin-3-ylmethoxy)- phenyl]-3- (5-methyl-pyrazin-2-yl)-urea ; 1- [5-fluoro-2- (l-methyl-piperidin-4-yloxy)-phenyl]- 3- (5-methyl-pyrazin-2-yl)-urea ; 1- [4-fluoro-2- (l-methyl-piperidin-4-yloxy)-phenyl]- 3- (5-methyl-pyrazin-2-yl)-urea ; 1- (2-methoxy-4-methylaminomethyl-phenyl)-3- (5- methyl-pyrazin-2-yl)-urea; l- (4- { [ (furan-3-ylmethyl)-amino]-methyl}-2-methoxy- phenyl)-3- (5-methyl-pyrazin-2-yl)-urea ; and 1- {2-methoxy-4- [ (4-methoxy-benzylamino)-methyl]- phenyl}-3- (5-methyl-pyrazin-2-yl)-urea.

Example 15 Identification of Chkl Inhibitors The human Chkl cDNA was identified and cloned as described previously in International Application No. PCT/US98/18558, filed September 4, 1998. A FLAG tag was inserted in frame with the amino terminus of the full-length Chkl. The 5' primer contains an EcoRI site, a Kozak sequence, and also encodes a FLAGO tag for affinity purification using the M2 Antibody (Sigma, Saint Louis, IL). The 3'primer contains a SalI site. The PCR-amplified fragment was cloned into pCI-Neo as an EcoRI-SalI fragment (Invitrogen, Carlsbad, CA), then subcloned as an EcoRI-NotI fragment into pFastBacI (Gibco-BRL, Bethesda, MD). Recombinant baculovirus was prepared as described in the Gibco-BRL Bac-to-Bac manual and used to infect Sf-9 cells grown in CCM3 medium (HyClone Laboratories, Logan, UT) for expression of FLAG@-tagged Chkl protein.

FLAG'-tagged Chkl was purified from frozen pellets of baculovirus-infected SF9 cells. Frozen

cell pellets were mixed with an equal volume of 2X lysis buffer containing 100 mM Tris-Cl pH 7.5,200 mM NaCl, 50 mM B-glycerophosphate, 25 mM NaF, 4 mM MgCl2, 0.5 mM EGTA, 0.2% TWEEN@-20, 2 mM sodium vanadate, 2 mM DTT, and a cocktail of protease inhibitors (Complete mini, Boehringer Mannheim 2000 catalog #1836170). Cells were then dounced 20 times with the loose pestle of a dounce homogenizer and centrifuged at 48,400 x g for 1 hour. The M2 affinity was prewashed with 10 column volumes of 50 mM glycine pH 3.5 followed by 20 mM Tris pH 7.5,150 mM NaCl alternating three times and ending with a Tris NaCl wash. The column was then washed with 25 column volumes of 20 mM Tris pH 7.5,150 mM NaCl, 0.1% TWEEN@-20, 1 mM EGTA, 1 mM EDTA and 1X complete mini protease tablets. The cleared lysate was then bound to M2 affinity resin in batch at 4°C for 4 hours. The mixture of resin and lysate was then poured into a column and the flow through collected.

The resin was washed with 10 column volumes of 20 mM Tris pH 7.5,150 mM NaCl, and 3 mM N-octyl gluco- side. FLAG@-tagged Chkl was then eluted from the column with 6 column volumes of cold 20 mM Tris pH 7.5,150 mM NaCl, 3 mM N-octyl glucoside containing 0. 5 mg/mL FLAG° peptide (Sigma, 2000 Catalog # F- 3290). Three fractions were collected an analyzed for the presence of FLAG-tagged Chkl.

The protein kinase was used to developed an assay for Chkl kinase activity that includes 100 ng purified FLAG@-Chkl (150 pmol of ATP/min), 20 Am Cdc25C peptide (H-leu-tyr-arg-ser-pro-ser-met-pro- glu-asn-leu-asn-arg-arg-arg-arg-OH) (SEQ ID NO: 1), 400 Am ATP, 2 yCi [32P]-gATP, 20 mM Hepes pH 7.2,5 mM MgCl2, 0.1% NP40 and 1 mM DTT. This assay was used to screen approximately 100,000 small molecule

inhibitors. Reactions were initiated by the addi- tion of ATP-containing reaction mix and carried out at room temperature for 10 min. Reactions were stopped by the addition of phosphoric acid (150 mM final concentration) and transferred to phospho- cellulose discs. The phosphocellulose discs were washed five times with 150 mM phosphoric acid and air-dried. Scintillation fluid was added and discs were counted in a Wallac scintillation counter. The screen identified a number of Chkl inhibitors having IC, o values in the range of 1 to 100 yM.

Example 16 Chkl Kinase Inhibitors are Selective Chkl inhibitors of the invention were tested for selectivity as against one or more other protein kinases, i. e., DNA-PK, Cdc2, Casein Kinase I (CKI), Chk2, p38 MAP kinase, Protein Kinase A (PKA), and calcium-calmodulin protein kinase II (CaM KII).

Assay procedures for all of these kinases except Chk2 have been previously described in the litera- ture, including U. S. provisional patent application 60/229,899, filed September 1, 2000, and U. S. patent application 08/184,605, filed January 21,1994, both of which are herein incorporated by reference.

Activity of the compounds against Chk2 was assayed as follows: 128 ng of purified His-tagged Chk2 was incubated with up to 100 mM Chkl inhibitor in the presence of 4 mM ATP, 1 mCi [32P] g-ATP, 20 mM Hepes pH 7.5,5 mM MgCl2, and 0.25% NP40 for 20 minutes at room temperature. Reactions were stopped with a final concentration of 150 mM phosphoric acid, and 5/8 of the reaction mixture was transferred to phos- phocellulose discs. The discs were washed five

times with 150 mM phosphoric acid, and air-dried.

Scintillant was added and radioactivity was counted using a Wallac beta counter. p38 MAP kinase, PKA, CaM KII, and Cdc2 were purchased from New England Biolabs, and assays were performed according to the manufacturer's instructions using 4-50 uM ATP and testing Chkl inhibitor concentrations as high as 100 uM. All inhibitors tested were showed at least 5- fold selectivity for Chkl over the other enzymes.

Example 17 Chkl Inhibitors Block Chkl Function in Cells Chk 1 is activated in response to ionizing radiation and certain chemical DNA damaging agents.

In the presence of DNA damage, Chkl is activated and causes a cell cycle arrest. In mammalian cells, the best-characterized cell cycle arrest invoked by Chkl is a G2 arrest. Activation of Chk 1 by DNA damage results in the phosphorylation and inactivation of Cdc25C, the dual specificity phosphatase that normally dephosphorylates cyclin B/cdc2 as cells progress into mitosis (Funari et al., Science, Sep.

5,1997; 277 (5331) 1495-7; Sanchez et al.; Matsuoka et al.; and Blasina et al.). This negative regula- tion of Cdc2 activity causes cell cycle arrest in order to prevent cells from entering mitosis in the presence of DNA damage or unreplicated DNA. Inhibi- tion of Chkl, therefore, allows cells to progress through the cell cycle in the presence of DNA damage of unreplicated DNA.

To establish that the Chkl inhibitors prevented Chkl function in cells, inhibitors were tested in molecular cell-based assays. Since mammalian Chkl has been shown to phosphorylate

Cdc25C in vitro, suggesting that it negatively regulates cyclin B/cdc2 in response to DNA damage, the ability of the Chkl inhibitors to enhance the activity of CyclinB/cdc2 was analyzed. The experi- ment was designed as follows: HeLa cells were irradiated with 800 rads and incubated for 7 hours at 37°C. Because these cells are functionally p53 negative, they arrest exclusively in G2. Then, nocodazole was added to a concentration of 0.5 ug/mL and incubated for 15 hours at 37°C. The addition of nocodazole was designed to trap any cells that pro- gressed through the G2 arrest into M. Finally, a Chkl inhibitor was added for 8 hours, the cells harvested, lysed and immunoprecipitated equal amounts of protein with an antibody to Cyclin B1 (New England Biolabs) as suggested by the manufac- turer. IPs then were analyzed for CyclinB-associ- ated cdc2 kinase activity by assaying histone H1 kinase activity (Yu et al., J Biol Chem. Dec. 11, 1998 ; 273 (50) : 33455-64). The results demonstrated that Compound 29 overrides the IR-induced inacti- vation of Cyclin B/Cdc2.

In addition, whether the Chkl inhibitors abrogate the IR-induced G2 DNA damage checkpoint as assayed by mitotic index experiments was tested.

HeLa cells (approximately lx106) were treated as described above. Cells were harvested by centrifu- gation, washed once with PBS, then resuspended in 2.5 mL 75 mM KC1 and centrifuged again. The cells then were fixed in 3 mL of freshly prepared cold, acetic acid: methanol (1: 3) and incubated on ice for 20 minutes. Cells were pelleted, fix solution aspirated and resuspended in 0.5 mL of PBS. Mitotic spreads were prepared by pipeting 100 uL of the fixed cells onto a glass microscope slide and flood-

ing the sample with 1 ml of fix solution. Slides were then air dried, stained with Wrights stain (Sigma) for 1 minutes, followed by one wash in water and one wash in 50% methanol. The presence of con- densed chromosomes and lack of nuclear envelope identified mitotic cells. Both Compounds 12 and 29 showed an increase in the number of mitotic cells in the presence of irradiation demonstrating abrogation of the IR-induced G2 arrest.

Abrogation of the IR-induced G2 checkpoint allows the cells to continue through the cell cycle, presumably in the presence of DNA damage, as demon- strated by analysis of DNA content by FACS profile.

293T cells were treated with 800 rads of ionizing radiation and increasing concentrations (up to 80 mM) of some of the Chkl inhibitors. The cells then were harvested and fixed with 5 mL of cold 70% ethanol at-20°C overnight. The cells then were pelleted by centifugation at 1000 x g for 10 minutes, and stained with 1 mL of solution contain- ing 50 mg/mL propidium iodide and 250 mg/mL RNase for 30 minutes at room temperature. Stained cells were then analyzed by FACS on FL2 using a Becton- Dickinson apparatus. These experiments demonstrated that, while the cells treated with radiation and vehicle alone remained arrested in G2, the Chkl inhibitor treated cells were distributed in Gl and S phase. These data, taken together with the data above, suggest that the Chkl inhibitors allow cells to continue cycling in the presence of ionizing radiation.

Example 18 Chkl Inhibitors enhance killing of cells by cancer treatments To test the hypothesis that inhibition of Chkl potentiates the killing effect of DNA-damaging agents, cells were incubated in the presence of selective Chkl inhibitors and either irradiation or chemical DNA-damaging agents. Cells plated at a density of 1000-2000 per well in 96-well microtitre plates were grown in RMPI 1640 containing 10% FBS, 100 U/mL penicillin and 00 yg/mL streptomycin for 18 hours at 37°C in a humidified incubator with 5% CO2. Cells tested included HeLa, ACHN, 786-0, HCT116, SW620, HT29, Colo205, SK-MEL-5, SK-MEL-28, A549, H322, OVCAR-3, SK-OV-3, MDA-MB-231, MCF-7, PC- 3, HL-60, K562, and MOLT4. All cell line designa- tions refer to human cell lines and refer to the following: HeLa cervical adenocarcinoma ACHN renal adenocarcinoma 786-0 renal adenocarcinoma HCT116 colon carcinoma SW620 colon carcinoma, lymph node metastasis HT-29 colonrectal adenocarcinoma Colo205 colon adenocarcinoma SK-MEL-5 melanoma SK-MEL-28 malignant melanoma A549 lung carcinoma H322 broncholoalveolar carcinoma OVCAR-3 ovarian adenocarcinoma SK-OV-3 ovarian adenocarcinoma MDA-MB-231 breast adenocarcinoma MCF-7 breast adenocarcinoma

PC-3 prostate adenocarcinoma, from metastasis to bone HL-60 acute promyelocytic leukemia K562 chronic myelogenous leukemia MOLT4 acute lymphoblastic leukemia; T lymphoblast Cells were treated with media containing chemotherapeutic drugs alone or chemotherapeutic drugs and Compounds 12 and 29. Cells were incubated for approximately 5 days before growth was measured by determination of levels of 3H-thymidine uptake.

Chemotherapeutic drugs included etoposide, doxorubi- cin, cisplatin, chlorambucil, 5-fluorouracil (5-FU).

The drug concentration necessary to inhibit cell growth to 90% of untreated control cells was defined as the GLgo. At concentrations less than 100 AM, Compounds 12 and 29 enhanced the killing of 5-FU from 2-to 10-fold.

Compounds 2 and 12 were tested with addi- tional antimetabolites, including methotrexate, hydroxyurea, 2-chloroadenosine, fludarabine, azacytidine, and gemcitibine for an ability to enhance killing of the agents. These Chkl inhibi- tors were found to enhance the killing of cells to hydroxyurea, fludarabine, 5-azacytidine, and metho- trexate suggesting that the combination of inhibi- tion of Chkl and blocking of DNA synthesis leads to increased cell death by these agents.

In addition, the ability of the Chkl inhibitor to enhance killing by irradiation was tested. In HeLa cells, Compounds 12 and 29 were found to enhance killing by irradiation 2-3 fold.

Example 19 Animal Tumor Models To test the ability of the Chkl inhibitors to enhance the killing of tumors by 5-FU in mice, xenograft tumor models using colon tumor cell lines were established. Colo205 and HT29 cells (human colon carcinoma) were used to propagate xenograft tumors in 6-8 week old female thymic Balb/c (nu/nu) mice. Mice were maintained in a laminar airflow cabinet under pathogen-free conditions and fed sterile food and water ad libitum. Cell lines were grown to subconfluence in RPMI 1640 media supple- mented with 10% FBS, 100 U/mL penicillin, 100 yg/mL streptomycin, and 1.5 mM L-glutamine in a 5% C02 humidified environment. Single cell suspensions were prepared in CMF-PBS, and cell concentration adjusted to lx108 cells/mL. Mice were inoculated subcutaneously (s. c). on the right flank or right leg with a total of 1X107 cells (100, uL).

Mice were randomized (5 mice/group) into four treatment groups and used when tumors reached a weight of 75-100 mg (usually 7-11 days post-inocula- tion). Tumors were measured with vernier calipers and tumor weights were estimated using the empir- ically derived formula: tumor weight (mg) = tumor length (mm) x tumor width (mm) 2/3. 3. Treatment consisted of i) 100 AL intraperitoneal (i. p). injec- tion of 5-FU at 50 mg/kg, 100 mg/kg, or 150 mg/kg.

A dose-dependent delay in tumor growth was observed in the mice treated with 5-FU. Tumor size was monitored every other day for the duration of the experiment.

Obviously, many modifications and varia- tions of the invention as hereinbefore set forth can be made without departing from the spirit and scope thereof, and, therefore, only such limitations should be imposed as are indicated by the appended claims.