Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ALPHA-HYDROXY AMIDES AS BRADYKININ ANTAGONISTS OR INVERSE AGONISTS
Document Type and Number:
WIPO Patent Application WO/2005/063690
Kind Code:
A1
Abstract:
&agr -Hydroxy amide derivatives of the general formula (I) are bradykinin B1 antagonists or inverse agonists useful in the treatment or prevention of symptoms such as pain and inflammation associated with the bradykinin B1 pathway. R2a is selected from (1) a group selected from Ra. (2) (CH2)nNRbC(O)Ra. (3)(CH2)nNRbSO2Rd.(4)(CH2)nNRbCO2Ra.(5)(CH2)k-heterocycle optionally substituted with 1 to 3 groups independently selected from halogen.nitro, cyano.ORa.SRa.C1-4 alkyl and C1-3 haloakyl wherein said heterocycle is (a) a 5-membered heteroaromatic ring having a ring heteroatom selected from N.O and S. and optionally having up to 3 additional ring nitrogen atoms wherein said ring is optionally benzo-fused; or(b) a 6-membered heteromatic ring containing from 1 to 3 ring nitrogen atoms and N-oxydes thereof. Wherein said ring is optionally benzo-fused. (6)(CH2)kCO2Ra.and (7)(CH2)C(O)NRbRc. R2b is OH or a group selected from R2a; or R2a and R2b together with the carbon atom to which they are attached form a 3-to 7-membered carbocyclic ring optionally substituted with 1 to 4 groups independently selected from halogen. ORa. C1-4 alkyl and C1-4 haloalkyl;

Inventors:
WOOD MICHAEL R (US)
ANTHONY NEVILLE J (US)
BOCK MARK G (US)
KUDUK SCOTT D (US)
Application Number:
PCT/US2004/042691
Publication Date:
July 14, 2005
Filing Date:
December 17, 2004
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MERCK & CO INC (US)
WOOD MICHAEL R (US)
ANTHONY NEVILLE J (US)
BOCK MARK G (US)
KUDUK SCOTT D (US)
International Classes:
C07C235/06; C07C235/08; C07C235/12; C07C235/26; C07C235/34; C07C235/40; C07D209/12; C07D213/56; C07D213/61; C07D239/26; C07D257/04; C07D271/06; C07D275/06; C07D401/10; C07D413/10; (IPC1-7): C07C235/40; C07D271/06; C07C235/12; C07C235/34; C07C235/26; C07C235/06; C07C23/08; C07D213/61; A61K31/235; A61K31/4245; A61K31/165; A61K31/44; A61P29/00
Domestic Patent References:
WO2003066577A12003-08-14
WO2002076964A12002-10-03
Attorney, Agent or Firm:
MERCK & CO., INC. (Rahway, New Jersey, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS :
1. A compound of formula (I) and pharmaceutically acceptable salts thereof wherein Y is CH or N ; RI is R2a is selected from (1) a group selected from Ra, (2) (CH2) nNRbC (O) Ra, (3) (CH2) nNRbS02Rd, (4) (CH2) nNRbCO2Ra, (5) (CH2) kheterocycle optionally substituted with 1 to 3 groups independently selected from halogen, nitro, cyano, ORa, SRa, C1 4 alkyl and C1 3 haloalkyl wherein said heterocycle is (a) a 5membered heteroaromatic ring having a ring heteroatom selected from N, O and S, and optionally having up to 3 additional ring nitrogen atoms wherein said ring is optionally benzofused; or (b) a 6 membered heteroaromatic ring containing from 1 to 3 ring nitrogen atoms and Noxides thereof, wherein said ring is optionally benzofused, (6) (CH2) kCO2Ra, and (7) (CH2) kC (O) NRbRc, R2b is OH or a group selected from R2a; or R2a and R2b together with the carbon atom to which they are attached form a 3to 7membered carbocyclic ring optionally substituted with 1 to 4 groups independently selected from halogen, ORa, C14 alkyl and C1 4 haloalkyl ; R3a and R3b are independently selected from hydrogen, C1 4 alkyl, and C1 4 haloalkyl ; R4a and R4b are independently selected from hydrogen and halogen; R6 is selected from (1) Cl8 alkyl optionally substituted with 15 groups independently selected from halogen, nitro, cyano, CORa, C02Ra, C (O) NRbRc, ORa, OC (O) Ra, SRa, S02Rd, S (O) Rd, NRbRc, NRbC (O) Ra, NRbS02Rd, and NRbC02Ra, (2) C38 cycloalkyl, (3) C28 alkenyl optionally substituted with C02Ra, (4) halogen, (5) cyano, (6) nitro, (7) NRbRc, (8) NRbC (O) Ra, (9) NRbCO2Ra, (10) NRbC (O) NRbRc, (11) NRbC (O) NRbC02Ra, (12) NRbS02Rd, (13) C02Ra, (14) CORa, (15) C (O) NRbRc, (16) C (O) NHORa, (17) C (=NORa) Ra, (18) C (=NORa) NRbRc, (19) ORa, (20) OC (O) Ra, (21) S (O) vRd, (22) S02NRbRc, (23) optionally substituted heterocycle where the heterocycle is (a) a 5 membered heteroaromatic ring having a ring heteroatom selected from N, O and S, and optionally having up to 3 additional ring nitrogen atoms, (b) a 6membered heteroaromatic ring having 1 to 3 ring N atoms, (c) 4,5dihydrooxazolyl or (d) 4, 5dihydro1, 2,4oxadiazolyl, and wherein said substituent is 1 to 3 groups independently selected from C14 alkyl optionally substituted with 1 to 5 halogen atoms, ORa or OC (O) Ra, (24) phenyl optionally substituted with 1 to 3 groups independently selected from halogen, nitro, cyano, ORa, SRa, C14 alkyl and C14 haloalkyl, and (25) OS02Rd ; R7 is selected from hydrogen and halogen; R8 and R9 are independently selected from hydrogen and a group from R6 ; with the proviso that not more than one of R6, R8, and R9 is a heterocycle; Ra is selected from (1) hydrogen, (2) C17 alkyl optionally substituted with 1 to 5 halogen atoms, OH, SH, OC14alkyl, or SCl 4alkyl, (3) (CH2) kphenyl optionally substituted with 1 to 3 groups independently selected from halogen, cyano, nitro, OH, Cl4 alkyloxy, C36 cycloalkyl, C14 alkyl and Cl 4haloalkyl, and (4) C36 cycloalkyl ; Rb and Rc are independently selected from (1) hydrogen, (2) C14 alkyl optionally substituted with 1 to 5 groups independently selected from halogen, amino, CO2Ra, ORa, monoCl4alkylamino, and di Cl 4alkylamino, (3) (CH2) kphenyl optionally substituted with 1 to 3 groups selected from halogen, cyano, nitro, ORa, C02Ra, C36 cycloalkyl, C14 alkyl and Cl 4haloalkyl, and (4) C36 cycloalkyl, or Rb and Rc together with the nitrogen atom to which they are attached form a 4, 5, or 6membered ring optionally containing an additional heteroatom selected from NRe, O, S, S (O) and S (O) 2; Rd is selected from (1) C1 4 alkyl, (2) Cl 4haloalkyl, (3) C14 alkyloxy, (4) (CH2) kphenyl optionally substituted with 1 to 3 groups selected from halogen, cyano, nitro, ORa, C02Ra, C36 cycloalkyl, Cl4 alkyl and Cl 4haloalkyl, (5) pyridyl, and (6) pyridyl Noxide ; Re is selected from hydrogen, C14 alkyl, C14 haloalkyl, C (O) H and C (O) Cl 4alkyl ; nis 1, 2, or 3 ; k is 0, 1,2, 3, or 4 ; and v is 0, 1, or 2.
2. A compound of Claim 1 wherein R2a, R2b and the carbon atom to which they are attached form a 3to 7membered carbocyclic ring optionally substituted with 1 to 4 groups independently selected from halogen, ORa, C1 4 alkyl and Cl4 haloalkyl.
3. A compound of Claim 1 wherein Rl is wherein R6, R8 and R9 are as defined in Claim 1.
4. A comopound of Claim 3 wherein R6 is selected from (1)C02Cl4alkyl, (2) C 1 4alkoxy, and (3) a 5membered heteroaromatic ring having a ring heteroatom selected from N, O and S, and optionally having up to 3 additional ring nitrogen atoms, said ring being optionally substituted with a Cl 4alkyl group.
5. A compound of Claim 4 wherein R8 is hydrogen or 3halo, and R9 is hydrogen or 5 halo.
6. A compound of Claim 1 wherein RI is wherein R4a, R4b and Ra are as defined in Claim 1.
7. A compound of Claim 6 wherein R4a and R4b are each fluoro.
8. A compound of Claim 1 having the formula (Ia) and pharmaceutically acceptable salts thereof : (Ia) wherein m is 1 to 5; Y is N or CH; one of R3a and R3b is hydrogen and the other is hydrogen or methyl; R7 is hydrogen or fluorine; R6 is selected from (1)C02Cl4alkyl, (2) Cl 4alkoxy optionally substituted with 1 to 5 halogen atoms, and (3) a 5membered heteroaromatic ring having a ring heteroatom selected from N, O and S, and optionally having up to 3 additional ring nitrogen atoms, said ring being optionally substituted with a Cl 4alkyl group; and R8 and R9 are independently hydrogen or halogen.
9. A compound of Claim 1 having the formula Ib and pharmaceutically acceptable salts thereof : Ib where R3a, R3b, R6, R7, R8 and R9 are as defined in Claim 1, and R2a'and R2b'are independently selected from (1) hydrogen, (2) C17 alkyl optionally substituted with 1 to 5 halogen atoms, SH, OH, S Cl 4alkyl or OCl4alkyl, (3) (CH2) kphenyl optionally substituted with 1 to 3 groups independently selected from halogen, cyano, nitro, OH, Cl4 alkyloxy, C36 cycloalkyl, C1 4 alkyl and Cl 4haloalkyl, (4) C36 cycloalkyl, (5) (CH2) kpyridyl, and (6) (CH2) kindolyl.
10. A compound of Claim 9 wherein R2a'and R2b'are independently Cl 7alkyl optionally substituted with 1 to 5 halogen atoms.
11. A compound of Claim 10 wherein one of R3a and R3b is hydrogen and the other is hydrogen or methyl ; R7 is hydrogen, chlorine or fluorine; R6 is selected from (1)CO2Cl 4alkyl, (2) Cl 4alkoxy optionally substituted with 1 to 5 halogen atoms, and (3) a 5membered heteroaromatic ring having a ring heteroatom selected from N, O and S, and optionally having up to 3 additional ring nitrogen atoms, said ring being optionally substituted with a Cl 4alkyl group; and R8 and R9 are indepnendently hydrogen or halogen.
12. A compound of Claiml having the formula Ic and pharmaceutically acceptable salts thereof : Ic wherein Y is N or CH; R7 is H, chlorine or fluorine; R3a is H or methyl; R6 is selected from (1)CO2 Cl4alkyl, (2) Cl 4alkoxy, (3) Cl 4haloalkyloxy, and (4) a 5membered heteroaromatic ring having a ring heteroatom selected from N, O and S, and optionally having up to 3 additional ring nitrogen atoms, said ring being optionally substituted with a Cl 4alkyl group; and R8 and R9 are independently hydrogen or halogen.
13. A pharmaceutical composition comprising a therapeutically effective amount of a compound of Claim 1 and a pharmaceutically acceptable carrier.
14. A method for the treatment or prevention of a condition mediated by bradykinin B 1 receptor in a mammal which comprises administering to said mammal a therapeutically effective amount of a compound of Claim 1.
15. A method for the treatment or prevention of pain in a mammal which comprises administering to said mammal a therapeutically effective amount of a compound of Claim 1.
16. A method for the treatment or prevention of pain selected from acute pain, inflammatory pain and neuropathic pain in a mammal which comprises administering to said mammal a therapeutically effective amount of a compound of Claim 1.
17. Use of a compound of Claim 1 or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment or prevention of diseases or conditions mediated by bradykinin B 1 receptor.
18. Use of Claim 17 wherein said diseases or conditions are acute pain, inflammatory pain and neuropathic pain.
Description:
TITLE OF THE INVENTION ALPHA-HYDROXY AMIDES AS BRADYKININ ANTAGONISTS OR INVERSE AGONISTS BACKGROUND OF THE INVENTION This invention is directed to a-hydroxyamide compounds. In particular, this invention is directed to a-hydroxy amide compounds that are bradykinin antagonists or inverse agonists.

Bradykinin ("BK") is a kinin which plays an important role in the pathophysiological processes accompanying acute and chronic pain and inflammation. Bradykinin (BK), like other kinins, is an autacoid peptide produced by the catalytic action of kallikrein enzymes on plasma and tissue precursors termed kininogens. The biological actions of BK are mediated by at least two major G- protein-coupled BK receptors termed B 1 and B2. It is generally believed that B2 receptors, but not B 1 receptors, are expressed in normal tissues and that inflammation, tissue damage or bacterial infection can rapidly induce B 1 receptor expression. This makes the B 1 receptor a particularly attractive drug target.

The putative role of kinins, and specifically BK, in the management of pain and inflammation has provided the impetus for developing potent and selective BK antagonists. In recent years, this effort has been heightened with the expectation that useful therapeutic agents with analgesic and anti-inflammatory properties would provide relief from maladies mediated through a BK receptor pathway (see e. g. , M. G.

Bock and J. Longmore, Current Opinion in Chem. Biol., 4: 401-406 (2000) ). Accordingly, there is a need for novel compounds that are effective in blocking or reversing activation of bradykinin receptors. Such compounds would be useful in the management of pain and inflammation, as well as in the treatment or prevention of diseases and disorders mediated by bradykinin; further, such compounds are also useful as research tools (in vivo and in vitro).

SUMMARY OF THE INVENTION The present invention provides a-hydroxy amide derivatives which are bradykinin antagonists or inverse agonists, pharmaceutical compositions containing such compounds, and methods of using them as therapeutic agents.

DETAILED DESCRIPTION OF THE INVENTION The present invention provides compounds of formula (I) and pharmaceutically acceptable salts thereof

wherein Y is CH or N; Rl is R2a is selected from (1) a group selected from Ra, (2) (CH2) nNRbC (O) Ra, (3) (CH2) nNRbSO2Rd, (4) (CH2) nNRbC02Ra, (5) (CH2) k-heterocycle optionally substituted with 1 to 3 groups independently selected from halogen, nitro, cyano, ORa, SRa, C1-4 alkyl and C1 3 haloalkyl wherein said heterocycle is (a) a 5-membered heteroaromatic ring having a ring heteroatom selected from N, O and S, and optionally having up to 3 additional ring nitrogen atoms wherein said ring is optionally benzo-fused; or (b) a 6- membered heteroaromatic ring containing from 1 to 3 ring nitrogen atoms and N-oxides thereof, wherein said ring is optionally benzo-fused, (6) (CH2) kCO2Ra, and (7) (CH2) kC (O) NRbRc, R2b is OH or a group selected from R2a ; or R2a and R2b together with the carbon atom to which they are attached form a 3-to 7-membered carbocyclic ring optionally substituted with 1 to 4 groups independently selected from halogen, ORa, C1-4 alkyl and C1 4 haloalkyl ; R3a and R3b are independently selected from hydrogen, C1-4 alkyl, and C1 4 haloalkyl ; R4a and R4b are independently selected from hydrogen and halogen; R6 is selected from (1) C1-8 alkyl optionally substituted with 1-5 groups independently selected from halogen, nitro, cyano, CORa, C02Ra, C (O) NRbRc, ORa, OC (O) Ra, SRa, S02Rd, S (O) Rd, NRbRc, NRbC (O) Ra, NRbS02Rd, and NRbC02Ra, (2) C3-8 cycloalkyl, (3) C2-8 alkenyl optionally substituted with CO2Ra, (4) halogen, (5) cyano, (6) nitro, (7) NRbRc, (8) NRbC (O) Ra, (9) NRbC02Ra, (10) NRbC (O) NRbRc, (11) NRbC (O) NRbCO2Ra, (12) NRbS02Rd, (13) C02Ra, (14) CORa, (15) C (O) NRbRc, (16) C (O) NHORa, (17) C (=NORa) Ra, (18) C (=NORa) NRbRc, (19) ORa, (20) OC (O) Ra, (21) S (O) vRd, (22) S02NRbRc, (23) optionally substituted heterocycle where the heterocycle is (a) a 5-

membered heteroaromatic ring having a ring heteroatom selected from N, O and S, and optionally having up to 3 additional ring nitrogen atoms, (b) a 6-membered heteroaromatic ring having 1 to 3 ring N atoms, (c) 4,5-dihydro-oxazolyl or (d) 4, 5-dihydro-1, 2,4-oxadiazolyl, and wherein said substituent is 1 to 3 groups independently selected from Cl-4 alkyl optionally substituted with 1 to 5 halogen atoms, ORa or OC (O) Ra, (24) phenyl optionally substituted with 1 to 3 groups independently selected from halogen, nitro, cyano, ORa, SRa, C1 4 alkyl and C1 4 haloalkyl, and (25) OS02Rd ; R7 is selected from hydrogen and halogen; R8 and R9 are independently selected from hydrogen and a group from R6 ; with the proviso that not more than one of R6, R8, and R9 is a heterocycle; Ra is selected from (1) hydrogen, (2) C1 7 alkyl optionally substituted with 1 to 5 halogen atoms, OH, SH, O-Cl 4alkyl, or S-Cl 4alkyl, (3) (CH2) k-phenyl optionally substituted with 1 to 3 groups independently selected from halogen, cyano, nitro, OH, C1-4 alkyloxy, C3-6 cycloalkyl, Cl_4 alkyl and Cl 4haloalkyl, and (4) C3-6 cycloalkyl ; Rb and Rc are independently selected from (1) hydrogen, (2) C1 4 alkyl optionally substituted with 1 to 5 groups independently selected from halogen, amino, CO2Ra, ORa, mono-Cl 4alkylamino, and di- Cl-4alkylamino, (3) (CH2) k-phenyl optionally substituted with 1 to 3 groups selected from halogen, cyano, nitro, ORa, CO2RA, C3-6 cycloalkyl, C1-4 alkyl and Cl 4haloalkyl, and (4) C3-6 cycloalkyl, or Rb and RC together with the nitrogen atom to which they are attached form a 4-, 5-, or 6-membered ring optionally containing an additional heteroatom selected from NRe, O, S, S (O) and S (O) 2; Rd is selected from (1) C1-4 alkyl, (2) Cl 4haloalkyl, (3) Ci-4 alkyloxy, (4) (CH2) k-phenyl optionally substituted with 1 to 3 groups selected from halogen, cyano, nitro, ORa, CO2Ra, C3-6 cycloalkyl, C1-4 alkyl and Cl 4haloalkyl, (5) pyridyl, and (6) pyridyl N-oxide ; Re is selected from hydrogen, C1 4 alkyl, C1 4 haloalkyl, C (O) H and C (O) Cl 4alkyl ; n is 1,2, or 3 ; k is 0, 1,2, 3, or 4 ; and v is 0, 1, or 2.

Examples of R2a and R2b include, without limitation, hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, t-butyl, n-pentyl, neo-pentyl, n-hexyl, n-heptyl, trifluoromethyl, 2,2, 2-trifluoroethyl, 1, 1-difluoroethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, phenyl, benzyl, o-, m-or p-tolyl, xylyl, (trifluoromethyl) phenyl, cyanophenyl, bromophenyl, chloro- phenyl, dichlorophenyl, fluorophenyl, difluorophenyl, hydroxyphenyl, dihydroxyphenyl, methoxyphenyl, dimethoxyphenyl, (hydroxy) (methoxy) phenyl, 3-indolylmethyl, pylwolyl, thiazolyl, oxazolyl, imidazolyl, 2-benzothienyl, isoxazolyl, isothiazolyl, quinolinyl, 2- (acethylamino) ethyl, (methylsulfonylamino)- methyl, carboxy, carbamoylmethyl, and 2- (methoxycarbonylamino) ethyl. R2a, R2b and the carbon to

which they are attached together may form cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl, each of which may be optionally substituted with 1 to 4 groups selected from, for example, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, t-butyl, chlorine, fluorine, bromine, chloromethyl, trifluoromethyl, 1-chloroethyl, hydroxy, methoxy, and ethoxy.

Examples of R3a and R3b include, without limitation, hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, t-butyl, chloromethyl, fluoromethyl, difluoromethyl, trifluoro- methyl, 2,2, 2-trifluoroethyl, 2,2-difluoroethyl, 1, 1,2, 2,2-pentafluoroethyl, and the like.

Examples of R6 include methyl, ethyl, 1-methylethyl, 1-hydroxyethyl, carboxy, methoxy- carbonyl, ethoxycarbonyl, 2-fluoroethoxycarbonyl, isopropoxycarbonyl, phenoxycarbonyl, cyclopentoxy- carbonyl, cyclobutoxycarbonyl, cyclopropoxycarbonyl, 2,2, 2-trifluoroethoxycarbonyl, 4-trifluoromethyl- phenoxycarbonyl, methoxyaminocarbonyl, methoxycarbonylmethyl, formyl, hydroxy, 5-methyl-1, 2,4- oxadiazol-3-yl, 3-methyl-1, 2,4-oxadiazol-5-yl, 1-methyl-5-tetrazolyl, 2-methyl-5-tetrazolyl, cyano, methoxy, ethoxy, difluoromethoxy, trifluoromethoxy, 2,2-difluoroethoxy, 1, 3-difluoropropoxy, 2,2, 2- trifluoroethoxy, trifluoromethyl, chloro, fluoro, methylaminosulfonyl, dimethylaminosulfonyl, methoxy- carbonylamino, ethoxycarbonylamino, 2-fluoroethoxycarbonylamino, methylaminocarbonylamino, dimethylamino, methylaminocarbonyl, isopropylaminocarbonyl, ethylaminocarbonyl, cyclopropylamino- carbonyl, cyclobutylaminocarbonyl, dimethylaminocarbonyl, and aminocarbonyl. Examples for R8 include hydrogen, chloro, fluoro and methoxycarbonyl; examples of R9 include hydrogen, chloro, fluoro and methyl.

In one subset of formula I are compounds wherein R2a, R2b and the carbon atom to which they are attached form a 3-to 7-membered carbocyclic ring, said ring being optionally substituted with 1 to 4 groups independently selected from halogen, ORa, C1 4 alkyl and Cl-4 haloalkyl. In one embodiment the carbocyclic ring is substituted by methyl or halogen.

In a second subset of formula I are compounds wherein one of R3a and R3b is hydrogen and the other is hydrogen or methyl.

In a third subset of formula I are compounds wherein Rl is In one embodiment within this subset are compounds wherein R6 is selected from (1)-CO2- C1 4alkyl, (2) Cl 4alkoxy, and (3) a 5-membered heteroaromatic ring having a ring heteroatom selected

from N, O and S, and optionally having up to 3 additional ring nitrogen atoms, said ring being optionally substituted with a Cl 4alkyl group. In one embodiment R6 is selected from methoxycarbonyl, ethoxy, 1,2, 4-oxadiazolyl optionally substituted with a methyl group, and tetrazolyl optionally substituted with a methyl group. In another embodiment within this subset are compounds wherein R8 is hydrogen or 3- halogen. In yet another embodiment within this subset are compounds wherein R8 is hydrogen or 3- halogen, and R9 is hydrogen or 5-halogen.

In a fourth subset of formula I are compounds of formula (Ia) and pharmaceutically acceptable salts thereof:

wherein m is 1 to 5; Y is N or CH; one of R3a and R3b is hydrogen and the other is hydrogen or methyl; R7 is hydrogen or fluorine; R6 is selected from (1)-C02-Cl-4alkyl, (2) Cl 4alkoxy optionally substituted with 1 to 5 halogen atoms, and (3) a 5-membered heteroaromatic ring having a ring heteroatom selected from N, O and S, and optionally having up to 3 additional ring nitrogen atoms, said ring being optionally substituted with a Cl 4alkyl group; and R8 and R9 are independently hydrogen or halogen.

In a fifth subset of compounds of formula I are compounds wherein RI is

In one embodiment within this subset Ra is H. In another embodiment, R4a and R4b are each a halogen exemplified by fluorine.

In a sixth subset of compounds of formula I are compounds of formula Ib and pharmaceutically acceptable salts thereof :

where R3a, R3b, R6, R7, R8 and R9 are as defined under formula I. R2a'and R2b'are independently selected from (1) hydrogen, (2) C1 7 alkyl optionally substituted with 1 to 5 halogen atoms, SH, OH, S- C1-4alkyl or OCl 4alkyl, (3) (CH2) k-phenyl optionally substituted with 1 to 3 groups independently selected from halogen, cyano, nitro, OH, C1-4 alkyloxy, C3-6 cycloalkyl, C1-4 alkyl and Cl 4haloalkyl, (4) C3-6 cycloalkyl, (5) (CH2) k-pyridyl, and (6) (CH2) k-indolyl. In one embodiment within this subset are compounds wherein R2a'and R2b'are independently Cl 7alkyl optionally substituted with 1 to 5 halogen atoms. In another embodiment are compounds wherein one of R3a and R3b is hydrogen and the other is hydrogen or methyl; R7 is hydrogen, chlorine or fluorine; R6 is selected from (1) -CO2- C 1 4alkyl, (2) C 1 4alkoxy optionally substituted with 1 to 5 halogen atoms, and (3) a 5-membered heteroaromatic ring having a ring heteroatom selected from N, O and S, and optionally having up to 3 additional ring nitrogen atoms, said ring being optionally substituted with a Cl 4alkyl group; and R8 and R9 are indepnendently hydrogen or halogen.

In a seventh subset of compounds of formula I are compounds of formula Ic and pharmaceutically acceptable salts thereof: wherein Y is N or CH; R7 is H, chlorine or fluroine; R3a is H or methyl; R6 is selected from (1)-C02- Cl 4alkyl, (2) C1-4alkoxy, (3) Cl 4haloalkyloxy, and (4) a 5-membered heteroaromatic ring having a ring heteroatom selected from N, O and S, and optionally having up to 3 additional ring nitrogen atoms, said ring being optionally substituted with a Cl 4alkyl group; and R8 and R9 are independently hydrogen or halogen. In one embodiment, R8 is fluorine or chlorine. In another embodiment R8 and R9 are each

independently fluorine or chlorine. In yet another embodiment, R6 is optionally methyl substituted tetrazolyl or optionally methyl substituted 1,2, 4-oxadiazolyl. In another embodiment R7 is chlorine or fluroine, R6 is optionally methyl substituted tetrazolyl or optionally methyl substituted 1,2, 4-oxadiazolyl, R8 is fluorine or chlorine, and R9 is H, chlorine or fluorine.

A second aspect of the present invention provides a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula (I) or a phannaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

A third aspect of the present invention provides the use of a compound of formula I or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of. conditions mediated by bradykinin B 1 receptor. In one subset said condition is pain including acute, inflammatory and neuropathic pain.

A fourth aspect of the present invention provides a method for the treatment of a condition mediated by bradykinin B 1 receptor in a mammal comprising administering to said mammal a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.

Unless otherwise stated, the following terms have the meanings indicated below: "Alkenyl"means carbon chains which may be linear or branched or combination thereof and containing at least one C=C bond. Examples of alkenyl include allyl, 2-butenyl, 3-butenyl, 1-methyl-2- propenyl, and the like.

"Alkyl"as well as other groups having the prefix"alk"such as, for example, alkoxy, alkanoyl, and the like, means carbon chains which may be linear or branched or combinations thereof.

Examples of alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec-and tert-butyl, pentyl, hexyl, heptyl and the like.

"Cycloalkyl"means carbocycles containing no heteroatoms, and includes mono-, bi-and tricyclic saturated carbocycles, as well as fused ring systems. Such fused ring systems can include one ring that is partially or fully unsaturated such as a benzene ring to form fused ring systems such as benzofused carbocycles. Cycloalkyl includes such fused ring systems as spirofused ring systems.

Examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, decahydronaphthalene, adamantane, indanyl, indenyl, fluorenyl, 1,2, 3,4-tetrahydronaphthalene and the like.

"Haloalkyl"means an alkyl radical as defined above wherein at least one and up to all of the hydrogen atoms are replaced with a halogen. Examples of such haloalkyl radicals include chloromethyl, 1-bromoethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 2,2, 2-trifluoroethyl and the like.

"Halogen"means fluorine, chlorine, bromine and iodine.

"5-Membered heteroaromatic ring"and"benzo-fused 5-membered heteroaromatic ring" include, without limitation, pyrrole, imidazole, pyrazole, 1,2, 3-triazole, 1,2, 4-triazole, tetrazole, furan, thiophene, thiazole, oxazole, isothiazole, isoxazole, 1,2, 4-oxadiazole, 1,2, 5-oxadiazole, 1,2, 4-thiadiazole, 1,2, 5-thiadiazole, benzofuran, benzothiophene, indole, benzimidazole, benzothiazole, benzoxazole, benzisothiazole, and benzisoxazole.

"6-Membered heteroaromatic ring"and"benzo-fused 6-membered heteroaromatic ring" include, without limitation, pyridine, pyrimidine, pyridazine, pyrazine, triazine, quinoline, isoquinoline and quinoxaline.

Optical Isomers-Diastereomers-Geometric Isomers-Tautomers.

Compounds described herein may contain an asymmetric center and may thus exist as enantiomers. Where the compounds according to the invention possess two or more asymmetric centers, they may additionally exist as diastereomers. The present invention includes all such possible stereoisomers as substantially pure resolved enantiomers, racemic mixtures thereof, as well as mixtures of diastereomers. The above Formula I is shown without a definitive stereochemistry at certain positions.

The present invention includes all stereoisomers of Formula I and pharmaceutically acceptable salts thereof. Diastereoisomeric pairs of enantiomers may be separated by, for example, fractional crystallization from a suitable solvent, and the pair of enantiomers thus obtained may be separated into individual stereoisomers by conventional means, for example by the use of an optically active acid or base as a resolving agent or on a chiral HPLC column. Further, any enaritiomer or diastereomer of a compound of the general Formula I may be obtained by stereospecific synthesis using optically pure starting materials or reagents of known configuration.

Some of the compounds described herein contain olefinic double bonds, and unless specified otherwise, are meant to include both E and Z geometric isomers.

Some of the compounds described herein may exist with different points of attachment of hydrogen, referred to as tautomers. Such an example may be a ketone and its enol form known as keto- enol tautomers. The individual tautomers as well as mixture thereof are encompassed with compounds of Formula I.

Salts.

The term"pharmaceutically acceptable salts"refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids. When the compound of the present invention is acidic, its corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic bases, including inorganic bases and organic bases. Salts derived from such inorganic bases include aluminum,

ammonium, calcium, copper (ic and ous), ferric, ferrous, lithium, magnesium, manganese (ic and ous), potassium, sodium, zinc and the like salts. Preferred are the ammonium, calcium, magnesium, potassium and sodium salts. Salts prepared from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines derived from both naturally occurring and synthetic sources.

Pharmaceutically acceptable organic non-toxic bases from which salts can be formed include, for example, arginine, betaine, caffeine, choline, N, N-dibenzylethylenediamine, diethylamine, 2-diethyl- aminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N- ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, dicyclohexylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like.

When the compound of the present invention is basic, its corresponding salt can be conveniently prepared from pharmaceutically acceptable non-toxic inorganic and organic acids. Such acids include, for example, acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluene- sulfonic acid and the like. Preferred are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, and tartaric acids.

Prodrugs.

The present invention includes within its scope prodrugs of the compounds of this invention.

In general, such prodrugs will be functional derivatives of the compounds of this invention which are readily convertible in vivo into the required compound. Thus, in the methods of treatment of the present invention, the term"administering"shall encompass the treatment of the various conditions described with the compound specifically disclosed or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the patient. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs, "ed. H. Bundgaard, Elsevier, 1985. Metabolites of these compounds include active species produced upon introduction of compounds of this invention into the biological milieu.

Pharmaceutical Compositions.

Another aspect of the present invention provides pharmaceutical compositions which comprises a compound of Formula I, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. The term"composition", as in pharmaceutical composition, is intended to encompass a product comprising the active ingredient (s), and the inert ingredient (s)

(pharmaceutically acceptable excipients) that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of Formula I, additional active ingredient (s), and pharmaceutically acceptable excipients.

The pharmaceutical compositions of the present invention comprise a compound represented by Formula I (or pharmaceutically acceptable salts thereof) as an active ingredient, a pharmaceutically acceptable carrier and optionally other therapeutic ingredients or adjuvants. The compositions include compositions suitable for oral, rectal, topical, and parenteral (including subcutaneous, intramuscular, and intravenous) administration, although the most suitable route in any given case will depend on the particular host, and nature and severity of the conditions for which the active ingredient is being administered. The pharmaceutical compositions may be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.

In practice, the compounds represented by Formula I, or pharmaceutically acceptable salts thereof, of this invention can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e. g., oral or parenteral (including intravenous). Thus, the pharmaceutical compositions of the present invention can be presented as discrete units suitable for oral administration such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient. Further, the compositions can be presented as a powder, as granules, as a solution, as a suspension in an aqueous liquid, as a non- aqueous liquid, as an oil-in-water emulsion or as a water-in-oil liquid emulsion. In addition to the common dosage forms set out above, the compound represented by Formula I, or pharmaceutically acceptable salts thereof, may also be administered by controlled release means and/or delivery devices.

The compositions may be prepared by any of the methods of pharmacy. In general, such methods include a step of bringing into association the active ingredient with the carrier that constitutes one or more necessary ingredients. In general, the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both. The product can then be conveniently shaped into the desired presentation.

Thus, the pharmaceutical compositions of this invention may include a pharmaceutically acceptable carrier and a compound or a pharmaceutically acceptable salt of Formula I. The compounds of Formula I, or pharmaceutically acceptable salts thereof, can also be included in pharmaceutical compositions in combination with one or more other therapeutically active compounds.

The pharmaceutical carrier employed can be, for example, a solid, liquid, or gas. Examples of solid carriers include lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid. Examples of liquid carriers are sugar syrup, peanut oil, olive oil, and water. Examples of gaseous carriers include carbon dioxide and nitrogen.

In preparing the compositions for oral dosage form, any convenient pharmaceutical media may be employed. For example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like may be used to form oral liquid preparations such as suspensions, elixirs and solutions; while carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like may be used to form oral solid preparations such as powders, capsules and tablets. Because of their ease of administration, tablets and capsules are the preferred oral dosage units whereby solid pharmaceutical carriers are employed. Optionally, tablets may be coated by standard aqueous or nonaqueous techniques A tablet containing the composition of this invention may be prepared by compression or molding, optionally with one or more accessory ingredients or adjuvants. Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent. Each tablet preferably contains from about 0. lmg to about 500mg of the active ingredient and each cachet or capsule preferably containing from about 0. lmg to about 500mg of the active ingredient.

Pharmaceutical compositions of the present invention suitable for parenteral administration may be prepared as solutions or suspensions of the active compounds in water. A suitable surfactant can be included such as, for example, hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Further, a preservative can be included to prevent the detrimental growth of microorganisms.

Pharmaceutical compositions of the present invention suitable for injectable use include sterile aqueous solutions or dispersions. Furthermore, the compositions can be in the form of sterile powders for the extemporaneous preparation of such sterile injectable solutions or dispersions. In all cases, the final injectable form must be sterile and must be effectively fluid for easy syringability. The pharmaceutical compositions must be stable under the conditions of manufacture and storage; thus, preferably should be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e. g. glycerol, propylene glycol and liquid polyethylene glycol), vegetable oils, and suitable mixtures thereof.

Pharmaceutical compositions of the present invention can be in a form suitable for topical use such as, for example, an aerosol, cream, ointment, lotion, dusting powder, or the like. Further, the compositions can be in a form suitable for use in transdermal devices. These formulations may be prepared, utilizing a compound represented by Formula I of this invention, or pharmaceutically acceptable salts thereof, via conventional processing methods. As an example, a cream or ointment is prepared by mixing hydrophilic material and water, together with about 5 wt% to about 10 wt% of the compound, to produce a cream or ointment having a desired consistency.

Pharmaceutical compositions of this invention can be in a form suitable for rectal administration wherein the carrier is a solid. It is preferable that the mixture forms unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art. The suppositories may be conveniently formed by first admixing the composition with the softened or melted carrier (s) followed by chilling and shaping in moulds.

In addition to the aforementioned carrier ingredients, the pharmaceutical formulations described above may include, as appropriate, one or more additional carrier ingredients such as diluents, buffers, flavoring agents, binders, surface-active agents, thickeners, lubricants, preservatives (including anti-oxidants) and the like. Furthermore, other adjuvants can be included to render the formulation isotonic with the blood of the intended recipient. Compositions containing a compound described by Formula I, or pharmaceutically acceptable salts thereof, may also be prepared in powder or liquid concentrate form.

The following are examples of representative pharmaceutical dosage forms for the compounds of Formula I : Inj. Suspension (I. M.) mg/mL Tablet mg/tab Capsule mg/cap. Cmpd of Formula I 10 Cmpd of Formula I 25 Cmpd of Formula I 25 Methylcellulose5.0 Microcryst. Cellulose 415 Lactose Powder573. 5 Tween 80 0.5 Povidone 14.0 Magnesium Stearate 1.5 Benzyl alcohol 9.0 Pregelatinized Starch 43.5 600 Benzalkonium chloride 1.0 Magnesium Stearate 2.5 Water for injection to a total 500 volume of 1 mL Utilities.

Compounds of this invention are antagonists or inverse agonists of bradykinin receptor, in particular the bradykinin B 1 receptor, and as such are useful in the treatment and prevention of

diseases and conditions mediated through the bradykinin receptor pathway such as pain and inflammation. The compounds would be effective in the treatment or prevention of pain including, for example, visceral pain (such as pancreatitis, interstitial cystitis, renal colic, prostatitis, chronic pelvic pain), neuropathic pain (such as postheipetic neuralgia, acute zoster pain, nerve injury, the"dynias", e. g., vulvodynia, phantom limb pain, root avulsions, radiculopathy, painful traumatic mononeuropathy, painful entrapment neuropathy, carpal tunnel syndrome, ulnar neuropathy, tarsal tunnel syndrome, painful diabetic neuropathy, painful polyneuropathy, trigeminal neuralgia), central pain syndromes (potentially caused by virtually any lesion at any level of the nervous system including but not limited to stroke, multiple sclerosis, spinal cord injury), and postsurgical pain syndromes (eg, postmastectomy syndrome, postthoracotomy syndrome, stump pain) ), bone and joint pain (osteoarthritis), spine pain (e. g., acute and chronic low back pain, neck pain, spinal stenosis), shoulder pain, repetitive motion pain, dental pain, sore throat, cancer pain, burn pain, myofascial pain (muscular injury, fibromyalgia), postoperative, perioperative pain and preemptive analgesia (including but not limited to general surgery, orthopedic, and gynecological), chronic pain, dysmenorrhea (primary and secodnary), as well as pain associated with angina, and inflammatory pain of varied origins (e. g. osteoarthritis, rheumatoid arthritis, rheumatic disease, teno-synovitis and gout, ankylosing spondylitis, bursitis).

Further, the compounds of this invention can also be used to treat hyperreactive airways and to treat inflammatory events associated with airways disease e. g. asthma including allergic asthma (atopic or non-atopic) as well as exercise-induced bronchoconstriction, occupational asthma, viral-or bacterial exacerbation of asthma, other non-allergic asthmas and"wheezy-infant syndrome".

Compounds of the present invention may also be used to treat chronic obstructive pulmonary disease including emphysema, adult respiratory distress syndrome, bronchitis, pneumonia, allergic rhinitis (seasonal and perennial), and vasomotor rhinitis. They may also be effective against pneumoconiosis, including aluminosis, anthracosis, asbestosis, chalicosis, ptilosis, siderosis, silicosis, tabacosis and byssinosis.

Compounds of the present invention may also be used for the treatment of inflammatory bowel disease including Crohn's disease and ulcerative colitis, irritable bowel syndrome, pancreatitis, nephritis, cystitis (interstitial cystitis), uveitis, inflammatory skin disorders such as psoriasis and eczema, rheumatoid arthritis and edema resulting from trauma associated with burns, sprains or fracture, cerebral edema and angioedema (including hereditary angioedema and drug-induced angioedema such as that caused by angiotensin converting enzyme (ACE) or ACE/neutral endopeptidase inhibitors, e. g. omepatrilat). They may be used to treat diabetic vasculopathy, diabetic neuropathy, diabetic retinopathy, post capillary resistance or diabetic symptoms associated with insulitis (e. g. hyperglycemia, diuresis, proteinuria and increased nitrite and kallikrein urinary excretion). They may be used as smooth muscle

relaxants for the treatment of spasm of the gastrointestinal tract or uterus. Additionally, they may be effective against liver disease, multiple sclerosis, cardiovascular disease, e. g. atherosclerosis, congestive heart failure, myocardial infarct; neurodegenerative diseases, eg. Parkinson's and Alzheimer disease, epilepsy, septic shock e. g. as anti-hypovolemic and/or anti-hypotensive agents, headache including cluster headache, migraine including prophylactic and acute use, stroke, closed head trauma, cancer, sepsis, gingivitis, osteoporosis, benign prostatic hyperplasia and hyperactive bladder. Animal models of these diseases and conditions are generally well known in the art, and may be suitable for evaluating compounds of the present invention for their potential utilities. Finally, compounds of the present invention are also useful as research tools (irv vivo and ill vitro).

The compounds of this invention are useful in the treatment of pain and inflammation by the administration of a tablet, cachet, or capsule each containing, for example, 0. lmg, 0. 5mg, lmg, 3mg, 5mg, lOmg, 25mg, 50mg, 100mg, 125mg, 250mg, or 500mg of a compound of this invention once every three to four hours, once, twice or three times a day, or (in an extended release formulation) once, twice or three times a week.

The compounds would be effective in the treatment or prevention of pain including, for example, bone and joint pain (osteoarthritis), repetitive motion pain, dental pain, cancer pain, myofascial pain (muscular injury, fibromyalgia), perioperative pain (general surgery, oral surgery, gynecological), neuropathic pain (post-herpetic neuralgia), and chronic pain by the administration of a tablet, cachet, or capsule each containing, for example, 0. lmg, 0.5mg, lmg, 3mg, 5mg, lOmg, 25mg, 50mg, 100mg, 125mg, 250mg, or 500mg of a compound of this invention once every three to four hours, once, twice or three times a day, or (in an extended release formulation) once, twice or three times a week.

In particular, inflammatory pain such as, for example, inflammatory airways disease (chronic obstructive pulmonary disease) would be effectively treated by the compounds of this invention by the administration of a tablet, cachet, or capsule each containing, for example, 0. lmg, 0. 5mg, lmg, 3mg, 5mg, lOmg, 25mg, 50mg, 100mg, 125mg, 250mg, or 500mg of a compound of this invention once every three to four hours, once, twice or three times a day, or (in an extended release formulation) once, twice or three times a week.

Further, the compounds of this invention can additionally be used to treat asthma, inflammatory bowel disease, rhinitis, pancreatitis, cystitis (interstitial cystitis), uveitis, inflammatory skin disorders, rheumatoid arthritis and edema resulting from trauma associated with burns, sprains or fracture by the administration of a tablet, cachet, or capsule each containing, for example, 0. lmg, 0. 5mg, lmg, 3mg, 5mg, lOmg, 25mg, 50mg, 100mg, 125mg, 250mg, or 500mg of a compound of this invention once every three to four hours, once, twice or three times a day, or (in an extended release formulation) once, twice or three times a week.

They may be used subsequent to surgical intervention (e. g. as post-operative analgesics) and to treat inflammatory pain of varied origins (e. g. osteoarthritis, rheumatoid arthritis, rheumatic disease, teno-synovitis and gout) as well as for the treatment of pain associated with angina, menstruation or cancer by the administration of a tablet, cachet, or capsule each containing, for example, 0. lmg, 0.5mg, lmg, 3mg, 5mg, lOmg, 25mg, 50mg, 100mg, 125mg, 250mg, or 500mg of a compound of this invention once every three to four hours, once, twice or three times a day, or (in an extended release formulation) once, twice or three times a week.

They may be used to treat diabetic vasculopathy, post capillary resistance or diabetic symptoms associated with insulitis (e. g. hyperglycemia, diuresis, proteinuria and increased nitrite and kallikrein urinary excretion) by the administration of a tablet, cachet, or capsule each containing, for example, O. lmg, 0. 5mg, lmg, 3mg, 5mg, lOmg, 25mg, 50mg, 100mg, 125mg, 250mg, or 500mg of a compound of this invention once every three to four hours, once, twice or three times a day, or (in an extended release formulation) once, twice or three times a week.

They may be used to treat inflammatory skin disorders such as psoriasis and eczema by the administration of a tablet, cachet, or capsule each containing, for example, 0. lmg, 0. 5mg, lmg, 3mg, 5mg, lOmg, 25mg, 50mg, 100mg, 125mg, 250mg, or 500mg of a compound of this invention once every three to four hours, once, twice or three times a day, or (in an extended release formulation) once, twice or three times a week.

They may be used as smooth muscle relaxants for the treatment of spasm of the gastrointestinal tract or uterus or in the therapy of Crohn's disease, ulcerative colitis or pancreatitis by the administration of a tablet, cachet, or capsule each containing, for example, O. lmg, 0. 5mg, lmg, 3mg, 5mg, lOmg, 25mg, 50mg, 100mg, 125mg, 250mg, or 500mg of a compound of this invention once every three to four hours, once, twice or three times a day, or (in an extended release formulation) once, twice or three times a week.

Such compounds may be used therapeutically to treat hyperreactive airways and to treat inflammatory events associated with airways disease e. g. asthma, and to control, restrict or reverse airways hyperreactivity in asthma by the administration of a tablet, cachet, or capsule each containing, for example, O. lmg, 0.5mg, lmg, 3mg, 5mg, lOmg, 25mg, 50mg, 100mg, 125mg, 250mg, or 500mg of a compound of this invention once every three to four hours, once, twice or three times a day, or (in an extended release formulation) once, twice or three times a week.

They may be used to treat intrinsic and extrinsic asthma including allergic asthma (atopic or non-atopic) as well as exercise-induced bronchoconstriction, occupational asthma, viral or bacterial exacerbated asthma, other non-allergic asthmas and"wheezy-infant syndrome"by the administration of a tablet, cachet, or capsule each containing, for example, 0. lmg, 0. 5mg, lmg, 3mg, 5mg, lOmg, 25mg,

50mg, 100mg, 125mg, 250mg, or 500mg of a compound of this invention once every three to four hours, once, twice or three times a day, or (in an extended release formulation) once, twice or three times a week.

They may also be effective against pneumoconiosis, including aluminosis, anthracosis, asbestosis, chalicosis, ptilosis, siderosis, silicosis, tabacosis and byssinosis was well as adult respiratory distress syndrome, chronic obstructive pulmonary or airways disease, bronchitis, allergic rhinitis, and vasomotor rhinitis by the administration of a tablet, cachet, or capsule each containing, for example, O. lmg, 0. 5mg, lmg, 3mg, 5mg, lOmg, 25mg, 50mg, 100mg, 125mg, 250mg, or 500mg of a compound of this invention once every three to four hours, once, twice or three times a day, or (in an extended release formulation) once, twice or three times a week.

Additionally, they may be effective against liver disease, multiple sclerosis, atherosclerosis, Alzheimer's disease, septic shock e. g. as anti-hypovolemic and/or anti-hypotensive agents, cerebral edema, headache including cluster headache, migraine including prophylactic and acute use, closed head trauma, irritable bowel syndrome and nephritis by the administration of a tablet, cachet, or capsule each containing, for example, O. lmg, 0. 5mg, lmg, 3mg, 5mg, lOmg, 25mg, 50mg, 100mg, 125mg, 250mg, or 500mg of a compound of this invention once every three to four hours, once, twice or three times a day, or (in an extended release formulation) once, twice or three times a week.

Combination Therapy.

Compounds of Formula I may be used in combination with other drugs that are used in the treatment/prevention/suppression or amelioration of the diseases or conditions for which compounds of Formula I are useful. Such other drugs may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with a compound of Formula I. When a compound of Formula I is used contemporaneously with one or more other drugs, a pharmaceutical composition containing such other drugs in addition to the compound of Formula I is preferred. Accordingly, the pharmaceutical compositions of the present invention include those that also contain one or more other active ingredients, in addition to a compound of Formula I. Examples of other active ingredients that may be combined with a compound of Formula I, either administered separately or in the same pharmaceutical compositions, include, but are not limited to: (1) morphine and other opiate receptor agonists including codeine, oxycodone, propoxyphene (Darvon) and tramadol; (2) non-steroidal antiinflammatory drugs (NSAIDs) including COX-2 inhibitors such as propionic acid derivatives (alminoprofen, benoxaprofen, bucloxic acid, carprofen, fenbufen, fenoprofen, fluprofen, flurbiprofen, ibuprofen, indoprofen, ketoprofen, miroprofen, naproxen, oxaprozin, piiprofen, pranoprofen, suprofen, tiaprofenic acid, and tioxaprofen), acetic acid derivatives (indomethacin, acemetacin, alclofenac,

clidanac, diclofenac, fenclofenac, fenclozic acid, fentiazac, furofenac, ibufenac, isoxepac, oxpinac, sulindac, tiopinac, tolmetin, zidometacin, and zomepirac), fenamic acid derivatives (flufenamic acid, meclofenamic acid, mefenamic acid, niflumic acid and tolfenamic acid), biphenylcarboxylic acid derivatives (diflunisal and flufenisal), oxicams (isoxicam, piroxicam, sudoxicam and tenoxican), salicylates (acetyl salicylic acid, sulfasalazine) and the pyrazolones (apazone, bezpiperylon, feprazone, mofebutazone, oxyphenbutazone, phenylbutazone), and the coxibs (celecoxib, valecoxib, rofecoxib and etoricoxib); (3) corticosteroids such as betamethasone, budesonide, cortisone, dexamethasone, hydrocortisone, methylprednisolone, prednisolone, prednisone and triamcinolone; (4) histamine H1 receptor antagonists such as bromopheniramine, chlorpheniramine, dexchlorpheniramine, triprolidine, clemastine, diphenhydramine, diphenylpyraline, tripelennamine, hydroxyzine, methdilazine, promethazine, trimeprazine, azatadine, cyproheptadine, antazoline, pheniramine pyrilamine, astemizole, terfenadine, loratadine, cetirizine, desloratadine, fexofenadine and levocetirizine; (5) histamine H2 receptor antagonists such as cimetidine, famotidine and ranitidine; (6) proton pump inhibitors such as omeprazole, pantoprazole and esomeprazole; (7) leukotriene antagonists and 5-lipoxygenase inhibitors such as zafirlukast, montelukast, pranlukast and zileuton; (8) drugs used for angina, myocardial ischemia including nitrates such as nitroglycerin and isosorbide nitrates, beta blockers such as atenolol, metoprolol, propranolol, acebutolol, betaxolol, bisoprolol, carteolol, labetalol, nadolol, oxprenolol, penbutolol, pindolol, sotalol and timolol, and calcium channel blockers such as diltiazam, verapamil, nifedipine, bepridil, felodipine, flunarizine, isradipine, nicardipine and nimodipine; (9) incontinence medications such as antimuscarinics, e. g. , tolterodine and oxybutinin); (10) gastrointestinal antispasmodics (such as atropine, scopolamine, dicyclomine, antimuscarinics, as well as diphenoxylate); skeletal muscle relaxants (cyclobenzaprine, carisoprodol, chlorphenesin, chlorzoxazone, metaxalone, methocarbamol, baclofen, dantrolene, diazepam, or orphenadrine); (11) gout medications such as allopurinol, probenicid and colchicine; (12) drugs for rheumatoid arthritis such as methotrexate, auranofin, aurothioglucose and gold sodium thiomalate; (13) drugs for osteoporosis such as alendronate and raloxifene; (14) decongestants such as pseudoephedrine and phenylpropanolamine; (15) local anesthetics; (16) anti-herpes drugs such as acyclovir, valacyclovir and famcyclovir; (17) anti-emetics such as ondansetron and granisetron; (18) migraine drugs such as the triptans (e. g. rizatriptan, sumatrip- tan), ergotamine, dihydroergotamine, CGRP antagonists, (19) antidepressants (e. g. , tricyclic antidepres- sants (such as doxepin, clomipramine, imipramine, amitriptyline, maprotiline, nortriptyline), serotonin- selective/serotonin and norepinephrine reuptake inhibitors (such as paroxetine, fluoxetine, duloxetine, vanlafexine), beta-adrenergic blockers; (20) VR1 antagonsits; (21) anticonvulsants (e. g. , gabapentin, pregabalin, lamotrigine, topiramate, carbamazepine, oxcarbazepine, phenytoin); (22) glutamate antagonists (e. g. , ketamine and other NMDA antagonists, NR2B antagonists); (23) acetaminophen ; (24) CCR2 antagonists; (25) PDE4 antagonists such as roflumilast ; (26) tegaserod; (27) alosetron; (28) topiramate; (29) cathepsin K inhibitors; and (30) ACE/NEP inhibitors such as omepatrilat.

Biological Evaluation.

(a) Assessing the Affinity of Selected Compounds to Bind to the Bradykinin B 1 or B2 Receptor.

Radioligand binding assays are performed using membranes from CHO cells that stably express the human, rabbit, rat, or dog B 1 receptors or CHO cells that express the human B2 receptor. For all receptor types, cells are harvested from culture flasks in PBS/ImM EDTA and centrifuged at 1000xg for 10 minutes. The cell pellets are homogenized with a polytron in ice cold 20mM HEPES, 1mM EDTA, pH 7.4 (lysis buffer) and centrifuged at 20, 000xg for 20 minutes. The membrane pellets are rehomogenized in lysis buffer, centrifuged again at 20, 000xg and the final pellets are resuspended at 5mg protein/ml in assay buffer (120mM NaCl, 5mM KC1, 20mM HEPES, pH 7.4) supplemented with 1% BSA and frozen at-80°C.

On the day of assay, membranes are centrifuged at 14, 000xg for 5 minutes and resuspended to the desired protein concentration in assay buffer containing 100nM enaliprilat, 140, ug/rnL bacitracin and 0.1% BSA. 3H-des-arglO, leu9 kallidin is the radioligand used for the human and rabbit B1 receptors, 3H-des-arglO kallidin is used for the rat and dog B l receptors, and 3H-bradykinin is used to label the human B2 receptor.

For all assays, compounds are diluted from DMSO stock solutions with 4, uL added to assay tubes for a final DMSO concentration of 2%. This is followed by the addition of 100/1L radioligand and 100µL of the membrane suspension. Nonspecific binding for the B 1 receptor binding assays is determined using llim des-arglo kallidin and nonspecific binding for the B2 receptor is determined with l, uM bradykinin. Tubes are incubated at room temperature (22°C) for 60 minutes followed by filtration using a Tomtec 96-well harvesting system. Radioactivity retained by the filter is counted using a Wallac Beta-plate scintillation counter.

The compounds of this invention have affinity for the B l receptor in the above assay as demonstrated by results of less than 5jHM. It is advantageous that the assay results be less than litm, even more advantageous for the results be less than 0. 5tM. It is further advantageous that compounds of this invention have affinity for the bradykinin B l receptor over the bradykinin B2 receptor; more advantageously, the affinity for the B 1 receptor is at least 10 fold, and preferably over 100 fold, over that for the B2 receptor.

(b) Assay for Bradykinin B 1 Antagonists.

B 1 agonist-induced calcium mobilization was monitored using a Fluorescence Imaging Plate Reader (FLIPR). CHO cells expressing the B 1 receptor were plated in 96 or 384 well plates and allowed to incubate in Iscove's modified DMEM overnight. Wells were washed two times with a physiological buffered salt solution and then incubated with 4uM Fluo-3 for one hour at 37°C. The plates were then washed two times with buffered salt solution and 100uL of buffer was added to each well. Plates were placed in the FLIPR unit and allowed to equilibrate for two minutes. The test compound was then added in 50ul volumes followed five minutes later by 50ul of agonist (des-argl° kallidin). Relative fluorescence peak heights in the absence and presence of antagonist were used to calculate the degree of inhibition of the B 1 receptor agonist response by the test compound. Eight to ten concentrations of test compound were typically evaluated to construct an inhibition curve and determine IC50 values using a four- parameter nonlinear regression curve fitting routine.

(c) Assay for Bradykinin Inverse Agonists.

Inverse agonist activity at the human B 1 receptor was evaluated using transiently transfected HEK293 cells. One day following transfection cell flasks were labeled overnight with 6uCi/ml [3H] myo- inositol. On the day of assay, the media was removed and the attached cells were gently rinsed with 2x20ml of phosphate-buffered saline. Assay buffer (HEPES buffered physiological salts, pH 7.4) was added and the cells were detached by tapping of the flask. The cells were centrifuged at 800xg for five minutes and resuspended at lx106 cells/ml in assay buffer supplemented with lOmM lithium chloride.

After 10 minutes at room temperature, one-half ml aliquots were distributed to tubes containing test compound or vehicle. After an additional 10 minutes the tubes were transferred to a 37°C water bath for 30 minutes. The incubation was terminated by the addition of a 12% perchloric acid solution and the tubes were placed on ice for 30 minutes. The acid was then neutralized with KOH and the tubes centrifuged to pellet precipitated material. [3H] Inositol monophosphate formed was recovered by standard ion exchange chromatographic techniques and quantitated by liquid scintillation counting.

Inverse agonist activity was determined by the degree to which a test compound reduced basal (cells incubated with vehicle) levels of [3H] inositol monophosphate accumulation.

Abbreviations Used.

The following abbreviations have the meanings indicated, unless stated otherwise in the specification: Ac=acetyl; Boc=t-butoxycarbonyl; Cat. =catalyst; DCM=dichloromethane; DMF=dimethylformamide; DMADMA=dimethylacetamide dimethyl acetal; DMSO=dimethyl sulfoxide; EDCI=1- (3- dimethylaminopropyl)-3-ethylcarbodiimide ; Et=ethyl; EtOAc=ethyl acetate; HOBT=1-

hydroxybenzotriazole; LAH=lithium aluminum hydride; LDA=lithium diisopropylamide; LiHMDS=lithium hexamethyldisilazide; Me=methyl; NBS=N-bromosuccinimide; Ph=phenyl; Rt=room temperature; Tf=triflyl (trifluoromethanesulfonyl); THF=tetrahydrofuran.

Methods of Synthesis.

Compounds of formula I may be prepared by the general procedure depicted in Scheme 1.

The a-hydroxycarboxylic acid (1) is coupled with the arylmethylamine (2) using standard reagents and reaction conditions for amide bond formation, such as EDCI/HOBt.

SCHEME 1 The carboxylic acids (1) are commercially available or may be prepared from commercially available reagents using conventional chemical reactions well known in the art. The amines (2) may be prepared as outlined in Schemes 2 to 6. In Scheme 2, the biaryl derivative (5) is assembled using a Suzuki reaction between an aromatic boronic acid derivative (3), or an appropriate boronic ester derivative, and an aromatic halide (4) in the presence of a triarylphosphine, like triphenylphosphine, and a metal catalyst, like palladium acetate. The resultant cyano biphenyl intermediate (5), is then catalytically reduced to the biaryl amine derivative (2a) using hydrogen and a metal, like Raney Ni, in an appropriate solvent.

SCHEME 2 R6 CN, NH2 NC R y y OH R9 (4) NC Y// Y , I/OH Rs 4) \ \ I g Raney Ni R7 Pd cat. Raney Ni J R Rs \ J R6 (5) R (2a) Rs

Alternatively, as illustrated in Scheme 3, the amine derivative (6), after primary amine protection with an appropriate protecting group, like Boc, is elaborated to the pinacol boron ester (8) using a palladium catalyst in an appropriate solvent, like dimethyl sulfoxide. This boron ester (8) is coupled to an aryl halide derivative (4) employing Suzuki reaction conditions to yield (2a) following deprotection.

SCHEME 3

A third method for the preparation of biarylmethanamine derivatives is depicted in Scheme 4. The biaryl moiety (11) is first assembled using a palladium catalyzed coupling of (9) with an aryl zinc compound (10) as shown. The methyl group of biaryl (11) is then elaborated according to the three step sequence of halogenation, nucleophilic displacement of the halogen with azide, and reduction to provide

the corresponding amine intermediate (2a). Alternatively, in a fourth method, the biarylmethanamine (2a) can also be prepared starting from the arylcarbonitrile (13) and aryl zinc compound (10) as previously discussed. The resulting biarylcarbonitrile (5) is then reduced using hydrogen to provide (2a).

SCHEME 4

A fifth method for the preparation of biarylmethanamine derivatives is depicted in Scheme 5. Reduction of the nitro group and nitrile hydrolysis of known pyridine (14) (J. Chenu. Soc, (1952), 2042-2046. ) is followed by conversion of the resultant amine to the fluoride to afford (15). The amide is converted in a 3 step sequence to aldehyde (16). Imine formation with t-butyl sulfinamide is followed by addition of methyl Grignard to produce (17), which may be further elaborated to provide the biarylmethanamine as shown above.

SCHEME 5. Br NO Br F 1-HCI Br F Br N02 1-SnC12 2-EDC N H HNMe (OMe) N CHO co N CN 2-NOBF4 N CNH2 3-LAH (14) (15) Br F 1-t-butyl sulfinamide 2-MeMgCI N l S (17) CH3 O

The reaction steps in Scheme 6 illustrates the preparation of a compound of Formula (I) wherein RI is the benzisothiazoline moiety. While Scheme 6 depicts the preparation of a specific compound, the procedure may be used to prepare other similar compounds of Formula (I).

SCHEME 6 Cu20 H pyridine 0 NaH N\ S02CI S02NH2// CI NH40H 9\CI 180 °C </o allyl-Br WJSt0 nab LiHMDS, THF Ns, 0 LiHMDS, THF N 0 NaBH4 _ S-- ; FN SO Ph, S FN (S02Ph) 2, w (Pd (PPh3) 4 (2) 2 F -78 °C F-78 °C F HCINH2 F 1-cru (OAc) 2 ! ! \_"0 | triethylamine Me o DCM, 2. 5 d/ F 2-HCI F EtOAc B02H2

Reference Examples.

The following examples are provided to illustrate the preparation of the biarylmethylamine starting materials. Other biarylmethylamines may be similarly prepared.

I. Methvl 2-r6-(aminomethyl)-5-fluoropyridin-3-yll-6-fluorobenzoate.

A solution of LDA (40.9 mmol, prepared from 11.4 mL of diisopropylamine and 16.4 mL of 2.5 M n-butyl lithium in hexanes) in 200 mL THE at-78°C was treated with 2-cyano-3-fluoropyridine (5.0 g, 40.9 mmol) in 50 mL of THF drop-wise. After 10 minutes a solution of iodine (10.4 g, 40.9 mmol) in 10 mL of THF was added. After 30 minutes the reaction was quenched with 40 mL of water followed by workup with aqueous sodium thiosulfate. The mixture was diluted with ether, washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was subjected to silica gel chromatography eluted with 0-20% ethyl acetate in hexanes to provide 3-fluoro-4-iodopyridine-2- carbonitrile that gave proton NMR spectra consistent with theory- A solution of LDA (16.9 mmol) in 200 mL THF at-78 °C was treated with the above carbonitrile (4.2 g, 16.9 mmol) in 50 mL of THF drop-wise. After 2 hours the reaction was quenched with water and warmed to room temperature. The mixture was diluted with ether, washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was subjected to silica gel chromatography eluted with 0-20% ethyl acetate in hexanes to provide 3-fluoro-5-iodopyridine-2- carbonitrile that gave proton NMR spectra consistent with theory.

A solution of the above carbonitrile (1.08 g, 3.87 mmol) in 10 mL of THF and palladium tetrakistriphenylphosphine (0.18 g, 0.16 mmol) was added to a solution of [3-fluoro-2- (methoxy- carbonyl) phenyl] (iodo) zinc (prepared from methyl 2-fluoro-6-iodobenzoate and Reike Zinc) in 20 mL of THF via cannula. The mixture was heated to reflux for one hour, cooled, and partitioned between ethyl acetate and water. The organic extract was washed with brine, dried over MgSO4, filtered and concentrated under vacuum. The residue was subjected to silica gel chromatography eluted with 0-20%

ethyl acetate in hexanes to provide methyl 2- (6-cyano-5-fluoropyridin-3-yl)-6-fluorobenzoate that gave proton NMR spectra consistent with theory.

To a stirred solution of the above ester (0.75 g, 2.7 mmol) in NH3/MeOH (5 mL, 2.0 M) was added Raney 2800 nickel (slurry in water). The mixture was stirred under a H2 atmosphere (balloon) at room temperature for 6 hours. The mixture was then filtered through glass filter paper, washing with additional MeOH. The resultant solution was concentrated under vacuum and azeotroped three times with toluene to provide crude title compound (0.65 g, 2.34 mmol).

II. (lR)-l- 5-F3, 5-dichloro-2- (methoxycarbgLnyl) phenyll-3-fluoropyridin-2- yl} ethanaminium chloride.

A solution of 2, 4-dichloro-6-hydroxybenzaldehyde (5. 00g, 26.18 mmol) in 125 mL methanol was cooled to 0 °C. Perchloric acid (70%, 1.47 mL, 16.23 mmol) was added, and the solution was stirred for 10 minutes. To a separate flask, vanadium (V) oxide (0.190 g, 1.05 mmol) was added to a hydrogen peroxide solution (30% in H20, 11.90 mL, 104.7 mmol) at 0 °C. This solution was stirred until the catalyst was dissolved, resulting in a clear orange solution, which was added dropwise to the methanol solution. The reaction was allowed to slowly warm to room temperature and stir overnight. The solution was concentrated under vacuum, and the residue dissolved in ethyl acetate. The organic extract was washed with aqueous sodium bicarbonate and brine, dried over Na2S04, filtered and concentrated. The residue was filtered through a silica gel plug with 10% ethyl acetate in hexanes to provide methyl 2,4- dichloro-6-hydroxybenzoate that gave proton NMR spectra consistent with theory.

A solution of the above ester (4.64g, 20.99 mmol) and pyridine (1.87 mL, 23.09 mmol) in 100 mL CH2C12 was cooled to 0 °C. Trifluoromethanesulfonic acid (4.94 mL, 29.39 mmol) was added, and the solution was stirred for 2h. The reaction mixture was washed with aqueous sodium bicarbonate, aqueous copper sulfate and brine, dried over Na2SO4, filtered and concentrated to provide methyl 2,4- dichloro-6- { [ (trifluoromethyl) sulfonyl] oxy} benzoate that gave proton NMR spectra consistent with theory.

To a solution of 5-bromo-3-nitropyridine-2-carbonitrile (4.71 g, 20.7 mmol) in MeOH (319 rnL) under N2 was added tin (II) chloride dihydrate (27.97 g, 123.9 mmol). The reaction was heated to 40 °C for 40 minutes, concentrated in vacuo, and azeotroped with toluene. The residue was dissolved in ethyl acetate, and 10% aqueous sodium bicarbonate was added till the solution was basic. The aqueous layer was extracted 3x with CHC13 and the combined organics were dried over Na2SO4, filtered and concentrated under vacuum to provide 3-amino-5-bromopyridine-2-carboxamide that gave a mass ion (ES+) of 218.2 for M+H+ (8lBr)

To a solution of the above carboxamide (40.0 g, 185.2 mmol) in CH2C12 was added nitrosonium tetrafluoroborate (22. 7 in, 191. 4 mmol). The reaction was stirred at room temperature for 4.5 hours, then concentrated in vacuo and azeotroped with toluene. The residue was suspended in toluene (1100 mL) and heated to 100 °C for 2 hours. The reaction was concentrated in vacuo, and the residue suspended in CH2C12. The solid was collected to provide 5-bromo-3-fluoropyridine-2- carboxamide that gave proton NMR spectra consistent with theory and a mass ion (ES+) of 219.1 for M+H+ (79Br).

A solution of the above crude carboxamide (40. 50 g, 184.9 mmol) in HC1 (12N, 539.4 mL, 6.472 mol) was heated to 120 °C for 1 hour. The reaction was cooled to room temperature, and NaOH (20%) was added slowly to pH ~6. The solution was concentrated in vacuo, and azeotroped 3x with toluene. The residue was extracted thoroughly with 40% MeOH/CHCl3 and filtered. This was repeated three times. The combined filtrates were concentrated and azeotroped 3x with toluene to provide 5- bromo-3-fluoropyridine-2-carboxylic acid that gave a mass ion (ES+) of 218.1 for M+H+ (79Br).

A solution of the above crude arboxylic acid (40.65 g, 184.8 mmol), O, N-dimethylhydroxyl- amine hydrochloride (21.63 g, 221.7 mmol), 1-ethyl- (3-dimethylaminopropyl)-carbodiimide hydrochloride (70.85 g, 369. 6 mmol), 1-hydroxy-7-azabenzotriazole (2.497 g, 18.48 mmol), and triethylamine (16.48 mL, 118.3 mmol) in 200 mL DMF was stirred at RT overnight. The solution was partially concentrated in vacuo and partitioned between ethyl acetate and 10% aqueous sodium bicarbonate. The aqueous layer was extracted 4x with ethyl acetate, and the combined organics were washed with water and brine, dried over Na2SO4, filtered and concentrated under vacuum to provide 5- bromo-3-fluoro-N-methoxy-N-methylpyridine-2-carboxamide that gave proton NMR spectra consistent with theory and a mass ion (ES+) of 263.01 for M+H+ (79Br) To a solution of the above crude carboxamide (27.94 g, 106.2 mmol) in THF (350 mL) at - 78 °C was added LAH (1M in THF, 45.67 mL, 45.67 mmol) dropwise. The reaction was stirred at-78 °C for 2 hours, then H20 (100 mL) and brine (100 mL) were added. The mixture was warmed to RT and partially concentrated in vacuo, diluted with ethyl acetate and filtered through celite. The aqueous layer was extracted 4x with ethyl acetate. The combined organics were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was filtered through a silica gel plug with 10% ethyl acetate in hexanes to provide 5-bromo-3-fluoropyridine-2-carbaldehyde that gave proton NMR spectra consistent with theory.

To a solution of (R)- (+)-2-methyl-2-propanesulfinamide (9.898 g, 81.67 mmol) in CH2C12 (160 mL) was added 5-bromo-3-fluoropyridine-2-carbaldehyde (16.66 g, 81.67 mmol), pyridiniump- toluenesulfonate (1.026 g, 4.08 mmol), and magnesium sulfate (49.15 g, 408.3 mmol). The reaction was stirred at room temperature overnight, then filtered through celite and concentrated in vacuo. The

residue was subjected to silica gel chromatography eluted with 0 to 10% ethyl acetate in hexanes to provide N- [ (lE)- (5-bromo-3-fluoropyridin-2-yl) methylidene] -2-methylpropane-2-sulfinamide that gave proton NMR spectra consistent with theory and a mass ion (ES+) of 307.0 for M+H+ (8lBr).

A solution of the above sulfinamide (18. 63 g, 60.65 mmol) in CH2C12 (375 mL) was cooled to-50 °C under N2. Methylmagnesium chloride (3M in THF, 30.32 mL, 90.97 mmol) was added dropwise, the reaction was stirred for lh. Additional methylmagnesium chloride (5.0 mL, 15.0 mmol) was added after 30 minutes to drive the reaction to completion. Water (200 mL) and brine (200 mL) were added, and the reaction allowed to warm to room temperature. The aqueous layer was extracted 4x with CH2C12, and the combined organics were dried over Na2SO4, filtered and concentrated under vacuum. The residue was subjected to silica gel chromatography eluted with 10-40% ethyl acetate in hexanes to provide N- [ (lR)-l- (5-bromo-3-fluoropyridin-2-yl) ethyl] -2-methylpropane-2-sulfinamide that gave proton NMR spectra consistent with theory and a mass ion (ES+) of 325. 0 for M+H+ (81Br) A mixture of the above. sulfinamide (0.500 g, 1.55 mmol), bis (pinacolato) diboron (0.412 g, 1.62 mmol), potassium acetate (0.456 g, 4.64 mmol), and [1, 1'-bis (diphenylphosphino) ferrocene]- palladium (II) dichloride (0.030 g, 0.041 mmol) in 5 mL DMF was heated to 90 °C under N2 for 4 hours.

Additional bis (pinacolato) diboron (0.295 g, 1.16 mmol) and 3-chloroperoxybenzoic acid (861 mg, 4.99 mmol) and [l, l'-bis (diphenylphosphino) ferrocene] palladium (ll) dichloride (0.030 g, 0.041 mmol) were added to drive the reaction to completion. The reaction mixture was cooled to room temperature, and methyl 2, 4-dichloro-6-{[(trifluoromethyl) sulfonyl] oxy} benzoate (0.546 g, 1.55 mmol), sodium carbonate (2M, 2.32 mL, 4. 64 mmol), and [1, l'-bis (diphenylphosphino) ferrocene] palladium (II) dichloride (0.003 g, 0.041 mmol) were added. The reaction mixture was heated to 90 °C 1.5 hours, then cooled to room temperature and partitioned between ethyl acetate and water. The organic extract was washed with water and brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was subjected to silica gel chromatography eluted with 0-30% ethyl acetate in hexanes to provide methyl 2- (6- { (1R)-1- [(tert-butylsulfinyl) amino] ethyl}-5-fluoropyridin-3-yl)-4, 6-dichlorobenzoate that gave proton NMR spectra consistent with theory.

To a solution of the above product in methanol (1.2 mL) was added HCl/dioxane solution (4M, 1.2 mL, 4.6 mmol). The solution was stirred at room temperature 30 minutes, then concentrated in vacuo to provide the title compound (ES+) of 343.01 for M+H+. m. Methyl 4'-rflR)-l-aminoethyll-3-fluoro-1. 1'-biphenyl-2-carboxylate HCI.

Commercially available (lR)-1-(4-bromophenyl) ethanamine was Boc protected, using standard procedures known to those skilled in the art, to produce tert-butyl (lR)-1-(4-bromophenyl) ethyl- carbamate.

To a solution of the above carbamate (7.6 g, 25.3 mmol) in DMSO (20 mL) was added bis (pinacolato) diboron (7.07 g, 27.9 mmol), dichloro [l, l'-bis (diphenylphosphino) ferrocene] palladium (II) dichloromethane adduct (2.06 g, 2.53 mmol), and potassium acetate (7.45 g, 76.0 mmol) at room temperature under N2. The resulting mixture was heated at 80 °C for 1 hour. The reaction was quenched by addition of EtOAc and filtered through celite. The organic extract was washed with water three times, saturated NaCl, dried over MgSO4, filtered and concentrated under vacuum. The residue was purified on silica gel eluted with 0-20% ethyl acetate in hexane to provide tert-butyl (lR)-l- [4- (4, 4,5, 5-tetramethyl- 1, 3,2-dioxaborolan-2-yl) phenyl] ethylcarbamate as a clear light yellow oil with a mass ion (ES+) of 333.

To a stirred solution of the above carbamate (1.0 g, 2.9 mmol) and methyl 2-fluoro-6- iodobenzoate (1.2 g, 4.32 mmol) in 25 mL of a 5: 1 THF : water mixture was added potassium carbonate (1.2 g, 8.64 mmol), tri-o-tolylphosphine (350 mg, 1.15 mmol) and lastly palladium acetate (65 mg, 0.29 mmol). The reaction vessel was then sealed and placed into a 90 °C oil bath for overnight stirring and heating. After about 18 hours the reaction mixture was cooled to ambient temperature and then diluted with EtOAc. The organics were washed with brine (x4), dried over sodium sulfate, filtered, and concentrated under reduced pressure to give an oil. This oil was subject to silica gel chromatography eluting with 10-60% EtOAc in hexanes to provide methyl 4'-{(lR)-l-[(tert-butoxycarbonyl) amino] - ethyl}-3-fluoro-1, 1'-biphenyl-2-carboxylate (205 mg), found to be pure by LC/MS and proton NMR.

The above carboxylate (205 mg, 0.60 mmol) dissolved in MeOH (15 mL) was cooled to 0 °C. This homogenous solution was saturated with anhydrous HC1 and allowed to sit for 20 minutes.

Dry nitrogen was then bubbled through the solution for about 30 minutes. Solvent was then removed under reduced pressure to yield an oily residue. The oil was then dissolved in DCM and the solvent removed. This process was repeated until a solid amine hydrochloride was obtained.

IV. N-r (lR)-1- (5-Bromo-3-fluoropyridin-2-yl) ethyllacetamide.

Step 1. 2s5-Dibromo-3-nitropyridine.

2-Hydroxy-3-nitro-5-bromopyridine (1 eq) was suspended in toluene (3 vol) and N, N- dimethylformamide (0.1 eq) was added. The mixture was protected from light. A solution of phosphorus oxybromide (1. 2 eq) in toluene (2 vol) was added to the pyridine mixture over 1.5 h at about 90 °C and the reaction was aged for about 14 h at 90 °C. The reaction mixture was cooled to room temperature and water (10 vol) and toluene (5 vol) were added. The layers were cut and the organic layer was washed with 1N NaOH (2 x 10 vol) and H20 (5 vol). The batch was concentrated under reduced pressure to yield the desired product.

Step 2. 5-Bromo-3-nitropyridine-2-carbonitrile.

The compound of Step 1 (1 eq) was suspended in propionitrile (3 vol). Copper cyanide (1. 1 eq) was added and the mixture was heated to 90 °C and aged for about 17 h. The reaction mixture was cooled to room temperature and isopropyl acetate (12 vol) and saturated brine (8 vol) were added.

The mixture was stirred for 15 min and the layers were cut. The top organic layer was washed with brine (4 x 6 vol). The batch concentrated under reduced pressure to yield the desired product.

Step 3. 5-Bromo-3-fluoropyridine-2-carbonitrile.

Sulfuric acid (0.02 eq) was added to a solution of tetrabutylammonium fluoride (3 eq) in DMF (5 vol) and the mixture cooled to-40 °C. A solution of the compound of Step 2 (1 eq) in DMF (2 vol) was added maintaining the temperature <-35 °C. After about 20 minutes 2N HCl (3 vol) was added followed by 1N HC1 (15 vol). The precipitated product was collected by filtration to give the desired product.

Step 4. N-f l- (5-Bromo-3-fluoropyridin-2-yl) vinyllacetamide.

Compound of Step 3 (1 eq) was dissolved in toluene (10 vol). The batch was cooled to- 10 °C and MeMgCI (1.5 eq) was added maintaining the temperature < 0 °C. The mixture was aged for 1 h and acetic anhydride was added over approximately 30 min maintaining the temperature <0 °C. The reaction was aged for 18 h at-10 °C. The mixture was quenched with half-saturated NaHC03 (6 vol) and aged at 20-25 °C for 30 min. The layers were separated and the organic layer was washed with water (5 vol), 10% aqueous Na2SO4 (2 x 5 vol) and water (2 x 5 vol). The batch was concentrated under vacuum to give the title compound.

Step 5. N-r (lR)-1-(5-Bromo-3-fluoropyridin-2-yl) ethellacetamide In a nitrogen filled glovebox (<10 ppm 02), (S, S, R, R) -Tangphos (1.05 equivalents relative to Rh) was combined with (COD) 2RhBF4 and dissolved in methanol to make a solution that was 0.107M in Rh. The catalyst solution was aged for lh.

In an nitrogen filled glovebox the catalyst solution ( ( (S, S, R, R)- (Tangphos) Rh (COD) BF4, 0.00284 eq, S/C = 352) was transferred to a stainless steel cylinder along with methanol rinse (1 volume).

To a separate stainless steel cylinder an additional charge of methanol (1 volume) was added. These two cylinders were connected via a ball-valve. The enamide of Step 4 (54 wt% in MeOH) was drawn into a stirred autoclave via vacuum followed by a methanol (10 mL/g enamide) rinse. The solution was then degassed with nitrogen (3 X). The stainless steel vessels containing the catalyst solution were connected to the autoclave via flexible tubing. The autoclave was placed under partial vacuum and the catalyst

solution was drawn into the autoclave followed by the MeOH rinse. The solution was degassed with H2 (100 psig) 3X and the final pressure adjusted to 20 psig. The reaction temperature was set to 25 °C and agitation initiated. The reaction pressure was increased to 98 psig after 20 minutes. The mixture was hydrogenated for an additional 4h. Enantiomeric excess was 99.5%.

The batch was removed from the autoclave and concentrated under vacuum and solvent switched to isopropyl acetate (IPAc) to a final concentration of 10 mL/g. The IPAc solution was filtered through silica gel (300 wt%), and washed with 1 volume of IPAc. Darko KB-B (50 wt%) was added and the mixture aged for 16 h at 20-25 °C. The batch was filtered through Solka Floc and the cake washed with IPAc (1.1 volumes). The batch was concentrated under vacuum to give the title compound.

V. 3- (2-bromo-4-chloro-6-fluorophenyl)-5-methyl-1, 2, 4-oxadiazole.

A solution of 2-bromo-4-chloro-6-fluoroaniline (25.0 g, 111 mmol) in 200 mL of anhydrous DCM, in a 2 liter round-bottom flask equipped with a bubbler, was treated with nitrosonium tetrafluoroborate (14.3 g, 123 mmol), at ambient temperature. After 1 hour complete consumption of the aniline was observed. The reaction mixture was cooled to 0 °C prior to the addition of potassium cyanide (14.5 g, 223 mmol). With rapid stirring, an aqueous solution of cupric sulfate hexahydrate (55.6 g, 223 mmol in 125 mL of water) was slowly added producing a large volume of gas evolution. After stirring for 40 minutes, the ice bath was removed and the reaction was allowed to warm to ambient temperature. After 1 hour at ambient temperature, the reaction mixture was diluted with additional DCM and then slowly quenched by the addition of aqueous saturated sodium bicarbonate until additional gas evolution is no longer observed. The resulting heterogeneous mixture was then filtered through a large pad of celite, washing with DCM as needed. The organic filtrate was then washed twice with saturated brine prior to drying over sodium sulfate. Filtration and solvent removal provided material which was subjected to silica gel chromatography eluting with 0-25% DCM in heptane. Collection of product containing fractions and removal of solvent yielded 9.76 grams of 2-bromo-4-chloro-6-fluorobenzo- nitrile, which gave LC/MS and proton NMR spectra consistent with theory.

To 21 mL of ethanol was added the above nitrile (5.00 g, 21.3 mmol) followed by excess hydroxylamine (21 mL of a 50% aqueous hydroxylamine solution). This mixture was then headed to 60 °C while open to the atmosphere. After 1.5 hours the reaction was judged complete by LC/MS analysis.

The mixture was allowed to cool to ambient temperature before being diluted with diethyl ether and water. The ethereal layer was washed with additional water, then twice with saturated brine. The organic layer was dried over sodium sulfate, filtered and then concentrated to obtain 5.5 g of unpurified hydroxylamine adduct. This material was combined with an equivalent quantity of unpurified hydroxyl- amine adduct prepared in parallel to give a total mass of ~ 11. 1 grams. This 11.1 grams of material was

dissolved in DMADMA (38.7 g, 290. mmol) and allowed to stir at ambient temperature for 30 minutes.

The reaction mixture was then diluted with diethyl ether and washed with water, half-saturated brine and then brine prior to drying over sodium sulfate. Filtration and solvent removal provided material which was subjected to silica gel chromatography eluting with 40-85% DCM in hexanes. Collection of product containing fractions and removal of solvent yielded 5.08 grams of the title compound, which gave LC/MS and proton NMR spectra consistent with theory.

The following examples are provided to illustrate the invention are are not to be construed as limiting the scope of the claims in any manner.

EXAMPLE 1 Methyl 4'-(f [(l-hydroxecyclopropel) carbonYllamino lmethyl) biphenal-2-carboxelate Methyl 4'- (aminomethyl) biphenyl-2-carboxylate (0.325 g, 1.35 mmol), 1-hydroxycyclo- propanecarboxylic acid (0.151 g, 1. 48 mmol) and 1-hydroxybenzotriazole hydrate (0.248 g, 1.62 mmol) were suspended in a mixture of THF/DCM (13 mL of THF/6. 5 mL of DCM), followed by the addition of 1- (3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (0. 336 g, 1.75 mmol). Rapidly the solution turned homogenous, but returned to a heterogeneous mixture after 5 minutes. After 3 hours the solvent mixture was exchanged for pure DCM, prior to purification by silica gel chromatography eluted with 1-5% methanol in DCM to provide the title compound.

LRMS (ES, M+H+): 326. 1H NMR (400 MHz, CDC13): 8 7.84 (dd, J = 8. 0, 1.2 Hz, 1H), 7.53 (td, J = 7.6, 1.6 Hz, 1H), 7.41 (td, J = 7.6, 1.2 Hz, 1H), 7.34 (m, 3H), 7.29 (bd, J = 8.4 Hz, 2H), 7.19 (bs, 1H), 4.55 (d, J = 6.0 Hz, 2H), 3.69 (s, 3H), 2.55 (bs, 1H), 1.43 (m, 2H), 1.07 (m, 2H).

EXAMPLE 2 <BR> <BR> (2R)-N-((1R)-1-{5-r5-chloro-3-fluoro-2-(5-methyl-1, 2, 4-oxadiazol-3-el) phenYll-3-fluoropyridin-2- yl ethyl)-3, 3, 3-trifluoro-2-hydroxv-2-methylpropanamide

A solution of the compound of Reference Example IV (20. 6 g, 79.1 mmol) in 210 mL of anhydrous MeOH was cooled to 0 °C prior to saturation with anhydrous hydrogen chloride. A condenser and oil bubbler were then attached, and the solution was heated to 85 °C for 5 hours. After cooling to ambient temperature, solvent was removed under reduced pressure to obtain a yellow solid. This solid was dissolved in 200 mL of DCM and 210 mL of aqueous sodium hydroxide (1M, 210 mmol). Boc- anhydride (23 g, 105 mmol) was then added and the reaction was allowed to stir for 16 hours. The layers were then separated and the organic layer was extracted once with DCM. The combined organics were washed with saturated brine prior to drying over sodium sulfate. Filtration and solvent removal provided material which was subjected to silica gel chromatography eluting with 5-30% EtOAc in hexanes.

Collection of product containing fractions and removal of solvent yielded 25.1 grams of tert-butyl [ (1R)- 1- (5-bromo-3-fluoropyridin-2-yl) ethyl] carbamate, which gave LC/MS and proton NMR spectra consistent with theory.

A mixture of potassium acetate (3.43 g, 35.0 mmol), the above carbamate (3.72 g, 11.7 mmol) and 4,4, 4', 4', 5,5, 5', 5'-octamethyl-2, 2'-bi-1, 3,2-dioxaborolane (3.85 g, 15.2 mmol) in a sealed tube reaction vessel caped with a rubber septum was dissolved in 24 mL of DMSO. This heterogeneous mixture was then evacuated and purged with nitrogen three times prior to introduction of [l, l'-bis- (diphenylphosphino) ferrocene] dichloro-palladium (II), complex with DCM (1 : 1) (0.426 g, 0.583 mmol).

The heterogeneous mixture was then evacuated and purged with nitrogen twice before the septum was replaced with a teflon screw-top cap. This sealed vessel then heated to 90 °C for 1.5 hours. After cooling to ambient temperature potassium carbonate (3.22 g, 23.3 mmol), water (3 mL), compound of Reference Example V (3.74 g, 12.8 mmol) and additional palladium catalyst (0.426 g, 0.583 mmol) were added and the reaction was sealed again. After heating to 80 °C for 3 hours the reaction was cooled and then quenched with water and EtOAc. This aqueous layer was extracted once with EtOAc. The

combined organic layers were washed with additional water and then brine prior to drying over sodium sulfate. Filtration and solvent removal provided material which was subjected to silica gel chromatography eluting with 10-35% EtOAc in hexanes. Collection of product containing fractions and removal of solvent yielded 3.62 grams of tert-butyl ( (1R)-1- {5- [5-chloro-3-fluoro-2- (5-methyl-1, 2,4- oxadiazol-3-yl) phenyl]-3-fluoropyridin-2-yl} ethyl) carbamate.

The above Boc-protected material was dissolved in 100 mL of MeOH and then cooled to 0 °C. Anhydrous HC1 gas was then bubbled through the solution for 3 minutes. After allowing this reaction mixture to stand at 0 °C for 30 minutes solvent was removed. Additional MeOH was added and then removed under vacuum, followed by addition and removal of DCM (twice) to provide 3.10 g of the amine hydrochloride as a foaming solid.

To the above amine hydrochloride (4.54 g, 11.7 mmol) in a 200 mL round-bottom flask was added anhydrous DCM (31 mL), (2R)-3, 3,3-trifluoro-2-hydroxy-2-methylpropanoic acid (2.78 g, 17.6 mmol), HOBt*H2O (0.898 g, 5.86 mmol), triethylamine (2.97 g, 29.3 mmol) and lastly EDCI (3.60 g, 18.8 mmol). This reaction mixture was allowed to stir at ambient temperature for 16 hours. After the reaction was judged complete by LC/MS analysis, the reaction mixture (without work-up or solvent removal) was subjected to silica gel chromatography eluting with 20-60% EtOAc in hexanes. Collection of product containing fractions and removal of solvent yielded 3.61 grams of the title compound as a foaming solid. Purity was determined by LCMS (ES MS, M + H+ found: 491) and proton NMR (400 MHz, CD30D: 8 8.208, 8.204, 7.569, 7.564, 7.558, 7. 546, 7.541, 7. 537,7. 533,7. 518,7. 515,7. 513, 7.510, 5.381, 5. 366, 5. 363,5. 349,5. 346, 5.331, 2. 546, 1. 576, 1. 468, 1.451).

The following compounds were prepared by following the general procedures provided in Examples 1 and 2. The a-hydroxycarboxylic acid starting materials are either commercially available of may be prepared from commercially available reagents using conventional reactions well known in the art. The biarylmethylamine starting materials may be prepared by the procedures provided in the Reference Examples section and in the general reaction schemes, using reagents that are either commercially available or that may be prepared from commercially available reagents using conventional reactions well known in the art.

TABLE 1 Ex. R2a R2b * LRMS (M+H+) 3 CH3 CH3 - 364 4 H H - 336 5 CH2CH3 CH2CH3 - 392 6 3- (OCH3)-Ph H 442 CHCH3 CH3 ~ 378 8 CH (CH3) 2 H ~ 378 9 CH3 H S 350 10 Ph cyclopentyl 480 11 4-Br-Ph H 490 12 4- (CF3)-Ph H 480 13 Ph H S 412 14 4- (OCH3)-Ph H 442 15 CH2Ph H 426 16 cyclohexyl H R 418 17 3-(OH)-4-(OCH3)-Ph H ~ 458 18 3, 4-diOH-Ph H ~ 444 19 CF3 CF3 - 472 20 CH2CH (CH3) 2 H ~ 392 21 Ph CH3 ~ 426 22 (1H-indol-3-yl)methyl H ~ 465 23 CH2 (CH2) 4CH3 H 420 24 3, 5-diF-Ph H ~ 448 25 CF3 H ~ 404 CF3 CH3 S 418 Ex. R2a R2b * LRMS (M+H+) 27 CF3 CH3 R 418 28 CH3 H R 350 29 (CH2) 3CH3 H ~ 392 30 cyclohexyl H S 418 31 CH2CH (CH3) 2 H S 392 32 CH (CH3) 2 H R 378 33 CH (CH3) 2 H S 378 34 CH (CH3) CH2CH3 H S 392 35 C (CH3)3 H S 392 36 CF3 H S 404 CH2CH2SCH3 H ~ 410 38 CH2CH (CH3) 21 H R 392 39 (pyridin-3-yl) methyl H 427 40 OH CF3 - 420 41 CF2CF3 CH3 - 468 42 CF2HCH3 400 TABLE 2

LRMS Ex. m R3 Y 1 R6 R7 R8 R9 LRMS (M+H+) 43 1 H CH-C02CH3 F F H 362 44 2 H CH-CO2CH3 F F H 376 45 1 CH3 N R C02CH3 F Cl Cl 427 46 5 H CH-C02CH3 F F H 418 Ex. m R3a Y R6 R7 R8 R9 LRMS (M+H+) 47 1 CH3 NR OCH2CH3P Cl Cl 413 48 1 CH3 CH R C02CH3 F Cl H 392 49 1 CH3 CH R C02CH3 H F H 358 50 1 CH3 CH S C02CH3 F F H 376 51 1 CH3 N R OCH2CHF2 F Cl Cl 449 52 1 CH3 CH R C02CH3 Cl Cl H 408 53 1 CH3 CH R OCH2CHF2 F Cl Cl 448 54 1 CH3 CH R 2-CH3-2H-tetrazol-5-yl F F H 400 55 1 CH3 CH R CO2CH3 P C1 C1 426 56 2 CH3 N R C02CH3F Cl Cl 441 57 1 CH3 N R 2-CH3-2H-tetrazol-5-yl F F H 401 TABLE 3

TUA/TQ Example Y * P R7 R8 R9 (M+H+) 58 CH R C02CH3 F Cl H 448 59 N R C02CH3 F F Cl 467 60 N R 2-CH3-2H-tetrazol-5-yl F F g 4S7 61 N R C02CH2CH3 F F Cl 481 62 N R C02CH3 F Cl ci 483 63 N R OCH2CHF2 F ci Cl 505 64 N R OCH2CH3 F Cl Cl 469 65 CH R 2-CH3-2H-tetrazol-5-yl F F H 456 66 CH Ph F H H 432 < o n LRMS Example Y * P"R7 R8 R9 (M+H+) 67 CH RC02CH3F Cl Cl 482 68 N R CN F Cl Cl 450 70 CH R OCH2CF3 F F ci 504 71 CH R 3-CH3-1, 2, 4-oxadiazole F H Cl 470 72 N R OCH2CF3 F F ci 505 73 CH R OCH2CHF2 F Cl ci 504 74 CH R 2-CH3-2H-tetrazol-5-yl F H F 456 75 CH R CF3 F H H 424 76 CH R 5-CH3-1, 2, 4-oxadiazole F H F 456 77 N R OCH2CF3 Cl Cl Cl 539 78 N R 2-CH3-2H-tetrazol-5-yl Cl F H 471 79 N R OCH (CH3) 2 F Cl ci 483 80 N R 2-CH3-2H-tetrazol-5-yl F Cl Cl 507 81 CH R 3-CH3-1, 2, 4-oxadiazole F F H 456 82 CH R 2-CH3-2H-tetrazol-5-yl F Cl CI 506 83 CH R OCH2CP3 P C1 C1 522 84 CH R OCH (CH3) 2 F Cl ci 482 85 CH R C02CH3 F F H 432 86 N R CONH2 F Cl C1 468 87 N R SCH2CH3 F Cl H 449 88 N R OCH2CP3 F C1 C1 523 89 N R 5-CH3-1, 2, 4-oxadiazole F Cl Cl 507 90 N R C02CH3 Cl F H 447 91 CH R 2-CH3-2H-tetrazol-5-yl F Cl H 470 92 CH R OCH (CH2CH3) 2 F Cl 510 93 CH R OCH2CH2CH3 F CI CI 482 94 CH R OCH2CHF2 F F F 472 95 CH R CO2CH2CH3 F Cl H 460 96 N R 2-CH3-2H-tetrazol-5-yl Cl CI H 489 97 N R OCH2CHF2 F F Cl 487 98 N R CO2CH3 Cl Cl Cl 499 Example Y _ R7 R8 R9 LRMS (M+H+) 99 N R OCH2CH (CH3) 2 F Cl Cl 497 100 N R C02CH2CH3 F Cl H 461 101 N R C02CH2CH3 102 N R C02CH3 F Cl H 447 103 N R 2-CH3-2H-tetrazol-5-yl F Cl H 471 104 N R S (O) CH2CH3 F Cl H 465 105 N R C02CH2CH3 ci ci H 479 106 CH R C02CH3 H F H 414 107 N R 5-CH3-1, 2, 4-oxadiazole F F H 457 108 CH R C02CH2CH3 F Cl Cl 496 109 N R 2-CH3-2H-tetrazol-5-yl Cl Cl Cl 521 110 N R 5-CH3-1, 2, 4-oxadiazole ci Cl Cl 521 111 N R 2-CH3-2H-tetrazol-5-yl F F CI 489 112 N R 3-CH3-1, 2, 4-oxadiazole F F H 457 113 N R 5-CH3-1, 2, 4-oxadiazole F Cl H 471 114 N R 3-CH3-1, 2, 4-oxadiazole F F ci 489 115 CH R 2-CH3-2H-tetrazol-5-yl F F Cl 488 116 N R 3-CH3-1, 2, 4-oxadiazole Cl F H 471 117 N R 3-CH3-1, 2, 4-oxadiazole Cl F Cl 507 118 N R 5-CH3-1, 2, 4-oxadiazole cl Cl H 489 119 CH R 5-CH3-1, 2, 4-oxadiazole F F ci 488 120 N R 5-CH3-1, 2, 4-oxadiazole Cl F H 471 121 N R 2-CH3-2H-tetrazol-5-yl Cl F Cl 507 122 CH RC02CH3Cl Cl H 464 123 N R 3-CH3-1, 2, 4-oxadiazole F Cl H 471 124 N R 5-CH3-1, 2, 4-oxadiazole F Cl F 489 125 N R 2-CH3-2H-tetrazol-5-yl F Cl F 489 126 N R C02CH2CH3 cl ci Cl 511 19 127 N R 5-CH3-1, 2, 4-oxadiazole C1 F C1 507 128 N R 3-CH3-1, 2, 4-oxadiazole F Cl Cl 507 129 CH R 5-CH3-1, 2, 4-oxadiazole F F Me 470 r o n LRMS Example Y _ R R8 R9 LRMS (M+H+) 130 CH R l-CH3-2H-tetrazol-5-yl F F Cl 488 131 CH R 2-CH3-2H-tetrazol-5-yl Cl F Cl 506 132 N R 3-CH3-1, 2, 4-oxadiazole Cl Cl Cl 521 133 N R 2-CH3-2H-tetrazol-5-yl F F F 475 134 N RC02CH3H Cl Cl 465 135 CH R 2-CH3-2H-tetrazol-5-yl F F Me 470 136 N R 5-CH3-1, 2, 4-oxadiazole Cl Cl Me 503 137 N R 2-CH3-2H-tetrazol-5-yl Cl Cl Me 503 138 CH R OCH2CH2CF3 F Cl ci 536 139 CH R 5-CH3-1, 2, 4-oxadiazole F Cl CF3 538 140 CH R 5-CH3-1, 2, 4-oxadiazole F H H 438 141 CH R 2-CH3-2H-tetrazol-5-yl F Cl CF3 538 142 N R 5-CH3-1, 2, 4-oxadiazole F F F 475 143 N R 5-CH3-1, 2, 4-oxadiazole Cl F F 489 144 N R 2-CH3-2H-tetrazol-5-yl Cl F F 489 145 N R S02CH2CH3 F Cl H 481 5-CH (CH3) 2-1, 2, 4- 146 N R oxadiazole F Cl Cl 534 147 CH R NH (CH2CF3) F Cl Cl 503 148 CH R 2-CH3-2H-tetrazol-5-yl F Cl OCF3 554 149 N R 3-CH3-1, 2, 4-oxadiazole F Cl F 489 150 N R 3-CH3-1, 2, 4-oxadiazole Cl F F 489 151 N R 3-CH3-1, 2, 4-oxadiazole F F F 475 152 N R 5-CH3-1, 2, 4-oxadiazole F F F 451 153 CH R 5-CH3-1, 2, 4-oxadiazole F Cl SOzMe 548 154 CH R cyclopropyl F Cl ci 464 155 CH R 5-pyrimidinyl F Cl ci 502 156 CH R 2-CH3-2H-tetrazol-5-yl F Cl C02CH3 528 TABLE 4 LRMS Example Y I-R6 R6 R7 R8 LRMS (M+H+) 157 CH R CONHCH3 F F H 417 158 N R C02CH3 Cl F H 433 159 CH R 2-CH3-2H-tetrazol-5-yl Cl F ci 492 160 N R 2-CH3-2H-tetrazol-5-yl F Cl Cl 493 161 N R 2-CH3-2H-tetrazol-5-yl F F Cl 475 162 N R 2-CH3-2H-tetrazol-5-yl Cl F Cl 493 163 CH R 5-CH3-1, 2, 4-oxadiazole Cl F Cl 492 164 N R 5-CH3-1, 2, 4-oxadiazole Cl F C1 493 165 N R 2-CH3-2H-tetrazol-5-yl Cl Cl Cl 507 166 N R 3-CH3-1, 2, 4-oxadiazole Cl F Cl 493 167 N R 2-CH3-2H-tetrazol-5-yl Cl F H 457 168 N R 2-CH3-2H-tetrazol-5-yl F F F 461 169 N R 5-CH3-1, 2, 4-oxadiazole F F Cl 475 170 N R 2-CH3-2H-tetrazol-5-yl F F H 443 171 CH RC02CH3Cl Cl Cl 482 172 N R 2-CH3-2H-tetrazol-5-yl Cl F F 475 173 N R 2-CH3-2H-tetrazol-5-yl Cl Cl H 475 174 N R 3-CH3-1, 2, 4-oxadiazole ci Cl ci 507 175 N R 5-CH3-1, 2, 4-oxadiazole Cl F F 475 176 N R 3-CH3-1, 2, 4-oxadiazole Cl F F 475 177 N R 2-CH3-2H-tetrazol-5-yl Cl Cl F 493 178 N R 3-CH3-1, 2, 4-oxadiazole Cl Cl F 493 179 N R CO2CH3 Cl F ci 468 6 R Rg R9 LRMS Example Y (M+H+) 180 N R 5-CH3-1, 2, 4-oxadiazole Cl Cl Cl 507 181 N R 5-CH3-1, 2, 4-oxadiazole F Cl Cl 493 182 N R 3-CH3-1, 2, 4-oxadiazole Cl Cl H 475 183 N R 5-CH3-1, 2, 4-oxadiazole Cl Cl F 493 184 N R 3-CH3-1, 2, 4-oxadiazole F F Cl 476