AU2011244844A1 - Benzazole derivatives, compositions, and methods of use as B-secretase inhibitors - Google Patents

Benzazole derivatives, compositions, and methods of use as B-secretase inhibitors Download PDF

Info

Publication number
AU2011244844A1
AU2011244844A1 AU2011244844A AU2011244844A AU2011244844A1 AU 2011244844 A1 AU2011244844 A1 AU 2011244844A1 AU 2011244844 A AU2011244844 A AU 2011244844A AU 2011244844 A AU2011244844 A AU 2011244844A AU 2011244844 A1 AU2011244844 A1 AU 2011244844A1
Authority
AU
Australia
Prior art keywords
carboxylic acid
amide
imidazol
isoquinoline
benzoimidazol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2011244844A
Inventor
Robert C. Andrews
Devi Reddy Gohimmukkula
Guoxiang Huang
David Jones
Adnan M. M. Mjalli
Mohan Rao
Tan Ren
Jeff Zhu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
vTv Therapeutics LLC
Original Assignee
High Point Pharnaceuticals LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by High Point Pharnaceuticals LLC filed Critical High Point Pharnaceuticals LLC
Priority to AU2011244844A priority Critical patent/AU2011244844A1/en
Publication of AU2011244844A1 publication Critical patent/AU2011244844A1/en
Abandoned legal-status Critical Current

Links

Abstract

The present invention is directed to benzazole compounds that inhibit p-site amyloid precursor protein-cleaving enzyme (BACE) and that may be useful in the treatment or prevention of diseases in which BACE is involved, such as Alzheimer's disease. The invention is also directed to pharmaceutical compositions comprising these compounds and the use of these compounds and compositions in the prevention or treatment of such diseases in which BACE is involved.

Description

AUSTRALIA Regulation 3.2 Patents Act 1990 Complete Specification Standard Patent APPLICANT: High Point Pharmaceuticals, LLC Invention Title: BENZAZOLE DERIVATIVES, COMPOSITIONS, AND METHODS OF USE AS B-SECRETASE INHIBITORS The following statement is a full description of this invention, including the best method of performing it known to me: WO 2006/099379 PCT/US2006/009049 TITLE Benzazole Derivatives, Compositions, and Methods of Use as p-Secretase Inhibitors STATEMENT OF RELATED APPLICATION The present application claims the benefit of priority of US Provisional Application 5 No. 60/661,349, filed March 14, 2005. FIELD OF INVENTION The present invention relates to benzazole derivatives useful as inhibitors of secretase, the p-site amyloid precursor protein-cleaving enzyme (BACE). BACKGROUND 10 Alzheimer's disease is characterized by the abnormal deposition of p-amyloid (AP) in the brain in the form of extra-cellular plaques and intra-cellular neurofibrillary tangles. The rate of amyloid accumulation is a combination of the rates of AP formation, aggregation and egress from the brain. It is generally accepted that the main constituent of amyloid plaques is the 4kD amyloid protein (pA4, also referred to as AfP, p-protein and PAP) which is a 15 proteolytic product of a precursor protein of much larger size. Amyloid precursor protein (APP) is a 695-770 amino acid glycoprotein, expressed in the neurons and glial cells in peripheral tissues. APP has a receptor-like structure with a large ectodomain, a membrane spanning region, and a short cytoplasmic tail. Al is a 39-42 amino acid peptide, constitutes part of the ectodomain of APP, and extends partly to the 20 transmembrane domain of APP. At least two secretory mechanisms exist which release APP from the membrane and generate soluble, truncated forms of APP (sAPP). Proteases that release APP and its fragments from the membrane are termed "secretases." Most sAPP is released by a putative a-secretase that cleaves within the As protein to release sAPPa and precludes the release of 25 intact Ap. A smaller portion of sAPP is released by a P-secretase that cleaves near the NHl 2 terminus of APP and produces COOH-terminal fragments (CTFs) which contain the complete AP domain. Thus, the activity of p-secretase or p-site amyloid precursor protein-cleaving enzyme ("BACE") leads to the abnormal cleavage of APP, production of AP, and accumulation of 30 p-amyloid plaques in the brain, which is characteristic of Alzheimer's disease. In addition, the processing of APP by p-secretase is thought to be the rate determining step in Af production. Therefore, therapeutic agents that can inhibit BACE may be useful for the treatment of Alzheimer's disease. 1 WO 2006/099379 PCT/US2006/009049 The compounds of the present invention may be useful for treating Alzheimer's disease by inhibiting the activity of the BACE, thus preventing or reducing the rate of formation of insoluble Af. SUMMARY OF THE INVENTION 5 The present invention is directed to benzazole compounds that inhibit P-site ainyloid precursor protein-cleaving enzyme (BACE) and that may be useful in the treatment or prevention of diseases in which BACE is involved, such as Alzheimer's disease. The invention is also directed to pharmaceutical compositions comprising these compounds and the use of these compounds and compositions in the prevention or treatment of such diseases 10 in which BACE is involved. In one aspect, the present invention provides compounds of Formula (I) as shown below. In another aspect, the present invention provides methods for the preparation of compounds of Formula (I). In another aspect, the present invention provides pharmaceutical compositions 15 comprising a compound of Formula (I). In an embodiment, the pharmaceutical composition comprises a compound of Formula (1) and a pharmaceutically acceptable carrier, excipient, diluent, or mixture thereof. In another aspect, the present invention provides a method for the preparation of a pharmaceutical composition comprising a compound of Formula (I). In another aspect, the present invention provides methods of treatment or prevention 20 comprising administering a compound of Formula (I) or a pharmaceutical composition comprising a compound of Formula (1) to a subject having a disorder or disease in which BACE is involved. In another aspect, the present invention provides methods of treatment or prevention comprising administering a compound of Formula (1) or a pharmaceutical composition 25 comprising a compound of Formula (I) to a subject having a disorder or disease or at risk for having a disorder or disease, wherein the disorder or disease is selected from the group consisting of: Alzheimer's disease, mild cognitive impairment, Down's syndrome, Hereditary Cerebral Hemorrhage with Amyloidosis of the Datch-Type, cerebral amyloid angiopathy, degenerative dementia, diffuse Lewy body type of Alzheimer's disease or central or 30 preipheral amyloid diseases. Additional features of the present invention are described hereinafter. 2 WO 2006/099379 PCT/US2006/009049 DETAILED DESCRIPTION In a first aspect, the present invention provides certain substituted benzazole compounds. Such compounds are useful in the reduction of the proteolytic activity of BACE, as will be discussed in more detail below. 5 In another aspect, the present invention provides a compound of Formula (1) Ri R2 N Li-Q1G- L -Qg-Ly- G, Ra A R4 () wherein A is -0-, -S-, or -N(R 5 )-, 10 wherein R5 is selected from the group consisting of: a) -hydrogen; b) -alkyl; c) -aryl; d) -heteroaryl; 15 e) -cycloalkyl; f) -heterocyclyl; g) -alkylene-aryl; h) -alkylene-heteroaryl; i) -alkylene-cycloalkyl; and 20 j) -alkylene-heterocyclyl;
L
1 , L 6 , and L7 are independently selected from the group consisting of: a direct bond, -CH 2 -, -0-, -N(R 6 )-, -C(O)-, -CON(R 6 )-, -N(Rr)C(O)
-N(R
6 )CON(R)-, -N(R 6 )C(O)O-, -OC(O)N(R6)-, -N(R 6 )S0 2 -, -SO 2
N(R
6 )-, -C(O)-O O-C(O)-, -S-, -S(O)-, -S(0) 2 -, and -N(R 6
)SO
2 N(Ry)-, 25 wherein R6 and R 7 are independently selected from the group consisting of: -hydrogen, -alkyl, -aryl, and -alkylene-aryl; Q, and Q6 are independently selected from the group consisting of direct bond, alkylene, alkenylene, and alkynylene; 3 WO 2006/099379 PCT/US2006/009049
G
3 is selected from the group consisting of: heterocyclylene, cycloalkylene, heterocyclylene, arylene, heteroarylene, fused arylcycloalkylene, fused cycloalkylarylene, fused cycloalkylheteroarylene, fused heterocyclylarylene, and fused heterocyclylheteroarylene group wherein G may be optionally substituted I to 7 5 times, wherein the substituents are independently selected from the group consisting of: a) -halo; b) -cyano; c) -nitro; 10 d) -perhaloalkyl; e) -R8; f) -L 2 -Rg; g) -L-Q,-Rg; and h) -Q 2
-L
2 -Rs; 15 wherein Rs is selected from the group consisting of hydrogen, -alkyl, -aryl, and alkylene aryl; Q2 is selected from the group consisting of a direct bond, alkylene, alkenylene, and alkynylene; 20 L 2 is selected from the group consisting of a direct bond, -CH 2 -, -0-, -N(R 9 )-, C(O)-, -CON(R 9 )-, -N(R 9 )C(O)-, -N(R 9 )CON(Ro)-, -N(R 9 )C(O)0 , -OC(O)N(R 9 )-, -N(R 9 )S0 2 -, -S0 2
N(R
9 )-, -C(O)-O-, -0-C(O)-, -S-, -S(O)-, S(O)-, and -N(R 9
)SO
2 N(RiO)-, wherein R9 and Rio are independently selected from the group consisting 25 of: -hydrogen, -alkyl, -aryl, and -alkylene-aryl; or R 9 and R]Q are taken together with the atoms to which they are attached to form a heterocyclic ring of 5 to 7 members containing 0-2 additional heteroatoms independently selected from oxygen, nitrogen, and sulfur; G6 is selected from the group consisting of: hydrogen, heterocyclyl, cycloalkyl, heterocyclyl, 30 aryl, heteroaryl, fused arylcycloalkyl, fused cycloalkylaryl, fused cycloalkylheteroaryl, fused heterocyclylaryl, and fused heterocyclylheteroaryl group, wherein G6 is optionally substituted 1 to 7 times, wherein the substituents are independently selected from the group consisting of: a) -halo; 4 WO 2006/099379 PCT/US2006/009049 b) -cyano; c) -nitro; d) -perhaloalkyl; e) -Rios; 5 f -LIorRIos; g) -LI 02
-QI
02 -Rios; and h) -Q;o 2 -Lo 2 -Rio; wherein R1os is selected from the group consisting of hydrogen, -alkyl, -aryl, and alkylene 10 aryl; Q102 is selected from the group consisting of a direct bond, alkylene, alkenylene, and alkynylene; L1 02 is selected from the group consisting of a direct bond, -CH 2 -, -0-, -N(R 1 09 )-, -C(O)-, -CON(Rioo)-, -N(Rio9)C(O)-, -N(R ioq)CON(Ruo1)-, -N(Ri09)C(O)O 15 , -OC(O)N(Rio 9 )-, -N(R 1 o 9
)SO
2 -, -SO 2 N(Rio 9 )-, -C(O)-O-, -O-C(O)-, -S-, S(O)-, -S(0) 2 -, and -N(R] 09 )S0 2 N(Ri to)-, wherein R, 09 and Rio are independently selected from the group consisting of: -hydrogen, -alkyl, -aryl, and -alkylene-aryl; or R 09 and Ri1o are taken together with the atoms to vhich they are attached to form a heterocyclic 20 ring of 5 to 7 members containing 0-2 additional heteroatoms independently selected from oxygen, nitrogen, and sulfur; Ri, R2, R3, and R4 are independently selected from the group consisting of a) -H; b) -NH 2 ; 25 c) -carboxy; d) -cyano; e) -halogen; f -nitro; g) -OH 30 h) -alkyl; i) -aryl; j) -alkylene-aryl; k) -K-alkyl; 1) -K-aryl; 5 WO 2006/099379 PCT/US2006/009049 m) -K-alkylene-aryl; n) -L 3
-G
2
-
3 ; and o) -Ls-Q-Ls-GrLis-Qis-L 19 -Gs; wherein at least one of R 1 , R 2 , R3, and R 4 is not hydrogen; and 5 wherein K is selected from the group consisting of: -C(O)-O-, -O-C(O)-, -C(O)-NH-, NH-C(O)-, -S02-, -SO 2 -NH-, -NH-SO 2 -, and -C(O)-; Qg and Qis are independently selected from the group consisting of a direct bond, alkylene, alkenylene, and alkynylene; 10 L3, Ls, L9, Lig, and L, 9 are independently selected from the group consisting of: a direct bond, -CH2-, -0-, -N(R 26 )-, -C(O)-, -CON(R26)-, -N(R 26
)C(O)
, -N(R 26
)CON(R
27 )-, -N(R 26 )C(0)0-, -OC(O)N(R 26 )-, -N(R 26
)SO
2 , SO 2
N(R
26 )-, -C(O)-O-, -0-C(O)-, -S-, -S(O)-, -S(0) , -N(R 26 )SO2N(R2)-, -C(O)-N(R 26
)-C(=NH)-N(R
2 7)-, and -C(O) 15 N(R26)-N(R27) wherein R 2 6 and R27 are independently selected from the group consisting of: hydrogen, -alkyl, -aryl, -alkylene-aryl, or R26 and R27 are taken together with the atoms to which they are attached to form a heterocyclic ring of 5 to 7 members 20 containing 0-2 additional heteroatoms independently selected from oxygen, nitrogen, and sulfur;
G
2 is selected from the group consisting of: direct bond, aIkylene, alkenylene, alkynylene, and 10 L 1 25 wherein
L
1 o is selected from the group consisting of alkyline, cycloalkyline, heteroaryline, aryline, and heterocyclyline; L12 is selected from the group consisting of -0-, -C(O)-N(R 1 )-, C(O)-0-, -C(O)-, and -N(R )-CO-N(R 12 )-, wherein R, and R,2 30 independently comprise hydrogen, -aryl, -alkyl, and -alkylene-aryl; Li is selected from the group consisting of hydrogen, -alkyl, -alkenyl, alkynyl, -aryl, -alkylene-aryl, -alkylene -heteroaryl, alkylene-0 6 WO 2006/099379 PCT/US2006/009049 alkylene-aryl, -alkylene-S-alkylene-aryl, -alkylene-O-alkyl, alkylene-S-alkyl, -alkylene-NH 2 , -alkylene-OH, -alkylene-SH, alkylene-C(O)-ORu, -alkylene-C(O)-NR3RI4, -alkylene-NR 13
R
1 4 , -alkylene-N(Rn)-C(O)-RI4, -alkylene-N(R 3 )-S(O)-R14, and the 5 side chain of a natural or non - natural amino acid, wherein R1 3 and R 1 4 independently comprise hydrogen, -aryl, -alkyl, and -alkylene-aryl; or
R
13 and R 14 may be taken together to form a ring having the formula - (CH 2 )q-Y-(CH 2 )c bonded to the nitrogen atom to 10 which R 13 and R1 4 are attached, wherein q and r are, independently, 1, 2, 3, or 4; Y is -CH 2 -, -C(O)-, -0-, -N(H)-, -S-, -S(O)-, -SO2-, -CON(H)-, -NHC(O)-, -NHCON(H)-, NHSO2-, -SO 2 N(H)-, -(O)CO-, -NHSO 2 NH-, -OC(O)-, N(R 15 )-, -N(C(O)Rs)-, -N(C(O)NHRs)-, -N(SO 2 NHRs)-, 15 -N(SO 2 RIs)-, and -N(C(0)OR 15 )-, wherein R 15 is selected from the group consisting of hydrogen, -alkyl, -aryl, and alkylene-aryl; or
R
13 and R 14 may be taken together, with the nitrogen atom to which they are attached, to form a heterocyclyl or 20 heteroaryl ring; and
G
3 and G 18 are independently selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, fused arylcycloalkyl, fused cycloalkylaryl, fused cycloalky1heteroaryl, fused heterocyclylaryl, and fused heterocyclylheteroaryl group, wherein G 3 and G 1 8 may be 25 optionally substituted 1 to 7 times, wherein the substituents are independently selected from group consisting of: a) -halo; b) -NH 2 ; c) -carboxy; 30 d) -cyano; e) -nitro; f) -OH; g) -haloalkyl; h) -perhaloalkyl; 7 WO 2006/099379 PCT/US2006/009049 i) -Ri 1 ; j) -L 4
-R
16 ; k) -L 4
-Q
4 -Ri 0 ; and 1) -Q 4
-L
4 -Ri 6 ; 5 wherein
R
16 is selected from the group consisting of hydrogen, -alkyl, cycloalkyl, -aryl, -heterocyclyl, -heteroaryl, and -alkylene-aryl; Q4 is selected from the group consisting of direct bond, alkylene, alkenylene, and alkynylene; 10 L 4 is selected from the group consisting of direct bond, -CH 2 -, -0-, N(Ris)-, -C(O)-, -CON(Ris)-, -N(R, 8 )C(O)-, -N(Ris)CON(R 9
)
-N(R
1 )C(0)0-, -OC(0)N(Rig)-, -N(R 18
)SO
2 -, -S0 2
N(RS)
-C(0)-O-, -O-C(0)-, -S-, -S(O)-, -S(0)2-, and -N(R 1
S)SO
2
N(R
9 )-; 15 wherein Rig and R 1 9 are independently selected from the group consisting of: -hydrogen, -alkyl, -aryl, and -alkylene aryl;
G
8 is selected from the group consisting of alkylene, cycloalkylene, 20 heterocyclylene, arylene, heteroarylene, fused arylcycloalkylene, fused cycloalkylarylene, fused cycloalkylheteroarylene, fused heterocyclylarylene, and fused heterocyclylheteroarylene group, wherein
G
8 may be optionally substituted I to 7 times, wherein the substituents are independently selected from group consisting of: 25 a) -halo; b) -NH 2 ; c) -carboxy; d) -cyano; e) -nitro; 30 f) -OH; g) -haloalkyl; h) -perhaloalkyl; i) -R 1 6 ; j) -LI14-Ru6; 8 WO 2006/099379 PCT/US2006/009049 k) -Lj 14 -Qj 4 -Rj 6 ; and 1) -Q 114
-L
14
-R
16 ; wherein
R(
16 is selected from the group consisting of hydrogen, -alkyl, 5 cycloalkyl, -aryl, -heterocyclyl, -heteroaryl, and -alkylene-aryl; Q 114 is selected from the group consisting of direct bond, alkylene, alkenylene, and alkynylene;
L]
1 4 is selected from the group consisting of direct bond, -CHr 2 -, -0-, N(R 1 s)-, -C(O)-, -CON(RI s)-, -N(R 18
)C(O)
10 , -N(R I)CON(Ru g)-, -N(R 1 Ig)C(0)0-, -OC(O)N(Ru s) , -N(RI s)SOr-, -SO2N(Rlus)-, -C(O)-0-, -O-C(0)-, -S-, -S(O)-, -S(0) 2 -, and -N(R 1
)SO
2 N(Rus)-; wherein
R
18 and R, 9 are independently selected from the group 15 consisting of: hydrogen, -alkyl, -aryl, and -alkylene-aryl; wherein the alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, fused arylcycloalkyl, fused cycloalkylaryl, fused cycloalkylheteroaryl, fused heterocyclylaryl, and fused heterocyclyliheteroaryl groups in R] to Ru 19 , LI to LI 1 4 , GI to G 1 s, and Qi to Q14 may be optionally substituted 1-4 times with a substituent selected from the group consisting of: 20 a) -I; b) -halo; c) -hydroxyl; d) -amino; e) -cyano; 25 f) -carbamoyl; g) -carboxyl; h) -Z-alkyl; i) -Z-haloalkyl; j) -Z-perhaloalkyl; 30 k) -Z-aryl; 1) -Z-alkylene-aryl; m) -Z-cycloalkyl; n) -Z-alkylene-cycloalkyl; o) -Z-heterocyclyl; 9 WO 2006/099379 PCT/US2006/009049 p) -Z-alkylene-heterocyclyl; q) -Z-heteroaryl; and r) -Z-alkylene-heteroaryl; wherein Z is selected from the group consisting of a direct bond, -CH 2 -, -0-, -N(H), 5 S-, SO2-, -CON(H)-, -NHC(O)-, -NHCON(H)-, -NHSO 2 -, -SO 2 N(H3)-, -C(O)-O-, NHSO 2 NHI-, and -0-CO-, or a pharmaceutically acceptable salt, ester, or prodrug thereof In an embodiment of the compound of Formula (), A is -N(R 5 )-. In another embodiment of the compound of Fonnula (I), A is -N(R 5 )-, and Q1, G;, L-6, 10 Q6, L 7 , and G6 are taken together to form a group selected from the group consisting of: isoquinoline-3-yl, and (phcnylethynyl)-pyridine-2-yl. In another embodiment of the compound of Formula (I), A is -N(Rs)-, and R 1 or R 4 is the group imidazole-2-ylcarbamoyl. In another embodiment of the compound of Formula (I), A is -N(R 5 )-, and R, or R 4 is the group imidazole-2-ylcarbamoyl and R2 and R3 are hydrogen. 15 In another embodiment of the compound of Formula (1),
L
1 is -NH-C(O)-, -NI-, -C(O)-NH-, -NH-C(O)-NH-, or a direct bond; Q, is a direct bond; G, is phenyl, indole, isoquinoline, pyridine, pyrimidine, benzofuran, benzothiophene, benzimidazole, furan, imidazole, 1 -oxo-1,2-dihydroisoquinoline, 1,2,3,4 20 tetrahydroisoquinoline, cinnoline, quinoxaline, {1,8]-naphthyridine, 2,3 dihydro-[1,4]-dioxino-[2,3-g]-isoquinoline, 1,3-dioxolo-[4,5-g]-isoquinoline, 1,3,4,9-tetrahydro-beta-carboline, thieno-[2,3-c]-pyridine, imidazole-[1,2-a] pyridine, imidazole-[2,1-b]-thiazole, wherein Gi is substutited or unsubstituted. In another embodiment of the compound of Formula (I), 25 L, is -NH-C(O)-, -NH-, -C(0)-NH-, -NH-C(O)-NH-, or a direct bond; QI is a direct bond; G, is phenyl, indole, isoquinoline, pyridine, pyrinidine, benzofuran, benzothiophene, benzimidazole, furan, imidazole, 1-oxo-1,2-dihydroisoquinoline, 1,2,3,4 tetrahydroisoquinoline, cinnoline, quinoxaline, [1,8]-naphthyridine, 2,3 30 dihydro-[1,4]-dioxino-[2,3-g]-isoquinoline, 1,3-dioxolo-[4,5-g]-isoquinoline, 1,3,4,9-tetrahydro-beta-carboline, thieno-[2,3-c] -pyridine, imidazole-[ 1,2-al pyridine, imidazole-[2, 1-b]-thiazole, wherein substutited or unsubstituted;
L
6 , Q6, L 7 are direct bonds, and G is hydrogen. 10 WO 2006/099379 PCT/US2006/009049 In another embodiment of the compound of Formula (I), L, is -NH-C(O)-, -NH-, -C(O)-NH-, -NH-C(O)-NH-, or a direct bond; Q1 is a direct bond; 5 G1 is phenyl, indole-2-yl, indole-3-yl, isoquinoline-1-yl, isoquinoline-3-yl, isoquinoline-5-y, isoquinoline-6-yl, pyridine-2-yl, pyridine-3-yl, pyridine-4-yl, pyrimidine-2-yI, pyrimidine-4-yl, benzimidazole-2-yl, 1 -oxo- 1,2 dihydroisoquinoline-3-yl, 1,2,3,4-tetrahydroisoquinoline-1-yl, 1,2,3,4 tetrahydroisoquinoline-3-yl, 1,2,3,4-tetrahydroisoquinoline-5-yl, 1,2,3,4 10 tetrahydroisoquinoline-6-yI, cinnoline-3-yl, quinoxaline-2-yl, [1,81 naphthyridine-2-yl, 2,3-dilydro[1,4]dioxino[2,3-g]-isoquinoline-8-yl, 1,3 dioxolo[4,5-g]-isoquinoline-7-yl, 1,3,4,9-tetrahydro-beta-carboline-3-yl, thieno-[2,3-cJ-pyridine-5-yl, thieno-[2,3-c]-pyridine-6-yl, imidazole-[1,2-a] pyridine-2-yl, imidazole- [1,2-a]-pyridine-3-yl, or imidazole- [2,1 -b]-thiazole 15 6-yl, wherein G, is substituted or unsubstituted; L6, Q6, L7 are direct bonds; and G6 is hydrogen. In another embodiment pf the compound of Formula (I), L, is -NH-C(O)-, -NH-, -C(O)-NH-, -NH-C(O)-NH-, or a direct bond; 20 Q I is a direct bond; G, is phenyl, isoquinoline-1-yl, isoquinoline-3-yl, isoquinoline-5-yl, isoquinoline-6-yl, pyridine-2-yl, pyridine-3-yl, pyridine4-yl, pyrimidine-2-yl, or pyrimidine-4-yl, wherein GI is substituted or unsubstitated;
L
6 , Q6, L7 are direct bonds; and 25 G6 is hydrogen. In another embodiment of the compound of Fonnula (1), L, is -NH-C(O)-, -NH-, -C(O)-NH-, -NH-C(O)-NH-, or a direct bond; Q I is a direct bond; G is phenyl, pyridine-2-yl, pyridine-3-yl, or pyridine-4-yl, wherein G1J is substituted 30 or unsubstituted; L6 is a direct bond, -CH 2 -, or -0-; Q6 is a direct bond, lower alkylene, or -C=-C-; L7 is a direct bond, -CH 2 -, or -0-; and 11 WO 2006/099379 PCT/US2006/009049
G
6 is phenyl, faran, pyridine, thiophene, cyclopropyl, cyclopentyl, cyclohexyl, imidazole, pyrazole, or isoxazole, wherein Gs is substituted or unsubstituted. hi another embodiment of the compound of Fonrula (I), Li is -N(H)-C(O)-, and Q1,
G
1 , Lr, Q6, L7, and G 6 are taken together to form a group selected from the group consisting 5 of [1,3]Dioxolo[4,5-g]isoquinoline-7-yl, 1,2,3,4-tetrahydro-isoquinoline-6-yl, 1,2,3,4 tetrahydro-isoquinoline-3-yl, 1-(2-alkylsulfanyl-ethyl)-isoquinoline-3-yl, 1-(2-alkylsulfonyl ethyl)-isoquinoline-3-yl, 1-(tetrahydro-pyran-4-y1)-isoquinoline- 3 -yl, 1-alkyl-6-haloalkoxy isoquinoline-3-yl, 1-alkyl-7-haloalkoxy-isoquinoline-3-yl, 1-alkyl-isoquinoline-3-yl, 1 cycloalkylmethyl-7-alkoxy-isoquinoline-3-yl, 2,3-dihydro-[1,4}dioxino[2,3-g]isoquinoline-8 l0 yl, 4-alkoxy-quinoline-2-yl, 5,8-dialkoxy-isoquinoline-3-yl, 6,7-Bis-(atkoxy-ethoxy) isoquinoline-3-yl, 6,7-alkoxy-isoquinoline-1-yl, 7-alkoxy-1-alkyl-isoquinoline-3-y1, 7 alkoxy-isoquinoline-1-yl, 7-alkylsulfonyl-1-alkyl-isoquinoline-3-yl, 7-halo-8-alkoxy isoquinoline-3-yl, 7-hydroxy-1-alkyl-isoquinoline-3-yl, 7-methanesulfonylamino isoquinoline-3-yl, alkoxy-isoquinoline-3-y1, alkyl-isoquinoline-3-yl, benzyloxy-isoquinoline 15 3-yl, cycloalkoxy-isoquinoline-3-yl, isoquinoline-3-yl, 1-oxo-1,2-dihydro-isoquinoline-3-yl, isoquinoline-5-yl, and nitro-isoquinoline-3-yl. In another embodiment of the compound of Fonnula (1), Li is -N(H)-C(O)-, and Q1, G1, L 6 , Q(, L 7 , and G6 are taken together to form a group selected from the group consisting of: 2-(alkoxycarbonyl)-1,2,3,4-tetrahydro-isoquinoline-6-yl, 2-(alkoxycarbonyl)-1,2,3,4 20 tetrahydro-isoquinoline-3-yl, 2-(alkoxycarbonyl)-6,7-dialkoxy-1,2,3,4 tetrahydroisoquinoline-1-yl, 2-alkyl-1,2,3,4-tetrahydro-isoquinotine-3-yl, 2-alkylsulfonyl 1,2,3,4-tetrahydro-isoquinoline-6-y, 2-alkylsulfonyl-1,2,3,4-tetrahydro-isoquinoline-3-yl, 2 benzenesulfonyl-1,2,3,4-tetrahydro-isoquinoline-6-yl, and 2-benzyl-1,2,3,4-tetrahydro isoquinoline-6-yl. 25 In another embodiment of the compound of Formula (I), Li is -N(H)-C(O)-, and Q1, GI, L6, Q6, L 7 , and G6 are taken together to form a group selected from the group consisting of: ((alkyloxyphenyl)-ethynyl)-pyridine-2-yl, ((alkylphenyl)-ethynyl)-pyridine-2-yl, ((halophenyl)-ethynyl)-pyridine-2-yl, ((thiophen-3-yl)-ethynyl)-pyridine-2-yl, (1-phenyl ethoxy)-pyridine-3-yl, (3-cycloalkyl-prop-1-ynyl)-pyridine-2-yl, (3-hydroxy-3-nethyl-but-I 30 ynyl)-pyridine-2-yl, (alkoxy)-pyridine-3-yl, (alkoxy-phenyl)-pyridine-3-yl, (alkoxy phenylethynyl)-pyridine-2-yl, (alkyl-phenylethynyl)-pyridine-2-yl, (alkylsulfonyl-phenyl) pyridine-2-yl, (alkylsulfonyl-phenylethyny1)-pyridine-2-yl, (alkynyl)-pyridine-2-yl, (alkynyl)-pyridine-3-yl, (aininomethyl-phenyl)-pyridine-2-yl, (benzyloxy)-pyridine-2-yl, (benzyloxy)-pyridine-3-yl, (carbamoyl-phenyl)-pyridine-2-yl, (cyanomethyl-phenyl) 12 WO 2006/099379 PCT/US2006/009049 pyridine-2-yl, (cyano-phenyl)-pyridine-2-yl, (cyano-phenyl)-pyridine- 3 -yl, (cyano-phenyl) pyridine-4-yl, (cycloalkoxy)-pyridine-3-yI, (cycloalkyl-alkoxy)-pyridinie-2-yl, (cycloalkyl alkoxy)-pyridine-3-yl, (cycloalkylethynyl)-pyridine-2-yl, (cycloalkylethynyl)-pyridine-3-yl, (dialkylamino-phenylethynyl)-pyridine-2-yl, (dihalo-phenyl)-pyridine-3-yl, (haloalkoxy 5 phenyl)-pyridine-2-yl, (haloalkoxy-phenyl)-pyridine-3-yl, (haloalkyl-phenyl)-pyridine-2-yl, (halo-phenyl)-pyridine-2-yl, (halo-phenyl)-pyridine-3-yl, (halo-phenylethynyl)-pyridine-2-yl, (phenylalkoxy)-pyridine-2-yl, (phenyl-alkyl)-pyridine-3-yl, (phenylethynyl)-pyridine-2-yl, (phenylethynyl)-pyridine-3-yl, (phenylethynyl)-pyridine-4-yl, (pyridin-3-ylethynyl)-pyridine 2-yl, (thiophen-2-yl)-pyridine-2-yl, [2,3']bipyridinyl-6-yl, [2,4']bipyridinyl-6-yl, 2-(cyano 10 phenyl)-pyridine-4-yl, 2-(halo-phenoxy)-pyridine-3-yl, 2-(phenoxy)-pyridine-3-yl, phenethyl pyridine-2-yl, 3-halo-6-(alk- 1-ynyl)-pyridine-2-yl, 5-(phenoxy)-pyridine-3-yl, 6-(3-cyano phenyl)-pyridine-2-yl, cyano-pyridine-2-yl, cyano-pyridine-3-yl, cyano-pyridine-4-yl, ethynyl-pyridine-2-yl, ethynyl-pyridine-3-yl, ethynyl-pyridine-4-yl, halo-pyridine-2-yl, halo pyridine-3-yl, phenyl-pyridine-2-yl, phenyl-pyridine-3-yl, and phenyl-pyridine-4-yl. 15 In another embodiment of the compound of Fonnula (1), Li is -N(H)-C(O)-, and Q1, G1, L 6 , Q6, L7, and G6 are taken together to form a group selected from the group consisting of: cinnoline-3-yl, quinoxaline-2-yl, [1,8]Naphthyridine-2-yl, cyano-phenyl, (cyano-phenyl) phenyl, (pyridin-3-yl)-phenyt, (pyridin-4-yl)-phenyl, (haloalkyl-pyridin-2-yl)-phenyl, (haloalkyl-phenoxy)-phenyl, 2-(alkoxycarbonyl)-2,3,4,9-tetrahydro-IH-beta-carboline-3-yl, 20 1-phenyl-2-(alkoxycarbonyl)-2,3,4,9-tetrahydro-1H-beta-carboline-3-yl, 2,3,4,9-tetrahydro 1H-beta-carboline-3-yl, 6-(phenyl)-pyrimidine-4-yl, 6-(halophenyl)-pyrimidine-4-yl, 2,6 dialkoxy-pyrimidine-4-yl, thieno[2,3-c]pyridinc-5-yl, thieno[3,2-c]pyridine-6-yl, 2-alkyl imidazo[1,2-a]pyridine-3-yl, imidazo[1,2-a]pyridine-2-yl, 5-alkyl-inidazo[1,2-a]pyridine-2 yl, imidazo[2,1-b]thiazole-6-yl, and 8-alkyl-imidazo[1,2-a]pyridine-2-yl. 25 In another embodiment of the compound of Formula (I), A is -N(R 5 )-, and
R
1 or R 4 is -L 8 -Q-L-Gs-LIr-Qs-Lpj-Gis, wherein
L
8 is a direct bond, -CH 2 -, -C(O)-O-, -C(O)-N(R 2 6 )-, or -N(R26)-C(O)-, Q8 is a direct bond or alkylene, 30 L9 is a direct bond, GS is imidazole, 4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridine, imidazolepyridine, piperidine, 1,2,3,4-tetrahydroisoquinoline, phenyl, cycloalkyl, purine, isoquinoline, benzimidazole, oxazole, pyrrazole, pyrimidine, 4,5,6,7 13 WO 2006/099379 PCT/US20061009049 tetrahydro-1H-benzimidazole, or furan, wherein Gs is substituted or unsubstituted, LIS is a direct bond, -SO 2 -, -0-, or -C(O)-, Qig is a direct bond or alkylene, 5 L 1 9 is a direct bond, and G13 is hydrogen, phenyl, quinoline, isoquinoline, cycloalkyl, pyridine, furan, alkyl, pyrrole, thiazole, imidazole, thiophene, or pyrrolidine, wherein
G
1 g is substituted or unsubstituted. In another embodiment of the compound of Fonnula (I), A is -N(R 5 )-, and .0 R 1 or 14 is -LS-QS-Lr-GrLiS-QIs-L9-Gis, wherein L9, Gs, Lis, Qis, Lig, and Gi 8 are taken together to form a group selected from the group consisting of: piperidin-4-yl, and 1 (alkoxycarbonyl)-piperidin-4-yl. In another embodiment of the compound of Formula (I), A is -N(R 5 )-, and R, or R 1 4 is -Ls-Qs-L-G-LiS-Q is-LiS-Gis, wherein L), Gs, Lis, Q is, L 1 9 , and Gis are taken together to [5 form a group selected from the group consisting of: 1H-pyrazol-3-yl, imidazol-2-yl, imidazol-l-yl, 4,5-dialkyl-imidazol-2-yl, 4-phenyl-f1H-imidazol-2-yl, 4-(halo-phenyl)-1H imidazol-2-yl, 5-adamantan-1-yl-1H-imidazol-2-yl, 5-alkyl-1H-imidazol-2-yl, 5-benzyl-1H imidazol-2-yl, 4-alkyl-oxazol-2-yl, and furan-2-ylmethyl. In another embodiment of the compound of Formula (I), A is -N(R 5 )-, and RI or l4 is 20 -Ls-Qg-L-Gs-Ls-QIS-L 9 -Gis, wherein L9, Gs, Lis, Qis, Li, and G 1 8 are taken together to form a group selected from the group consisting of: cycloalkyl, alkyl, 2-alkylsulfanyl-ethyl, (alkoxy-phenyl)-ethyl, (alkoxy-phenyl)-methyl, 2-(alkoxyphenyl)-ethyl, 2-(phenoxy-phenyl) ethyl, 1-(alkylsulfonyl-phenyl)-ethyl, benzyl, cyano-benzyl, alkoxy-benzyl, alkyl-benzyl, alkylsulfonyl-benzyl, dihalo-benzyl, haloalkyl-benzyl, haloalkyl-halo-benzyl, haloalkoxy 25 benzyl, halo-benzyl, sulfamoyl-benzyl, alkoxyphenyl, halo-phenyl, and alkylsulfonyl-phenyl. In another embodiment of the compound of Formula (I), A is -N(Rs)-, and Ri or R4 is -L-Qs-Lq-Gr-Lr-Q-LirGis, wherein L 9 , G 8 , Lis, Q18, Lig, and G 18 are taken together to form a group selected from the group consisting of: benzimidazole-2-yl, benzimidazole-S-yl, benzimidazole-6-yl, isoquinoline-3-yl, isoquinoline-6-yl, 4,5,6,7-tetrahydro-benzimidazole-2 30 yl, 4,5,6,7-tetrahydro-benzothiazole-2-yl, 5,5-dialkyl-7-oxo-4,5,6,7-tetrahydro-benzothiazol 2-yl, 6-(alkyl)-4,5,6,7-tetrahydro-benzothiazol-2-yl, 1,2,3,4-tetrahydro-isoquinolin-6-yl, 2 (alkoxycarbonyl)-1,2,3,4-tetrahydro-isoquinolin-6-yl, 7H-purin-8-yl, and 2-amino-pyrimidin 4-yL 14 WO 2006/099379 PCT/US2006/009049 In another embodiment of the compound of Fonnula (I), A is -N(Rs)-, and R 1 or R 4 is
-L
8 -Qg-L 9
-G
8
-L
18 -Qis-Liv-Gis, wherein Qs, L9, G 8 , Lis, Qis, L 19 , and Gig are taken together to form a group selected from the group consisting of: (piperidin-3R-yl)methyl, (piperidin-3S yl)methyl, (piperidin-4-yl)-methyl, (1 -(alkoxycarbonyl)-pip eridin-3 -yl)-methyl, (1 5 (cycloalkylnethyl)-piperidin-3-yl)methyl, (1-(cycloalkylmethyl)-piperidi-4-yl)methy1, (piperidin-4-yl)-mcthyl, (1-(alkoxycarbonyl)-piperidin-4-yl)-methyl, [1-(carboxylic acid methyl ester)-piperidin-3R-yl]methyl, [I -(carboxylic acid methyl ester)-piperidin-3 S yl]methyl, [1-(cycloalkylmethyl)-piperidin-3S-yl]methyl, [1-(cycloalkyhmethyl)-piperidin 3R-yl]methyl, 1-(1-alkyl-IH-imidazol-2-ylmethyl)-piperidin- 3 -ylmethyl, 1-(1-alkyl-1H 10 imidazol-4-ylmethyl)-piperidin-3-ylmethyl, 1-(IH-imidazol-2-ylnethyl)-piperidin-3 yhmethyl, 1-(cycloalkylmethyl)-piperidin-3R-yljImethyl, 1-{1-(alkoxycarbonyl)-pyrrolidine 2R-ylmethyl)-piperidin-3-ylmethyl, 1-[1-(alkoxycarbonyl)-pyrrolidine-2S-yl1methyl] piperidin-3 -ylmethyl, 2-(piperidin-3 -yl)-ethyl, 2-(piperidin-4-yl)-ethyl, 2-[ 1 (alkoxycarbonyl)-piperidin-3-yl]-ethyl, and 2-f1-(alkoxycarbonyl)-piperidin-4-yl]-ethyl. 15 In another embodiment of the compound of Formula (I), A is -N(R 5 )-, and R2 or R4 is -LS-Q-LrGS-LI-Qjs-LI 4 is, wherein Qs, L9, Gs, Lig, Qjs, Ls, and Gig are taken together to form a group selected from the group consisting of: 4,5,6,7-tetrahydro-thiazolo[5,4-C]pyridin 2-yl, 3H-inidazo[4,5-clpyridin-2-yl, 3H-imidazo[4,5-c]pyridin-2-yl, [5-(2,4-dialkoxy benzyl)-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-yl, (5-alkyl-3H-imidazo1-4-ylmethyl) 20 4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-yl, 5-[l-(alkoxycarbonyl)-pyrrolidin- 2
S
ylmethyl]-4,5,6,7-tetrahydro-thiazolo(5,4-c]pyridin-2-yl, 5-[1-(alkoxycarbonyl)-pyrrolidin 2R-ylmethyl]-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-yl, 5-(1-alkyl-1H-imidazol-2 ylnethyl)-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-yl, 5-(1-alkyt-1H-pyrrol-2-ylmethyl) 4,5,6,7-tetrahydro-thiazolo[5,4-clpyridin-2-yl, 5-(3-alkylsulfanyl-propyl)-4,5,6,7-tetrahydro 25 thiazolo[5,4-c]pyridin-2-yl, 5-(3-alkylsulfinyl-propyl)-4,5,6,7-tetrahydro-thiazolo{5,4 c]pyridin-2-yl, 5-(3-alkylsulfonyl-propyl)-4,5,6,7-tetrahydro-thiazolo{5,4-c]pyridin-2-yl, 5 (3H-imidazol-4-ylmethyl)-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-yl, 5-(alkoxy-benzyl) 4,5,6,7-tetrahydro-thiazolo{5,4-c]pyridin- 2 -yl, 5-(alkylsulfonyl)-4,5,6,7-tetrahydro thiazolo[5,4-c]pyridin-2-yl, 5-(alkoxycarbonyl)-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-y1, 30 5-(cycloalkylmethyl)-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-yl, 5-(halo-benzyl)-4,5,6,7 tetrahydro-thiazolo[5,4-c]pyridin- 2 -yl, 5-(imidazol-2-ylmethyI)-4,5,6,7-tetrahydro thiazolo[5,4-c]pyridin-2-yl, 5-(quinolin-3-ylmethyl)-4,5,6,7-tetrahydro-thiazolo[5,4 c]pyridin-2-yl, 5-alkyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-yl, 5-alkylcarbamoyl 4,5,6,7-tetrahydro-thiazolo(5,4-c]pyridin-2-yl, 5-alkylsulfonyl-4,5,6,7-tetrahydro 15 WO 2006/099379 PCT/US20061009049 thiazolo[5,4-clpyridin-2-y1, 5-benzoyl-4,5,6,7-tetrahydro-thiazo1o[5,4-c]pyridin-2-yl, 5 benzyl-4,5,6,7-tetrahydro-thiazolo[5,4-c)pyridin-2-yl, 5-cycloalkylnethyl-4,5,6,7-tetrahydro thiazolo[5,4-o]pyridin-2-yI, 5-furan-3-ylmethyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-yl, 5-phenethyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-y, 5-pyridin-3-ylmethyl-4,5,6,7 5 tetrahydro-thiazolo[5,4-c]pyridin-2-yl, 5-pyrrolidin-28-ylmethyl-4,5,6,7-tetrahydro thiazolo[5,4-c]pyridin-2-yl, 5-thiazol-2-ylhnethyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2 yl, 5-thiophen-2-ylmethyl-4,5,6,7-tctrahydro-thiazolo[5,4-c]pyridin-2-yl, and 5-thiophen-3 ylmethyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-yl. In another embodiment, the present invention provides a compound of Formula (Ia) Ri R2 N I \> L-Q~ R iN 10 4 (Ia) wherein
L
1 is selected from the group consisting of: a direct bond, -CH 2 -, -0-, -N(R 6 )-, -C(O)-, -CON(R 6 )-, -N(R 6
)C(O)
15 , -N(RJ)CON(R 7 )-, -N(R 6 )C(O)O-, -OC(O)N(Ro)-, -N(R 6
)SO
2 -, -SO 2
N(R
6 )-, -C(0)-0 , -0-C(O)-, -S-, -S(O)-, -S(0) 2 -, and -N(R,)S0 2
N(R
7 )-, wherein R 6 and R 7 are independently selected from the group consisting of: hydrogen, -alkyl, -aryl, and -alkylene-aryl; Q, is selected from the group consisting of direct bond and alkylene; 20 G1 is selected from the group consisting of: aryl, heteroaryl, fused arylcycloalkyl, fused cycloalkylaryl, fused cycloalkylheteroaryl, fused heterocyclylaryl, and fused heterocyclylheteroaryl group, and G1 is optionally substituted 1 to 7 times, wherein the substituents are independently selected from the group consisting of: 25 a) -halo; b) -cyano; c) -nitro; d) -perhaloalkyl; 16 WO 2006/099379 PCT/US20061009049 e) -Rs; f) -L2-RR; g) -L 2 -Qz-Rg; and 1) -Qr LrRs; 5 wherein Rs is selected from the group consisting of hydrogen, -alkyl, -aryl, and alkylene aryl; Q2 is selected from the group consisting of alkylene, alkenylene, and alkynylene;
L
2 is selected from the group consisting of -CHr, -0-, -N(R)-, -C(O)-, 10 CON(R 9 )-, -N(R 9 )C(O)-, -N(R 9 )CON(RI)-, -N(R)C(0)O-, -OC(O)N(R) , -N(R 9 )S0 2 -, -SO 2
N(R
9 )-, -C(O)-O-, -0-C(O)-, -S-, -S(O)-, -S(0)2-, and -N(R 9
)SO
2 N(Ro)-, wherein R9 and RIO are independently selected fl-om the group consisting of: hydrogen, -alkyl, -aryl, and -alkylene-aryl; 15 R1, R2, R3, and R 4 are independently selected from the group consisting of a) -H; b) -alkyl; c) -aryl; d) -alkylene-aryl; 20 e) -K-alkyl; f) -K-aryl; g) -K-alkylene-aryl; and h) -L 3
-G
2
-G
3 ; wherein 25 K is selected from the group consisting of: -C(O)-0-, -0-C(O)-, -C(0)-NH-, NH-C(O)-, -S02-, -SO 2 -NH-, -NH-SO 2 -, and -C(O)-;
L
3 is selected from the group consisting of: a direct bond, -CH 2 -, -0-, -N(R 26 )-, -C(O)-, -CON(R 26 )-, -N(R 26 )C(O)-, -N(R26)CON(R27)-, -N(R 26 )C(0)Q , -OC(O)N(R 26 )-, -N(R 2 6
)SO
2 -, -SO 2
N(R
2 6 )-, -C(O)-O-, -0-C(O)-, -S-, 30 -S(O)-, -S(O) 2 -, and -N(R 26
)SO
2 N(R27) wherein R26 and R 27 are independently selected from the group consisting of: -hydrogen, -alkyl, -aryl, and -alkylene-aryl; G2 is selected from the group consisting of: a direct bond, and 17 WO 2006/099379 PCT/US2006/009049 L 110 L1 wherein L 0 is selected from the group consisting of alkyline, cycloalkyline, heteroaryline, aryline, and heterocyclyline; 5 L12 is selected from the group consisting of -0-, -C(O)-N(R)- , C(O)-O-, -C(O)-, and -N(Rni)-CO-N(R 2 )-, wherein R, and R 12 independently comprise hydrogen, -aryl, -alkyl, and -alkylene-aryl; L , is selected from the group consisting of hydrogen, -alkyl, -alkenyl, -alkynyl, -aryl, -alkylene-aryl, -alkylene -heteroaryl, alkylene-O 10 alkylene-aryl, -alkylene-S-alkylene-aryl, -alkylene-0-alkyl, alkylene-S-alkyl, -alkylene-NH 2 , -alkylene-OH, -alkylene-SH, alkylene-C(O)-ORis, -alkylene-C(O)-NR13R4, -alkylene-NR1 3 R14, -alkylene-N(R 3
)-C(O)-R
1 4 , -alkylene-N(R 3
)-S(O)-R
1 4 , and the side chain of a natural or non - natural amino acid, wherein 15
R
13 and R 1 4 independently comprise hydrogen, -aryl, -alkyl, and -alkylene-aryl; or R 13 and R 4 may be taken together to fonn a ring having the formula - (CH2)q-Y-(CH2)r bonded to the nitrogen atom to which R 3 and R 14 are attached, wherein q and r are, 20 independently, 1, 2, 3, or 4; Y is -CHr, -C(O)-, -0-, -N(H)-, -S-, -S(O)-, -SOr, -CON(H)-, -NHC(O)-, -NHCON(H)-, NHSO 2 -, -SO 2 N(H)-, -(O)CO-, -NHSO 2 NH-, -OC(O)-, N(R 1 5 )-, -N(C(O)R 15 )-, -N(C(O)NHR 15 )-, -N(SO 2
NHR
1 5 )-, -N(S0 2
RI
5 )-, and -N(C(O)OR 5 )-, wherein R 15 is selected 25 from the group consisting of hydrogen, -alkyl, -aryl, and alkylene-aryl; or
R
3 and Rt 4 may be taken together, with the nitrogen atom to which they are attached, to fonn a heterocyclyl or heteroaryl ring; and 30 G3 is selected from the group consisting of hydrogen, alkyl, aryl, heteroaryl, fused arylcycloalkyl, fused cycloalkylaryl, fused cycloalkylheteroaryl, fused heterocyclylaryl, and fused heterocyclylheteroaryl group, wherein 18 WO 2006/099379 PCT/US2006/009049 G3 is optionally substituted 1 to 7 times, wherein the substituents are independently selected from group consisting of: a) -halo; b) -cyano; S c) -nitro; d) -perhaloalkyl; e) -R 16 ; f) -L 4
-R]
6 ; g) -L 4
-Q
4
-R
1 6 ; and 10 h) -Q 4
-L
4 -Rii; wherein
R[
6 is selected from the group consisting of hydrogen, -alkyl, -aryl, and -alkylene-aryl; Q4 is selected from the group consisting of alkylene, alkenylene, 15 and alkynylene;
L
4 is selected from the group consisting of -CH 2 -, -0-, -N(RLS)-, C(O)-, -CON(Ria)-, -N(Ris)C(O)-, -N(R is)CON(Rig) -N(Ris)C(0)0-, -OC(O)N(Ris)-, -N(RI 8
)SO
2 -, -SO 2 N(Ris) -C(0)-0-, -O-C(O)-, -S-, -S(O)-, -S(0)r, 20 and -N(Ris)S0 2 N(Riq)-; wherein Ris and Rig are independently selected from the group consisting of: -hydrogen, -alkyl, -aryl, and -alkylene-aryl; and 25 wherein at least one of R,-R 4 is the group -L 3
-G-G
3 ; and
R
5 is selected from the group consisting of: a) -hydrogen; and b) -alkyl; and wherein the aryl and/or alkyl group(s) in Ri to R27, LI to L 12 , GI to G3 may be optionally 30 substituted 1-4 times with a substituent selected from the group consisting of: a) -H; b) -halo; c) -hydroxyl; d) -cyano; 19 WO 20061099379 PCT/US2006/009049 e) -carbamoyl; f) -carboxyl; g) -Z-alkyl; h) -Z-aryl; 5 i) -Z-alkylene-aryl; wherein Z is selected from the group consisting of -CH-, -0-, -N(H), -S-, SO 2 -, CON(H)-, -NHC(O)-, -NHCON(H)-, -NHSOr, -SO 2 N(H)-, -C(O)-O-, -NHSO 2 NI-, and -0-CO-, or a phannaceutically acceptable salt, ester, or prodrug thereof. 10 In an embodiment of the compound of Formula (Ia), Li is -NH-C(O)-, -NH-, or a direct bond; Q1 is a direct bond; Gi is phenyl, biphenyl, naphthyl, indole, isoquinoyl, pyridine, or pyrimidine; and wherein 15 at least one of R-R 4 is the group -L 3
-G
2
-G
3 , wherein
L
3 is selected from the group consisting of a) -C0r; b) -C(O)NH-; and c) -NH 20 G2 is a direct bond, or alkylene; and G3 is selected from the group consisting alkyl, phenyl, naphthyl, biphenyl, alkylene-phenyl, pyrole, thiophene, indole, imidazole, tetrazole, thiazole, 1,3,4-thiadiazole, pyrimidine, pyridine, benzimidazole, and benzthiazole, wherein G3 is optionally substituted 1 to 7 times with a group selected from 25 the group consisting of: a) -halo; b) -cyano; c) -nitro; d) -perhaloalkyl; 30 e) -R 16 ; 1) -L 4 -Ri 6 ; g) -L 4
-Q
4
-R
6 ; and h) -Q 9
-L
4
-R
6 ; wherein 20 WO 2006/099379 PCT/US2006/009049
R
16 is selected from the group consisting of hydrogen, -alkyl, -ayl, and -alkylene-aryl; Q4 is selected from the group consisting of -alkylene, -alkenylene, and -alkynylene; 5 L4 is selected from the group consisting of-CH 2 -, -0-, -N(Rig)-, C(O)-, -CON(Ris)-, -N(Ris)C(O)-, -N(Ris)CON(R,) -N(Ris)C(O)O-, -OC(O)N(Ris)-, -N(RIS)SO 2 -, -SO 2
N(RI,)
-C(O)-O-, -O-C(O)-, -S-, -S(O)-, -S(O) 2 -, and -N(Rig)SO 2 N(Rg)-; 10 wherein Ris and R 19 are independently selected from the group consisting of: hydrogen, -alkyl, -aryl, and -alkylene-aryl. In another embodiment of the compound of Formula (Ia), LI is a direct bond, -NH-C(O)-, or -NH-; 15 Q is a direct bond; Gi is phenyl, biphenyl, isoquinoyl, pyridine, or pyrimidine; and at least one of R 1
-R
4 is the group -L3-G2-G3, wherein L3 is -C(O)-NH-; L L Ito. G2 is ; 20 wherein Lio is defined as above,
L,
1 is defined as above, and L12 is -C(O)-NH-; and G3 is selected from the group consisting alkyl, phenyl, naphthyl, biphenyl, 25 alkylene-phenyl, pyrole, imidazole, tetrazole, thiazole, 1,3,4-thiadiazole, pyrimidine, pyridine, benzimidazole, and beazthiazole, wherein G3 is optionally substituted 1 to 7 times with a group selected from the group consisting of a) -halo; 30 b) -cyano; c) -nitro; 21 WO 2006/099379 PCT/US2006/009049 d) -perhaloalkyl; e) -Ri6; f) -L4-Ri 6 ; g) -L 4
-Q
4
-R
6 ; and 5 h) -Q 2
-L
4 -Ri 6 ; wherein
R
16 is selected from the group consisting of hydrogen, -alkyl, -aryl, and -alkylene-aryl; Q4 is selected from the group consisting of alkylene, alkenylene, 10 and alkynylene;
L
4 is selected from the group consisting of -CH 2 -, -0-, -N(RIg)-, C(O)-, -CON(Ris)-, -N(R 1 )C(O)-,.-N(Ris)CON(R 19
)
-N(Ris)C(0)O-, -OC(O)N(RIs)-, -N(Rig)SOr, -SO 2
N(R
1 s) -C(O)-O-, -0-C(O)-, -S-, -S(O)-, -S(0)2-, 15 and -N(R 1 s)SO 2 N(Rig)-; wherein Ri 8 and Ri 9 are independently selected from the group consisting of: hydrogen, -alkyl, -aryl, and -alkylene-aryl In another embodiment of the compound of Formula (Ia), G3 is imidazole or 20 benzimidazole. In another embodiment of the compound of Formula (Ta), G, is isoquinoline. In another embodiment of the compound of Formula (Ia), the compound of Formula (Ia) has the formula N N G-G2N \P H 0 25 wherein G1 is unsubstituted 2-isoquinoline-3yl or pyridin-2yl; and G2 and G3 together forn a group selected from phenyl, isobutyl, n-butyl, 1H-iinidazol-2-yl, [1,3,4]-thiadiazol-2yl, thiazol-2y1, 1H-imidazol-2-yl, benzothiazol-2-yl, pyridin-2-yl, pyridin-3 -yl, and 4-phenyl- lH-imidazol-2-yl. 30 22 WO 2006/099379 PCT/US2006/009049 In another embodiment of the compound of Formula (Ia), the compound of Formula (Ia) has the formula O L N 0
L
11 I N G-N N N N H H O H wherein 5 L I is 2,2 dimethyl propyl, 2-chloro-benzyl, 2-fluorobenzy, 2-phenyl-ethyl, 3,4,5-trifluoro benzyl,3,5-difluoro -benzyl, 3-chloro benzyl, 3-fluoro benzyl, 3-fluoro -phenyl, 3 methoxy phenyl, 3-trifluoromethyl benzyl, 4-chloro benzyl, 4-fluoro benzyl, 4 methoxy phenyl, 4-methyl benzyl, 4-phenyl-benzyl, Benzyl, Butyl-2yl, Isobutyl, Isopropyl, cyclopropyl, or phenyl, 10 n is 0 or 1 and G3 is IIH -benzimidazol-2-yl, 1H-imidazol-2-yl, pyridin-2yl, or thiazol-2yl. In another embodiment of the compound of Formula (Ia), the compound of Formula (Ia) has the formula H \G 3 L11 N H a N a N N N H 15 wherein
L,
1 is benzyl or 3,5-difluorobenzyl, and G3 is methyl, isobutyl, lH-imidazol-2yl, tbiazol-2yl, benzthiazol-2yl, IH-Benzimidazol-2-yl, 2-methyl-piperidine-1-carboxylic acid tert-butyl ester, or 3-methyl-piperidine-l carboxylic acid tert-butyl ester. 20 In another embodiment of the compound of Formula (Ia), the compound of Formula (1a) has the formula 23 WO 2006/099379 PCT/US2006/009049 N NG N H H H wherein G is unsubstituted 2-isoquinolin-3-yl, 5-bromopyridin-2-yl, phenyl, or pyridin-2-yl; 5 G2 and G3 are taken together to form a group selected from the group consisting of (benzthiazol-2yl)methyl, 1 H-imidazol-2-yl, 2,4-dichlorobenzyl, 2,5-dichlorophenyl, 3-hydroxypropyl, 3-tert-butylphenyl, 4'biphenyl, 4-chlorophenyl, 4-fluorophenyl, 4 methoxyphenyl, 4-methyl-benzyl, 4-phenoxyphenyl, 4-phenylthiazol-2yl, 4-phenyl thiazol-2-yl, 4-tert-butyl-benzyl, benziH-imidazol-2-yl, benzothiazol-6-yl, [0 benzothiazol-2yl, benzyl, methyl, phenyl, pyridin-2-yl, and thiazol-2-yl. In another embodiment of the compound of Formula (Ia), the compound of Formula (la) has the formula N H II G N G / N \N 3 1 P'H O H 0 G1 is 1,5-dimethyl- indole-2yl, 1-benzyl- indole-2-yl, IH-imidazol-2-yl, IH-Indazoline-3yl, 15 1HW-indole-2y1, 1i-methyl-Indazoline-3yl, I-methyl-indole-2-yl, 1-n-propyl- indole-2y1, 2,4-difluorophenyl, 2-Chloro-phenyl, 2-fluoro-phenyl, 2-isoquinolin-3-yl, 3,4 dimethoxyphenyl, 3-Chloro-pheny, 3-fluoro-phenyl, 3-methoxy-4-fluoro-phenyl, 4' biphenyl, 4-fluorobenzyl, 4-fluorophenyl, 4-isopropyl-phenyl, 4-inethoxy-phenyl, 4 nitro-phenyl, 4-phenyoxyphenyl, 4-tert-butyl-phenyl, 4-trifluoromethylphenyl, 5 20 Chloro-benzofuran-2-yl, 5-methyl-indole-2-y, 5-phenoxy-pyridin-2-yl, 6,7 dimethoxy-2-isoquinolin-3-yl, benzofuran-2-yl, benzothiophene-2-yl, furan-3yl, naphthalin-2-yl, phenyl, quinolin-2yl, or quinolin-3yl; and G2 and G3 are taken together to form a group selected from the group consisting of (1 methylbenzimidazole-2-yl)mcthyl, (1R)-phenyl ethyl, (1S)-phenyl ethyl, (5 25 methylfuran-2-yl)-methyl, (benzothiophene-2-yl)methyl, (benzthiazol-2-yl)methyl, (thiophene-2-yl)ethyl, (thiophene-2-yl)methyl, {1,3,4}-thiadiazol-2-yl, 111 benziimidazol-2-yl, IH-benzimidazol-2-yl, 1H-benzimidazol-2-yl, 1H-benzimidazole 24 WO 2006/099379 PCT/US2006/009049 2-yl methyl, 1H-imidazol-2-yl, 1H-indazole-5-yl, 2-(1H-benzimidazole-2-yl)-ethyl, 2,3,4,9-tetrahydro-1H-beta-carboline-2-yl, 2-phenyoxy-phenyl, 3-phenoxy-phenyl, 4 phenoxy-phenyl, 4-phenyl-1H-imidazol-2-yl, benzthiazol-2yl, benzyl, ethyl, furan-2 yl methyl, phenethyl, phenyl, pyridin-2-yl, pyridin-3-yl, quinolin-3-yl, tert-butyl, 5 thiazol-2-yl, and thiazole-2yl-methyl. In another embodiment of the compound of Formula (Ia), the compound of Formula (Ia) has the formula N NH 0 O N 0 1 N r~ N N H H 0 G3G2 wherein, 10 G2 and G3 are taken together to form a group selected from the group consisting of isopropyl, isobutyl, and phenyl. In another embodiment of the compound of Formula (Ia), the compound of Formula (1a) has the formula RI R2 N H I \>-N Gi R N >G 3 \0 R5 o NH G1 G3 15 wherein R 1 , R2, R 3 , R 5 , G1, G2, and G3 are defined as above for the compound of Formula (Ia). 25 WO 2006/099379 PCT/US2006/009049 In another embodiment of the compound of Formula (Ia), the compound of Formula (Ia) has the formula Ri R2 N H H N G 1 G3-G N N 2 \0 0 R 4 R5 wherein R 1 , R2, R 3 , Rs, GI, G2, and G 3 are defined as above for the compound of Formula (Ia). 5 In another embodiment of the compound of Fonnula (La), the compound of Fonnula (Ia) has the formula O G N R
G
2 ,N N H H N Gi R N S 3 \0 R5 O NH G IG wherein R1, R3, R 5 , G1, G 2 , and G 3 are defined as above for the compound of Formula (Ia). In another embodiment, the present invention provides a compound of Formula (Ib) Ri R2 N R N R R1 10 (Ib) wherein L, is -N(R 6 )-, wherein R 6 is selected from the group consisting of: hydrogen, -alkyl, -aryl, and -alkylene-aryl; 15 Q i is a direct bond; Gi is hydrogen:
R
1 , R2, R3, and R4 are independently selected from the group consisting of a) -H; 26 WO 2006/099379 PCT/US2006/009049 b) -alkyl; c) -aryl; d) -alkylene-aryl; e) -K-alkyl; 5 f) -K-aryl; g) -K-alkylene-aryl; and h) -L-G 2
-G
3 ; wherein at least one of RI-R 4 is not hydrogen; and wherein [0 K is selected from the group consisting of: -C(O)-O-, -O-C(O)-, -C(O)-NH-, NH-C(O)-, -SO 2 -, -S0 2 -NH-, -NH-SO 2 -, and -C(O)-;
L
3 is selected from the group consisting of: a direct bond, -CH 2 -, -0-, -N(R 2 )-, -C(O)-, -CON(R 26 )-, -N(R 26 )C(O)-, -N(R 2 r)CON(R27)-, -N(R 26 )C(O)0
-OC(O)N(R
26 )-, -N(R 26 )S0 2 -, -SO 2
N(R
26 )-, -C(O)-O-, -O-C(O)-, -S-, 15 -S(O)-, -S(0)-, and -N(R 26
)SO
2 N(R27) wherein R 2 6 and R27 are independently selected from the group consisting of: -hydrogen, -alkyl, -aryl, and -alkylene-aryl; G2 is selected from the group consisting of: a direct bond, and LL ; 110 20 L wherein
L
1 0 is selected from the group consisting of alkyline, cycloalkyline, heteroaryline, aryline, and heterocyclyline;
L
1 2 is selected from the group consisting of -0-, -C(O)-N(RI I)- , 25 C(O)-O-, -C(O)-, and -N(R i)-CO-N(Rz)-, wherein R, 1 and R 12 independently comprise hydrogen, -aryl, -alkyl, and -alkylene-aryl; L 1 is selected from the group consisting of hydrogen, -alkyl, -alkenyl, -alkynyl, -aryl, -alkylene-aryl, -alkylene -heteroaryl, alkylene-O alkylene-aryl, -alkylene-S-alkylene-aryl, -alkylene-O-alkyl, 30 alkylene-S-alkyl, -alkylene-NH 2 , -alkylene-OH, -alkylene-SH, alkylene-C(O)-OR 13 , -alkylene-C(O)-NR 3 Ri 4 , -alkylene-NR 13 R]4, 27 WO 2006/099379 PCT/US2006/009049 -alkylene-N(Ru)-C(O)-RI4, -alkylene-N(R )-S(0) 2
-RI
4 , and the side chain of a natural or non. - natural amino acid, wherein
RI
3 and R[ 4 independently comprise hydrogen, -aryl, -alkyl, and -alkylene-aryl; or 5 R 13 and R 1 4 may be taken together to form a ring having the formula - (CH 2 )-Y-(CH2) bonded to the nitrogen atom to which R 13 and R 14 are attached, wherein q and r are, independently, 1, 2, 3, or 4; Y is -CH 2 -, -C(O)-, -0-, -N(H)-, -S-, -S(O)-, -902-, -CON(H)-, -NHC(O)-, -NHCON(H)-, 10
NHSO
2 -, -SO 2 N(H)-, -(0)CO-, -NHSO 2 NH-, -OC(O)-, N(R 5 )-, -N(C(O)RI 5 )-, -N(C(O)NHRis)-, -N(SO 2
NHR
15 )-, -N(S0 2
R
5 )-, and -N(C(O)OR 15 )-, wherein R, 5 is selected from the group consisting of hydrogen, -alkyl, -aryl, and alkylene-aryl; or 15 RU and R 14 may be taken together, with the nitrogen atom to which they are attached, to form a heterocyclyl or heteroaryl ring; and G3 is selected from the group consisting of hydrogen, alkyl, aryl, heteroaryl, fused arylcycloalkyl, fused cycloalkylaryl, fused cycloalkylheteroaryl, 20 fused heterocyclylaryl, and fused heterocyclylheteroaryl group, wherein G3 is optionally substituted 1 to 7 times, wherein the substituents are independently selected from group consisting of: a) -halo; b) -cyano; 25 c) -nitro; d) -perhaloalkyl; e) -R 16 ; f) -L4-Ri6; g) -L 4
-Q
4 -Rt6; and 30 h) -Q 4
-L
4
-RI
6 ; wherein
R
16 is selected from the group consisting of hydrogen, -alkyl, -aryl, and -alkylene-aryl; 28 WO 2006/099379 PCT/US2006/009049 Q4 is selected from the group consisting of alkylene, alkenylene, and alkynylene;
L
4 is selected from the group consisting of -CH 2 -, -0-, -N(R 8 )-, C(O)-, -CON(Ris)-, -N(R g)C(O)-, -N(Ris)CON(R 1 9) 5 , -N(Rs)C(0)O-, -OC(O)N(RI 8), -N(R s)SO-, -SO 2 N(Ria) -C(0)-O-, -O-C(O)-, -S-, -S(O)-, -S(0)2-, and -N(Ris)SO 2 N(Riv)-, wherein R, 8 and R, are independently selected from the group 0 consisting of: -hydrogen, -alkyl, -aryl, and -alkylene-aryl; and R5 is selected from the group consisting of: a) -hydrogen; and b) -alkyl; and 15 wherein the aryl and/or alkyl group(s) in R, to Rig, Li to L 12 , G 2 and G3 may be optionally substituted 1-4 times with a substituent selected from the group consisting of: a) -H; b) -halo; c) -hydroxyl; 20 d) -cyano; e) -carbamoyl; f) -carboxyl; g) -Z-alkyl; h) -Z-aryl; 25 i) -Z-alkylene-aryl; wherein Z is selected from the group consisting of -CH 2 -, -0-, -N(H), -S-, S02, CON(H)-, -NHC(0)-, -NHCON(H)-, -NHSO-, -SO 2 N(H)-, -C(O)-O-, -NHSO 2 NH-, and -0-CO-, or a pharmaceutically acceptable salt, ester, or prodrug thereof 30 In an embodiment of the compound of Formula (Tb), L I is -NH-C(O)-, -NH-, or a direct bond; and at least one of R 1
-R
4 is the group -L 3
-G
2
-G
3 , wherein
L
3 is selected from the group consisting of a) a direct bond; 29 WO 20061099379 PCT/US2006/009049 b) -CO-; c) -C(O)NH-; and d) -NH G2 is a direct bond, or alkylene; and 5 G3 is selected from the group consisting alkyl, phenyl, naphthyl, biphenyl, alkylene-phenyl, pyrole, thiophene, indole, imidazole, tetrazole, thiazole, 1,3,4-thiadiazole, pyrimidine, pyridine, benzimidazole, and benzthiazole, wherein G3 is optionally substituted 1 to 7 times with a group selected from the group consisting of: 10 a) -halo; b) -cyano; c) -nitro; d) -perhaloalkyl; e) -R 1 6 ; 15 f-L4-Ri6; g) -L4-Q4-Ris; and h) -Q 2
-L
4 -Ri; wherein
R
16 is selected from the group consisting of hydrogen, -alkyl, -aryl, 20 and -alkylene-aryl; Q4 is selected from the group consisting of -alkylene, -alkenylene, and -alkynylene;
L
4 is selected from the group consisting of -CI 2 -, -0-, -N(Ris)-, C(0)-, -CON(Rig)-, -N(Ris)C(O)-, -N(Ris)CON(R 19
)
25 , -N(R 1 s)C(O)0-, -OC(0)N(Rs)-, -N(Ris)SO 2 -, -SO 2 N(Ris) -C(O)-0-, -O-C(O)-, -S-, -S(O)-, -S(O) 2 -, and -N(RIg)SO 2
N(R
19 )-; wherein Rig and Rig are independently selected from the group consisting of: hydrogen, -alkyl, 30 -aryl, and -alkylene-aryl. In the compounds of Formula (I), the various functional groups represented should be understood to have a point of attachment at the functional group having the hyphen. In other words, in the case of -C.
0 alkylene-aryl, it should be understood that the point of attachment 30 WO 2006/099379 PCT/US2006/009049 is the alkylene group; an example would be benzyl. hi the case of a group such as -C(O) NH-C1-oo alkylene-aryl, the point of attachment is the carbonyl carbon. The term "BACE inhibitor" or "inhibitor of BACE" is used to signify a compound having a structure as defined herein, which is capable of interacting with BACE and 5 inhibiting its enzymatic activity. Inhibiting BACE enzymatic activity means reducing the ability of BACE to cleave a peptide or protein. The peptide or protein may be APP, and a BACE inhibitor may reduce the ability of BACE to cleave APP near the NH 2 terminus of APP and produce COOH-terminal fragments (CTFs) that contain the complete AP domain. In various embodiments, such reduction of BACE activity is at least about 50%, at least about 10 75%, at least about 90%, at least about 95%, or at least about 99%. In various embodiments, the concentration of BACE inhibitor required to reduce a BACE's enzymatic activity is less than about 30 pM, less than about 10 pM, or less than about 1 p.M. Also included within the scope of the invention are the individual enantiomers of the compounds represented by Formula () above as well as any wholly or partially racemic 15 mixtures thereof. The present invention also covers the individual enantioners of the compounds represented by Formula (I) above as mixtures with diastereoisomers thereof in which one or more stereocenters are inverted. Unless otherwise stated, structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for 20 the replacement of a hydrogen atom by a deuterium or tritium, or the replacement of a carbon atom by a " 3 C- or 4 C-enriched carbon are within the scope of the invention. In another aspect, the present invention provides a pharmaceutically acceptable salt, solvate, or prodrug of compounds of Formula (I). In an embodiment, the prodrug comprises a biohydrolyzable ester or biohydrolyzable amide of a compound of Fonnula (1). 25 Examples of compounds of Formula (I) of the present invention having potentially useful biological activity are listed by name below in Table 1. The ability of compounds Formula (I) to inhibit the proteolytic activity of BACE was established with representative compounds of Formula (I) listed in Table 1 using the enzyme assay described in the Examples section. The compounds of Formula (I) in Table 1 inhibit BACE in the enzyme 30 assay with an IC 50 of less than or equal to 30 p.M. Examples of compounds of Formula (I) of the present invention are shown in Table 1 and in the Examples section. 31 WO 2006/099379 PCT/US20061009049 Table 1. Ex Structure Name _ 2-Isoquinolin-3-yl-1H 0 N benzoimidazole-4-carboxylic acid NA N \ / (4-phenyl-1H-inidazol-2-yl) amide 2 0 2-Isoquinolin-3-yl-lH N \ / benzoimidazole-5-carboxylic acid isobutyl-amide 3 0 N 2-Pyridin-2-yl-1H NeN benzoimidazole-5-carboxylic acid HH I-i ,A (1 H-bcnzoirnidazol-2-yl)-arnidt 4 0 N 2-Naphthalen-2-yl-IH /_N N N benzoimidazole-5-carboxylic acid - N H (1H-benzoimidazol-2-yl)-amide H 5
H
3 C 0 OH, 2-Isoquinolin-3-yl-lH benzoimidazole-4-carboxylic acid isobutyl-amide 0 /2-Isoquinolin-3-yl-1H N benzoimidazole-4-carboxylic acid phenylamide 32 WO 2006/099379 PCT/US20061009049 7 CH 3 7 02-Isoquinolin-3-yl-IH benzoimidazole-4-carboxylic acid butylamide M 8 2-Isoquinolin-3-yl-1H benzoinidazole-5-carboxylic acid H I[1-cyclopropyl-2-(1H-imidazol-2 ylcarbamoyl)-ethyl]-amide 9 2-Isoquinolin-3-yl-1H benzoimidazole-5-carboxylic acid N \ / [1-(1H-imidazol-2-ylcarbamoyl) cyclopentyl}-amide 10 F 2-Isoquinolin-3-yl-1H H N-<) benzoimidazole-5-carboxylic acid [2-(2-fluoro-phenyl)-iS-(1IH iindazol-2-ycarbanoyl)-ethyl] amide 11 2-Isoquinolin-3-yl-IH 0 N benzoimidazole-5-carboxylic acid N O H [1 S-(I H-inidazol-2-ylcarbamoyl) F ~ ~/2-phenyl-ethyl]-amide N N H 12 2-Isoquinolin-3-yl-1H N- benzoimidazole-5-carboxylic acid N 0l H C [2-(2-chloro-phenyl)- 1 S-(1 H inidazol-2-ylcarbainoyl)-ethyl] amide 33 WO 2006/099379 PCT/US2006/009049 13 2-Isoquinolin-3-yl-1H F 0 HN benzoimidazole-5-carboxylic acid [1-(2-fluoro-phenyl)-2-(IH imidazol-2-ylcarbamoyl)-ethyl] amide 14 2-Isoquinolin-3-yl-lH benzoinidazole-5-carboxylic acid N N10 N\ 1-biphenyl-4-yl-2-(1 H-imidazol N II H N H 2-ylcarbamoyl)-ethyl -amide H 15 F /\F 2 -Isoquinolin-3-yl-1- N \ -{ 111benzoimidazole-5-carboxylic acid H -N 0 [2-(3,5-difluoro-phenyl)-lS-(IHT N imidazol-2-ylcarbamoyl)-ethyl] N aide H 16 2-Isoquinolin-3-yl-1H 9 benzoimidazole-5-carboxylic acid o 0 HN {1R-(lH-imidazol-2 NN ylcarbanoyl)-methyl]-3-phenyl -N N X H propyl)-amide 17 2-Isoquinolin-3-yl-TH benzoimidazole-5-carboxylic acid 0 O HN {2-biphenyl-4-yl-1-[(1H-imidazol z ilN 2-ylcarbamoyl)-methyl]-ethyl} N N 34 WO 2006/099379 PCT/US2006/009049 18 F 2-Isoquinolin-3-yl-1H H
-
benzoinidazole-5-carboxylic acid HN o ~{2-(3-fluoro-pheny1)-I~S-( 1H H imidazol-2-ylcarbanoyl)-ethyl) N aide H 19 0 CH, 2-Isoquinohin-3-yl-lJ benzoimidazole-5-carboxylic acid o 0 H [2-(H-inidazot-2-ylcarbamoyl) N IS -(4-methoxy-phenyl)-ethYl} amide 20 2-Isoquinolin- 3 -yl-1H o 0 5 benzoimidazole-5-arboxylic acid N N [2-(lf-imidazol-2-ylcarbaloyl) N 1--phenyl-ethyl]-amide 21 2-Isoquinolin- 3 -yl-IH 1 N benzoimidazole-5-carboxylic acid 7 oN [2-(4-chloro-pheny1)-lS-(1H H N /imidazol-2-ylcarbamoyl)-ethYll N amide 22 CH, 2-Isoquinolin- 3 -yl-lIH benzoimidazoe-5-carboxylic acid H [I S-(1H-imidazol-2-ylcarbamoyl) N \ / 2-p-tolyl-ethyl]-anide N. H 35 WO 2006/099379 PCT/US2006/009049 23-7 s 2-Isoquinolin-3-yl-1H o N benzoimidazole-5-carboxylic acid -N a H [2-phenyl-1 S-(thiazol-2 N yl carhanoyl)-ethyl]-amide N N CH 24 S 2-Isoquinolifn- 3 -yl-lH o N benzoimidazole-5-carboxylic acid _ [2-phenyl- l S-(pyridin-2 N \ / ylcarbamoyl)-ethyl]-amide -N H 25 F F \ FH 2-soquinolif- 3 -yl-lH N benzoimidazole-5-carboxylic acid N O [IS-(1H-imidazol-2-ylcarbamoyl) H N 2-(3,4,5-trifluoro-phenlyll)-ethYl] [ N amide 26 2-Isoquinolin- 3 -yl-IH H' benzoinidazole-5-carboxylic acid 0 0 HN. / N [2-(1H-iidazol-2-y1carbamoyl) SNH 1-(3-methoxy-phenyl)-ethYl] amide 27 Frig i7FF 2-Isoquinolin- 3 -yl-lH N H benzoimidazole-5-carboxylic acid N [1 S-(1 H-imidazol-2-ylcarbamoyl) N 2-(3-trifluoromethyl-phenyl) H. ethyl]-amide 36 WO 2006/099379 PCT/US2006/009049 28 CH H, 2-Isoquinolin-3-yl-IlH o CH benzoimidazole-5-carboxylic acid ~~ N N N NN/ 1S-(IH-imidazol-2-ylcarbamoy1) H b 3,3-dimethyl-butyl]-amide 29 9 H 2-Isoquinolin-3-yl-1HI H N aci N benzoimidazole-5-carboxylic acid O N [1S-(1H-benzoinidazol-2 N ylcarbanoyl)-2-phenyl-ethy1] N aide 30 F 2-Isoquinolin-3-yl-1H H benzoinidazole-5-carboxylic acid N 0 [2-(4-fluoro-pheny)-1S-(1H N \ / imidazol-2-ylcarbamoyl)-ethyl] H amide 31 2-Isoquinolin-3-yl-1H benzoimidazole-5-carboxylic acid ilyN II KF[2-biphenyl-4-yl- 1 -(1 H-imidazol -N 2-ylcarhamoyl)-ethyl]-amide 32 CFt 2-Isoquinolin-3-yl-1H O CH3 benzoimidazole-5-carboxylic acid N NO [IS-(IH-imidazol-2-ylcarbamoyl) H 3-methyl-butyl]-amide 33 F2-Isoquinolin-3-ylIH benzoinidazole-5-carboxylic acid O HN 0,DN[ S-(3-fluoro-phenyl)-2-(1H imidazol-2-ylcarbamoyl)-ethyl amide 37 WO 2006/099379 PCT/US2006/009049 34 F3CH H--C 2-Isoquinolin-3-yl-lH N o beizoimidazole-5-carboxylic acid H N [ S-(lH-imidazol-2-ylcarbamoyl) N N N 2-iethyl-butyll-amide N
H
3 5O 2-Isoquinolin-3-yl-1H 0 N benzoimidazole-5-carboxylic acid N [lS-(IH-imidazol-2-ylcarbamoyl) 2-methyl-propyl]-amide 36 ci 2-Isoquinolin-3-yl-1H benzoimidazole-5-carboxylic acid [2-(3-chloro-phenyl)-l S-(lH N / iinidazol-2-ylcarbamoyl)-ethyl Samide 37 F I CH 3 F 0 . to 03-(3,5-Difluoro-phenyl)-2S-[(2 N isoquinolin-3-yl-l1H H benzoimidazole-4-carbonyl) N ~ /amino]-propionic acid methyl ester N N N H 38 F F\ O 3-(3,5-Difluoro-phenyl)-2S-[(2 isoquinolin-3-yl-lH-. N H -benzoimidazole-4-carbonyl) N /amino]-propionic acid N N 38 WO 2006/099379 PCT/US2006/009049 39 N F - HN 2-Isoquinolin-3-yl-1H N 0 benzoimidazole-4-carboxylic acid N [2-(3,5-difluoro-phenyl)-1S-(1H N inidazol-2-ylcarbamoyl)-ethyl] amide N 40 0 N 2-Isoquinolin-3-yl-1H NH benzoimidazole-4-carboxylic acid H [1S-(1H-benzoimidazol-2 ylcarbamoyl)-2-phenyl-ethyl amide H 41 F - CR -- CH, F-, HN -o 2-Isoquinolin-3-yl-1H N benzoinidazole-4-carboxylic acid H [2-(3,5-difluoro-phenyl)-IS '-) N Nmethylcarbamoyl-ethyl]-amide (]) ..-N H 42 H 3 C CHS HC (0 2S-({2-[(2-Isoquinolin-3-yl-1H 0 N benzoimidazole-4-carbonyl) 0 amino]-3-phenyl N propionylamino}-methyl) H N piperidine-l-carboxylic acid tert N f N butyl ester 9N 39 WO 2006/099379 PCT/US2006/009049 43 N 2-Isoquinolin-3-yl-1H N benzoinidazole-4-carboxylic acid H - [2-phenyl-1 S-(thiazol-2 N /ylcarbamoyl)-ethyl)-amide N ' 'N N 44 CH 0 N OH N CH 3 2-Isoquinolin-3-yl-lH 0 N benzoimidazole-4-carboxylic acid H (I S-isobutylcarbamoyl-2-phenyl ethyl)-amide CN 45 / 3-({2S-[(2-Isoquinolin-3-yl-1H H CH N N \4-CH benzoimidazole-4-carbonyl) q_ \-O CH, NH amino]-3-phenyl propionylamino}-methyl) N piperidine 1-carboxylic acid tert H X N butyl ester 46 / N S 2-Isoquinolin-3-yl-IH N H
-
benzoimidazole-4-carboxylic acid H [1S-(benzothiazol-2-ylcarbamoyl) N \2-phcnyl-ethyl]-amide H 47 N NH 2-(Isoquinolin-3-ylanino)-3H HN benzoimidazole-4-carboxylic acid N (lH-imidizol-2-yl)-amide 4N N 40 WO 2006/099379 PCT/US2006/009049 48 0 HN 2-(Isoquinolin-3-ylamino)-3H N m N benzoimidazole-5-carboxylic acid N N (IH-imidazol-2-yl)-amide 49 F 2-(Isoquinolin-3-ylamnino)-3H a benzoimidazole-4-carhoxylic acid N N '(4-fluoro-phenyl)-amide -N N 50 2-(Isoquinolin-3-ylamino)- 3
H
HN 0 benzoinidazole-4-CaThoXylic acid phenylamide r xN N 51 0 HN 2-(Pyridin-2-ylamno)- 3
H
N N N N N benzoimidazole-5-carboxylic acid N' N (1H-imidazol-2-yl)-amide 52 CI 2-(Isoquinolin-3-ylamino)-3H HN O benzoimidazole-4-CaboXyliC acid (4-chloro-phenyl)-amide 53 -CH 2-(Isoquinolin-3-ylamino)-3H HN 0 benzoinidazole-4-arboXyliC acid (4-methoxy-phenyl)-amide 54 2-(Isoquinolin-3-ylamino)- 3
H
HN 0 benzoimidazole-4-carboxylic acid benzylamide 41 WO 2006/099379 PCT/US2006/009049 55H O HIN 2-(Pyridin-2-ylamino)-3H N N N benzoimidazole-5-carboxylic acid N Nl (1H-benzoimidazol-2-yl)-amide 56 HO 2-(Isoquinolin-3-ylamino)-3H HN 0 benzoimidazole-4-carboxylic acid (3-hydroxy--propyl)-amide 57 OH HN o 2-(Isoquinolin-3-ylamino)-3H iI H NN benzoimidazole-4-carboxylic acid methylamide 58 0 NH Isoquinoline-3-carboxylic acid [7 N N N (IH-imidazol-2-ylcarbamoyl)-1H N benzoimidazol-2-yl]-amide 59 H N2-(4-Chloro-benzoylamino)-3H SHN \ benzoimidazole-4-carboxylic acid N N (1 H-imidazol-2-yl)-amide 60 NIsoquinoline-3-carboxylic acid [7 N ft (1H-benzoimidazol-2 N NHI ylcarbamoyl)-1H-benzoimidazol 2-ylJ-amide 61 0 NH Isoquinoline-3-carboxylic acid [7 N N 1 NH N (benzothiazo-2-ylcarbanoyl)-1H \/benzoinidazol-2-yl]-anide 0 42 WO 2006/099379 PCT/US2006/009049 62 0 NH Isoquinoline-3-carboxylic acid [7 CN NH N ([1,3,4]thiadiazol-2-ylcarbamoyl) S1H-benzoimidazol-2-yl] -amide 63 J o Jsoquinoline-3-carboxylic acid [4 N N (pyridin-2-ylcarbanoyl)-IH N N benzoimidazol-2-ylJ-amide H 0 64 H Isoquinoline-3-carboxylic acid [4 N N (pyridin-3-ylcarbamoyl)-1H \ > benzoimidazol-2-yl]-amide 65 65 2-[(IH-Indole-2-carbonyl)-amiol 0 NH l 1H-benzoimidazole-4-carboxylic acid (1H-benzoimidazol-2-yl) amide 66 Isoquinoline-3-carboxylic acid [5 -- 1 (1H-benzoimidazol-2 0 N \/Nb 'ylcarbamoyl)-1H-benzoimidazol x N 2-yl)-amide 67 H N 0 N,/ N ONIsoquinoline-3-carboxylic acid [4 N N N H HN N (1H-indazol-5-ylcarbamoyl)-1H \>N H -benzoimidazol-2-yl]-amide 40 43 WO 2006/099379 PCT/US2006/009049 68 N6 N Isoquinoline-3-carboxylic acid [4 N N_ (quinolin-3-ylcarbamoyl)-tlH N o \ benzoimidazol-2-yl]-amide 69 0 - S N'Isoquinoline-3-carboxylic acid [5 o N (benzothiazol-2-ylcarbamoyl)-IH N N benzoimidazol-2-yl]-ainide 70 0 H N s Isoquinoi-ne-3-carboxylic acid [5 O N (thiazol-2-ylcarbamoyl)-lH N 4" N benzoinidazol-2-y]-amide 71 0 NH 2-Benzoylamino-3H N NH benzoimidazole-4-carboxylic acid N (lI--imidazol-2-yl)-anide 0 72 / HIsoquinoline-3-carboxylic acid {4 N >-tN [(furan-2-ylmethyl)-carbamoylj N H /\ -1H-benzoimidazol-2-yl}-anide 7 Isoquinolinc-3-carboxylic acid {4 (2-phenoxy-phenylcarbamoyl)-1H benzoimidazol-2-y]-amide 44 WO 2006/099379 PCT/US2006/009049 74 Isoquinoline-3-carboxylic acid [4 SN (3-phenoxy-phenylcarbamoyl)-IH benzoimidazol-2-yl]-amide 75 Isoquinoline-3-carboxylic acid [4 N N N (4-phenoxy-phenylcarbamoyl)-1H \ /benzoimidazol-2-yl]-amide 76 H 77 N 0 Isoquinoline-3-carboxylic acid [4
CH
3 N H (I S-phenyl-ethylcarbamoyl)-1IH N benzoimidazol-2-yI]-arnide o 77 N 0 Isoquinoline-3-carboxylic acid [4 IH N N (I1S-phenyl-ethylcarbamoyl)-1UH N -bezoimidazol-2-yl-amide H 78 2-[(Benzo[bthiophene-2 carbonyl)-amino]-3 I-I aio]be -boimidazole-4-carboxylic / \ (IfH-imidazol-2-yl)-arnide NH 2-BlNaphthalene-2-carbonyl)y NH 0 amino] -1 H-benzoimidazole-4 N carboxylic acid (IH-imidazol-2 / yl)-amide H 45 WO 2006/099379 PCT/US2006/009049 80 N NH 2-[(Biphenyl-4-carbonyl)-amino] 1 H-benzoimidazole-4-carboxylic acid (IH-imidazol-2-yl)-amide 81 Nl:' NHH NH 2-[(IH-Indole-2-carbonyl)-amino] 1H-benzoinidazole-4-carboxylic N N acid (1H-inidazol-2-yl)-amide N N' N H H 0 H 82 N o N 2-(3,5-Difluoro-benzoylamino) 0 H \ / 3H-benzoimidazole-4-carboxylic F acid (IH-irnidazol-2-yl)-amide 83 0 N 2-(4-Fluoro-benzoylamino)-3H o HE benzoiniidazole-4-carboxylic acid F %N (1H-inidazol-2-yl)-amide Fe 84 H N 0 Isoquinoline-3-carboxylic acid {4 ] N (2-thiophen-2-yl-ethylcarbamoyl) 1Hl-benzoimidazol-2-yl]-amide 85 NH soquinoline-3-carboxylic acid (7 N / NH phenylcarbamoyl-Iff benzoimidazol-2-yl)-amide 46 WO 2006/099379 PCT/US20061009049 86 /c isoquinoline-3-carboxylic acid (4 N [(5-methyl-furan-2-ylmethyl) N- carbamoyl]-1H-benzoimidazol-2 0/, % yl}-amide 87 ' NHI /Isoquinoline-3-carboxylic acid (7 N NH benzylcarbanoyl-IH benzoimidazol-2-yl)-amide 88 Isoquinoline-3-carboxylic acid {4 N [(thiophen-2-ylmethyl) Carbamoylj-IH-benzoimidazol-2 0 ~yl}-amide 89 0 NH Isoquinoline-3-carboxylic acid (7 N H phenethylcarbamoyl-1H4 N (O benzoimidazol-2-yl)-arnide 900 90 0 sNH oquinolinc-3-carboxylic acid (7 N / NH CH ethylcarbamoyl-lH benzoinidazol-2-yI)-amide 91 0 NzIo NH quinoline-3-carboxylic acid (7 N/ NH CH 3 tert-butylcarbamoyl- 1 H O CH benzoimidazol-2-yl)-amide 47 WO 2006/099379 PCT/US2006/009049 92 d H 2-[(Benzofuran-2-carbonyl) O NH amino]- 1H-benzoimidazole-4 N carboxylic acid (1H-imidazol-2 yl)-amide 93b 93 Isoquinoline-3-oarboxylic acid [4 (1,3,4,9-tetrahydro-beta-carboline N N 2-carbonyl)-1H-benzoimidazol- 2 0- yl]-amide 94 Isoquinoline-3-carboxylic acid (4 s [(benzo[b]thiophen-2-ylmethyl) N N carbanoyl] -1 H-beuzoimidazol-2 o /yl}-amide 95 \/ N Isoquinoline-3-carboxylic acid {4 s [(benzothiazol-2-ylmethyl) N carbamoyl]-IH-benzoimidazol-2 o ~ / yl}-amide 96 N Isoquinoline-3-carboxylic acid {4 [( -inethyl-1H-benzoimidazol-2 IC N , j N ylmethyl)-carbamoyl -1H &N H aenzoimidazol-2-yl}-amide 97 N 0 Isoquinoline-3-carboxylic acid (4 / \ N [2-(1H-benzoimidazol-2-yl) ethylcarbamoyl]-tH / benzoimidazol-2-yl}-amide 48 WO 2006/099379 PCT/US2006/009049 98 2-[2-(4-Fluoro-phenyl) o N acetylamino]-3H-benzoimidazole F O HN 4-carboxylic acid (1H-imidazol-2 N yl)-amide 99 2-(4-Fluoro-3-metlioxy 0 0 HN benzoylamino)-3H XN benzoimidazole-4-carboxylic acid (IH-imidazol-2-yl)-amide 100 2-(3,4-Dimethoxy-benzoylamino) o 14\ 3H1-benzoirnidazole-4-carboxylic HaC H acid (IH-imidazol-2-yl)-amide HC 101 N s Isoquinoline-3-carboxylic acid {4 N [(thiazol-2-ylnethyl)-carbamoylj I H-benzoinidazol-2-yl}-amide 102 K Isoquinoline-3-carboxylic acid {4 [(1H-benzoimidazol-2-ylmethyl) N carbamoyl]-1H-benzoimidazol-2 yl}-amide 103 2-(4-Trifluoromethyl ' o' benzoylaiino)-1H benzoimidazole-4-caTboxylic acid (IH-imnidazol-2-yl)-anide 49 WO 2006/099379 PCT/US2006/009049 104 NH 2-(4-Phcnoxy-benzoylamino)-1H -l 0benzoinidazole-4-carboxylic acid (1 H-imidazol-2-yl)-anide 105 Quinoline-3-carboxylic acid [7 o HN (1H-inidazol-2-y1carbamoy1)-1H N benzoimidazol-2-yl)-amide 106 H O N Qinoline-2-carboxylic acid [7 0 HN \(1 H-imidazol-2-ylcarbamoyl)- 1 H N benzoimidazol-2-yl]-amide 107 O N 2-[(Furan-3-carbonyl)-aminol-3H benzoinidazole-4-carboxylic acid o HN / 0 HN (IH-imidazol-2-yl)-amide 108 o N 2-(4-tert-Butyl-benzoylamino)-3H o HN \ / benzoimidazole-4-carboxylic acid HC N N N (1H-imidazol-2-yl)-atnide H,C C H
H
3 0;
CH
3 109 10N 2-(4-Nitro-benzoylamino)-3H o HN benzoimidazole-4-carboxylic acid 50(H-iinidazol-2-yl)-amide 50 WO 2006/099379 PCT/US2006/009049 110 o 4 i2-(4-Methoxy-bcnzoylamnino)-3 o HN benzoimidazole-4-carboxylic acid HaCN (IH-imidazol-2-yl)-amide 2-[(1H-Inidazole-2-carbonyl) o N anino]-3-benzoimidazole-4 0 <\ / carboxylic acid (1H-imidazol-2 yl)-anide 112 H H4D o N 2-(4-Isopropyl-benzoylanino)-3H- 0 H\ benzoimidazole-4-carboxylic acid HH e (1H-imidazol-2-yl)-amide 113 o N 2-(3-Fluoro-benzoylamino)-3Hf o HN \ / benzoimidazole-4-carboxylic acid N ( H-imidazol-2-yl)-amide F 114 o 2-(2-Fluoro-benzoylamino)-3H o H ~/benzoimidazole-4-carboxylic acid HF N (1H-imidazol-2-yl)-amide 115 O% NH 2-(5-Chloro-benzOfuran-2 H carbonyl)-amino]-1H 0 benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide Ci 51 WO 2006/099379 PCT/US2006/009049 116 H N4 D 6,7-Dimethoxy-isoquinoline-3 0 N carboxylic acid [7-(1H-imidazol-2 CH, 0 Htd\/ HN NI ylcarbanoyl)-1H-benzoimidazol 2-yi]-amide CH, 117 -4 6,7-Dimethoxy-isoquinoline-3 carboxylic acid [7-(1H-imidazol-2 ylcarbamoyl)-1H-benzoimidazol H 2-yl]-amide 118 1 o N 2-(2-Chloro-benzoylamino)-3H C H \ benzoimidazole-4-carboxylic acid N (1H-imidazol-2-yl)-amide 119 N 1H-Indazole-3-carboxylic acid [7
N
1 NH (1H-imidazol-2-ylcarbamoyl)-11-1 benzoimidazol-2-yl]-amide 120 0NH 2-[(1-Methyl-IH-indole-2 - N H N carbonyl)-amino]-311 CH3 N benzoimidazole-4-carboxylic acid \ / N (ff-imidazol-2-yl)-amide 121 0 1-Methyl-iH-indazole-3 N NH carboxylic acid {7-(1H-imidazol-2 H, NH ylcarbamoyl)-1H-benzoiiidazol 2-yl]-amide 52 WO 2006/099379 PCT/US2006/009049 122 o soquinoline-3-carboxylic acid [4 N (5-phenyl- 1H-imidazol-2 N ylcarbamoyl)-lH-benzoinidazol H 2-yl]-amide 23HC / NH2-[(5-Methyl-1HI-indole-2 NNH carbonyl)-amino]-3H N benzoimidazole-4-carboxylic acid N (1 H-inmidazol-2-yl)-amide 124 2-[(6-Phenoxy-pyridine-2 0 N carbonyl)-amino]-3H N /benzoimidazole-4-carboxylic acid \ N ~ (1H-iinidazol-2-yl)-amnide 125 0 2-[(I-Benzyl-1H-indole-2 / N NH carbonyl)-amino]-3H N / NH benzoimidazole-4-carboxylic acid S(IH-imidazol-2-yl)-amide 126 2-[(1-Propyl-1H-indole-2 N carbonyl)-aminoj-3H benzoimidazole-4-carboxylic acid \ N (lH-imidazol-2-yl)-amide 127 2-[(1,5-Dimethyl-1H-indole-2 H3C / \NH N /a NH harbonyl)-amino]-3H Ha N/NHbenzoimidazole-4-carboxylic acid (3 (1H-imidazol-2-yl)-amide 53 PCT/Us2o06100 9049 WO 2006/099379 128 N K I-I soquinoine-3-CarboxYUOc acid [4 0 NH (IH-ifldazol-2ylcalballioyl)- 6 N itro- 41Vbpflzoinfdazo2Yli "' N ;\ ~amide 129 IsoqullOlinC- 3 -carboxylic acid [6 N a lm in o 4 -K j Hfim id a z o l 2 N y~carbarnoyl)UHbenzoimidazol N N 2-y1I1-amide 130 N,. NN 1S0qaiflinfl 3 -carhoxyliC acid. [4 0 NH (Il-imidazo2y1clbam"oY I >'ri :: isabutytylamiflo 1 H~ON ZAbenzoilfidazol2-yl-amide o N ylcarbamoyD)-InHbenizoimidazob H 2-y1I1-amfide 132 isoquin o in e -3ca -b x Y lic a i 4 J7 NH (lH ilidazo1 2-ylcarbanoy 0
)
6 N0
(
3 nhethy1butYIYain o-l ' 14 N benzoimidazol2-Ymiiaidc 133 Nlsoquinol1ine3-carboxYlic acid [7 0 NN \ (ln -iI idazob 2Ycatbam nOYl) -5 HN ~phenylacetylaminolfl A benzoimnidazol-2-Yllamide 54 WO 2006/099379 PCTIU S2006/0090 4 9 W O 2 0 6 1 0 9 3 7 91 -B e n z y 1 -2 -( ( i s o q u i n o l i n e -3 NH carbonyl)-amino]i- 1 1 134 m N benzoimidazole-4-carboxylic acid methyl ester isoquinoline-3-carboxylic acid N H N[l-bcnzyl-4-(l1H-imidazol- 2 135 N 3 ylcarbamoy 1
)-
1
H
/ 0 N benzoimidazol-2-yl-amide 0, N NHIsoquinoline-3-carboxyliC acid N14 {4-(1H-imidazol-2 136 H N ylcar bamoyl)-1methyl-IH N benzoimidazol-2-y1-ainde 0 NH Isoquinoline3--carboxylic acid 7 N /t/ (4-benzylcaTbamoyl-1-methyl 137 Hr c H-benzoimidazol-2-y1)-amide 0 2-(3-Isoquinolin-3-Yl-ureido) \ 1H-benzoimidazole- 4 138 carboxylic acid (IH-imidazol 2-yl)-amide 2-[(Isoquino1in-3-yinethy1) 139 rN N H aninof-1H-benzoimidazole-4 139 HNN N carboxylic acid (1H-imidazol 2-yl)-amide 0 Isoquinoline-3-carboxylic acid N N Na{ [5-(iH-imidazol- 2 140 N ylcarbanoyl)-1H
H
0 benzoimidazol-2-y1}-amide 55 WO 20061099379 PCT/US2006/00 9 0 4 9 0 F Isoquinoline- 3 -carboxylic acid NH (7-[3-(4-fluoro-phelyl) 141 N urcido]-1H-benzoimidazol- 2 yl}-amide N Isoquinoline- 3 -carboxylic acid 142 ~ N~ / 0oH,(7- {342-(4-methloxy-phflYI) 142 etyl-ureidoA-i benzoimidazol-2-yl)-amide oH, Isoquinoline-3-CaTbOXyliC acid HN {74[3-(4-methoxy-btfl)l o-N ... 143 N : ureido]-IH-benzoinidazol-2 H A yl} -amide INH H-Benzoimnidazole-2, 4 SNH dicarboxylic acid 44(11 144 NH iridazo1-2-yl)-amide] 2 N' isoqurnolifl- 3 -ylamide ~ 0 Isoquinoline-l-carboxylic acid k NH 'Z, N(11,3'1- 145 Nm-lH [2,4']bibenzoimidazOlyl-2yl) anide 0 lsoquinoline-l-Carboxylic acid N. NH 146 - N H [7-(3H-iidazo[4,5-clpyridin N 2-yl)-1H-benZoimidaZOl-2-yl amide 56 WO 2006/099379 PCT/US2006/0 0 9 0 49 Isoquinoline-l-carboxylic acid NH [7-(4-pheny1-1H-imidazol- 2 147 NH y1)-1H-benzoimidazol-2-yl] \ 'amide A NH Isoquinoline-3-carboxylic acid 148 N HN N [4-(1H-imidazol-2-y1)-lH 18benzoimidazol-2-y1]-amide osoquinoline-3-carboxylic acid NH {4-[4-(4-chlorO-Phenyl)-1H 149 N N N imidazol-2-yl1-1H benzoimidazol-2-Y}-anide NH Isoquinaline-3-carboxYlic acid 150 N HN NN (4-imidazol-1-y1-IH 150 N benzoiidazol-2-yl)-amide 0 NH CH, Isoquinoline-3-carboxYlic acid 151N RN H N H C H [4-(4-tert-butyl-oXaZo1-2-Yl N 1H-benzoimidazo1-2-yl1-anide HN OH 1-{4-[N'-(3-HydroxY-proPY1) 0 NH H guanidinocarbonyl]-1H 152 N A benzoimidazol-2-Yl}-3 isoquinolin-3uxea 57 WO 2006/099379 PCT/US2006/009049 HN CH I-Isoquinolin-3-y1-3-[4-(N' 153 05( NH propyl-guanidinocarbonyl)-1H benzoimidazol-2-yl]-urea OH H1-{4-[N'-(4-Hydroxy-butyl) guanidinocarbony]-1H 154 NH 1- bnoirniclazol-2-yl} -3 isoquinolhn-3-yl-urea 0 SC Isoquinoline-3-carboxylic acid N [4-(4-methanesulfbnyl 155 0 NHbenzylcarbamoyl)-H N. z benzoimidazo-2-y1]-amide Isoquinoline-3-carboxylic acid N NH [4-(4,5,6,7-tetrahydro-1H 156 HN / benzoimidazol-2-ylcarbamoyl) '0 1H-benzoimidazol-2-yl]-amide O H2-({2-{(Isoquinoline-3 Nc'G carbonyl)-amino]-1H 157 benzoimidazole-4-carbonyl} anino)-3H-imidazol-4-yl] acetic acid methyl ester 0 [2-({2-[(Isoquinoline-3 N OH carbonyl)-aminol-1H 158 .
benzoimidazole-4-carbonyl} S N amino)-3H-imidazol- 4 -yl] acetic acid O CH3 Isoquinoline-3-carboxylic acid 159 N N 0 -CH, (4- {5-(methoxy-methyl carbamoyl)-methyl]-lH 58 PCT/US2006/0 0 9 0 4 9 WO 2006/099379 irnidazo-2-1caramoy1}-1H benzoimidazol-2-Yl)-anide lsoquinoline-3-carboxyic acid X NHN [4-(5-isopropYl- H-imidazol- 2 160 HN N CH, yloarhamoy1)-1H H, benzoimidazol-2-yl-anide F lsoquinolilne-3-carboxyliG acid 0 H j4-(4-fluor-phlenYlcarbamoyl) 161 N N1-benzoimidazol-2-Y1-amide 13 P, Isoquinoline-3-carboxylic acid "4o 4-nethaleslfbfYl H3 c' ([1-(4 162 phenyl)-ethylca-rbamoyl)-1H ) Nbenzoimidazol-2-yl}-amide A ~ Isoquinoline-3-carboxylic acid I o [4-(3-methanesulfOnyl 163 0 NH phenylcarbaml)-1 benzoinidazol-2-yl)-amide OF F lsequlinlline-3-carboxylic acid NH F [7-(4-trifluorOmethoXY N N ),NH / \-1 164 A AN INNH - benzylcarbamoy1-H N benzoimidazol-2-y11-amide o o-cHS Isoquinoline-3-carboxylic acid l NH / \ 7-(4-methoxY 165 N NH belnzylcarbamOyl)-1
H
benzoinidazo1-2-Y1}-amide 59 WO 2006/099379 PCT/US2006/009049 CH, Isoquinoline-3-carboxylio acid NZ NH [7-(4-niethyl 166 N//NHi bcnzylcarbamoyl)-1H benzoimidazol-2-yi]-amide FNH Isoquinoline-3-carhoxylic acid 167 N NH [7-(4-fluoro-benzylcarbamoyl) IH-benzoimidazol-2-yI]-anide 0 o ,cL NH Isoquinoline-3-carboxylic acid 168 N / NH [7-(4-chloo-benzylcarbamoyl) IH-benzoiimidazol-2-yl)-amide F o F JSoquinoline-3-carboxylic acid 169 NH 7-(3-trifluoromethyl 169 x -N N"NH enzylcarbamoyl)-IH \ / benzoimidazol-2-yl]-amide F 0 F Isoquinoline-3-carboxylic acid 170 NH/N\F [7.(3-fluoo-4.trifluoromethyl N)" H benzylcarbamoyl)-1H benzoimidazol-2-yl-amide Isoquinoline-3-carboxylic acid 17-N N[7-(3,5-difluoro 171 Nbenzylcarbamoyl)-1H \ / ~benzoimidazol-2-yl]-amide 60 WO 2006/099379 PCT/US2006/009049 N - NH Isoquinoline-3-carboxylic acid 172 / z N NH 7-(4-cyano-benzylcarbamoyl) 1H-benzoimidazol-2-yl]-amide o Isoquinoline-3-carboxylic acid HN/ {7-[N'-(4,5-dihydro-1H 173 N NH H imidazol-2-yl) N-f hydrazinocarbonyl]-IH benzoimidazol-2-yl}-amide H HC- 0 Isoquinoline-3-carboxylic acid N [7-(3-methoxy 174N/ HH NH benzylcarbamoyl)-1 H / \ benzoinidazol-2-yl]-amide O N N Isoquinoline-3-carboxylic acid N CH[7-(4-methoxy 175 N// NH /H 175N ( O phenylcarbamoyl)-1H benzoimidazol-2-yl]-anide 0 Isoquinoline-3-carboxylic acid N N NH {7-[2-(4-methoxy-phenyl) K- 0 176 N76 ethylcarbamoyll-1H ./ benzoiindazol-2-yl}-amide F NH Isoquinoline-3-carboxylic acid 177 7 AN NH [ 17 N7-(3-fluoro-benzylcarbamoyl) 1H-benzoimidazol-2-yl]-amide 0 61 WO 2006/099379 PCT/US2006/009049 NH b s Isoquinoline-3-carboxylic acid 178 N NH [7-(4-sulfamoyl N NH benzylcarbamoyl)-1H benzoimidazol-2-yl-amide Isoquinoline-3-carboxylic acid N N [7-(2-methoxy 179 N NH O benzylcarbamoyl)-1l HsC benzoimidazol-2-yl]-amide Isoquinoline- 3 -carboxylic acid N N[7-(2-ethoxy 180 N /NH benzylcarbamoyl)-tH CHK benzoimidazol-2-yl]-amide lsoquinoline-3-carboxylic acid ' NH 0 [7-(2,2-dioxo-1,3 181 N ' NH dihydrobenzo[c]thiophen-5 ylcarbamoyl)-1H benzoimidazol-2-yl]-amide 0 Isoquinoline-3-carboxylic acid 182 ZN NN {4-[2-(4-phenoxy-phenyl) 182 Cethylcarbamoyl]-ilH benzoimidazol-2-yl}-amide H C H 2 0 N Isoquinoline-3-carboxylic acid 183 N (4-butylcarbamoyl-1 H N benzoinidazol-2-yl)-amide N 0 0 Isoquinoline-3-carboxylic acid 4NH N H[4-(3H-imidazo[4,5-c]pyridin NN 2-ylcarbamoyl)-1H H beizoimidazol-2-yl)-amide 62 WO 2006/1099379 PCT/US2006/009049 N 0O N- NIsoquinoline-3-carboxylic acid NH N 185 N N N [4-(7H-purin-8-ylcarbamoyl) H - IH-benzoimidazol-2-yl]-amide H N 0 NIsoquinoline-3-carboxylic acid 186 N N [4-(isoquinolin-6-ylcarbamoyl) IH-benzo.imidazol-2-yl]-amide H S Isoquinoline-3-carboxylic acid 0 N 0 [4-(2-methylsulfanyl N ethylcarbamoyl)-1H H N A benzoimidazol-2-yll-amide C Isoquinoline-3-carboxylic acid NH O [4-(5,5-dinethyl-7-oxo-4,5,6,7 .188 N HN N s tetrahydro-benzothiazol-2 CH, N CH, ylcarbamoyl)-lH \ >benzoimidazol-2-yl]-amide Isoquinoline-3-carboxylic acid [4-(1H-benzoimidazol-5 ylcarbamoyl)-IH -- -benzoimidazol-2-yl]-amide 0 '- z NH Isoquinoline-3-carboxylic acid 190 ' N N-t [4-(IH-pyrazol-3-ylcarbamoyl) 1 H-benzoimidazol-2-yl]-anide 63 WO 20061099379 PCT/US2006/0 0 9 0 49 N N0 3H 2 Isoqu in olin e-3 -C arb ox ylic acid {4-(2-amino-pyrimidin-4 191 0 NH ylcarbamoyl)- 1 NN -- Nbenzoimidazo1-2-y1}-amide Isoquinoline-3-carboxylic acid NH 'N NHG CH 3 [4-(4-tert-butyl-1 H-imidazol-2 192 HN N N- CH, ylcarbamoyl)-lH \ /benzoimidazol-2-yl-amide 0
-
Isoquinolifne-3-carboxylic acid A N NH H, {4-(4,5-ditnethyl-1H-imidazol 193 HN N 2-ylcarbamoyl)-1H HN Obenzoimidazol-2-yl]-anide Isoquinoline- 3 -carboxylic acid 19N N [4-(5-ethyl-1H-imidazol-2 194 N N N ylcarbanoyl)-l 1 0 / CH, benzoimidazol-2-yl]-anide 0 'Isoquinoline-3-carboxylic acid A AN [4-(5-adamantan-1-yl-1H 195 imidazol-2-ylcarbamoyl)-1H benzoinidazo1-2-yl]-amide isoquinoline-3-carboxylic acid NH [4-(5-benzyl-1H-imidazol- 2 196 N NN N ylcarbamoyl)-1H benzoimidazol-2-yl]-amide 0 64 WO 2006/099379 PCT/US2006/009049 O H3C CH 2-({2-[(Isoquinoline-3 NH" VQ CH carbonyl)-anino]-31H 197 N NH benzoimidazole-4-carbonyl) N amino)-6,7-dihydro-4H thiazolo[5,4-c]pyridine-5 carboxylic acid tert-butyl ester o Isoquinoline-3-carboxylic acid NH H [7-(4,5,6,7-tetrahydro 198 N; NH thiazoto[5,4-c]pyridin-2 N ylcarbamoyl)-IH \ 0 / ~benzoimidazol-2-yl]-amide O Isoquinoline-3-carboxylic acid N H 1
CH
3 [7-(5-ethyl-4,5,6,7-tetrahydro 199 N N NH thiazolo[5,4-c]pyridin-2 N ylcarbamoyl)-lfH benzoimidazol-2-yl]-amide 0 CH3 Isoquinoline-3-carboxylic acid ZZ "'- NH ,so {7-[5-(propane-1-sulfonyl) 200 NH 4,5,6,7-tetrahydro-thiazolo[5,4 N c]pyridin-2-ylcarbamoyl]-1H benzoimidazol-2-yl} -arnide o Isoquinoline-3-carboxylic acid N N 3 4-(5-tert-butylcarbamoyl 201 N HNNN N 4,5,6,7-tetrahydro-thiazolo[5,4 c\ N cjpyridin-2-ylcarbamoyl)-lH benzoinidazol-2-yl]-amide o Isoquinoline-3-carboxylic acid '_ N H 1H [4-(5-benzoyl-4,5,6,7 x N ), 202 H N tetrahydro-thiazolo[5,4 N cpyridin-2-ylcarbamoyl)-lH benzoimidazol-2-yl]-amide 65 WO 2006/099379 PCT/US2006/009049 o Isoquinoline- 3 -carboxylic acid NH
,CH
3 {4-[5-(3-methy1sulfanYl HN N H N propy1)-4,5,6,7-tetrahydro HNN 20 NN thijazolo(5,4-c]pyridin-2 ylcarbanoyl]-1H benzoimidazol-2-yl}-amide Isoquinoline-3-carboxyliO acid NH ri-CH, {4-[5-(3-methaneulfOInYl HN N s propyl)-4,5,6,7-tetrahydro 204 thiazolo[5,4-C}pyridin-2 ylcarbanioyl]-1H benzoimidazol-2-yl}-amide 4-({2-[(Jsoquinoline-3 carbonyl)-amino]-lH 205 p benzoimidazole-4-carbonYl} o anino)-piperidi-ne-1-carboxylic H N-1 acid tert-butyl ester H N Isoquinoline-3-carboxYlic acid 206 o NH [4-(piperidin-4-ylcarbamoy1) N 1H-benzoinidazol-2-y1-amide NA H esH 4-({2-{(Isoquinoline-3 Y carbonyl)-amino-i1 207 benzoimidazole-4-carbonyl} NH arnino)-piperidine- 1 -carboxylic acid isopropyl ester o Isoquinoline-3--carboxyliC acid NH OC7 {7-(5-methanesulfonyl-4,5,6,7 208 N NH W O tetrahydro-thiazolo[5, 4 N c]pyridin-2-ylcarbamfloyl)-H benzoimidazol-2-y1]-amide 66 WO 2006/099379 PCT/US2006/0090 49 Isoquinoline-3-carboxylic acid NH [7-(5-benzyl-4,5,6,7-tetrahydro 209 N thiazolo[5,4-c]pyridin-2 N ylcarbamoyl)-IH (o / benzoimidazol-2-yll-amnide N 0 Isoquinoline-3-carboxylic acid s H [4-(4,5,6,7-tetrahydro 210 -N N benzothiazol-2-ycabamoyl) >N H o 1H-benzoimidazo1-2-y1)-amide H Isoquinoline-3-carboxylic acid [4-(6-methyl-4,5,6,7 S N 211 \N N tetrahydro-benzothiazol- 2 N HC 0 ylcaTbamoyl)-1H benzoimidazol-2-yi}-amide 0 Isoquinoline-3-carboxylic acid NH [4-(5-methyl-4,5,6,7 212 HN N N tetrahydro-ftliazolo[5,4 cpyridin-2-ylcarbamoy1)-IH benzoinidazol-2-yl]-amide 0 Isoquinoline-3-carboxylic acid NH [4-(5-cyclopentylmethyl 213 N N H 4,5,6,7-tetrahydro-thiazolo[5,4 N -~ o N cjpyridin-2-ylcarbanoyl)- 1H enzoimidazol-2-yl}-amide o Isoquinoline-3-carboxylic acid NH [4-(5-pyridin-3-ylmethyl A N 7. s N 214 HN -N 4,5,6,7-tetrahydro-thiazolo[5,4 O N c]pyridin-2-ylcarbamoyl)-1H benzoimidazol-2-yl]-amide 67 WO 2006/099379 PCT/US2006/009049 o Isoquinoline-3-carboxylic acid NN [4-(5-cyclohexylmethyl 215 x N N N S 4,5,6,7-tetrahydro-thiazolo[5,4 N c]pyridin-2-ylcarbamoyl)-1H benzoimidazol-2-yl]-amide 0 Isoquinoline-3-carboxylic acid NH [4-(5-furan-3-ylnetlhyl-4,5,6,7 216 N HN N H N tctahydro-thiazolo[5,4 N \N cjpyridin-2-ylcarbamoyl)-lH b / o benzoimidazol-2-yl]-amide 3-[({2-[(Isoquinoline-3 HC0 0 N O carbonyl)-amino]--1H 217 benzoimidazole-4-carbonyl} 0 amino)-methyl)-piperidine-1 carboxylic acid tert-butyl ester CH c 4-[({2-[(Isoquinoline-3 H3 N carbonyl)-amino]-1H 218 N benzoimidazole-4-carbonyl) amino)-nethyl]-piperidine-1 carboxylic acid tert-butyl ester 1H lsoquinoline-3-carboxylic acid {4-[(piperidin-3-ylmethyl) 219 N N carbamoyl]- IH-benzoimidazol 6N H \ /2-yl}-amide H N NIsoquinoline-3-carboxylic acid N 0 Jounln--ahxlcai {4-[ (piperidin-4-ylmethyl) N N carbamoyl)-IH-benzoimidazol
N
H 0 /2-yl}-amide 68 WO 2006/099379 PCT/US2006/009049 Isoquinoline-3-carboxylic acid CH {4-{5-(1-methyl-1H-pyrrol-2 21HN N s N / ylmethyl)-4,5,6,7-tetrahydro 22, N thiazolo[5,4-c]pyridin-2 ylcarbanoy1}-IH benzoimidazol-2-yl}-amide o Isoquinoline-3-carboxylic acid NH CH {4-(5-propyl-4,5,6,7-tetrahydro 222 C C N HH S N thiazolo[5,4-c]pyridin-2 N ylcarbanoyl)-1H benzoimidazol-2-yl]-amide Isoquinoline-3-carboxylic acid NH CH {4-[5-(2-ethyl-butyl)-4,5,6,7 223 N HNNN N w tetrahydro-thiazolo{5,4 -/ o N Nc]pyridin-2-ylcarbamoyl]-1H benzoinidazol-2-yl}-amide o Isoquinoline-3-carboxylic acid NH CH 3 [4-(5-isobutyl-4,5,6,7 N H X OH 3 tetrahydro-thiazolo[5,4 N c]pyridin-2-ylcarbamoyl)-1H benzoinidazol-2-yl]-amide Isoquinoline-3-carboxylic acid N N 0 {4-[(1-cyclohexylmethyl 225 N H piperidin-3-ylmethyl) N-N N N carbamoyl]-1H-benzoimidazol 2-yl}-anide N Isoquinoline-3-carboxylic acid N 0 {4-[ (1-cyclohexylmethyl 226 N H piperidin-4-ylmethyl) N-N -N carhanoyl]- 1H-benzoimidazol 0 / 2-yl}-amide 69 WO 2006/099379 PCT/US2006/009049 CH o 3-[2-({2-[(Isoquinoline- 3 O 0 o carbonyl) -aniino)-IlH 227 N benzoinidazoe--4-carbonyl} / amino)-ethyl]-piperidine-1 carboxylic acid tert-butyl ester o 4-[2-({2-[(Isoquinoline-3 H0 cO N N carbonyl) -amino]-lH HOY~ Y N 228 W c , o benzoimidazole-4-carbolYfl amino)-ethyl]-piperidine-1 carboxylic acid tert-butyl ester N 0 isoquinoline-3-carboxylic acid N H [4-(2-piperidin-3-yl 229 N N ethylcarbanoyl)-lH benzoimidazol-2-yi]-amide H N o Isoquinoline-3-carboxylic acid H N N H {4-(2-piperidin- 4 -yl 230 N N ethylcarbamoyl)-lH H benzoimidazol- 2 -yl]-amide o lsoquinoline-3-carboxylic acid NH {4-[5-(2,4-dimethoxy-benzyl) 231 . NHN N Oc N4,5,6,7-tetrahydro-thiazolo[5, 4 231 CH, xN .c]pyridin-2-ylcarbamoyl]-IH benzoimidazol-2-yl} -amide o Isoquinoline-3-carboxylic acid NH {4-{5-(4-chloro-benzyl) 232 HN cl 4,5,6,7-tetrahydro-thiazoo( 5
,
4 d o / c]pyridiu-2-ylcarbamoyl]-1 benzoimidazol-2-yl}-amide 70 WO 2006/099379 PCT/US2006/009049 - Isoquinoline-3-carboxylic acid - NH {4-[5-(4-fluoro-benzyl)-4,5,6,7 N/ N -FN 233 HN JF tetrahydro-thiazolo{5,4 N c]pyridin-2-ylcarbamoylj-1H benzoimidazol-2-yl}.-amide o Isoquinoline-3-carboxylic acid N AN MH {4-[5-(4-isopropoxy-benzyl) H-N N 234 HNN N H / 0 4,5,6,7-tetrahydro-thiazolo[5, 4 c]pyridin-2-ylcarbamoyl]-IH benzoimidazol-2-yl}-amide o o-Isoquinoline-3-carboxylic acid NH (4-[5-(2-methoxy-benzyl) 235 HN/N N / 4,5,6,7-tetrahydro-thiazolo[5,4 b o N clpyridin-2-ylcarbamoyj- 1H1 benzoimidazol-2-y}-amide Isoquinoline-3-carboxylic acid NH
O-CH
3 {4-[5-(3-methoxy-benzyl) 236 N 4,5,6,7-tetrahydro-thiazolo[5,4 c]pyridin-2-ylcarbamoyl]-1H benzoimidazol-2-yl}-anide o Isoquinoline-3-carboxylic acid N NH {4-[5-(4-rmethoxy-benzyl) N A N /a 237 4,5,6,7-tetrahydro-thiazolo[5,4 S/ o c]pyridin-2-ylcarbamoyl]-1H benzoimidazol-2-yl} -amide Isoquinoline-3-carboxylic acid [4-(5-phenethyl-4,5,6,7 - N NH 238 N tetrahydro-thiazolo[5,4 H N c]pyridin-2-ylcarbamoyl)-1H S/ 0 benzoimidazol-2-yl-amide 71 WO 2006/099379 PCT/US2006/009049 0 Isoquinoline-3-carboxylic acid NH s [4-(5-thiophen-2-ylmethyl 239 NHN 4,5,6,7-tetrahydro-thiazolo[5,4 N c]pyridin-2-ylcarbamoyl)-lH benzoimidazol-2-y-amide Isoquinoline-3-carboxylic acid [4-(5-thiophen-3-ylmethyl 240 HN t N 4,5,6,7-tetrahydro-thiazolo[5,4 N c]pyridin-2-ylcarbamoyl)-1H benzoimidazol-2-yl]-amide Q CHIsoquinoline-3-carboxylic acid S N C{4-5-(3-methyl-butyl)-4,5,6,7 241 HNN I tetrahydro-thiazolo[5,4 N c]pyridin-2-ylcarbamoyl)-1H benzoimidazol-2-yl}-amide 3R-[( {2-[(Isoquinoline-3 C N N O carbonyl)-amino}-1IJ 242 C
H
, N N H benzoimidazole-4-carbonyl} IAN N amino)-methyl]-piperidine-1 carboxylic acid tert-butyl ester 3S-[({2-[(Isoquinoline-3 H~O 0 N N O carbonyl)-amino)-1IH 243 CH,30 N N H 243 03)-NN N benZoimridazole-4-carbonyl} N arnino)-methyl]-piperidine-1 carboxylic acid tert-butyl ester N 0Isoquinoline-3-carboxylic acid 244 N H (4-[ (piperidin-3S-ylmethyl) N- N carbamoyll-1H-benzoimidazol N \ /2-yl}-amide 72 WO 2006/099379 PCT/US2006/009049 HN N 0 Isoquinoline-3-carboxylic acid {4-[ (piperidin-3R-ylmethyl) 245 NH 25NN N carbamoyl}-1H-benzoimidazol H 0 \ /2-yl}-anide Isoquinoline-3-carboxylic acid NH {4-[5-(3H-imidazol-4 246 HN\N ylmethyl)-4,5,6,7-tetrahydro N thiazolo[5,4-c]pyridin-2 ylcarbainoyll-1H benzoimidazol-2-y}-amide isoquinoline-3-carboxylic acid
OH
3 NNH H {4-[5-(1-methyl-1H-imidazol AN HN N 2-ylmethyl)-4,5,6,7-tetrahydro 247 \ N thiazolo[5,4-c]pyridin-2 ylcarbamoyll-1H benzoimidazol-2-yl}-amide 0 Isoquinoline-3-carboxylic acid NH {4-[5-(IH-imidazol-2 NHN N \ ylmethyl)-4,5,6,7-tetrahydro 248 N thiazolo[5,4-clpyridin-2 ylcarbamoyl]-IH benzoimidazol-2-yl}-amide o Isoquinoline-3-carboxylic acid NH [4-(5-cyclopropylmethyl 249 HN NS 4,5,6,7-tetrahydro-thiazolo[5,4 N c]pyridin-2-ylcarbamoyl)-1H benzoinidazol-2-yl-anide Isoquinoline-3-carboxylic acid HH {4-[5-(3,3-dimethyl-butyl) 250 HN 4 C a 4,5,6,7-tetrahydro-thiazolo{5,4 N c]pyridin-2-ylcarbamoyl]-1H benzoimidazol-2-yl}-amide 73 WO 2006/099379 PCT/US2006/009049 6-({2-[(Isoquinoline-3 HaC 0 N carbonyl)-amino]-lH 25 3 CR 3 0 -- "' benzoimidazole-4-carbonyl} 251 amino)-3,4-diydro-1f isoquinoline-2-carboxylic acid tert-butyl ester H N 0 Jsoquinoline-3-carboxylic acid HN N [4(1,2,3,4-tetrahydro 252 \H isoquinoin-6-ylcarbamoyl)-IH H O benzoimidazol-2-yll-amide Isoquinoline-3-carboxylic acid N -. O {4-[(1-cyclohexylmethyl 253 N piperidin-3S-ylmethyl) N carbamoyl]-1H-benzoimidazol 2-yl}-amide Isoquinolinc-3-carboxylic acid N o {4-[(1-cyclohexylmethyl 254 H N piperidin-3R-ylmethyl) 25 \>N N H carbamoyl]-1H-benzoimidazol 2-yl}-ainide 0 Isoquinoline-3-carboxylic acid N H S [4-(5-thiazol-2-ylmethyl A yN S N 255 HN N 4,5,6,7-tetrahydro-thiazolo[5, 4 6 o N c]pyridin-2-ylcarbamoyl)-1
H
benzoimidazol-2-yl}-amide Isoquinoline-3-carboxyic acid NH N {4-[5-(5-niethyl-3H-imidazol HNNN S 4-ylmethyl)-4,5,6,7-tetrahydro 256 N HC thiazolo[5,4-clpyridin-2 ylcarbamoyl]-1H benzoimidazol-2-yl}-amide 74 WO 2006/099379 PCT/US2006/009049 o Isoquinoline-3-carboxylic acid ( N {4-[5-(2-ethyl-5-methyl-3H A NHN s N \ Cci HNI N imidazol-4-ylmethyl)-4,5,6,7 257
H
3 G 257 tetrahydro-thiazolo[5,4 c]pyridin-2-ylcarbamoyl]-1H benzoimidazol-2-y}-amide 0 O O H 3 2S-[2-({2-[(Isoquinoline-3 NH CH3 carbonyl)-amino}-1H x .NN N Nbenzoimidazole-4-carbonyl} H N H< 258 N anino)-6,7-dihydro-4H thiazolo[5,4-cjpyridin-5 y1methyl]-pyrrolidine-1 carboxylic acid tert-butyl ester 0 Isoquinoline-3-carboxylic acid S NH /N [4-(5-quinolin-3-ylmethyl - NA N /N 259 HN N 4,5,6,7-tetrahydro-thiazolo[5,4 \- o / ~c]pyridin-2-ylcarbamoyl)-1H benzoimidazol-2-yl]-anide H 2S- {3-[({2-[(Isoquinoline-3 N4 CHJ, N N0 carbonyl)-aminoj-1
H
0yN N benzoimidazole-4-carbonyl} 260 0 amino)-methyl]-piperidin-1 ylmethyl}-pyrrolidine-1 carboxylic acid tert-butyl ester H 2R-{3-[({2-[(Isoquinoline-3 C H N N N carbonyl)-aninol-1H 3, 040 N ,>A N- benzoimidazole-4-carbonyl} 261N \N/ amino)-inethyl] -piperidin-1 yhnethyl} -pyrrolidine-1 carboxylic acid tert-butyl ester 75 WO 2006/099379 PCT/US2006/009049 0 Isoquinoline-3-carboxylic acid H [4-(5-pyrrolidin-2S-ylmethyl 262 N HN S N 4,5,6,7-tetrahydro-thiazolo[5,4 N c]pyridin-2-ylcarbamoyl)-IH benzoimidazol-2-yl]-amide N Isoquinoline-3-carboxylic acid (4-{[1-(1H-imidazol-2 263 N ylnethyl)-piperidin-3 O ylmethyl]-carbamoyl1}-H benzoimidazol-2-yl)-amide N Isoquinoline-3-carboxylic acid N U4-([ 1-(1-methyl-1H-imidazol 264 3 0c NU N 2-ylnethyl)-piperidin-3 N /ylnethyl}-carbamoyl}-1H benzoimidazol-2-yl)-amide Isoquinoline-3-carboxylic acid HN N ON (4-{[I-(1H-imidazol-4 265 N ylnethyl)-piperidin-3 ylnethyl)-carbamoyl}-1H bcnzoirnidazol-2-yl)-amide F Isoquinoline-3-carboxylic acid N {7-[(4-fluoro-benzyl)-methyl 266 NIX- NH H3C N arbamoyl]-tH-benzoimidazol \ / (0 2-yl}-amide 0 NH Isoquinoline-3-carboxylic acid 267 HN N [ (4-dibenzylcarbamoyl-1H N \b /\benzoimidazol-2-yl)-amide 76 WO 2006/099379 PCT/US2006/009049 H H 1-Oxo-1,2-dihydro 0 N N O N isoquinoline-3-carboxylic acid 268 N -- [4-(1H-imidazol-2 H x NH ' ylcarbamoyl)-1H 0 benzoimidazol-2-yl]-amide SCH 6-(4-Methoxycarbonyl-1 H 269 I N benzoimidazol-2-ylcarbamoyl) N H N O CH 3,4-dihydro-1H-isoquinoline-2 H S -: H3 carboxylic acid tert-butyl ester 6-[4-(1H-Imidazol-2 ylcarbamoyl)-1H 270 N benzoimidazol-2-ylcarbamoyl] *-N oH 3,4-dihydro-1H-isoquinoline-2 0 Ccarboxylic acid tert-butyl ester 1,2,3,4-Tetrahydro N NH isoquinoline-6-carboxylic acid 0 NH 271 0 14-(l H-imidazol-2 N ylcarbanoyl)-1H N H NH benzoimidazol-2-yl]-amide H N NH2-Benzenesulfonyl-1,2,3,4 tetrahydro-isoquinoline-6 272 carboxylic acid [4-(IH 72 1 1- N I im idazol-2-ylcarb am oyl)-1H o b benzoimidazol-2-yl]-amide 2-Benzyl-1,2,3,4-tetrahydro isoquinoline-6-carboxylic acid 0 NI 273 {4-(1H-inidazol-2 273 ~ ~ 0; ylcarbanoyl)-IH benzoinidazol-2-yl)-amide 77 WO 2006/099379 PCT/US2006/009049 6-[4-(1H-Imidazol-2 ylcarbamoyl)-lH 274 0 benzoimidazol-2-ylcarbamoyl] 06CHN 3,4-dihydro-1H-isoquinoline-2 carboxylic acid methyl ester 2-Methanesulfonyl-1,2,3,4 N NH o atetrahydro-isoquinoline- 6 275 0 N carboxylic acid {4-(1H N CH imidazol-2-ylcarbamoyl)-1H benzoimidazol-2-yll-amide 6-[4-(1H-Inidazol-2 ylcarbamoyl)-1H 276 benzoimidazol-2-ylcarbamoyll 7 N O 3,4-dihydro-H-isoquinoline-2 o carboxylic acid ethyl ester PVH 6-[4-(lH-Imidazol-2 O Nylcarbamoyl)-1H 277 N benzoinidazol-2-ylcarbamoyl] N C3,4-dihydro-IH-isoquinoline-2 0 carboxylic acid propyl ester 6-[4-(1H-Imidazol-2 o NH ylcarbamoyl)-1 H 278 N benzoimidazol-2-ylcarbamoyl] OH2 3,4-dihydro-1H-isoquinoline-2 carboxylic acid isobutyl ester 6-[4-( 1H-Imidazol-2 ylcarbamoyl)-1H 0 NH 279 N 0 benzoimidazol-2-ylcarbamoyl 0 CH, 3,4-dihydro-1H-isoquinoline-2 Y H carboxylic acid isopropyl ester 78 WO 2006/099379 PCT/US2006/009049 6-{4-(1H-Imidazol-2 N NH ylcarbamoyl)-1H 280 N benzoimidazol-2-ylcarbamoyl] N1 O C 3,4-dihydro-IH-isoquinoline-2 carboxylic acid butyl ester H3C CH 3 3S-[4-(1H-Imidazol-2 K O H C ylcarbamoyl)-1H 281 NH N N benzoimidazol-2-ylcarbamoyll N - 3,4-dihydro-1H-isoquinoline-2 H carboxylic acid tert-butyl ester H 1,2,3,4-Tetrahydro N N 0 CNH isoquinoline-3S-carboxylic acid H H 282 N N {4-(1H-imidazol-2 H \ / ylcarbamoyl)-lH benzoimidazol-2-yl]-ainde H 2-Methanesulfonyl-1,2,3,4 JN N 0 NH O N H C 40 tetrahydro-isoquinoline-3S 283 NN carboxylic acid [4-(iH N H Oimidazol-2-ylcarbamoyl)-IH benzoinidazol-2-yl]-amide N N Q 2-Ethyl-1,2,3,4-tetrahydro NH ~ N H <CH, isoquinoline-3S-carboxylic acid 284 I N N [4-(1H-imidazol-2 N N H ylcarbamoyl)-lH benzoimidazol-2-yl]-amide N N O H C CH 3 3S-[4-(tH-ImidaZol-2 CNH H C- o ylcarbamoyl)-1H I ~ N N benzoimidao1-2-ylcarbamoyl] 285 0 / N 1,3,4,9-tetrahydro-beta carboline-2-carboxylic acid tert-butyl ester 79 WO 2006/099379 PCT/US2006/009049 H 3S-[4-(1 H-Imidazol-2 NH H ycarbamoyl)-1H N N H benzoimidazol-2-ylcarbamoyl] 286 N 1-phenyl-1,3,4,9-tetrahydro beta-carboline-2-carboxylic acid tert-butyl ester H N O 2,3,4,9-Tetrahydro-11-beta NH N carboline-3-carboxylic acid [4 287 N H H 2 N (1H-imidazol-2-ylcarbamoyl) 1H-benzoimidazol-2-yfl-amide N 1-[4-(1H-Imidazol-2 O NHH ylcarbamoyl)-1H o N benzoimidazol-2-ylcarbamoyl] 288-N 6,7-dimethoxy-3,4-dihydro-1H H N soquinoline-2-carboxylic acid
H
3 CH 0 tert-butyl ester H 3 C CEH3 N NH CH 6,7-Dimethoxy-isoquinoline-1 OCH, 0 NH carboxylic acid [4-(IH 289 N 0 iidazol-2-ylcarbamoyl)-IH N I benzoimidazol-2-yl]--anide NH N/ H 7-Nitro-isoquinoline-3 NH Os yN carboxylic acid [7-(1H 290 NN NH N Nimidazol-2-ylcarbamoyl)-1H \ c / Qbenzoimidazol-2-yl)-amide 80 WO 2006/099379 PC T/US2006/009049 o 7-Methanesulfonylamino NH isoquinoline-3-carboxylic acid 291 HN N / NH [7-(IH-imidazol-2 oC'o N ylcarbamoyl)-IH CH, \H / L~benzoimidazol-2-yl]-amide CH, 0 6 2-[(7-Benzyloxy-isoquinoline 0 292N- 3-carbonyl)-amino]-1H 292 N6 N O benzoimidazole-4-carboxylic acid methyl ester 7-Benzyloxy-isoquinoline-3 X N 0 carboxylic acid [4-(1l- 293 N> N d H ~imidazol-2-ylcarhamoyl)-1IH benzoimidazol-2-yl]-amide 6-Methoxy-isoquinoline-3 o /carboxylic acid {4-(1H 294 HC' N N imidazol-2-ylcarbamoyl)- IlH N benzoimidazol-2-yl]-amide N ' 8-Methoxy-isoquinoline-3 N N carboxylic acid [4-(IH 295 N NK 1 N imidazol-2-ylcarbamoyl)-IH 0'CHa benzoimidazol-2-yl]-amide o H 7-Methoxy-isoquinoline-3 o \ /carboxylic acid [4-(1H 296 H C N N imidazol-2-ylcarbamoyl)-H oN H benzoimidazol-2-yl]-amide 81 WO 2006/099379 PCT/US2006/009049 6-Isopropoxy-isoquinoline-3 NH1 N 29 N carboxylic acid [4-(1H 297 H imidazol-2-ylcaTbamoyl)-1H CH, \/CH, benzoimidazol-2-yl]-amide o I W 1-(2-Methylsulfanyl-ethyl) N ~ ~~-N 50oquinoline-3-carboxyliC aCid 298 N [4-(lH-imidazol-2 ylcarbamoyl)-1 H benzoimidazol-2-yl)-amide o I$ 1-(2-Methanesulfonyl-ethyl) N N isoqinoline-3-carboxylic acid HNN 299 N [4-(1H-imidazol-2 ylcarbamoyl)-1 H benzoimidazol-2-yl]-amide 0 1-Methyl-isoquinoline-3 N N carboxylic acid [4-(IH N3 00 imidazol-2-ylcarbamoyl)-1H cH benzoimidazol-2-yl]-anide o ~J7-Chloro-8-methoxy 0 N isoquinoline-3-carboxylic acid 301 (4-(1H-imidazol-2 ci ylcarbanoyl)-IH HC benzoimidazol-2-yl]-amide N' NH Thieno[2,3-c]pyridine-5 302 o NH carboxylic acid f4-(111 N imidazol-2-ylcarbamoyl)-1H benzoimidazol-2-yl]-amide 0 N Thieno[3,2-c]pyridine-6 0 -g 303 s N N carboxylicacid [7-(1H H N- inidazol-2-ylcarbamoyl)-1JI 82 WO 2006/099379 PCTfUS2006/009049 benzoimidazol-2-yl}-amide H H 2-[(2-Methyl-imidazo[1,2 \ ajpyridine-3-carbonyl)-amino N N WN 304 NN I IH-benzoimidazole-4 N CH H carboxylic acid (lIH-imidazol
CH
3 2-yl)-amide H N N \j 24(8-Methoxy-imidazo[1 ,2 o N a]pyridine-2-carbonyl)-amino] 305 0 HN 3H-benzoimidazole-4 N N N H carboxylic acid (1H-imidazol o-cH 2-yl)-amide 0-CH, 6,7-Bis-(2-ncthoxy-ethoxy) Y isoquinoline-3-caTboxylic acid 0 NH 306 0 [4-(1H-imidazol-2
CH
3 ylcarbanoyl)-1H CH, benzoimidazol-2-yl]-amide H N N 0 NH O Cinnoline-3-carboxylic acid [4 307 N\>-N N=N (lH-imidazol-2-ylcarbanoyl) H O 1H-benzoimidazol-2-ylJ-anide 0 u H N N 0 Quinoxaline-2-carboxylic acid NH N [4-(1IH-imidazol-2 308 N N ylcarbamoyl)-1H H o N benzoinidazol-2-yl]-amide NH C-DrN H2 [(6-Bromo-pyridine-2 309 NH carbonyl)-amino]-IH benzoimidazole-4-carboxylic N/ acid (IIH-imidazol-2-yl)-amide r 83 WO 2006/099379 PCT/LS2006/009049 H [1,8]Naphthyridine-2 o \ carboxylic acid [4-(1IH 310 N N N N N N N / imidazol-2-ylcarbamoyl)-IH HA benzoimidazol-2-y]-amide 0 Isoquinoline-4-carboxylic acid NH [4-(1H-imidazol-2 311 , NN L HN N ylcarbamoyl)-lH N benzoinidazol-2-yl]-anide 6,7-Dimethoxy-isoquinoline-3 oN carboxylic acid [4-(1H 312 cO C HN N imidazol-2-ylcarbamoyl)-1H 0H o benzoimidazol-2-y1]-amide
OH
3 H H 0 2-(4-Cyano-benzoylamino)-1H 313 benzoimidazole-4-carboxylic 0 / N acid (IH-inidazol-2-yl)-amide H N N 0 2-[(6-Cyano-pyridine-3 carbonyl)-amino]-1H NH N benzoimidazole-4-carboxylic 0 ~ /acid (1H-imidazol-2-yl)-anide Isoquinoline-5-carboxylic acid S NH [4-(IH-imidazol-2 315 3 N I ylcarbamoyl)-1H benzoinidazol-2-yl]-amide H N 0 2-[(2,6-Dimethoxy-pyrimidine NH N H O-CH 3 4-carbonyl)-amino]-1H 316 N N N N benzoimidazole-4-carboxylic 0oCH acid (1H-imidazol-2-yl)-anide 84 WO 2006/099379 PCT/JS2006/009049 6-Benzyloxy-isoquinoline-3 Sbcarboxylic acid {4-(1H 317 imidazo1.-2-ylcarbamoy1)-1H benzoimidazol-2-yl]-amide o 2,3-Dihydro-[1,4]dioxino(2,3 o \N g]isoquinoline-8-carboxylic NN N 318 O Z acid [4-(1H-imidazol-2 0N H ylcarbamoyl)-1 H benzoimidazol-2-yl]-amide [1,3]Dioxolo[4,5 o N / g]isoquinoline-7-carboxylic NN N 319 N < I acid [4-(1H-imidazol-2 O N H ylcarbamoyl)-1H benzoimidazol-2-y]-amide N 6-Cyclopentyloxy-isoquinoline 320 3-carboxylic acid [4-(1H imidazol-2-ylcarbamoyl)-IH benzoimidazol-2-yl]-amide 1-Cyclopentylnethyl-7 N N nethoxy-isoquinoline-3 321 H 3 C, ,,- -N carboxylic acid [4-(lH imidazol-2-ylcarbamoyl)-1H benzoimidazol-2-yl]-amide INN 1-Isopropyl-isoquinoline-3 322 N carboxylic acid [4-(iH S Nimidazol-2-ylcarbamoyl)-lH
H
3
CH
3 benzoimidazol-2-yl]-amide 85 WO 2006/099379 PCT/US2006/009049 N o 6-Ethoxy-isoquiloline-3 323 N Nc. carboxylicacid [4-(1 imidazol-2-ylcarbamoyl)-1H o-C benzoimidazol-2-yl)-amide 0 HaC, O NH 6-Butoxy-isoquinoline-3 324 HN N carboxylic acid [4-(1H 3 4HN -J\ m N imidazol-2-Icarbamoyl)-1H O/ N benzoimidazo-2-yl)-amide 325 Z .N n$carboXyliC acid [4-(1H imlidazol-2-carbamoyl)-H NT imi dazo - -cr a o l-H c Ubenzoimidazo-2-yl-amide o 4 M
-
a N -Obutyl-ISoquinoline-3 32N carboxylic acid [4-(IH imidazol-2-lcarbamoyl)-1H H1, beZoinidaZol-2-yl-amide S\-cobuty-isoquinoline-3 328 carboxylic acid [4-(fH imidazol-2-ylcarbamoyl)- 1H benzoimidazol-2-y1]-amide 86 -Iobety-isoquinojin3g 32. - < carboxylic acid [4-'IH N NCHimidazol-2-ylcarbamoyly
H
H, ~benzoimnidazo-2-yljyamide 86 WO 2006/099379 PCT/US2006/009049 N. o 7-Methoxy-1-methyl \NH N CHisoquinoline-3-carboxylic acid 329 N_ [4-(1H-imidazol-2 ylcarbamoyl)-1H CH, benzoimidazol-2-yl]-amide o 1 -Methyl-6-trifluoromethoxy N NH isoquinoline-3-carboxylic acid F 330 CH N N [4-(1H-imidazol-2 ylcarbamoyl)-lH o benzoimidazol-2-yl]-amide N o 7-Methanesulfonyl- 1-methyl N CN isoquinoline-3-carboxylic acid 331 [4-(1H-imidazol-2 / =0 ylcarbamoyl)-IH
CH
3 benzoimidazol-2-yl]-amide 1-(Tetrahydro-pyran-4-yl) N isoquinoline-3-carboxylic acid 332 [4-(IH-imidazol-2 ylcarbamoyl)-1H 0 benzoimidazol-2-yl]-amide o 1 -Methyl-7-trifluoromethoxy NH isoquinoline-3-carboxylic acid 333 F-N N [4-(1H-imidaZol-2 F CH ylcarbamoyl)-1H o benzoimidazol-2-yl]-amide 0' 0 5,8-Dimethoxy-isoquinoline-3 334 N carboxylic acid [4-(1H o N N imidazol-2-ylcarbamoyl)-1H \ / Nbenzoinidazol-2-yl]-amide 87 WO 2006/099379 PCT/US2006/009049 N NH 4-Methoxy-quinoline-2 N carboxylic acid [4-(lH 335 N imidazol-2-ylcarbamoyl)-1H H benzoimidazol-2-yl]-amide 7-Methoxy-isoquinoline-1 O N NH carboxylic acid [7-(IH N36H iidazol-2-ylcarbaoy-IH benzoimidazol-2-yl}-amide N& NH 24(hnidazo[1,2-a]pyridine-2 a N carbonyl)-amino}-1H 337 N 0 benzoimidazole-4-carboxylic ~N acid (1H-imidazol-2-yl)-amide H H 2-[(5-Methyl-imidazo[1,2 o \.N alpyridine-2-carbonyl)-amino] 338 N 1H-benzoimidazole-4 338NN ' H carboxylic acid (1H-imidazol 2-yl)-amide 0 2-[(Lnidazo[2,1-blthiazole-6 o carbonyl)-amino]-IH 339 N benzoimidazole-4-carboxylic H N N H acid (1H-imidazol-2-yl)-amirde 2-[(8-Methyl-imidazo[1,2 t a]pyridine-2-carbonyl)-amino] 340 N 1fH-benzoimidazole-4 N N- carboxylic acid (I H-imidazol H20 2-yl)-amide 88 WO 2006/099379 PCT/US2006/009049 oa 2I-{2,2-Bis-(4-chloro-pheny1) / \ acetylanino]-IH 341 H benzoimidazole-4-caboxylic acid (IH-imidazol-2-yl)-amide NNH 2-[(6-Phenyl-pyridine-2 N carbonyl)-amino]-1H 342 N N N benzoimidazole-4-carboXylic acid (1H-inidazol-2-yl)-amide N NH 2- { [6-(3-Cyano-phenyl) N " 0 pyridine-2-carbonyl-amilO} 343 IH-benzoimidazole-4 H carboxylic acid (1 H-imidazol 2-yl)-amide NC , o 2-[(5-Phenoxy-pyridine-3 344 N N N carbonyl)-amino]-lI 3 benzoitnidazole-4-carboxylic o acid (lH-imidazol-2-yl)-amide N N o N N N 2-[(5 -Benzyloxy-pyridine- 3 - -N carbonyl)-amino-lfH N 345 H b /enzoinidazole-4-carbOXylic /b \acid (1H-imidazol-2-yl)-anide 89 WO 2006/099379 PCT/US2006/009049 N NH 2-{[6-(2-Carbamoyl-phenyl) 346 N IE-benzoimidazole-4 H N carboxylic acid (IH-imidazol NH, 2-yI)-ainide NH 2- {[6-(2-Trifluoromethoxy N phenyl)-pyridine-2-carbonyll 347 amino}-1H-benzoimidazole- 4 0> Fcarboxylic acid (1H-inidazol F '~ F 2-yl)-amide H 2- {f6-(4-Fluoro-phenyl) N N 0 ~NH Npyridine-3 -carbonyl]-amino} ~- N H 348 N N 1H-benzoimidazole-4 H \F\ carboxylic acid (IH-imidazol 2-yl)-amide 2- {{6-(4-Trifluorornethyl H phenyl)-pyridine-3-carbonyll 349 N F amino)-1H-benzoimidazole-4 ~ F F carboxylic acid (1H-imidazol 2-yl)-amide N o 2-{[6-(4-Trifluoromethoxy HX N phenyl)-pyridine-3-carbonyl] 350 N amino}-1H-benzoimidazole-4 O FVF carboxylic acid (1H-imidazol 2-yl)-amide 2- {[5-(3-Fluoro-phenyl) NH pyridine-3-carbonyl]-amino} O NH 351 0 1H-benzoimidazole-4 N F carboxylic acid (1H-imidazol N 2-yl)-amide 90 WO 2006/099379 PCT/US2006/009049 o fl2-{{5-(4-Trifluoromethoxy F O N phenyl)-pyridine-3-carbonyl] 352 N amino]}-1I-benzoimidazole-4 N carboxylic acid (1H-inidazoi 2-yl)-amide 2-{[5-(2,4-Difluoro-phenyl) pyridine-3-carbonyl]-amino} 353 1H-benzoimidazole-4 F carboxylic acid (IlH-imidazol F 2-yl)-amide N 0 2-{[5-(3-Trifluoromethoxy CIL N pheniy1)-pyridine-3-carbonyl] 354 amino}-lH-benzoimidazole-4 o carboxylic acid (IlH-imidazol F F 2-yl)-amide NY NH 2-((6-Furan-2-yl-pyridine-2 o NH 0 carbonyl)-amino]-1H N benzoimidazole-4-carboxylic N acid (1H-imidazol-2-yl)-anide N NH 2- {{6-(2-Fluoro.-pheny1) ON NH pyridine-2-carbonyl]-amino} 356 ) NIH-benzoimidazole-4 H carboxylic acid (IH-imidaZol ; IF 2-yl)-amide o 2- {[6-(4-Methoxy-phenyl) 0 N pyridine-3-carbonyl]-amino) N N 357 1H-benzoimidazole-4 ~N N H carboxytic acid (IH-imidazol 0 2-yl)-amide 91 WO 2006/099379 PCT/US2006/009049 0 /2-[(5-Phenyl-pyridine-2 N N carbonyl)-amino]-11 N I benzoinidazole-4-carboxyliC acid (1H-imidazol-2-yl)-amide 2-[(5-Plienyl-pyridine-3 carbonyl)-amino]-1H 359 benzoimidazole-4-carboxylic / \ acid (IH-imidazol-2-yl)-amide N 2- {5-(4-Methoxy-phenyl) NH NHN _N pyridine-3-carbonyl]-amino} 360 / \ / 1H-benzoimidazole-4 / \ carboxylic acid (1H-imidazol O-CH2-yl)-amie o f2-[(4-Phenyl-pyridine-2 0 \carbonyl)-amino]-1lH 361 N -ibenzoimidazole--4-carboxylic N acid (1H-imidazol-2-yI)-amide N NH. 2- { [6-(2-Methoxy-phenyl) o NH pyridine-2-carbonyl]-ainino} 362 I 1H-benzoimidazole-4 H N carboxylic acid (1H-imidazol CH 1 2-yl)-amide N NH 2- {[6-(3 -Methanesulfonyl Os 1 NHN 1 phenyl)-pyridine-2-carbonyl 363 II- N amino}-IH-benzoimidazole-4 carboxylic acid (I H-imidazol 2-yl)-a2ide 92 WO 2006/099379 PCT/US2006/009049 N NH 2- {[6-(3-Aminomethyl phenyl)-pyridine-2-carbonyll 364 N arnino} -1 H-benzoimidazole-4 H N carboxylic acid (1H-imidazol 2-yl)-anide
NH
2 P-\ N NH 2-{[2-(3-Cyano-phenyl) pyridine4-carbonyl]-amino} 365 N IH-benzoimidazole-4 H N carboxylic acid (I H-imidazo 2-yl)-amide CN 2-[(6-Phenyl-pyridine-3 N 3 N carbonyl)-amino]-1H 366 -' benzoinidazole-4-carboxylic N acid (tH-imidazol-2-yl)-amide N N ox 2-(3-Cyano-benzoylamino)-IH 367 N benzoimidazole-4-carboxylic A N acid (lH-imidazol-2-yl)-amide CN o k AI t2-{[6-(3-Cyanomethyl-phenyl) N N41 pyridine-2-carbonyl]-amino} 368 -N 1H-benzoimidazole-4 carboxylic acid (IH-imidazol e ON2-yl)-amide 93 WO 2006/099379 PCT/US2006/009049 H H 0 N o N 2-{[6-(4-Methanesulfonyl N N - )helyl)-pyridine-2-carbonlyl] H N 369 H amino}-1H-benzoimidazole-4 carboxylic acid (1 H-imidazol 2-yl)-amide o H 2-[(3'-Cyano-biphenyl-3 370 N)-- carbonyl)-anino)-1H 370 H /benzoimnidazole-4-carboxylic / N acid (1H-inidazol-2-yl)-amide H H N H 2-((4'-Cyano-biphenyl-3 3N carbonyl)-amino]-1H 371 H / benzoimidazole-4-carboxylic - acid (1H-imidazol-2-yl)-amide N [2,4']Bipyridinyl-6-carboxylic 37'N acid[7-(1 H-imidazol-2 372 N NH ylcarbamoyl)-1H / o benzoimidazol-2-yl]-amide 0 [2,3']Bipyridinyl-6-carboxylic 373 -N N// acid[7-(1H-imidazol-2 ylcarbamoyl)-IH s N benzoimidazol-2-yl]-amide N NH 2-[(2-Phenoxy-pyridine-3 o carbonyl)-amino]-lH 3740 0 N benzoimidazole-4-carboxylic 3N acid (1H-imidazol-2-yl)-amide 94 WO 2006/099379 PCT/US2006/009049 2-[(3'-Cyano-biphenyl-4 carbonyl)-amino]-1HI 375 -benzoimidazole-4-carboxylic acid (lH-inidazol-2-yl)-arnide - -2-[(4'-Cyano-biphenyl-4 N carbonyl)-anino]-lH 376 benzoimidazole-4-carboxylic o) acid (1H-inidazol-2-yl)-amide o 2-{[6-(3-Cyano-phenyl) o N pyridine-3-carbonyl]-amino) 377 1f-benzoimidazole-4 carboxylic acid (1H-imidazol N 2-yl)-.amide 2-(3-Pyridin-3-yl 378 benzoylamino)-1H benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide H o 0 2-(4-Pyridin-3-y1 379 N H benzoylanino)-1H -N - /- benzoimidazole-4-carboxylic N H O - acid (1IH-imidazol-2-yl)-amide tH F 2- { [2-(4-Fluoro-phenoxy) 7 / pyridine-3-carbonyl]-anino} 380 a o IIH-benzoimidazole-4 " N carboxylic acid (1H-imidazol 2-yl)-amide N O 2-(4-Pyridin-4-y 381 NN benzoylamino)-1 H N / N benzoimidazole-4-carboxylic H O acid (1H-imidazol-2-yl)-amide 95 WO 2006/099379 PCT/US2006/009049 D U2-{[6-(2-Cyano-phenyl) N / pyridine-2-carbonyl]-aninlo} 382 N 1H-benzoimidazole-4 CN carboxylic acid (lH-imidazol 2-yl)-amide 2-[(3-Benzyloxy-pyridine- 2 carbonyl)-amino]-lH 383 benzoinidazole-4-carboxylic acid (1 H-inidazol-2-y1)-anide N N O 2-[(6-Benzyloxy-pyridine-2 carbonyl)-amino]-1H 384 CNH N O_ \> N N benzoinidazole-4-carboxylic H \ / acid (IH-imidazol-2-yl)-amide
-
2-[(6-Thiophen-2-yl-pyridine N N 2-carbonyl)-amino]-IH 385 benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide 2-{(5-Cyclopentyloxy-pyridine 6 N 3-carbonyl)-anino]-1H 386 /benzoimidazole-4-carboxylic acid (IH-inidazol-2-yl)-amide 2-[(5-Cyclopentylnethoxy 387 T ~ pyridine-3-carboniyl-aninol N 387 1H-benzoimidazole-4 \ carboxylic acid (1 H-imidazol 2-yl)-amide 96 WO 2006/099379 PCT/US2006/009049 q H 0 2-{[ 5 -(2-Cyclopentyl-ethoxy) N Pyridine-3-carbonyl]-aminoj 388 x1 /H-benzoimidazole4 0 carboxylic acid (1H-imidazol 2-yl)-amide 2-{{5-(1-(2R)-Phenyl-ethoxy) NH N - pyridine-3-carbonyl]-amino} 389 N l CIH-benzoimidazole-4 o carboxylic acid (IH-imidazol 2-yl)-amide N N 0 2- {[5-(1 -(I S)-Phenyl-ethoxy)~ N N pyridine-3-carbonyl]-amino} 390 l CH 1H-benzoimidazole-4 o carboxylic acid (lH-inidazol 2-yl)-anide -N N 2 -[(3-Phenethyloxy-pyridine-2 N N 391 li ocarbonyl)-amino]-H benzoimidazole-carboxylic acid (LH-iniidazol-2-yl)-amide U r2-[(5-Benzyloxy-pyridine-2 392 I N carbonyl)-amino]-l \ benzoimidazole-4-carboxylic acid (IHf-imidazol-2-yl)-aniide N H0 N 2-[(5-Phelethyloxy-pyridiiie-2 393 carbonyl)-amino]-IH 0 benzoimidazole-4-carboxylic / 'acid (1H-imidazol-2-yl)-amide 97 WO 2006/099379 PCT/US2006/009049 N NH 2 -[(5-Cyclopentylmethoxy NH N pyridine-2-carbonyl)-amino] 394 N 1IH-benzoimidazole-4 NN carboxylic acid (I H-imidazol 2-yl)-amide 2- {[5-(2-Cyclopentyl-ethoxy) NH 6 N 4pyridine-2-carbonyl]-aminol 395 3950 H-benzoimidazole-4 carboxylic acid (IH-imidazol 2-yl)-amide NH N 2-[(5-Isopropoxy-pyridine--3 396 N carbonyl)-amino]-1H CH, benzoimidazole-4-carboxylic CH, acid (IH-imidazol-2-yl)-amide H NN ON o 2-{[5 -(1 -Ethyl-propoxy) \>H H pyridine-3-carbonyl]-amino I N ~N 397 'NN H \97H, 1H-benzoinidazole-4 o carboxylic acid (1H-imiidazol HaC 2-yl)-amide N 2-[(5-Cyclopropylmethoxy NH pyridine-3-carbonyl)-amino] 398 N1H-benzoimidazole-4 o-l carboxylic acid (1H-imidazol 2-yl)-amide 2- {[5-(1 -Cyclopropyl-ethoxy) NH pyridine-3-carbonyl]-anino} 399 NC/ 1H-benzoimidazole4 carboxylic acid (IH-imidazol 2-yl)-amide 98 WO 2006/099379 PCT/US2006/009049 N 2 -[(5-Propoxy-pyridine-3 400 N carbonyl)-ainino]-[H 4benzoinidazole4-carboxylic CH, acid (1 H-imidazol-2-yl)-amide N 2
-[(
5 -Butoxy-pyridine-3 401 N carbonyl)-amino]-IH- 0 benzoimidazole-4-carboxylic acid (lH-imidazol-2-yl)-amide N N 2-[(5-Isobutoxy-pytidine-3 402 --Ncarbonyl)-aninoyH benzoinidazole-4-carboxylic H CR acid (IH-imidazol-2-yl)-amide F2-{[ 4
-(
2 -Cyclopentyl-ethoxy) iN dine-2-carbonyl-amino} 403 \IH-benzoimidazole-4 carboxylic acid (lH-imidazol 2 -yl)-amide Ne 2
-[(
6 -PhnyI-pyrimidine-4 404 N carbonyl)-amino]-1H benzoimidazole-4-carboxylic acid (IH-imidazol-2-yl)-amide N N2- {[6-( 4 -Fluoro-phenyl) NH&N -- N N\Pyrimidine-4-carbonyl] 405 6N N anino}-1Hbenzoimidazole-4 / carboxylic acid (lH-imidazol 2 -yl)-amide 99 WO 2006/099379 PCT/US2006/009049 H HN' 2
-[
4 -(5-Tritluoromethyt O HN 40-0H Pyfidii-2-yl)-henzoylarninjO] 40 3 H-benzoimidazole-4 | N carboxylic acid (iH-imidazol F FN F 2-yl)-amide H N 0 N O HN 2
-[
4 -(4-Trifluoromethyl 407 N N phenoxy)-benzoylamino]-3H benzoimidazole-4-carboxylic acid (1H-imidazol-2-y)-amide F F 0 NH 2 -{[6-(3-Cyano-phenyl) 408 N N NCH pyridine-2-carbonyl] -amino} 408N NH H -N 3H-benzoimidazole-4 NN Ncarboxylic acid butylamide 2-{[ 6 -(3-Cyano-phenyl) 409 NHN pyridine-2-carbonyl-amino} N N N / 3H-benzoiniidazole-4 N / O carboxylic acid phenylamide NH 2 -{[6-(3-Cyano-phenyl) 410 pyridine-2-carbony]]-amino) N_ 3H-benzoimidazole-4 N0H o carboxylic acid isopropylamide 100 WO 2006/099379 PCT/UJS2006/009049 2-f [6-(3 -Cyano-plienyl) NH pyridine-2-carbonyl]-amino} 411 /N N NH H 3H-benzoinidazole-4 carboxylic acid cyclohexylamide 0 2- { [6-(3-Cyano-phenyl) NH pyridine-2-carbonyl]-amino) 412 N NH 3H-benzoimidazole-4 carboxylic acid (furan-2 N 0ylmethyl)-amide 0 IN, 2-[(4-Phenylethynyl-pyridine 413 0 N N 2-carbonyl)-amino]-1IH 413 K-NN N- benzoimidazole-4-carboxylic H acid (1H-imidazol-2-yl)-amide N N NH 2-[(5-Phenylethynyl-pyridine o H 3-carbonyl)-amino]-1H 414 N benzoimidazole-4-carboxylic acid (1iH-imidazol-2-yl)-amide N' H 2-[(5-Phenethyl-pyridine-3 o H carbonyl)-amino]-1H 415a 415 Ibenzoimidazole-4-carboxylic Nl Hacid (1 H-imidazol-2-yl)-amide N N 2-[(6-Phenylethynyl-pyridine N H 2-caTbonyl)-amino]-1H 416 H benzoimidazole-4-carboxylic acid (1 H-imidazol-2-yl)-amide 101 WO 2006/099379 PCT/US2006/009049 0 NH 2-{ [6-(3-Methanesulfonyl HN N phenylethynyl)-pyridine-2 417 carbonyl]-amino}-I H benzoimidazole-4-carboxylic es, acid (IH-imidazol-2-yl)-amide 0 N N2-[(2-Phenylethynyl-pyridine 418 4-carbonyl)-amino}-l
H
benzoimidazole-4-carboxylic acid (IH-imidazol-2-yl)-amide 2-[(5-Phenylethynyl-pyridine NN 419 N 2-carbonyl)-aimino]-H N beDZoimidaZole-4-carboxylic acid (lH-imidazol-2-yl)-amlide 2-[(6-Cyclohexylethynyl N 0 pyridine-2-carbonyl)-amino} 420 NH N I H-benzohnidazole-4 N N carboxylic acid (ilH-imidazol 2-yl)-amide a NH 2-{[6-(4-Fluoro N HN N H N phenylethynyl)-pyridine-2 421 X carbonyl]-amino}-1H benzoinidazole-4-carboxylic F acid (IH-imidazol-2-yl)-amide 102 WO 2006/099379 PCT/US2006/009049 0 NH 2-{[6-(4-Ethyl-phenylethynyl) RN N N pyridinp-2-carbony]-amaino} 422 N 1H-benzoimidazole-4 carboxylic acid (liH-imidazol NC 2-yl)-amnide 2-{[6-(4-Methoxy N phenylethynyl)-pyridine-2 423 . carbonyl]-amino}-IH benzoimidazole-4-carboxylic o e acid (lH-imidazol-2-yl)-anide 2-{{6-(4-Chloro NHN N N phenylethynyl)-pyridine-2 424 N carbonyfl-anino}-1IH \/ H benzoimidazole-4-carboxylic cl acid (IH-imidazol-2-yl)-amide, 2-[(3-Phenylethynyl-pyridine 425 0 o 2-carbonyl)-amino]-1H N benzoimidazole-4-carboxylic H x acid (1H-imidazol-2-yl)-ainide o 2-{[6-(3-Methyl-but-1-ynyl) NH pyridina-2-carbonyl] -arnino} 426 HN 'N N 1H-benzoimidazole-4 carboxylic acid (IlH-imidazol H8c CH 3 2-yl)-amide 2-[(6-Thiophen-3-ylethynyl NH pyridine-2-carbonyl)-amino] HNHN 427 1H-benzoimidazole-4 0 carboxylic acid (liH-imidazol 2-yl)-amide 103 WO 2006/099379 PCT/US20061009049 o 2-{[6-(3,3-Dimethyl-but-1 'N N ynyl)-pyridine-2-carbonyl] 428 amino)-IH-benzoimidazole-4 0 /carboxylic acid (lH-imidazol
H
3 C CH Ha 2-yl)-amide 2-{[6-(3-Cyclopentyl-prop-1 ynyl)-pyridine-2-carbonyl] 429 H amino }-1H-benzoimidazole4 0 carboxylicc acid (lH-imidazol 2-yl)-amide o 2-{[6-(3-Hydroxy-3-methyl N NH Nbut- -ynyl-pyridine-2 430 - carbonyl]-amino}-1H \ / H benzoimidazole-4-carboxylic HC OH H, Acid (1H-imidazol-2-yl)-amide 2- {[G-(4-Methyl-pent-1 -ynyl) NHN pyridine-2-carbonyl]-aminfo 431 1 H-benzoimidazole-4 Hc /carboxylic acid (I H-imidazol itc 2-yl)-amide NH 2-[(6-Pent-1-ynyl-pyridine-2 NHN N carbonyl)-amino]-1H 432 - /-) benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide H N NH 2- {[6-(4-Dimevhylamino -N H HN N HN phenylethynyl)-pyridine-2 433 caibonyl]-amino}-1H benzoimidazole-4-carboxylic N acid (IH-imidazol-2-yl)-amide HtC Co, 104 WO 2006/099379 PCT/US2006/009049 0 - NH 2-[(6-Pyridin-3-ylethynyl NHN N N pyridine-2-carbonyl)-amino] 434 1 H-benzoimidazole-4 \ / Hcarboxylic acid (1H-imidazol N 2-yl)-amide NH 2-{[6-(3-Methoxy NHNI N N phenylethynyl)-pyridine-2 435 IINcarbonyl)-amino}-lH1 benzoimidazole-4-caiboxylic acid (lH-imidazol-2-yl)-amide CH, 2-{[6-(2-Methoxy N phenylethynyl)-pyridine-2 H- N "NN 436 carbonyl]-amino}-iH H0C benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-anide 2-[(3-Cyclohexylethynyl pyridine-2-carbonyl)-amino] 437 1 IH-benzoimidazole-4 CN T) carboxylic acid (lil-imidazol 2-yl)-amide 2-[(3-Thiophen-3-ylethynyl pyridine-2-carbonyl)-amino) 438 N 1H-benzoinidazole-4 carboxylic acid (IR-imidazol C 2-yl)-amide H 2-[(6-Cyclopropylethynyl N N 0 pyridine-2-carbonyl)-amino] 439 NH N H 1H-benzoimidazole-4 N N \Z carboxylicc acid (H.-inidazol 2-yl)-amide 105 WO 2006/099379 PCT/US2006/009049
H
3 C CH 3 2-{[3-(3,3-Dimethyl-but-1 II H3 H H Nynyl)-pyridine-2-carbonyl] 440 \ (\anino}-1H-benzoimidazole-4 N /Ncarboxylic acid (1H-imidazol N H 2-yl)-amide H 2-{[6-(2-Fluoro N~NH K NH phenylethynyl)-pyridine-2 HN ~N H 441 carbonyl]-amino}-IH F > benzoimidazole-4-carboxylic acid (IH-imidazol-2-yl)-amide H H 0 NN o 2-[(6-m-Tolylethynyl-pyridine IN H N 2-carbonyl)-amino]-1H 442 benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide cHH oN 0 "2-{[6-(3-Fluoro N N / phenylethynyl)-pyridine-2 443 rN carbonyl)-aminol -1 H benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-anide H H C 0 0 N 2-[(3-Chloro-6-pent-1-ynyl KN ss pyridine-2-arboay1)-aminOu H N 444 HH -benzoimidazole-4 carboxylic acid (1H-inidazol CH, 2-y1)-arnide N N H 2-[(6-Ethynyl-pyridine-2 N N 4NHN./ carbonyl)-amino]-1H 5N N benzoimidazole-4-carboxylic N H O /acid (1 H-inidazol-2-yl)-amide 106 WO 2006/099379 PCT/US2006/009049 H H 0 NN N 2-[(6-Phenethyl-pyridine-2 N H carbonyl)-amino]-1H 446H benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide NH 2-[(6-Phenylethynyl-pyridine N HN N 2-carbonyl)-amino]-1H 447 0-OHs benzoimidazole-4-carboxylic acid methyl ester 2-[(6-Phenylethynyl-pyridine | NH 2-carbonyl)-amino]- 1I 448 - N-1 benzoimidazole-4-carboxylic acid 4-methanesuIlfonyl benzylarmide NH 2-[(6-Phenylethynyl-pyridine HN \N 2-carbonyl)-aminoj-111 449 Ibenzoimidazole-4-carboxylic 0 acid cyclopentylamide 0 NH 2-[(6-Phenylethynyl-pyridine NHN N CH, 2-carbonyl)-aminoj-1H 450 el benzoimidazole-4-carboxylic 0 acid tert-butylamide Isoquinoline-3-carboxylic acid o [7-(1H-irnidazol-2 451 0 HN ylcarbamoyl)-5-(3-phenyl N a pTOpionylamino)-IH N benzoimidazol-2-yll-amide 107 WO 2006/099379 PCT/US2006/009049 Isoquinoline-3-carboxylic acid 4N5 [5-benzenesulfonylamino-7 452 o HN (1l-irnidazol-2-ylcarbamoyl) N 0 0 / lH-benzoimidazol-2-yl]-amide NNH Isoquinoline-3-carboxylic acid Y [4-(1 H-imidazol-2 453 ylcarbamoyl)-6 0s methanesulfonylamino-1IH S.benzoimidazol-2-yl-amnide H HN 0 Isoquinoline-3-carboxylic acid 44 N \ CH, {7-(1H-imidazol-2 4 N N <N ylcarbamoyl)-5-methyl- 1H 54e H benzoimidazol-2-yl]-amlide CH, 2-[(Isoquinoline- 3 -carbonyl) 0 arnino]-6-propyl-lH 455N benzoimidazole-4-carboXylic NCN acidd methyl ester 0. CH 3 isoquinoline-3-carboxylic acid [4-(4-methanesulfonyl 456 benzylcarbamoyl)-6-propyl-lH H, bezoimidazol-2-yll-amide O CH 2- (Isoquinoline-3-carbonyl) 457 -~N 0 amino]-6-pyridin- 4 -yl- 1H 457 Ibenzoinidazole-4-carboxylic N acid methyl ester 108 WO 2006/099379 PCT/US2006/009049 Isoquinoline-3-carboxylic acid 0, {4-(4-methanesulfonyl 458 0 NH benzylcarbamoyl)-6-pyridin- 4 NN -- Nyl-1H-benzoimidazol-2-yl] N amide Isoquinoline-3-carboxylic acid 0 N H S F [7-(1IH-imidazol-2 459N F ylcarbamoyl)-5 N N H trifluoromethyl-1H benzoimidazol-2-yl]-amide H C a 6-Cyano-2-[(isoquinoline- 3 40 AH caTbonyl)-amino]-IH NH benzoimidazole4-carbOXylic acid methyl ester oIsoquinoline-3-carboxylic acid N [6-cyano-4-(1H-imidazol-2 461 NA NA H 4ylcarbamoy1)-1H benzoimidazol-2-ylJ-amide 00 H Isoquinoline-3-carboxylic acid NHA [7-(4-methanesulfonyl 462 _ N benzylcarbamoyl)-5-phenyl 1H-benzoimidazol-2-yl]-amide e, Isoquinoline-3-carboxylic acid A [6-isopropyl-4-(4 463 0NH metanesulfonyl N benzylcarbamoyl)-1H N HNd G Hbenzoimidazol-2-yl]-amide 0 NN NH Isoquinoline-3 -carboxylic acid 464 A .Nr 'H N )[7-( IH-irnidazol-2 0 ylcarbamoyl)-5-phenyl-1Hf benzaimidazol-2-yl]-amide 109 WO 2006/099379 PCT/US2006/009049 HIsoquinoline-3-carboxylic acid % [6-furan-3-yl-4-(4 465 0 NH methanesulfonyl benzylcarbamoyl)-IH 0'I benzoimidazol-2-yl]-anide Isoquinoline-3-carboxylic acid NH {7-(4-methanesulfonyl 466 0- benzylcarbamoyl)-5 F trifluoromethyl-1H benzoimidazol-2-yl]-amide N H Isoquinoline-3-carboxylic acid O NH {4-(1H-imidazol-2 467 ylcarbamoyl)-6-pyridin- 4 -yl N N H-benzoimidazol-2-yl]-amide 9Che Isoquinoline-3-carboxylic acid p [4-(4-methanesulfonyl 468 o H I benzylcarbamoyl)-6-thiophe-n 1 N - 3-yl-1H-benzoimidazol-2-yl] s amide Isoquinoline-3-carboxylic acid 469 0 (\/ N (4-ethy1carbamoyl-6-pyridin-4 m yl-1H-benzoimidazol-2-yl) I9- amide Isoquinoline-3-carboxylic acid 470 N N (4-cyclopentylcarbamoyl-G 470N -N N pyridin-4-yl- il H- H benzoimidazol-2-yl)-amide 110 WO 2006/099379 PCT/US2006/009049 o CH, 6-Ethoxy-2-[(isoquinoline-3 o HN \ / carbonyl)-amino]-3H 471 N N benzoimidazole-4-carboxylic acid ethyl ester ] Isoquinoline-3-carboxylic acid 472 o HN o [5-ethoxy-7-(lH-imidazol- 2 47N N H 3 0 ylcarbamoyl)-IH benzoimidazol-2-y1]-amide ] Isoquinoline-3-carboxylic acid o73 R / [5-benzyloxy-7-(1H-imnidazol 43N N 2-ylcarbamoyl)-lU benzoimidazol-2-yl]-amide H -4 Isoquinoline-3-carboxylic acid O N 474 0 NN C, /[7-(1H-imidazol-2 N N N H ylcarbanoyl)-5-methoxy-IH benzoimidazol-2-yl]-amide ocH 3 S=O H lsoquinoline-3-carboxylic acid Ho N [7-(4-methanesulfonyl 475 HN \ benzylcarbamoyl)-5-methoxy N N 5 N H 1 H-benzoimidazol-2-yl]-amide KN Isoquinoline-3-carboxylic acid 476 o HN ~/[7-(IH-inidazol-2 N476N ylcarbamoyl)-5-propoxy-IH AF benzoimidazol-2-yl]-amide 111 WO 20061099379 PCT/US2006/009049 H N ) Isoquinoline-3-carboxylic acid 0 N [7-(1 H-imidazol-2 477 o 0N \1 -H 477N NC ylcarbamoyl)-5-isopropoxy-1H benzoimidazol-2-yl]-anide H- Isoquinoline-3-carboxylic acid [5-butoxy-7-(IH-imidazol-2 47 NN N ylcarbamoyl)-IFI H H,C benzoimidazol-2-yl}-amide o-cH, o O-2-[(Isoquinoline-3-carbonyl) 0 HN anino]-7-methcixy-3H 479 N N -CH3 benzoimidazole-4-carboxylic acid methyl ester 0 Isoquinoline-3-carboxylic acid NH[7-(4-mneth-anesulfonyl 480 0 benzylcarbamoyl)4-methoxy N JN C IH-benzoimidazol-2-yl]-amide H HC O-CH, o 0OH 3 2-[(Isoquinoline-3-carbonyl) SRN \ amino] -5-nethoxy-3 H benzoimidazole-4-carboxylic acid methyl ester CNN N NH Isoquinoline-3-carboxylic acid o 0-CR" [7-(1H-imidazol-2 482 0 HN / ylcarbamoyl)-6-methoxy-lH H benzoimidazol-2-yll-amide 112 WO 2006/099379 PCT/US2006/009049 0 Isoquinoline-3-carboxylic acid Nr 7-(4-nethanesulfonyl 483 0= 0J-CH. Ho benzylcarbamoyl)-6-methoxy N ' 1H-benzoimidazol-2-yl]-anide H N N I o N 6,7-Dimethoxy-isoquinoline-3 N \ / \-CH carboxylic acid [5-ethoxy-7 484 NaN N 484 H (1H-imidazol-2-ylcarbamoyl) HCsO IH-benzoimidazol-2-yl]-anide O'CH, O Isoquinoline-3-carboxylic acid [5-ethoxy-4-fluoro-7-(4 NH 485 -methanesulfonyl HN \ / -Ci benzylcarbamoyl)-1H H benzoimidazol-2-yl]-amide NH Isoquinoline-3-carboxylic acid 0 [5-ethoxy-4-fluoro-7-(1IH 486 O HN\/ 4N N 0 -CH, imidazol-2-ylcarbamoyl)-1I N N F benzoimidazol-2-yl]-amide NH N NH Isoquinoline-3-carboxylic acid 0 F [4,6-difluoro-7-(1H-imidazol-2 487 o HN oi 2 N FCH 3 ylcaTbamoyl)-5-methoxy-1H benzoinidazol-2-yl]-amide 113 WO 2006/099379 PCT/US2006/009049 Nl Isoquinoline-3-carboxylic acid o8 N[7-(1H-imidazol-2 48 H ylcarbamoyl)-benzooxazol-2 yl]-amide 0 N)fIsoquinoline-3-carboxylic acid 489 0 HN \/ [5-ethylsulfanyl-7-(H 49 N H 3 C imidazol-2-ylcarbamoyl)-iH benzoimidazol-2-yl}-anide Isoquinoline-3-carboxylic acid o90 0 H / [5-butylsulfanyl-7-(tH 490 N NAN cH, inidazol-2-ylcarbamoyl)-1H benzoimidazol-2-yl]-anide H Isoquinoline-3-carboxylic acid O N 491 0 IN\[5-ethanesulfonyl-7-(IH NN1N H C imidazol-2-ylcarbamoyl)-JH benzoimidazol-2-yl]-amide H N Isoquinoline-3-carhoxylic acid o NOt [7-(1 H-imidazol-2 492 cN \ ylcarbamoyl)-5 N N H methanesulfonyl-1H benzoimidazol-2-yl]-amide ) Isoquinoline-3-carboxylic acid 493 fN 5-benzenesulfonyl-7-(IH N N imidazol-2-ylcarhamoyl)-1H benzoimidazol-2-yl]-amide 114 WO 2006/099379 PCT/US2006/009049 H 1-b Isoquinoline-3 -carfboxylic acid 494 o [5-(butane-1-sulfonyl)-7-(IH Simidazol-2-ylcarbamoyl)- I H benzoimidazol-2-yl]-amide 0 0 Hd / Isoquiiline-3-carboxylic acid 495 0 (7-benzenesulfonylamino-iH N benzoinidazol-2-yl)--amide o o 0- -S.1
H
3 C NH Isoquinoline-3-carboxylic acid 496 N N (4-ethanesulfonylamino-LH benzoimidazol-2-yl)-amide N N Isoquinoline-3-carboxylic acid 497 N (5-amino-1H-benzoiniidazol-2 H yl)-amide s D N N Isoquinoline-3-carboxylic acid 498 H 3 C N (6-ethanesulfonylamino- 1 H H / benzoimidazol-2-yl)-amide o, -- Isoquinoline-3-carboxylic acid 499 [6-(biphenyl-4-sulfonylamino) IH-benzoimidazol-2-yl)-amide NN Oo N Isoquinoline-3-carboxylic acid 500 H-N H [6-(propane-1 -sulfonylamino) H H 1H-benzoimidazol-2-yl]-amide HN Isoquinoline-3-carboxylic acid 501 / 6propane-2-sulfonylamino)
CH
3 0 H-benzoimidazol-2-yl]-amide 115 WO 2006/099379 PCT/US2006/009049 Incomplete valences for heteroatoms such as oxygen and nitrogen in the chemical structures listed in Table 1 are assumed to be completed by hydrogen. Compounds in Table 1 having a basic group or acidic group are depicted and named 5 as the free base or acid. Depending on the reaction conditions and purification conditions, various compounds in Table 1 having a basic group were isolated in either the free base form, as a salt (such as HCI salt), or in both forms. In another aspect, the present invention comprises a pharmaceutical composition comprising the compound of Fonnula (I) and a phannaceutically acceptable carrier, excipient, 10 diluent, or a mixture thereof. As used herein, the term "lower" refers to a group having between one and six carbons. As used herein, the term "alkyl" refers to a straight or branched chain hydrocarbon having from one to ten carbon atoms, optionally substituted with substituents selected from 15 the group consisting of halo, lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, oxo, hydroxy, mercapto, amino optionally substituted by alkyl, carboxy, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally substituted by alkyl, silyloxy optionally substituted by alkoxy, alkyl, or aryl, silyl optionally substituted by alkoxy, alkyl, or aryl, nitro, cyano, halogen, or lower perfluoroalkyl, multiple 20 degrees of substitution being allowed. Such an "alkyl" group may containing one or more 0, S, S(O), or S(0)2 atoms. Examples of "alkyl" as used herein include, but are not limited to, methyl, n-butyl, t-butyl, n-pentyl, isobutyl, and isopropyl, and the like. As used herein, the term "alkylene" refers to a straight or branched chain divalent hydrocarbon radical having from one to ten carbon atoms, optionally substituted with 25 substituents selected from the group consisting of halo, lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, oxo, hydroxy, mercapto, amino optionally substituted by alkyl, carboxy, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally substituted by alkyl, silyloxy optionally substituted by alkoxy, alkyl, or aryl, silyl optionally substituted by alkoxy, alkyl, or aryl, nitro, cyano, halogen, or lower 30 perfluoroalkyl, multiple degrees of substitution being allowed. Such an "alkylene" group may containing one or more 0, S, S(O), or S(0)2 atoms. Examples of "alkylene" as used herein include, but are not limited to, methylene, ethylene, and the like. As used herein, the term "alkenyl" refers to a hydrocarbon radical having from two to ten carbons and at least one carbon - carbon double bond, optionally substituted with 116 WO 2006/099379 PCT[US2006/009049 substituents selected from the group consisting of lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, oxo, hydroxy, mercapto, amino optionally substituted by alkyl, carboxy, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally substituted by alkyl, silyloxy optionally substituted by alkoxy, alkyl, 5 or aryl, silyl optionally substituted by alkoxy, alkyl, or aryl, nitro, cyano, halogen, or lower perfluoroalkyl, multiple degrees of substitution being allowed. Such an "alkenyl" group may containing one or more 0, S, S(O), or S(0) 2 atoms. As used herein, the term "alkenylene" refers to a straight or branched chain divalent hydrocarbon radical having from two to ten carbon atoms and one or more carbon - carbon 10 double bonds, optionally substituted with substituents selected from the group consisting of halo, lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, oxo, hydroxy, mercapto, amino optionally substituted by alkyl, carboxy, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally substituted by alkyl, silyloxy optionally substituted by alkoxy, alkyl, or aryl, silyl optionally substituted by alkoxy, alkyl, 15 or aryl, nitro, cyano, halogen, or lower perfluoroalkyl, multiple degrees of substitution being allowed. Such an "alkenylene" group may containing one or more 0, S, S(O), or S(0)2 atoms. Examples of "alkenylene" as used herein include, but are not limited to, ethene-1,2 diy1, propene-1 ,3-diyl, methylene-1, 1-diyl, and the like. As used herein, the tenn "alkynyl" refers to a hydrocarbon radical having from two to 20 ten carbons and at least one carbon - carbon triple bond, optionally substituted with substituents selected from the group consisting of halo, lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, oxo, hydroxy, mercapto, amino optionally substituted by alkyl, carboxy, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally substituted by alkyl, silyloxy optionally substituted by alkoxy, alkyl, 25 or aryl, silyl optionally substituted by alkoxy, alkyl, or aryl, nitro, cyano, halogen, or lower perfluoroalkyl, multiple degrees of substitution being allowed. Such an "alkynyl" group may containing one or more 0, S, S(O), or S(O)2 atoms. As used herein, the term "alkynylene" refers to a straight or branched chain divalent hydrocarbon radical having from two to ten carbon atoms and one or more carbon - carbon 30 triple bonds, optionally substituted with substituents selected from the group consisting of halo, lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, oxo, hydroxy, mercapto, amino optionally substituted by alkyl, carboxy, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally substituted by alkyl, silyloxy optionally substituted by alkoxy, alkyl, or aryl, silyl optionally substituted by alkoxy, alkyl, 117 WO 2006/099379 PCT/US20061009049 or aryl, nitro, cyano, halogen, or lower perfluoroalkyl, multiple degrees of substitution being allowed. Such an "alkynylene" group may containing one or more 0, S, S(O), or S(0)2 atoms. Examples of "alkynylene" as used herein include, but are not limited to, ethyne-1,2 diyl, propyne-1,3-diyl, and the like. 5 As used herein, the terms "haloaliphatic", "haloalkyl", "haloalkenyl" and "baloalkoxy" refer to an aliphatic, alkyl, alkenyl or alkoxy group, as the case may be, substituted with one or more halogen atoms. As used herein, "cycloalkyl" refers to a alicyclic hydrocarbon group optionally possessing one or more degrees of unsaturation, having from three to twelve carbon atoms, 10 optionally substituted with substituents selected from the group consisting of halo, lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, oxo, hydroxy, mercapto, amino optionally substituted by alkyl, carboxy, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally substituted by alkyl, nitro, cyano, halogen, or lower perfluoroalkyl, multiple degrees of substitution being allowed. "Cycloalkyl" includes 15 by way of example cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl, and bicyclic and tricyclic structures such as adamanatane. As used herein, the term "cycloalkylene" refers to an non-aromatic alicyclic divalent hydrocarbon radical having from three to twelve carbon atoms and optionally possessing one or more degrees of unsaturation, optionally substituted with substituents selected from the 20 group consisting of halo, lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, oxo, hydroxy, mercapto, amino optionally substituted by alkyl, carboxy, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally substituted by alkyl, nitro, cyano, halogen, or lower perfluoroalkyl, multiple degrees of substitution being allowed. Examples of "cycloalkylene" as used herein include, but are not limited to, cyclopropyl-1,1 25 diyl, cyclopropyl- 1,2-diyl, cyclobutyl- 1,2-diyl, cyclopentyl- 1,3 -diyl, cyclohexyl- 1,4-diyl, cycloheptyl-1,4-diyl, or cyclooctyl-1,5-diyl, and bicyclic and tricyclic structures. As used herein, the term "heterocyclic" or the term "heterocyclyl" refers to a non aromatic three to twelve-membered heterocyclic ring optionally possessing one or more degrees of unsaturation, containing one or more heteroatomic substitutions selected from S, 30 SO, SO 2 , 0, or N, optionally substituted with substituents selected from the group consisting of halo, lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, oxo, hydroxy, mercapto, amino optionally substituted by alkyl, carboxy, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally substituted by alkyl, nitro, cyano, halogen, or lower perfluoroalkyl, multiple degrees of substitution being allowed. 118 WO 2006/099379 PCT/US2006/009049 Such a ring may be optionally fused to one to three benzene rings or to one to three of another "heterocyclic" ring(s) or cycloalkyl ring(s). Examples of "heterocyclic" include, but are not limited to, tetrahydrofuran, 1,4-dioxane, 1,3-dioxane, piperidine, pyrrolidine, morpholine, piperazine, and the like. 5 As used herein, the term "heterocyclylene" refers to a three to twelve-membered heterocyclic ring diradical optionally having one or more degrees of unsaturation containing one or more heteroatoms selected from S, SO, S02, 0, or N, optionally substituted with substituents selected from the group consisting of halo, lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, oxo, hydroxy, mercapto, amino 10 optionally substituted by alkyl, carboxy, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally substituted by alkyl, nitro, cyano, halogen, or lower perfluoroalkyl, multiple degrees of substitution being allowed. Such a ring may be optionally fused to one to three benzene rings or to one to three of another "heterocyclic" rings or cycloalkyl rings. Examples of "heterocyclylene" include, but are not limited to, tetrahydrofuran-2,5-diyl, 15 morpholine-2,3-diyl, pyran-2,4-diyl, 1,4-dioxane-2,3-diyl, 1,3-dioxane-2,4-diyl, piperidine 2,4-diyl, piperidine- 1,4-diyl, pyrrolidine- 1,3 -diyl, morpholine-2,4-diyl, piperazine- 1,4-dyil, and the like. As used herein, the term "aryl" refers to a benzene ring or to an optionally substituted benzene ring system fused to one to three optionally substituted benzene rings, optionally 20 substituted with substituents selected from the group consisting of halo, lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, di(lower alkyl)aminoalkyl, aminoalkyl, oxo, hydroxy, mercapto, amino optionally substituted by alkyl, carhoxy, tetrazolyl, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally substituted by alkyl, acyl, aroyl, heteroaroyl, acylamino, acyloxy, aroyloxy, heteroaroyloxy, 25 alkoxycarbonyl, silyloxy optionally substituted by alkoxy, alkyl, or aryl, silyl optionally substituted by alkoxy, alkyl, or aryl, nitro, cyano, halogen, or lower perfluoroalkyl, multiple degrees of substitution being allowed. Examples of aryl include, but are not limited to, phenyl, 2-naphthyl, 1 -naplithyl, I -anthracenyl, and the like. As used herein, the term "arylene" refers to a benzene ring diradical or to a benzene 30 ring system diradical fused to one to three optionally substituted benzene rings, optionally substituted with substituents selected from the group consisting of halo, lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, di(lower alkyl)aminoalkyl, aminoalkyl, oxo, hydroxy, mercapto, amino optionally substituted by alkyl, carboxy, tetrazolyl, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally 119 WO 2006/099379 PCT/US2006/009049 substituted by alkyl, acyl, aroyl, heteroaroyl, acylamino, acyloxy, aroyloxy, heteroaroyloxy, alkoxycarbonyl, silyloxy optionally substituted by alkoxy, alkyl, or aryl, silyl optionally substituted by alkoxy, alkyl, or aryl, nitro, cyano, halogen, or lower perfluoroalkyl, multiple degrees of substitution being allowed. Examples of "arylene" include, but are not limited to, 5 benzene-1,4-diyl, naphthalene-1,8-diyl, and the like. As used herein, the term "heteroaryl" refers to a five - to seven - membered aromatic ring, or to a polycyclic heterocyclic aromatic ring (up to 3 rings), containing one or more nitrogen, oxygen, or sulfur heteroatoms, where N-oxides and sulfur monoxides and sulfur dioxides are permissible heteroaromatic substitutions, optionally substituted with 10 substituents selected from the group consisting of halo, lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, oxo, hydroxy, mercapto, amino optionally substituted by alkyl, carboxy, tetrazolyl, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally substituted by alkyl, acyl, aroyl, heteroaroyl, acyloxy, aroyloxy, heteroaroyloxy, alkoxycarbonyl, silyloxy optionally substituted by alkoxy, alkyl, or aryl, silyl 15 optionally substituted by alkoxy, alkyl, or aryl, nitro, cyano, halogen, or lower perfluoroalkyl, multiple degrees of substitution being allowed. For polycyclic aromatic ring systems, one or more of the rings may contain one or more heteroatoms. Examples of "heteroaryl" used herein are furan, thiophene, pyrrole, imidazole, pyrazole, triazole, tetrazole, thiazole, oxazole, isoxazole, oxadiazole, thiadiazole, isothiazole, pyridine, pyridazine, pyrazine, pyrimidine, 20 quinoline, isoquinoline, quinazoline, benzofuran, benzothiophene, indole, and indazole, and the like. As used herein, the term "heteroarylene" refers to a five - to seven - membered aromatic ring diradical, or to a polycyclic heterocyclic aromatic ring diradical (up to 3 rings), containing one or more nitrogen, oxygen, or sulfur heteroatoms, where N-oxides and sulfur 25 monoxides and sulfur dioxides are pennissible heteroaromatic substitutions, optionally substituted with substituents selected from the group consisting of halo, lower alkyl, lower alkoxy, lower alkylsulfanyl, lower alkylsulfenyl, lower alkylsulfonyl, oxo, hydroxy, mercapto, amino optionally substituted by alkyl, carboxy, tetrazolyl, carbamoyl optionally substituted by alkyl, aminosulfonyl optionally substituted by alkyl, acyl, aroyl, heteroaroyl, acyloxy, 30 aroyloxy, heteroaroyloxy, alkoxycarbonyl, silyloxy optionally substituted by alkoxy, alkyl, or aryl, silyl optionally substituted by alkoxy, alkyl, or aryl, nitro, cyano, halogen, or lower perfluoroalkyl, multiple degrees of substitution being allowed. For polycyclic aromatic ring system diradicals, one or more of the rings may contain one or more heteroatoms. Examples of "heteroarylene" used herein are furan-2,5-diyl, thiophene-2,4-diyl, 1,3,4-oxadiazole-2,5 120 WO 2006/099379 PCT/US2006/009049 diyl, 1,3,4-thiadiazole-2,5-diyl, 1,3-thiazole-2,4-diyl, 1,3-thiazole-2,5-diyl, pyridine-2,4-diyl, pyridine-2,3-diyl, pyridine-2,5-diyl, pyrimidine-2,4-diyt, quinoline-2,3-diyl, and the like. As used herein, the term "fused cycloalkylaryl" refers to a cycloalkyl group fused to an aryl group, the two having two atoms in common, and wherein the aryl group is the point 5 of substitution. Examples of "fused cycloalkylaryl" used herein include 5-indanyl, 5,6,7,8 tetrahydro-2-naphthyl, and the like. As used herein, the term "fused cycloalkylarylene" refers to a fused cycloalkylaryl, wherein the aryl group is divalent. Examples include 10 ,and the like. As used herein, the term "fused arylcycloalkyl" refers to an aryl group fused to a cycloalkyl group, the two having two atoms in common, and wherein the cycloalkyl group is the point of substitution. Examples of "fused arylcycloalkyl" used herein include 1 -indanyl, 2-indanyl, 1-(1,2,3,4-tetrahydronaphthyl), 15 , and the like. As used herein, the term "fused arylcycloalkylene" refers to a fused arylcycloalkyl, wherein the cycloalkyl group is divalent. Examples include and the like. As used herein, the term "fused heterocyclylaryl" refers to a heterocyclyl group fused 20 to an aryl group, the two having two atoms in common, and wherein the aryl group is the point of substitution. Examples of "fused heterocyclylaryl" used herein include 3,4 121 WO 2006/099379 PCT/US2006/009049 methylenedioxy-1 -phenyl, N H , and the like As used herein, the term "fused heterocyclylarylene" refers to a fused heterocyclylaryl, wherein the aryl group is divalent. Examples include N 5 H and the like. As used herein, the term "fused aryiheterocyclyl" refers to an aryl group fused to a heterocyclyl group, the two having two atoms in common, and wherein the heterocyclyl group is the point of substitution. Examples of "fused arylbeterocyclyl" used herein include 2-(1,3-benzodioxolyl), N to H ,and the like. As used herein, the tenn "fused arylheterocyclylene" refers to a fused arylheterocyclyl, wherein the heterocyclyl group is divalent. Examples include N H , and the like, As used herein, the term "fused cycloalkylheteroaryl" refers to a cycloalkyl group 15 fused to a heteroaryl group, the two having two atoms in common, and wherein the heteroaryl group is the point of substitution. Examples of "fused cycloalkylheteroaryl" used herein include 5-aza-6-indanyl, N ,and the like. 122 WO 2006/099379 PCT/US2006/009049 As used herein, the term "fused cycloalkylheteroarylen&' refers to a fused cycloalkylheteroaryl, wherein the heteroaryl group is divalent. Examples include , and the like. As used herein, the term "fused heteroarylcycloalkyl" refers to a heteroaryl group 5 fused to a cycloalkyl group, the two having two atoms in common, and wherein the cycloalkyl group is the point of substitution. Examples of "fused heteroarylcycloalkyl" used herein include 5-aza- 1 -indanyl, N and the like. As used herein, the term "fused heteroarylcycloalkylene" refers to a fused 10 heteroarylcycloalkyl, wherein the cycloalkyl group is divalent. Examples include , and the like. As used herein, the term "fused heterocyclylheteroaryl" refers to a heterocyclyl group fused to a heteroaryl group, the two having two atoms in conon, and wherein the heteroaryl group is the point of substitution. Examples of "fused heterocyclylheteroaryl" used herein 15 include 1,2,3,4-tetrahydro-beta-carbolin-8-yl, H N N and the like. As used herein, the term "fused heterocyclylheteroarylene" refers to a fused heterocyclylheteroaryl, wherein the heteroaryl group is divalent. Examples include H N FIN N , and the like. 20 As used herein, the term "fused heteroarylheterocyclyl" refers to a heteroaryl group fused to a heterocyclyl group, the two having two atoms in common, and wherein the 123 WO 2006/099379 PCT/US2006/009049 heterocyclyl group is the point of substitution. Examples of "fused heteroarylheterocyclyl" used herein include -5-aza-2,3 -dihydrobenzofiran-2-yl, NN H , and the like, As used herein, the term "fused heteroarylheterocyclylene" refers to a fused 5 heteroarylheterocyclyl, wherein the heterocyclyl group is divalent. Examples include N H ,and the like. As used herein, the term "direct bond", where part of a structural variable specification, refers to the direct joining of the substituents flanking (preceding and succeeding) the variable taken as a "direct bond". Where two or more consecutive variables 10 are specified each as a "direct bond", those substituents flanking (preceding and succeeding) those two or more consecutive specified "direct bonds" are directly joined. As used herein, the tern "alkoxy" refers to the group RaO-, where Ra is alkyl. As used herein, the term "alkenyloxy" refers to the group RaO-, where P 4 is alkenyl. As used herein, the term "alkynyloxy" refers to the group RaO-, where R, is alkynyl 15 As used herein, the term "alkylsulfanyl" refers to the group RaS-, where Ra is alkyl. As used herein, the term "alkenylsulfanyl" refers to the group RaS-, where Ra is alkenyl. As used herein, the term "alkynylsulfanyt" refers to the group RaS-, where R" is alkynyL 20 As used herein, the term "alkylsulfenyl" refers to the group RaS(O)-, where Ra is alkyl. As used herein, the tern "alkenylsulfenyl" refers to the group RaS(O)-, where Ra is alkenyl. As used herein, the term "alkynylsulfenyl" refers to the group RaS(O)-, where Ra is alkynyl. 25 As used herein, the term "alkylsulfonyl" refers to the group RaSO2-, where Ra is alkyl. As used herein, the term "alkenylsulfonyl" refers to the group RaSO2, where Ra is alkenyl. 124 WO 2006/099379 PCT/US2006/009049 As used herein, the term "alkynylsulfonyl" refers to the group RaSO2, where Ra is alkynyl. As used herein, the term "acyl" refers to the group RaC(O)- , where Ra is alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, or heterocyclyl. 5 As used herein, the tent "aroyl" refers to the group R 11 C(O)- , where Ra is aryl. As used herein, the term "heteroaroyl" refers to the group RaC(O)-, where Ra is heteroaryl. As used herein, the term "alkoxycarbonyl" refers to the group RaOC(O)-, where Ra is alkyl. 10 As used herein, the term "acyloxy" refers to the group R 2 C(O)O- , where Ra is alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, or heterocyclyl. As used herein, the term "aroyloxy" refers to the group RaC(O)O- , where Ra is aryl As used herein, the term "heteroaroyloxy" refers to the group RaC(O)O- , where Ra is heteroaryl. 15 As used herein, the term "optionally" means that the subsequently described event(s) may or may not occur, and includes both event(s) which occur and events that do not occur. As used herein, the term "substituted" refers to substitution with the named substituent or substituents, multiple degrees of substitution being allowed unless otherwise stated, provided that the substitution results in a stable or chemically feasible compound. A 20 stable compound or chemically feasible compound is one in which the chemical structure is not substantially altered when kept at a temperature from about -80" C to about +40* C, in the absence of moisture or other chemically reactive conditions, for at least a week, or a compound which maintains its integrity long enough to be useful for therapeutic or prophylactic administration to a patient. The phrase "one or more substituents", as used 25 herein, refers to a number of substituents that equals from one to the maximum number of substituents possible based on the number of available bonding sites, provided that the above conditions of stability and chemical feasibility are met. As used herein, the terms "contain" or "containing" can refer to in-line substitutions at any position along the above defined alkyl, alkenyl, alkynyl or cycloalkyl substituents with 30 one or more of any of 0, S, SO, SO 2 , N, or N-alkyl, including, for example, -CH 2
-O-CH
2 ,-CH 2
-SO
2
-CH
2 -, -CH-NH-CH3 and so forth. As used herein, the term solvatee" is a complex of variable stoichiometry formed by a solute (in this invention, a compound of Formula (I)) and a solvent. Such solvents for the 125 WO 2006/099379 PCT/US2006/009049 purpose of the invention may not substantially interfere with the biological activity of the solute. Solvents may be, by way of example, water, ethanol, or acetic acid. As used herein, the term "biohydrolyzable ester" is an ester of a drug substance (in this invention, a compound of Formula (I)) which either a) does not interfere with the 5 biological activity of the parent substance but confers on that substance advantageous properties in vivo such as duration of action, onset of action, and the like, or b) is biologically inactive but is readily converted in vivo by the subject to the biologically active principle. The advantage is that, for example, the biohydrolyzable ester is orally absorbed from the gut and is transformed to Formula (I) in plasma. Many examples of such are known in the art 10 and include by way of example lower alkyl esters (e.g., C-C 4 ), lower acyloxyalkyl esters, lower alkoxyacyloxyalkyl esters, alkoxyacyloxy esters, alkyl acylamino alkyl esters, and choline esters. As used herein, the term "biohydrolyzable amide" is an amide of a drug substance (in this invention, a compound of general formula (I)) which either a) does not interfere with the 15 biological activity of the parent substance but confers on that substance advantageous properties in vivo such as duration of action, onset of action, and the like, or b) is biologically inactive but is readily converted in vivo by the subject to the biologically active principle. The advantage is that, for example, the biohydrolyzable amide is orally absorbed from the gut and is transformed to Formula (I) in plasma. Many examples of such are known in the art and 20 include by way of example lower alkyl amides, .alpha-amino acid amides, alkoxyacyl amides, and alkylaminoalkylcarbonyl amides. As used herein, the tenn "prodrug" is a) a biohydrolyzable amide or a biohydrolyzable ester and encompasses compounds in which the biohydrolyzable functionality in such a prodrug is encompassed in the compound of Formula (I) or b) a compound that may be 25 oxidized or reduced biologically at a given functional group to yield drug substance of Formula (I). Examples of these functional groups include, but are not limited to, 1,4 dihydropyridine, N-alkylcarbonyl-1,4-dihydropyridinc, 1,4-cyclohexadiene, tert-butyl, and the like. Whenever the terms "alkyl" or "aryl" or either of their prefix roots appear in a name of 30 a substituent (e.g. arylalkoxyaryloxy) they shall be interpreted as including those limitations given above for "alkyl" and "aryl". Designated numbers of carbon atoms (e.g. C- 1 ]) shall refer independently to the number of carbon atoms in an alkyl, alkenyl or alkynyl or cyclic 126 WO 2006/099379 PCT/US2006/009049 alkyl moiety or to the alkyl portion of a larger substituent in which the term "alkyl" appears as its prefix root. As used herein, the term "oxo" shall refer to the substituent =0. As used herein, the term "halogen" or "halo" shall include iodine, bromine, chlorine 5 and fluorine. As used herein, the term "mercapto" shall refer to the substituent -SH. As used herein, the term "carboxy" shall refer to the substituent -COOH. As used herein, the term "cyano" shalt refer to the substituent -CN. As used herein, the term "aminosulforyl" shall refer to the substituent -SO 2
NH
2 . 10 As used herein, the term "carbamoyl" shall refer to the substituent -C(O)NH 2 . As used herein, the term "sulfanyl" shall refer to the substituent -S-. As used herein, the term "sulfenyl" shall refer to the substituent -S(O)-. As used herein, the term "sulfonyl" shall refer to the substituent -S(0)2-. The compounds can be prepared according to the following reaction Schemes (in 15 which variables are as defined before or are defined) using readily available starting materials, reagents and conventional synthesis procedures. In these reactions, it is also possible to make use of variants which are themselves known to those of ordinary skill in this art, but are not mentioned in greater detail. The present invention also provides a method for the synthesis of compounds useful 20 as intermediates in the preparation of compounds of Formula (I) along with methods for the preparation of compounds of Formula (I). The general procedures used in the methods to prepare the compounds of Formula (I) of the present invention are described below where the definitions of the variable groups are the same as those described for the compound of Fonnula (I). 25 As shown in Scheme I, diamino methyl benzoate (1) is treated with a carboxylic acid in the presence of a coupling reagent such as, but not limited to, HBTU to form amide (2). Amide (2) is then refluxed in a solvent such as, but not limited to, AcOH to form benzimidazole (3). The methyl ester of benzimidazole (3) is hydrolyzed using a base such as, but not limited to, LiOH to provide the free carboxylic acid, which is then coupled with an 30 amine in the presence of a coupling reagent such as, but not limited to, JBTU to form aide (4). 127 WO 2006/099379 PCT/US2006/009049 Scheme I 0 0 0 NH 2 G,-COOH 0 NH 2 AcOH K
-
-I 0
NH
2 a NK (2) H Gi G G O NH O O N 1.Hydrolysis N N GG, N 2. G 3
-G
2
-NH
2 N H H (3) (4) 5 As shown in Scheme II, the methyl ester of benzimidazole (3) is hydrolyzed using a base such as, but not limited to, LiOH to provide the free carboxylic acid, which is then coupled with compound (5) in the presence of a coupling reagent such as, but not limited to HBTU, to form the amide (6). Compound (5) is prepared by first coupling an amino protected amino acid, such as a BOC protected amino acid, with an amine and then removing 10 the amino protecting group to provide compound (5). 128 WO 2006/099379 PCT/US2006/009049 Scheme II 0 G N H n O NH 0 0 > N 1. Hydrolysis N \>-Ar IAr - N N H 2. Couple with compound (5) H (3) (6) n=0or1 L OH 10 3
-NH
2 R N H HN 2. 2 M HCI in Dioxane H 2 N boo (5) As shown in Scheme IIt diamino methyl benzoate (1) is reacted with isothiocyanate 5 (7) by refluxing in a solvent such as, but not limited to, THF to provide intermediate (8). Isothiocyanate (7) is either commercially available or is prepared from a corresponding amine by reacting with 1,1'-thiocarbonyldiimidazole in a solvent such as, but not limited to, THF. The intermediate (8) is treated with coupling reagent such as, but not limited to, DCC to form benzimidazole (9). The methyl ester of benzimidazole (9) is hydrolyzed using a base such as, 10 but not limited to, LiOH to provide the free carboxylic acid, which is then coupled with an amine in the presence of a coupling reagent such as, but not limited to, HBTU to form the amide (10). 129 WO 2006/099379 PCT/US2006/009049 Scheme III 0 0 O0N Gr-N=C=S 0
NH
2 (7)
NH
2 DCC I>H
NH
2 N (1)(8 HN I o NH 0- 0 N 1.Hydrolysis N N \> N-G N-C N N H N 2. Gr-G2-NH2H (9) (10) As shown in Scheme IV, diamino methyl benzoate (1) is treated with cyanogen 5 bromide in a mixture of solvents such as, but not limited to, EtOH:H 2 0 to forn 2-amino benzimdazole (11). The amine of benzimidazole (11) is coupled with a carboxylic acid in the presence of a coupling reagent such as, but not limited to, HBTU to form the amide (12). The methyl ester of amide (12) is hydrolyzed using a base such as, but not limited to, LiOH to provide the free carboxylic acid, which was then coupled with an amine in the presence of 10 a coupling reagent such as, but not limited to, HTBTU to form the amide (13). 130 WO 2006/099379 PCT/US2006/009049 Scheme IV O 0 0 NH 2 CNBr O N G<COOH
NH
2
NH
2 N H (1) (1 O NH O 0 O N 0 1.Hydrolysis N N N SG1 2. G 3
-G
2
-NH
2 H (12) (13) Scheme V shows an alternate general method to prepare amide (13). As shown in 5 Scheme V, nitro-amino benzoic acid (14) is coupled with an amine in the presence of a coupling reagent such as, but not limited to, HBTU to form the aide, which is then reduced under conditions such as, but not limited to, Pd/C under hydrogen atmosphere to provide diamine (15). The resulting diamine (15) is reacted with cyanogen bromide in a mixture of solvents such as, but not limited to, EtOH: H20 to form 2-amino benzimdazole (16). The 10 amino group of benzimidazole (16) is coupled with a carboxylic acid in the presence of a coupling reagent such as, but not limited to, HBTU to provide aide (13). 131 WO 2006/099379 PCT/US2006/009049 Scheme V OH NG2-G NH O
NH
2 1. G-G-NH 2 O N NH 2 CNBr
NO
2 2. H2/Pd/C in MeOH
NH
2 (14) (15) G -G IG 2-G 312 3 HN NH 'N N G -COO H- > N O NH2 ~G OO N GI N 2 N H H H (16) (13) As shown in Scheme VI, compound (I1a) is treated with a nitrating reagent such as, but not limited to, KNO 3
/H
2
SO
4 to provide benzimidazole (17). The amine of benzimidazole 5 (17) is coupled with a carboxylic acid in the presence of a coupling reagent such as, but not limited to, HBTU to form the aide (18). The methyl ester of amide (18) is hydrolyzed using a base such as, but not limited to, LiOH to provide the free carboxylic acid, which is then coupled with an amine in the presence of a coupling reagent such as, but not limited to, HBTU to forn the amide (19). The nitro group of amide (19) is reduced under conditions 10 such as, but not limited to, Pd/C under hydrogen atmosphere and the resulting aide is then reacted with an acid chloride in the presence of a base such as, but not limited to, pyridine to provide compound (20). 132 WO 2006/099379 PCT/US2006/009049 Scheme VI O O A 0 0 0 N KNO, H 2 S0 4 N G-COOH 1 INH- 2 _ I NH 2 N 0N N H- 2H (11a) (17) 0 0 0 N -G 2 G3 H N 0 1. Hydrolysis N 0 N N H G 2. W-NH 2 HIGN H H (18) (19) G -G 1,2 3 NH 0 1. Reduction -N G_ G,\ NJ 2. G -%COC I N 7 N H G 2.O0 H H (20) As shown in Scheme VII, compound (21) is reacted with cyanogen bromide in a 5 mixture of solvents such as, but not limited to, EtOH-H 2 0 to fonn 2-amino benzimidazole (22). The amino group of benzimidazole (22) is coupled with a carboxylic acid in the presence of a coupling reagent such as, but not limited to, HBTU to fonn an amide, and the nitro group is then reduced under conditions such as, but not limited to, Pd/C under hydrogen atmosphere to provide amine (23). The amine (23) is then reacted with a sulfonyl chloride in 10 the presence of a base such as, but not limited to, pyridine to provide compound (24). In Scheme VII, the variable R is a group such as but not limited to aryl, arylalkylene, heteroarylalkylene, or heteroaryl, or their optionally substituted forms. 133 WO 2006/099379 PCT/US2006[009049 Scheme VII O2 . NH 2 02 N G 1 -COOH CNBr \ NH 2 N N H Pd-C in MeOH NH 2 H 2 (21) (22) O e'S-R HN o H 2 N N 0 O N H N H Gi R-SO 2 CI H H (24) (23) As shown in Scheme VII, amine (23) may also be treated with a coupling reagent 5 such as CDI and then reacted with an alkyl or aryl amine to give urea (25). In Scheme VIII, the variable R is a group such as but not limited to aryl, arylalkylene, heteroarylalkylene, or heteroaryl, or their optionally substituted forms. Scheme VIII R O 1 NH H-/ H-N 0 N G CDI N 1>-N K GI R-NH 2 N H G 1 H 10 (23) (25) As shown in Scheme IX, compound (26) is reacted with bromine in solvent such as, but not limited to, AcOH to provide aryl bromide (27). The aryl bromide (27) is then coupled 15 to a boronic acid in the presence of palladium reagent such as, but not limited to, palladium tetraldstriphenylphosphine to form aryl compound (28) that is then subjected to the sequence of reactions as shown in Scheme V to provide benzimidazole (29). The aryl bromide (27) may also be coupled with aryl and alkenyl tin reagents to provide aryl compounds that are 134 WO 2006/099379 PCT/US2006/009049 then subjected to the sequence of reactions as shown in Scheme V to provide benzimidazole (29). In Scheme IX, the variable R is a group such as but not limited to aryl, heteroaryl, arylalkenylene, alkenyl, heteroarylalkenylene, or their optionally substituted forms. 5 Scheme IX 0 00 0 OH 0 0
NH
2 Br 2
NH
2 R OH N NH 2
NO
2 Br NO 2 R NO 2 (26) (27) (28) 62-G, 0 NH N N G R N H H (29) As shown in Scheme X, compound (30) is treated with a nitrating reagent such as, but not limited to, 1NO 3
/H
2
SO
4 to provide nitro compound (31). The nitro compound (31) is 10 treated with ammonium carbonate in a solvent such as DMF to provide an amino nitro compound (32), which is then treated with a sodium alkoxide or sodium aryloxide to provide aryl ether (33). The aryl ether (33) is then subjected to a sequence of reactions as shown in Scheme V to provide benzimidazole (34). In Scheme X, the variable R is a group such as but not limited to aryl, heteroaryl, arylalkylene, alkyl, heteroarylalkylene, or their optionally 15 substituted forms. 135 WO 2006/099379 PCT/US2006/009049 Scheme X O 0 O 0 0O. HNO, H 2 S0 4 NO 2
NH
4 CO3 - NO 2 F F F F F
NH
2 (30) (31) (32) G -G, S0 N NaOR N NO 2 N 0 I I " N R' 0 NH 2 O N H R H (33) (34) 5 As shown in Scheme XI, compound (32) is reacted with a sodium thiolate in the presence or absence of a base such as triethylamine to provide compound (35) which is then subjected to a sequence of reactions as shown in Scheme V to provide the benzimidazole (36). Benzimidazole (36) may then be treated with an oxidizing agent such as a peroxide including, but not limited to, MCPBA to provide sulfone (37). In Scheme XI, the variable R is a group 10 such as but not limited to aryl, heteroaryl, arylalkylene, alkyl, heteroarylalkylene, or their optionally substituted forms. 136 WO 2006/099379 PCT/US20061009049 Scheme XI o a U NO2 NaSR
NO
2 F NH 2 R NH 2 (32) (35) G-G G 0 NH O NH N MCPBA oN G N G H N H N ' H (36) (37) 5 As shown in Scheme XII, acid inputs used in above schemes may be prepared from bromo carboxylic acids (38) by reacting with an alkenyl or aryl boronic acid in the presence of a palladium reagent such as, but not limited to, palladium tetrakistriphenylphosphine to form aryl compound (39). fn Scheme XII, the variable R is a group such as but not limited to aryl, heteroaryl, arylalkenylene, alkenyl, heteroarylalkenylene, or their optionally substituted 10 forms. The variable X in Scheme XII may be a group such as but not limited to N or CH, or their optionally substituted forms. Scheme XII Br O OH R 0 OH R OH OH (38) (39) 15 137 WO 2006/099379 PCT[US2006/009049 As shown in Scheme XIII, acid inputs used in the above schemes may also be prepared from the bromo acid (40), where X is a group such as but not limited to CH or N, by reacting with an acetylene in the presence of a palladium reagent such as, but not limited to, dichlorobis(triphenylphosphine)-palladium (II) to afford acetylene (41). In Scheme XIII, the 5 variable R is a group such as but not limited to aryl, heteroaryl, alkyl, cycloalkyl, or their optionally substituted forms. Scheme XIII R Br O O OH R OH X pX (40) (41) 10 The tenn "amino-protecting group" as used herein refers to substituents of the amino group commonly employed to block or protect the amino functionality while reacting other functional groups on the compound. Examples of such amino-protecting groups include the formyl group, the trityl group, the phthalimido group, the trichloroacetyl group, the 15 chloroacetyl, bromoacetyl and iodoacetyl groups, urethane-type blocking groups such as benzyloxycarbonyl, 4-phenylbenzyloxycarbonyl, 2-methylbenzyloxycarbonyl, 4 methoxybenzyloxycarbonyl, 4-fluorobenzyloxycarbonyl, 4-chlorobenzyloxycarbonyl, 3 chlorobenzyloxycarbonyl, 2-chlorobenzyloxycarbonyl, 2,4-dichlorobeuzyloxycarbonyl, 4 bromobenzyloxycarbonyl, 3-bromobenzyloxycarbonyl, 4-nitrobenzyloxycarbonyl, 4 20 cyanobenzyloxy-carbonyl, 2-(4-xenyl)iso-propoxycarbonyl, 1,1-diphenyleth-1-yloxycarbonyl, 1,1-diphenylprop-1-yloxycarbonyl, 2-phenylprop-2-yloxycarbonyl, 2-(p-toluyl)prop-2 yloxycarbonyl, cyclopentanyloxycarbonyl, 1-methylcyclopentanyloxycarbonyl, cyclohexanyloxycarbonyl, 1-methylcyclohexanyloxycarbonyl, 2 niethylcyclohexanyloxycarbonyl, 2-(4-toluylsulfonyl)ethoxycarbonyl, 25 2(methylsulfonyl)ethoxycarbonyl, 2-(triphenylphosphino)ethoxycarbonyl, 9 fluorenylmethoxycarbonyl ("FMOC"), t-butoxycarbonyl ("BOC"), 2 (trimethylsilyl)ethoxycarbonyl, allyloxycarbonyl, 1-(trimethylsilyhnethyl)prop-1 enyloxycarbonyl, 5-benzisoxalylmethoxycarbonyl, 4-acetoxybenzyloxycarbonyl, 2,2,2 138 WO 2006/099379 PCT/US2006/009049 trichloroethoxycarbonyl, 2-ethynyl-2-propoxycarbonyl, cyclopropylmethoxycarbonyl, 4 (decyloxy)benzyloxycarbonyl, isobomyloxycarbonyl, 1 -piperidyloxycarbonyl and the like; the benzoylnethylsulfonyl group, the 2-(nitro)phenylsulfenyl group, the diphenylphosphine oxide group and like amino-protecting groups. The species of amino-protecting group 5 employed is not critical so long as the derivatized anino group is stable to the condition of subsequent reaction(s) on other positions of the compound of Formula (I) and can be removed at the desired point without disrupting the remainder of the molecule. Commonly used amino-protecting groups are the allyloxycarbonyl, the t-butoxycarbonyl, 9 fluorenylnethoxycarbonyl, and the trityl groups. Similar amino-protecting groups used in 10 the cephalosporin, penicillin and peptide art are also embraced by the above terms. The related term "protected amino" or "protected amino group" defines an amino group substituted with an amino-protecting group discussed above. Further examples of progroups referred to by the above terms are described by J. W. Barton, "Protective Groups In Organic Chemistry", J. G. W. McOmie, Ed., Plenum Press, 15 New York, N.Y., 1973, and T. W. Greene, "Protective Groups in Organic Synthesis", John Wiley and Sons, New York, N.Y., 1981. The invention further provides pharmaceutical compositions comprising a compound of Formula (I) or a pharmaceutically acceptable salt, ester, or prodrug thereof. The term "pharmaceutical composition" is used herein to denote a composition that may be 20 administered to a mammalian host, e.g., orally, topically, parenterally, by inhalation spray, or rectally, in unit dosage formulations containing conventional non-toxic carriers, diluents, adjuvants, vehicles and the like. The term "parenteral" as used herein, includes subcutaneous injections, intravenous, intramuscular, intracisternal injection, or by infusion techniques. The term "therapeutically effective amount" is used herein to denote that amount of a 25 drug or pharmaceutical agent that will elicit the therapeutic response of a subject that is being sought. In an embodiment, a therapeutically effective amount is an amount capable of inhibiting the interaction of BACE with its physiological ligands such as, but not limited to, amyloid precursor protein (APP). As used herein, the phrase "a subject" or "a subject in need thereof' includes 30 mammalian subjects, such as humans, but may also encompass other mammals, such as dogs, cats, mice, rats, cattle, horses, sheep, rabbits, monkeys, chimpanzees or other apes or primates, who either suffer from one or more of the aforesaid diseases or disease states or are at risk for such. In an embodiment, a subject is one for which inhibition of p-secretase enzyme activity or treatment of the above noted disorders is desired. Accordingly, in the 139 WO 2006/099379 PCT/US2006/009049 context of the therapeutic methods of this invention, this method also is comprised of a method for treating a mammalian subject prophylactically, or prior to the onset of diagnosis such disease(s) or disease state(s). Factors which may influence what constitutes an effective amount will depend upon the size and weight of the subject, the biodegradability of the 5 therapeutic agent, the activity of the therapeutic agent, as well as its bioavailability. The pharmaceutical compositions containing a compound of the invention may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous, or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixits. Compositions intended for oral use may be prepared according to any known method, 10 and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents, and preserving agents in order to provide phannaceutically elegant and palatable preparations. Tablets may contain the active ingredient in admixture with non-toxic pharmaceutically-acceptable excipients which are suitable for the manufacture of tablets. These excipients may be for example, inert diluents, 15 such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example corn starch or alginic acid; binding agents, for example, starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and 20 thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated by the techniques to form osmotic therapeutic tablets for controlled release. Fonnulations for oral use may also be presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, 25 calcium phosphate or kaolin, or a soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil. Aqueous suspensions may contain the active compounds in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, mnetbylcellulose, hydroxypropylmethylcellulose, 30 sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide such as lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example, heptadecaethyl eneoxycetanol, or condensation products of ethylene oxide with partial esters derived from 140 WO 2006/099379 PCT/US2006/009049 fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more coloring agents, one or more flavoring agents, and one or more sweetening 5 agents, such as sucrose or saccharin. Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as a liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and 10 flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid. Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active compound in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or 15 wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example, sweetening, flavoring, and coloring agents may also be present. The phannaceutical compositions of the invention may also be in the form of oil-in water emulsions. The oily phase may be a vegetable oil, for example, olive oil or arachis oil, 20 or a mineral oil, for example a liquid paraffin, or a mixture thereof Suitable emulsifying agents may be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of said partial esters with ethylene oxide, for example polyoxyethylene 25 sorbitan monooleate. The emulsions may also contain sweetening and flavoring agents. Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such fonnulations may also contain a demulcent, a preservative and flavoring and coloring agents. The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension may be 30 formulated according to the known methods using suitable dispersing or wetting agents and suspending agents described above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In 141 WO 2006/099379 PCT/US2006/009049 addition, sterile, fixed oils are conveniently employed as solvent or suspending medium. For this purpose, any bland fixed oil may be employed using synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. The compositions may also be in the form of suppositories for rectal administration of 5 the compounds of the invention. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will thus melt in the rectum to release the drug. Such materials include cocoa butter and polyethylene glycols, for example. For topical use, creams, ointments, jellies, solutions of suspensions, etc., containing 10 the compounds of the invention are contemplated. For the purpose of this application, topical applications shall include mouth washes and gargles. The compounds of the present invention may also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles. Liposomes may be formed from a variety of phospholipids, such as 15 cholesterol, stearylamine, or phosphatidylcholines. Also provided by the present invention are prodrugs of the invention. Pharmaceutically-acceptable salts of the compounds of the present invention, where a basic or acidic group is present in the structure, are also included within the scope of the invention. The tenn "pharmaceutically acceptable salts" refers to non-toxic salts of the 20 compounds of this invention which are generally prepared by reacting the free base with a suitable organic or inorganic acid or by reacting the acid with a suitable organic or inorganic base. Representative salts include the following salts: Acetate, Benzenesulfonate, Benzoate, Bicarbonate, Bisulfate, Bitartrate, Borate, Bromide, Calcium Edetate, Camsylate, Carbonate, Chloride, Clavulanate, Citrate, Dihydrochloride, Edetate, Edisylate, Estolate, Esylate, 25 Fumarate, Gluceptate, Gluconate, Glutamate, Glycollylarsanilate, Hexylresorcinate, Hydrabamine, Hydrobromide, Hydrochloride, Hydroxynaphthoate, Iodide, Isothionate, Lactate, Lactobionate, Laurate, Malate, Maleate, Mandelate, Methanesulfonate, Methylbromide, Methylnitrate, Methylsulfate, Monopotassium Maleate, Mucate, Napsylate, Nitrate, N-methylglucamine, Oxalate, Painoate (Embonate), Palmitate, Pantothenate, 30 Phosphate/diphosphate, Polygalacturonate, Potassium, Salicylate, Sodium, Stearate, Subacetate, Succinate, Tannate, Tartrate, Teoclate, Tosylate, Triethiodide, Trimethylammonium and Valerate. When an acidic substituent is present, such as-COOH, there can be formed the ammonium, morpholinium, sodium, potassium, barium, calcium salt, and the like, for use as the dosage forn. When a basic group is present, such as amino or a 142 WO 2006/099379 PCT/US2006/009049 basic heteroaryl radical, such as pyridyl, an acidic salt, such as hydrochloride, hydrobromide, phosphate, sulfate, trifluoroacetate, trichloroacetate, acetate, oxlate, maleate, pyruvate, malonate, succinate, citrate, tartrate, fumarate, mandelate, benzoate, cinnamate, methanesulfonate, ethanesulfonate, picrate and the like, and include acids related to the 5 pharmaceutically-acceptable salts listed in the Journal of Pharmaceutical Science, 66, 2 (1977) p. 1-19. In one embodiment, a compound of Formula (I) having one or more basic groups may be used and/or formulated as an HCi salt Other salts which are not pharmaceutically acceptable may be useful in the 10 preparation of compounds of the invention and these form a further aspect of the invention. In addition, some of the compounds of Formula (1) may form solvates with water or common organic solvents. Such solvates are also encompassed within the scope of the invention. Thus, in another aspect of the present invention, there is provided a pharmaceutical 15 composition comprising a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, solvate, or prodrug thereof, and a pharmaceutically acceptable carrier, excipient, diluent, or mixture thereof. In another aspect of the present invention, there is provided a pharmaceutical composition comprising a therapeutically effective amount of a compound of Fonnula (I), or 20 a pharmaceutically acceptable salt, solvate, or prodrug thereof, and a pharmaceutically acceptable carrier, excipient, diluents, or mixture thereof wherein said therapeutically effective amount of the compound of Fonnula (I) preferentially inhibits the interaction of BACE with its physiological ligands relative to the interaction of other secretases, such as a secretase, with its physiological ligands. 25 In another aspect, the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of the compound of Formula (I), and a pharmaceutically acceptable carrier, excipient, diluent, or mixture thereof, and further comprising one or more therapeutic agents. The compounds of Formula (I) may be used in combination with one or more other 30 drugs in the treatment, prevention, control, amelioration, or reduction of risk of diseases or conditions for which the compounds of the present invention have utility. In an embodiment, the combination of the drugs together may be safer or more effective than either drug alone. Additionally, the compounds of Formula (I) may be used in combination with one or more other drugs that treat, prevent, control, ameliorate, or reduce the risk of side effects or toxicity 143 WO 2006/099379 PCT/US2006/009049 of the compounds of the present invention. Such other drugs may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with the compounds of Formula (I). Accordingly, the pharmaceutical compositions of the present invention include those 5 that contain one or more other active ingredients, in addition to the compounds of Formula (I). The combinations may be administered as part of a unit dosage form combination product, or as a kit or treatment protocol wherein one or more additional drugs are administered in separate dosage forms as part of a treatment regimen. Examples of combinations of the compounds of the present invention with other drugs 10 in either unit dose or kit form include combinations with: anti-Alzheimer's agents, other betasecretase inhibitors, gamma-secretase inhibitors, HMG-CoA reductase inhibitors, non steroidal anti-inflanimatory drugs (NSAID's) including ibuprofen, naproxen, diclofenac, N methyl-D-aspartate (NMDA) receptor antagonists, such as memantine, cholinesterase inhibitors such as galantamine, rivastigmine, donepezil, and tacrine, vitamin E, CB-I receptor 15 antagonists or CB- 1 receptor inverse agonists, antibiotics such as doxycycline and rifampin, agents that bind AP or that induce antibodies that bind Ap, anti-Ap antibodies, AQ vaccines, RAGE/RAGE ligand interaction antagonists, and other drugs that affect receptors or enzymes that either increase the efficacy, safety, convenience, or reduce unwanted side effects or toxicity of the compounds of the present invention. 20 The foregoing list of combinations is illustrative only and not intended to be limiting in any way. In another embodiment, the present inventin provides a method comprising: administering to a subject a compound of Formula (I), or a phannaceutically acceptable salt, ester, or prodrug thereof 25 In another embodiment, the present invention provides a method for inhibiting the interaction of BACE with its physiological ligands comprising: administering to a subject a compound of Formula (I), or a pharmaceutically acceptable salt, ester, or prodrug thereof. An example of a physiological ligand of BACE includes, but is not limited to, amyloid precursor protein (APP). 30 In another embodiment, the present invention provides a method for increasing the o secretory pathway in a subject comprising: administering to a subject a compound of Formula (I), or a pharmaceutically acceptable salt, ester, or prodrig thereof. In another embodiment, the present invention provides a method of treating or preventing a BACE mediated disease comprising: administering to a subject a therapeutically 144 WO 2006/099379 PCT/US2006/009049 effective amount of a compound of Formula (1) or a pharmaceutically acceptable salt, ester, or prodrug thereof. In another embodiment, the present invention provides a method for treating a disorder or condition selected from Alzheimer's disease, mild cognitive impairment, Down's 5 syndrome, Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch-Type, cerebral amyloid angiopathy, degenerative dementia, diffuse Lewy body type of Alzheimer's disease or central or preipheral amyloid diseases comprising administering to a subject an amount of a compound of Formula (I), or a pharmaceutically acceptable salt, ester, or prodrug thereof, that is effective in treating such disorder or condition. 10 In another embodiment, the present invention provides a method for treating a disorder or condition method, wherein the disorder or condition being treated is a dementia of the Alzheimer's type and is selected from the group consisting of dementia of the Alzheimer's type with early onset uncomplicated, dementia of the Alzheimer's type with early onset with delusions, dementia of the Alzheimer's type with early onset with depressed mood, dementia 15 of the Alzheimer's type with late onset uncomplicated, dementia of the Alzheimer's type with late onset with delusions and dementia of the Alzheimer's type with late onset with depressed mood, comprising administering to said mammal an amount of a compound of Formula (I), or a pharmaceutically acceptable salt, ester, or prodrug thereof, that is effective in treating such disorder or condition. 20 In another embodiment, the present invention provides a method for treating one or more conditions associated with plaque accumulation comprising administering to a subject an effective amount of a compound of Formula (I), or a phannaceutically acceptable salt, ester, or prodrg thereof. In an embodiment, administering the compound of Formula (I) reduces the rate of neurofibrillary tangle formation in a subject. In another embodiment, 25 administering the compound of Formula (I) reduces the rate of plaque accumulation in a subject. In the methods of the present invention, the compound of Formala (1) may be administered alone or in combination with a therapeutic agent selected from the group consisting of anti-Alzheimer's agents, other betasecretase inhibitors, gamma-secretase 30 inhibitors, HMG-CoA reductase inhibitors, non-steroidal anti-inflammatory drugs (NSAID's) including ibuprofen, naproxen, diclofenac, N-methyl-D-aspartate (NMDA) receptor antagonists, such as memantine, cholinesterase inhibitors such as galantamine, rivastigmine, donepezil, and tacrine, vitamin E, CB-1 receptor antagonists or CB-i receptor inverse agonists, antibiotics such as doxycycline and rifarpin, agents that bind AP or that induce 145 WO 2006/099379 PCT/US2006/0090 49 antibodies that bind Ap, anti-AP antibodies, Ap vaccines, RAGE/RAGE ligand interaction antagonists, and other drugs that affect receptors or enzymes that either increase the efficacy, safety, convenience, or reduce unwanted side effects or toxicity of the compounds of the present invention. 5 The compound of Formula (1) of the present invention, may be administered at a dosage level of from about 0.01 to 1000 mg/kg of the body weight of the subject being treated. In another embodiment, the compound of Formula (I) of the present invention, may be administered at a dosage range between 0.01 and 100 mg/kg. In another embodiment, the compound of Formula (I) of the present invention, may be administered at a dosage range 10 between 0.5 to 10 mg/kg of body weight per day. The amount of active ingredient that may be combined with the carrier materials to produce a single dosage will vary depending upon the host treated and the particular mode of administration. For example, a formulation intended for oral administration to humans may contain 1 ing to 2 grams of a compound of Formula (I) with an appropriate and convenient amount of carrier material which may vary 15 from about 5 to 95 percent of the total composition. Dosage unit forms will generally contain between from about 5 mg to about 500 mg of active ingredient. This dosage may be individualized by the clinician based on the specific clinical condition of the subject being treated. Thus, it will be understood that the specific dosage level for any particular patient may depend upon a variety of factors including the activity of the specific compound 20 employed, the age, body weight, general health, sex, diet, time of administration, route of administration, rate of excretion, drug combination and the severity of the particular disease undergoing therapy. EXAMPLES The present invention may be further understood by reference to the following non 25 limiting examples. Examples of compounds of the present invention and procedures that may be used to prepare and identify useful compounds of the present invention are described below. Abbreviations used in the Examples are as follows: AcOH = acetic acid 30 Boc = tert-butoxycarbonyl CDI = carbonyl di-imidazole DCC = NN-Dicyclohexylcarbodiimide DCE = 1,2-dichloroethane DCM = dichloroiethane 146 WO 2006/099379 PCT/US2006/009049 DDQ =2,3-Dichloro-5,6-dicyano-1,4-benzoquinone DIAD = diisopropyl azodicarboxylate DIEA = diisopropylethylamine DME = dimethoxyethane 5 DMF = N, N-dimethylformamide DMSO= dimethyl sulfoxide EDCI = 1-ethyl-3 -(3-dimethylaminopropyl)-carbodiimide hydrochloride EDTA = ethylenediaminetetracetic acid Et 2 O = diethyl ether 10 EtOAc ethyl acetate h =hour HBTU = 0-benzotriazol-1-yl-N,NN ,N'-tetranethyluronium hexafluorophosphate m-CPBA = meta-chloroperbenzoic acid NMP = N-methyl pyrrolidine 15 r.t. = room temperature TEA = triethyl amine THF = tetrahydrofuran LC-MS data was obtained using gradient elution on a parallel MUXTM system, running four Waters 1525 binary HPLC pumps, equipped with a Mux-UV 2488 multichannel 20 UV-Vis detector (recording at 215 and 254 nM) and a Leap Technologies HTS PAL Auto sampler using a Waters Xterra MS C 18 4.6x50 mm column. A three minute gradient was run from 25% B ( 9 7.5%acetonitrile, 2.5% water, 0.05% TFA) and 75% A (97.5% water, 2.5% acetonitrile, 0.05% TFA) to 100% B. The system is interfaced with a Waters Micromass ZQ mass spectrometer using electrospray ionization. All MS data was obtained in the positive 25 mode unless otherwise noted. 1H NMR data was obtained on a Varian 400 MHz spectrometer. General procedure A: Amide fonnation To a solution of carboxylic acid (1.0 mmol) in dry DMF (2.5 mL) was added HBTU (1.2 mmol) in one portion, the reaction mixture was stirred at room temperature for 10 min, 30 and then an amine (1.0 minol) and DIEA (0.8 mL) were added subsequently. The resulting reaction mixture was stirred at room temperature for 12 h or in some cases at 70 'C for 1-3 h. The reaction mixture was diluted with water (50 mL) and the product was precipitated. The product was either isolated after filtration, subsequent washings with water and ethyl acetate or silica gel column chromatography. 147 WO 20016/099379 PCT/US20061 0 0 9 0 49 General procedure B: Hydrolysis of methyl benzoate To a suspension of a methyl ester (1.0 mmol) in methanol (5 mL) was added lithium hydroxide monohydrate (5.0 mimol), water (5 mL) and THF (5 mL). The reaction mixture 5 was refluxed for 2h. After cooling to room temperature, the reaction mixture was neutralized with AcOH (3 mL). After removal of the organic solvent under vacuum, the crude product was suspended in water (50 mL), and collected by filtration. The solid was further washed with water and dried under high vacuum to afford corresponding acid. General procedure C: Reduction of Nitro to Amine: 10 10.0 mmol of the nitro compound was dissolved in a mixture of 20 mL of CH30H and 5 mL of acetic acid. To this stirring solution 100 mg of 10% Pd on carbon was added, and the resulting mixture was hydrogenated at 42 psi of H2 at room temperature for 3.0 h. The reaction mixture was filtered, and the solid was washed with portions of methanol. The filtrate and washings were combined and evaporated to give the corresponding amine. This 15 product was directly used in subsequent reactions without further purification. Example I The 2,3-diamino-benzoic acid methyl ester was synthesized from 2-amino-3-nitro benzoic acid methyl ester (2.0 g, 10.2 miol) as described in general procedure C 1.7g (10.2 mmol) of the above synthesized diamine was reacted with 1.7 g (10.0 mmol) of 20 isoquinoline-3-carboxylic acid and 4.6 g (12.0 mumol) of HBTU as described in general procedure A to provide 2-amino-3 - [(isoquinoline- 3 -carbonyl)--amino]-benzoic acid methyl ester. LCMS: 322 (M+1)+. To a solution of the above crude product 2-amino-3- {(isoquinoline-3-carbonyl) amino]-benzoic acid methyl ester in AcOH (25 mL) was added AcONH4 (16 g). The reaction 25 mixture was refluxed for 3 h. After removal of AcOH under vacuum, the reaction mixture was washed with water (150 mL) The crude solid product 2-isoquinolin-3-yl-1H benzoimidazole-4-carboxylic acid methyl ester was collected after filtration. LCMS: 304 (M+1) . This product was hydrolyzed according to the general procedure C to provide 2 isoquinolin-3-yl-1H-benzoiniidazole-4-carboxylic acid (1.6 g, 55% yield over three steps). 30 LCMS: 290 (M+l). To a solution of 2.0 g (10 mmol) of 2-bromoacetophenone in DMF (30 mL) was added 3.0 g (29 nunol) 1-acetylguanidine in one portion, the reaction mixture was stirred at rt for 2 days. The reaction mixture was diluted with EtOAc (250 IL) and washed with saturated NH 4 C1 aq. solution (50 maL). The organic phase was dried over MgSO4 and 148 WO 2006/099379 PCT/US2006/009049 concentrated under vacuum. The residue was purified by a flash column chromatography eluting with EtOAc to give N-(4-phenyl-LH-imidazol-2-yl)-acetamide. (0.5 g, 24%). LCMS: 203 (M+1) . To a solution of the above 0.5 g (2.4 mmol) N-(4-phenyl-1H-imidazol-2-yl) 5 acetamide in MeOH (20 mL) was added water (20 mL) and cone. H 2
SO
4 (1 mL). The mixture was refluxed under nitrogen for 2 days. After cooling to rt, the organic solvent was removed under vacuum. The reaction mixture was neutralized with saturated sodium bicarbonate aq. solution (50 mL) and extracted with ethyl acetate (150 mL). The organic layer was dried and condensed. The residue was purified by a flash column chromatography 10 eluting with EtOAc then EtOAc/MeOI= 7:1 to give 4-phenyl- 1 H-imidazol-2-ylanine (160 mg, 41%). LCMS: 160 (M + 1)+ 120 mg (0.4 mmol) of 2-isoquinolin-3-yl-1H-benzoinidazole-4-carboxylic acid was reacted with 400 mg (1.1 mmol) of HBTU and 160 mg (1.0 mmol) of 4-phenyl-1H-imidazol 2-ylamine (160 ng, 1 mmol) according to the general procedure A. The residue was purified 15 by flash column chromatography (EtOAc) to provide 2-isoquinolin-3-yl-1H-benzoimidazole 4-carboxylic acid (4-phenyl-lH-imidazol-2-yl)-amide (30 mg, 17.5%). LCMS: 431 (M + 1). Analogous procedures to those used to prepare the compound of Example 1 were used to prepare the compounds of Examples 2-7. N \>'Arl WN N I H 20 H O Amide LCMS Ex. position W Arl (M+I)' 2 5 isobutyl 2-isoquinolin-3-yl 355 3 5 1H -benzimidazol-2-y1 pyridin-2y1 355 4 5 1H benzimidazol-2-yl 2-naphthalen-2y1 404 5 4 isobutyl 2-isoquinolin-3-yl 345 6 4 phenyl 2-isoquinolin-3-yl 365 7 4 n-butyl 2-isoquinolin-3-yl 345 149 WO 2006/099379 PCT/US2006/009049 Example 8 1.6 g of 2-isoquinolin-3-yl-1H-benzoimidazole-5-carboxylic acid was made from 1.7 g of (10 mmol) methyl 3,4-diamino benzoate as described in Example 1 for the synthesis of 2 -isoquinolin-3-yl-l H-benzoimidazole-4-carboxylic acid. 5 203 mg (1.0 mmol) of 3 -tert-butoxycarbonylamino-3 -cyclopropyl-propionic acid (0.23 g, 1 mmol) was reacted with 380 mg (1.0 mmol) of HBTU and 130 mg (1.0 mmol) of 2-aminoimidazole sulfate as described in general procedure A. The desired aide product was isolated by column chromatography (EtOAc/silica) and carried on directly to the next step. The BOC compound in DCM (3 mL) was treated with 4.0 M HC1/dioxane (15 mL) for 10 approximately I h. The solid 3-Amino-3-cyclopropyl-N- (11-imidazol-2-yl)-propionamide (180 mg, 100%) was collected by filtration and used directly in the next step. 289 mg (1 mmol) of 2-isoquinolin-3-yl-1H-benzoimidazole-5-carboxylic acid (289 mg, 1 mmol) was reacted with 380 ing (I mmol) of HBTU and 3-amino-3-cyclopropyl-N (IH-imidazol-2-yl)-propionamide (230 mg, 1 mnol) as described in general procedure A. A 15 solid precipitated from the solution and was collected by filtration, washed with H20 and Et 2 0, to give 2 -isoquinolin-3-yl-1H-benzoimidazole-5-carboxylic acid [1-cyclopropyl-2 (l1H-imidazol-2-ylcarbamoyl)-ethyl]-amide (20 mg, 4%). LCMS: 466 (M+1)*. 'IH NMR (DMSO-d 6 , 400 MHz): 8 9.20 (s, 1H), 8.56 (s, 11H), 8.09-7.34 (in, 9H), 6.39 (s, 2H), 3.71 (t, 1H), 2.44 (m, 2H), 0.81 (m, 1H), 0.10 (m, 4H) ppm. 20 Example 9 229 mg (1.0 mmol) of 1-tert-butoxycarbonylarnino-cyclopentaneearboxylic acid (0.23 g, 1 mmol) was reacted with 420 mg (1.2 mmol) of HBTU and 200 mg (1.5 mmol) of 2 aminioimidazole sulfate as described in general procedure A. The BOC compound in DCM (3 mL) was treated with 4.0 M HC1/dioxane (15 mL) for approximately I h. The solid 1 25 Amino-cyclopentanecarboxylic acid (1H-imidazol-2-yl)-aimide HCI salt (230 mg, 100%) was collected by filtration and used directly in the next step. 145 mg (0.5 mmol) of 2-isoquinolin-3-yl-lH-benzoinidazole-5-carboxylic acid (Synthesized in Example 1, step 3) was reacted with 280 ing (0.75 mmol) of IBTU and 1 amino-cyclopentanecarboxylic acid (IH-imidazol-2-yl)-amide HC1 salt (120 mg, 0.5 mmol) 30 as described in general procedure A. A solid precipitated from the solution and was collected by filtration, washed with H20 and EtOAc, to give 2-isoquinolin-3-yl-1H-benzoinidazole-5 carboxylic acid (1-(lH-imidazol-2-ylcarbamoyl)-cyclopentyl]- amnide (24 rug, 10%). LCMS: 466 (M+1)*. 150 WO 2006/099379 PCT/ULS2006/009049 Analogous procedures to those used to prepare the compound of Example 9 were used to prepare the compounds of Examples 10-36. 0 X N B-N N N N H H 0 H Stereo LCMS Ex. X B n chemistry (M+1) 4 10 2-fluoro benzyl S 1H-imidazol-2-yl 0 520 11 Benzyl S I H-imidazol-2-yl 0 502 12 2.-chloro-benzyl S 1H-imidazol-2-yl 0 536 13 2-fluorophenyl racemic IT-imidazol-2-yl 1 520 14 Biphenyl racemic IH-imidazol-2-yl 1 578 15 3,5-difluoro -benzyl S 1H-imidazol-2-yl 0 538 16 2-phenyl-ethyl R 1H-imidazol-2-yl 1 530 17 4-phenyl-benzyl racemic 1H-imiidazol-2-yl 1 592 18 3-fluorobenzyl S JH-imidazol-2-yl 0 520 19 4-methoxyphenyl racemic JH-imidazol-2-yl 1 532 20 Phenyl racemic IH-imidazol-2-yl 1 502 21 4-chlorobenzyl S 1H-imidazol-2-yl 0 536 22 4-methylbenzyl S 1H-imidazol-2-yl 0 516 23 Benzyl S Thiazol-2y1 0 519 24 Benzyl S Pyridin-2yl 0 513 25 3, 4 ,5-trifluorobenzyl S 1H-imidazol-2-yl 0 556 26 3-methoxyphenyl racemic IH-imidazol-2-yl 1 532 27 3-trifluoromethylbenzyl S 1H-inidazol-2-yl 0 570 28 2,2 dimethylpropyl S 1H-imidazol-2-yl 0 482 29 Beazyl S 1H -benzimidazol-2-yl 0 552 30 4-fluorobenzyl S 1H-imidazol-2-yl 0 520 151 WO 2006/099379 PCT/US2006/009049 31 4-phenyl-benzyl racemic IH-imidazol-2-yl 0 578 32 Isobutyl S 1H-imidazol-2-yl 0 468 33 3-fluorophenyl racemic 1H-imidazol-2-yl 1 520 34 Butyl-2-yl S 1H-imidazol-2-yl 0 468 35 Isopropyl S 1H-imidazol-2-yl 0 454 36 3-chlorobenzyl S 1H-imidazol-2-yl 0 536 Example 37 1.05g (5.0 mmol) of 2-amino-3-(3,5-difluoro-phenyl)-propionic acid (1.05 g, 5 mmol) was dissolved in 30.0 mL of MeOH, then 1.0 mL of thionyl chloride was added dropwise. 5 The mixture was refluxed overnight. Evaporation of the solvent yielded the solid 2-Amino-3 (3,5-difluoro-phenyl)-propionic acid methyl ester HC1 salt (1.26 g, 100%), and it was used directly in the next step. 145 mg (0.5 nnnol) of 2-isoquinolin-3-yl-1H-benzoimidazole-4-carboxylic acid (synthesized in Example 1, Step 3)) was reacted with 280 mg (0.75 mmol) of HBTU and 2 10 amino-3-(3,5-difluoro-phenyl)-propionic acid methyl ester HCI salt (126 mg, 0.5 mmol) as described in general procedure A. After cooling to room temperature, the reaction mixture was diluted with ethyl acetate (30 mL). The organic phase was washed with 1% NaOH aq. solution, then dried over Na 2
SO
4 and concentrated under vacuum. The residue was purified by flash column chromatography (DCM/EtOAc =1:1 then DCM/EtOAc/MeOH= 15:15:1) to 15 give 3-(3,5-difluoro-phenyl)-2-[(2-isoquinolin-3-yl-1H-benzoimidazole-4-carbonyl)-amino] propionic acid methyl ester (120 mg, 50 %). LCMS: 487 (M+1)t Example 38 Hydrolysis of 100 mg (0.2 mmol) of (3-(3,5-difluoro-phenyl)-2-f(2-isoquinolin-3-yl 1 H-benzoimidazole-4-carbonyl)-amino]-propionic acid methyl ester as described in general 20 procedure B gave 3-(3,5-difluoro-phenyl)-2-{(2-isoquinolin-3-yl-1H-benzoimidazole-4 carbonyl)-amino]-propionic acid (97 mg, 100 %). LCMS: 473 (M + 1)t Example 39 602 mg (2.0 mmol) of 2-tert-butoxycarbonylamino-3-(3,5-difluoro-phenyl)-propionic acid (0.60 g, 2 mmol) was reacted with 1140 mg (3.0 mnol) of HBTU and 400 mg (3.0 mmol) 25 of 2-aminoimidazole sulfate as described in general procedure A. The BOC compound in DCM (3 mL) was treated with 4.0 M HC1/dioxane (15 mL) for approximately 1 h. The solid 152 WO 2006/099379 PCT/US2006/009049 2-Amino-3-(3,5-difluoro-phenyl)-N-(1H-imidazol-2-yl)-propionamide HCi salt (303 mug, 50%) was collected by filtration and used directly in the next step. 145 mg (0.5 mmol) of 2-isoquinolin-3-yl-lH-benzoimidazole-4-carboxylic acid was reacted with 280 mg (0.75 mmol) of HBTU and 2-Amino-3-(3,5-difluoro-pheny)-N-(1H 5 imidazol-2-yl)-propionamide HCl salt (152 mg, 0.5 n-nol) as described in general procedure A. A solid precipitated from the solution and was collected by filtration, washed with H20 and EtOAc, to give 2-isoquinolin-3-yl- I H-benzoimidazole-4-carboxylic acid [2-(3,5 difluoro-phenyl)-1-(IH-imidazol-2-ylcarbamoyl)-ethyl]-amide (60 mg, 22 %). LCMS: 538 (M + 1). 10 H\ ,B X N H o N 0 N N N H Analogous procedures to those used to prepare the compound of Example 39 were 15 used to prepare the compounds of Examples 40-46. LCMS Ex. X B 40 Benzyl IH-benzimidazol-2-yl 552 3,5 41 difluorobenzyl methyl 486 2-methyl-piperidine-1-carboxylic acid tert 42 benzyl 633 butyl ester 43 benzyl thiazol-2yl 519 44 benzyl isobutyl 492 45 heuzyt 3-methyl-piperidine-t-carboxylic acid tert- 633 butyl ester 153 WO 2006/099379 PCT/US2006/009049 LCMS Ex. X B (M+1)* 46 benzyl benzthiazol-2y 569 Example 47 To a stirring solution of 0.86 g (6.0 mmol) of isoquinolin-3-ylamine in dry THF (30 mL) was added 1.25 g (7.0 mmol) of di-imidazol-1-yl-methanethione. The reaction mixture 5 was refluxed for 30 min. After cooling to room temperature, a solution of 1.7 g (10 mmol) of 2,3-diamino-benzoic acid methyl ester (for synthesis see example 1) in THF (40 mL) and DIEA (2 mL) was added subsequently. The reaction mixture was stirred at room temperature for I h and diluted with hexanes (40 mL). The resulting precipitate was filtered. The solid was washed with water and ether and dried under vacuum to give 2-amino-3-(3-isoquinolin 10 3-yl-thioureido)-benzoic acid methyl ester as a solid. To a solution of above thiourea (6.0 mmol) in DCE (50 nil) was added polymer supported-DCC (6.0 g, 7.8 mmol) and DMF (25 mL). The reaction mixture was stirred at 85 *C for lh. The hot reaction mixture was filtered, and the solid resin on the filter paper was further washed with hot DMF (25 mL). The combined organic solution was concentrated 15 under vacuum to give the 2-(isoquinolin-3-ylamino)-3H-benzoimidazole-4-carboxylic acid methyl ester. This methyl ester was hydrolyzed according to the general procedure C to give 2-(isoquinolin-3-ylamino)-3H-benzoinidazole-4-carboxylic acid (1.3 g, 71% overall yields in 3 steps). LCMS: 305 (M+1)*. 100 mg (0.33 mmol) of the above carboxylic acid was reacted with 200 mg (0.53 20 mmol) of HBTU and 200 mg (1.2 mmol) of 2-aminoimidazole sulfate according to the general procedure A. The crude product was purified by flash column chromatography (EtOAc then EtOAc/MeOH= 10:1) to give 2-(isoquinolin-3-yamino)-3H-benzoimidazole-4 carboxylic acid (1H-imidazol-2-yl)-amide (20 mg, 15%). LCMS: 370 (M+1)*. 'H NMR (DMSO-dr, 400 MHz): 8 6.82 (s, 1H), 6.89 (s, 1H), 7.20 (dd, 1H), 7.47 (dd, 1H), 7.71 (m, 25 2H), 7.84 (dd, 1H), 8.04 (s, 111), 8.08 (d, 1H), 8.18 (d, 1H), 9.21 (s, 1H), 11.14 (s, 1H), 11.84 (s, 111), 12.11 (s, 111), 12.60 (s, 1H-) ppm. Analogous procedures to those used to prepare the compound of Example 47 were used to prepare the compounds of Examples 48-57. 154 WO 2006/099379 PCT/US2006/009049 N GN \ N- 2 G N H | H H 0 Amide LCMS Ex.G12 position (M+1)* 48 5 1H-imidazol-2-yl 2-isoquinolin-3-yl 370 49 4 4-fluorophenyl 2-isoquinolin-3-yl 398 50 4 phenyl 2-isoquinolin-3-yl 380 51 5 IH-imidazol-2-yl pyridin-2-yl 320 52 4 4-chlorophenyl 2-isoquinolin-3-yl 414 53 4 4-methoxyphenyl 2-isoquinolin-3-yl 410 54 4 benzyl 2-isoquinolin-3-yl 394 55 5 benzlH-imidazol-2-yl pyridin-2-yl 370 56 4 3-hydroxypropyl 2-isoquinolin-3-yl 362 57 4 methyl 2-isoquinolin-3-yl 318 5 Intermediate A - 2-Amino-3H-benzoimidazole-4-carboxylic acid methyl ester 0 0 H N
H
2 N N 2,3-diamino-benzoic acid methyl ester was synthesized in a quantitative yield (1.7 g) from 2-amino-3-nitro-benzoic acid methyl ester (2.0 g, 8.8 mmol) as described in general procedure C. 10 1.7 g (10.2 mmol) of the above methyl ester was dissolved in a mixture of 20 mL of ethanol and 10 mL of H 2 0. To this stirring solution, 1.6 g (15.3 mmol) of BrCN was added in one portion, and the resulting mixture was refluxed for 1.0 h. The reaction mixture was cooled to room temperature, concentrated to 10 mL, and washed with ethyl acetate (20 mL). 155 WO 2006/099379 PCT/US2006/009049 The EtOAc layer was extracted with water (20 mL). The combined water extracts were neutralized with saturated NaHCO 3 solution to -pI 8, and the resulting suspension was extracted with EtOAc (50 mL). The organic layer was washed with water and brine solution, dried (Na 2
SO
4 ), and evaporated to give 1.7 g (90%) of the title compound. LCMS: 192 5 (M+1)* Intermediate B - 2-Amino-3H-benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl) amide: HN N HN 0 H N
H
2 N N 5.0 g (27.4 mmol) of 2-amino-3-nitro-benzoic acid, 12.48 g (32.9 mmol) of HBTU 10 were suspended in 20 mL of DMF. To this suspension 10 mL of DIEA was added, and the resulting mixture was stirred for 10 minutes to form a paste. To this paste, 10 nL of DMF was added followed by 9 g (68.2 mmol) of 2-amino imidazole sulfate. The resulting mixture was heated at 75 0 C for 3.0 h. The reaction mixture was cooled to room temperature and 5 mL of 2N NaOH was added followed by 200 mL of brine solution. A solid precipitated from 15 the solution and was collected by filtration, washed with water (300 ml), ethyl acetate (100 ml), and dried under vacuum to provide 5.3 g (75%) of 2-amino-N- (IH-imidazol-2-yl)-3 nitro-benzamide as a yellow solid. The product was hydrogenated according to the general procedure C to give 2,3-diamino-N-(lH-imidazol-2-yl)-benzamide (4.5 g). 4.5 g (21.2 mmol) of the above solid was dissolved in a mixture of 40 mL of ethanol 20 and 8 mL of H20. To this stirring solution 3.3 g (31.8 mmol) of BrCN was added and the resulting mixture was refluxed for 1.0 h. The reaction mixture was cooled to room temperature and the solvent was evaporated and adjusted to -pH 8.0 with aqueous NH40H. The resulting solid was filtered. The solid was further washed with water (100 mL), ethyl acetate (50 mL) and dried under vacuum to give 2.5 g (50%) of 2-amino-3H-benzoimidazole 25 4-carboxylic acid (1H-imidazol-2-yl)-amide. LCMS: 243 (M+I)+. 156 WO 2006/099379 PCT/US2006/009049 Example 58 2.6 g (75%) of the 2-[(isoquinoline-3-carbonyl)-amino]-1H-benizoimidazole-4 carboxylic acid methyl ester was synthesized from isoquinoline-3-carboxylic acid (1.7 g, 10 mmol), HBTU (4,6 g, 12 mmol), and the above synthesized 2-amino-3H-benzoimidazole-4 5 carboxylic acid methyl ester (1. 9 g, 10 mnim) as described in general procedure A. This ester was then hydrolyzed according to general procedure B to give 2.2 g (88%) of acid. LCMS: 333 (M+1)t 332 mg (1.0 nunol) of above 2-[(isoquinoline-3-carbonyl)-amifo]-IH benzoimidazole-4-carboxylic acid (0.33 g, 1.0 mmol) was reacted with 800 mg (2.0 mmol) of 10 HBTU and 2-aminoimidazole sulfate (400 mg, 3.0 mmol) in 3.0 mL of DMF and 1.0 mL of DIEA as described in general procedure A. After cooling to room temperature, the reaction mixture was diluted with 0.6 mL of 5 NNaOH aq, solution and 4.0 mL of brine. The solid was collected by filtration and washed with 2 NNaOH aq. (3.0 mL), followed by H20 and EtOAc (3X of each), to give 224.0 mg of the crude compound. The crude was heated in 3.0 15 mL of DMF at 80 0 C for 20 minutes. The filter cake was washed by another 1.0 mL of hot DMF. The combined filtrate was reheated to a clear solution, then cooled and addition of 2.0 mL of EtOAc, followed by 4.0 mL of H20. The mixture was stirred, and a solid was gradually formed between EtOAc and DMF/H 2 0 layers. The solid was collected by filtration and washed with H20, EtOAc and MeOH (3X of each), then dried, to yield isoquinoline-3 20 carboxylic acid [4-(1H-imidazol-2-ylcarbamoyl)-1H-benzoimidazol-2-yl]-amide (83.0 ng, 21%). LCMS: 398.0 (M+1)+. 'H NMR (DMSO-d 6 , 400 MHz): 8 12.29 (s, 1H), 11. 86 (s, 1H), 9.52 (s, 1H), 8.81 (s, 1H), 8.31 (t, 211), 7.80-7.95 (m, 4H), 7.32 (t, 111), 6.88 (s, lH), 6.77 (s, 1H) ppm. Example 59 25 70 mg (0.45 mmol) of 4-chloro-benzoic acid 170 ing (0.45 mmol) of HBTU in a mixture of 3 ml of DMF and I mL of DIEA was reacted with 100 mg (0.4 mmol) of 2-amino 3H-benzoimidazole-4-carboxylic acid (lH-inidazol-2-yl)-amide synthesized above as described in general procedure A and 2-(4-chloro-benzoylamino)- 1 H-benzoimidazole-4 carboxylic acid (1H-imidazol-2-yl)-amide (30 mg, 17.24 %) was isolated. LCMS: 382 30 (M+l). 'H NMR (DMSO-d 6 , 400 MHz): 5 11.99 (d, 1H), 8.16 (d, 2H), 7.94 (d, 111), 7.70 (dd, 3H), 7.30 (t, 1H), 6.81 (d, 2H) ppm. 157 WO 2006/099379 PCT/US2006/009049 Analogous procedures to those used to prepare the compound of Example 59 were used to prepare the compounds of Examples 60-127. N H ',-N Ar, NN H 0 H 0 5 Amide LCMS Ex. position W Ar, (M+1)* 60 4 1H-benzimidazol-2-yl isoquinolin-3-yl 448 61 4 benzthiazol-2-yl isoquinolin-3-yl 465 62 4 {1,3,4}-thiadiazol-2yl isoquinolin-3-yl 416 63 4 pyridin-2-yl isoquinolin-3-yl 409 64 4 pyridin-3yl isoquinolin-3-yl 409 65 4 1H-benzimidazol-2-yl IH-indole-2yl 435 66 5 1H-benzimidazol-2-yl isoquinolin-3-yl 448 67 4 1H-indazole-5-yl isoquinolin-3-yl 448 68 4 quinolin-3-yl isoquinolin-3-yl 459 69 5 benzothiazol-2-yl isoquinolin-3-yl 465 70 5 thiazol-2-yl isoquinolin-3-yl 415 71 4 IH-imidazol-2-yl phenyl 347 72 4 furan-2-yl methyl isoquinoline-3yl 412 73 4 2-phenyoxy-phenyl isoquinolin-3-yl 500 74 4 3-phenoxy-phenyl isoquinolin-3-yl 500 75 4 4-phenoxy-phenyl isoquinolin- 3 -yl 500 76 4 (1S)-I-phenyl ethyl isoquinolin-3-yl 436 77 4 (1R)-l-phenyl ethyl isoquinolin- 3 -yl 436 78 4 1H-imidazol-2-yl benzothiophene-2-yl 403 79 4 1H-imidazol-2-yl naphthalen-2-yl 397 80 4 1 H-imidazol-2-yl 4'-biphenyl 423 81 4 1H-imidazol-2-yl 1H-indole-2yl 386 158 WO 2006/099379 PCT/[JS2006/009049 82 4 1H-inidazol-2-yl 3,5-difluorophenyl 383 83 4 1H-imidazol-2-y1 4-fluorophenyl 365 84 4 (thiophene-2-yl)ethyl isoquinolin-3-yl 442 85 5 phenyl isoquinolin-3-y 408 86 4J (5-methylfuran-2-y1)-methyl isoquinolin-3-y1 360 7 4 ben-zyl isoquinolin- 3 -yl 422 8 4 (thiophene-2-yl)mothyl isoquinolin-3-yl 38 89 4 phenethyl isoquinolin-3-yl 436 90 4 ethyl isoquinolin-3-y1 360 91 4 tert-butyl isoquinolin- 3 -y1 388 9 2 4 1H-imidazol-2-y1 benzofuran-2-yl 387 93 4 ~~2,3,4,9-tetrahiydro-1IH-b eta- iounln3y 8 carboline-2-yl 944 (bnotiphn-2y)methyl isoquinolin-3--yl 478 95 4 (benlzthiazol--2-yl)methiyl isogainolin-3-y1 7 96 4 (1-nethylbenzimidazole- 2 - isoquinolin- 3 -y1 476 yl)methyl S2-(H-benzimidazole-2-yl)- isoquinolin- 3 -yl 476 ethyl 98 4 1H-imidazol-2-yl 4-fluorobenzyl 379 3-mnethoxy-4-fluoro 99 4 1H-imidazol-2-yl eyl395 phenyl 100 4 1l-imidazol-2-yl 3,4-dimethoxyphenyl 407 101 4 thiazole-2-yl-methyl isoquinolin- 3 -yl 429 102 4 f{-benzirnidazole-2-y. isoquinolin-3-yi 462 methyl 103 4 1H-imidazol-2-yl 4-trifluoromethylphenyl 415 104 4 1H-imidazol-2-yl 4-phenyoxyphenyl 439 105 4 1H-imidazol-2-y1 quinolin-3-yl 398 106 4 1H-inidazol-2-y1 quinolin- 2 -yl 398 107 4 1H-imidazol-2-yl furan-3-yl 334 108 4 1H-imidazol-2-yl 4-tert-butyl-phenyl 403 109 4 1H-imidazol-2-yl 4-nitro-phenyl 392 159 WO 2006/099379 PCT/US2006/009049 1H-imidazol-2-yl 4-nethoxy-phenyl 377 4 H-imidazol-2-yl 1H-imnidazol-2-yl 337 112 4 1H-imidazol-2-y1 4-isopropyl-phenyl 389 113 4 1H-imidazol-2-y1 3-fluoro-phenyl 365 114 4 1H-imidazol-2-yl 2-fluoro-phenyl 365 115 4 1H-inidazol-2-yl 5-chloro-benzofutan-2-yl 421 116 4 1 H-imidazol-2-yl 6,7-dimethoxy-2 isoquinolin- 3 -y4 117 4 1H-imidazol-2-yl 3-chloro-phenyl 381 118 4 1H-imidazol-2-y1 2-chloro-phelinyl 381 susenedin10no e 11dat- 1H-indazol e -3-y1 387 112 4 1H-imidazol-2-yl 1-ehyl-indolen 400 121 4 1H-imidazo1-2-yl 1-methy-indaole--ye 401 1022 ove 30-p.heny-H mixtdazourd-2-yl beke isotainoni-y ic 474nuraie 3 4 1H-iidazol-2-yl -mthyl-ingdoleid ws l0 124 4 g170%) H-iacidazol-2-y 5hoxpidmin-h-yl 4 4 125 4 1H-imidazo1-2-y1 -benzyl- indole-2-y1 476 126 4 1H-imidazol-2-y1 1-n-propyl- indole-2y1 428 127 4 H-imidazol-2-yl 15 -dimethy1- indole-2y1 - 4 Example 128 1.0 g (5.2 mmol) of 2-amino-3H-benzoimidazole-4-carboxylic acid methyl ester was suspended in 10 mL of cone. H2SO4 at -15 OC and cooled to -30 "C. Potassium nitrate (0.589 g, 5 5.72 mmol) dissolved in 10 ml of sulfuric acid and cooled to 0 tC was then added dropwise to the reaction mixture. The reaction mixture was stirred -30 "C for 15 min then warmed to 10 "C over 30 min. Then the mixture was poured into a beaker containing ice and neutralized (~pH = 8) with concentrated aqueous amm-onia solution. The resulting solid was filtered. The solid obtained was washed with cold water and ether, and dried under vacuum to provide 10 0.85 g (70%) 2-amino-6-nitro-1H-benzoim-idazole-4-carboxylic acid methyl ester as a solid. LCMS: 239 (M + 1)*. 0.5 g (2.12 mmol) of 2-amino-6 -nitro-1IH-benzoimiidazole-4-carboxylic acid methyl ester was reacted with 0.37 g (2.12 mmol) of isoquinoline 3-carboxylic acid and 0. 89 g (2.33 mmol) of HBTU as described in general procedure A to provide 0.67 g (80%) of 2 160 WO 2006/099379 PCT/US2006/009049 [(Isoquinoline-3-carbonyl1)-amino]-6-nitrTo-H-benzoimidazole-4-carboxylic acid methyl ester. LCMS: 392 (M + 1)'. The ester was dissolved in dry DMF (5 mL) and 0.22 g (5-1 mmol) of LiC1 in one portion was added. The resulting mixture was stirred at 120 OC for 12 h, diluted with water, and the solid was collected by filtration. The solid was washed with water (20 5 mL), ether (10 mL) and dried under vacuum to provide 0.6 g (93%) of 2-[(isoquinoline- 3 carbonyl)-amino}-6-nitro-1H-benzoimidazole-4-carboxylic acid. LCMS: 378 (M + 1)*. 0.5 g (1.3 mmol) of the above acid (Example 128) was reacted with 0.6 g (1.6 mmol) of HBTU and 0.35 g (1.3 mmol) 2-amino imidazole as described in general procedure A. After the reaction was complete, the reaction mixture was diluted with water 20 (mL), 3.0 mL 10 of 2 N NaOH, and stirred for 15 min. The resulting solid was filtered, washed with water, diethyl ether and dried under vacuum to provide 0.1 g (17.2 %) of isoquinoline-3-carboxylic acid [4-(l -imidazol-2-ylcarbamoyl)-6-nitro- IH-benzoimlidazo1-2-yl]-amide. LCMS: 443 (M + 1)* Example 129 15 0.1 g (0.2 nnol) of isoquinoline-3-carboxylic acid [4-(1H-imidazol-2-ylcarbamoyl) 6-nitro-1H-benzoimidazol-2-y1]-amide (Example 129) was suspended in a mixture of 2 mL of DMF and 5 mL of acetic acid. To this stirring solution, -20 mg of 10% Pd on carbon was added, and the resulting mixture was hydrogenated at 42 psi of H2 at room temperature for 3.0 h. The reaction mixture was filtered, and the solid was washed with 5 mL of DMF. The 20 filtrate and washings were combined and evaporated to reduce the volume to 10 mL, then diluted with saturated NaHCO 3 (10 mL), and extracted with ethyl acetate. The organic layer was washed with water and brine and evaporated to provide 85 mg (91 %) of isoquinoline-3 carboxylic acid [6-amino- 4 - (1H-imidazol-2-ylcarbamoyl)-lH-benzoimidazol-2-yl]-amide. LCMS: 413 (M + 1). 25 Example 130 20 mg (0.04 mmol) of isoquinoline-3-carboxylic acid [6-amino- 4 - (lH-imidazol-2 ylcarbamoyl)-1-H-benzoimidazol-2-yl}-amide (Example 129) was dissolved in pyridine (2 mL). To this stirring solution at 0 *C, 5.6 mg (0.053 muol) of isobutyryl chloride was added in one portion. The reaction mixture was stirred for 10 min at 0 4C and warmed to room 30 temperature. The pyridine was evaporated and water (5 mL) was added to the reaction mixture. The mixture was stirred for 10 min. and the resulting solid was filtered, washed with water (2 mL) and diethyl ether (5 mL), and dried to provide 15.5 mg (66.5%) of isoquinoline 3-carboxylic acid [4-(1H-limidazol-2-ylcarbamoy)- 6 -isobutyrylamin-o-1H-benzoimidazol- 2 161 WO 2006/099379 PCT/US2006/009049 yl]-amide. LCMS: 483 (M + 1)*. 'H NMR (CD 3 0D, 400 MHz): 6 9.43 (s, IR), 8.77 (s, 1H), 8.06-8.31 (m, 5H), 7.80-7.93 (m, 3H), 7.08 (s, 2H), 2.45 (m, 1H), 1.1 (d, 6H) ppm. Analogous procedures to those used to prepare the compound of Example 130 were used to prepare the compounds of Examples 131-133. N NH 0 N, N H I N \ H H 0 R 5 Ex. R LCMS (M+1)+ 131 phenyl 517 132 isobutyl 497 133 benzyl 531 Example 134 To a solution of 0.1 g (0.52 minmol) of 2-anino-IbH--bnzoimidazole4-carboxylic acid methyl ester (intermediate A) in DMSO (1 nL) was added 0.089. g (0.52 mmol) of benzyl 10 bromide, the reaction mixture was stirred at room temperature for 3h. The conversion was monitored by LCMS. After usual work up, the residue was purified by column chromatography eluting with DCM/methanol (vlv== 10:1) to afford 0.08 g (55%) of 2-amino 1-benzy1-1H-benzoinidazole-4-carboxylic acid methyl ester. LCMS: 281 (M + 1)*; 'H NMR (DMSO-d 6 , 400 MHz): 6 7.49 (dd, 1H), 7.29 (t, 2H), 7.25 (in, 2H), 7.16(m, 2H), 7.05 (bs, 15 2H), 6.85 (t, 1H), 5.28 (s, 2H), 3.79 (s, 3H) ppm. To a solution of 0.031 g (0.18 mmol) of isoquinoline-3-carboxylic acid in DMF (1 mL) was added 0.085 g (0.22 mmol) of HBTU and 0.1 mL of DIEA. The mixture was stirred for 5 min, then added 0.04 g (0.18 mmol) of 2-amino.-1-benzyl-1H-benzoimidazole.4-carboxylic acid methyl ester in one portion. The reaction mixture was stirred for 1 h. After usual workup, 162 WO 2006/099379 PCT/US2006/009049 45 mg (73%) of 1-benzyl-2-[(isoquinoline-3-carbonyl)-amino-1IH-benzoimidazOle- 4 carboxylic acid methyl ester was isolated after purification by colunm chromatography eluting with DCM/methanol (v/v from 15:1 to 10:1). LCMS: 437 (M + 1); '1H NMR (DMSO-d 6 , 400 MHz): 5 12.8 (s, 1H1), 9.41 (s, IH), 8.92(s, 111), 8.20 (d, lH), 8.14 (d, 1H), 5 7.84 (t, 1H), 7.77 (in, 3H), 7.51 (d, 2H), 7.32 (m, 4H), 5.63 (s, 2H), 3.99 (s, 3H) ppm Example 135 To a solution of 45 mg (0.12 mmol) of (1-benzyl-2-[(isoquinoline-3-carbonyl) amino-1lH-benzoimidazole-4-carboxylic acid methyl ester (Example 134) in THF (1mL) was added methanol (I mL) and I N LiOH solution (lmL). The reaction was heated at 65 C for 10 45 min., and the conversion was monitored by TLC. After cooling to room temperature, the organic solvents were removed under reduced pressure, the reaction residue was acidified (pH = 2) by addition of 1 N HC1 soluiton. The solid product was collected by filtration and further dried to afford 43 mg (100%) 1-benzyl-2-[(isoquinoline-3-carbony1)-amino]-1H benzoimidazole-4-carboxylic acid. LCMS: 423 (M + 1). 15 To a solution of 43 mg (0.12 mmol) of the above crude acid in DMF (0.7 mL) was added 75 mg (0.2 mmol) of HBTU and DIEA (0.1 nL). The mixture was stirred for 5 min, then 40 mg (0.3 mmol) of 2-aminoimidazole sulfate was added in one portion. The reaction mixture was heated at 80 "C for 1 h. After usual workup as described in General Procedure A, 25 mg (50%) of isoquinoline-3-carboxylic acid [1-benzyl-4-(1H-imidazol-2-ylcarbamoyl) 20 1H-benzoimidazol-2-yl]-amide was isolated after purification by column chromatography eluting with DCM/methanol (v/v from15:1 to 7:1). LCMS: 488 (M + 1). Example 136 15 mg of isoquinoline-3-carboxylic acid [4-(1H-imidazol-2-ylcarbamoyl)-1 -methyl 1H-benzoimidazol-2-yl]-amide was synthesized following the same procedures described for 25 above Example 135, with the exception that methyl iodide was used instead of benzyl bromide. LCMS: 412 (M+1)4. Example 137 20 mg of isoquinoline-3-carboxylic acid (4-benzylcarbamoyl-l-methyl-iH benzoimidazol-2-yl)-amide was synthesized following the same procedures described for 30 Example 135, with the exception that methyl iodide was used instead of benzyl bromide and benzyl amine was used instead of 2-aminoimidazole sulfate. LCMS: 436 (M+1)+. 163 WO 2006/099379 PCT/US2006/009049 Example 138 To a solution of 48.4 mg (0.2 mmol) of 2-amino-3H-benzoiiidazole-4-carboxylic acid (1HJ-imidazol-2-yl)-amide (Intermediate B) in dry DMF (1.0 mL) was added 49 mng (0.3 mmol) of di-imidazol-1 -yl-inethanone in one portion. The mixture was heated at 50 "C for 30 5 nin, after cooling to room temperature, 23 mg (0.16 nnol) of isoquinolin-3 -ylamine was added to the reaction mixtures. The reaction mixture was stirred at 45 C for 45 min. After cooling to r.t., the organic solvent was removed under reduced pressure. The residue was purified by column chromatography eluting with DCM/methanol (v/v= 10:1 to 8:1) to give 17 mg (20%) of 2-(3-isoquinolin-3-yl-ureido)-1H-benzoimidazole-4-carboxylic acid (1H 10 imidazol-2-yl)-amide. LCMS: 413 (M+1)*. Example 139 To a solution of 150 mg (0.95 minol) of isoquinoline-3-carbaldehyde in dry DCE (1 mL) was added 60 mg (0.25 mmol) of 2-amino-3H-benzoimidazole-4-carboxylic acid (1H imidazol-2-yl)-amide (intennediate B) , and Ti(OiPr)4 (3 mL). The mixture was sonciated for 15 30 min then stirred at room temperature for 6 h. NaBH4 (0.5 g) was added, and the mixture was stirring for 12 h. The reaction was quenched by addition of methanol (2 mL). After usual workup, the residue was purified by column chromatography eluting with DCM then EtOAc/methanol (v/v=100:5 ) to give 7 mg (7%) of 2-(isoquinolin-3-ylmethyl)-amino1-IH benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-ainide. LCMS: 384 (M+1)*. 'H NMR 20 (DMSO-d 6 , 400 MHz): 8 12.58 (s, 1HT), 11.8 (s, 1H), 11.6 (s, iH), 9.07 (s, 1H, 8.69 (s, 1H), 8.22 (t, 1H), 7.96 (d, 1H), 7.91 (d, 1H), 7.67 (in, 2H), 7.53 (t, 1H), 7.36 (d, 1H), 7.02 (t, IH), 6.90 (s, 1H), 6.85 (s, 1H), 4.78 (d, 2H) ppm. Example 140 3.4 g (20.5 mmol) of methyl 3,4-diaminobenzoate was dissolved in a mixture of 50 25 mL of ethanol and 10 mL of H20. To this stirring solution 2.6 g (24.5 mmol) of BrCN was added in one portion, and the resulting mixture was heated under reflux for 1.0 h. The reaction mixture was cooled to room temperature, concentrated to 10 mL, then washed with ethyl acetate (20 mL). The EtOAc layer was extracted with water (20 mL). The combined water extracts were neutralized with saturated NaHCO 3 solution to -pH 8, and the resulting 30 suspension was extracted with EtOAc (50 mL). The organic layer was washed with water and brine solution, dried with Na 2
SO
4 , and evaporated to give 2-amino-IH-benzoimidazole-5 carboxylic acid methyl ester, which was used directly in the next step without further purification. 164 WO 2006/099379 PCT/US2006/009049 2-[(Isoquinolint-3-carbonyl)-amino]-1IH-benzoimidazole-5-carboxylic acid methyl ester was prepared from 1.9 g (10 immol) of isoquinoline-3-carboxylic acid monohydrate, 4.6 g (12 inmol) of HBTU, and 1.9 g, (10 mmol) of 2-anino-1H-benzoimidazole-5-carboxylic acid methyl ester as described in general procedure A. A 0.35 g (1.0 mmol) portion of 2 5 [(isoquinoline-3-carbonyl)-amino]-1H-benzoinidazole-5-carboxylic acid methyl ester was subsequently hydrolyzed according to general procedure B to give 2-{(isoquinoline-3 carbony)-amino]-1H-benzoimidazole-5-carboxylic acid as a light yellow solid, which was used in the final coupling step without further purification. 0.15 g (0.45 mmol) of 2-[(isoquinoline-3-carbonyl)-amino1 4 H-benzoimidazole-5 10 carboxylic acid was reacted with 0.26 g (0.68 mmol) of HBTU and 0.06 g (0.45 mmol) of 2 aminoimidazole sulfate in 3.0 mL of DMF and 1.0 mL of DIEA as described in general procedure A. After cooling to room temperature, the reaction mixture was diluted with water, the solid was collected by filtration, dissolved with MeOH and evaporated onto silica gel, and isolated by flash column chromatography (500 mL DCM/20 mL NH3/MeOH) to yield - 10 15 mg of the desired isoquinoline-3-carboxylic acid [5-(1H-imidazol-2-ylcarbamoyl)-IH benzoimidazol-2-yll-ainide. LCMS: 398.0 (M+1)* Example 141 To a solution of 1.53 g (10 mmol) of 3-nitro-benzene-1,2-diaminie in ethanol (20 mL) was added 1.3 g (12.5 mmol) of BrCN and water (5 mL). The mixture was refluxed for 1.0 h. 20 After cooling to room temperature, the solvent was concentrated, then the residue was dissolved in water, the pH was adjusted to 8.0 with sat. NaHCO3 solution. The resulting solid was filtered, washed with water and dried under vacuum to afford 1.42 g (80%) of the 7 nitro-1H-benzoimidazol-2-ylamine. LCMS: 179 (M+1) t . 1.42 g (8 mmol) of above 7-nitro-1H-benzoimidazol-2-ylamine was reacted with 1.38 25 g (8 mmol) of isoquinoline-3-carboxylic acid, 3.4 g (9 mmol) of HBTU in 30.0 mL of DMF and 5.0 mL of DIEA as described in general procedure A. After cooling to room temperature, the reaction mixture was diluted with 60 mL of saturated NaHCO 3 and 100 mL of water. The solid was collected by filtration and washed with water and dried under reduced pressure, and subsequently subjected to hydrogenation as described in general procedure C to give 1.0 g 30 (41 %) of the isoquinoline-3-carboxylic acid (7-amino- 1 H-benzoimidazol-2-yl) -amide. LCMS: 304 (M+1) t To a solution of 30 mg (0.1 mmol) of above isoquinoline-3-carboxylic acid (7-amino 1l-benzoimidazol-2-yl)-amide in dry DMF (1 niL) was added 24 mg (0.15 mmol) of di imidazol-1-yl-methanone in one portion. The mixture was heated at 50 "C for 30 min. After 165 WO 2006/099379 PCT/US2006/009049 cooling to room temperature, 16 mg (0.15 mmol) of 4-fluoro-phenylanine was added to the reaction. The reaction mixture was stirred at 45 ' C for 45 min. After cooling to room temperature, the organic solvent was removed under reduced pressure, and the residue was purified by column chromatography eluting with DCM/methanol (v/v= 10:0.5 to 10:1) to 5 provide 17 mg (39%) of isoquinoline-3-carboxylic acid {7-[3-(4-fluoro-phenyl)-ureido]-1H benzoimidazol-2-yl}-amide. LCMS: 441 (M+1). By analogous methods to those used to prepare Example 141 and those in the relevant above schemes, the following compounds were synthesized. o Ar %-NH HN 0 HN N N N I H EX Ar LCMS: (M+ 1)* 142 4-methoxy phenethyl 481 143 4-methoxy benzyl 467 10 Example 144 To a stirring solution of 200 mg (0.92 mmol) of 2,3-diamino-N-(lH-imidazol-2-yl) benzamide (synthesized as described in Intermediate-B) in 2.0 mL of acetic acid was added 200 mug (1.15 mmol) of 2,2,2-trichloroacetamide and stirring continued for 30 min at r.t. Then 15 0.5 mL of 1% HCl in methanol was added and stirred for another 45 min at rt. Evaporation of the solvent gave the crude ester, which was dissolved in 4:1 THF and methanol (5 mL) and then 1.0 mL of 2N LiOH was added at r.t. The reaction mixture was stirred at r.t. for 30 min, the pH was adjusted to 3.0 and then extracted with ethyl acetate twice (2 x 25 mL). The combined organic solvents were dried, evaporated to provide 100 mg (40%) of 4~11H 20 imidazol-2-ylcarbamoyl)-1 H-benzoimidazole-2-carboxylic acid, which was used, for the next step without further purification. LCMS: 272 (M + 1)'. To a stirring solution of 100 mg (0.36 mmol) of above acid in 2.0 ml of DMF was added 140 mg (0.72 mmol) of EDCI. The reaction mixture was stirred for 10 min then 53 mg (0.36 mmol) of Isoquinolin-3-ylamine was added. The reaction mixture stirred for another 20 25 h and cooled to room temperature 5.0 mL of water and 5.0 mL of saturated NaHCO 3 was 166 WO 2006/099379 PCT/US2006/009049 added. The resulting solid was filtered washed with water, ether and dried to provide 20 mg (13.6%) of lH-benzoimidazole-2,4-dicarboxylic acid 4-[(1H-imidazol-2-yl)-amide] 2 isoquinolin-3-ylamide. LCMS: 398 (M + 1)*. Example 145 5 To a solution of 0.33 g (1.0 mmol) of -[(isoquinoline-1-carbonyl)-aminol- 3
H
benzoimidazole-4-carboxylic acid (from Example 58) in DMF (5 mL) was added 0.47 g (1.25 mmol) of HBTU and DIEA (0.5 mL). The mixture was stirred for 5 min, then 0.22 g (2.0 mmol) of benzene--1,2-diamine was added in one portion. The reaction mixture was heated at 50 *C for lh, after cooling to room temperature, the coupling product was precipitated by 10 addition of sat. aq. sodium carbonate solution (15 mL). The solid was filtered, washed with water, and dried to provide 0.35 g (80%) of isoquinoline-1-carboxylic acid [7-(2-amino phenylcarbamoyl)-1H-benzoimidazol-2-yl]-anide. LCMS: 443 (M+1)*, To a solution of 169 mg (0.4 mmol) of isoquinoline-l-carboxylic acid [7-(2-amino phenylcarbamoyl)-1 H-benzoimidazol-2-ylJ-amide in AcOH (15 mL) was added NU 4 OAc 15 (0.5 g). The mixture was heated at 130 *C for 30 min. After cooling to room temperature, the AcOH was removed under reduced pressure, extracted with EtOAc (50 mL), washed with sat. aq. sodium bicarbonate solution and brine. The organic phase was dried over sodium sulfate and concentrated. The residue was dissolved in DCM (3 mL), and 5 mL of hexane was added. The resulting solid was filtered and dried to afford 120 mg (75 %) of 20 isoquinoline-l-.carboxylic acid (1H,3'H-[2,4']bibenzoimidazolyl-2'yl)-amide. LCMS: 405 (M+1). By analogous methods to those used to prepare Example 145 and those in the relevant above schemes, the following compounds were synthesized. R Q HN N N N H LCMS Ex. R 146 3H-Imidazo[4,5-c]pyridine-2-y1 406 147 4-phenyl-1H.-imidazol-2-yl 431 148 lH-imidazol-2-yl 355 167 WO 2006/099379 PCT/US20061009049 149 4-(4-chloro-phenyl)-1H-imidazol-2-yI 465 Example 150 To a solution of 61 mg (0.2 rmol) of isoquinoline-3-carboxylic acid (4-amino-1H benzoimidazol-2-yl)-amide in AcOH (5 mL) was added formaldehyde (0.7 mL, 37% aq. 5 solution), 0.3 mL of ethanedial and NH 4 OAc (0.5 g). The mixture was heated at 120 "C for 1h. After cooling to room temperature and usual workup, the residue was purified by column chromatography eluting with EtOAc/methanol (v/v=1 0:1) to give 13 mg (18 %) of isoquinoline-3-carboxylic acid (4-imidazol-1-yl-1H-benzoinidazol-2-y1)-amide. LCMS: 355 (M+1). 10 Example 151 To a solution of 0.17 g (0.5 mmol) of 2-[(isoquinoline-3-carbony)-amino]-lH benzoimidazole-4-carboxylic acid (from Example 58) in DMF (3 mL) was added 0.13 g (1 mmol) of 1-chloro-3,3-dimethyl-butan-2-one and DIEA (1 mL). The mixture was stirred at room temperature for 1 h., extracted with ethyl acetate, washed with water and brine. The 15 organic layer was dried over magnesium sulfate, evaporated under reduced pressure to provide the 0.17 g (85%) of the keto ester which was dissolved in AcOH (5 mL), 0.5 g of NH4 0 Ac was added, the mixture was stirred at 100 'C for 1 h under microwave (150 W, 250 psi), The solvent was evaporated and the residue was purified by column chromatography eluting with DCMIEtOAc (v/v 1:1) to give 90 ing (73%) of isoquinoline-3-carboxylic acid 20 [4-(4-tert-buty1-oxazol-2-y1)-lH-benzoimidazo-2-y]-amide. LCMS: 412 (M+l). Example 152 To a stirring mixture of 1.Og (5.2 mmol) of 2-anino-3H-benzoimidazole-4 carboxylic acid methyl ester (Intermedate A) and 0.86 g (5.2 mmol) of CDI in 3.0 mL of DMF was added 0.75 g (5.2 mmol) of Isoquinolin-3-ylamine. The reaction mixture was 25 heated at 60 0 C for 30 min, and then cooled to room temperature. 5.0 mL of water was added. The resulting solid was filtered washed with water (10 ml) and ether (10 mL), and dried to provide the 2-(3-isoquinolin-3-yl-ureido)-IH-benzoimidazole-4-carboxylic acid methyl ester as a solid which was hydrolyzed as described in general procedure B to provide 0.75 g (40%) of 2-(3-isoquinolin-3-yl-ureido)-1H-benzoimidazole-4-carboxylic acid. LCMS: 348 (M + 30 W). 0.2 g (0.57 mmol) of the above acid was reacted with 0.24 g (0.63 mmol) of HBTU and 0.083 g (0.57) of 1H-pyrazole-1-carboxamdine as described in general procedure A. 168 WO 2006/099379 PCT/US2006/009049 After the reaction was complete the reaction mixture was diluted with water (5.0 mL), and saturated NaHCO 3 was added and stirred for 15 min. The resulting solid was filtered, washed with water, ether, and dried under vacuum to provide 0.15 g (62.5 %) of isoquinoline-3 carboxylic acid {4-[(inino-pyrazol-1--yl-methyl)-carbamoyl]-1H-benzoimidazol-2-yl}-amide 5 as a solid. LCMS: 425 (M + 1). To a stirring solution of 0.05 g (0.12 mmoL) of above amide and 0.088 g (1.12 mmol) of 3-amino-propan-1-ol in DCM (5.0 mL) was added 0.077 g (0.68 mrmol) of DIEA. The reaction mixture was stirred at r.t. overnight. 5.0 nL of water was added, and the resulting solid was filtered, washed with water and ether, and dried to provide 0.03 g (57.6 %) of 1- 4 10 [N'-(3- hydroxy-propyl)-guanidinocarbonyl}-1H-benzoimidazol-2-yl}-3-isoquinolin-3-yl-urea. LCMS: 447 (M + 1). By analogous methods to those used to prepare Example 152 and those in the relevant above schemes, the following compounds were synthesized. R HN YNH NH 0 0 HN Na N N H LCMS EX R 153 n-propyl 431 154 4-hydroxy-butyl 461 15 Example 155 100 mg (0.3 mmol) of 2-[(isoquinoline-3-carbonyl)-amino]-H-benzoimidazole- 4 carboxylic acid (from Example 58) was reacted with 130 mg (0.33 mmol) of HBTU and 55 mg (0.3 mmol) 4-methanesulfonyl-benzylamine in 3.0 mL of DMF and 1.0 mL of DIEA as 20 described in general procedure A. After the usual work up as described in general procedure 169 WO 2006/099379 PCT/US2006/009049 A, 50 mg (33 %) of isoquinoline-3-carboxylic acid [4-(4-methanesulfonyl-benzylcarbamoy) I1-benzoimidazol-2-ylnamide was isolated. LCMS: 500 (M+1)*. Example 156 To a solution of 5 g (38 mmol) of 2-chloro-cyclohexanone in MeCN (150 mL) was 5 added 7 g (69 mmol) of N-acetyl-guanidine. The mixture was refluxed for 24h. After removal of all the solvent, the residue was stirred with sat. aq. Na 2
CQ
3 (50 mL) for 5 min, filtered, and the solid was dried to provide 1 g (14%) of N-(4,5,6,7-tetrahydro-1H-benzoimidazol-2 yl)-acetamide. LCMS: 180 (M+l)*. IH NMR (DMSO-d 6 , 400 MHz): 5 2.38 (m, 4H), 2.00(s, 3H), 1.65 (in, 411) ppm. 10 To a solution of 150 mg (0.83 mmol) of N-(4,5,6,7-tetrahydro-H-benzoimidazol-2 yl)-acetamide in methanol (3 mL) was added water (3 nL) and cone. H 2
SO
4 (0.3 mL). The mixture was heated at 100 "C under microwave (150 w, 250 psi) for 1h. After removal of all the solvents, the residue was neutralized with sat. aq. Na 2
CO
3 solution then diluted with methanol (50 mL). After removal of the inorganic salts through filtration, the organic phase 15 was concentrated and dried under vacuum to afford 100 ing (86%) of the 4,5,6,7-tetrahydro 1 H-benzoimidazol-2-ylamine, which was used for the next step without further purification. To a solution of 70 ing (0.2 mmol) of 2-[(isoquinoline-3-carbony)-amino]- 3
H
benzoimidazole-4-carboxylic acid in DMF (2 mL) was added 100 mg (0.26 mmol) of HBTU and DIEA (0.3 mL). The mixture was stirred for 5 min, then 100 mg (0.7 mmol) of (4,5,6,7 20 tetrahydro-1H-benzoimidazol-2-ylamine was added. The resulting reaction mixture was heated at 80 "C for 1.5h. After usual workup, the residue was purified by column chromatography eluting with DCM/EtOAc (v/v= 1:1) to afford 12 mg (13 %) of isoquinoline 3-carboxylic acid [4-(4,5,6,7-tetrahydro-lH-benzoimidazol-2-ylcarbamoyl)-1H benzoimidazol-2-yl]-amide. LCMS: 452 (M+1)4. 25 Example 157 A mixture of 4.9 g (29.4 rmmol) of (1H-imidazol-4-yl)-acetic acid hydrochloride, 4 N HCI in dioxane (22.5 mL) and methanol (70 mL) was refluxed overnight Evaporation of the solvents gave (1H-imidazol-4-yl)-acetic acid methyl ester hydrochloride. LCMS: 141 (M+1)l. An aqueous solution (5.1 mL) of sodium nitrite (4.5 mmol, 311 mg) was cooled in an 30 ice bath and added to a mixture of 694 mg (4.5 mmol) of 4-amino-benzoic acid methyl ester in 2.36 N aq. HCl (7.5 mL) at 0 "C. The mixture was stirred at 0 "C for 30 min, and the reaction mixture was added to a mixture of 795 mg (4.5 mmol) of (1H-imidazol-4-yl)-acetic acid methyl ester hydrochloride in saturated aqueous sodium tetraborate solution (150 mL). 170 WO 2006/099379 PCT/US2006/009049 The mixture was stirred at 0 0 C for 1. 5 h and slowly warmed up to room temperature. The 4 (4-Methoxycarbonylmethyl-1H-imidazol-2-ylazo)-benzoic acid methyl ester was obtained after filtering and washing with water in a quantitative yield. LCMS: 303 (M+1)t A mixture of 4-(4-methoxycarbonylmethyl- 1 H-imidazol-2-ylazo)-benzoic acid 5 methyl ester (2 mnol, 604 mg) synthesized above, platinum (IV) oxide (60 mg) and methanol (50 mL) was hydrogenated (60 psi) for two days (another portion of PtO 2 was added after one day). Trifluoroacetic acid (TFA) (2 mmol, 0.15 mL) was added to the reaction mixture and it was filtered. The filtrate was concentrated and diluted with water, washed with ether (2 x 50 mL). Evaporation of the water from the aqueous solution gave the (2-Amino-1H-imidazol-4 10 yl)-acetic acid methyl ester as a TFA salt. (273.8 ng, 5 1%). LCMS: 156 (M41)'. 2-({2-[(Isoquinoline-3-carbonyl)-amino] -1 H-benzoimidazole-4-c arbonyl } -amino) 3H-imidazol-4-yl] -acetic acid methyl ester was synthesized according to the general procedure A using 339 mg (1.02 mmol) of 2-[(isoquino1ine-3-carbonyl)-amino]-iH benzoimidazole-4-carboxylic acid, 1.02 mmol of (2-amino-1H-imidazol-4-yl)-acetic acid 15 methyl ester trifluoro-acetic acid, 387 mg (1.02 mmol) of HBTU, 3 mL of DMF and 0.704 mL (4 mmol) of DIEA heated at 80 'C for 2 h. Purification by column chromatography gave 234 mg (0.50 nmmol, yield 49%) of the desired compound. LCMS: 470 (M+)t. Example 158 [2-({2-[(Isoquinoline-3-carbonyl)-amino]-1H-benzoimidazole-4-carbonyl}-amino) 20 3H--imidazol-4-yl]-acetic acid was synthesized according to general Procedure B from 225 mg (0.479 mmol) of 2-((2-[(isoquinoline-3-carbony)-amino]-1H-benzoimidazole- 4 carbonyl}-amino)-3H-imidazol-4-yll-acetic acid methyl ester, 0.5 mL of 2 N aq. LiOH, 2 mL of THF and 0.5 mL of methanol stirring at room temperature for 3 h. 116.9 mg (54%) of the product was obtained. LCMS: 456 (M+1)t. 25 Example 159 Isoquinoline-3 -carboxylic acid (4- {5 -[(methoxy-methyl-carbamoyl)-methyl]-1
H
imidazol-2-ylcarbamoyl} -IH-benzoinidazol-2-yl)-amide was synthesized according to the general procedure A from 109 mg (0.239 mmol) of 2-({2-{(isoquinoline-3-carbonyl)-amino] 1H-benzoimidazole-4-carbonyl} -amino)-3H-imidazol-4-yl]-acetic acid, 50 mg (0.5 mmol) of 30 0,N-dimethyl-hydroxylamine hydrochloride, 91 mg (0.24 mnol) of IBTU, 0.176 mL (1 mmol) of DIEA and 1 mL DMF stirring at room temperature for 1 h. 64.4 mg (54%) of the product was obtained. LCMS: 499 (M+1 )t 171 WO 2006/099379 PCT/US2006009049 By analogous methods to those used to prepare Examples 155 to 159 and those in the relevant above schemes, the following compounds were synthesized. H o N, N H N N H 0 N 5 LCMS Ex. W(M + 1)+ 160 5-Isopropyl-1H-imidazol-2-yl 440 161 4-fluoro-phenyl 426 162 1-(4-methanesulfonyl-pheny1)-ethyl 514 163 3-methanesulfonyl-phenyl 486 164 4-trifluoromethoxy-benzyl 506 165 4-methoxy-benzyl 452 166 4-methyl-benzyl 436 167 4-fluoro-benzyl 440 168 4-chloro-benzyl 456 169 3-trifluoromethyl-benzyl 490 170 3-fluoro-4-trifluoromethyl-benzyl 508 171 3,5-difluoro-benzyl 458 172 4-cyano-benzyl 447 173 N'(4,5-dihydro-IH-imidazol-2-yl)-hydrazino 415 174 3-methoxy-benzyl 452 175 4-methoxy-phenyl 438 176 2-(4-methoxy-phenyl)-ethyl 466 177 3-fluoro-benzyl 440 178 4-sulfamoyl-benzyl 501 179 2-inethoxy-benzyl 452 172 WO 2006/099379 PCT/US2006/009049 180 2-ethoxy-benzyl 466 181 2,2-dioxo-1,3-dihydrobezo[cjthiophen-5-y 498 182 2-(4-phenoxy-phenyl)-ethyl 528 183 butyl 388 184 3H-imidazo[4,5-cpyridin-2-y 449 185 7H-purin-8-y 450 186 isoquinolin-6-yl 459 187 2-methylsulfanyl-ethyl 406 188 5,5-dimethyl-7-oxo-4,5,6,7-tetrahydro-benzothiazol- 2 -yl 511 189 1H-benzoimidazol-5-yl 448 190 1H-pyrazol-3-yl 398 191 2-amino-pyrimidin-4-yl 425 192 4-tert-butyl-1H-imidazol-2-yl 454 193 4,5-dimethyl-1H-imidazol-2-yl 426 194 5-ethyl-1H-imidazol-2-yl 426 195 5-adamantan-1-yl-lH-imidazol-2-yl 532 196 5-benzyl-1H-imidazol-2-yl 488 Example 197 To a solution of 4 g (20 mmol) of 4-oxo-piperidine-l-carboxylic acid tert-butyl ester in THF (60 mL) was added 10 g (20 mmol) of pyrrolidone hydrotribromide. The reaction 5 mixture was refluxed for 5 min, after cooling to room temperature, the solid (pyrrolidone HBr salt) was filtered off, the organic layer was concentrated and used for the next step without further purification. The above crude material was dissolved in ethanol (50 mL), and 2.2 g (30 mmol) of thiourea was added. The mixture was refluxed for 3h, and the solvent was evaporated under 10 reduced pressure. The residue was dissolved in methanol (50 mL), then 4 M HGI solution in 1,4-dioxane (24 mL) was added. The mixtures were refluxed for 5 min. After removal of all solvent, the residue was dissolved in DCM (20 mL), and hexane (20 mL) was added. The resulting solid was filtered and dried to afford 4,5,6,7-tetrahydro-thiazolo [5,4-c] pyridin-2 ylamine HC1 salt which was then dissolved in dioxane (25 mL) and sodium carbonate 15 solution (1M, 25 mL). To this stirring solution, 2.2 g (10 mmol) of di-tert-butyl dicarbonate was added. Then the reaction mixture was stirred at room temperature for 2 h., extracted with 173 WO 2006/099379 PCT/US20061009049 ethyl acetate, washed with water and brine. The organic layer was dried over magnesium sulfate, concentrated under reduced pressure and purified by column chromatography eluting with hexanes/ ethyl acetate (v/v = 1:1 then 1:2) to give 1.2 g (24 %, 3 steps) of 2-amino-6, 7 dihydro-4H-thiazolo [5,4-c] pyridine-5-carboxylic acid tert-butyl ester. H NMR (DMSO-d 6 , 5 400 MHz): 5 6.82 (bs, 2H), 4.29 (s, 2H), 3.58 (t, 2H), 2.41 (t, 2H), 1.41 (s, 9H) ppm. To a solution of 0.25 g (0.75 mmol) of 2-[(isoquinoline-3-carbonyl)-amino]- 3
H
benzoimidazole-4-carboxylic acid in DMF (4 nL) was added 0.4 g (1.05 mmol) of HBTU and DIEA (1 mL). The mixture was stirred for 5 min and then 0.18 g (0.7 mmol) of 2-amino 6,7-dihydro-4H-thiazolo[5,4-c]pyridine-5-carboxylic acid tert-butyl ester was added. The 10 resulting reaction mixture was heated at 80 "C for 2h. After usual workup, the residue was purified by column chromatography to afford the 0.2 g (55%) of 2-({2-[(isoquinoline-3 carbonyl)-amino]-3H-benzoimidazole-4-carbonyl)-amino)-6,7-dihydro-4H-thiazolo[5,4 c]pyridine-5-carboxylic acid tert-butyl ester. LCMS: 570 (M+1). 'H NMR (DMSO-d 3 , 400 MHz): 5 9.56 (s, 1H), 8.84 (s, 1H), 8.35 (m, 2H), 7.96 (in, 4H), 7.84 (d, 1H), 7.35 (t, 1H), 15 4.58 (bs, 2H), 3.70 (t, 2H), 2.73 (t, 2H), 1.44 (s, 911) ppm. Example 198 To a solution of 0.2 g (0.35 mmol) of 2-({2-[(isoquinoline-3-carbonyl)-amino]- 3
H
benzoimidazole-4-carbonyl}-amino)-6,7-dihydro-4H-thiazolo[5,4-c]pyridine-5-carboxylic acid tert-butyl ester in methanol (inL) was added HCI solution (5 mL, 4M HCl in 1,4 20 dioxane). The mixture was stirred at room temperature for 30 min. The conversion was checked by LCMS. After removal of the organic phase, the residue was diluted with DCM (50 mL) and the organic phase was washed with sat. aq. Na2CO 3 solution, water, brine, dried over Na 2
SO
4 and concentrated to afford 160 mg (100%) of isoquinoline-3-carboxylic acid [7 (4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-ylcarbamoyl)- 1H-bcnzoimidazol-2-yl]-amide. 25 LCMS: 470 (M+1) Example 199 To a solution of 20 mg (0.042 mmol) of isoquinoline-3-carboxylic acid [7-(4,5,6,7 tetrahydro-thiazolo [5,4-c]pyridin-2-ylcarbamoyl)-1H-benzoimidazol-2-yl]-amide (above example) in DCE (3 mL) was added 0.2 g (0.94 mmol) of sodium triacetoxyborohydride and 30 acetaldehyde (0.1 mL). The mixture was stirred at room temperature over night. After LCMS showed that the starting material was completely consumed, the reaction was quenched by addition of methanol (2 nL). After usual workup, the residue was dissolved in DCM (1 mL). The desired product crashed out by addition of hexanes and 12 mg (57%) of 174 WO 2006/099379 PCT/US2006/009049 isoquinoline-3-carboxylic acid [7-(5-ethyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2 ylcarbanoyl)- lH-benzoimidazol-2-yl]-amidewas collected after filtration. LCMS: 498 (M+l)t. Example 200 5 To a solution of 25 mg (0.05 mmol) of isoquinoline-3-carboxylic acid [7-(4,5,6,7 tetrahydro-thiazolo [5,4-c] pyridin-2-ylcarbanoyl)-IH-benzoimidazol-2-yl]-amide (Example 198) in pyridine (2 mL) was added 21.3 mg (0.15 imol) of 1-propanesulfonyl chloride. The mixture was stirred at room temperature for 12 h, extracted with ethyl acetate, washed with IN HCl, water and brine. The organic layer was dried over magnesium sulfate, evaporated 10 under reduced pressure. The crude product was dissolved in DCM (0.5 mL), hexane was added (4 ml) and the resulting precipitate was filtered and dried to provide 12 mg (57%) of isoquinoline-3-carboxylic acid [7-(5-propanyl- 1 -sulfonly-4,5,6,7-tetrahydro-thiazolo[5, 4 cJpyridini-2-ylcarbanoyl)-1H-benzoimidazol-2-yl]-ainide. LCMS: 576(M+). Example 201 15 To a solution of 20 mg (0.042 mmol) of isoquinoline-3-carboxylic acid [7-(4,5,6,7 tetrahydro-thiazolo [5,4-c]pyridin-2-ylcarbamoyl)-1H-benzoimidazol-2-yl]-amide (Example 198) in TIF (2 mL) was added 0.1 mL of 2-isocyanato-2-methyl-propane and DIEA(0.2 mL). The mixture was stirred at room temperature for 30 min. After LCMS showed that the starting material was completely consumed, the reaction mixture was diluted with ethyl 20 acetate (20 mL). The organic phase was washed with sat. aq. Na 2
CO
3 solution, dried over Na 2
SO
4 , and concentrated under reduced pressure. The crude product was dissolved in DCM (0.5 mL), hexane was added (4 ml) and the resulting precipitate was filtered and dried to provide 11 mg (46 %) of isoquinoline-3-carboxylic acid [4-(5-tert-butylcarbamoyl-4,5,6,7 tetrahydro-thiazolo[5,4clpyridin-2-ylcarbamoyl)-1H-benzoimidazol-2-yl]-anide. LCMS: 25 569 (M+1) 4 . Example 202 To a solution of 30 mg (0.063 mmol) of isoquinoline-3-carboxylic acid [7-(4,5,6,7 tetrahydro-thiazolo {5,4-c]pyridin-2-ylcarbainoyl)-1H-benzoimidazol-2-yl]-amide (Example 198) in DCM (2 mL) was added pyridine (0.5 mL) and benzoyl chloride (0.05 mL). The 30 mixture was stirred at room temperature overnight. The reaction mixture was diluted with ethyl acetate (20 mL), the organic phase was washed with sat. aq. Na 2
CO
3 solution, dried over Na 2
SO
4 . and concentrated under reduced pressure. The crude product was dissolved in DCM (0.5 mL), hexane was added (4 mL), and the resulting precipitate was filtered and dried 175 WO 2006/099379 PCT/US2006/009049 to provide 17 mg (47 %) of isoquinoline-3-carboxylic acid [4-(5-benzoyl-4,5,6,7-tetrahydro thiazolo[5,4-c]pyridin-2-ylcarbamoyl)-1H-benzoimidazol-2-yll-amide. LCMS: 574 (M+1)*. Example 203 To a solution of 30 mg (0.064 mmol) of isoquinoline-3-carboxylic acid [7-(4,5,6,7 5 tetrahydro-thiazolo [5,4-c]pyridin-2-ylcarbamoyl)-IH-benzoimidazol-2-yl]-amide (Example 198) in DCE (0.7 mL) was added 3-methylsulfanyl-propionaldehyde (0.1 mL), 0.2 g (0.94 mamol) of sodium triacetoxyborohydride, the mixture was stirred at room temperature for I h. The reaction was quenched by addition of methanol (0.5 mL). After usual workup, the reaction was quenched by addition of methanol (2 mL). After usual workup, the residue was 10 dissolved in DCM (1 mL) and the desired product was precipitated by addition of hexanes. 21 mg (58%) of isoquinoline-3-carboxylic acid {4-{5-(3-methylsnlfanyl-propyl)- 4 ,5, 6
,
7 tetrahydro-thiazolo{5,4-c]pyridin-2-ylcarbamoyl]- 1 H-benzoimidazol-2-yl}-amide was collected after filtration. LCMS: 558 (M+1)*. Example 204 15 To a solution of 20 mg (0.037 mmol) of thioether (Example 203) in DCM (2.5 mL) was added 25 mag (0.14 mmol) of m-CPBA. The reaction was stirred at room temperature for 2h. The solvent was evaporated under reduced pressure. The crude product was dissolved in DCM (0.5 mL), hexane was added (4 mL), and the resulting precipitate was filtered and dried to provide 11 rag (47 %) of isoquinoline-3-carboxylic acid {4-[5-(3-methanesulfonyl-propyl) 20 4,5,6,7-tetrahydro-thiazolo[5,4-clpyridin-2-ylcarbamoyl]--H-benzoimidazol-2-yl}-amide. LCMS: 590 (M+1)+ By analogous methods to those used to prepare Examples 197 to 204 and those in the relevant above schemes, the following compounds were synthesized. H o N.s N H N 25 H 0 N 176 WO 2006/099379 PCT/US2006/009049 LCMS (M + 1)+ 205 piperidin-4-yl-1-carboxylic acid tert-butyl ester 515 206 piperidin-4-yl 488 207 piperidin-4-yl-1-carboxylic acid isopropyl ester 501 208 5-methanesulfonyl-4,5,6,7-tetrahydro-thiazolo[5,4-cpyridin- 2 -yl 548 209 5-benzyl-4,5,6,7-tetrahydro-thiazolo{5,4-c]pyridin- 2 -yl 560 210 4,5,6,7-tetrahydro-benzothiazol-2-yl 469 211 6-methy1-4,5,6,7-tetrahydro-benzothiazol- 2 -yl 483 212 5-methyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin- 2 -yl 484 213 5-cyclopentylmethyl-4,5,6,7-tetrahydro-thiazolO[5,4-c]pyridin-2-y 662 214 5-pyridin-3-ylmethyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-yl 707 215 5-cyclohexylmethyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-Yl 676 216 5-furan-3-ylmethyl-4,5,6,7-tetrahydro-thiazolo[5,4-c)pyridin-2-yl 660 217 piperidin-3-ylmethyl-1-carboxylic acid tert-butyl ester 529 218 piperidin-4-ylmethyl-1-carboxylic acid tert-butyl ester 529 219 piperidin-3-ylmethyl 538 220 piperidin-4-ylmethyl 538 221 5-(1-methyl-1H-pyrrol-2-ylmethyl)-4,5,6,7-tetrahydro-thiazolo[5,4- 563 c] pyridin-2-yl 222 5-propyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin- 2 -yl 621 223 5-(2-ethyl-buty1)-4,5,6,7-tetrahydro-thiazolo(5,4-c]pyridin-2-yl 664 224 5-isobutyl-4,5,6,7-tetrahydro-thiazolo[5,4-c~pyridin- 2 -yl 636 225 1 -cyclohexylmethyl-piperidin-3-ylnethyl 635 226 1-cyclohexyhmethyl-piperidin-4-ylmethyl 635 227 2-piperidin-3-yl-ethyl-1-carboxylic acid tert-butyl ester 543 228 2-piperidin-4-yl-ethyl-- -carboxylic acid tert-butyl ester 543 229 2-piperidin-3-yl-ethyl 552 230 2-piperidin-4-yl-ethyl 552 231 5-(2,4-dimethoxy-benzyl)-4,5,6,7-tetrahydro-thiazolo[5,4-c] 620 pyridin-2-yl 232 5-(4-chloro-benzyl)-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-yl 595 177 WO 2006/099379 PCT/US2006/009049 233 5-(4-fluoro-benzy1)-4,5,6,7-tetrahydro-thiazolo[5, 4 -]pyridin- 2 -yl 578 234 5-(4-isopropoxy-benzyl)-4,5,6,7-tetrahydro-thiazolo{5,4-c]pyridil- 618 2-yl 235 5-(2-methoxy-benzyl)-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2- 590 y1 236 5-(3-methoxy-benzy1)-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2- 590 y 1 237 5-(4-methoxy-benzyl) -4, 5,6,7-tetrahydro-thiazolo [5,4-c]pyridin-2- 590 23 -hA ty-,,,-erhdotizl[,-lyiii2y 7 239 5-thiophen-2-ylethyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2- 566 yl 240 5-thiophen-3-ylmethyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin- 2 566 241 5-(3-methyl-butyl)-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-y1 540 242 piperidin-3 S-ylmethyl-1 -carboxylic acid tert-butyl ester 529 243 piperidin-3R-ylmethyl-1-carboxylic acid tert-butyl ester 529 244 piperidin-3S-ylmethyl 429 245 piperidin-3R-ylmethyl 429 246 5-(3H-imidazol-4-yhnethyl)-4,5,6,7-tetrahydro-thiazolo[5,4- 550 c]pyridin-2-yl 247 5-(1-methyl-i1H-inidazol-2-ylmethyl)-4,5,6,7-tetrahydro- 564 thiazolo[5,4-c] pyridin-2-yl 5-(1H-imidazol-2-ylmethyl)-4,5,6,7-tetrahydro-thiazolo[5, 4 248 550 c]pyridin-2-y1 249 5-cyclopropylmethyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2-y1 524 5-(3,3-dimethyl-butyl)-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin- 2 250 554 251 1,2,3,4-tetrahydro-isoquinolin-6-yl-2-carboxylic acid tert-butyl 563 ester 252 1,2,3,4-tetrahydro-isoquinolin-6-y1 572 253 1 -cyclohexylmethyl-piperidin-3 S-ylmethyl 635 254 1 -cyclohexylmethyl-piperidin-3R-ylmethyl 635 178 WO 2006/099379 PCT/US2006/009049 255 5-thiazol-2-ylmethyl-4,5,6,7-tetrahydro-thiazolo[5,4-cjpyridin-2-y 567 256 5-(5-methyl-3H-imidazol-4-ylmethyl)-4,5,6,7-tetrahydro- 564 thiazolo[5,4-c] pyridin-2-yl 257 5-(2-ethyl-5-methyl-3H-imidazol-4-ylmethyl)-4,5,6,7-tetrahydro- 592 thiazolo[5,4-c]pyridin-2-yl 258 2S-[2-amino-6,7-dihydro-4H-thiazolo[5,4-cjpyridin-5-ylhnethyl]- 653 pyrrolidine-1-carboxylic acid tert-butyl ester 259 5-quinolin-3-ylmethyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin-2 611 yl 260 2S-{3-[amino-methyl]-piperidin-1-ylmethyl}-pyrrolidine-1- 612 carboxylic acid tert-butyl ester 2R-{3-[amino-methyl]-piperidin-1-ylmethyl}-pyrrolidine-1 261 carboxylic acid tert-butyl ester 612 262 5-pyrrolidin-2S-ylmethyl-4,5,6,7-tetrahydro-thiazolo[5,4-c]pyridin- 699 2 -yl 263 1-(1H-imidazol-2-ylmethyl)-piperidin-3-ylmethyl 509 264 1-(1-methyl-1H-imidazol-2-ylmethyl)-piperidin-3-ylmnethyl 523 265 1 -(1 H-imidazol-4-ylmethyl)-piperidin-3 -ylmethyl 509 Example 266 To a solution of 100 mg (0.3 mmol) of 2-[(isoquinoline-3-carbonyl)-amino]-3H benzoimidazole-4-carboxylic acid (from Example 58) in DMF (2 nL) was added 200 mg 5 (0.53 mmol) of HBTU and DIEA (0,5 mL). The mixture was stirred for 5 min then 140 mg (1 0 mmol) of N-(4-fluoro-benzyl)-N-methyl-amine was added. The resulted reaction mixture was heated at 70 "C for 1 h. After usual workup as described in general procedure A, the residue was purified by column chromatography eluting with DCM/EtOAc (v/v=1:1) to afford the 90 rug (66%) of isoquinoline-3-carboxylic acid {7-[(4-fluoro-benzyl)-methyl 10 carbamoyl]-1H-benzoimidazol-2-yl}-amide. LCMS: 454 (M+1)+. Example 267 Isoquinoline-3-carboxylic acid (4-dibenzylcarbamoyl-1H--benzoimidazol-2-yl)-anide (40 mg) was synthesized according to the general procedure described for the synthesis of 179 WO 2006/099379 PCT/US2006/009049 Example 266, with the exception that NN-dibenzylamine was used instead of N-(4 fluorobenzyl)-N-nethyl-amine. LCMS: 512 (M+1). Example 268 3.00 g (10 mmol) of tert-Butoxycarbonylamino-(dimethoxy-phosphoryl)-acetic acid 5 methyl ester was added to a solution containing 1.69 g (10 mmol) of 2-formyl-benzoic acid methyl ester and 1.51 mL (10 mmol) of DBU in 50 nL of dichloromethane at 0 C. The reaction was slowly warmed to room temperature overnight, then diluted with dichloromethane (100 mL), washed once with water (150 mL) and dried over sodium sulfate. Purification by column chromatography on silica gel gave 2.12 g (70%) of the product as 10 white solid. LCMS: 304 (M+1)+. 6 mL of 4 N HCl in dioxane (6 mL) was added to a solution of 1-oxo-1lH isoquinoline-2,3-dicarboxylic acid 2-tert-butyl ester 3-methyl ester from the previous step and 10 mL of dichloromethane and stirred overnight. The reaction mixture was concentrated and partitioned between ethyl acetate (100 nL) and saturated NaHCO 3 solution (100 mL). 15 The aqueous layer was separated and extracted again with ethyl acetate (100 mL). The combined organic layers were dried over sodium sulfate. Evaporation of solvents in vacuo gave 1.39 g (98%) of the 1-oxo-1,2-dihydro-isoquinoline-3-dicarboxylic acid methyl ester as a white solid. LCMS: 204 (M+ I ). 663 mg (81%) of 1-Oxo-1,2-dihydro-isoquinoline-3-dicarboxylic acid was 20 synthesized according to General Procedure B using above synthesized 877 mg (4.32 mmol) of 1-oxo-1,2-dihydro-isoquinoline-3-dicarboxylic acid methyl ester, 4.3 mL of 2 N aq. LiOH, 16 mL of THF and 4 mL of methanol stirring at r.t. overnight. LCMS: 190 (M+1t 76 mg (0.4 mmol) of 1-oxo-1,2-dihydro-isoquinoline-3-dicarboxylic acid was reacted with 73 mg (0.3 mmol) of intermediate B (2-amino-3H-benzoimidazole-4-carboxylic acid 25 (IH-imidazol-2-yl)-amide), 152 mg (0-4 mmol) of HBTU, 0.176 mL (1 mumol) of DIEA and 1.2 mL of DMF stirring at 85'C for 2 h according to general Procedure A 54.5 mg (0.132 mmol, 44%) of 1-oxo-1,2-dihydro-isoquinoline-3-carboxylic acid [4-(lIH-imidazol-2 ylcarbamoyl)-1H-benzoimidazol-2-yl]-amide was obtained. LCMS: 414 (M+tI-. Example 269 30 6-(4-Methoxycarbonyl-1H-benzoimidazol-2-ylcarbamoyl)-3,4-dihydro-1H isoquinoline-2-carboxylic acid tert-butyl ester (114 mg, 90%) was synthesized according to General Procedure A using 798 mg (2.82 nmol) of 3,4-dihydro-IH-isoquinoline-2,6 dicarboxylic acid 2-tert-butyl ester, 540 mug (2.82 mmol) of intermediate A (2-amino-iH 180 WO 2006/099379 PCTfUS2006/009049 benzoimidazole-4-carboxylic acid methyl ester), 1.28 g (3.38 mmol) of HBTU, 0.992 mL (5.64 mmol) of DIEA and 15 nL of DMF stirring at room temperature overnight. LCMS: 451 (M+1)#. Example 270 5 109 ing (0.242 mmol) of 6-(4-methoxycarbonyl-1H-benzoimidazol-2-ylcarbamoyl) 3,4-dihydro-IH-isoquinoline-2-carboxylic acid tert-butyl ester was hydrolyzed using I mL of 2 N aq. LiOH, 4 mL of THF and I mL of methanol stirring at room temperature for one day as described in general procedure B. 76.5 mg (72%) of the 6-(4-Carboxy-1H-benzoimidazol 2-ylcarbamoyl)-3,4-dihydro- I 1H-isoquinoline-2-carboxylic acid tert-butyl ester product was 10 obtained. LCMS: 437 (M+1)+ 72.9 mg (0.167 nnol) of above acid was reacted with 47 mg (0.35 mmol) of 2 aminoimidazole sulfate, 95 mg (0.25 mmol) of HBTU, 0.176 mL (1 mmol) of DIEA and I mL DMF. The mixture was stirred at 80CC for 18 h according to General Procedure A and gave 54.7 mg (65%) 6-[4-(1 H-imidazol-2-y1carbanoyl)-lH-benzoimidazol- 2 -ylcarbamoyl] 15 3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester. LCMS: 502 (M+1)+ Example 271 49.8 mg (0.10 mmol) of 6-{4-(l H-imidazol-2-ylcarbamoyl)- IH-benzoimidazol-2 ylcarbamoyl]-3,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester in 2 mL of 4 N HCI in dioxane was stirred at r.t. for 2 h. Evaporation of solvents in vacuo gave 1,2,3,4 20 tetrahydro-isoquinoline-6-carboxylic acid [4-(1 H-imidazol-2-ylcarbamoyl)- 1H benzoimidazol-2-yl]-aniide in quantitative yield. LCMS: 402 (M+1)t Example 272 25 mg (0.049 mmol) of 1,2,3,4-tetrahydro-isoquinoline-6-carboxylic acid [4-(1H imidazol-2-ylcarbamoyl)-1H-benzoimidazol-2-yl]-anide, 0.01 mL of (0.07 mmol) 25 benzenesulfonyl chloride, and 0.07 mL of (0.5 nmol) triethylamine in 0.5 mL DMF was stirred at room temperature for 1 h. The reaction mixture was then treated with 0.024 mL of hydrazine hydrate and diluted with water. The precipitated solid was filtered and washed with water to give 2-benzenesulfonyt-1,2,3,4-tetrahydro-isoquinoline-6-carboxylic acid [4 (lH-imidazol-2-ylcarbamoyl)-lH-benzoimidazol-2-yl]-amide (17.2 mg, 65%). LCMS: 542 30 (M+1). Example 273 A mixture of 25 mg (0.049 nnol) of 1,2,3,4-tetrahydro-isoquinoline-6-carboxylic acid [4-(1 H-imidazol-2-ylcarbamoyl)- 1 H-benzoimidazol-2-yl] -amide, 0.0152 mL (0.15 mmol) 181 WO 2006/099379 PCT/US2006/009049 of benzaldehyde and 0.6 mL of DMF was stirred for 10 min. 67 mg (0.3 mmol) of sodium triacetoxyborohydride was added and the reaction mixture was stirred at room temperature for 24 h. The reaction mixture was partitioned between dichloromethane (10 mL) and water (10 mL). The organic layer was separated and dried over sodium sulfate. Purification by 5 column chromatography on silica gel gave 11.7 ing (49%) of 2-benzyl-1,2,3,4-tetrahydro isoquinoline-6-carboxylic acid [4-(1H-imidazol-2-ylcarbamoyl)-1 H-benzoimidazol-2-yl} amide. LCMS: 492 (M+l)*. Example 274 41 mg (0.08 mmol) of 1,2,3,4-tetrahydro-isoquinoline-6-arboxylic acid [4-(IH 10 imidazol-2-ylcarbamoyl)-1H-benzoimidazol-2-yl]-amide in 0.6 mL of DMF and 0.176 mL (1 mmol) of DIEA was treated with 0.024 mL (0.3 mmol) of methyl chloroformate and stirred at room temperature for 14 h. The reaction mixture was then treated with 0.15 mL of 2 N aq. LiOH. Water was added to the reaction mixture. The precipitated solid was collected by filtration and washed with water to yield 11.4 mg (0.0248 nmol, 31%) of 6-{4-( lH-imidazol 15 2-ylcarbamoyl)-lH-benzoimidazol-2-ylcarbanoyl-3,4-dihydro-1H-isoquinoline-2 carboxylic acid methyl ester. LCMS: 460 (M+1)-. By analogous methods to those used to prepare Examples 269 to 274 and those in the relevant above schemes, the following compounds were synthesized. H-N N O N, N H \-N N Ar 20 H 0 W LCMS Ex. (M ± 1) 275 2-Metbanesulfonyl-1,2,3,4-tetrahydro-isoquinoline-6-yl 480 276 2-Ethoxycarbonyl-3,4-dihydro- 1H-isoquinoline-6-yl 474 277 2-Propoxycarbonyl-3,4-dihydro-lH-isoquinoline-6-yl 488 278 2-Isobutoxycarbonyl-3,4-dihydro- 1 H-isoquinoline-6-yl 502 182 WO 2006/099379 PCT/US2006/009049 279 2-Isopropoxycarbonyl-3,4-dihydro-1H-isoquinoline-6-y1 488 280 2-Butoxycarbonyl-3,4-dihydro-1H-isoquinoline-6-yl 502 281 (3S)-2-tert-Butoxycarbonyl-3,4-dihydro-IH-isoquinoline-3-yl 502 282 (3S)-1,2,3,4-Tetrahydro-isoquinoline-3-y 511 283 (3S)-2-Methanesulfonyl-1,2,3,4-tetrahydro-isoquinoline-3-y 480 284 (3S)-2-Ethyl-1,2,3,4-tetrahydro-isoquinoline-3-yl 539 285 (3S)-2-tert-Butoxycarbonyl-1,3,4,9-tetrahydro-beta-carboline-3-yl 541 286 (3S)-2-tert-Butoxycarbonyl-1-phenyl-1,3,4,9-tetrahydro-beta-carboline- 617 3 -yl 287 2,3,4,9-Tetrahydro-1H-beta-carboline-3-y1 550 Example 288 To a stirring solution of 5.0 g (27.5 mmol) of 2-(3,4-dimethoxy-phenyl)-ethylamine was added 3.7 g (27.5 mmol) of ethyl oxalyl chloride at 0 0 C. The reaction mixture was 5 warmed to r.t. and stirred for 1.0 h and 100 mL of DCM was added. The solvent was washed with 1.0 N HCi, water and brine, dried over the magnesium sulfate, and evaporated under reduced pressure to afford 7.76 g (100%) of the N-[2-(3,4-dimethoxy-phenyl)-ethyl] oxalamic acid ethyl ester which was used for the next step without further purification. LCMS: 282 (M+i). 10 To a refluxing solution 5.0 g (17.7 mmol) of above oxalamide in 100 mL of CH 3 CN was added 20 mL of POC1 3 . After 12 h at reflux, the reaction mixture was poured onto ice and extracted 3 times with ether (3 X 50 mL). The ether extracts were washed twice with I N HCL. The combined aqueous extracts were brought to pH 8 with solid NaHCO 3 . The aqueous extracts were then washed five times with EtOAc (5 x 50 nL). The combined organic layers 15 were dried over magnesium sulfate and evaporated under reduced pressure to provide 3.2 g (69%) of 6,7-dimethoxy-3,4-dihydro-isoquinoline-1-carboxylic acid ethyl ester. LCMS: 264 (M+i)* A mixture of 0.26 g (1.0 mmol) of the above imine and 10% Pd-C in 5.0 mL of EtOH was stirred at r.t. under hydrogen (1 atm). After 5 h, the reaction mixture was filtered and 20 evaporated to give a residue that was dissolved in DCM (5.0 mL) containing 0.2 g (2.0 mmol) of triethyl amine. To this stirring solution at r.t., 0.24 g (1.1 nmol) of di-tert-butyl dicarbonate was added and stirring continued for another 2 h. The reaction mixture was extracted with DCM (10 mL), washed with water, brine, dried over magnesium sulfate, and 183 WO 2006/099379 PCT/US2006/009049 evaporated to provide 0.35 g (97%) of the 6,7-dimethoxy-3,4-dihydro-IH-isoquinoline-1,2 dicarboxylic acid 2-tert-butyl ester 1-ethyl ester. LCMS: 366 (M + 1)+ To a stirring solution of 0.35 g (0.95 nnnol) of above ester in a mixture of THF (10 mL) and methanol (3.0 mL) was added 3.0 mL of 2N LiOH at r.t. The reaction mixture was 5 stirred at r.t. for 45 min and neutralized (pH = 6.0) with 2.0 N HCl and extracted with ethyl acetate. The organic layer was washed with water and brine, dried over magnesium sulfate, and evaporated under reduced pressure to afford 0.3 g (90%) of the 6,7-dimethoxy-3,4 dihydro-1H-isoquinoline-1,2-dicarboxylic acid 2-tert.-butyl ester. LCMS: 338 (M + 1).
0.25 g (0.74 mmol) of above acid, 0.30 g (0.81 mmol) of HBTU in a mixture of 4.0 10 ml of DMF and 0.5 mL of DIEA was reacted with 0.18 g (0.74 mmol) of 2-Ainino-3H benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide (Intermediate B) as described in general procedure A. The reaction mixture was cooled to rt. 5.0 mL of water and 5.0 mL of saturated NaHCO 3 was added. The resulting solid was filtered and the solid was washed with water, ether and dried to provide 0.2 g (48%) of 1-[4-(lH-iniidazol-2-ylcarbamoyl)-lH 15 benzoimidazol-2-ylcarbamoyl]-6,7-diniethoxy- 3 ,4-dihydro-1H-isoquinoline-2-carboxylic acid tert-butyl ester. LCMS: 562 (M+1)*. Example 289 To a refluxing solution of 2.0 g (8.0 nunol) of 6,7-dimethoxy-3,4-dihydro isoquinoline-1-carboxylic acid ethyl ester was added to 2.0 g of 10 % Pd-C. After 12 h at 20 reflux, the reaction mixture was cooled, filtered through celite, and evaporated under reduced pressure to provide 2.0 g (98%) of 6,7-dimethoxy-isoquinoline-1 -carboxylic acid ethyl ester. LCMS: 262 (M+1) 4 . To a stirring solution of 0.35 g (1.3 mmol) of above ester in a mixture of THF (10 mL) and methanol (3.0 mL) was added 3.0 mL of 2N LiOH at r.t. The reaction mixture was stirred 25 at r.t. for 45 rmin and neutralized (pH = 6.0) with 2.0 N HC1 and extracted with ethyl acetate. The organic layer was washed with water, brine and dried over magnesium sulfate, evaporated the solvent under reduced pressure to afford 0.3 g (96%) of the 6,7-Dimethoxy isoquinoline-1-carboxylic acid. LCMS: 234 (M + 1)*. 0.1 g (0.43 mmol) of above acid, 0.17 g (0.47 mmol) of HBTU in a mixture of 2.0 ml 30 of DMF and 0.24 mL of DIEA was reacted with 0.10 g (0.43 mmol) of 2-Amino-3H benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-ainide (Intennediate B) as described in general procedure A. The reaction mixture was cooled to r.t. 5.0 mL of water, 5.0 mL of saturated NaHCO 3 was added and the resulting solid was filtered and the solid was washed with water, ether and dried to provide 0.005 g (2.6 %) of 6,7-dimethoxy-isoquinoline-I 184 WO 2006/099379 PCT/US2006/009049 carboxylic acid [4-(1H-imidazol-2-ylcarbamoyl)-IH-benzoinidazol-2-y1]-amide. LCMS: 458 (M+1)*. 'H NMR (DMSO-d 6 , 400 MHz): 5 8.86 (s, 1H), 8.57 (d, 1H), 8.11 (d, 1H), 7.96 (d, 1H), 7.88 (d, 1H), 7.57 (s, 1H), 7.38(m, 311), 3.98 (s, 3H, 3.97 (s, 311) ppm. Example 290 5 To a solution of 2.1 g (10 nnol) of 1,2,3,4-Tetrahydro-isoquinolie-3-caboxylic acid HCI salt (2.1 g, 10 mmol) in cone. H 2 S0 4 (15 mL) was added HN0 3 (70%, 2 mL) dropwise at -10 "C. The mixture was stirred at -10 *C for 1.5 h then poured into ice. The reaction mixture was neutralized with conc. NH 4 0H. The white precipitate was collected after filtration, dried under vacuum, dissolved in methanol (50 mL), and 4M HC1 in dioxane was 10 added. After refluxing overnight, the solvent was evaporated under reduced pressure to provide 1.5 g (63%) of the 7-nitro-1,2,3,4-tetrahydro-isoquinoliine-3-carboxylic acid methyl ester. To a solution of the above ester in 1,4-dioxane (100 mL) was added 2.8 g (12.7 mmol) of DDQ, and the reaction mixture was refluxed for 6 h. After removal of solvents, the 15 residue was dissolved with ethyl acetate (250 mL) and washed with aq. NaHSO 3 solution (1M solution, 100 mL), water and brine. The ethyl acetate layer was dried over sodium sulfate, evaporated under reduced pressure and purified by column chromatography elutingg with ethyl acetate then EtOAc/methanol v/v=100:5) to provide 1.0 g (71%) of the 7-nitro isoquinoline-3-carboxylic acid methyl ester. LCMS: 233 (M+1). The methyl ester was 20 hydrolyzed according to the general procedure B to give 0.9 of 7-nitro-isoquinoline- 3 carboxylic acid. LCMS: 219 (M+1)*. To a solution of 44 mg (0.2 mmol) of 7-nitro-isoquinoline-3-carboxylic acid in DMF (1 mL) was added 113 nig (0.3 mmol) of HBTU and DIEA (0.3 mL). The mixture was stirred for 10 min and then 40 mg (0.16 mmol) of 2-amino-3H-benzoimidazole-4-carboxylic 25 acid (1 H-imidazol-2-yl)-amide (intermediate B) was added. The reaction was heated at 80 *C for lh. After the usual work up, the residue was purified by column chromatography eluting with DCM/methanol (v/v-10:1) to afford 8 mg (9%) of 7-nitro-isoquinoline-3-carboxylic acid [7-(1H-imidazol-2-ylcarbamoyl)-1IH-benzoimidazol-2-yl]-amide. LCMS: 443 (M+1)+. Example 291 30 0.5 g (2.1 mmol) of 7-nitro-isoquinoline-3-carboxylic acid methyl ester in methanol (25 ml) was hydrogenated according to the general procedure C to provide 0.42 g of 7-amino isoquinoline-3-carboxylic acid methyl ester in quantitative yield. LCMS: 203 (M+1)I 185 WO 2006/099379 PCT/US2006/009049 To a solution of 0,42 g (2.1 mmol) of above amine in pyridine (2 mL) was added methanesulfonyl chloride (0.2 mL) at 0 C. The reaction mixture was stirred at 0 "C for 30 min, warmed to r.t., and extracted with ethyl acetate (20 mL). The organic phase was washed with I N HCI, water, dried over Na 2 SO4, and concentrated. The residue was purified by 5 column chromatography eluting with ethyl acetate to afford the 0.35 g (59 %) of 7 methanesulfonylamino-isoquinoline- 3 -carboxylic acid methyl ester. The methyl ester was hydrolyzed according to the general procedure B to give 0.3 g (90%) of 7 nethanesulfonylamino-isoquinoline- 3 -carboxylic acid. LCMS: 267 (M+1)*. To a solution of 30 mg (0.11 mmol) of 7-methanesulfonylaino-isoquinoline- 3 10 carboxylic acid in DMF (0.8 mL) was added 60 mg (0.15 mmol) of HBTU and DIEA (0.1 mL). The mixture was stirred for 10 min and then 24 mg (0.1 mmol) of 2-amino-3H benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide (intermediate B) was added. The reaction was heated at 80 C for 1h. After usual work up, the residue was purified by column chromatography eluting with DCM/methanol (v/v-- 10:1 to 8:1) to afford 5 mg (9%) 15 of 7-methanesulfonylamino-isoquinoline- 3 -carboxylic acid [7-(lH-imidazol-2-ylcarbamoyl) 1H-benzoimidazol-2-yl]-amide. LCMS: 491 (M+1)-. Example 292 To a stirring solution of 2.0 g (6.8 mmol) of Boc-7-hydroxy-tetrahydroisoquilnoline -3 carboxylic acid in a mixture of DCM (20 mL) and DIEA (2.0 mL) was added 1.93 g (13.7 20 mmol) of iodomethane at r.t. The reaction mixture was stirred at r.t. for 6.0 h. DCM (20 nL) was added, and the solvent was washed with water, brine, dried over magnesium sulfate, and evaporated under reduced pressure to provide 2.09 g (100%) of 7-hydroxy-3,4-dihydro-1
H
isoquinoline-2,3-dicarboxylic acid 2-tert-butyl ester 3-methyl ester which was used for the next step without further purification. LCMS: 308 (M + 1)*. 25 2.09 g (6.8 mmol) of above ester, 1.28 g (0.89 mmol) of benzyl bromide and 1.38 g (10.2 nmnol) of K 2 C0 3 in DMF (30 mL) was stirred at 70 aC for overnight and extracted with ethyl acetate (50 mL). The organic layer was washed with water and brine, dried over magnesium sulfate, and evaporated under reduced pressure to provide 2.2 g (81 %) 7 benzyloxy-3,4-dihydro-1H-isoquinoline-2,3-dicarboxylic acid 2-tert-butyl ester 3-methyl 30 ester. LCMS: 398 (M + 1)+. 2.2 g (5.5 mmol) of the above ester was dissolved in 4.0 M HCl in dioxane (20 ml) and stirred for 1.0 h at r.t. The solvent was evaporated under vacuum, and the residue was dissolved in toluene (20 mL). To this stirring solution 2.5 g (11.0 mmol) of DDQ was added and heated to reflux for 30 min. The reaction mixture was cooled, and the precipitate was 186 WO 2006/099379 PCT/US20061009049 filtered. The filtrate was washed with sat. NaHSO 3 (25 mL), water and brine. The organic layer was dried over magnesium sulfate, evaporated under reduced pressure and purified by column chromatography using 20% ethyl acetate in hexane as an eluent to afford 1 .0 g (62.5%) of 7-benzyloxy-isoquinoline-3-carboxylic acid methyl ester. LCMS: 294 (M + I)-. 5 To a stirring solution of 1.0 g (3.4 mmol) of above ester in a mixture of THF (30 mL) and methanol (10 mL) was added 10 mL of 2N LiOH at r.t. The reaction mixture was stirred at r.t. for 45 min., neutralized (pH = 6.0-7.0) with 2.0 N HCl, and extracted with ethyl acetate. The organic layer was washed with water and brine, dried over magnesium sulfate, and evaporated under reduced pressure to afford 0.8 g (8 1%) of the 7-benzyloxy-isoquinoline- 3 10 carboxylic acid. LCMS: 280 (M + 1)+. 0.70 g (2.5 mmol) of 7-benzyloxy-isoquinoline-3-carboxylic acid synthesized above, 1.0 g (2.7 mmol) of HBTU in a mixture of 7.5 ml.of DMF and 1.7 ml, of DIEA was reacted with 0.48 g (2.5 mmol) of 2-Amino-3H-benzoimidazole-4-carboxylic acid methyl ester (Intermediate A) as described in general procedure A. 2-[(7-Benzyloxy-isoquinoline- 3 15 carbonyl)-amino}-1H-benzoimidazole- 4 -carboxylic acid methyl ester (1.0 g, 88 %) was isolated after column chromatography using 1:1 ethyl acetate and hexane as an eluents. LCMS: 453 (M+l)7. Example 293 0.13 g (0.45 mmol) of 7-benzyloxy-isoquinoline-3-carboxylic acid 0.19 g (0.5 mmol) 20 of HBTU in a mixture of 2.0 ml of DMF and 0.26 mL of DIEA was reacted with 0.1 g (0.41 nnol) of 2-amino-3H-benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide (Intermediate B) as described in general procedure A. The reaction mixture was cooled to r.t., and 5.0 mL of water and 5.0 mL of saturated NaHCO 3 was added. The resulting solid was filtered, and the solid was washed with water and ether, and dried to provide 0.055 g (27%) 25 of 7-benzyloxy-isoquinoline-3-carboxylic acid [4-(IH-imidazol-2-ylcarbamoyl)-IH benzoimidazol-2-1]-amide. LCMS: 504 (M+1). Example 294 A mixture of 10.12 g (55.9 mmol) of 2-amino-3-(3-hydroxy-phenyl)-propionic acid, 20 mL of 37% formaldehyde solution in water, 10 mL of conc. aq. HCl and 100 mL of 30 methanol was refluxed for 3 h. Evaporation of the solvents gave a mixture of 6-hydroxy 1,2,3,4-tetrahydro-isoquinoline-3-carboxylic acid methyl ester and 8-hydroxy-1,2,3,4 tetrahydro-isoquinoline-3 -carboxylic acid methyl ester as light yellow solid in quantitative yield. LCMS: 208 (M+1)*. 187 WO 2006/099379 PCT/US2006/009049 A mixture of the previously described 6-hydroxy-1,2,3,4-tetrahydro-isoquinoline-3 carboxylic acid methyl ester and 8-hydroxy- 1,2,3,4-tetrahydro-isoquinoline-3 -carboxylic acid methyl ester and 15.3 g (70 mmol) of di-tert-butyl dicarbonate in 40 mL of dioxane and 40 mL of 2 N Na2CO 3 solution was stirred at room temperature for 80 min. The reaction 5 mixture was partitioned between ethyl acetate (300 mL) and brine (300 mL). The aqueous layer was separated and extracted again with ethyl acetate (150 mL). The combined organic layers were dried over sodium sulfate and evaporated. Isolation by column chromatography on silica gel gave 6.186 g (36%) of a mixture of 6-hydroxy-3,4-dihydro-1H-isoquinoline-2, 3 dicarboxylic acid 2-tert-butyl ester 3-methyl ester and 8-hydroxy-3,4-dihydro-1H 10 isoquinoline-2,3-dicarboxylic acid 2-tert-butyl ester 3-methyl ester. LCMS: 308 (M-1). The mixture of 6-hydroxy-3,4-dihydro-1H-isoquinoline-2,3-dicarboxylic acid 2-tert butyl ester 3-methyl ester and 8-hydroxy-3,4-dihydro-1H-isoquinoline-2,3-dicarboxylic acid 2-tert-butyl ester 3-methyl ester from the previous step, 5.04 mL (80 mmol) of methyl iodide, 5.53 g (40 mmol) of potassium carbonate, and 50 mL of DMF was heated at 90'C overnight. 15 The reaction mixture was partitioned between ethyl acetate (400 mL) and water (400 mL) and separated. The organic layer was dried over sodium sulfate, evaporated and used directly for next step. LCMS: 322 (M+1). A solution of 6-methoxy-3,4-dihydro-1H-isoquinoline-2,3-dicarboxylic acid 2-tert butyl ester 3-methyl ester and 8-methoxy-3,4-dihydro-1 H-isoquinoline-2,3-dicarboxylic acid 20 2-tert-butyl ester 3-methyl ester from the previous step in 20 mL of dichloromethane was treated with 20 mL of 4 N HCI in dioxane and stirred at room temperature for 3 h. Evaporation of solvents in vacuo gave a mixture of 6-methoxy-1,2,3,4-tetrahydro isoquinoline-3-carboxylic acid methyl ester hydrochloride and 8-methoxy-1,2,3,4-tetrahydro isoquinoline-3-carboxylic acid methyl ester hydrochloride, which was washed with hexanes 25 and used directly for next step. LCMS: 222 (M+1)*. The mixture of 6-methoxy-1,2,3,4-tetrahydro-isoquinoline-3-carboxylic acid methyl ester hydrochloride and 8-methoxy-1,2,3,4-tetrahydro-isoquinoline-3-carboxylic acid methyl ester hydrochloride from the previous step in 100 mL of toluene was treated with 9.50 g (41 mmol) of DDQ and was heated under reflux for 1 h. The reaction mixture was partitioned 30 between 2 M NaS 2
O
3 (100 nL), 1 M Na 2 COI (100 mL) and ethyl acetate (150 mL) and separated. The aqueous layer was extracted again with ethyl acetate (200 mL). The combined organic layers were dried over sodium sulfate and evaporated. Purification by column chromatography on silica gel separated the desired products as follows: 188 WO 2006/099379 PCT/US2006/009049 6-Methoxy-isoquinoline-3-carboxylic acid methyl ester: 2.35 g (52 % in 3 steps). 'I NMR (400 MHz, CDCl 3 ): 9.19 (s, 111), 8.51 (s, IH), 7.95 (d, 1H), 7.37 (dd, 1H), 7.21 (d, 1H), 4.07 (s, 3H), 3.98 (s, 3H) ppm; LCMS: 218 (M+1). 8-Methoxy-isoquinoline-3-carboxylic acid methyl ester: 90 mg (2 % in 3 steps). 'H 5 NMR (400 MHz, CDC1 3 ): 9.69 (s, 1H), 8.53 (s, 1H), 7.69 (dd, 1H), 7.52 (d, 1H), 7.04 (d, 1H), 4.066 (s, 3H), 4.064 (s, 3H) ppm; LCMS: 218 (M+1)+. 2.35 g (10.82 mmol) of 6-methoxy-isoquinoline-3-carboxylic acid methyl ester, was hydrolyzed according to the general procedure B to provide 1.99 g (90%) 6-methoxy isoquinoline-3-carboxylic acid LCMS: 204 (M+1)+. 10 224 mg (1.1 mmol) of 6-methoxy-isoquinoline-3-carboxylic acid was reacted with 242 mg (1 mmol) of intermediate B (2-amino-3H-benzoimidazole-4-carboxylic acid (1H imidazol-2-yl)-amide), 569 mg (1.5 mmol) of HBTU, 0.704 ml (4 mmol) of DIEA and 5 mL of DMF stirring at 804C for 2 h as describe in general procedure A to provide 212.4 mg (50%) of the title compound. 'H-NMR (400 MHz, DMSO): 9.37(s, 1H), 8.71(s, 1H), 8.24(d, 15 1M), 7.94(d, 111), 7.82(d, 1H), 7.73(d, IH), 7.49(dd, IH), 7.34(t, 1H), 6.88(s, 1H), 6.76(s, 1H), 3.98 (s, 3H) ppm; LCMS: 428 (M+i)+. Example 295 90 mg (0.46 mmol) of 8-methoxy-isoquinoline-3-carboxylic acid methyl ester (synthesized in above examples) was hydrolyzed according to the general procedure B to 20 provide 84 mg (100%) of 8-methoxy-isoquinoline-3-carboxylic acid. LCMS: 204 (M+1)*. 32.3 mg (38%) of the 8-methoxy-isoquinoline-3-carboxylic acid [4-(IH-imidazol-2 ylcarbamoyl)-1H-benzoimidazol-2-yl-amide was synthesized according to General Procedure A from 41 mg (0.2 mmol) of 8-methoxy-isoquinoline-3-carboxylic acid, 48 mg (0.2 mmol) of intermediate B (2-amino-3H-benzoinidazole-4-carboxylic acid (lH-imidazol 25 2-yl)-amide), 100 mg (0.264 mmol) of HBTU, 0.167 mL (0.97 nmol) of DIEA and 1 mL of DMF stirring at 80*C for 2 h. LCMS: 428 (M+1)+ Example 296 0.25 g (17% in 4 steps) 7-methoxy-isoquinoline-3-carboxylic acid was prepared according to procedure described for the synthesis of 6-methoxy-isoquinoline-3-carboxylic 30 acid (in Example 294) starting from 2.0 g (6.8 mmol) of boc-7-hydroxy tetrahydroisoquinoline carboxylic acid using 179 mg (0.824 mmol). LCMS: 204 (M+1)t. 71.8 mg (84%) of 7-mnethoxy-isoquinoline-3-carboxylic acid [4-(lH-imidazol-2 ylcarbamoyl)-IH-benzoimidazol-2-yl}-amide was prepared according to General Procedure 189 WO 2006/099379 PCT/US2006/009049 A using 41 mg (0.2 mmol) of 7-methoxy-isoquinoline-3-carboxylic acid, 48 mg (0.2 mrnol) of intermediate B (2-amino-3H-benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide), 100 mg (0.246 mmol) of HBTU, 0-07 mL (0.4 mmol) of DIEA and 2 mL of DMT stirring at 80 0 C for 2 h. LCMS: 428 (M+1)*. 5 Example 297 0.2 g (9.1%, 4 steps) of the 6-isopropoxy-1,2,3,4-tetrahydro-isoquinoline- 3 -carboxylic acid was prepared according to procedure described for the synthesis of 6-methoxy isoquinoline-3-carboxylic acid (in Example 294) starting from 0.67 g (2.18 mmol) of 6 hydroxy-3,4-dihydro-1HI-isoquinoline-2,3-dicarboxylic acid 2-tert-butyl ester 3-methyl ester. 10 LCMS: 232 (M+1)*. 0.05 g (0.23 mmol) of 6-isopropoxy-isoquinoline-3-carboxylic acid and 0.19 g (0.50 mmol) of HBTU were combined in a flask charged with DMF (3 mL) and DIEA (1 mL). This reaction mixture was then treated with 0.1 g (0.4 mmol) 2-amino-3H-benzoimidazole-4 carboxylic acid (1IH-imidazol-2-yl)-amide (Intermediate B,) and stirred at 80"C for 1h. The 15 reaction mixture was diluted with water, and the resultant solid precipitate was filtered and washed with saturated NaHCO 3 . The solid was dissolved with MeOH, evaporated onto silica gel and isolated by flash column chromatography (500 mL DCM/20 mL NH3/MeOH) to yield 10 mg (9%) of 6-isopropoxy-isoquinoline-3-carboxylic acid [4-(IH-imidazol-2 ylcarbamoyl)-1IH-benzoimidazol-2-yl]-amide. LCMS: 456 (M+1)*. 20 Example 298 1.556 g (11 mmol) of 3-methylsulfanyl-propionyl chloride was added slowly to a mixture of 2.20 g (10 mmol) of phenylalanine methyl ester hydrochloride, 4.18 mL (30 mmol) of triethyl amine and 50 mL of dichloromethane and stirred at room temperature for 2 h. The reaction mixture was partitioned between DCM (50 mL) and saturated NaHCO 3 solution (100 25 mL). The aqueous layer was separated and extracted again with dichloromethane (100 mL). The combined organic extracts were dried over sodium sulfate and evaporated. Purification by column chromatography on silica gel gave 2.574 g (92%) of 2-(3-methylsulfanyl propionylamino)-3-phenyl-propionic acid methyl ester. LCMS: 282 (M+1)*. 0.882 mL (10.07 mmol) of oxalyl chloride was added dropwise to a solution of 2-(3 30 methylsulfanyl-propionylamino)-3-phenyl-propionic acid methyl ester (from previous step) in 30 mL of dichloromethane and stirred at room temperature for I h. This reaction mixture was then cooled to -10 C and treated with 1.82 g (11 mmol) of FeC 3 at this temperature in portions. This reaction mixture was allowed to warmi up to room temperature overnight. 30 190 WO 2006/099379 PCT/US2006/009049 mL of 2 N aq. HC1 was added slowly while stirring for 1 h. 20 mL of dichloromethane was added and separated, washed again with water (50 mL), and dried over sodium sulfate. The organic solvents were evaporated in vacuo. The residual red solid was mixed with 30 mL of methanol and 1.5 mL of conc. H 2 S0 4 and heated under reflux overnight. Water was added to 5 the reaction mixture and the methanol was evaporated under reduced pressure. Ammonium hydroxide was added to adjust pH to basic. The aqueous layer (-50 mL) was extracted with dichloromethane (2 x 50 mL). The combined organic layers were dried over sodium sulfate and evaporated. Purification by column chromatography on silica gel gave 923 rmg (3 8%) of 1-.(2-methylsulfanyl-ethyl)-3,4-dihydro-isoquinoline-3-carboxylic acid methyl ester. LCMS: 10 264 (M+1)*. 1,297 g (7 mmol) of copper acetate was added to a solution of 923 mg (3.5 mmol) of 1-(2-methylsulfanyl-ethyl)-3,4-dihydro-isoquinoline-3-carboxylic acid methyl ester in 10 mL of dichloromethane, stirring at room temperature overnight. This reaction mixture was concentrated and purified by column chromatography on silica gel to give 266 mg (29%) of 15 1-(2-methylsulfanyl-ethyl)-isoquinoline-3-carboxylic acid methyl ester. LCMS: 262 (M+l). 1-(2-methylsulfanyl-ethyl)-isoquinoline-3-carboxylic acid was prepared according to General Procedure B using 49 mg (0.187 mnol) of 1-(2-methylsulfanyl-ethyl)-isoquinoline 3-carboxylic acid methyl ester, 0.28 mL of 2 N aq. LiOH, 2 mL of THF and 0.5 mL of methanol stirring at room temperature for 1 h. 42.4 mg (91%) of 1-(2-methylsulfanyl-ethyl) 20 isoquinoline-3-carboxylic acid was obtained. LCMS: 248 (M+1)*. 42.4 mg (0.171 mmol) of 1-(2--methylsulfanyl-ethyl)-isoquinoline-3-carboxylic acid was reacted with 34 mg (0.14 mmol) of intennediate B (2-amino-3H-benzoimidazole-4 carboxylic acid (1H-imidazol-2-yl)-amide), 114 mg (0.3 mmol) of HBTU and 0.106 mL (0.6 mmol) of DIEA in 0.8 mL of DMF stirring at 851C for 3 h according to the general procedure 25 A. 44.3 mg (55%) of 1-(2-methylsulfanyl-ethyl)-isoquinoline-3-carboxylic acid [4-(IH imidazol-2-ylcarbamoyl)-1H-benzoimidazo-2-yl}-amide was obtained. LCMS: 472 (M+1) 4 . Example 299 559 mg (0.91 nmol) of oxone monopersulfate was added to a solution of 217 mg (0.83 mmol) of 1-(2-inethylsulfanyl-ethyl)-isoquinoline-3-carboxylic acid methyl ester 30 (synthesized in above Example 298) in 4 nL of methanol and stirred at room temperature for 1 5 h. The reaction mixture was partitioned between dichloromethane (40 mL) and water (40 mL). The aqueous layer was separated and extracted again with dichloromethane (40 mL). The combined organic extracts were dried over sodium sulfate and evaporated. Purification 191 WO 2006/099379 PCT/US2006/009049 by column chromatography on silica gel gave 169 mg ( 69%) of 1-(2-methylsulfony-ethyl) isoquinoline-3-carboxylic acid methyl ester as off-white solid. LCMS: 294 (M+1). The ester was hydrolyzed according to the general procedure B to provide 145 mg (90%) of the 1 (2-methylsulfonyl-ethyl)-isoquinoline-3-carboxylic acid. 5 51.7 mg (57%) of 1-(2-methanesulfony-ethyl)-isoquinoliie-3-carboxylic acid [4-(1H imidazol-.2-ylcarbamoyl)-1H-benzoimidazol-2-yll-amide as prepared according to General Procedure A using 50 .3 mg (0.18 mnol) of 1-(2-inethylsulfonyl-ethyl)-isoquinoline- 3 carboxylic acid, 36.3 mg (0.15 mmol) of intermediate B (2-amino-3H-benzoimidazole-4 carboxylic acid (IH-imidazol-2-y1)-amide), 114 mg (0.3 mnol) of HBTU, 0.106 nL (0.6 10 mmol) of DIEA and 0.8 mL DMF (0.8 mL) stirring at 85 0 C for 3 h. LCMS: 504 (M+1)*. Example 300 0.25 g (11 % overall yield) of 1-methyl-isoquinoline-3-carboxylic acid was synthesized according to the procedure described for the synthesis of 1-(2-methylsulfanyl ethyl)-isoquinoline-3-carboxylic acid (in Example 298) starting from 2.20 g (10 mmol) of 15 phenylalanine methyl ester hydrochloride. LCMS: 188 (M+1)*. 17.6 mg (15%) of 1 -methyl-isoquinoline-3 -carboxylic acid [4-(1 H-imidazol-2 ylcarbamoyl)-1H-benzoimidazol-2-yl]-amide was synthesized according to General Procedure A using 55 mg (0.29 nmol) of 1-methyl-isoquinoline-3-carboxylic acid, 70 mg (0.29 mmol) of intermediate B (2-amino-3H-benzoimidazole-4-carboxylic acid (1H 20 imidazol-2-yl)-amide), 144 mg (0.38 mmol) of HBTU, 0.264 mL (1.5 mmol) of DIEA in 0.8 mL of DMF stirring at 85 0 C for 2 h. 1 H-NMR (400 MHz, DMSO): 8.66 (s, 1H), 8.38 (d, 1H), 8.28 (d, 1H1), 7.89 (m 4H), 7.34 (m, 1H), 6.85 (b, 211), 3.07 (s, 31) ppm. LCMS: 412 (M+1)j. Example 301 0.083 mL (I mmol) of sulfuryl chloride was added to a solution of 90 mg (0.41 mmol) 25 of 8-methoxy-isoquinoline-3-carboxylic acid methyl ester (from Example 295) in 1 mL of acetic acid and stirred at room temperature overnight. Ether was added to the reaction mixture, and the precipitated solid was filtered, washed with ether, then partitioned between ethyl acetate (30 mL) and saturated NaHCO 3 solution (30 mL) and extracted. The organic layer was dried over sodium sulfate, and evaporation of the solvent in vacuo gave 47.3 mg 30 (46%) of 7-chloro-8-methoxy-isoquinoline-3-carboxylic acid methyl ester. LCMS: 252 (M+i)+. The ester was hydrolyzed according to the general procedure B to provide 40 mg (88%) of the 7-chloro-8-methoxy-isoquinoline-3-carboxylic acid. LCMS: 238 (M+1)*. 192 WO 2006/099379 PCT/US2006/009049 10 mg (13%) of 7-chloro-8-methoxy-isoquinoline-3-carboxylic acid [4-(1 H-imidazol 2-ylcarbamoyl)-1H-benzoimidazol-2-yl]-amide was synthesized according to the general procedure A using 7-chloro-8-inethoxy-isoquinoline-3-carboxylic acid (from previous step), 36.3 mg (0.15 mmol) of intermediate B (2-amino-3H-benzoimidazole-4-carboxylic acid (111 5 imidazol-2-yl)-amide), 69 mg (0.18 mmol) of HBTU, 0.106 mL (0.6 mmol) of DIEA in 2 mL of DMF stirring at 80'C for 2 h. LCMS: 462 (M+1)t. Example 302 1.0 g (6.8 mmol) of thiophene-2, 3-dicarbaldehyde was dissolved in DCM (50 mL) and cooled to 0 0 C. 2.0 g (7.4 rnmol) of (±) Boc-alpha-phosphonoglycine trimethyl ester was 10 dissolved in DCM (25 mL) and 1. 14 g (7.5 mmol) of DBU was added at ambient temperature under stirring. This solution was added drop wise to the cold solution of the dialdehyde under stirring. The solution was stirred at 0 0 C for I h and overnight at ambient temperature. The reaction mixture was concentrated, and the residue was dissolved in 4 M HCI in dioxane. The resulting white suspension was stirred at room temperature for 1 h. Solvent was 15 evaporated, extracted with ethyl acetate (100 ml), washed with sat NaHCO3, water and brine. The organic layer was dried over magnesium sulfate, evaporated purified by column chromatography using 20 % ethyl acetate in hexane as an eluent to give two regioisomers as follows. 0.34 g (24 %) of Thieno (2,3-c] pyridine-5-carboxylic acid methyl ester. 1 H-NMR 20 (400 MHz, CDCI 3 ): 9.20 (s, 1H), 8.27(s, 1H), 7.82 (d, IH), 7.50(d, 1H), 4.03 (s, 3H) ppm; LCMS: 180 (M+1)*. 0.17 g (12 %) of thieno L3,2-c] pyridine-6-carboxylic acid methyl ester. 'H-NMR (400 MHz, CDCI 3 ): 9.191 (s, 1H), 8.7(s, 1H), 7.72 (d, 1H), 7.55 (d, 1H) ppm; LCMS: 180 (M+1). The methyl esters were hydrolyzed as shown in general procedure B to give corresponding 25 acids in a quantitative yield. 2.6 mg (3 %) of thieno[2,3-c]pyridine-5-carboxylic acid [4-(1H-imidazol-2 ylcarbamoyl)- 1 H-benzoimidazol-2-yl}-amide was prepared according to general procedure A using 54 mg (0.3 mmol) thieno[2,3-c]pyridine-5-carboxylic acid, 61 mg (0.25 nimol) intermediate B (2-amino-3H-benzoimidazole-4-carboxylic acid (IH-imidazol-2-yl)-amide), 30 114 mg (0.3 mmol) of HBTU, 0.106 mL (0.6 mmol) of DIEA in 1 mL of DMF stirring at 85 0 C for 1 h. LCMS: 404 (M+1)1. 193 WO 2006/099379 PCT/US2006/009049 Example 303 5.0 mg (2.9 %) of thieno[3,2-c]pyridine-6-carboxylicacid [7-(lH-imidazol-2 ylcarbamoyl)-1H-benzoimidazol-2-yl]-amide was prepared according to general procedure A using 54 mg (0.3 mmol) thieno[3,2-c]pyridine-6-carboxylic acid (synthesized in above 5 example), 61 mg (0.25 mmol) intermediate B (2-amino-3H-benzoimidazole-4-carboxylic acid (lH-imidazol-2-yl)-amide), 114 mg (0.3 mumol) of HBTU, 0.106 mL (0.6 mmol) of DIEA in 1 mL of DMF stirring at 851C for 1 h. LCMS: 404 (M+1)j. Example 304 100 mg (0.56 mmol) of the 2-methyl-1,8a-dihydro-imidazo[1,2-a]pyridine-3 10 carboxylic acid and 234 mg (0.61 mmol) of HBTU in a mixture of 1.5 mL of DMF and 0.6 mL of DIEA was reacted with 137 mg (0.56 nmol) of the 2-amino-3H-benzoimidazole-4 carboxylic acid (1H-imidazol-2-yl)-amide (Intermediate B) as described in general procedure A. After the reaction was complete, it was diluted with brine (20 mL), and the resulting solid was filtered, washed with water, ethyl acetate and dried under vacuum to give 40 mg (17 %) 15 of 2-[(2-methyl-imidazo[1,2-a~pyridine-3-carbonyl)-amino]-1H-benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide as a solid. LCMS: 401 (M + 1). Example 305 1.54 g (10.0 mmol) of 3-methoxy-2-nitropyridine was hydrogenated according to the general procedure C to give 2-amino-3-methoxypyridine. This product was directly treated 20 with 2.5 g (12.0 mmol) of ethyl bromopyruvate in 20.0 mL of TIF at r.t. overnight and refluxed for 8 h after addition of 20.0 mL of ethanol. The solvents were evaporated, and the resulting residue was diluted with TIF, filtered and washed with THF to give 1.76 g (80%) of 8-methoxy-imidazo[1,2-a]pyridine-2-carboxylic acid ethyl ester. This ester was hydrolyzed according to general procedure B to give 1.5 g (95%) of the 25 corresponding 8-methoxy-imidazo[1,2-a]pyridine-2-carboxylic acid. 300 mg (3.0 mmol) of the above 8-methoxy-iiidazo[l,2-a]pyridine-2-carboxylic acid and 200 mg (0.5 mmol) of HBTU in a mixture of 1.5 mL of DMF and 0.6 mL of DIEA was reacted with 60 mg (0.25 mmol) of the 2-amino-3H-benzoimidazole-4-carboxylic acid (IH imidazol-2-yl)-amide (Intennediate B) as described in general procedure A. After the 30 reaction was complete, it was diluted with brine (20 mL), and the resulting solid was filtered, washed with water, ethyl acetate and dried under vacuum to give 41 mg (40 %) of 2-[(8 methoxy-imidazo[1,2-a]pyridine-3-carbonyl)-amino]-1H-benzoimidazole-4-carboxylic acid (lH-imidazol-2-yl)-amide as a solid. LCMS: 417 (M + 1). 194 WO 2006/099379 PCT/US20061009049 By analogous methods to those used to prepare Examples 290 to 305, and those in the relevant above schemes, the following compounds were synthesized. H'N 7 N o NsH N H \ N N A r H O LCMS Ex. Ar ( 306 6,7-Bis-(2-methoxy-ethoxy)-isoquinoline-3-y 546 307 Cinnoline-3-yl 417 308 Quinoxaline-2-yl 399 309 6-Bromo-pyridine-2-yl 427 310 [1,8]Naphthyridine-2-yl 399 311 Isoquinoline- 1 -yl 398 312 6,7-Dimethoxy-isoquinoline-3-yl 458 313 4-Cyano-phenyl 372 314 6-Cyano-pyridine-3-yl 373 315 Isoquinoline-5-yl 398 316 2,6-Dimethoxy-pyrimidine-4-y1 518 317 6-Benzyloxy-isoquinoline-3-yl 504 318 2,3-Dihydro-[ 1,4] dioxino[2,3 -g]isoquinoline-8-yl 456 319 [1,3]Dioxolo[4,5-g]isoquinoline-7-y1 442 320 6-Cyclopentyloxy-isoquinoline-3-yl 482 321 1-Cyclopentyhnethyl-7-methoxy-isoquinoline-3-yI 510 322 1 -Isopropyl-isoquinoline-3-yI 440 323 6-Ethoxy-isoquinoline-3-yl 442 324 6-Butoxy-isoquinoline-3-yl 470 325 1-Propyl-isoquinoline-3-yl 440 195 WO 2006/099379 PCT/US2006/009049 326 1-Butyl-isoquinoline-3-yl 454 327 1 -Isobutyl-isoquinoline-3-yl 454 328 1-Cyclopentyl-isoquinoline-3-yl 466 329 7-Methoxy-1-methyl-isoquinoline-3-y1 442 330 1-Methyl-6-trifluoromethoxy-isoquinlifne- 3 -y1 496 331 7-Methanesulfonyl-1 -methyl-isoquinoline-3-yl 490 332 1-(Tetrahydro-pyran-4-y)-isoquinoline- 3 -yl 482 333 1-Methyl-7-trifluoromethoxy-isoquinoline-3-yl 496 334 5,8-Dimethoxy-isoquinoline-3-yI 458 335 4-Methoxy-quinoline-2-yl 428 336 7-Methoxy-isoquinoline-1 -yl 428 337 Imidazo[1,2-a]pyridine-2-yl 387 338 5-Methyl-imidazo[1,2-a]pyridine-2-yl 401 339 Imidazo[2,1-blthiazole-6-yl 393 340 8-Methyl-imidazo[1,2-a]pyridine-2-yl 401 341 [Bis-(4-chloro-phenyl)]-methyl 505 Example 342 To a solution 1.0 g (4.9 mmol) of 6-bromo-pyridine-2-carboxylic acid, 0.9 g (7.4 mmol) of phenylboronic acid, 0.4 g (0.36 mmol) of palladium tetrakistriphenyl phosphine in 5 25 mL of DME was added 9.9 mL of 2 N Na 2
CO
3 solutions under nitrogen. After 12h reflux under nitrogen, the reaction mixture was cooled and filtered through a filter paper. The filtrate was acidified using 2 N HCI (pH = 5). The resulting white precipitate was filtered, washed with water and dried to afford 0.5 g (51%) of 6-phenyl-pyridine-2-carboxylic acid. LCMS: 200 (M+1)t 10 0.1 g (0.5 mmol) of above acid, 0.19 g (0.5 mmol) of HBTU in a mixture of 2.0 ml of DMF and 0.24 mL of DIEA was reacted with 0.12 g (0.5 nnol) of 2-amino-3H benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide (Intermediate B) as described in general procedure A. The reaction mixture was cooled to r.t. and 5.0 mL of water and 5.0 mL of saturated NaHCO 3 was added. The resulting solid was filtered, washed with water and 15 ether, and dried to provide 0.04 g (20 %) of 2-{(6-phenyl-pyridine-2-carbonyt)-amino]-lH benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide. LCMS: 424 (M+1). 196 WO 2006/099379 PCT/US2006/009049 Example 343 0.12 g (0.51 mmol) of 6-(3-cyano-phenyl)-pyridine-2-carboxylic acid, 0.19 g (0.5 mmol) of H3TU in a mixture of 2.0 ml of DMF and 0.26 nL of DIEA was reacted with 0.1 g (0.41 mmol) of 2-amino-3H-benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide 5 (Intermediate B) as described in general procedure A. The reaction mixture was cooled to r.t. and 5.0 mL of water and 5.0 mL of saturated NaHCO 3 was added. .The resulting solid was filtered, and the solid was washed with water and ether and dried to provide 0.04 g (17.5 %) of 2-{ [6-(3 -cyano-phenyl)-pyridine-2-carbonyl]-amino} -IH-benzoimidazole4-carboxylic acid (lH-imidazol-2-yl)-amide. 'H-NMR (400 MHz, DMSO): 9.15 (s, IH), 8.86 (s, IH), 10 8.45(m, 111), 8.24 (m, 2H), 7.99 (d, 1H), 7.92 (d, 1H), 7.87 (d, 1H), 7.72 (t, 1H), 7,41(t, 1H), 7.30 (s, 2H) ppm. LCMS: 449 (M+1). Example 344 0.31 g (2 mmol) of 5-hydroxy-nicotinic acid methyl ester, 0.73 g (4 mmol) of copper acetate, -1 g of 4A molecular sieves and 0.85 g (7 mmol) of phenylboronic acid were 15 combined in a flask charged with 20 mL of DCM and 2.0 mL (14 mmol) of Et 3 N. This reaction slurry was stirred overnight at r.t. The reaction mixture was then diluted with 200 mL of EtOAc and washed 3 times with 10% K 2
CO
3 . The organic layer was then dried with Na 2
SO
4 , evaporated and purified by flash column chromatography (EtOAc/hexanes = 1:2) to give 5-phenoxy-nicotinic acid methyl ester, which was subsequently hydrolyzed according to 20 general procedure B to give 5-phenoxy-nicotinic acid and used in the next step without further purification. 0.07 g (0.30 mmol) of 5-phenoxy-nicotinic acid and 0.12 g (0.32 mmol) of HBTU were combined in a flask charged with 2 mL of DMF and 0.25 mL of DIEA. The reaction mixture was then treated with 0.05 g (0.20 mmol) of 2-amino-3H-benzoimidazole-4 25 carboxylic acid (1H-imidazol-2-yl)-amide (Intermediate B) and stirred at 80"C for lh. The reaction mixture was diluted with water, and the resultant solid precipitate was filtered and washed with saturated NaHCO 3 . The solid was dissolved with MeOH, evaporated onto silica gel and isolated by flash column chromatography (500 mL DCM/20 mL NH 3 /MeOH) to yield 10 mag (11 %) of 2-[(5-phenoxy-pyridine-3-carbonyl)-amino]-lH-benzoimidazole-4 30 carboxylic acid (1H-imidazol-2-yl)-amide. LCMS: 441 (M+l)*. Example 345 0.31 g (2 mmol) of 5-hydroxy-nicotinic acid methyl ester and 3.1 g (4 mmol) of polymer-supported triphenylphospbine were combined in a flask charged with 45 mL of THF and 0.31 mL (3 mmol) of benzyl alcohol, and shaken at r.t. until the starting materials 197 WO 2006/099379 PCT/US20061009049 dissolved. The reaction mixture was then cooled to 0 0 C with a water/brine bath, 0.58 mL (3 mmol) of DIAD was added and the reaction slurry then stirred overnight at r.t. The reaction mixture was filtered and the resin was washed 3 times with DCM/MeOH = 1:1. The organic layer was then evaporated and flashed (EtOAc/hexanes = 1:2) to give the desired 5 5 benzyloxy-nicotinic acid methyl ester, which was subsequently hydrolyzed according to general procedure B to give 5-benzyloxy-nicotinic acid which was used in the next step without further purification. LCMS: 230 (M+1)*. 0.07 g (0.30 mmol) of 5-benzyloxy-nicotinic acid and 0.12 g (0.32 mmnol) of HBTU were combined in a flask charged with 2 mL DMF and 0.25 mL of DIEA. This reaction 10 mixture was then treated with 0.05 g (0.20 mmol) of 2-Amino-3H-benzoinidazole-4 carboxylic acid (lH-imidazol-2-yl)-amide (Intermediate B) and stirred at 80"C for lh. The reaction mixture was diluted with water, and the resultant solid precipitate was filtered and washed with saturated NaHCO 3 . The solid was dissolved with MeOH, evaporated onto silica gel and isolated by flash column chromatography (500 mL DCM/20 mL NH/MeOH) to 15 yield 10 mg (10%) of 2-{(5-benzyloxy-pyridine-3-carbonyl)-amino]-1H-benzoimidazole-4 carboxylic acid (1H-imidazol-2-yl)-amide. LCMS: 455.0 (M+)t. By analogous methods to those used to prepare Examples 342 to 345 and those in the relevant above schemes, the following compounds were synthesized. H'N xN 0 NI H N H \>- N N Ar H O 20 Ar LCMS Ex. (M + 1)+ 346 6-(2-Carbamoyl-phenyl)-pyridine-2-y 467 347 6-(2-Trifluoromethoxy-phenyl)-pyridine-2-yl 508 348 6-(4-Fluoro-phenyl)-pyridine-3--y1 442 198 WO 2006/099379 PCT/US2006/009049 349 6-(4-Trifluoromethyl-phenyl)-pyridine-3-yl 492 350 6-(4-Trifluoromethoxy-phenyl)-pyridine-3-y1 508 351 5-(3-Fluoro-phenyl)-pyridine-3-yl 442 352 5-(4-Trifluoronethoxy-phenyl)-pyridine-3-yl 508 353 5-(2,4-Difluoro-phenyl)-pyridine-3-yl 460 354 5-(3-Trifluoromethoxy-phenyl)-pyridine-3-yl 508 355 6-(Furan-2-yl)-pyridine-2-yl 414 356 6-(2-Fluoro-phenyl)-pyridine-2-yl 442 357 6-(4-Methoxy-phenyl)-pyridine-3-yl 454 358 5-Phenyl-pyridine-2-yl 424 359 5-Phenyl-pyridine-3-yl 424 360 5-(4-Methoxy-phenyl)-pyridine-3-yl 454 361 4-Phenyl-pyridine-2-yl 424 362 6-(2-Methoxy-phenyl)-pyridine-2-yl 454 363 6-(3-Methanesulfonyl-phenyl)-pyridine-2-yl 502 364 6-(3-Aniinomethyl-phenyl)-pyridine-2-yI 599 365 2-(3-Cyano-phenyl)-pyridine-4-yl 449 366 6-Phenyl-pyridine-3-yl 424 367 3-Cyano-phenyl 372 368 6-(3-Cyanomethyl-phenyl)-pyridine-2-y1 463 369 6-(4-Methanesulfonyl-phenyl)-pyridine-2-y 502 370 3'-Cyano-biphenyl-3-yl 448 371 4'-Cyano-biphenyl-3-yl 448 372 [2,4']Bipyridinyl-6-yl 425 373 {2,3']Bipyridinyl-6-y 425 374 2-Phenoxy-pyridine-3-yl 440 375 3'-Cyano-biphenyl-4-yi 448 376 4'-Cyano-biphenyl4-yl 448 377 6-(3-Cyano-phenyl)-pyridine-3-y 449 378 3-Pyridin-3-phenyl 424 379 4-Pyridin-3-phenyl 424 380 2-(4-Fluoro-phenoxy)-pyridine-3-yl 458 199 WO 2006/099379 PCT/US2006/009049 381 4-Pyridin-4-phenyl 424 382 6-(2-Cyano-phenyl)-pyridine-2-yl 449 383 3-Benzyloxy-pyridine-2-yl 454 384 6-Benzyloxy-pyridine-2-yl 454 385 6-Thiophen-2-yl-pyridine-2-y1 430 386 5-Cyclopentyloxy-pyridine-3-yl 432 387 5-Cyclopentylmethoxy-pyridine-3-yl 446 388 5-(2-Cyclopentyl-ethoxy)-pyridine-3-yI 460 389 (R)-5-(1 -Phenyl-ethoxy)-pyridine-3 -yl 468 390 (S)-5-(l-Phenyl-ethoxy)-pyridine-3-yl 468 391 3-(2-Phenethyloxy)-pyridine-2-yl 468 392 5-Benzyloxy-pyridine-2-yl 454 393 5-(2-Phenethyloxy)-pyridine-2-yl 468 394 5-Cyclopentylmethoxy-pyridine-2-yI 446 395 5-(2-Cyclopentyl-ethoxy)-pyridine-2-yt 460 396 5-Isopropoxy-pyridine-3-yl 406 397 5-(1-Ethyl-propoxy)-pyridine-3-yI 434 398 5-Cyclopropylmethoxy-pyridine-3-y1 418 399 5-(1 -Cyclopropyl-ethoxy)-pyridine-3 -yl 432 400 5-Propoxy-pyridine-3-yI 406 401 5-Butoxy-pyridine-3-yl 420 402 5-Isobutoxy-pyridine-3-yl 420 403 4-(2-Cyclopentyl-ethoxy)-pyridine-2-yl 460 404 6-Phenyl-pyrimidine-4-yl 425 405 6-(4-Fluoro-phenyl)-pyrimidine-4-y 443 406 4-(5-Trifluoromethyl-pyridin-2-yl)-phenyl 492 407 4-(4-Trifluoromethyl-phenoxy)-phenyl 507 Example 408 To a solution of 0.66 g (3.0 mmol) of 6-(3-cyano-phenyl)-pyridine-2-carboxylic acid in DMF (10 mL) was added 1.5 g (3.9 mmol) of HBTU and DIEA (1 mL). The mixture was 5 stirred at room temperature for 10 min then 0.8 g (4.1 mmol) of 2-amino-3H-benzoimidazole 4 -carboxylic acid methyl ester (Intermediate A) was added. The reaction was heated at 90 0 C 200 WO 2006/099379 PCT/US2006/009049 for lh. Usual work up as described in general procedure A provided 1 g (83%) of 2-{[6-(3 cyano-phenyl)-pyridine-2-carbonyl]-anino}-3H-benzoimidazole-4-carboxylic acid methyl ester. LCMS: 398 (M+1)+. '1 NMR (DMSO-d 6 , 400 MHz): 5 8.85 (s, 1H), 8.60 (d, 1H), 8.23 (m, 2H), 8.11 (m, 111), 7.95 (m, 2H), 7.76 (dd, 1H), 7.60 (m, 2H), 3.91 (s, 311) ppm. 5 0.7 g (1.7 mmol) of above ester was hydrolyzed according to the general procedure B to give 0.53 g (82%) of the 2-{[6-(3-cyano-phenyl)-pyridine-2-carbonyl]-amino}-3H benzoimidazole-4-carboxylic acid. LCMS: 384 (M+1)*. To a solution of 76 mg (0.2 mmol) of above acid in DMF (1 mL) was added 113 mg (0.3 mmol) of HBTU and DIEA (0.2 mL), the mixture was stirred at room temperature for 10 10 min then 0.1 g (1.2 mmol) of butylamine was added. The mixture was heated at 60 "C for 30 min. After cooling to room temperature, usual work up as shown in general procedure A followed by column chromatography purification (eluting with DCM then ethyl acetate) gave 20 mg (22%) of 2-{(6-(3-cyano-phenyl)-pyridine-2-carbonyl]-amino}-3H-benzoimidazole-4 carboxylic acid butylamide. LCMS: 438 (M+1). 15 By analogous methods to those used to prepare Example 408 and those in the relevant above schemes, the following compounds were synthesized. H o N'W CEN N H \ N N. N H O LCMS Ex. W 409 phenyl 459 410 isopropyl 425 411 cyclohexyl 465 412 furan-2-ylmethyl 463 20 201 WO 2006/099379 PCT/US2006/009049 Example 413 0.728 mL (6,5 mmol) of phenyl acetylene was added to a mixture of 10.1 g (5 mmol) of 4-bromopicolinic acid, 91 mg (0.13 mmol) of dichlorobis(triphenylphosphine) palladium(II), 29 ing (0.15 mmol) of copper(I) iodide, 10 mL of triethylamine and 2 mL of 5 DMF and heated at 80"C overnight. The reaction mixture was filtered. The filtrate was concentrated and partitioned between 5 mL of 2 N Na 2
CO
3 solution, 25 nL of water and 30 mL of ether. The aqueous layer was separated and washed again with ether (30 mL). 10% aq. HCI was added to adjust the pH to 5. The aqueous layer was extracted with dichloromethane (2 x 50 mL), and the combined organic layers were dried over sodium sulfate. Evaporation 10 of the solvents in vacuo gave 1.028 g (4.61 mmol, 92%) of 4-phenylethynyl-pyridine-2 carboxylic acid. LCMS: 224 (M+)*. 0.05 (0.23 mmol) of the above synthesized 4-phenylethynyl-pyridine-2-carboxylic acid, 0.095 g (0.25 mmol) of HBTU in a mixture of 2.0 ml of DMF and 0.20 mL of DIEA was reacted with 0.05 g (0.20 mmol) of 2-amino-3H-benzoimidazole-4-carboxylic acid (1H 15 imidazol-2-yl)-amide (Intermediate B) as described in general procedure A. The reaction mixture was cooled to r.t. 5.0 mL of water and 5.0 mL of saturated NaHCO 3 was added, and the resulting solid was filtered. The solid was washed with water and ether, and dried to provide 0.04 g (43 %) of 2-[(4-phenylethynyl-pyridine-2-carbonyl)-amnino]-lH benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide. LCMS: 448 (M+1)*. 20 Example 414 0.1 g (0.46 mmol) of 5-Phenylethynyl-nicotinic acid, 0.19 g (0.5 mmol) of HBTU in a mixture of 2.0 ml of DMF and 0.30 mL of DIEA was reacted with 0.1 g (0.40 mmol) of 2 Amino-3H-benzoimidazole-4-carboxylic acid (IH-imidazol-2-yl)-amide (Intermediate B) as described in general procedure A. The reaction mixture was cooled to r.t. 5.0 mL of water 25 and 5.0 mL of saturated NaHCO 3 was added, and the resulting solid was filtered. The solid was washed with water, ether and dried to provide 0.08 g (43 %) of 2-[(5-phenylethynyl pyridine-3-carbonyl)-amino}-1H-benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl) amide. LCMS: 448 (M+1)+. Example 415 30 20 mg of (0.44 mmol) of 2-[(5-phenylethynyl-pyridine-3-carbonyl)-amino]-lH benzoimidazole-4-carboxylic acid (I H-imidazol-2-yl)-amide was dissolved in a mixture of 10 mL of CH 3 0H and 0.5 mL of acetic acid. To this stirring solution 10 mg of 10% Pd on carbon was added and the resulting mixture was hydrogenated (I atm) at room temperature 202 WO 2006/099379 PCT/US2006/009049 for 3.0 h. The reaction mixture was filtered, and the solid was washed with portions of methanol. The filtrate and washings were combined and evaporated to give 18 mg (88%) of 2-[(5-phenethyl-pyridine-3-carbonyl)-amino]-1H-beizoimidazole-4-carboxylic acid (1H imidazol-2-yl)-amide. LCMS: 452 (M+1)*. 5 Example 416 To a stirring solution 0.5 g (2.4 mmol) of 6-bromo-pyridine-2-carboxylic acid, 0.05 g (0.07 mmol) of dichlorobis(triphenylphosphine)-palladium(II), and 0.015 g (0.07 mmol) of copper (I) iodide in 10 mL of NEta, 9.9 mL of 2 N Na 2
CO
3 solution was added 0.38 g (3.7 mmol) of phenyl acetylene under nitrogen. After 12h reflux under nitrogen, the reaction 10 mixture was cooled and filtered. The filtrate was acidified using 2 N HCl (pH = 5), and the resulting white precipitate was filtered, washed with water and dried to afford 0.5 g (5 1%) of 6-phenylethynyl-pyridine-2-carboxylic acid. LCMS: 224 (M+1)*. 0.05 (0.23 mmol) of the above synthesized 6-Phenylethynyl-pyridine-2-carboxylic acid, 0.095 g (0.25 mmol) of HBTU in a mixture of 2.0 ml of DMF and 0.20 mL of DIEA 15 was reacted with 0.05 g (0.20 mmol) of 2-Amino-3H-benzoimidazole-4-carboxylic acid (1H imidazol-2-yl)-amide (Intermediate B) as described in general procedure A. The reaction mixture was cooled to r.t. 5.0 mL of water and 5.0 mL of saturated NaHCO 3 was added, and the resulting solid was filtered. The solid was washed with water and ether and dried to provide 0.04 g (43 %) of 2-[(6-phenylethynyl-pyridine-2-carbonyl)-amino]-lH 20 benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide. 'H-NMR (400 MHz, DMSO): 8.3 (d, 1H), 8.21 (t, 1H), 8.12 (bs, 1H), 8.02 (d, 1H1), 7.94 (d, 1H), 7.72(m, 2H), 7.50 (m, 4H), 7.40 (s, 2H) ppm; LCMS: 448 (M+1)*. Example 417 To a solution of 0.23 g (1 mmol) of 1-bromo-3-methanesulfonyl-benzene in benzene 25 (2 mL) was added 40 mg (0.1 iniol) of PdC1 2 (Ph3P)2, 30 rmg (0.1 mmol) CuI, triethylainine (1 mL) and 0.3 mL of ethynyl-trimethyl-silane. The reaction mixture was heated at 70 *C for 3h After removal of solvent, the residue was purified by column chromatography elution with hexanes/ethyl acetate (v/v=7:1 then 5:1) to give 0.24 g (95 %) of (3-methanesulfonyl phenylethynyl)-trimethylsilane. 'H NMR (CDCl 3 , 400 MHz): 5 8.4 (s, 1H), 7.88 (d, 1H), 7.70 30 (d, 1H), 7.52(dd, 1H), 3.05 (s, 3H), 0.27 (s, 9H) ppm. To a solution of 0.24 g (0,95 nmol) of the above (3-methanesulfonyl-phenylethynyl) trimethyl-silane in methanol (2 mL) was added K 2 C0 3 aq. solution (I M, 1 mL), the mixture was stirred at room temperature for lh. After usual workup, the residue was purified by 203 WO 2006/099379 PCT/US2006/009049 column chromatography eluted with hexanes/ethyl acetate (v/v=5:1 then 3:1) to give 140 mg (82 %) 1-ethynyl-3-methanesulfonyl-benzene. 136 mg (0.45 mmol) of 6-(3-methanesulfonyl-phenylethynyl)-pyridine-2-carboxylic acid was synthesized as described for the synthesis of 6-Phenylethynyl-pyridine-2-carboxylic 5 acid in the above example starting with 100 mg of 6-bromo-pyridine-2-carboxylic acid. LCMS: 302 (M+1)t. To a solution of 136 mg (0.45 mmol) of 6-(3-methanesulfonyl-phenylethynyl) pyridine-2-carboxylic acid in DMF (2 mL) was added 210 mg (0.55 mmol) of HBTU (210 mg) and DIEA (0.2 mL). The mixture was stirred for 5 min then 96 mg (0.5 mmol) of 2 10 amino-1H-benzoimidazole-4-carboxylic acid methyl ester (Intermediate A) was added. The reaction mixture was stirred at 30 *C for 30 min. After usual workup as described in general procedure A, 142 mg (60%) of 2-{[6-(3-Methanesulfonyl-phenylethynyl)-pyridine-2 carbonyl]-amino}-1H-benzoimidazole-4-carboxylic acid methyl ester was obtained after column chromatography eluting with DCM/EtOAc (v/v= 1:1) then EtOAc. LCMS: 475 15 (M+1)4. This ester was hydrolyzed as described in general procedure B to provide 135 rmg (100 %) of 2- {[6-(3-methanesulfonyl-phenylethynyl)-pyridine-2-carbonyl]-amino }-1H benzoimidazole-4-carboxylic acid. LCMS: 461 (M+1). To a solution of 46 mg (0.1 mmol) of the above acid in DMF (Im L) was added 60 mg (0.15 mmol) of HBTU and DIEA (0- 1 mL), The mixture was stirred for 5 min then 40 mug 20 (0.3 mmol) of 2-aminoimidazole sulfate was added in one portion. The reaction mixture was heated at 80 "C for 40 min. After usual workup as described in general procedure A, 12 mg (20%) of 2- { [6-(3-methanesulfonyl-phenylethynyl)-pyridine-2-carbonyl]-amino} -1 H benzoimidazole-4-carboxylic acid (1H-imidazol-2-yl)-amide was obtained after purification by column chromatography eluting with DCM/methanol (v/v from15:1 to 7:1). LCMS: 526 25 (M+1)*. By analogous methods to those used to prepare Examples 413 to 417 and those in the relevant above schemes, the following compounds were synthesized. 204 WO 2006/099379 PCT/US2006/009049 H'N N 0 Ns N H \-N N Ar H O LCMS Ex. (M 1) 418 2-Phenylethyny1-pyridine-4-yl 448 419 5-Phenylethynyl-pyridine-2-yl 448 420 6-Cyclohexylethynyl-pyridine-2-y1 454 421 6-(4-Fluoro-phenylethyny1)-pyridine-2-y 466 422 6-(4-Ethyl-phenylethynyl)-pyridine-2-yl 476 423 6-(4-Methoxy-phenylethynyl)-pyridine-2-yl 478 424 6-(4-Chloro-phenylethynyl)-pyridine-2-y1 482 425 3-Phenylethynyl-pyridine-2-yl 448 426 6-(3-Methyl-but-1-yny)-pyridine-2-y 414 427 6-(Thiophen-3-ylethynyl)-pyridine-2-yl 454 428 6-(3,3-Dimethyl-but- 1 -ynyl)-pyridine-2-yl 428 429 6-(3-Cyclopentyl-prop- I -ynyl)-pyridine-2-yl 454 430 6-(3-Hydroxy-3-methyl-but- I -ynyl)-pyridine-2-yl 430 431 6-(4-Methyl-pent-1-ynyl)-pyridine-2-yl 428 432 6-(Pent-1-ynyl)-pyridine-2-yl 414 433 6-(4-Dimethylamino-phenylethynyl)-pyridine-2-yl 491 434 6-(Pyridin-3-ylethynyl)-pyridine-2-yI 449 435 6-(3-Methoxy-phenylethynyl)-pyridine-2-y1 478 436 6-(2-Metboxy-phenylethynyl)-pyridine-2-y1 478 437 3-(Cyclohexylethynyl)-pyridine-2-yl 454 438 3-(Thiophen-3-ylethynyl)-pyridine-2-y1 454 439 6-(Cyclopropylethynyl)-pyridine-2-yl 521 205 WO 2006/099379 PCT/US2006/009049 440 3-(3,3-Dimethyl-but-1-ynyl)-pyridine-2-y1 428 441 6-(2-Fluoro-phenylethynyl)-pyridine-2-y1 466 442 6-(n-Tolylethynyl)-pyridine-2-yl 462 443 6-(3-Fluoro-phenylethynyl)-pyridine-2-yl 575 444 3-Chloro-6-pent-1 -ynyl-pyridine-2-yl 448 445 6.-Ethynyl-pyridine-2-yl 372 446 6-phenethyl-pyridine-2-yl 452 Example 447 To a solution of 4.4 g (20 mmol) of 6-phenylethynyl-pyridine-2-carboxylic acid in DMF (30 mL) was added 7.6 g (20 mmol) of HBTU and DIEA (4 mL). The resulted mixture 5 was stirred at room temperature for 10 min, then 4 g (21 mmol) of 2-amino-iH benzoimidazole-4-carboxylic acid methyl ester (Intermediate A) was added, and stirred at room temperature for 1 h. After usual workup as described in general procedure A, 7 g (88 %) of 2-[(6-phenylethynyl-pyridine-2-carbonyl)-amino]-1H-benzoimidazole-4-carboxylic acid methyl ester was obtained after column chromatography purification eluting with 10 DCM/EtOAc. LCMS: 397 (M+1)t 'H NNMR (DMSO-d,, 400 MHz): 6 8.20 (dd, 1K), 8.13 (dd, 1H), 7-96 (d, 1H), 7.81 (d, 1H), 7.74 (d, 1H), 7.60 (m, 2H), 7.50 (m, 3H), 7.31 (t, 1H), 3.95 (s, 31) ppm. Example 448 4.2 g (10.6 mmol) of 2-[(6-phenylethynyl-pyridine-2-carbonyl)-amino-1IH 15 benzoimidazole-4-carboxylic acid methyl ester was hydrolyzed as described in general procedure B to provide 3.5 g (86%) of 2-[(6-phenylethynyl-pyridine-2-carbonyl)-amino]-1H benzoimidazole-4-carboxylic acid. LCMS: 383 (M+ 1)-. 'H NMR (DMS O-d 6 , 400 MHz): 6 8.29 (d, 1H), 8.23 (dd, 1H), 8.06 (d, 1H), 7.95 (m, 2H), 7.68 (m, 2H), 7.52 (m, 4H) ppm. To a solution of 30 mg (0.07 mmol) of above 2-[(6-Phenylethynyl-pyridine-2 20 carbonyl)-amino]-l1H-benzoimidazole-4-carboxylic acid in DMF (1 mL) was added 40 mg (0.1 mmol) of HBTU and DIEA (0.1 mL), the resulting mixture was stirred at room temperature for 10 min, then 40 mg (0.1 mmol) of 4-methanesulfonyl-benzylamine HCl salt was added. After usual work up as described in general procedure A, 21 ing (48 %) of 2-[(6 phenylethynyl-pyridine-2-carbonyl)-amino]-IH-benzoimidazole-4-carboxylic acid 4 25 methanesulfonyl-benzylamide was obtained after column chromatography purification eluting with DCMIEtOAc (v/v from 5:1 to 1:1). LCMS: 550 (M+1)t 206 WO 2006/099379 PCT/US2006/009049 By analogous methods to those used to prepare Example 448 and those in the relevant above schemes, the following compounds were synthesized. H 0 Nw H N N, N H O Ex. Ar LCMS (M + 1)' 449 cyclopentyl 450 450 tert-butyl 438 5 Example 451 60 mg (0.4 mmol) of 3-phenyl-propionic acid and 170 mg (0.45 mmol) of HBTU in a mixture of 1.0 mL of DMF and 0.5 mL of DIRA was reacted with 120 mg (0.3 mmol) of the isoquinoline-3-carboxylic acid [6-amino-4- (1H-irnidazol-2-ylcarbamoyl)-1H-benzoinidazol 10 2-ylJ-amide (See Example 130) as described in general procedure A. After the reaction was complete, it was diluted with brine (20 mL). The resulting solid was filtered, washed with water, ethyl acetate and dried under vacuum to give 101 mg (66%) of isoquinoline-3 carboxylic acid [7-(1H-imidazol-2-ylcarbamoyl)-5-(3-phenyl-propionylamino)-1H benzoimidazol-2-yl]-amideas a solid. LCMS: 545 (M + 1)+. 15 Example 452 82 mg (0.2 mmol) of the isoquinoline-3-carboxylic acid [6-amino-4- (1H-imidazol-2 ylcarbamoyl)-1H-benzoimidazol-2-y-amide (See Example 130) was dissolve in pyridine (0.6 mL). To this stirred solution at 0" C, 0.2 mmnol of phenylsulfonyl chloride in 0.2 mL of DCM was added in one portion. The reaction mixture was stirred at r.t. for 2 h. Brine (5 mL) 20 was added and stirred for 10 min. The resulting solid was collected, washed with water (3 x 2 mL) and ethyl acetate (3 x 3 mL) and dried to give 55 mg (50%) of isoquinoline-3-carboxylic acid [5-benzenesulfonylamino-7-(IH-imidazol-2-ylcarbamoyl)-1H-benzoimidazol-2-yl] amide as a solid. LCMS: 553 (M + 1)*. 207 WO 2006/099379 PCT/US2006009049 Example 453 Isoquinoline-3-carboxylic acid [4-(IH-iinidazol-2-ylcarbamoyl)-6 methanesulfonylamino- I H-benzoimidazol-2-yl]-anide (20 mg) was synthesized according to 5 the same procedure described above, with the exception that methane sulfonylchloride was used instead of phenyl sulfonylchloride. LCMS: 491 (M + I)-. Example 454 15.1 g (100 nmol) of 2-amino-5-methyl-benzoic acid was refluxed with 60 mL of ethyl chloroformate for 6h, and then was refluxed for another 3 h after addition of 60 ML of 10 acetyl chloride. The solid was collected, washed with hexane to give 14.2 g (80%) of the 5 methylisatoic anhydride. 6.9 g (40 mmol) of the above 5-Methylisatoic anhydride was suspended in 40 mL of cone. H2SO 4 at -10" C and cooled to -20" C. Next, 1.05 eq of potassium nitrate dissolved in 12 nL of sulfuric acid and cooled to 0" C, was then added dropwise. The reaction mixture 15 was stirred -20" C for 15 min then kept between -5* C and 00 C for 3h. The mixture was poured into a beaker containing ice. The resulting solid was collected, washed with cold water and dried to give 3.9 g (44 %) of 5 -methyl-3 -nitroisatoic anhydride as a solid. 3.9 g (17.6 mmol) of the above 5-methyl-3-nitroisatoic anhydride in ethyl acetate (50 mL) was treated with NaOH at r.t. for 2 h. The mixture was neutralized with dilute HC 20 solution and the resulting solid was filtered and washed with water and dried to give 2.5 g (72 %) of 2-amino-5-methyl-3-nitrobenzoic acid. 0.25 g of 2-amino-6-methyl-1H-benzoimidazole-4-carboxylic acid (lH-imidazol-2 yl)-amide was synthesized according to procedures described for the synthesis of 2-amino 3H-benzoimidazole-4-carboxylic acid (I H-imidazol-2-yl)-amide (intennediate B) starting 25 from 2.5 g (12.7 mmol) of above synthesized 2-amino-5-methyl-3-nitrobenzoic acid. LCMS: 257 (M + 1)*. 96 mg (0.5 mmnol) of isoquinoline-3-carboxylic acid and 200 mg (0.52 unol) of HBTU in a mixture of 3.0 mL of DMF and 0.3 mL of DIEA was reacted with 40 mg (0.16 mmol) of the above synthesized 2-amino-6-nethyl- 1 H-benzoimidazole-4-carboxylic acid 30 (lH-imidazol-2-yl)-amide as described in general procedure A. After the reaction was complete, it was diluted with brine (20 mL). The resulting solid was filtered, washed with water, ethyl acetate and dried under vacuum to give 32 nig (50%) of isoquinoline-3 carboxylic acid [7-iH-imidazol-2-ylcarbamoyl)-5-maethyl-IH-benzoimidazol-2-ylJ-amide. LCMS: 412 (M+ I)*. 208 WO 2006/099379 PCT/US2006/009049 Example 455 To a solution of 2.23 g (10.4 nimol) of 2-amino-3-nitro-benzoic acid methyl ester in 12 mL of acetic acid was added dropwise a solution of 0.53 niL (10.4 imnol) of Br 2 in 2.0 mL of acetic acid. The mixture was stirred at r.t. for 30 min and poured into 100 g of ice. The 5 solid was collected by filtration and dried to afford 2.5 g (82 %) of 2-Amino-5-bromo-3 nitro-benzoic acid methyl ester. 'IH NMR (CDCl 3 , 400 MHz): 6 8.6 (d, 1H), 8.53 (s, 1H), 3.95 (s, 3H) ppm. A solution of 0.5 g (1.8 mmol) of the above ester 0.9 g (2.7 mmol) of tributyl propenyl-stannane, 0.18 g (0.18 mmol mmol) of (tetrakistriphenyl phosphine) palladium (0) 10 in 25 mL of dioxane was degassed with nitrogen for 20 min then refluxed under nitrogen for 12 h. The reaction mixture was cooled, 20 mL of 2 M KF solution was added. The mixture was stirred for 20 min, extracted with ethyl acetate washed with water and brine. The residue was purified by column chromatography using 8 : 2 hexane and ethyl acetate to afford 0.35 g (77 %) of the 2-amino-3-nitro-5-propenyl-benzoic acid methyl ester as a solid LCMS: 239 15 (M+i)t. 0.3 g (85% overall yield) 2-amino-6-propyl-1H-benzoimidazole-4-carboxylic acid methyl ester was synthesized according to procedures described for the synthesis of 2 Amino-1H-benzoimidazole-4-carboxylic acid methyl ester (intermediate A) starting from 0.35 g (1.5 mmol) of above synthesized 2-amino-5-methyl-3-nitrobenzoic acid methyl ester. 20 LCMS: 234 (M + 1)+. 0.4 g (75%) of 2-[(isoquinoline-3-carbonyl)-amino]-6-propyl-1H-benzoimidazole-4 carboxylic acid methyl ester was synthesized from 0.25 g (1.4 minol) of isoquinoline-3 carboxylic acid, 0.6 g (1.6 mmol) of HBTU, and 0.3 g (1.3 mmol) of the above synthesized 2 Amino-6-propyl-1H-benzoimidazole-4-carboxylic acid methyl ester as described in general 25 procedure A. LCMS: 389 (M + 1). Example 456 0.2 g (0.5 mmol) of 2-[(isoquinoline-3-carbonyl)-amino]-6-propyl-1H benzoimidazole-4-carboxylic acid methyl ester was hydrolyzed according to the general procedure B to provide 0.17 g (89%) of the 2-[(Isoquinoline-3-carbonyl)-amino]-6-propyl 30 IH-benzoimidazole-4-carboxylic acid as a solid. LCMS: 375 (MN1 + 1)*. 0.1 (0.27 inmol) of the above synthesized acid, 0.11 g (0.29 mmol) of HBTU in a mixture of 2.0 ml of DMF and 0.20 mL of DIEA was reacted with 0.05 g (0.27 mrol) of 4 Methanesulfonyl-benzylanilne as described in general procedure A. The reaction mixture 209 WO 2006/099379 PCT/US2006/009049 was cooled to r.t. 5.0 mL of water and 5.0 mL of saturated NaHCO3 was added, and the resulting solid was filtered. The solid was washed with water, ether and dried to provide 0 .05 g (33 %) of isoquinoline-3-carboxylic acid [4-(4-nethanesulfonyl-benzylcarbamoyl)-6 propyl-IH-benzoimidazol-2-yl]-amide. LCMS: 542 (M+I)*. 5 Example 457 A solution of 1.0 g (3.6 mmol) of 2-amino-5-brono-3-nitro-benzoic acid methyl ester, 0.9 g (7.2 mmol) of pyridine-4-boronic acid, 0.42 g (0.36 inmol) of (tetrakistriphenyl phosphine) palladium (0) in 50 maL of DME and 8.4 mL of 2 N Na2,C3 was degassed with nitrogen for 20 min and then heated at 90 0 C for 12 h. The reaction mixture was cooled to r.t., 10 extracted with ethyl acetate (100 mL), washed with water, brine, dried over magnesium sulfate, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by column chromatography using 8 : 2 hexane and ethyl acetate to afford 0.5 g (50 %) of the 2-Amino-3-nitro-5-pyridin-4-yl-benzoic acid methyl ester as a solid. LCMS: 273 (M+1)+ 15 0.25 g (50 % overall yield) of 2-amino-6-pyridin-4-yl-IH-benzoimidazole-4 carboxylic acid methyl ester was synthesized according to procedures described for the synthesis of 2-Amino-1H-benzoimidazole-4-carboxylic acid methyl ester (intermediate A) starting from 0.5 g (1.8 nmol) of above synthesized 2-Amino-3-nitro-5-pyridin-4-yl-benzoic acid methyl ester. LCMS: 269 (M + 1)+. 20 0.3 g (75%) of 2-[(isoquinoline--3-carbonyl)-amino}-6-pyridin- 4 -yl-31 benzoimidazole-4-carboxylic acid methyl esterwas synthesized from 0.16 g (0.09 mmol) of isoquinoline-3-carboxylic acid, 0.4 g (1 mmol) of HBTU, and 0.25 g (0.09 mmol) of the above synthesized 2-amino-6-pyridin-4-yl-1H-benzoimidazole-4-carboxylic acid methyl ester as described in general procedure A. LCMS: 424 (M + 1). 25 Example 458 0.2 g (0.5 nimol) of above ester was hydrolyzed according to the general procedure B to provide 0.17 g (89%) of 2-[(isoquinoline-3-carbonyl)-amino]- 6 -pyridin-4-y -11H benzoimidazole-4-carboxylic acid as a solid. LCMS: 414 (M + l)+. 0.1 (0.25 nmol) of the above synthesized acid, 0.11 g (0.29 minol) of HBTU in a 30 mixture of 2.0 ml of DMF and 0.20 mL of DIEA was reacted with 0.05 g (0.27 mol) of 4 Methanesulfonyl-benzylamine as described in general procedure A. The reaction mixture was cooled to r.t. 5.0 mL of water and 5.0 mL of saturated NaHCO 3 was added and the resulting solid was filtered. The solid was washed with water, ether and dried to provide 210 WO 2006/099379 PCTIUS2006/009049 0 .05 g (33 %) of isoquinoline-3-carboxylic acid [4-(4-methanesulfonyl-benzylcarbamoyl)-6 pyridin-4-yl-1H-benzoimidazol-2-yl]-amide. LCMS: 577 (M+1). Example 459 To a stirring solution of 4.48 g (20 minol) of 2-chloro-5-trifluoromethyl-benzoic acid 5 suspended in 40 mL of conc. H 2 S0 4 at -20" C was added dropwise a cold solution of 1.05 eq of potassium nitrate dissolved in 12 mL of sulfuric acid. The reaction mixture was stirred 20" C for 15 min then warmed up to r.t. overnight. The mixture was poured into a beaker containing ice. The resulting solid was collected, washed with cold water and dried to give 4.8 g (90%) of 2-chloro-3-nitro-5-trifluoromethyl-benzoic acid as a solid. LCMS: 270 (M + 10 1)*. To a stirring solution of 0.81 g,(3.0 tmnol) of the above 2-chloro-3-nitro-5 trifluoromethyl-benzoic acid in 6.0 mL of DMF was added 6.0 mL of TEA, followed by 3.0 mmol of Benzylamine. The mixture was heated at 80" C for 6 h. After cooling to r.t., 3.0 mL of 6N HCl solution was added, The solid was collected, washed with fresh water to give 2 15 benzylamino-3-nitro-5-trifluoromethyl-benzoic acid. LCMS: 341 (M + 1)-. This product was hydrogenated according to the general procedure C to give 0.25 g of 2,3-diamino-5 trifluoromethyl-benzoic acid. LCMS: 221 (M + 1). 0.25 g of 2-amino-6-trifluoromethyl-1H-benzoimidazole-4-carboxylic acid was synthesized according to procedures described for the synthesis of 2-Amino-3H 20 benzoimidazole-4-carboxylic acid methyl ester starting from 0.25 g of above 2,3-diamino-5 trifluoromethyl-benzoic acid. This acid was refluxed in methanol and HCl/ether to give 0.26 g of 2-amino-6-trifluoromethyl -1H-benzoimidazole-4-carboxylic acid methyl ester. LCMS: 260 (M+ 1)+. 166 mg (0.9 mmol) of Isoquinoline-3-carboxylic acid and 364 mg (0.9 mmol) of 25 H1BTU in a mixture of DMF (5 ml) and DIEA (1.0 mL) was reacted with 0.25 g (0.9 rnmol) of the above amino methyl ester as described in general procedure A to give 2-[(isoquinoline 3-carbonyl)-anino}-6-trifluoromethyl-lH-benzoimidazole-4-carboxylic acid methyl ester. LCMS: 416 (M + 1)*. This ester was hydrolyzed according to general procedure B to give 0.2 g of 2 -[(isoquinoline-3-carbonyl)-amino]-6-trifluorometiyl-lH-benzoimidazole-4 30 carboxylic acid. LCMS: 401 (M + I)+. 50 mg (0.25 nmol) of the above acid and 190 ng (0.5 mmol) of HBTU in a mixture of 1.0 mL of DMF and 0.5 mL of DIEA was reacted with 100 mg (0.75 mmol) of 2 aminoimidazole sulfate as described in general procedure A. After the reaction was complete, diluted with brine (20 mL). The resulting solid was filtered, washed with water, ethyl acetate 211 WO 2006/099379 PCT/US2006/009049 and dried under vacuum to give 29 mg (25%) of isoquinoline-3-carboxylic acid [7-(1H imidazol-2-ylcarbamoyl)-5-trifluoromethyl-1H-benzoinidazol-2-yl]-amide as a solid. LCMS: 467 (M+ 1)*. Example 460 5 5.0 g (18.2 mmol) of 2-Amino-5-bromo-3-nitro-benzoic acid methyl ester was dissolved in 50 mL of NMP and treated with 2.5 g (27.3 mmol) of CuCN, then heated under reflux until the disappearance of starting material was observed by TLC (EtOAc/hexanes = 1:3). Upon completion, the contents of the reaction were poured into EtOAc followed by water. The organic layer was extracted, dried, evaporated and isolated by flash column 10 chromatography (EtOAc/hexanes = 1:9 to 3:7 gradient) to yield 1.0 g (22%) of 2-amino-5 cyano-3-nitro-benzoic acid methyl ester. 0.28 g (50 % overall yield) of 2-Ainino-6-cyano-3H-benzoimidazole-4-carboxylic acid methyl ester was synthesized according to procedures described for the synthesis of 2 Amino-3H-benzoimidazole-4-carboxylic acid methyl ester starting from 1.0 g of 2-amino-5 15 cyano-3-nitro-benzoic acid methyl ester. LCMS: 269 (M + 1)+ 0.28 g (1.3 mmol) of 2-amino-6-cyano-1H-benzoimidazole-4-carboxylic acid methyl ester was added to a mixture of 0.22 g (1.3 mrol) of isoquinoline-3-carboxylic acid monohydrate, 0.49 g (1.3 mmol) of HBTU, 3 mL of DMF and 1 mL of DIEA to produce 6 cyano-2-[(isoquinoline-3 -carbonyl)-arnino]- 1H-benzoimidazole-4-carboxylic acid methyl 20 ester, according to General Procedure A. LCMS: 373.0 (M+l)*. Example 461 0.2 g (0.56 mmol) of 6-cyano-2-[(isoquinoline-3-carbonyl)-amino]-l benzoimidazole-4-carboxylic acid methyl ester was hydrolyzed according to general procedure B to produce 2-amino-6-cyano-1H-benzoimidazole-4-carboxylic acid, which was 25 used in the next step without further purification. 0.20 g (0.56 mmol) of 6-cyano-2-[(isoquinoline-3-carbonyl)-amino]-1H benzoimidazole-4-carboxylic acid and 0.21 g (0.56 mmol) of HBTU were combined in a flask charged with 3 mL of DMF and I mL of DIEA. This reaction mixture was then treated with 0.07 g (0.56 mmol) of 2-amino-imidazole sulfate and stirred at 80*C for lh. The 30 reaction mixture was diluted with water, and the resultant solid precipitate was filtered and washed with saturated NaHC0 3 . The solid was dissolved with MeOH, evaporated onto silica gel and isolated by flash column chromatography (500 mL DCM/20 mL NH3/MeOH) to yield 10 mg of isoquinoline-3-carboxylic acid [6-cyano-4-(IH-imidazol-2-ylcarbamoy1)-1
H
benzoimidazol-2-yl]-amide. LCMS: 424.0 (M+I)*. 212 WO 2006/099379 PCT/US2006/009049 By analogous methods to those used to prepare Examples and 454-461 and those in the relevant above schemes, the following compounds were synthesized. H 0 N,. N /H N N. R N H O 5 Ex. W R LCMS (M+1)* 462 4-methanesulfonyl-benzyl phenyl 577 463 4-methanesulfonyl-benzyl isopropyl 543 464 1H-imidazol-2-yl phenyl 475 465 4-methanesulfonyl-benzyl firan-3-yl 567 466 4-methanesulfonyl-benzyl trifluoromethyl 569 467 lH-imidazol-2-yl pyridin-4-yl 476 468 4-methanesulfonyl-benzyl thiophen-3.-yl 583 469 ethyl pyridin-4-yl 438 470 cyclopentyl pyridin-4-yl 478 Example 471 10.0 mL of conc. HNO 3 at 00 C was treated with 20.0 mL of conc. H 2 S0 4 , then 6.4 g 10 (40 mmol) of 3,5-difluorobenzoic acid. The reaction mixture was warmed to room temperature and stirred overnight, then poured into a beaker containing ice. The resulting solid was collected, washed with cold water and dried to produce 8.0 g (80%) of 3,5-difluoro 2-nitro-benzoic acid. LCMS: 204 (M + 1)+, 6.0 g (30.0 mmol) of 3,5-difluoro-2-nitro-benzoic acid was dissolved in DMF, treated 15 with 8.5 g (60.0 mmol) of Mel and 9.5 g (90.0 mmol) of Na2CO3, and heated at 80 0 C for 2h. Water was added to the reaction mixture, and extraction with EtOAc produced 3,5-difluoro 2-nitro-benzoic acid methyl ester. LCMS: 218 (M + I)*. 213 WO 2006/099379 PCT/US2006/009049 6.0 g (27.5 mmol) of 3,5-difluoro-2-nitro-benzoic acid methyl ester was dissolved in DMF and treated with 2.9 g (30.25 mmol) of amimonium carbonate. The reaction was heated at 60" C for 6 h. Water was added to the reaction mixture. The solid produced was collected by filtration and washed with water to produce 3-anino-5-fluoro-2-nitro-benzoic acid methyl 5 ester. LCMS: 215 (M + 1)*. 5.0 g (23.3 mmol) of 3-amino-5-fluoro-2-nitro-benzoic acid methyl ester was dissolved in ethanol and treated with 2.6 g (25.6 mmol) of triethylamine and 1.8 g (25.6 mmol) of NaOEt. The reaction was heated at 60 * C for 0.5 h to produce 3-amino-5-ethoxy 2-nitro-benzoic acid ethyl ester. LCMS: 255 (M + 1)*. 10 5.0 g (19.7 mmol) of 3-axnino-5-ethoxy-2-nitro-benzoic acid ethyl ester was hydrogenated according to general procedure C to produce 2,3-dianino-5-ethoxy-benzoic acid ethyl ester. LCMS: 225 (M + 1). 4.0 g (17.9 mmol) of 2,3-diamino-5-cthoxy-benzoic acid ethyl ester was subjected to a similar cyclization condition as illustrated in intermediate A by using 5.7 g (53.7 mmol) of 15 BrCN in methanol to produce 2-amino-6- ethoxy- 1 H-benzoimidazole-4-carboxylic acid ethyl ester. LCMS: 250 (M + 1)+. 600 mg (3.0 mmol) of isoquinoline-3-carboxylic acid and 1200 mg (3.1 mmol) of UBTU were dissolved in 3.0 mL of DMF and 0.6 mL of DIEA and reacted with 500 mg (2.0 mmol) of 2-amino-6-ethoxy-lH-benzoimidazole-4-carboxylic acid ethyl ester as described in 20 general procedure A. Upon completion, the reaction was diluted with brine. The resulting solid was filtered, washed with water and EtOAc, and dried under vacuum to produce 500 mg (62%) of 6-ethoxy-2-[(isoquinoline-3-carbonyl)-amino]-3H-benzoimidazole- 4 -carboxylic acid ethyl ester. LCMS: 406 (M + 1)*. Example 472 25 0.3 g (0.74 mmol) of 6-ethoxy-2-[(isoquinoline-3-carbonyl)-aminol-3H benzoimidazole-4-carboxylic acid ethyl ester was hydrolyzed according to general procedure B to produce 2-[(isoquinoline-3-carbonyl)-anino]-6-ethoxy-lHbenzoiiidazole- 4 -carboxylic acid. LCMS: 378 (M + 1). 100 mg (0.3 mmol) of 2-(isoquinoline-3-carbonyl)-amino]-6-ethoxy-lH 30 benzoimidazole-4-carboxylic acid and 200 mg (0.53 mmol) of HBTU were dissolved in 1.0 mL of DMF and 0.5 mL of DIEA and reacted with 132 ng (1.0 mmol) of 2-aminoimidazole sulfate as described in general procedure A. Upon completion, the reaction was diluted with brine. The resulting solid was filtered, washed with water and EtOAc, and dried under 214 WO 2006/099379 PCT/US2006/009049 vacuum to produce 30 mg (23%) of isoquinoline-3-carboxylic acid [5-ethoxy-7-(1H imidazol-2-ylcarbamoyl)-liH-benzoinidazol-2-yl]-amide. LCMS: 443 (M + 1)*. Example 473 3.0 g (14.0 mmol) of 3-amino-5-fluoro-2-nitro-benzoic acid methyl ester in 1.6 g 5 (15.4 mmol) of triethylamine and 2.1 g (15.4 irmiol) of NaOBn/BnOH was heated at 60"C for 0.5 h to produce 3-amino-5-benzyloxy-2-nitro-benzoic acid benzyl ester. LCMS: 380 (M + 1)W. 3.0 g (7.9 mmol) of 3-anino-5-benzyloxy-2-nitro-benzoic acid benzyl ester was reduced by 5.4 g (23.7 mmol) of SnClj2H 2 0 in 30 mL of ethanol at 80"C for 3 h to produce 10 2,3-diamino-5-benzyloxy-benzoic acid benzyl ester. LCMS: 350 (M + 1)-. 2.5 g (7.2 mmol) of 2,3-diamino-5-benzyloxy-benzoic acid benzyl ester was subjected to a similar cyclization condition as illustrated in intermediate A by using 2.3 g (21.6 mmol) of BrCN in methanol to produce 2-amino-6-benzyloxy-1H-benzoimidazole-4-carboxylic acid benzyl ester. LCMS: 375 (M + 1)*. 15 600 mg (3.0 mmol) of isoquinoline-3-carboxylic acid and 1200 ing (3.1 mmol) of HBTU were dissolved in 3.0 mL of DMF and 0.6 mL of DIEA and reacted with 1.1 g (3.0 mmol) of 2-amino-6-benzyloxy- 1 H-benzoimidazole-4-carboxylic acid benzyl ester as described in general procedure A to produce 6-benzyloxy-2-{(isoquinoline-3-carbonyl) amino]-3H-benzoimidazole-4-carboxylic acid benzyl ester. LCMS: 530 (M + 1)*. 20 0.7 g (1.3 mmol) of 6-benzyloxy-2-[(isoquinoline-3-carbonyl)-anino]-3H benzoimidazole-4-carboxylic acid benzyl ester was hydrolyzed according to general procedure B to produce 6-benzyloxy-2-[(isoquinoline-3-carbonyl)-amino]-3H benzoimidazole-4-carboxylic acid. LCMS: 440 (M + 1)+. 300 mg (0.75 mmol) of 6-benzyloxy-2-[(isoquinoline-3-carbonyl)-amino]-3H 25 benzoimidazole-4-carboxylic acid and 600 mg (1.58 mmol) of HBTU were dissolved in 2.0 mL of DMF and 1.0 mL of DIEA and reacted with 390 mg (3.0 nmol) of 2-aminoimidazole sulfate as described in general procedure A. Upon completion, the reaction was diluted with brine. The resulting solid was filtered, washed with water and EtOAc, and dried under vacuum to produce 300 mg (80%) of isoquinoline-3-carboxylic acid [5-benzyloxy-7-(IH 30 imidazol-2-ylcarbamoyl)-lH-benzoimidazol-2-y]-amide. LCMS: 505 (M + 1)t By analogous methods to those used to prepare Examples 471-473 and those in the relevant above schemes, the following compounds were synthesized. 215 WO 2006/099379 PCT/US2006/009049 H I 0 N' W N /H \N
N
R, H O Ex. W R LCMS (M+1)+ 474 1H-imidazol-2-yl methyl 429 475 4-nethanesulfonyl-benzyl methyl 531 476 1H-imidazol-2-yl propyl 457 477 1H-imidazol-2-yl isopropyl 457 478 1H-imidazol-2-yl butyl 471 Example 479 A solution of 2.65 mL (52.0 mmol) of bromine in 10 mL of acetic acid was added 5 dropwise over 15 minutes to a solution of 8.89 g (52.0 mmol) of 2-amino-4-chloro-benzoic acid in 60 mL of acetic acid. The mixture was stirred at room temperaturefor 1 h and poured into 500 grams of ice. The solid was collected by filtration, washed with cold water and dried to produce 11 2 g (86%) of 2-amino-5-bromo-4-chloro-benzoic acid. LCMS: 251 (M + 1). 10 7.5 g (30 mmol) of 2-amino-5-bromo-4-chloro-benzoic acid was heated under reflux with 18 mL of ethyl chloroformate for 6 h, and was further heated under reflux for another 3 h after addition of 18 mL of acetyl chloride. The solid was collected and washed with hexanes to produce 5.8 g (70%) of 5-bromo-4-chloroisatoic anhydride. LCMS: 277 (M + 1). 5.4 g (20 mmol) of 5-bromo-4-chloroisatoic anhydride was suspended in 20 ml of 15 conc. H 2 S0 4 at -10 C and cooled to -20* C. 2.1 g (21 mmol) of potassium nitrate was dissolved in 6 mL of sulfuric acid and cooled to 00 C, was then added dropwise to the reaction mixture. The reaction mixture was then stirred -104 C for 15 min and warmed to room temperature overnight. The mixture was poured in to a beaker containing ice. The resulting solid was collected by filtration, washed with cold water and dried to produce 3.2 g 20 (50%) of 5-bromo-4-chloro-3-nitroisatoic anhydride. LCMS: 323 (M+ 1)+. 216 WO 2006/099379 PCT/US2006/009049 2.0 g (6.2 mmol) of 5-bromo-4-chloro-3-nitrolisatoic anhydride was dissolved in methanol and treated with 2.02 g (37.4 mmol) of NaOMe. The reaction mixture was heated at 100* C for 12 h to produce 2-amino-5-bromo-4-methoxy-2-nitro-benzoic acid. LCMS: 292 (M + I)-. 5 1.5 g (5.15 mmol) of 2-amino-5-bromo-4-niethoxy-2-nitro-benzoic acid was hydrogenated according to general procedure C to produce 2,3-diamino-4-methoxy-benzoic acid. LCMS: 183 (M + 1)'. 0.9 g (4.9 mmol) of 2,3-diamino-4-methoxy-benzoic acid was subjected to a similar cyclization condition as illustrated in intermediate A by using 2.6 g (24.5 mmol) of BrCN in 10 methanol to produce 2-amino-7-nethoxy-IH-benzoinidazole-4-carboxylic acid. LCMS: 208 (M + 1). 1.0 g (4.8 mmol) of 2-amino-7-methoxy-IH-benzoimidazole-4-carboxylic acid was heated under reflux in methanol and HC1/ether to produce 2-amino-7-methoxy-1H benzoimidazole-4-carboxylic acid methyl ester. LCMS: 222 (M + I). 15 300 mg (1.5 mmol) of isoquinoline-3-carboxylic acid and 570 mg (1.50 mmol) of HBTU were dissolved in 1.0 nL of DMF and 0.8 mL of DIEA and reacted with 110 mg (0.5 mmol) of 2-amino-7-methoxy- 1 H-benzoimidazole-4-carboxylic acid methyl ester as described in general procedure A. Upon completion, the reaction was diluted with brine. The resulting solid was filtered, washed with water and EtOAc, and dried under vacuum to 20 produce 70 mg (37%) of 2-[(isoquinoline-3-carbonyl)-amino]-7-metboxy-3H benzoimidazoe-4-caboxylic acid methyl ester. LCMS: 378 (M + 1)+. Example 480 0.06 g (0.16 mmol) of 2-[(Isoquinoline-3-carbonyl)-amino}-7-methoxy-34H benzoimidazole-4-carboxylic acid methyl ester was hydro lyzed according to general 25 procedure B to produce 0.05 g (0.14 mmcol, 87%) of 2-[(isoquinoline-3-carbonyl)-amino]-7 inethoxy-1H-benzoimidazole-4-carboxylic acid. LCMS: 363 (M + I)-. 33 mg (0.1 mmol) of 2-{(isoquinoline-3-carbonyl)-amino]-7-methoxy-1H benzoimidazole-4-carboxylic acid and 100 mg (0.25 mmol) of HBTU were dissolved in 1.0 nmL of DMF and 0.4 mL of DJEA and reacted with 32 mg (0.15 mmol) of 4-methanesulfonyl 30 benzylamine hydrochloride as described in general procedure A. Upon completion, the reaction was diluted with brine. The resulting solid was filtered, washed with water and EtOAc, and dried under vacuum to produce 14 mg (26%) of isoquinoline-3-carboxylic acid
[
7
-(
4 -methanesulfonyl-benzylcarbamoyl)-4-imethoxy-IH-benzoinidazol-2-yl-amide. LCMS: 530 (M + l)-. 217 WO 2006/099379 PCT/US2006/ 0 0 9 0 49 Example 481 5.0 mL of conc. HN0 3 at 0" C was slowly treated with 10.0 mL of cone. H 2
SO
4 , then 3.2 g (20 mmol) of 2,6-difluoro-benzoic acid. The reaction mixture was warmed to room temperature and stirred overnight. The mixture was poured into a beaker containing ice. The 5 resulting solid was collected by filtration, washed with cold water and dried to produce 4.0 g (80%) of 2,6-difluoro-3-nitro-benzoic acid as a white solid. LCMS: 204 (M + W. 2.0 g (9.9 mmol) of 2,6-difluoro-3-nitro-benzoic acid in methanol and 3.0 g (30.0 mmol) of ammonium acetate was stirred at room temperature for 12 h. The solvent was evaporated and dilute HC1 solution was added. The precipitated solid was collected by 10 filtration and washed with water to produce 2-amino-6-fluoro-3--nitro-benzoic acid. LCMS: 201 (M + 1). 1.8 g (9.0 mmol) of 2-amino-6-fluoro-3-nitro-benzoic acid in methanol and 2.9 g (54 mmol) of NaOMe was heated at 900 C overnight to produce 1.6 g (7.5 mmol 83%) of 2 amino-6-methoxy-3-nitro-benzoic acid. LCMS: 213 (M + 1). 15 1.0 g (4.7 mmol) of 2-amino-6-methoxy-3-nitro-benzoic acid was hydrogenated according to general procedure C to produce 2,3-diamino-6-methOxy-benzoic acid. LCMS: 183 (M+ 1). 0.8 g (4.4 mmol) of 2,3-diamino-6-methoxy-benzoic acid was subjected to a similar cyclization condition as illustrated in intermediate A by using 0.7 g (6.6 nimol) of BrCN in 20 methanol to produce 2-amino-5-methoxy-1H-benzoimidazole-4-carboxylic acid. LCMS: 208 (M + 1)t 0.9 g (4.3 mmol) of 2-amino-5-methoxy-1H-benzoimidazole-4-carboxylic acid was heated under reflux in methanol and HC1/ether to produce 2-amino-5-.methoxy-1H benzoimidazole-4-carboxylic acid methyl ester. LCMS: 222 (M + 1)> 25 400 mg (2.0 mumol) of isoquinoline-3-carboxylic acid and 800 mg (2.1 mmol) of HBTU were dissolved in 2.0 mL of DMF and 1.3 ml of DIEA and treated with 300 mg (1.5 mmol) of 2-amino-5-methoxy-1H-benzoimidazole-4-carboxylic acid methyl ester as described in general procedure A. Upon completion, the reaction was diluted with brine. The resulting solid was filtered, washed with water and EtOAc, and dried under vacuum to 30 produce 160 mg (28%) of 2--(isoquinoline-3-carbonyl)-amino]-5-methoxy-3H benzoimidazole-4-carboxylic acid methyl ester. LCMS: 377 (M + 1)4, Example 482 0.14 g (0.38 mmol) of 2-[(Isoquinoline-3-carbonyl)-aminol-5-methoxy-3H benzoimidazole-4-carboxylic acid methyl ester was hydrolyzed according to general 218 WO 2006/099379 PCT/US2006/009049 procedure B to produce 0.12 g (0.33 mmol, 87%) of 2-[(isoquinoline-3-carbonyl)-amino]-5 methoxy-1H-benzoimidazole-4-carboxylic acid. LCMS: 363 (M + I)-. 66 mg (0.18 mmol) of 2-[(isoquinoline-3-carbonyl)-amino]-5-methoxy-1H benzoimidazole-4-carboxylic acid and 200 mg (0.53 mmol) of HBTU were dissolved in 1.0 5 mL of DMF and 0.5 mL of DIEA was treated with 132 mg (1.0 nmol) of 2-aminoimidazole sulfate as described in general procedure A. Upon completion, the reaction was diluted with brine. The resulting solid was filtered, washed with water and EtOAc, and dried under vacuum to produce 26 ing (34%) of isoquinoline-3-carboxylic acid {7-(IH-imidazol-2 ylcarbanoyl)-6-nethoxy-1H-benzoimidazol-2-yl]-amide. LCMS: 428 (M + 1)*. 10 Example 483 33 mg (0.1 rmol) of 2-[(isoquinoline-3-carbonyl)-aminol-5-methoxy-lH benzoimidazole-4-carboxylic acid and 100 mg (0.25 mmol) of HBTU were dissolved in 1.0 mL of DMF and 0.4 mL of DIEA was treated with 32 mg (0.15 mmol) of 4-methanesulfonyl benzylamine hydrochloride as described in general procedure A. Upon completion, the 15 reaction was diluted with brine. The resulting solid was filtered, washed with water and EtOAc, and dried under vacuum to produce 25 mg (47%) of isoquinoline-3-carboxylic acid [7-(4-methanesulfonyl-benzylcarbamoyl)-6-methoxy-IH-benzoimidazol-2-yl]-amide. LCMS: 530 (M+ 1)+. Example 484 20 2.0 g (8.44 mmol) of 6,7-dimethoxy-1,2,3,4-tetrahydro-isoquinoline-3-carboxylic acid was heated under reflux in methanol and HCI/ether to produce 2.1 g (8.40 mmol, 99%) of 6,7-dimethoxy-1,2,3,4-tetrahydro-isoquinoline-3-carboxylic acid methyl ester, LCMS: 252 (M + 1)*. 1.8 g (6.26 mmol) of 6,7-dimethoxy-1,2,3,4-tetrahydro-isoquinoline-3-carboxylic acid 25 methyl ester and 3.3 g (37.6 mmol) of MnO 2 in toluene/dioxane/THF was heated under reflux overnight. After gravity filtration, the hot filtrate was evaporated to produce 0.4 g (1.62 mmol, 26%) of 6,7-dimethoxy-isoquinoline-.3-carboxylic acid methyl ester. LCMS: 248 (M + 1). 0.4 g (1.62 mmol) of 6,7-dimethoxy-isoquinoline-3-carboxylic acid methyl ester was 30 hydrolyzed according to general procedure 13 to produce 0.38 g (1.60 nnnol, 98%) of 6,7 dimethoxy-isoquinoline-3-carboxylic acid. LCMS: 234 (M + 1)*. 235 mg (1.0 mmol) of 6,7-dimethoxy-isoquinoline-3-carboxylic acid and 400 mg (1.0 mmol) of HBTU were dissolved in 1.5 mL of DMF and 0.5 mL of DIEA and treated with 249 mg (1.0 mmol) of 2-amino-6-ethoxy-IH-benzoimidazole-4-carboxylic acid ethyl ester 219 WO 2006/099379 PCT/US2006/009049 (prepared in Example 471) as described in general procedure A. Upon completion, the reaction was diluted with brine. The resulting solid was filtered, washed with water and EtOAc, and dried under vacuum to produce 290 mg (0.63 mnol, 63%) of 2-[(6,7-dimethoxy isoquinoline-3-carbonyl)-amino]-6-ethoxy-1 H-b enzoimidazole-4-carboxylic acid ethyl ester. 5 LCMS: 465 (M + 1). This 2-[(6,7-dimethoxy-isoquinoline-3-carbonyl)-amino]-6-ethoxy-1H benzoimidazole-4-carboxylic acid ethyl ester was hydrolyzed according to general procedure B to produce 2-[(6,7-dimetlioxy-isoquinoline-3-carbonyl)-anino]-6-ethoxy-lH benzoimidazole-4-carboxylic acid. LCMS: 437 (M + 1)t. 10 100 mg (0.25 mmol) of 2-[(6,7-dimethoxy-isoquinoline-3-carbonyl)-amino}-6 ethoxy-l fH-benzoimidazole-4-carboxylic acid and 200 mug (0.53 mmol) of HBTU were dissolved in 1.0 mL of DMF and 0.5 mL of DIEA and treated with 200 mg (1.5 mmol) of 2 aminoimidazole sulfate as described in general procedure A. Upon completion, the reaction was diluted with brine. The resulting solid was filtered, washed with water and EtOAc, and 15 dried under vacuum to produce 60 mg (47%) of 6,7-dimethoxy-isoquinoline-3-carboxylic acid [5-ethoxy-7-(1H-imidazol-2-ylcarbamoyl)-1H-benzoimidazol-2-yl]-amide. LCMS: 502 (M + 1)-. Example 485 5.0 mL of conc. HN0 3 at 0" C was slowly treated with 10.0 ml of conc. H 2
SO
4 , then 20 3.52 g (20.0 mmol) of 3,4,5-trifluoro-benzoic acid. The reaction mixture was warmed up to room temperature and stirred overnight. The mixture was poured into a beaker containing ice. The resulting solid was collected by filtration, washed with cold water and dried to produce 3.4 g (16.0 mmol, 80%) of 2-nitro-3,4,5-trifluoro-benzoic acid. LCMS: 222 (M + 1)+. 1.4 g (6.3 mmol) of 2-nitro-3,4,5-trifluoro-benzoic acid was treated with ammonium 25 hydroxide to produce 3-amino-4,5-difluoro-2-nitro-benzoic acid. LCMS: 219 (M + 1). 1.2 g (5.5 mmol) of 3-amino-4,5-difluoro-2-nitro-benzoic acid was heated with excess 21% NaOEt in ethanol at 90" C for 6 h to yield 3-amino-5-ethoxy-4-fluoro-2-nitro-benzoic acid. LCMS: 245 (M + 1) . 1.0 g (4.1 mmiol) of 3 -amino-5-ethoxy-4-fluoro-2-nitro-benzoic acid was 30 hydrogenated according to general procedure C to produce 2,3-diamino-5-ethoxy-4-fluoro benzoic acid. LCMS: 215 (M + 1)t. 0.8 g (3.7 nmol) of 2 ,3-diamino-S-ethoxy-4-fluoro-benzoic acid was subjected to a similar cyclization condition as illustrated in intermediate A by using 1.2 g (11.2 mmol) of 220 WO 2006/099379 PCT/US2006/009049 BrCN in methanol to produce 2-amino-6-ethoxy-7-fluoro-lH-benzoimidazole-4-carboxylic acid. LCMS: 240 (M + 1)*. 0.8 g (3.3 mmol) of 2-amino-6-ethoxy-7-fluoro-IH-benzoimidazole-4-carboxylic acid was heated under reflux in methanol and HCI/ether to produce 2-amino-6-ethoxy-7-fluoro 5 1H-benzoimidazole-4-carboxylic acid methyl ester. LCMS: 254 (M + 1)+. 0.3 g (1.5 mmol) of isoquinoline-3-carboxylic acid and 0.6 g (1.6 mmol) of HIBTU were dissolved in 3.0 mL of DMF and 0.6 mL of DIEA and treated with 0.25 g (1.0 mmol) of 2-amino-6-ethoxy-7-fluoro-1H benzoimidazole-4-carboxylic acid methyl ester as described in general procedure A to produce 0.18 g (0.44 mmol, 44%) of 6-ethoxy-7-fluoro-2 10 [(isoquinoline-3-carbonyl)-amino]-3H-benzoimidazole-4-carboxylic acid methyl ester. LCMS: 409 (M + 1). 0.17 g (0.42 mmol) of 6-ethoxy-7-fluoro-2-{(isoquinoline-3-carbonyl)-amino]-3H benzoimidazole-4-carboxylic acid methyl ester was hydrolyzed according to general procedure B to produce 0.16 g (0.41 mmol, 98%) of 6-ethoxy-7-fluoro-2-{(isoquinoline-3 15 carbonyl)-amino]-3H-benzoimidazole-4-carboxylic acid. LCMS: 395 (M + I). 30 mg (0.08 mmol) of 6-ethoxy-7-fluoro-2-[(isoquinoline-3-carbonyl)-amino]-3H benzoimidazole-4-carboxylic acid and 100 mg (0.25 mmol) of HBTU were dissolved in 1.0 mL of DMF and 0.3 mL of DIEA and treated with 30 mg (0.14 mmol) of 4-methanesulfonyl benzylamine hydrochloride as described in general procedure A. Upon completion, the 20 reaction was diluted with brine. The resulting solid was filtered, washed with water and EtOAc, and dried under vacuum to produce 19 mg (45%) of isoquinoiine-3-carboxylic acid [5-ethoxy-4-fluoro-7-(4-methanesulfonyl-benzylcarbamoyl)-1H-benzoimidazol-2-yl]-amide. LCMS: 562 (M+ 1) Example 486 25 60 mg (0.15 mmol) of 6-ethoxy-7-fluoro-2-{(isoquinoline-3-carbonyl)-amino]-3H benzoiinidazole-4-carboxylic acid (from Example 485) and 100 mg (0.25 mmol) of HBTU were dissolved in 1.0 mL of DMF and 0.2 mL of DIEA and treated with 50 mg (0.5 mmol) of 2-aminoimidazole sulfate as described in general procedure A. Upon completion, the reaction was diluted with brine. The resulting solid was filtered, washed with water and 30 EtOAc, and dried under vacuum to produce 32 mug (46%) of Isoquinoline-3-carboxylic acid [5-ethoxy-4-fluoro-7-(1H-imidazol-2-ylcarbamoyl)-1H-benzoimidazol-2-yl]-amide. LCMS: 460 (M + I)t 221 WO 2006/099379 PCT/US2006/009049 Example 487 5.0 mL of conc. HN0 3 at 00 C was slowly treated with 10.0 mL of cone. H 2 S0 4 , then 4.12 g (20 mmol) of 3-methoxy-2,4,5-trifluoro-benzoic acid. The reaction mixture was warmed to room temperature and stirred overnight. The mixture was poured in to a beaker 5 containing ice. The resulting solid was collected by filtration, washed with cold water and dried to produce 4.0 g (16 mmol, 80%) of 3-methoxy-6-nitro-2,4,5-trifluoro-benzoic acid. LCMS: 252 (M + 1). 2.0 g (8 mmol) of 3-methoxy-6-nitro-2,4,5-trifluoro-benzoic acid was treated with ammonium hydroxide to produce 1.6 g (6.45 mmol, 80%) of 3-amino-4,6-difluoro-5 10 methoxy-2-nitro-benzoic acid. LCMS: 249 (M + 1). 1.6 g (6.45 mmol) of 3-amino-4,6-difluoro-5-methoxy-2-nitro-benzoic acid was hydrogenated according to general procedure C to produce 2,3-diamino-4,6-difluoro-5 methoxy-benzoic acid. LCMS: 219 (M + 1), 1.2 g (5.5 mmol) of 2,3-diamino-4,6-difluoro-5-methoxy-benzoic acid was subjected 15 to a similar cyclization condition as illustrated in intermediate A by using 1.7 g (16.5 mmol) of BrCN in methanol to produce 2-amino-5,7-difluoro-6-methoxy- 1 H-benzoimidazole-4 carboxylic acid. LCMS: 244 (M + 1)+. I.Og (4.1 mmol) of 2-amino-5,7-difluoro-6-methoxy-1H-benzoimidazole-4-carboxylic acid was heated under reflux in methanol and HCi/ether to produce 0.9 g (3.6 mmol, 88%) of 20 2-ainino-5,7-difluoro-6-meithoxy-IH-benzoimidazole-4-carboxylic acid methyl ester. LCMS: 258 (M+ 1). 0.4 g (2.0 mmol) of isoquinoline-3-carboxylic acid and 800 mg (2.1 mmol) HBTU were dissolved in 3.0 mL of DMF and 0.6 mL of DIEA and treated with 0.35 g (1.5 mmol) of 2-amino-5,7-difluoro-6-methoxy-1 H-benzoimidazole-4-carboxylic acid methyl ester as 25 described in general procedure A to produce 0.3 g (0.73 mmol, 49%) of 5,7-difluoro-2 [(isoquinoline-3 -carbony1)-amino]-6-methoxy-3H-benzoimidazole-4-carboxylic acid methyl ester. LCMS: 413 (M + 1). 0.21 g (0.5 mnmol) of 5,7-difluoro-2-[(isoquinoline-3-carbonyl)-amnino]-6-methoxy 3H-benzoimidazole-4-carboxylic acid methyl ester was hydrolyzed according to general 30 procedure B to produce 0.20 g (0.5 mmol, 100%) of 5,7-difluoro-2-[(isoquinoline-3 carbonyl)-amino]-6-methoxy-3H-benzoinidazole-4-carboxylic acid. LCMS: 399 (M + 1)+. 60 mg (0.15 mmol) of the above 5,7-difluoro-2-[(isoquinoline-3-carbonyl)-amino]-6 methoxy-3H-benzoimidazole-4-carboxylic acid and 100 mg (0.25 mmol) of HBTU were dissolved in 1.0 mL of DMF and 0.5 mL of DIEA and treated with 68 mg (0.5 nmol) of 2 222 WO 2006/099379 PCT/US2006/009049 aminoimidazole sulfate as described in general procedure A. Upon completion, the reaction was diluted with brine. The resulting solid was filtered, washed with water and EtOAc, and dried under vacuum to produce 24 mg (35%) of isoquinoline-3-carboxylic acid [4,6-difluoro 7-(lH-imidazol-2-ylcarbamoyl)-5-methoxy-1H-benzoimidazol-2-yl-amide. LCMS: 464 (M 5 + 1. Example 488 A mixture of 2.01 g (10 mmol) 2-hydroxy-3-nitro-benzoic acid methyl ester, 0.8 g of palladium (10 wt.% on carbon powder) and 30 mL of methanol was hydrogenated at 40 psi overnight. The reaction mixture was filtered over celite. Evaporation of the solvents in 10 vacuo gave 3-Amino-2-hydroxy-benzoic acid methyl ester, LCMS: 168 (M+l)*. A mixture of 913 mg (5.46 mmol) 3-amino-2-hydroxy-benzoic acid methyl ester, 894 mg (8.i19 mmol) of cyanogen bromide, 14.4 mL of ethanol and 1.6 mL of water was heated under reflux for 1 h. Ethanol was evaporated under reduced pressure and 20 mL of 2 N aq. Na 2
CO
3 was added to the residue. The precipitated solid was filtered and washed with water. 15 The remaining product was treated with methanol and the soluble filtrate containing the product was evaporated in vacuo to provide 797 mg (4-15 mmol, 76%) of 2-amino benzooxazole-7-carboxylic acid methyl ester. LCMS: 193 (M+)*. 2 -[(Isoquinoline- 3 -carbonyl)-amino]-benzooxazole-7-carboxylic acid methyl ester was prepared according to general procedure A using 1.052 g (5.5 mmol) of isoquinoline 3 20 carboxylic acid hydrate, 797 mg (4.15 mnnol) of 2-amino-benzooxazole-7-carboxylic acid methyl ester, 2.09 g (5.5 mmol) of HBTU, 1.58 mL (9 nmnol) of DIEA and 14 mL of DMF stirring at 901C for 3 h. 684 mg (1.97 mmol, 48%) of the product was obtained. LCMS: 348 (M+l). 2 -[(Isoquinoline-3-carbonyl)-amino]-benzooxazole-7-carboxylic acid was synthesized 25 according to General Procedure B using 417 mg (1.2 nniol) 2 -[(isoquinoline-3-carbonyl) amino]-benzooxazole-7-carboxylic acid methyl ester, 2.4 mL of 2 N aq. LiOH, 12 mL of THF and 3 mL of methanol at 40*C for 4 b. 141 mg (0.423 mmol, 35%) of the product was obtained. LCMS: 334 (M+1)*. Isoquinoline-3-carboxylic acid [7-(IH-imidazol-2-ylcarbamoyl)-benzooxazol-2-y] 30 aide was synthesized according to General Procedure A using 137 rmg (0.411 mmol) of 2 [(isoquinoline-3-carbonyl)-aiinol-benzooxazole-7-carboxylic acid, Il1 mg (0.822 mmol) 2 aminoimidazole sulfate, 171 mg (0.45 mmol) HBTU, 0.29 nL (1.65 mmol,) of DIEA and 2 223 WO 2006/099379 PCT/US2006/009049 mL of DMF stirring at 90 0 C for 2 h. 98.9 mg (0.248 mmol, 60%) of the product was obtained. LCMS: 399 (M+1)+. Example 489 10.0 mL of conc. HNO 3 at 0" C was slowly treated with 20.0 mL of conc. H 2 S0 4 , then 5 7.6 g (40 mmol) of 3,5-difluoro-benzoic acid ethyl ester. The reaction mixture was wanted up to room temperature and stirred overnight. This mixture was poured into a beaker containing ice. The resulting mixture was extracted with ethyl acetate, washed with cold water and evaporated to produce 7.4 g (32 mmol, 80%) of 3,5-difluoro-2-nitro-benzoic acid ethyl ester. LCMS: 232 (M + 1)*. 10 4.64 g (20 mmol) of 3,5-difluoro-2-nitro-benzoic acid ethyl ester in DMF and 2.1 g (22 mmol) of ammonium carbonate was heated at 60* C for 6 h. The reaction mixture was treated with water. The resulting solid was collected by filtration and washed with water to produce 3.4 g (15.0 mmol, 75%) of 3-amino-5-fluoro-2-nitro-benzoic acid ethyl ester. LCMS: 229 (M+ 1)+. 15 2.31 g (10 mmol) of 3-amino-5-fluoro-2-nitro-benzoic acid ethyl ester in DMF, 1.1g (II mmol) of triethylamine and 1.4 g (16 mmol) of NaSEt were heated at 80" C for 6 h. After cooling to room temperature, EtOAc extraction provided 3-amino-5-ethylsulfanyl-2-nitro benzoic acid ethyl ester. LCMS: 271 (M + 1)*. 1.5 g (5.5 mmol) of 3-amino-5-ethylsulfanyl-2-nitro-benzoic acid ethyl ester was 20 reduced by 2.9 g (16.6 mmol) of Na 2
S
2
O
4 in 28 mL of ethanol/ethyl acetate/water (3:3:1) at 80* C for 3 h to produce 2,3-diamino-5-ethylsulfanyl-benzoic acid ethyl ester. LCMS: 241 (M + 1)t. 0.9 g (3.75 mmol) of 2,3-diamino-5-ethylsulfanyl-benzoic acid ethyl ester was subjected to a similar cyclization condition as illustrated in intermediate A by using 1.2 g 25 (11.3 mmol) of BrCN in methanol to produce 0.9 g (3.4 mmol, 90%) of 2-amino-6 ethylsulfanyl-IH-benzoimidazole-4-carboxylic acid ethyl ester. LCMS: 266 (M + 1). 0.4 g (2.0 mmol) of isoquinoline-3-carboxylic acid and 800 mg (2.1 mmol) HBTU were dissolved in 3.0 mL of DMF and 0.6 mL of DIEA and treated with 0.26 g (1.0 mmol) of 2-amino-6-ethylsulfanyl-1H-benzoimidazole-4-carboxylic acid ethyl ester as described in 30 general procedure A to produce 0.35 g (0.83 mmol, 83%) of 6- ethylsulfanyl-2 [(isoquinoline-3-carbonyl)-amino]-31-benzoimidazole-4-carboxylic acid ethyl ester. LCMS: 421 (M + 1). 224 WO 2006/099379 PCT/US2006/009049 0.34 g (0.81 mmol) of 6- ethylsulfanyl-2-[(isoquinoline-3-carbonyt)-amino}-3H benzoimidazole-4-carboxylic acid ethyl ester was hydrolyzed according to general procedure B to produce 0.30 g (0.77 mmol, 95%) of 6-ethylsulfanyl-2-[(isoquinoline-3-carbonyl) amino]-3H-benzoimidazole-4-carboxylic acid. LCMS: 393 (M + )*. 5 80 ing (0.2 mmol) of 6-ethylsulfanyl-2-[(isoquinoline-3-carbonyl)-anino]-3H benzoimidazole-4-carboxylic acid and 300 mg (0.76 mmol) of HBTU were dissolved in 1.0 mL of DMF and 0.5 mL of DIEA and treated with 132 mg (1.0 mmol) of 2-aminoimidazole sulfate as described in general procedure A. Upon completion, the reaction was diluted with brine. The resulting solid was filtered, washed with water and EtOAc, and dried under 10 vacuum to produce 66 mg (71%) of isoquinoline-3-carboxylic acid [5-ethylsulfanyl-7-(1H imidazol-2-ylcarbamoyl)-1H-benzoimidazol-2-yl]-amide. LCMS: 458 (M+ 1)*. Example 490 Isoquinoline-3-carboxylic acid [5-butylsulfanyl-7-(IH-imidazol-2-ylcarbamoyl)-1H benzoimidazol-2-yl]-amide (15 mg) was synthesized by analogous procedures to those used 15 to prepare the Example 489, with the exception that NaS(CH 2
)
3
CH
3 was used instead of NaSEt, Example 491 0.26 g (1.0 mmol) of 2-amino-6-ethylsuLfanyl-1H-benzoimidazole-4-carboxylic acid ethyl ester (See Example 489) in DCM was stirred with 0.35 g (2.0 nnol) of MCPBA and 20 0.34 g (4.0 mmol) of NaHCO 3 at room temperature for 3h. The reaction mixture was treated with Na 2
S
2
O
3 solution and stirred for 0.5 h at room temperature. EtOAc extraction, followed by washing with dilute NaOH solution and brine, and evaporation of the solvent produced 0.28 g (0.94 mmol, 94%) of 2 -amino-6-ethylsulfonyl-1H-benzoiinidazole-4-carboxylic acid ethyl ester. LCMS: 298 (M + 1)*. 25 0.30 g (1.5 mmol) of isoquinoline-3-carboxylic acid and 800 mg (2.1 mmol) HBTU were dissolved in 3.0 mL of DMF and 0.6 muL of DIEA and treated with 0.28 g (0.94 mmol) of 2-amino-6-ethylsulfonyl-1H-benzoimidazole-4-carboxylic acid ethyl ester as described in general procedure A to produce 6 -ethylsulfonyl-2-[(isoquinoline-3-carbonyl)-amino]-3H benzoimidazole-4-carboxylic acid ethyl ester. LCMS: 453 (M + 1)-. 30 0.36 g (0.79 mmol) of 6-ethylsulfonyl-2-[(isoquinoline-3-carbonyl)-amino]-3H benzoimidazole-4-carboxylic acid ethyl ester was hydrolyzed according to general procedure B to produce 0.30 g (0.71 mmol, 90%) of 6 -ethylsulfonyl-2-[(isoquinoline-3-carbonyl) amino]-3H-benzoimidazole-4-carboxylic acid. LCMS: 425 (M + 1)*. 225 WO 2006/099379 PCT/US2006/009049 62 mg (0.15 mmol) of 6-ethylsulfonyl-2- [(isoquinoline- 3 -carbonyl)-amino]- 3
H
benzoimidazole-4-carboxylic acid and 200 mg (0.53 mmol) of HBTU were dissolved in 1.0 mL of DMF and 0.5 mL of DIEA and treated with 132 mg (1.0 mnol) of 2-aminoimidazole sulfate as described in general procedure A. After the reaction was complete, diluted with 5 brine 10 (mL). The resulting solid was filtered, washed with water, EtOAc and dried under vacuum to produce 36 mg (57%) of isoquinoline-3-carboxylic acid [5-ethanesulfonyl-7-(IH imidazol-2-ylcarbainoyl)- 1H-benzoimidazol-2-yll-amide . LCMS: 490 (M + 1). By analogous methods to those used to prepare Example 491 and those in the relevant above schemes, the following compounds were synthesized. H 0 N'W N H \N N R, / N 10 Ex. W R LCMS: (M+1)+ 492 1H-imidazol-2-yl methyl 477 493 1H-imidazol-2-yl phenyl 539 494 1H-imidazol-2-yl butyl 519 Example 495 A solution of 1.53g (10 mmol) of 3-nitro-benzene-1,2-diamine in 20 mL of methanol 15 was treated with 1.3 g (12.5 mmol) of BrCN , and the mixture was heated under reflux for 1.0 h. After cooling to r.t., the reaction was concentrated and the residue were washed with sat. NaHCO 3 solution. The solid was collected, washed with water and dried to produce 1H benzoimidazole-4-nitro-2-amine. LCMS: 179 (M + 1)*. 1.6 g (8 mmol) of isoquinoline-3-carboxylic acid and HBTU were dissolved in DMF 20 and DIBA was treated with 1.4 g (8 mmol) of 1H-benzoimidazole-4-nitro-2-amine as described in general procedure A to produce 1.9 g (6 mmol, 75%) of isoquinoline-3 carboxylic acid (1H-benzoimidazol-4-nitro-2-yl)-amide. LCMS: 335 (M + 1)+ 226 WO 2006/099379 PCT/US2006/009049 1.6 g (6 nmol) of isoquinoline-3-carboxylic acid (1H-benzoimidazol-4-nitro-2-yl)-amide was hydrogenated according to general procedure C to produce 0.9 g (3 mmol, 50%) of isoquinoline-3-carboxylic acid (4-amino-1H-benzoimidazol-2-yl)-amide. LCMS: 305 (M + 1). 5 66 mg (0.2 minol) of isoquinoline-3-carboxylic acid (4-amino-iH-benzoimidazol-2 yl)-amide was dissolved in pyridine (0.6 mL). To this stirred solution at 0" C, 0.2 mmol of phenylsufonyl chloride in 0.2 mL of DCM was added in one portion. The reaction mixture was stirred at room temperature for 2 h. Brine (5 mL) was added and stirred for 10 min. The resulting solid was collected by filtration, washed with water (3 x 2 ml) and EtOAc (3 x 3 mL) 10 and dried to yield 46 mg (52%) of isoquinoline-3-carboxylic acid (7-benzenesulfonylamino 1H-benzoimidazol-2-yl)-amide. LCMS: 444 (M + 1). Example 496 0.10 g (0.30 mmol) of isoquinoline-3-carboxylic acid (4-amino-1H-benzoimidazol-2 yl)-amide in 2 mL of pyridine was treated with 0.06 mL (0.75 mmol) of methanesulfonyl 15 chloride and shaken at room temperature for -4 hours. After such time, this crude reaction mixture was treated with 0.10 mL hydrazine monohydrate and heated at 60"C for - 1 hour. The reaction mixture was diluted with water and the resulting solid isoquinoline-3-carboxylic acid (4-methanesulfonylamino-lH-benzoimidazol-2-yl)-amide was collected by filtration and required no further purification. LCMS: 382.9 (M+1)t. 20 Example 497 7.6 g (50 mmol) of 4-nitro-ortho-pbenylenediamnine was dissolved were dissolved in 100 mL of ethanol and 20 mL of H 2 0. 7.9 g (75 mmol) of BrCN was added in one portion to this stirring solution, and the resulting mixture was heated under reflux for 1.0 h. The reaction mixture was cooled to room temperature, concentrated to 20 mL then neutralized 25 with saturated NaHCO 3 solution to -pH 8, and resulting solid 5-nitro-1 H-b enzoimidazol-2 ylamine collected via filtration was used directly in the next step without further purification. Isoquinoline-3-carboxylic acid (5-nitro-IH-benzoimidazol-2-yl)-amide was synthesized from 3.5 g (20 mmol) of isoquinoline-3-carboxylic acid monohydrate, 7.6 g (20 mmol) of HBTU, and 3.56 g (20 mmol) of the above described 5-nitro-1H-benzoimidazol-2 30 ylamine as described in general procedure A. A 0.50g (1.5 mmol) portion of this isoquinoline-3-carboxylic acid (5-nitro- 1 H-benzoinidazol-2-yl)-anide was subsequently hydrogenated according to general procedure C to produce isoquinoline-3-carboxylic acid (5 amino-I1H-benzoimidazol-2-yl)-amide. LCMS: 304 (M+1)*. 227 WO 2006/099379 PCT/US20061009049 Example 498 0.10 g (0.30 mmol) of isoquinoline-3-carboxylic acid (5-amino-lH-benzoimidazol-2 yl)-amide in 2 mL of pyridine was treated with 0.06 mL (0.75 mmol) of methanesulfonyl chloride and shaken at room temperature for -4 hours. After such time, this crude reaction 5 mixture was treated with 0.10 mL hydrazine monohydrate and heated at 60 0 C for - 1 hour. The reaction mixture was diluted with water, and the resulting solid isoquinoline-3 carboxylic acid (5-methanesulfonylamino- 1 H-benzoimidazol-2-yl)-amide was collected by filtration and required no further purification. LCMS: 382 (M+-1)*. 'H NMR (DMSO-d 6 , 400 MHz): 5 9.49 (s, 1H), 9.45 (s, 1H), 8.78 (s, 1H), 8.30 (t, 2H), 7.94-7.87 (m, 2H), 7.46 10 7.44 (m, 2H), 7.03 (d, 1H), 2.89 (s, 3H) ppm. By analogous methods to those used to prepare Example 498 and those in the relevant above schemes, the following compounds were synthesized. ON H o1.o j O~ N' N R'N N H H O Ex. R LCMS (M+1)+ 499 4-biphenyl 521 500 propyl 411 501 isopropyl 411 15 Biological Assay The following assay methods were used to identify and evaluate compounds of Formula (I) that are effective in reducing the proteolytic activity of BACE. BACE Fluorescence Resonance Energy Transfer (FRET) Assay 20 In the following assay, the proteolytic activity of BACE is measured by observing cleavage of a fluorescent group from a peptide substrate containing a rhodamine fluorescent donor and a quenching acceptor. The inhibitory activity of compounds of Formula (I) is compared to a statine derived control inhibitor STA200 (KTEEISEVN(Statine)VAEF-OH, MP Biomedical Cat. # STA 228 WO 2006/099379 PCT/US2006/009049 200). The cleavage reaction occurs when a BACE-I substrate (Rhodamine-EVNLDAEFK Quencher, Invitrogen, Cat.# P2947) was added to a reaction mixture containing BACE-i and allowed to proceed for one hour. Fluorescence, used as a marker of BACE activity, is monitored using 540 rim excitation and 585 inm emission wavelengths (Envision, Perkin 5 Elmer). A typical assay reaction contains BACE- in assay buffer (50 mM sodium acetate, pH 4-4.5, 0.0125% CHAPS (3-[(3-Cholamidopropyl)dimethylammonio)-1-propanesulfonate), 0.0125% TritonX-100, 0.006% EDTA) which is pre-incubated for 30 minutes with test compound in 7.5% DMSO. The reaction is initiated with the addition of BACE- 1 substrate 10 in assay buffer and allowed to proceed for one hour at room temperature. Assays are conducted in black 384-well microtiter plates and scanned at room temperature using 540 nm excitation and 585 nrm emission wavelengths. A test compound's activity is reported as the ICSO. Compounds in Examples 1-501 inhibited the proteolytic activity of BACE in the FRET assay with an IC 50 of less than 30 pM. 15 AB Cell Based Assay Procedure In the following assay, the proteolytic activity of BACE in cells exposed to varying concentrations of a compound of interest is measured by observing the amount of A 1 o secreted from HEK293 cells (Human Embryonic Kidney epithelial cell line) stably expressing either wildtype human APP695 protein (HEK-APPwt cells). 20 HEK-APPwt cells were grown in high glucose DMEM (Dulbecco's Modified Eagles Medium) with 4500 mg glucose/L,L-glutamine, NaH-C0 3 , pyrdoxin HC1, 10 mM HEPES (4 (2-hydroxyethyl)-1-piperazineethanesulfonic acid) (pH 7.4), 0.1 mM NEAA (Non-essential Amino Acids) (GIBCO Cat#i 1140-050), 10% fetal bovine serum and 250 pg/ml hygromycin in T-225 flasks at 37 0 C with CO 2 and humidity control. 25 Test compounds were initially prepared in DMSO and diluted with DMEM media containing 2% PBS (Fetal bovine serum) and 0.05% Tween20. Ten standard compound solutions were prepared having a range of concentrations. The standard compound solutions were used to determine the EC 50 of the test compound. The range of concentrations chosen may depend on the compound's predicted potency. 30 To prepare the cells for the assay, a flask containing HEK-APPwt cells were trypsinized briefly (1 mL trypsin), and once the cells detached, 4 mL of 10% FBS-DMEM was added to the flask. The detached cells were centrifiged at 900 rpm for 5 min to form a pellet. 229 WO 2006/099379 PCT/US2006/009049 The HEK-APPwt cell pellet was re-suspended with 10 mL DMEM media containing 2% FBS. 80 gL of the cell suspension was added to each well of a 96-well cell culture plate to give 100,000 cells/well. 10 gL of a standard compound solution was added to each well of the 96-well cell culture plate followed by 10 pL of Alamar blue solution. The cells were 5 incubated at room temperature for 1 hi, followed by a five-hour incubation in the CO 2 incubator at 37* C. At the end of the incubation, the plates were removed from incubator, and the supernatant was collected. Ap13-4 concentration in the medium was measured by using a commercial AP 1
.
4 0 ELISA kit (IBL, Japan). Briefly, the ELISA plates were coated with anti 10 human Ap 35 -o mouse IgG monoclonal antibody. A horseradish peroxidase enzyme conjugated anti-human AP1 1-28 mouse IgG monoclone antibody was used for detection. The cell culture supernatant was diluted 1:4 fold with (EIA buffer + Protease inhibitor (buffer containing Protease Inhibitor (I mL PI/ 30 mL buffer). A 100 pL aliquot of the diluted supernatant was added to each well of the ELISA plate and incubated for 6 hrs at 4 0 C. The 15 ELISA plate was washed 8 times with phosphate buffered saline (PBS) containing 0.05% Tween 20. A 100 pL of detection antibody was then added and incubated for 1 hour at 4 0 C. The plate was washed 8 times with PBS buffer containing 0.05% Tween 20 followed by addition of 100 iL of the substrate with the chromogen tetramethyl benzidine (TMB). The plate was 20 incubated in the dark at room temperature for about 30 min and a stop solution (IN H2SO4) was added. The intensity of the color developed was measured at 450 nrm. The optical density at 450 nm (OD450) is proportional to the concentration of human Ap iA secreted by the cell. As a reference, N-[N-(3,5-difluorophenacetyl-L-alanyl)]-S-phenylglycine t-butyl ester 25 (DAPT, a y-secretase inhibitor) was used to indicate 100% inhibition of BACE activity. Thus, the assay measures the ability of a compound of interest to reduce ApIAo secretion. Compound potency was reported as the ECSO by calculating the percent inhibition at all concentration levels and the data were fit with non-linear curve fitting algorithm using in GraphPad Prism. 30 Various compounds of the present invention including the compounds of Examples 58, 140, 155, 165, 176, 193, 198, 212, 247, 289, 294, 295, 296,300, 311,312, 318, 343, 351, 355, 414, 416, 417, 428, 434, 439, 458, 472, and 491 exhibited an EC 50 value of less than or equal to 2.0 pM in the cell based assay described above. 230 While the invention has been described and illustrated with reference to certain embodiments thereof, those skilled in the art will appreciate that various changes, modifications and substitutions can be made therein without departing from the spirit and scope of the invention. For example, effective dosages other than the dosages as set forth 5 herein may be applicable as a consequence of variations in the responsiveness of the subject being treated. Likewise, the specific pharmacological responses observed may vary according to and depending on the particular active compound selected or whether there are present pharmaceutical carriers, as well as the type of formulation and mode of administration employed, and such expected variations or differences in the results are contemplated in accordance with the objects and practices of the present invention. Moreover, all compounds that are recited in the written description are contemplated as possibilities for any of the recited methods, processes, compositions, and/or compounds as appear in the written description and the appended claims. Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps. The reference to any prior art in this specification is not, and should not be taken as, an acknowledgement or any form of suggestion that the prior art forms part of the common general knowledge in Australia. 231

Claims (23)

1. A compound, which is a compound selected from the group consisting of: 1 -methyl-1I-indazole-3-carboxylic acid [7-( IH-imid azol-2-ylcarbamoyl)-l H benzoimidazol-2-yl]-amide; isoquinoline-3 -carboxylic acid [4-(4,5,6,7-tetrahydro-I H--benzoimidazol-2-ylcarbamoyl) 1H -benzoimidazol-2-yl]-amide; and
2-[(6-Cyclopropylethynyl-pyridine-2-carbonyl)-amino]-1 H-benzoimidazole-4-carboxylic acid (1 H-imidazol-2-yl)-amide; or a pharmaceutically acceptable salt thereof 2. The compound of claim 1, wherein the compound is 1-methyl-1I H-indazole-3-carboxylic acid [7-(lH-imid azol-2-ylcarbamoyl)-1 H-benzoimidazol-2-yl]-amide, or a pharmaceutically acceptable salt thereof
3. The compound of claim 1, wherein the compound is isoquinoline-3-carboxylic acid [4 (4,5,6,7-tetrahydro-1H-benzoimidazol-2-ylcarbamoyl)- 1 H-benzoimidazol-2-yl]-amide, or a pharmaceutically acceptable salt thereof
4. The compound of claim 1, wherein the compound is 2-[(6-cyclopropylethynyl-pyridine-2 carbonyl)-amino]-1 H-benzoimidazole4-carboxylic acid (1 H-imidazol-2-yl)-amide, or a pharmaceutically acceptable salt thereof
5. A pharmaceutical composition comprising a compound of any one of claims I to 4 and a pharmaceutically acceptable carrier.
6. The pharmaceutical composition of claim 5, where the compound is 2-[(6-cyclopropyl ethynyl-pyridine-2-carbonyl)-amino]-1H-benzoimidazole-4-carboxylic acid ( H-imidazol-2-yl) amide or a pharmaceutically acceptable salt thereof. 232
7. The pharmaceutical composition of claim 5 further comprising one or more therapeutic agents selected from the group consisting of: an anti-Alzheimer's agent, another beta-secretase inhibitor, a gamma-secretase inhibitor, a HMG-CoA reductase inhibitor, ibuprofen, naproxen, diclofenac, memantine, galantamine, rivastigmine, donepezil, tacrine, vitamin E, a CB-i receptor antagonist, a CB-1 receptor inverse agonist, doxycycline, rifampin, an agent that binds A P or that induces antibodies that bind AP, an anti-Ap antibody, an AP vaccine, and a RAGE/RAGE ligand interaction antagonist.
8. Use of a compound of any one of claim I to 4 in the manufacture of a medicament for treating dementia of the Alzheimer's type.
9. Use of a compound of any one of claims 1 to 4 in the manufacture of a medicament for treating mild cognitive impairment.
10. Use of a compound of any one of claims 1 to 4 for treating or preventing a BACE mediated disease.
11. Use of a compound of any one of claims I to 4 in the manufacture of a medicament for treating Alzheimer's disease, mild cognitive impairment, Down's syndrome, Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch-Type, cerebral amyloid angiopathy, degenerative dementia, diffuse Lewy body type of Alzheimer's disease or central or peripheral amyloid diseases.
12. The use of claim 8, wherein dementia of the Alzheimer's type and is selected from the group consisting of dementia of the Alzheimer's type with early onset uncomplicated, dementia of the Alzheimer's type with early onset with delusions, dementia of the Alzheimer's type with early onset with depressed mood, dementia of the Alzheimer's type with late onset uncomplicated, dementia of the Alzheimer's type with late onset with delusions and dementia of the Alzheimer's type with late onset with depressed mood.
13. Use of a compound of any one of claims I to 4 in the manufacture of a medicament for treating one or more conditions associated with amyloid plaque accumulation. 233
14. The use of claim 13, wherein the treating comprises reducing the rate ofneurofibrillary tangle formation in a subject.
15. The use of claim 13, wherein the treating comprises reducing the rate of amyloid plaque accumulation in a subject.
16. A method of treating dementia of the Alzheimer's type comprising administering to a subject in need thereof a compound of any one of claims 1 to 4.
17. A method of treating mild cognitive impairment comprising administering to a subject in need thereof a compound of any one of claims 1 to 4.
18. A method of treating or preventing a BACE mediated disease comprising administering to a subject in need thereof a compound of any one of claims I to 4.
19. A method of treating Alzheimer's disease, mild cognitive impairment, Down's syndrome, Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch-Type, cerebral amyloid angiopathy, degenerative dementia, diffuse Lewy body type of Alzheimer's disease, or central or peripheral amyloid diseases, comprising administering to a subject in need thereof a compound of any one of claims I to 4.
20. The method of claim 16, wherein dementia of the Alzheimer's type is dementia of the Alzheimer's type with early onset uncomplicated, dementia of the Alzheimer's type with early onset with delusions, dementia of the Alzheimer's type with early onset with depressed mood, dementia of the Alzheimer's type with late onset uncomplicated, dementia of the Alzheimer's type with late onset with delusions, or dementia of the Alzheimer's type with late onset with depressed mood.
21. A method of treating one or more conditions associated with amyloid plaque accumulation comprising administering to a subject in need thereof a compound of any one of claims 1 to 4. 234
22. The method of claim 21, wherein the treating comprises reducing the rate ofneurofibrillary tangle formation in a subject.
23. The method of claim 21, wherein the treating comprises reducing the rate of amyloid plaque accumulation in a subject. 235
AU2011244844A 2005-03-14 2011-10-28 Benzazole derivatives, compositions, and methods of use as B-secretase inhibitors Abandoned AU2011244844A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2011244844A AU2011244844A1 (en) 2005-03-14 2011-10-28 Benzazole derivatives, compositions, and methods of use as B-secretase inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US60/661,349 2005-03-14
AU2011244844A AU2011244844A1 (en) 2005-03-14 2011-10-28 Benzazole derivatives, compositions, and methods of use as B-secretase inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2006223070A Division AU2006223070B2 (en) 2005-03-14 2006-03-14 Benzazole derivatives, compositions, and methods of use as B-secretase inhibitors

Publications (1)

Publication Number Publication Date
AU2011244844A1 true AU2011244844A1 (en) 2011-11-24

Family

ID=45442334

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2011244844A Abandoned AU2011244844A1 (en) 2005-03-14 2011-10-28 Benzazole derivatives, compositions, and methods of use as B-secretase inhibitors

Country Status (1)

Country Link
AU (1) AU2011244844A1 (en)

Similar Documents

Publication Publication Date Title
AU2006223070B2 (en) Benzazole derivatives, compositions, and methods of use as B-secretase inhibitors
JP4004737B2 (en) Heterocycle-substituted benzimidazoles, methods for their preparation and uses thereof
JP4887139B2 (en) Dipeptidyl peptidase inhibitor
AU760020B2 (en) Novel angiogenesis inhibitors
CA2758958A1 (en) Substituted imidazo[1,2-a]pyridine derivatives, pharmaceutical compositions, and methods of use as .beta.-secretase inhibitors
EA019027B1 (en) Fluorene derivatives, compositions containing said derivatives and the use thereof
EP2590950A1 (en) N-cyclyl-3 - (cyclylcarbonylaminomethyl) benzamide derivatives as rho kinase inhibitors
EP1017682A1 (en) Novel angiogenesis inhibitors
JP2009526072A (en) Benzazole derivatives, compositions, and methods of use as Aurora kinase inhibitors
AU2013339167A1 (en) Novel amine derivative or salt thereof
AU2019319835A1 (en) Indole and azaindole inhibitors of PAD enzymes
WO2000034277A1 (en) Sulfonamide compounds and uses thereof as medicines
AU2011244844A1 (en) Benzazole derivatives, compositions, and methods of use as B-secretase inhibitors
CN113754635A (en) Fused ring compound and preparation method and application thereof
CA2759162A1 (en) Benzazole derivatives, compositions, and methods of use as .beta.-secretase inhibitors
MX2008009811A (en) Benzazole derivatives, compositions, and methods of use as aurora kinase inhibitors

Legal Events

Date Code Title Description
MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application