AU2015203286A1 - C7-fluoro substituted tetracycline compounds - Google Patents

C7-fluoro substituted tetracycline compounds Download PDF

Info

Publication number
AU2015203286A1
AU2015203286A1 AU2015203286A AU2015203286A AU2015203286A1 AU 2015203286 A1 AU2015203286 A1 AU 2015203286A1 AU 2015203286 A AU2015203286 A AU 2015203286A AU 2015203286 A AU2015203286 A AU 2015203286A AU 2015203286 A1 AU2015203286 A1 AU 2015203286A1
Authority
AU
Australia
Prior art keywords
alkyl
compound
hydrogen
alkoxy
halo
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2015203286A
Other versions
AU2015203286B2 (en
Inventor
Roger B. Clark
Diana Katharine Hunt
Louis Plamondon
Xiao-Yi Xiao
Robert P. Zahler
Jingye Zhou
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Tetraphase Pharmaceuticals Inc
Original Assignee
Tetraphase Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Tetraphase Pharmaceuticals Inc filed Critical Tetraphase Pharmaceuticals Inc
Priority to AU2015203286A priority Critical patent/AU2015203286B2/en
Publication of AU2015203286A1 publication Critical patent/AU2015203286A1/en
Priority to AU2016259418A priority patent/AU2016259418A1/en
Application granted granted Critical
Publication of AU2015203286B2 publication Critical patent/AU2015203286B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Abstract

The present invention is directed to a compound represented by Structural Formula (A): or a pharmaceutically acceptable salt thereof. The variables for Structural Formula (A) are defined herein. Also described is a pharmaceutical composition comprising the compound of Structural Formula (A) and its therapeutic use.

Description

Australian Patents Act 1990 - Regulation 3.2A ORIGINAL COMPLETE SPECIFICATION STANDARD PATENT Invention Title C7-fluoro substituted tetracycline compounds The following statement is a full description of this invention, including the best method of performing it known to us: C:\NRPortbl\DCC\ACG\4895878_1.DOC - 31/1/13 C:\Users\ACG\AppData\Local\Temp\e05d3ca39224a6b956156adOI99fe45.7920109_1_2CFBICF.doc-6/15/2015 - 1 C7-FLUORO SUBSTITUTED TETRACYCLINE COMPOUNDS RELATED APPLICATION 5 This is a divisional of Australian Patent Application Nos. 2009279473 and 2013500565, the entire contents of which are incorporated herein by reference. BACKGROUND OF THE INVENTION The tetracyclines are broad spectrum anti-microbial agents that are widely used in 10 human and veterinary medicine. The total production of tetracyclines by fermentation or semi-synthesis is measured in the thousands of metric tons per year. The widespread use of tetracyclines for therapeutic purposes has led to the emergence of resistance to these antibiotics, even among highly susceptible bacterial species. Thefore, there is need for new tetracycline analogs with improved antibacterial 15 activities and efficacies against other tetracycline responsice diseases or disorders. SUMMARYT OF THE INVENTION The present invention is directed to a compound represented by Structural Formula (A): 20 or a pharmaceutically acceptable salt thereof, wherein: X is selected from hydrogen, -(C 1
-C
7 )alkyl, carbocyclyl, aryl and heteroaryl; Y is selected from hydrogen, -(C1-C 7 )alkyl, carbocyclyl, -(C1-C4)alkylene-N(R A)(R B), 25 -(C1-C4)alkylene-N(R F)C(O)-[C(RD)(RE)]o_4-N(RA)(R B), -CH=N-OR A, -N(R A)(R B), -N(RF)-C(O)-(C C6r)alkvl, -N(RF)-C(O)-heterocyclyl, -N(RF)-C(O)-heteroarvl,
-N(R
1 )-C(Oi-carboeyclyt1 -N(RF)-C(O)-aivl, -N(Rr)- S(0) 1
,(C
1 C4)alkylenec-N (iR')(R), 5 -N(I1)-S(O)m-(C ( C4)ilkylene-carbocyclyl, and -N(RF-SOm-(C- 4kenc-ar at least one of X and Y is not hydrogen; each R and R" are independently sele'ted firo hydrogen, (C-C 7 )alkyl, .- O-( C-C)alkyi, -(C-C 6 )aikyiene-carbocycl, -(CO-C 6 )alkvene-aryl,
-(C
0
-C
6 )'alkyl ene-heterocyclyl, -(C,-()alkylene-heteroaryl, 1 0 -( C;C,)alkylene-O-carbocyciyl, -(Cr-C 0 )aikylene-O-aryl, -(C-Cr)al kylene-0-heterocyclyl, -(C -C 6 )alkylene-0-heteroaryl, -S (Oj~(C -C )alkyl, -(C,C4)alkylene-S( 0 -carbocyclyl, -,(CjC4alkleneS()naryl, -(CrC4~)alkylene-S(O)m,-heteracy cly! and -(C C)alkvlene-S(O)-heteroaryl; or 15 R A and .RC taken together with the nitrogen atom to which they are bound form a lieterocyclyl or heteroaryl, wherein the heterocycle or heteroaryl optionally comprises I to 4 additional heteroatons independently selected from N, S and 0; each R 0 and each R)> is independently selected from hydrogen, (Cr -Cj)alkvl carbocyclyl, aryl, heterocyclyl, heteroaryl, or a naturally occurrmgT] amino acid side 20 chain moiety, or
.R
0 and Re taken together with the carbon atom to which they are bound form a 3-7 meibered carbocyclyl, or a 4-7 membered heterocyclyl, wherein the hetcrocyclyl formed by R1 and R. optionally comprises one to two additional heteroatonis independently selected from N, S and 0; 25 R is selected from hy drogen, (Ci-Ci)alkyl, carbocyclyl, aryl and helcroaryl, and m is I or 2, wherein: each carbocyclyl, aryl, heterocyclyl or heteroayl is optionally and independently substituted with one or more substituents independently selected from 30 halo, -(CrC4)alkyl, -OH1, =0, -0-(C C4I)alkyl, -(C,-C4)alkyvl-O-(CI-C4.)a'kyl, halo-substituted -(C-C4)alkyl, halo-substituted -O-(C -C4)al kv, -C(O)-(C -C 4 )alkyl, -C(0)-(fluoro-substituted-(C -C4)alkyl), -S(O)m-(, -C4.)alkvl, -N(R)(R'), and CN; 3 each alkyl in the group represented by R^, R", R 1 and RF is optionally and independently substituted with one or more substituents independenfly selected from halo, -(C 1
-C
4 )alkyl, -OH, -O-(C-Cj)alkyl, -(C-C4)alkyl-O-(CC4)akyl, Loro-sutbstituted-(C-C4)alkyl, -S(O), 1 -(C-sC)alky], and -N(R)(R ), whereil 5 each R is hydrogen or (C-C4)alkyl, wherein each alkyI in the group represented by R is optionally and independently substituted with one oir more substituents in dependently selected from -(CI -C4)alkyl, (Cr 3 -C()cycloal kyl, halo, -OH, -O-(C -C 4 )aikvl, and (CI -C)alkyl-O-(C - C 4 )alkyl. Another embodiment of the present invention is directed to a compound. 10 represented by Structural Formula (II) F X H3C, ,N'CH3 H HO R1 OH Kr NH 2
R
2 N 6 - OH 0 HOpHO 0 or a pharmaceutically acceptable salt thereof, wherein: R' and R are each independently selected roin hydrogen. (CI-C 7 )alkyl, (CrCs)cycloalkyl(C-C)alkyl, (Ci-C7)alkoxy(C-C4)alkyl, 15 (C!-C)cycloatkoxy(C-C4)alkyl, (C-C 6 )cycloaIkyl, aryl, aryl(Cj-C 4 )alkyl, aryloxy(C-C4)alkyl, arvlthio(C,-C,)alkyl, arylsufi nyi(Ci-C1)alkyI, aryisulfoiyl(C -C4)alkyl, and -O-(C-C7)dkvl, or R' and R2 taken together with the nitrogen atom to which they are bonded for m a tonocyclic or bicyclic heteroaryl, or a monocyclic, fused bicyclic, bridged 20 bicyclic or spiro bicyclic leterocycle, wherein the heteroaryl or heterocycle optionally contains one or two additional heteroatoms independently selected from N, 0 and S; and wherein each alkvl, cycloalkyl, alkoxy and cycloalkoxy moiety in the groups represented byR anrd R and each beterocvcle represented by NR R taken together 25 is optionally substituted with one or more substituents independently selected from the group consisting of (C-C4)alky l, halo, -01, (C-C4al koxy, (Ci-C4)alkyIthia, (C-C)alkyIsulfmnyi, (C-C 4 ) alkylsulfonyl, (C-C 4 )alkoxy(C -C 4 ) alkyI, and -N(R )(R 1 ); and -4 each aryi, aryloxy, arylthio, arylsufinyl and arysulfonyl moiety in the groups represented by R' and R2 and each heteroarvl represented by NR K2 taken together is optionally substituted with one or more substituents independently selected from the group consisting of (Ci-C4)akvl, halo, -OH1, (C-C4)alkoxy, -S-(C:-C4)aikvl, 5 -S(O)(C 1 -C4)alkyl, -S(O)(C- Cpialkyl, (C -C.
4 )alkoxy(C 1 -C)g lky -N(R)(R4); -CN. halo(C 1 -C.)alkyl, and halo(C -C)alkoxy, and
R
3 and R 4 are each independently selected from the group consisting of -H and (C 1
-C
4 )alkyl, wh erein the (C-C4)alkvl represented by R3 and R' is optionally substituted with one or more substituents independently selected from the group 10 consiting of (C C4)ailkvI, halo, -OH, (C-C 4 )alkoxy, an d
(C
4 -C4)alkoxy(CC4)alkyLI Values for X and R' are as described above for Structural Formula A A another nbodiment of the present inivention is directed to a compound represented by Structural formulaa (I): H3CN
CH
3 F N H H
R
1 ~ OH H NH 15 OH 0 HO 0 0 () or a pharmaceutically acceptable salt thereof, wherein: R' and R 2 are each independenrly selected from hydrogen, (C 1 C-)aikyl, (Ci)cycloa[lkyl (C 1 C4)alkyl, (C 1 -C7)aikoxy(CI-C4)aikyl. (CrC,)cycloakoxy(C-Csjalkyl, (Q-C()cycloalkyl, aryl, aryl(C-C 4 )alkyl, 20 aryloxy(Ci C4)aikyl, arylthio(CvC4)alkyI, arylsulfinyl(C-C 4 )alkyl, and arvlsulfonyl(C-C4)alkyl, or R' and R2 taken together with the nitrogen atom to which they are bonded forr a monocyclic or bicyclic heteroaryl, or a monocyclic, fused bicyclic, bridged bicyclic or spiro bicyclic heterocycle, wherein the heteroaryl or heterocycle 25 optionally contains one additional heteroatom independently selected from N, 0 and S; and the remainder of the variables are as described above for Structural Formula (11).
Another embodiment of the present invention is directed to a pharmaceutical composition comprising a pharmaceuticals acceptable carrier or diluent and a compound represented by Structural Formul a (A), (II) or (I) or a pharmaceuticalV acceptable salt thereof The pharmaceutical composition is used in therapy, such as § treating an infection in a subject. Another embodiment of the present invention is a method of treating an intfetion in a subject comprising administering to the subject an effective amount of a compound represented by Structural Formula (A), (Ii) or (T) or a pharmaceutically acceptable salt thereof 10 Another embodiment of the present invention is a method of preventing an infection in a subject comprising administering to the subject an effective amount of a compound represented by Structural Formula (A), (IT) or (1) or a pharmaceutically acceptable salt thereof Another embodiment of the present invention is the use of a compound 15 represented by Structural Formula (A), (TI) o r (1) or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating an infection in a subject, Another embodiment of the present invention s the use of a compound represented by Structural Formula (A), (11) or (I) or a pharmaceuticady acceptable 20 salt thereof for the manufacture of a medicament for preventing an infection in a subject. Another embodiment of the present invention is the use of a compound represented by Structural Formula (A), (1I) or (1) or a pharnaceutically acceptable salt thereof for therapy, such as treating or preventing an infection in a subject. 25 TFTAILED DESCRIPTION OF THE INVENTION The present invention is directed to a compound represented by Structural Formula (A) or a pharmaceutically acceptable salt thereof Values and alternative va] lies for the variables in Structural Formula (A) are defined as the following. X is selected from hydrogen, -(CC7)al.kyl, carbocyclyl, aryl and heteroaryl. 30 In oie embodiment, X is hydrogen, In an alternative embodiment, X is -(C I C2)alkyl. Alternatively, X is -(CrC4)alkyl in another embodiment, X is -6 carbocyclyl. In another alternative embodiment, X is ary or heteroayl. In another alternative embodiment, X is phenyl. Y is selected from hydrogen, -(CuC )alkyi, carbocyclyl, -(C] C4)alkylene-N(RAR)(e), 5 -(C,-C 4 )alkyiene-N4(RF)-C(O)-[C(R )(R)]wN(R^)(R), -CH=N -OR^, -N( )(R ) -NR--(4-C(-(R) N(R^61)(R"11), -N(R F).-C(0)-N(R")(RB) -N(RF )-C(O)(C C )alkyl, -N(RF)-C(O)-hcerocyclyl, -N(RF -N(R)-C(O)-carbocycly1, -N(R' )-C(O)-aryl, -N(R A)-S(O> (C C 4 ) rlkvlene-MR 5 )() 10 -N(R')-S(O', -(C, -C 4 )alkvlene-carbocyclyl, and N(R )-S(O),(C-C4aikylene-aryl, provided at ast one of X and Y is not hydrogen. In one embodiment, Y is selected from hy drogen, -(C -C 7 )alkyl, -(C] -C4)alkylene-N (R)R ) -(C C jaIky ene-N(RF -( F (0, -CH N-OR^ -N(R(R). -N(R)-C(O)-[C(R))(RI)]1 4 N(R^)(RB), -N(RF)-C(O)-N(R)(R 5 ), 5 -N(F-C(O)-(C 1 CQ)alkyi, -N (R')-C(O)-heterocyclyL -N(Rt)-C(O)-heteroaryi. -N(R)-C(0)-carbocyclyl, -N(RF)_C (0 -N(R')- S(O )r (C - C 4 )aikylonere-N(tR^)(R'E), -N(R%)-S(O)ir(C CC4)alkyl ene-carbocyclyi. and -N(R')-S(O) 1 w(C 1 -47akyleue-aryi provided at least one of X ard Y is not hydrogen.. In one emnbodi.mntnl, Y is selected 20 from hydrogen, -(C iC7)alky, -(C-CA)a1kylene-N(RA)(Ri), -(C-C)alkyiene-N(RF)-C(O) -[C(R )(RE) ]wN(R^)R), -CH-N-ORA -N(R^)(R), -N(R )-C(O)-N(R^)(R)), -N(R F)-C(O)-(Ce-Cj)alky1, -N(RF)-C(O)-heterocyclyl, -N(R$)-C(O)-heteroaiy], -N(R")-C(O)-carbocyclyI, -N(RF)-C(O)-aryl 25 -N(1<)-S(O r(C-C 4 )alkylene-N(RA(Rb) -- N(R)-S(O) -(C,-C 4 )alkylene-carbocyclyl. and -N(R )-S(O)m,-(C-C4)a1kylene-ary!. In another embodiment, Y is selected from hydrogen, -(C -Cr)alkyl, -(C, C4)a1kylene-N)(AXR!I), -(CrC 4 )alevene-N-C(O)-(CH.IwN(R^)R 5 ), -N(RU)(Ra), -NH-C(O)-carbocyclyl, -NH-C(O)-arl, -NH-C(O}-heterocyclyl, 30 -NH-C(O)-heteroaryl, -N-H-C(O)-N(R^:) ^ o -CO-C.N(^(R -NH-C(O)-C(R')(Ri)-N(R)(R") and -NH-S(O)n(C-C4)alkylene-N(R )(RE). Ahernatively, the -(C 1
-C
7 )alkvl represented by Y described above is a -(Ci-C4)alkyl. In yet another embodiment, Y is selected from -N(R)(R), -7 -N(T-)-C(O)-carbocy clyL, -N(L)-C(O)-mryl, -N(H) -C(O)-heterocyclet, and -N(1)-C(0)-hcteroaryl. Alternatively, Y is -NT-C(O)-CH7 -N (R)(R). More specifically, R and R" in -NH-C(0)-CIrN(Rt(RE) are R[ and R), respectively. Each R A' and R are independently selected from hy drogen, (C 1 -Cy)alkvl, 5 -0-(,-C)alkyl, -(CC )alkylene-carbocyclyl, -(Cr-C)alkylene-aryI, -(Cr Calkvlene- hetero cvciyl, -(Cc-C)alkylene-heteroaryl, -(C Cjdlklcne-0-carbocyclyl, -(C1G )alkylene-O-anl, (C C )alkylene-O0-heterocyclyl, -(C 1 C)al kylen 2-O-heteroary] -S(O)j (tiC salkyl, -(Cg-C4)alkylene-S(O0m-carbocyclyl, 10 -(C 0
-C
4 )alkylene-S(O)-aryl, -(C-C4)alkyleae- S(O-heterocyclyi and - (C 0
-C
4 )alkylin e- S(O)", -heteroaryl, or R^ an d R taken together with the nitrogen atom to which they are bound forn a heterocyclyl or heteroaryl, wherein the heterocycle or heteroaryl optionally comprises I to 4 additional heteroatoms independently selected from N, S and 0. In 1 5 one embodiment, each R^ is independently selected from hydrogen and methyl; Rt is selec ted from hydrogen, (C-C-,)alkyl, -(C)alkylene-carbocyclyl, -(C(k-C) alkykene-amy], -(C 1 C s)alkylene-lheterioyl, - S(0)ri(C -Co)alkyl, (JC-Cjalkylene S- )rcarbocyclyl -(Co-C4)alklene-S(0)m-aryl, -((C.)alkycvno-S(0) heterocycle and -(C :rC4[)alkylene-S(O)r-heteroaryl; or R^ 20 and R' taken together with the nitrogen atom to form a ieterocycle, wherein the heterocycle is optionally substituted with =0 and -N'R")(R in another embodiment R^ is hydrogen; and Rn is selected from (C-C 1 )a. kyl, and -S(O)-CI or R^ and RH taken together to form 4-7 membered heterocyclic ring. Each R) and each RE is independently selected from hydrogen, (C -C 0 )alkyl, 25 carbocyclyl, aryl, heterocycly1, heteroaryl, or a naturally occurring amino acid si de chain moiety, or R) and R' taken together with the carbon atom to which they are bound form a 3-7 membered carbocyclyl, or a 4-7 membered heterocyclyl, wherein the heterocyclyl formed by R- and RE optionally comprises one to two additional liereroatoms mdependentiy sciected from N, S and 0 In one embodiment, RD and 30 R" are both -R RF is selected from hydrogen (C -C7)alkyl, carbocycll, ary] and heteroaryl. In one embodniet, R is hydrogen. In another embodiment, k is selected from hydrogen, (Cj-CK)alkyl, ary] and heteroaryl. In another embodiment, R' is selected -8 from hydrogen. (Ci-C)alkyl and phenyl. In another embodiment, R> is selected from hydrogen (C 1 -C4)alkyl and phenyl Rl' is selected from (C-C)alkyl, carbocyclyl, aryl, heteroaryl, h eterocyclyl, and a naturally occurring amino acid side chain moiety, or R' and R 2 taken together 5 with the carbon atoni to which they are bound foi rm a 3-7 mieumbered carbocyclyl, or a 4-7 membered heterocyclyl, wherein the heterocyclyl formed by R t ' and RE optionally comprises one to two additional heteroatorns independently selected from N, S and 0 in one enbodinent, R) and RE taken together with the carbon atom to which they are bound form a (C C 7 )cycloalkyl. 10 m is I or 2. In one embodiment, mf is 2. Each carbocyclyl, aryl, heterocyclyl or heteroaryl described above (e.g, in the groups represented by Y, R< RB, R 0 . R and RE) is optionally and independently substituted with one or 111ore substituents independently selected from halo, -('C-C.)alky l -OH, =0, -O-(C 1 -C4)alkyi, -(CI-C4)alkyi-O-(C -Ca)alkyl, 15 halo-substituted -(CrC4alky4 halo-substituted -O-(C-C 4 alkyl -C(0)-(CpC4)alkyl, -C(O)-(fluoro-substituted-(C-C4)alkyl), -S(O)m-(C- C 4 )alkyl, -N(Rt)(R!) and CN. In one embodiment, each carbocyclyl, heterocyclyl or heteroaryl is optionally and independently substituted with one or more substitiuents independently selected from halo, -(C -C4)alIyI, hale-substituted -(C-C,)akv, -0-(C -C4)alkyl, and -N(Rt)(R') 20 In another embodiment, each carbocyclyl aryl, heteroaryl, or heterocycle is optionally and independently substituted with one or more substituents independently selected from -C1 3 , fluoro, and -N(CH3). Each alky] described above (e.g.. in the groups represented by Y, WR, R e , R' R", RE, R' and RF) is optionally and independently substituted with one or more 25 substituents independently selected from halo, -(Ci-C4)alkyl. -OH, -O-(C] -C 7 )a lkyl, -( C 1 -C4)alkv-O-(C-)C4)alkyL tlioro-substituted-(C 1
-C
4 )alkvl, -S(0),-(C-C )alkyl, and -N(R')(R). In one embodiment, each alkyl group (eg, in the group represented by Y or R) is optionally and independently substittMed with one or more substituents independently selected from halo, -OH, and -N( 3 0 Each R(4 is hydrogen or (Cj-Ci)alkyl, wherein each alkyl in the group represented by R is optionally and independently substituted with one or more substituents independently selected from -(C -C4)alky, (C 3 -C)cyclo alky , halo, -OH, -O-(C-C4)alkyL, and (CrC )alkyl-O-(C -C4alkyl. In one embodiment, each -9 allkyl in the group represented by R3 is optionally and independently substiued with
(C
3 -CS)cycloalkyl. As used herein, when RA and R 0 taken together with the nitrogen atom to which they are bound form a a heterocyclyl or heteroaryl, the heterocyclyl or 5 heteroaryl represented by -N RAR 13 can include a. ring system that has a ieteroaton adjacent to the nitrogen atom to which R^ and R" are bound For example, -NRAR can be, but is not limited to, the following ring systems: N N N N \N / Y N N-N/ Similarly, when R and RE or R 2 and RE" taken together with the carbon 10 atom to which they are bound form a heterocyclyl, the heterocyclyl can include a one system that has a heteroatom adjacent to the carbon atom to which R 0 and R or R.) and R' are bound The present invention is directed to 'a compound represented by Structural Formula (1) or (II) or a pharmaceutically acceptable salh thereof. Values and. 15 alternative values for the variables in Structural Formula (1) or (i) are defined as the fbllo wing: RI is selected from hydrogen, (C -C7)alkyl, (C-C)cycloadlkyl(C 1
-C.
4 )alkyl, (CI-C,alkoxy(C -C)alkyl, (C 3 -C)cycloalkoxy(C -C4)alkyl, (C-C)cycloalkyl, aryl, aryl(Cj-C 4 )alkyl, aryloxy(Ci C4)alkyl, arylthio(C-C 4 )alkyl, aryisulfinyl(C 20 C 4 )aikyl, arylsulfonyl(C-C4)alkyl and -O-(C-C7)alkyl Each alkyl, cycloakyl, alkoxy, cycloatkoxy, aryl, aryloxy, arylthio, ary Isulfinyl and aryisulfonyl moiety in the groups represented by R' can be optionally substituted wit one or more independently selected substituents defined above for Structural Formula (1). Alternatively, RI is selected from hydrogen, (Cj-C 7 )alkyl, 25 (CrtC6)cycloalkyl(C -C4)alkyl, (CI C 7 )alkoxy(C-C 4 )alky 1,
(C
3 -Cs)cycloalkoxy(C 1 -C4)alkyl, (C -Cs)cycloalkyl, aryl, auyl(C,-C4)akyl, arvloxy(CI-C 4 )alkyl, aryltuo(ClC4)ailkyi, arylsulfinyl(C -Cj)alkyl and arylsulfonyf(C 1 C4)alkyl In another alternative, R is -41, (C -C 7 )alkyl, or -O-(C C.)alkyl. In another alternative, R is -. H or (C-C 7 )akyi. In another alternative, R 30 is -H1, methyl or ethyl. In yet another alternate ve, RI is -OCB or -OC(CH 3
)
- 10 R2 is selected from hydrogen., (C-C-)alky, (C 3 Cjcycloalkyl(C-Caalkyi (C -C7)alkoxy(C Ci)alkyl, (C 3 -Cs~cycloalkoxy(C'i-C4alkyi, (C-Ct~cy cloalkyl, aryl, aryl(C-C4alkyl, aryloxy (C-C4a[ky, arylthio(C 1 -C4aik ryl sulfiyl(C
C
4 )alkvl, arylsulfonyl(C -C 4 )alkyl and -O-(C-C,)alkyl Each alkyl, cycloalkyl, 5 alkoxy, cycloalkox, ary1, and aryloxy moiety in the groups represented b Rb can be optionally substituted with one or more independently selected substituents defined above for Structural Formula (1). Alternatively, R is selected from hydrogen, (C -C-)alkyl, (C 3 -Cj)cycloalky(C -C)alky], (C-C-)alkoxy(Ci-C4)alkyl., (C-CS)cycloal koxy(CjC4)aIky, (C-Cs)cy cloalkyl, aryi, aryl(C-C4)alkyl, 10 aryloxy( CC4)alkyl, arylthio(C C 4 )alkyl, arylsulfiunyl(CI1 C4)alhyl and arylsulfonyl(CI-C4)alkyi Alternatively, R is selected from (C. -Cy)alkyl, (C -C6)cvcloalkl (C-C4)alkyl, (C-C7)aIkoxy(( C4)alky1, phenyl, pheny1(CI-C4) alkyl, (C-Co)cycloalkyI and .1alo(CC )alkyl, wheinfu each alkyl aikoxy and cycloalkyl moiety in the groups represented by .R 2 is optionally 15 substituted with one or more substituents independently selected f1011 the group consisting of (C-C4)alky'l and halo; and each phenyl moiety in the groups represented by R 2 is optionally substituted with one or more substituents independently selected from the group consisting of (C]C4)alkyl, halo, (CeC)alkoxy, (C-C4)a koxy (C 3
C
4 )alkyl, -CN, halo(C-C4)alkyl, and 20 halo(C -C4)alkoxy. In another alternative, R 2 is selected from the group consisting of cyclopropyl, cy dobutyI, cyclopentyl, cyclopropy methyl, cyclobutylmethyl, phenvl, benzyl, -(CH )-O-CH 3 -(CH )-OCTb -C(CH ). -CI (Cl)2 -CH-0CH ), C124CH(Cil3)2, -C]r 1-CF, -(CH 2 -)CH2F, and -(C.H,),CH. wherein is 0, 1 2, 3, 4, 5 or 6, and wherein the phenyl or benzyl group represented by R 2 is 2$ optionally substituted with one or more substituents independently selected from the group consisting of (CI-C4)ailkyl, halo, (C -C4)aIkoxy, (C C4)alkoxy(C-C}alkyl, -CN. halo(C-C4)alkyl, and halo(C-C )alkoxy. ir another alternaive, R" is a phenyi or bernyl group optionally substituted with one or more substituents independently selected from the group consisting of (C-C4)alkyl, halo, (C] -C4)al koxy, 30 (C -C 4 )alkoxy(CC4)alkyl, -CN, halo(C 1 -C4)aikyl, and halo(C -C.)alkoxy. Tn another aliemnaive, R is unsubstituted phenyl or benzyl. in another attnative, R is selected from cyclopropyl, cy clopropyt methyl, cycl obuty I, cy clopenty I, - 11 cyclohexy -(C2) 2 -- C.H, -C(CH 3 ) -CH(CH) -CH4CF, -CH2CH(CH; )2, -CR I; and -CH Alternatively, RI and R 2 taken together with the nitrogen atom to which they are bonded can also form a ionovclic or bicyclic heteroaryl, or a monocyclic, 5 fused Licyclic, bridged bicyclic or sprro bicyclic heterocycle, wherein the heteroaryl or heterocycle optionally contains one or two additional heteroatoms independently selected fro m N, 0 and S in addition to the N atom to which R 1 and R 2 are bonded. The heteroaryl or heterocycle can be optionally substituted with one or more independently selected substituents described above for Structural Formula (1). 10 Alternatively, the heteroaryl or heterocycle contains one additional heteroaton selected from N, 0 and S. Atenatively, R' and R 2 taken together with the nitrogen atom to which they are bonded form a heterocycle selected from the group consisting of azetidine, pyrroli dine, morpholine, piperidine, octahydrocyclopenta[cjpyrrol, isoindoline, and azabicyclo[3. 1 0]hexane, wherein the 15 heterocycle is optionally substituted with one or more substituents independently selected from i the group consisting of (CIC 4 )alkyl, halogen, -OH, (C -C4)alkoxy, (C -C4)alkyltli o, (CI C4)alkylsulfmyl, (C- C 4 )alkylsnlfony], (C -C4)alkoxy(C:-C4)alkyl, and -N(R)(R') In a more specific embodiment, theso heterocycles are optionally substituted with one or more substitLients independently 20 selected from the group consisting of halogen, (CI 1
C
4 )alkoxy, hydroxy, (C, -C 4 )alkoxy(C 1
"-C
4 )aikyl and -N(R 3
)(R
4 ). In ano her al teiii d ve em bod im ent these heterocycles are optionally substituted with one or more substituents indopend ently selected from the group consisting of halogen, merthoxy, hydoroxy, rnethoxymethyl and dim ethylamino Al tematively, R 1 and R 2 taken together with 25 the nitrogen aloI to wich they are bonded forn a ring selected fon pyrrolidinyl. morpholinyl, azet idinyi pi peridinyl, octah.vdrocyclopenta[cipyrrolyl, isoindolinyl. indazolyl, imidazolyl, pyrazolyl, triazolyl, and tetrazolyl, wherein the ring formed by RW and R2 taken together with the nitrogen atom to which they are bonded is optionally substituted with halogen, (C -C 4 )alkoxy, hydroxy, 30 (C - 4 )aIkoxy(C 1 -C4i)alkyl and -N(R )(R 4 ), More specifically, the ring formed by R and R 2 taken together with the nitrogen atom to which they are bonded is optionally substituted with fluoro, -OH-, -OCH 3 , or N(CH) 2
.
- 12 R. and R 4 are each independently selecLed froin the group consisting of - 1
-
1 and (C:-C 4 )alkyl, wherein the (C -C4)alkyl represented by RW and R 4 is optiona.1l substituted with one or more subsiltiuents independetly selected fromr the group consisting of (Ci-C 4 )alkyl. halo, -OH, (C-C 4 )alkoxy, and 5 (C -C4)alkoxyv(C-Ci)aiky I. Alternatively, RW and R 4 are both methyl. In other alternative, RJ and R 4 are both -H In yet another alternative, R' and R 4 are each unsubstitut4d (C-C 4 )alkyl In a first alternative embodiment, the compound of the present invention is represented by Structural Formula (I) or (I), or a pharmaceutically acceptable salt 10C thcreoft wherein: R' is -H or (C-C7')alkyl; and R2 is selected from (C 1 -C7)alkyl, (Cr, -Cjcycloalkyl(C -C.)alkyl,
(C
1 -C7)alkoxy(C 1 -C4)alkyl- phenl1, pheny|(C -C4)aky1, (C 3 -Cr)cyclo alkyl and halo(C 1 IC4alkyl, wherein each alkyl, alkoxy or cycloalkyl moiety in the groups 15 represented byl R is optionally substituted with one or more substituents independently selected from the group consisting of (C-C 1 )alkyl and halo; and each phenyl moiety in the groups represented by R 2 is optionally substituted with one or more substituents independently selected from the group consisting of (C-C4)alkyl, halo, (CiC4)alkoxy, (C 1 C4)akoxy(C 1 C4alkyl, -CN, halo(Ci-C4)aIky1, and 20 halo(C 1 -C4)alkoxy. Akernatively, R 2 is selected from the group consisting of Cyclopropyl, cyclobutv!, cyclopentyl, cyclopropylmethyl , cyciobutlvImethyl, phenyl, benzyl, -(CI2)O-CI. -(CH )-OCH> -C(CT Iiu -CH(C -CH2CH(tCl-I) 2 , -C -CI 3 , -(CH2)rCHF, and -(CH2) 1 C1 wherein n is 0, 1, 2, 3, A or 6 and wherein the phenyl or benzyt group represented by R is optionally 25 substituted with one or more substiuoents independently selected fron- the group consi sting of (C -C)alkyl, halo, (C -C4)alkoxy, (C) -C 4 )alkoxy(Ci-C4)akyl, -CN halo(C C4)alkyl, an r halo(Ci- C4)al koxy, In another alternative, the phenyl or benzvl group represented by RC is unsubstiluted, in yet another alternative, RS is selected from cyclopropyl, cy clopropylmedthyl, cy clobutyl, cy clopentyl, cyclohexyl, 30 -(CH 2
)
2
-CH
3 , -C(CH 3 ), -CH(C1 3
)
2 , -ClHr-CF 3 , -C.H2CH(CiH±,) -CHA and -Cl 12C L - 13 in a secoiid alternative embodimeni, for compounds represented by Structural Formula (I) or (II), R' is hydroger., methyl or ethyl; and. values and alternative values for R' are as described above for the first alternative embodiment. In a third alternative embodiment for compounds represented by Structural 5 Formula (I) or (II), R t is hydrogen, (Cj-C.,)alkvi or -0-(C-C4)alkyl; R 2 is selected from (C -C7)alkyl, (Cr Cc)cycloalkyl(C-)C4)alkyl, (C i-C7)alkoxy(Ci-C4)alkyl, phenyl, (C!-C cycloalkyi, and fluoro(C-C4)alkyl; or R 1 and R2 taken together wiih the nitrogen atom to which they are bonded form a ring selected from pyrrolidinyl, morpholinyl, azetidinyl, piperidinyl, octahvdrocyclo penta[cipyrrolyl, isoindolinyl, 10 indazoly imidazoly l pyrazolyl, triazolyl, and tetrazolyl, wherein the ring foned by R and R> taken together with the nitrogen atom to which they are bonded is optionally substituted vitli fluoro, -011, -OC-. or N(CW)M More specifically, R' is hydrogen, rihy, othyl, meihoxy or torl-buoxy In a fourth alternative embodiment, for compounds represented by Structural 15 Formula (J) or (R.), Ri hydrogen, methyl, or ethyl; R 2 is selected from methyl, ethyl, i-propy, isopropyl., n-butyl, 2,2-dim ethylpropyl, t-butyl, isobutyl, n-pentyl, (Ci-C 6 )cvcloalkl,. (C>-C5)cycloalkyln.ethyl, methoxyothyl, and 2-fluoroethyl; or R and R 2 taken together with the nitrogen atom to which they are bonded form a ring selected from aietidinyl, pyrrolidinyl, piperidinyl, tetrazolyl., or 20 octahydrocvclopenta[cpyrrolyl, and wherein the ring formed by R' and R2 taken together with the nitrogen atom to which they are bonded is optionaliy subsituted with fluoro. In a fifth alternative embodiment, for compounds represented by Structural Formula (A), when X is hydrogen, Y is selected from hydrogen, -(C-C 4 )alky, 25 -(C-C)alkylene-N(RA)(RB) -(C-C4)alkylene-NI(R-)-C(O)-[C(R')(RI)]sN(R)(R), -CH1=N-OR^, -N(R)(RtR), -NR' )-C(O)-[C(R )(R )]w-N(R.)(R), -N H-C(O)-C(R )(R)-N(R^)(RB), S(RR)C(O)-N(RR), -N(RF )-C()-C -C)alkyl -N(RF)-C(O)-heterocvclv, -N(RF)-C(O)-heteroaryl, -N (RF)-C(0)-caTbocyclyl1, -N(R)-C(O)-aryl 30 -N(Rr)-S(o)e(C C4)aikvylene-NR)R, -N(RF)-S(O,}~-(C, C4)alkylene-carbocy clyl, -N(RF Rr T h is selected from (Cr-Calkyi, carbocyclyl, arvl, heLeroaryl, heterocycily, and a naturally occurring amino acid side chain moiety, or - 14 R and R" taken together with the carbon atom to which they are bound form a 3-7 menibered carbocycly'l, or a 4-7 membered heterocyclyl, wherein the ieterocyclyl formed by RT and R' optionally comprises one to two additional heteroaoms independently selected from N, S and 0; and 5 Rr is selected from (C C7)alkyl, carbocyclyl, aryi and heteroaryl Values and alternative valLies for the remainder of the variables are as described above for Structural Formula (A). Alternatively, Rr is selected front (CI-C4)alkyl and phenyl and the remainder of the vanables are as described above in the fifth alternative embodiment. 10 It a sixth alternative emrboditment, for conpounds represented by Structural Formula (A), X is selected from hydrogen, methyl, ethyl and phenyl; and Y is selected from hydrogen -(Cr.-C 4 alkyl), -(CI -C4),Akyeone-N(R^)(RB),
-(C
1 -CY)alk-ylene-NI l-C()-[CH 2 ]w N(RA)(R), -N(R)(R"), -NHE-C(O0)-carbocy cll -NH-C(0) -aryl, -NH-C(O)-h eterocyclyl, 15 -NI-C(O)-heteroaryl -NH-C(0)-N(R -)(R^), -N(R' )-C(O)-CHAN(R^( RE), -N-COC R N(RA)(R) and -NH-S(O),(C -C4)alkyleneN(RA)(R) or X is selected from methyl, ethyl and phenyl; and Y is -NR--C(O)-C~RN(R^)(R), wherein: each R^ s mdependently selected from hydrogen and methyl: 20 R" is selected from hydrogen, (C] -C 7 )aikyi, -(C 1 -C)alkylene-carbocyciyl, -(CeCs~al kylen e-aryk -(C-Cs)aIlkylerie-heteroary I, -S(OyA(C 1 Cs)alkyi, -(C C4)alkvlene-S(O)a-carbocyclyl, -(CU-C4)alkylene-S(O),< aryl, -(Cc-C4)alkylene-S(O) 1 -hcterocycle and -(CO-Claikylenc-S(0)Lheteroaryl, or R. and R] when bound to a common nitrogen atom are taken together with 25 the nitrogen atom to form a heterocycle, wherein the heterocycle is optionally substituted with -0 and -N(Rf)(R); R) and R are taken together with the carbon atom to vi ich they are bound frm a (C:-C 7 )cycloalkyI; and mi) is 1 or 2; 30 each carbocyclyl, aryl, heterocyclyl or heteroaryl is optionally arid independently subs tied with one or more substituents independently selected from halo, -(-C)alkyl, halo-substituted -(C-C4)akyl. -O-(C, C4)alkyL and -N (R t
)(RG);
- 15 each alkyl portion in the group represented by Y or R" is optionally and independeitly substituted with one or more substituents independently selected from h alo, -Ol4, and -Rv)(R) wherein R is hydrogen or (C-C4)alky, and wherein each alkyl in the group 5 represented by Ra is optionally and independently substituted with ((,-C 6 ) cycloalkyi. The remainder of the variables are as described above in the fifth alternative embodiment. In a seventh alternative embodiment, for compounds represented by Structural Formula (A), X is selected from hydrogen and methyl: and 10 Y is selected from -N(R^)(R'), -N(H)-C(O)-carbocyclyl, -N(H)-C(O)-aryl, -N (H)-C(O)-heterocycle, and -N(H)-C(O)-heteroaryl; or X is methyl, and Y is -NHI-C(O)-CH J;N(RA)(Re), wherein: 15 R^ is hydrogen; and RL is selected from (Ci-C 4 )alkyl, and -S(O)-CH; or R^ and R taken together to form 4-7 membered heterocyclic ring; wherein each carbocycly, ary. heteroaryl, or heterocycle is optionally and independently substituted with onc or more substituents independently selected from -CH 3 - fluoro, and -N(Cl1) 20 In a cig th alternative embodiment, for compounds represented by Structural 0 R 3 N N H A Formula (A), Y is - , wherein ring A represents a 4-7 membered heterocclyl; and iR is hydrogcn or (CI-COalkyl. Values and alternative values for the remainder of the variables are as described above for Structural Formula (A) More specifically, ring A is selected from the group consisting of azetidinyl, 25 pyrrolidinyl, piperidinyl. or octahy drocyclopenta~c]pyrrolyi, each of which is optionally substituted with one or more substitutents independently selected frorn the group consisting of halo, -(C 1 -C4)akyl, hal.o-substituted-(C-C 4 )alkyl (e.g,
-CF
3 ), -Oli -O-(C -C 4 )alkyl, or -N(R)(RG), wherein R' is hydrogen or (C C)alkyl, Even more specifically, ring A described above is optionally substituted 30 with one or more fluoro, - 16 In a ninth alternative embodiment, for compounds represented by Structural Formula (A), Y is -NH-C(0)-heteroaryl. Values and alternative values for the remainder of the variables are as described above for Structural Formula (A). More specifically, he heteroaryl in -NH-C(0)-heteroawl is selected from the group 5 consisti n of thienyl., pyridinyl, pyrrolyl, oxazolyl, pyrazolyl and thiazolyl, each of which is optionally substituted with one or more substituents independently selected frorn -(C -C4)alky1, halo-substituted-(C-C4)alkyl (c g, -CF), -Ot, -O-(C 1 -C)akyl, and -N(R)(RG, wherein kG is hydrogen or (C 1 -C4)al.ki. More specifically, the pyrrolyl and pyrazolyl are optionally substituted wth a methyl group on the N atom 10 in the ring. In a tenth alternative embodiment, for compounds represented by Structural Formula (A), Y is HNI+C( p)-nhenyl, Values and alternative values for the remainder of the variables are as described above for Structura! Formula (A). More specifically, the phenyl in. -NH.-C(O)-phenyl is optionally substituted with one or 15 more substitutents independently selected from -(C 1 - C 4 )aikvl, halo-sibsftuted-(Cr C.4)alkyl (eg, -CF 3 ), -OH. -O-(C-C.j)alkyl, and -N(Ra'('), wherein Ri s hydrogen or (C -C 4 )a'lkVl More specifically, tle phenyl in -NIH-C(O)-phenyl is opuonaly substituted with one or more substitutents independently selected from
-CF
3 , -OCH3 and -N(CH3). 20 In a eleventh alternative embodinent, for compounds represented by Structural Formula (A), Y is represented by -NH-S(0)-(C 1 Csalkyi -NH1-S(O) phonyl, -NII-S(O1-heteroaryl. Values and alternative values for the rernaider of the variables are as described above for Structural Formula (Al More spefically, the phenyl, heteroaryl or alkyl in the group represented by Y is 25 opuonally sibstituted with one or more substueints independently selected from -(C -C4)aikyl. halo-substituted-(C-C4)alkyl (e. -CF 3 ), -OH. -O-(C-C4)al.kyi, and -N(R")(RD), wherein R$ is hydrogen or (C'-C 4 )alkyl. In a twelfth alternative embodiment, for compounds represented by Structural Formula (A), V is represented by -N(RA)(Ra). wherein IA and Re are 30 each independently selected from hydrogen, (CI-C7)alkvl, -(C 1 C.)alkyl (Cr3Q)cycloalkyl, wherein the (CI-C7)alkvl is optionally substituted with -N(Ro)(R), wherein RG' is hydrogen or (C 1 -C4)alkyl. Values and alternative values for the reminder of the variables are as described above for Structural Formula (A), - 17.
In a thirteenth alternative embodiment, for compounds represented by Structural Formula (A), Y is represented by -CH-N(R)(R 3 ), wherein R^ and Rr h are each independently selected from hydrogen, (Ci-C7)alkyl, -(C 1C 1 )alkyl (CrC 6 )cycloalkvl, or RA and R taken together with the nitrogen atom to which they 5 are bound form a heterocyclyl, wherein the (Cr-C7)alkyl represented by R^ or R" is optiorally and independently substituted with -N(R')(R), wherein R' is hydrogen or (C,- C4)akyl, and the (Ci-C)alkyl represented by R' is optionally substituted with -F. Values and alternative values for the remainder of the variables are as described above for Structural FormuLa (A) 10 In a fourteenth alternative embodiment, for compounds represented by Structural Fornula (A), Y is represented by -CH-N()(R'), wherein R'" and RB are each independently selected from hydrogen, (C,-C 7 )alkyl,
-(C-C
4 )aikyl-(C 2
-C
6 )cycloalkyl. Values and alternative values for the remainder of the variables are as described above for Structural Formula (A). 1 The compound of the present invention is exempli fied by the compounds shown in the Table below or a pharmaceutically acceptable salt thereof and compounds described in Examples 1-12 below or a pharmaceuticaly i acceptable salt thereof: COMPOUND Cl-HEK MICAL STRUCTURE NO. 11 H N'CH3 F H H H- OH HC N N c NH 2 H~t H OH
CH
3 HO O HO 0 0 12F
H
3 C,N CH3 H H O O N NH 2 H O O OH 0 0~H 0 18 3F
H
3 C N'CH 3 H H OH H3C NNH HH i H 14 F 3 TCHs
HNCNH
2 H OH 0 OH 0 0 14 F H 3 C N CH, H H H NH 2 H OHO OHOo 17 F HC, N'CH, H NHN OH8 F O3' H
-
NH
2 OH H - H 16 FH$C, CH 3 . C~H:OOH HaF NN H HH -N: ONH
H
3 0 N O2r
CH
3 OIH 0 O 17 F H 2 3C, N XH 3 H H H H
-
O NN NH 2 H6 H OH 0 O H) 0 0 is F HC, CH 3 H H:7 O NH H OH 0 OH0 0 19 F HC H H H 2 OH OH 0 OHH 0(0 -19 201 H-C,N CH F N H H Ha Of I HOHN O O NH OHH FaC NH2 OH o OH 0C0 21 F H-3C, 0H 3 H H H 3C N NH H N d O OH 24 F HCCN N CH H OH 0 = H2H HX HNH 3 N II OH : OH 0 23 HC- N-CH H H
H
3 O N hN
NH
2 H H 0H 2 0 N7 OH
H
3 C N NH HOH 0 OHP 2 5F H ,N C. H H:
H
3 0 f OHI
H
3 C- N >JN NH HOH 0: OH% 0 F H H H HO 0 ,N7 O NH H H C O 0- 0 - 20 27 FH 3 C CH H - O H3N H NrN OH NH 2 H OH 0 OH 0 0 28 F HsC N CH 3 H HF H H 3 C HN~
NH
2 HN
H
3 3 N C HNH HCH 6 , oto 29 H 3 C, r'., F N' H HNH H0 .:>S OH HO OOH HH HF N 'NH 2 CH O OH O 31 F F N H ' C FUH 0H 3 0 N O NN H OH 0 O Ho 0 32 {F H 2 CWX H3 N OH 0 N _NH 2 O f)H 0) O H0 0 33 1H 3 C, CH3 F N H H~ N
NH
2 H OH 'i OH0OH0 -21 34 F HjC' N'CH, H H O 7 OH D FN -~ NH 2 H OH 0 O 6 H NH2 N H OA W 0 NCH H OH F N F3 N N > N NH 2 N C H OH OH 0 OH 0 37 F H3C NCH 3 HFN NN N H OHO OH H N N NH Ho OHO OHO 47 C3N CH F N H Ht HO N-NH: OH O OHO 38F F3N CH Hac, OH HcNIO, N - NH 2 H X6 I OHO OH 0a -9F 13,NCH H HF 'maI NH HON N t z- NH N H H OH 0 % 9H 0 -22 41 F H3C CI1 H HHOH 0N 4N F H NH 2 HC HI HH2 NHNH 0-- H OH 0j FPH 42 F H 3 O CH 3 H, H N Z - OH N N H NH 2 46 F HC N o OHO OPH 0&
UH
3 . N~N OH
NH
2 H O OH 0 OH 0 F H N. H H H: - - OH 0 0 N >E , N NH- 2 H OH 0 OHP C 4 5F HN lA H H: 0~ OH 00 f'13C~ H 2 46F 'N H Hy CH N NH 2 NIO)H 0 0 H 0 "Alkyl" means a saturated aliphatic branched or straight-chain monovalent hydrocarbon radical having th spec'fied number of carbon ators. Thus, "(C1 C-)alkyl" means a radical having from 1-7 carbon atoms in a linear or branched 5 arrangerMent. "(C1-C7)alkvl" includes methyl, ethyl, propyl, butyl, pentyl, hexyl - 23 and heptyl. Suitable substiLtltions fbr a "substituted alkyl" include, but are not limited to, -halogen, -OH, (C -C 4 )alkyl, (C -4)al koxy, (CI -Ci)alkyl thio, (C: -C4)alkyl sulfinyi, (C' -C)alkylsu[fonV, (C-C 4 )aikoxy(C-Ca 1 aky I, and N(R3)(R4), wherein R" and R 4 are as described above. "Cycloalkyl" means a saturated aliphatic cyclic hydrocarbon radical having the specified number of carbon atoms. (C;-Cs)cycloalkvl includes cyclopropyl, cvclobutv cyclopentyl, and cyclohexyl, Suitable substituents for a "substituted cycloalkyl" include halogen, -OH, (C)-C 4 )alkyl, (C 1 -C4)alkoxy, (C-C4)alkylthio, (C -C4:)alkylsulfinVl, (C-C4)alkylsulfonyl, fC-C4alkoxy(C-C4)alkyl, and 10 -N(R )(R), wherein R' and R 4 are as described above. "Heterocycle" means a 4-12 membered partially unsaturated or saturated heterocyclic ring containing 1, 2, or 3 hetei-oatoms independently selected from N, 0 or S When one heteroatom is S, it can be optionally mono- or di-oxygenated (i e. -S(O)- or -S(O)-). The heterocycle can be monocyclic, fused bicychc, bridged 15 bicyclic, or spiro bicyclic. Examples of monocyclic heterocycle include, but not limited to, azetidinc, pyrroiidine, piperidine, piperazine, hexahydropyri idi ne, tetrahydrofuran. tetralydropyran, morpholine, thionorpholine, thiomorpholine 1,1-dioxide, tetrahydro-2H- I,2-thiazine, tetrahydro-2H-1 i,2-ihiazine 1-dioxide, isothiazoli dine, 20 i sothiazolidine i,1-dioxide. A fused bicyclic haterocycle has two rings which have two adjacent ring atons in common. The first ring is a monocyclic heterocycle and the second ring is a cycloalkyl, partiafly unsaturated carbocycle, phenyl, heteroaryl or a nionocycici heterocycle. For example, the second ring is a (C,;-Ccloalkyl, such as 25 cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. Alternatively, the second ring is phenyl. Example of fused bicycli c heterocvcles includes, but not limited to, indoline, isoindoline, 2,3-di hydro-1H-benzoed]imidazole, 2,3 dihy drobeinzo[dJoxazole, 2,3-dihy drobenzo d]tiazole, octahy drebenzo d]oxazole, ociahvdr o-iH-benzo[d]imidazole, octahydrobenzo[d]ihiazole, 30 ectahydrocyclopenta[]pyrrole, 3-azabicyclo[3. I 0]hexane, and 3 azabicycloj3 .2.0] heptane. A spiro bicyclic heterocycle has two rings which have only one ring atom in common The first ring is a monocyclic heterocycie and the second ring is a - 24 cycloalkyl partially unsaturated carbocycle or a monocyclic heterocycle For example, the second ring is a (Cr-C)cycloalkyl. Example of spiro bicyclic heterocycle includes, but not limited to, azaspiro[4.4]nonane, 7 azaspiro[4.4lnonane, azasprio[4.5]decane, 8-azaspiro(4.5]decane, 5 azaspiro[5.5}undecane, 3-azaspiro[5.5 ]undecane and 3,9 -diazaspiro[5.5]undec an e. A bridged bicyclic hoterocycle has two rings which have three or more adjacent ring atoms in common. The first rina is a monocyclic heterocycle and the other ring is a cycloalkyl (such as (C-C)cycloalkyl), partially unsaturated carbocvcle or a mnonocyclic heterocycle. Examples of bridged hi cyclic heterocycle s 10 include, but are not limited to, azabicyclo[3.3. 1]nonane, 3-azabicyclo[3 .3.1 ]nonane, azabicyclo[3.2.1joctane, 3-azabicyclo[3.2 I]octane, 6-azabicyclo[3 2.1 ]octane and azabicyclo[2.2,2]octane, 2-azab'cyclo[2.2.2]octane. When the heterocycle contains a N atom other than the nitrogen atom to which Ri and R 2 are bonded, the N atom can be substituted with H, alkyl, 15 cvcloalkvl, cycloalkylalkyl, arv, arylalkyl, heteroaryl, heteroarylalkyi, each of which can be optionally substituted with halogen, hydroxy, aikoxv, haloalkyl aikyl, etc. The hetorocycle can be optionally substituted with an oxo group (C=0) and oxo substituted heterocyclic rings include, but are riot limited to, thiomorpholine 1 oxide, thiomorpholine 1,1-dioxide, tetrahydro-H-1,2-rhiazine I 1-dioxide, and 20 isothiazolidine I I -dioxi de, pyrroli din-2-one, piperidin-2-one, piperazin-2-one., and morpholin-2-one. Other optional substituents for a heterocvcle include
(C
1 -C4)alkyl, halo, -01H, (C-C4)alkoxy, (CI -C4)alkythio, (CIrC 4 )alkvlsulfinyl,
(C
1
-C
4 )alkylsulfonyl, (C -C 4 )alk oxy(Ci-C4f)alkyl, -N(R'(R), -CN, halo(C-C4)alkyl, and halo(CI -C)alkoxy. 25 "Heteroaryl" means a 5-12 membered monovalent heteroaromatic rmonocyclic or bicylic ring radical. A herteroaryl contains 1, 2 or 3 heteroatoms independently selected from N4, 0, and S. IHeteroaryls include, bIu are not limited to pyrrole, indazole, pyrazole, oxazole, isoxazole, thiazole, isothiazol , (riavole, 1,2,4-triazole, 1,3 ,4-oxadiazole, 1,.5-thiadiazole, 1,2,5-thiadiazole l 30 oxide, .,2,5thi adiazole 1, 1 -dioxide, 1,3,4-thiadiazole, pyri dine, pyrazin, py rimi dine, pvridazine, 1,2,4-triazine, 1,3,5-triazmne, and tetrazole. Bicyclic heteroaryl rings include, but are not limited to., bicyclo[4.4 0] and bicyclo[4.3.0] fied ricg systerns such as indolizine, indole, isoindole, indazole, benzinidazole, - 25 bentLijazolc, prne , quinoline, isoquinoline, cinnoline, phthalazine, quinazolne, quinoxaline, 1,8-naphthyridine, and pteridine. "Carbocycl" means 4-12 mrembered saturated o: unsaturated aliphatic cyclic hydrocarbon ring. 5 "Alkoxy" means an alkyl radical attached through an oxygen linking atom. "Alkoxy" can also be depicted as -0-alkyl. For example, (C-C.4)-alkoxy can also depicted as -O-(C 1 -C4)alkyl. "(C-C4)-aikoxv" includes methoxy, ethoxy, propoxy, and butoxy. "Alkyltho" means an. alkyl radical attached through a sulfur linkir atom 10 "Alkvlthio'" can also be depicted as -S-aikyl. For example, "(C-C4)alkvithio" car be depicted as -S(C-C4)alkyl. "(CinCcalkylth in clude methylthio, ethylthio, propylthio and butythio. "Alkylsilfinyl" means an alkyl radical attached through a -S(O)- linking group. "Alkysulliny!" can be depicted as -S(0)-alkyl. For example, 15 "(CI 4,)alkylsulfinyi" can be depicted as -S(0)-(C-C 4 )alkvl. "(C C 4 )alkylsulfinyl" include methylsulfinyl, ethylilsulfi nyL. propylsulfinyl and "Alkyisulfonyl" means an alkyl radical attached through a -S(0)- hnliking group. "Alkylsulfonyl" can be depicted as -S(O)-alkvl. For example, 20 "(C -C 4 )alkylsul finyl" car be depicted as -S(0) 2
-(C-C
4 )aikv. "(C -C4)alkylsulfonyl" include methylsul fonyl, ethylsulfonyl, propyl suifonyl and buatyl sualfonyt. Haloalkyl and halocycloalkyl include mono, poly, and perhaloalkyl groups wrj e each halogen is iidependently selected froni fluorine, chlorine, and brormine. 25 Hlaloalkyl arid halocycloalkyl cmi also be referred as halo-substituted alkyl and halo-substituted cycloalkyl, respectively. "Cycloalkoxy" means a cycloalkyl radical attached through an oxygen linking atom. "Cycloalkoxy" can also be depicted as -0-cycloalkyl. For example, "(C-C)cy cloalkoxy" can be dep actedd as -0-(C3-Cs)cycloalkyl. "(C 30 C )cycoalkoxv" includes cyclopropyloxy, cyclobutyloxy, cyclopentyloxy and cyelolexyloxy.
- 26 "Arv" means an aromatic monocyclic or polycy click (e.g. bicyclic or tricyclic) carbocyclic ring system. In one embodiment, "aryl" is a 6-12 membered monocylic or bi cy clic systems. Aryl systems include, but not limited to, phenyl, naphihaleyti, fluoreny indeny1, azulenyl, and anthracenyl. 5 "Aryloxy" means ar ayl moiety altachted through an oxygen linking aton, "Aryloxy" can be also depicted as -O-arvl. Aryloxy includes, but not limited to, phenom xy "Arylthio" means an aryl moiery attached through a sulfur linking atom. "Arvlthio" can be also depicted as -S-aryl1. Arylthio includes, but not limited to, 10 phenylthio. "Arylsulfinyl" means an aryl moiety attached through a -S(O)- linking group. "Arvlsulinyl" can be also depicted as -S(O)-aryl. Arylsulfinyl includes, but not limited to, phenylsulfinyl. "Arylsulfonyl" means an aryI moiit attached through a -S(O)2- linking 15 group. "Atylsulfonyl" " can be also depicted as -S(O)2-aryl Arylsulfonyl includes, but not limited to, phenvsulfonyl. "Hetero" refers to the replacement of at least one carbon atom member in a ring system with at least one heteroatom selected from N, S, and 0. "Hetero" also refers to the replaceroent of at least one carbon atom member in a acyclic system. 20 A hetero ring system or a heteio acyclic system may have 1, 2, or 3 carbon atom members replaced by a heteroatoni "Haloqen"or "halo" used herein refers to fluorine, chlorine, bromine, or iodine. As used herein, cycloalkylalkyl can be depicted as -alkylene-cycloalkyl. For 25 example, (C3-C6)cvcloalkyl(C 1 -Ci)alkv can be depicted as -(CIC4)aikylon-(CC 6 )cycloalkyl. As use herein, alkoxyalkyl can be depicted as -alkylene-O-alkyl. For example., (C 1 -C7)alkoxv(C-Cj)alkyi can be depicted as
-(CI-C
4 )alkyiene-O-(C -C7)alkyl. 30 As use herein. cycloalkoxyalkyl can be depicted as -alkylene-0-cycloalkyl. For example, (C 3
-C
6 )cycloalkoxy(C 1
-C
4 )alkyl can be depicted as c-(C C)alkyiene-O-(C-Cs)lal ky.
- 27 As use herein, arvlalkyl can be depicted as -alkylnc-aryl. For example, aryl(C-C4)alkyl can be depicted as -(C-C4)alkylene-aryl. As used herein, aryloxyalkyl can be depicted as -alkylene-O-aryl. For example, aryloxy(Cr-C 4 )aikyi can be depicted as -(C C )aiky lene-O-aryl 5 As used herein, arvlthioaikyl can be depicted as -alkylene-S-aryl For example, arylthio(C C4 )alkyl can be depicted as -(C 1 -C4)alkylene-S-aryl. As used herein, aryisu linylalkyl can be depicted as -alkylene-S(O)-arv For example, aryl sufinyl (C.-C4)alkyl can be depicted as -(C -C4)al kylene-S(O)-aryl As used herein, arylsulfonvlalkyl can be depicted as -alkylene-S(O)-aryl. 10 For example, arylsulfonyl(C-C4)alkyl can be depicted as
-(
1 -C)aikvlene-S(O)-aryl. Another embodimert of the present invenrion is a pharmaceutical composition comprising one or more pharmaceutically acceptable carrier and/or diluenti and a compound disclosed herein or a pharmaceuticalfly acceptable salt 15 thereof. LPharmaceutically acceptable cancer" and "'pharmaceutically acceptable diluent" means non-therapeutic components that are of sufficient purity and quality for use in the formulation of a compomtion of the invention that, when appiropriatelv administered to an animal or human, typically do not produce an adverse reaction, 20 and that are used as a vehicle for a drug substance (i.e. a compound of the present invention). Pharmaceutically acceptable salts of the compounds of the present invention are also included. For example, an acid salt of a comripound of the presena invention containing an amine or other basic group can be obtained by reacting the compound 25 with a suitable organic nr inorganic acid, resulting in pharmaceutically acceptable anionic salt forms. Examples of anionic salts include the acetate, benzenesulfonate, henzoate, bicarbonate, bitarrate, bromide, calcium edetate, camsxlate, carbonate, chloride, cirate, dihydrochloide, edetate, cdisylate, estolate, esylate, fumarate, glyceptate, gluconate, glutamate gly collylarsanilate, hex y]resorc iate, 30 hydrobromide, hvdrochlori de, .hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, malate, maleate, mandolate, mesy late, methylsulfate, mucate, napsylate, nitrate, pamoate, pantothenate, phosphate/diphosphate, polygalacturonate, - 28 salicvlate, stearate, subacetate, succinate, sulfate, taiate, tartrate, teoclate, tosylate, and triethiodide salts, Salts of the compounds of the present invention containing a carboxylic acid or other acidic functional group can be prepared by reacting with a suitable base. 5 Such a pharnaceutically acceptable salt may be made with a base which affords a pharima cuti calIly acceptable caion, which includes alkali metal salts (especially sodium and potassium), alkaline earth metal salts (especially calcium and ragnesiun, aluminum salts and ammonium salts, as well as salts made from physiologically acceptable organic bases such as tinmethylanine, trie thylanine, 10 inorpholine, pyridine, piperidine, picoline, dicy clohexylanine, N,N'-dibenzylethiylenediamine, 2-hydroxy ethylatmine, bis- (2-ly droxyeihyf)aiine, tri-(2-hydroxyethyl)amine, procaine. dibeizylpiperidine, dlehydroabiet ylamir e, NN='-bisdehy droabietylainine, glucamine, N-methiLg! u camine, coilidine, quinine, quinoline, and basic amino aci ds such as lysine and arginine. 15 The invention also includes various isomers and mixtures thereof. Certain o fthe compounds of the present invention may exist in various stercoisomeric foris, Stereoisoiers are compounds which differ only in their spatial arrangement. Enantiomers are pairs of stereoisomers whose mirror images are not s uperimposable, most commonnly because they cotuin an asyntnetrically 20 substituted carbon atom that acts as a chiral center. "Enantiomer" means one of a pair of molecules that are mirror images of each other and are not superimposabLe. Diastereomers are stereoisorners that are not related as mirror images, most commonly because they contain two or more asyrmetrically substituted carbon atoms. "R" and "tS" represent the confi.guration of substituents around one or more 25 chiral carbon atoms, When a chiral center is not defined as R or S, either a pure enaniomer or a mixture of both configurations is present. "Racemate" or "racenic mixtur" means a compound of equimolar quantities of two enantiomers, wherein such mixtures exhibit no optical activity: i.e., they do not rotate the plane of polarized light. 30 The compounds o th invention may be prepared as individual isomers by either isomer-specific synthesis or resolved from an isomeric mixture. Conventional resolution fechtiiques include forming the salt of a free base of each isomer of an isomeric pair using an optically active acid (followed by fractional - 29 crystallization and regeneration of the free base), forming the salt of the acid form of each isomer of an isomneric pair using an optically active amine (followed by fractional cIystallizaion and regeneration of the free acid), forming an ester or amide of each of the isomers of an isomeric pair using an optically pure acid, amine 5 or alcohol (followed by chrtomatographic separation and removal of the chiral auxiliary), or resolving an isomeric mixture of either a starting material or a final product using various well known chronatographic niethods. When the stereochemistry of a disclosed compound is named or depicted by structure, the named or depicted stereoisomer is at least 60%, 70%, 80%. 90%,99% 10 or 99.9% by weight pure relative to the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% by weight optically pure. Percent. optical purity by weight is the ratio of the weight of the enantiomer that is present divided by the confined weight of the enantionier that is present and the weight of 15 its optical isomer. The present invention also provides a method of treating or preventing a subject with a tetracycline-responsive disease or disorder comprising administering to the subject an effective arriount of a com pounLd of the present invention or a pharmaceuticallv acceptable salt thereof 20 "Tetracycline-responsive disease or disorder" refers to a disease or disorder that can be treated, prevented, or otherwise ameliorated by the administration of a tetracycline cormpoLind of the present invention Tetracycine-responsive disease or disorder includes infections, cancer, inflarmiatory disorders, autoimrune disease, arieriosclerosis, corncal ulceration, emiphvsema, arthritis, osteoporosis, 25 ostcoarthritis, multiple sclerosis, osteosarcoma, ostonmyelitis, bronchicetasis, chronic pulmonary obstructive disease, skin and eye diseases, perodontitis, osteoporosis, rheumatoid arthritis, ulcerative colitis, prostatitis, tumor growthi and invasion, metastasis, diabetes, diabetic proteinuria, panbronchlolitis; aortic or vascular aneursms, skin tissue wounds, dry eye, bone, cartilage degradation, 30 malaria. senescence, diabetes, vascular stroke, neurodegenerative disorders, cardiac disease, juvenile diabetes, acute and chronic bronchitis, sinusitis, and respiratory infections, including the comrron cold; Wegener's grand iulomatosis; neutrophilic dermuatocss and other i ammatory diseases such as dermatitis herpetiformis, - 3 leukocytoclastic vasculitis, bullous lupus erythem atosus, pustular psoriasis, ervthena elevatum diutinurn; vitiligo; discoid lupus erythematosus; pyoderma gangrenosum; pustular psoriasis; blepharitis, or incibomianitis; Alzheimer s disease; degenerative maculopathy; acute and chronic gastroenteritis and colitis: acute and 5 chronic cystitis and urethritis; acute and chronic dermatitis; acute and chronic conjuictivitis; acute and chronic serositis; uremic pericarditis; acute and chronic choiecysis cystic fibrosis, acute and chronic vaginitis; acute and chronic uveitis; drug reactions; insect bites; burns and sunburn, bone mass disorder, acute lung injury, chronic lung disorders, isch eria, stroke or ischermic stroke, skin wound, 10 aortic or vascular aneurysm, diabetic reinopathy, hemorrhagic stroke, angiogenesis, and other states for which tetracycline compounds have been found to be active (see, for example, U S. Patent Nos. 5,789,395; 5,834,450; 6,277,061 and 5,532.227, each of which is expressly incorporated herein by reference). In addition, a method to treat any disease or disease state that could benefit 15 from modulating the expression and/or function of nitric oxide. metallop-oteases, proinflammatoty mediators and cytokines, reactive oxygen species, components of the immune response, including chemotaxis, lymphocyte transformation, delayed bypersensitivty, antibody production, phagocytosis, and oxidative metabolism of phagocytes. A method to treat any disease or disease state that could benefit from 20 muodulati ng the expression and/or function of C-reactive protein, signalin pathways (e.g, FAK signaling pathway), and/or augment the expression of COX-2 and PGE 2 production is covered. A method to treat amy disease or disease state that could benefit from inhibition ofneovascularization is covered Compounds of the invention can be used to prevent or treat important 25 mammalian and veterinary diseases such as diarrhea. uinary iract mfections, infections of skin and skin structure, ear, nose and throat infections, wound infection, mastitis and the like. In addition, methods for treating neoplasms using tetracycline compounds of the invention are also i included (van der Bozert et al., Cancer Res, 48: 6686-6690 (1988) ). 30 Infections that can be treated using compounds of the invention or a pharimaceutically acceptable salt thereof include, but are not limited to, skin infections, C infections, tuinary tract infections, genito-urinary infections, respiratory tract infections, sin uses infections, middle ear infections, systemic - 31 infections, cholera, influenza, bronchitis, acne. malaria, sexually transmitted disease including syphilis and gonorrhea, Legionnaires' disease, Lyme disease, Rocky Mountain spotted fever, Q fever, typhus, bubonic plague, gas gangrene, hospital acquired infections, leptospirosis, whooping cough, anthrax and infections caused by 5 the agents responsible for lymphogranuloma venereum, inclusion conjunctivitis, or psittacosis. Infections can be bacterial, fungal, parasitic and viral infections (including those which arc resistant to other tetracycline compounds). In one embodiment, the infection can be caused bacteria. In another ebodiment, lie infection is caused by a Gram-positive bacteria. In a specific 10 aspect of this embodiment, the infection is caused by a Gram-positive bacterium selected from Staphylococcus spp- Sireptococcus spp., Propionibacterium spp, Enterococcus spp., Baciluus spp., Corynebacterium spp. Aocardia s pp, Clostridium spp., Actinobacteria pp, amd ,isteria spp. In another embodiment, the infection is caused by a Gram-negative 15 bacterium. In one aspect of this embodiment, the infection is caused by a proteobacteria (e.g, Betaproteobacteria and Gamnaproteobacteria), includIng [Escherichia col, Salmonella, Shig/clla, other Enterobactcriaccae, Pseuadononus, Moraxella, i-clicobacter, Stenotrophomonas, .de/lovibrio, acetic acid bacteria, Legionela or aipha-proteobacteria such as Wolbachia. In another aspect, the 20 infection is caused by a Gram-negative bacteria selected from cyanobacteria, spirochaetes, green sulfur or green nion-sulfur bacteria. In a specific aspect of tbis embodiment, the infection is caused by a Gram-negative bacteria selected from Lnerobactericeae (e.g., . coh, Klebsiellapneumonia including those containing extended-spectrum -lf3 actamases and/or carbapenemases), Bacewroidaceae ( a, 25 BacVroids fragi. Vibtronaccae (Vibrio cholerae, Pasteure/ac (e., Hacrophilus influenza), Pseudomonadaceac (e.g, I've utdom onas aeruginosa), Neisscr/aceae (e.g. Neisseria meningiidis), Rickenusia Moraxel/uaca (e Muroraxella catarrhais), any species of Proieeae, Acinefobacter spp., f/elicobacter spp., and Campyl o b acrer spp, 30 In a particular embodiment, the infection is caused by Gram-negative bacterium selected from the group consisting of Jnerobaericea (e.g, E col, K/cbsiela pnumoniac), Psendononas, and Acinefobacwter sp - 32 In another embodiment, the infection is caused by an organism selected from the group consisting of K. pneumnoniae, &wlnoucia, L. hirac, A baumanii, MW. catarrhalis, H. iafltuenzac, P. aeruginosa, K fuecimn . coli, S. are us, and F. faecalis. In anotlcr embodiment, the infection is caused by an organism selected fira in the group consisting of rickeirsiae, chIarmydiae, fegione/la spp and Mycoplasma Spp. in another embodiment, the infection is caused by an organisn resistant to tetracycline or any member of first and second generation of tetracycline antibiotics (e.g. doxycycline or minocycline). 10 In another embodiment, the infection is caused by an organism resistant to methicillin. In another embodiment, the infection is caused by an organism resistant to vanco my cin, In another embodiment. the infection is caused by an organism resistant to a 15 quinolono or f1 uoroqui nolone. tn another embodiment, the infection is caused by an organism resistant to tigcycline. In another embodiment, the infection is caused by a multidrug-resistant pathogen (having intermediate or full resistance to any two or more aiitibiotcs) In 20 another embodiment the infection is a Bacillus anthracis infection "Bacilius anthracs infectionr" includes any state, diseases, or disorders caused or winch result from exposure or alleged exposure to Bacillus anyhracis or another member of the Bacdlur cereus group of bacteria. In another embodiment, the infection is caused by Bacillus anthracis (anthrax), Yers'inapestis (plague), or Francise/la tularensis 25 (tularemia) In yet another embodiment, the infection can be caused by more than one organisi described above. Examples of such infections include, but are not limited to, intra-abdorninal infections (often a rnixture of a gram-negative species like k. colt and an anaerobe like B. fragihs), diabetic foot (various cornbinations o1' 30 Streptococcus, Serratia. Staphylococcus and Enterocccus spp., anaerobes (S K Dowd, et a,, PoS one 2008;:e3325) and respiratory disease (especially in patients that have chronic infections like cystic fibrosis - e.g, S. aurcus plus P. aeruginosa or H in/hesna, atypical pathogens). wounds and abscesses (various gram-negaive - 33 and gram-positive bacteria, notably MSSA/ARSA, coagulase-negative staphylococci, enterococi, Acinelobacter, P, a.eruginosa F coi, B.and bloodstream infections (13% were polymicrobial (H. Wisplinghoff, et al, Cin. Infect, Dis. 2004:39:311-317)). In a further embodiment, the tetracycline responsive disease or disorder is not a bacterial infection. In another embodiment., the tetracycline compounds of tie' invenion are essential] non-antibacterial For example, non-antibacteral compounds of the invention may have MIC values greater than. about 4 g/m (as measured by assays known in the art and/or the assay given in Example 14, In 10 another onbodiment, the tetracycline compounds of the invention have both antibacterial and non-antibacterial effects. Tetracycline responsive disease or disorder also includes diseases or disorders associated with inflammrtoiy process as sociated states (IPAS). The tenn 'inflammatory process associated state" includes states in which inflannation or I5 inflammatory factors (e.g., matrix metalloproteinases (MNPs), nitric oxide (NO), TNF, interleukins, plasma proteins, cellular defense systems, cytokines, lipid metabolites, proleases, toxic radicals, adhesion molecules, etc.) are involved or are present in an area in aberrant amounts, e g., in amounts which may be advantageous to a ter, e.g, to benefit the subject. The infammatory process is the response of 20 living tissue to damage. The cause of inflammation may be due to physical damage chernical substances, mi cro-organlisns, tissue necrosis, cancer or other agents. Acute inflammation is short-lasting, lasting only a few days, If it is longer iastingr however, then it may be referred to as chronic in tlanimation. IPASs include infl ammatory disorders. Inflamiatory disorders are generally 25 characterized by heat, redness, swelling, pain and loss of function. Examples of causes of inflaminatoiy disorders include, but are not limited to, microbial infections (e g., bacterial and fungal infections), physical agents (e.g,, burns, radiation, and trauma), chemical agents (e.g., toxins and caustic substances), tissue necrosis and various types of immunologic reactions. 30 Examples of inflammatory disorders can be treated using the compounds of the invention or a pharmaceutically acceptable salt thereof include, but are not limited to, osteoansbritis, rheuiatoid arthritis, acute and chronic infection (bacterial and fungal, including diphtheria and portUssis); acute and chronic bronchitis, - 34 sinusitis, and upper respiratory infections, including the common cold; acite and chronic gastroenteriis and colitis; inflammatory bowel disorder; acute and chronic cystitis and ureth ritis; vasculitis; sepsis; nephritis; pancreatitis; hepatitis; ILptus; inflammatory skin disorders including, for example, eczema, dermatitis. psonasis, 5 pyoderma gangrenosum, acne rosacea, and acute and chronic dermatitis; acute and chronic conjunctivitis; acute and chronic serosiis (pericarditis, peritonitis. synovitis, pleuriis and tendinitis); uremic pericarditis, acute and chronic cholecystis; acute and chronic vaginitis; acute and chronic uveitis; drug reactions; insect bites; burns (th ermual, chemical, and electrical); and sunburn. 10 PASs also include matrix metalloproteinase associated states (MMPAS). MNVPAS include states characterized by aberrant amounts of MMPs or MMvP activity. Examples of matrix metalloproteinase associated states ("MMPAS's") can be treated using compounds of the invention or a pharmaceutcally acceptable salt thereof, 15 include, but are not limited to, arteriosci erosis, corneal ulceration, ernphy sema, ostcoarthrii is, mui]iple sclerosis (Liedtke et al., Ann Neurol. 1998, 44: 35-16; Chandler et al., J. Neuroimmunot. 1997, 72: 155-71), osteosarcoma, osteomyelitis., bronchiectasis, chronic pulmonary obstructive disease, skin and eye diseases, periodontitis, osteoporosis, rheumatoid arthritis, ulcerative colitis, inflarnmatoiy 20 disorders, tumor growth and invasion (Stetler-Stevenson ct al., Annu. Rev Cell Biol 1993, 9. 54 1-73; Tryggvason et a., Biochim Biophys. Acta 1987, 907: 191-21.7 ;1 i et a., Mol Carcillog, 1998, 22 81-89) ), metastasis, acute lung injury, stroke, ischenia, diabetes, aortic or vascular aneurysms, skin tissue wounds, dry eye, hone and cartilage degradation (Greenswald et al., Bone '998,22 : 33-38; Ryan et aL, 25 Curr. Op. Rheumatol. 1996, 8: 238- 247), Other MMPAS include those described in U. S. Pat Nos. 5,459,135; 5,321,017; 5,308,839; 5,258,371; 4,935,412; 4,704,383, 4,666.897, and RE 34,656, incorporated herein by reference in their entirety. In a further embodiment, the IPAS includes disorders described in U, S. Patents Nos. 5,929,055; and 5,532,227, incorporated herein by reference in their 0 entiretv Tetracycline responsive disease or disorder also includes diseases or disorders associated with NO associated states The term "NO associated states' incutter states which involve or are associated with nitric oxide (NO) or Tnducib[e - 35 nitric oxide synthase (iNOS). NO associated state includes states which are characterized by aberrant arnounts of NO and/or iNOS. Preferably, the NO associated state can be treated by administering tetracycline compounds of the invention. The disorders, diseases and states described in U S. Patents Nos 5 6,23 1,894, 6,01 5,804; 5,919,774; and 5,789,395 are also included as NO associated states. The entire contents of each of these patents are hereby incorporated herein by reference. Fxamples of diseases or disorders associated with NO associated states can be treated using the compounds of the present invention or a pharnaceutcaly 10 acceptable salt thereof include, but are not limited to, malaria, senescence, diabetes, vascular stroke, neurodegenerative disorders (Alzheinief's disease and Hunt ington's disease), cardiac disease (reperfusion-associated in ury following infarction), juvenile diabetes, inflaimmatory disorders, osteoarthritis, rheumatoid arthritis, acute, recurrent and chronic infections (bacterial, viral and fungal); acute and chronic 15 bronchitis, sinusitis, and respiratory infections, including the common cold, acute and chronic gastroenteritis and colitis; acute and chronic cystitis and urethritis, acute and chronic dermatitis; acute and chronic conjunctivitis; acute and chronic serositis (pericarditis, peritonitis, synovitis, pleuritis and tendonitis); urenic pericardiis. acute and chronic cliolecystis; cystic fibrosis, acute and chronic vaginitis; acute and 20 chronic uveitis; drug reactions; insect bites, burns (thermal, chemical, and elecifclal); and sunburn. In another embodiment, the tetracycline responsive disease or disorder is cancer. Examples of cancers that can be treated using the compounds of the invention or a phrmaceutically acceptable salt thereof include all solid tumors, i.e, 25 carcinomas e.g, adenocarcinonas, and sarcomas. Adenocarcinomas are carcinomas derived from glandular tissue or in which the tumor cells form recognizable giandulIr struck tures. Sarcomas broadly include tuinors whose cellIs are embedded in a fibriliar or homogeneous substance like embryon ic connclve tissuI-e. Examples of carcinomas which may be treated using the methods of the invention include, but are 3D not li mited to, carcinomas of the prostate, breast, ovary, tests, lung, colon, and breast. The methods of the invention are not limited to the treatment of these tumor types, but extend to any solid tumor derived front any organ system. Examples of treatable cancers include, but are not limited to, colon cancer, bladder cancer, breast - 36 cancer, melanoma, ovarian carcinoma, prostate carcinoma, lung cancer, and a variety of other cancers as well, The methods of the invention also cause th e inhibition of cancer growth in adenocarcinomas, such as, for example, those of the prostate. breast, kidney, ovary, testes, and colon. In one embodiment, the cancers a treated by methods of the inventiori include those descri bed in U S. Patent Nos. 6,100,248; 5,843,925; 5,837,696; or 5,668,122, incorporated herein by reference in their entirety Alternatively, the tetracycline compounds may be useful for preventing or reducing the likelihood of cancer recurrence, For example, to treat residual cancer 10 following surgical resection or radiation therapy. The tetracycline compounds useful according to the invention are especially advantageous as they are substantially non-toxic compared to other cancer treatments. In a further embodiment, the compounds of the invention are administered in combination with standard cancer therapy, such as, but not limited to, chemotherapy. 15 Examples of tetracycline responsive states can be treated using the compounds of the invention or a phannaceutically acceptable salt thereof also include neurological disorders which include both neuropsychiatic and neurodegenerative disorders, but are not limited to, such as Al zheimer's disease, dementias related to Alzheimer's disease (such as Pick's disease). Parkinsun's and 20 other Lw diffuse body diseases, senile dementia, Huntington's disease, Gilles de la Tourette's syndrome, multiple sclerosis, aiotrophi c lateral sclerosis (ALS), progressive suprIMuclear palsy, epilepsy, ana Creutzfeldt-Jakob disease; autonomic function disorders such as hypertension and sleep disorders, and neuropsychiatric disorders, such as depression, schizophrenia, schizoaffective disorder, KorsakolTs 25 psychosis, mania, anxiety disorders, or phobic disorders learning. or memory disorders, e. g, anesia or age-related memory loss, attention deficit disorder, dysthymic disorder, major depressive disorder, mania, obsessive-compulsive disorder, psychoactive substance use disorders, anxiety, phobias, panic disorder, as well as bipolar affective disorder, e. g., severe bipolar affective (tood) disorder 30 (BP- ), bipolar affective neurological disorders, e g. , migraine and obesity. Further neurological disorders include, for example, those listed in the American Psychiatric Association's Diagnostic and Statistical manualI of Mental - 37 Disorders (DSM), the most current version of which is incorporated herein by reference in its entirety. In another embodiment, the tetracycline responsive disease or disorder is diabetes. Diabetes that can be treated using the compounds of the invention or a 5 pharmaceutically acceptable sal thereof include, but aire not limited to, juvenile diabetes, diabetes muellitus, diabetes type I, or diabetes type I. In a further embodiment, protein glycosylation is not affected by the administration of the tetracycline compounds of the invention. In another embodiment, the tetracycline compound of the invention is administered in combination with standard diabetic 10 therapies, such as, but not limited to insulin therapy. In another etmbodiient, the tetracycline responsive disease or disorder is a bone mass disorder. Bone mass disorders that can be treated using the compounds of the invention or a pharmaceutical acceptable salt thereof include disorders where a subjects bones are disorders and states where the formation, repair or 15 remodeling of bon e is advantageous. For examples bone mass disorders inci ude osteoporosis (e. g. , a decrease in bone strength and density). bone fractures, bone formation associated with surgical procedures (e. g., facial reconstruction), osteogenesis imperfecta (brittle bone disease), hypophospharasi a, Paget's disease, 1ibrous dysplasia. osteopetrosis, myeloma bone disease, and the depIction of cal ciui 20 in bone, such as that which is related to primary hyperparathyroidism. Bone mass disorders include all states in which the formation, repair or remodeling of bone is advantageous to the subject as well as all other disorders associated with the bones or skeletal system of a subject which can be treated with the tetracycline compounds of the invention. In a further embodiment, the bone mass disorders include those 25 described in U. S Patents Nos. 5,459,135; 5,231,017. 5,998,390; 5,770,588; RE 34,656; 5,308,839; 4,925,833; 3.304,227; and 4.666,897, each of which is hereby incorporated herein by reference in its entirety. In another embodiment, the tetracycline responsive disease or disorder is acuto lung injury, Acute ung injuries that can be treated using the compounds of 30 the invention or a pharnraceutically acceptable salt thereof miude adult respiratory distress syndrome yards) , post-pump syndrome (PPS), and trauma. Trauma includes any injury to living tissue caused by an extrinsic agent or event. Examples - 38 of trauma include, but are not limited to, crush injuries, contact with a hard surface, or cutting or other damage to the lungs. The tetracycline responsive disease or disorders of the invention also include chrone lung disorders. Examples of chronic lung disorders that can be treated using 5 the compounds of the invention or a pharmaceutically acceptable salt thcreoaf include, but are not limited, to asthma, cystic fibrosis, chronic obstructI ve pulnionary disease (COPD), and emphysema. In a further embodiment, the acute and/or chronic lung disorders that can be treated using the compounds of the invention or a pharmaceutically acceptable salt thereof include those described in U. S, Patents No, 10 5,977,091; 6,043,23l; 5,523,297; and 5,773,430, each of which is hereby incorporated herein by reference in its entirety. In yet another embodi ment, the tetracycline responsive disease or disorder IS ischemi a, stroke, or ischem ic stroke. In a further embodimient, the tetracycline compounds of the invention or a 15 pharmaceutically acceptable salt thereof can be used to treat such disorders as described above and in U S. Patents No. 6,23 17894; 5,773,430; 5,919,775 and 5,789,395, incorporated herein by reference. In another embodiment, the tetracycline responsive disease or disorder is a skin wound 'Th inovention also provides a method for improving the healing 20 response of the epitelliaized tissue (e.g, skin, mucosae) to acute traumatic injury (e.g , cut, bLm scrp etc.). The method includes using a tetracycli re cormpould cf the invention or a pharmaceutically acceptable salt thereof to improve the capacity of the epitholialized tissue to heal acute wounds, The method may increase the rate of collagC accumulation of the healing tissue. The method may also decrease the 25 proteolytic activity in the epitheliaized tissue by decreasing the collagenolytic and/or gcllatinolytic activity of MMPs. In a further embodiment, the tetracycline compound of the invention or a pharmaccuticadly acceptable salt thereof is administered to the surface of the skin (e. g., topically). in a further embodiment, the tetracycline compound of the invention or a pharmaceuticaly acceptable salt 30 thereof is used to t-eat a skin wound, and other such disorders as described in, for example, U. S. Patent Nos. 5,827,840; 4,704,383; 4,935,412; 5,258,371; 5,308,839, 5,459,135; 5,532,227; and 6,015,804; each of whi ch is incorporated herein by reference in its entirety.
- 39 In yet another ermbodiment, the tetracycline responsive disease or disorder is en aortic or vascular aneurysm in vascular tissue of a subject (e.g, a subject having or at risk of having an artic or vascular aneurysm, etc.). The tetracycline compound or a pharmaceutically acceptable salt thereof may be effective to reduce 5 the size of the vascular aneurysm or it may be administered -o the subject prior to the onset of the vascular aneurysm such that the aIeuirysM is prevented. In one embodiment, the vascular tissue is an artery, e g, the aorta, e g., the abdominal aorta. ln a funher embodiment, the tetracycline compounds of the invention are used to treat disorders described in U. S. Patent Nos 6,043,225 and 5,834449, 10 incorporated herein by reference in their entirety. The compounds of the invention or a phannaceutically acceptable salt thereof can be used alone or in combination with one or more therapeutic agent in the methods of the invention disclosed herein. The language "it) coinbation with" another therapeutic agent or treatment 15 jiclides co-adminisration of the tetracycline compound and with the other therapeutic agent or treatment as either a single combination dosage form or as multiple, separate dosage forms, administration of the tetracycline compound first, followed by the other therapeutic agent or treatment and administration of the other therapeutic agent or treatment first, followed by the tetracycline compound 20 The other therapeutic agent may be any agent that is known in the art to treat, prevent, or reduce the symptoms of a tetracycl ine-responsive disease or disorder. The choice of additional therapeutic agent(s) is based upon the particular teracycline-responsive disease or disorder being treated, Such choice is within the knowledge of a treating physician. Furthermore, the other therapcutic agent iiiay be 25 any agent of benefit to the patient when administered in conbination with the administration of a tetracycline compound. As used herein, the term "subject" means a mammal in need of treatneit or prevention, e.g, companion animals (e.g., dogs, cats, and the like), farm animals (e g., cows, pigs, horses, sheep, goats and the like) and laboratory animals (e.g. rats, 30 mice, guinea pigs and the like). Typically, the subject is a human in need of the specified treatment. As used herein, the term "treating" or 'treatment" refers to oblaining desired pharmacological. and/or physiological effect. The effect can include achieving, - 40 partially or substantially, one or more of the following restilts: partially or totally reducing the extent of the disease, disorder or syndrome; ameliorating or improving a clinical symptom or indicator associated with the disorder; delaying, inhibiting or decreasing the likelihood of the progression of the disease, disorder or syndrome. 5 As used herein, "preventing" or "prevention" refers to reducing the likelihood of the onset or development of disease, disorder or syndrome. "Effective amount" means that amount of active compound agent that elicits the desired biological response in a subject. In one erbodinient, the effective amount of a compound of the invention is from about 0.01 mg/kg/day to 10 about 1000 mg/kg/day, front about 0.1 mg/kg/day to about 100 mng/kg/day, or from about 0.5 mg/kg/day to about 50 ng/k/day. The invention further includes the process for making the composition coinprising mixing one or more of the present compounds and an optional pharmaceutically acceptable carrier; and includes those compositions resulting from 15 such a process, which process includes conventional pharmaceutical techniques The compositions of the invention include ocular, oral, nasaL transdermal. topical with or without occlusion, intravenous (Iboth bolus and infusion), inialable. and injection (intraperiton eally, subcutaneously, intramuscularly, intratumo rally, or parenterally) formulations The composition may be in a dosage unit such as a 20 tablet, pill capsule, powder, granule, liposome, ion exchange resin, sterile ocular solution, or ocular delivery device (such as a contact lens aid the like facilitating immediate release, timed release, or sustained release), pareriteral soiLion or suspension, metered aerosol or liquid spray, drop, ampoule, auto-injector device, or suppository; for administration ocularly, orally, intranasally, sublingualv, 25 parenterally, or rectally, o: by inhalation or insufflation. Compositions of the invention suitable for oral administration include solid forms such as pills, tablets, caplets, capsules (each including immediate release, timed release, and sustained release formulations), granules and powders, and, liquid forms such as solutions, syrups, elixirs, emulsions, and suspensions. Fornis 30 useful for ocular administration include sterile solutions or ocular delivery devices. Forms useful for parenteral administration include sterile solutions, emulsions, and suspensi ons.
.- 41 The compositions of the invention may be administered in a form suitable [or once-weekly or once- monthly administration. For example, an insoluble salt of the active compound may be adapted to provide a depot preparation for inLramuscular injection (e.g., a decanoate salt) or to provide a solution fbr 5 0phIaliic administration. The dosage form containing the composition of the invention contains an effective amount of the active ingredient necessary to provide a therapeutic effect. The composition may contain frorn about 5,000 mg to about 0.5 mg (preferably, from about 1,000 ng to about 05 mg) of a comDound of the invention or salt forn 10 thereof and may be constituted into any form suitable for the selected mode of administration. The composition nay be administered about I to about 5 times per day. Daily administration or post-periodic dosing may be employed. For oral adrninistratioA the composition is preferably in the form of a tablet or capsule containing, e.g., 500 to 0.5 milligrams of the active compound. Dosages will 15 vary depending on factors associated with the particular patient being treated (e.g., age, weight, diet, and time of administration), the severity of the condition being treated, the cornpound being employed, the mode of administration, and the strength of the preparation. The oral composition is preferably formuated as a homogeneous 20 composition, wherein the active ingredient is dispersed evenly throughout the mixture, which may he readily subdivided into dosage anits containing equal amounts of a compound of the invention. Preferably, the compositions are prepared by mixing a compound of the invention (or pharmaceutically acceptable salt thereof) with one or more optionally present pharmaceutical carriers (such as a 25 starch. sugar, diluent, granulating agent, lubricant, glidant, binding agent, and disintegrating agent), one or more optionally present inert phanraceutical excipients (such as water, glycols, oils, alcohols, flavoring agents, preserva lives, coloring agents, and syrup), one or more optionally present conventional tableting ingredientss (such as corn starch, lactose, sucrose, sorbitol, tale, stearic acid, 30 magnesium stearate, dicalcium phosphate, and any of a variety of gums), and an optional diluent (such as water). Binder agents include starch, gelatin, natural sugars (e.g.. glucose and beta-lactose), corn sweeteners and natural and synthetic gums (e.g, acacia and - 42 tragacanth). Disintegrating agents include starch, methyl cellulose, agar, and bentonite. Tablets and capsules represent an advantageous oral dosage unit form. Tablets may be sugarcoated or filincoated using standard techniques. Tablets may 5 also be coated or otherwise compounded to provide a prolonged, control-release therapeutic effect The dosage form may comprse an inner dosage and an outer dosage component, wherein the outer component is in the form of an envelope over the inner component The two components may further be separated by a layer which resists disintegration in the stomach (such as an enteric layer) and permits the 10 inner component to pass intact into the duodenum or a layer whi ch delays or sustains release. A variety of enteric and non-enteric layer or coating materials (such as polymeric acids, shellacs., acetyl alcohol, and cellulose acetate or combinations thereof) may be used. Compyounds of the invention may also be administered via a slow release 15 composition; wherein the composition includes a compoun d of the invention arid a biodegradable slow release carrier (e.g., a polymeric carrier) or a pharmaceutically acceptable non-biodegradable slow release carrier (e.g. an ion exchange carrier). Biodegradable and non-biodegradable slow release carriers are well known Ui the art. Biodegradable carriers are used to form particles or matrices which retail 20 an active age nt(s) and which slowly degrade/dissolve in a suitable environment (e.g.. aqueous, acidic, basic and the like) to release the agent. Such particles degradeiissolve in body uiids to release the active compound(s) therein. The particles are preferably nanoparticles or nanoenulsions (e.g., in the range of about I to 500 nm in diameter, preferably about 50-200 run, in diameter, and most 25 preferably about 100 nm in diameter). In a process for preparing a slow release composition, a slow release carrier and a compound of the invention are first dissolved or dispersed in an organic solvent. The resulting mixture is added intlo an aqueous solution containing an optional surface-active agent(s) to produce an emulsion. The organic so lvnt is then evaporated from the enulsion to provide a 30 colloidal suspension of particles containing the slow release carrier and the compound of the invention, The compound disclosed herein may be incorporated for administration orally or by injection in a liquid form such as aqueous solutions, suitably flavored - 43 syrups, aqueous or oil suspensions, flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil or peanut oil and the like, or in elixirs or simlilIar pharmaceutical vehicles. Suitable dispersing or suspending agents for aqueous suspensions, include synthetic and natural gums such as tragacanth, acacia, 5 alginate, dextran, sodium carboxym erhylcellulose, methylcellulose, poly vinyl-pyrrcli done, and gelatin, The liquid forms in suitably flavored suspending or dispersing agents may also include synthetic and natural gums. For parenteral administration, sterile suspensions and. solutions are desired. Isotonic preparations, which generally contain suitable preservatives, are employed when intravenous 10 admnistrationis desired. The compounds may be administered parenterally via injection. A parenteral formulation may consist of the active ingredient dissolved in or mixed with an appropriate inert liquid carrier. Acceptable liquid carriers usually comprise aqueous solvents and other optional ingredients for aiding solubility or preservation 15 Such aqueous solvents include sterile water, Ringer's solution, or an isotonic aqueous salne solution. Other optional ingredients ine[ude vegetable oils (such as peanut oil, cottonseed oil, and sesame oil), and organic solvents (such as solketal, glycerol, and formy!). A sterile, non-volatile oil may be employed as a solvent or suspeicding agent The parenteral formulation is prepared by dissolving or 20 suspending the active ingredient in the liquid carrier whereby the final dosage unit contains from 0 005 to 10% by weight of the active i ngred ent. Other additives include preservatives, isotoni zers, solubilizers, stabilizers, and pain-soothing agents. Injectable suspensions may also be prepared, in which case appropriate liquid carriers, suspending agents and the like may be employed. 25 Compounds of the invention may bie administered intranasally using a suitable intranasal vehicle. in another embodiment, the compounds of this invention may be administered directly to the lungs by inhalation. Compounds of the invention may also be administered topically or enhanced 30 by using a suitable topical transdermal vehicle or a transdermal patch. For ocular administration, the composition is preferably iu the form of an ophthalmic composition. The ophthalmic compositions are preferably formulated as eve- drop formulations and filled. in. appropriate containers to facilitai administration - 44 to the eye, for example a dropper fitted with a suitable pipette. Preferably, the compositions are sterile and aqueous based, using purified water in addition to the compound of the invention, an ophthalmic composition may contain one or more of: a) a surfactant such as a polyoxyethylene fatty acid ester; b) a thickening agents 5 such as cellulose, cellulose derivatives, carboxvviny polyrners, polyvinyl polyiers, and polyvinylpyrrolidones, typically at a concentration n the range of about 0.05 to about 5.0% (wt/vol); c) (as an alternative to or in addition. to storing the composition in a container containing nitrogen and optionally including a free oxygen absorber such as Fe), an anti-oxidant suoh as butyiated hydroxyanisol, 10 ascorbic acid, sodium thiosulfate, or butylated hydroxytoluene at a concentration of about 0.00005 to about 0.] % (wt/vol); d) ethanol at a concentration of about 0.01 to 0 5% (wt/voi) and e) other excipienIs such as an isotonic agent, buffer, preservative, an d/or pH-controlling agent. The pH of the ophthalmric composition is desirable within the range of 4 to 8. 15 in cenain embodiments, the composition of this invention includes one or more additional agents. The other therapeutic agent may be ay agent that is capable of treating, preventing or reducing the symptoms of a tetracycline-responsive disease or disorder Alternatively, the other therapeutic agent may be any agent of benefit to a patient when administered in combination with the tetracycline compound in this 20 invention. While this invention has been particularly shown and described with references to example embodiments thereof, it will be understood by those skilled in the art that various changes in foTm and details may be made therein without departing from the scope of the invention encompassed by the appended claims. 25 EXE MPLI FICATION The following abbreviations and the terms have the indicated meanings: Abbrev aton/Term Meaning Acacetyl ABN 2-2'-azobis( 2 -methylpropionitrile) aaq aqueous Bn benzl brine saturated aqueous sodium chloride Boc tert-butoxy carbonyl or z-butoxv carbonyl - 45 (Boc)n0 di-ert-butyl dicarbonate B boron tribromide Bu butyl Cbz benzvloxycarbonyl
CH
2
C
2 methvlene chloride CJL;CN or MoCN acetonitrile Cy tricy clohexylphosph Ine dba dibenzylideneacetoiie DIB AL A diisobutylal uminum hydride DIE A A, N-d isoptopyl ethyl amne DMAP 4-(dimethIylamnino)pyridine DME 1 2-di methoxyethane D 1 _, NN-di methylformarnide DMPTJ l.3-dimethyl-3,4-5,6 tctrahydro-2( 1 1)-pyridone DMSO dnethyl sulfoxide EC N- (3 -di methyl aminopropyl )-N -ethvl carbodtmide ESi electrospray ionizat on equiv. equivalent Et ethyl E WO ethyl other EtOAc ethyl acetate h, hr hour HCl hydrochloric acid KHPO4 potassi um hydrogenphosphate ITPLC high performance I iqtiid chromatography HOBt I -hydroxybenzotriazole / ISO JBX 2-;odoxybenzoic acid L DA lithium diisopropylami de LMIDS lithium bis(trimcthylsilyi)aimde .TNP lithium 2.2, 6,6-tetramethvlpiperidida Me methyl NICOT Imethanol Mel methyl iodid -46 min minute methanesulfonyl M4S mass spectrum MT methyl wrt-butyl ether MW molecular weight NaHC& sodium bicarbonate NaO I sodium hy droxide Na2SO 4 sodium sulfate NBS -bromosuccinimide NCS N-chlor osuccinimi de NMR nuclear magnetic resonance spectrometry Ph phenyl Pr propyl s secondary i tertiary RIP reverse phase TMEDA tetiramethyl etl51enedi amine 1-il .'-- -. " ' 5:- -- : " 0 -0 :.. "|'|7:"1- - ||| |7.:" . . -- :--- -, -- - - . _ ... -_ . -...c... . - ..... TBS terr-butyldimethylsilyl TEA triethylamiine TF 'ri1 uoroma thanesul fo nyl TFA tifIloroacetic acid TFAA trIl uoroacetic anhydride THF tetrahydrofuran TI-C thin layer chromatography Ts para-toluenesulfonyi TsOH para-toluenesulfonic acid X antphos 4- 5bis(diphenyiphtosphino)9 9-dimdthylxanthene Example 1. Synthesis of Compounds of Structural Formula (I). T he compounds of the invent ton can be prepared according the synthetic scheme shown in Scheme L - 47 F F F F 1)sBuLi N OCH 0 1)(0C 2 CH CH OH 2MEDA 2' F)PhOH P O Br OPh CHCO 0 0H3O 0 CH 1 0 0 OH 0 1 2 F HC, N'CH$ F H3C N' CH 1 LDA F H H:i H H- TMEDA OH N q HF 2 )enone N' e 4 Oo CH * OFFO OBn Bo: o H O n HoC0 0 r)5 OTBS 4 H /PdI F C.N CHS HC CH H H H H OH HN0 0
/H
2
SO
4 N OH NH N OH 0 O O OHO O O a 7 HI/Pd FH3,C CHa F HC', Cri, F N 0 F N H C, H H Ht H2 NW - NH HS~ 6 o~- 0 O I109 or F H0 H 0 N ~ OH 2 HNH 2 Scheme 1 Specific conditions for the reactions depicted in Scheme 1 are provided in 5 the following examples. Compoumd 1 F C H C Ha0 0 1 - 418 To a TF [F solution of 5-fluoro-2-mothoxybenzoic acid (500 mg, 2.94 nmol, Al dri ch 523097) cooled at -78 "C was added a'THF solution of s-BuLi (4.60 mL, 1.40 M, 6,44 mmol, 22 equiv) an d TMFIDA (0.97 ml., 6.47 nuno l, 2.2 cquiv). The reaction was stirred at -78 'C for 2 h Mel (I .10 ml., 17.64 mmol (.0 equiv) was 5 added to the reaction mixttur'e dropwise. The reaction was allowed to warm to 25 0C over 1 hi and stitred at 25 "C for I h. NaOHF (6 N, 20 mL) was added. 'The resulting mixture was extracted with t-butylmeothyl ether (20 mL - 2). The aqueous layer was acidified with HCI (6 N) to p1 1 and extracted with EtOAc (20 ml x 4). The combined EItAc extracts were dried (Na2SO 4 ) and concentrated to give 510 mg of 10 crude product 1 'H N'MR (400 MHz, CDC 3 ) 6 7.06 (dd,J = 9.8, 8.5 Hz I H), 6.75 (dd, J= 98, 3.7 Hz, 1 I), 3.86 (s, 3 H), 2.34 (dJ 24 -Hz, 3 H), MS (ESI) mc 185.12 (MIT). Compound 2
CH
3 0 0 15 2 Oxalyl. cioride (0.95 mL, 11. 10 mmol, 5,5 equiv) was added to CH 2 C1 solution (1 5 mL, anhydrous) of 1 (5 10 mg, 2.00 mmol). DMF (0.1 miL) was added to the resulijng mixture. The reaction was stirred at 25 'C' for 1 h aid concentrated. The 20 resuhing solid was re-dissolved in. 15 mL of anhydrous CH 7 Cl 2 . Phenol (520 mg, 5.50 mnol, 2.8 equiv), DMAP (670 mg, 5.6 nmmol, 2.8 equiv), and triethylamine (1.90 mL, 13,90 mnmol, 7 0 equiv) were added to the reaction mixture, The reaction was stirred at 25 'C for 12 h and concentrated. EtOAc and 1120 were added to the residue The organic layer was washed with NaOHl (I N), H-O, and brine, dried 25 (Na 2 SO4), and concentrated, Flash chromatography on silica gel (40:1 )exanest/OAc) yielded 400 mlg of compound 2 (52% for 2 steps): '14 NMR (400 Mliz, CDCL) 5 7.47-7 41 (m, 2 H), 7.31-7.24 (m, 3 H), 708 (dd, J - 92, 9.2 Hz, I H), 6.77 (dd, .= 9,2, 3 7 Hz, 1 H), 3.88 (s, 3 H), 2.36 (d. J= 2.3 Hz, 3 1i)i; MS (EST) az 261 .12 (M-H), :30 - 49 Compound 3 F C H 3 O1Ph OH 0 3 BBr- (.85 niL, I M, 1.85 nmol, 1 2 equiv) was added to a CH2Cl 2 solution 5 (8 iL) of 2 (400 ng, 154 mmol) at -78 'C. The reaction was stirred from -78 'C to 25 'C for 1. i, quenched with saturated NaHCO and concentrated, EtOAc and 1120 were added to the reaction mixture. The aqueous layer was extracted with EtOAc. The combined EtOAc extracts were dried (Na 2
SO
4 ) and concentrated to yield 360 mg of crude 3: 11 NMR (400 MHz, CDClI) 8 10.66 (s, I H), 7.50-7.44 10 (m, 2 14), 7,36-731 (im , H), 7.26-7.18 (i, 3 H), 6.86 (dd, J= 9.3, 4.9 Hz, 1 11) 2.60 d,1 J: 2.4 lIz, 3 11); MS (ESI) nz 245.11 (M-H). Compound 4 F >cHS Oph Oco 0 4 Boc 2 O (350 mg, 1.60 minmol, 1 1 equiv) and DMAP (20 mg, 0 16 mmol, 0.1 equiv) were added to a CI ClA solution of crude 3 (360 mg). The reaction was stirred at 25 0 C for 1.5 h and concentrated Flash chromatography on silica gel (35:1 hexanes/EtOAc) yielded 400 mg of compound 4 (94% for 2 steps): 'H NMR (400 20 MEz, CDCk) 8 7.46-7.41 (m,, 2 HI, 7.31-7.23 (m, 3 H) 7.18 (dd, J= 8.8, 8.7 Hz, 1 H) 7.10 (dd, J= 8.8, 4.4 Hz, I H), 2, I (d = 2.3 Hz, 3 H), 1.44 (s, 9 H); MS (ESI) mI 345. 1 8 (N-H). Compound 5 H H - O~ BocO a OH 0 OBn 25 6 OTBS - 50 A THF solution (6 mL) of 4 (487 mg, 1.40 mmol, 2.0 equiv) was added To a TH-F solution (5 mL) of LDA (6.30 nL, 10% wt. 4.20 mniol, 6.0 equiv) and TMEDA (1 70 mL, 11.20 rumol, 16.0 equiv) at -78 'C, The reaction was stirred at 5 -78 0 C for 5 rmin A TIF solution of enone (339 mig, 0.70 mrnol, 1.0 equi) was added to the reaction mixture dropwise The reaction was stirred from -78 "C to 25 'C fbr I i, cquerched with saturated NH4CI, and extracted with EtOAc. The combined EtOAc extracts were dried (Na 2 S04) and concentrated to yield the crude product. Preparative reverse phase HPLC purification on a Waters Autopurificalion 10 system using a Sunfire Prep C 18 OBD column [5 min, 19 x 50 mm; flow rate, 20 mL/niia; Solvent A: [120 with 0.1% HC021; Solveit B: CH 2 CN with 0.1% 1C0 2 11 injection volume: 4.0 niL (CICN); gradient: 80-100%B over 15 minl; mass-directed fraction collection]. Fractions with the desired MW, eluting at 6.3 8.0 min, were collected and concentrated on a RotaVap at rt to remove most of the 15 acetonitrile. The resulting mostly aqueous solution was extracted with EtOAc. The combined EtOAc extracts were dried (NaSO4) and concentrated to give 185 nig of pure 5 (35%): lH NMR (400 MHz, CDCl) d 15.67 (s, I H), 7.51-7.46 (m, 2 H), 7 39-7.29 (m, 3 iH), 7.21 (dd, J- 8.9, 8 9 Hz, 1 H), 7.03 (dd, J = 8.9, 4.0 Hz, 1 1), 5 34 (s, 211), 3.93 (d, J= 104 Hz, I H), 3.30-3.21 (m, 'I [), 3.10-3.00 (m, I H), 20 2 57-2.41 (i 3 T4), 2,48 (s, 6 H), 2.17-2.12 (m, I H), 1.53 (s, 9 H), 0,82 (s, 9 H), 0 26 (s, 3 t) 0.12 (s, 3 H): MS (ESI) ur 735.45 (M+ 1) Compound 6 FInC. -OWK F H U H H r 1N OH 0 jPH Jn 25 Aqueous fTP (3 nil, 48%) and TFA (4 pL) were added to a ChCN solution (7 mL) of 5 (21 10 mg, 0.29 mmiol) in a polypropylene tube at 25 'C. The reaction was stirred at 25 .C for 18 i. The resulting mixture was poured into an aqueous solution of KJMPO(21 g, dissolved in 150 mrL water). The rrixture was extracted with EtOAc. 30 The combined EtOAc extracts were dried (Na2SOI) and concentrated to yield - 51 180 mg of crude: 11H NNR (400 M1zL CCl 3 ) 5 14.64 (s, I H),, 11.47 (s, I .). 7.49-7.45 (i, 2 H), 7.39-7.32 (in, 3 f), 7 14 (dd,.J= 9.2, 8.8 Hz, I H), 6 77 (dd, J= 9.2, 4.3 Hz, 1 H), 5.36 (s, 2 H), 3.68 (d, J= 3.7 Hz.1 H), 3.09 (dd, ]- 15.6, 4 6 iz, I .), 3.02-2.92 (m, I H), 2.84-2.79 (m, 1 H) 2 49 (s, 6 H), 2.34-2,22 (i I H), 2,09 5 2.02 (n, 1 H), 1.55-1.44 (m, 1 H); MS (ESI) mz 521.30 (M.+H). Compound 7 F H3CN CH3 |K HH OH. J NH2 OH 0 05H 7 10 Palladiurn on carbon (35 mg. 10 wt%) was added to a MeOH/dioxane sohition (4 mL/4 mL) of crude 6 (180 ag). The reaction was pured with hydrogen and stirred under H2 (balloon) at 25 C for I h The reaction mixture was filtered through a small Celite plug, The filtrate was concentrated to yield the crude product Prcparative reverse phase HWLC purification on a Waters Autopurification system 15 using a Phenomencex Polymvnerx 10 - RP-1 100A column [10 pm, 150 x 21.20 mm; flow rate, 20 inLmin; Solvent A: 0.05 N HC/water: Solvent B: CH-1 3 CN; injection volume: 4.0 nL (0.05 N HC/water); gradient: 0-100% B over 15 min; mass directed fraction collection]. Fractions with the desired MW, eling at 6.4-8.2 mii, were collected and freeze-dried 1o yield 51 mg of compound 7 (4 1% for 2 steps): 4H 20 NMR (400 M.z, CD 1 OD) 5 7.26 (dcd, J= 9.2, 9.2 Hz, 11), 6.80 (dd, J = 9.2, 4.3 Iz, 1 H.), 4.09 (br s, i H), 3.14 (dd, J 1 L5.0, 4.6 Hz, I H), 3.04 (s, 3 H), 2.96 (s, 3 l), 3.09-2.91 (m, 2 1), 2,3 1-2.18 (, 2 11) 1.68-1.56 (i, 111); MS (ESI) m/z 433.28 ( 1). 25 Compound 8 F
H
3 C COH 3 H 0-9H OHO 0 H O 0 8 - 52 A mixture of HNO 3 (8.5 pL, 69%) and H 2
SO
4 (0.5 mL) was added to a 112SO4 solution (1 mL) of 7 (51 mg, 0.12 mmol) at 0 C. The reaction was stirred at 0 C for 30 min. The resulting mixture was added dropwise to vigorously stirred diethvl ether (60 mL). The suspension was filtered through a small Celite pad and 5 washed several times with more diethyl ether. The Celite pad was then eluted with MeO! I until the fluent became colorless. The yellow MeCOH eluent was collected and concentrated under reduced pressure to afford crude 8: 'I NMR (400 MHz, CDOD) 8 8,03 (d, I= 8.5 Hz, 1 U), 4.09 (br s., 1 H), 3.50-2.97 (n, 3 H), 3.04 (s, 3 Hi), 2 96 (s, 3 l), 246-2.36 (in, 111), 2.29-2.20 (in, I H1), 1L71-1.59 (n, 1 .H) MS 10 (ESI) mw 478 20 (M+H). Compound 9 F Hac .o HOH -, OH C OH'0 0 9 15 Palladium on carbon (12 mg, 10 wt%) was added to a McOl solution (4 mL) of crude 8. The reaction was purged with hvdrogen and stored under H (balloon) at 25 'C for 2 h The catalyst was filtered off with a small Celite pad. The filtrate wkVas concentrated to y eki crude 9. Preparative reverse phase I IPLC pLrification on a Waters A utopurifi cation system using a Phenomenex Poly merx 20 10 P RP-1 IOA column [ 10 n, '150 21.20 mm; flow rate, 20 mL/min; Solvent A: 0.05 N 110C1 Solvent B: Cl CN; injection volume: 4.0 mL (0.05 N HC/water) gradient: 0-100% B over 15 rnin; mass-directed fraction collection]. Fractions with the desired M\W, cluting at 5.0-6.6 min, were collected and freeze-dried to yield 43 mg of pure 9 (81% for 2 steps): 1H NNIR (400 MIlz, CD-0D) 5 7.43 (d, /= 8 5 25 Hz- I H), 4.11 (br s, I H), 3.22-3. 16 (m, I i ), 3 15-3.08 (n, I H), 3.06-2.95 (im, . H), 3.04 (s, 3 11), 2.96 (s, 3 14), 2.40-2.31 (m, 1 H), 2 28-2,21 (m, 1 H), 1.71-1.59 (n I fl); MS (ESI) ml: 448.24 (M-H), ConmpoUid 11 30 - 53 F H3N~ H H H <OH HC N N NH H3C'H 3 H 0 HO 0 0 11 2-i-Butylaninoacetylchloride hydrochloride (4. mg, 0.022 mmol, 2.0 cquiv) was added to a DMF solution (0 1 miL) of 9 (5 mg, 0. 01 1 mumol) at 25 "C. The 5 reaction was stirred at 25 &C for 30 min, The reaction mixture was diluted with 0.05 N HC (2 mL) and injected into a Waters Autopurification system equipped WihI a Phenornenex Polymerx 10 p RP-1 100A colun (10 um, 150 x 21.20 mm; flow rate, 20 mL/min; Solvent A: 0.05 N HC!, Solvent B CH-CN; gradient: 0--l 00% B over 20 min; mass-directed fraction collection]. Fractions with the desired MW, cluting at 10 6.4-7.0 rin, were collected and freeze-dried to yield 3.9 mg of pure 11 (62%) 'H NMR (400 MHz, C.D-OD) 6 8.25 (dJ= 11.0 Hz, I H), 4.11 (br s, 1 H), 409(, 2 H), 3.22-2.86 (m, 3 H), 3.05 (s, 3 H), 2.97 (s, 3 H), 2.33-2.20 (m, 2 H), 1.69-1 57 (n, 1 Hl), 1 42 (s, 911), MS (ESI) mz 561.39 (Ni-H). 15 Compound 32 H, k G'CH 3 H H Ca HO N NH 2 OH 0 OHO 0 32 Anhydirous Na 2 CO (16 mog, C 15 tymnol 5.5 equiv) was added to an 20 anhydrous DMPU!acetonitrile (.150 p/50 l) solution of 9 (12 mg, 0.027 mmol). Brornoacetyl bromide (2.8 yL, 0.032 tmrol, 1.2 equiv) was added to the mixture. The reaction was stirred at 25 'C for 10 min. LC/MS analysis indicated complete formation of intermediate 10. Azetidine (36 uL. 0.54 inmo, 20 equiv) was added to the reaction mixture, The reaction was stirred at 25 "C for 2 h. The reaction 25 mixture was concentrated and acidified with HC (0.5 N to MeOH. 0 7 mL). The reSulting mixtice was added dropwise to vigorously stirred dicthyl eter (10 mL). The suspension was f itered through a small Celite pad and washed several times - 54 with more diethyl ether. The Celite pad was then eluted with MeOH until the eluet became colorless, The yellow MeOH fluent was collected and concentrated under reduced pressure to afford crude 32. Preparative reverse phase IPLC purification on a Waters Autopurification system using a Phenomenex Polymerx I V RP- I 5 1 OA colu mn [1.0 um, 1-50 21.20 mm: flow rate, 20 iL/m:in Solvent A: 0.05 N 111; Solvent B: CJALCN; injection volume: 2.0 mL (0.05 N HCl/water); gradi ent: 0-420% .B over 30 mirn mass-directed fraction collection]. Fractions with the desired MW, ehoting at 10.8-12.5 min, were collected and freeze-dried to yield 2.0 mg of pure 32: 'H NIMR (400 MHz, CD-,OD) 6 8.18 (d, .1 i 10 Hz, I H.), 4.41 10 4.31 (in, 2 H), 4.32 (s, 2 11), 4 24-4.13 (in, 2), 4.08 (br s, I I). 3.1 -2.86 (in, 3 H), 3. 03 (s, 3 H), 2.95 (s, 3 H), 2.71-2 57 (m, I H), 2,54-2.42 (m, I H), 2.33-2.16 (m 2 H), 1 69-1,57 (m, I H-); MS (ESI) mt' 545.20 (Mi-H). Compounds 12 -31 and Compounds 33-46 are prepared similarly to Compounds 15 11 or 32, substituting the appropriate acy I hal ide for 2-t-Butylaminoacetvlchloride in the synthesis of Compound II or cyclic amine for azeti dine in the synthesis of Compound 32 Compound 12 20 H H HC H3CN OH 0 O 12 H NIR (400 N1Viz, CD 3 0D) 6 8.23 (d, 1 11 2 H7, I1 H), 4.08 (s, 3 H), 3.17-2 97 (m, I 1I ). 2,31 (dd, - 14.8, 14.8 Hz, 11H), 2 24 (ddd, J - 14.0, 5.2, 2.8 25 1 z, I H). 1,79-1 72 (an, 2 H), 1.66 (ddd J= 13.6, 13.6, 13 6 Hz.1 .), 1.05 (t, J 7 2 Hz, 3 H); MS (ESI) m/z 547 2 (M+H) Compound 13 -55 F HiC, CH3 H HOH F Nn~. ~ OH 0 OH C 0 13 11 NMR (400 MHz, CD 3 OD) 6 8 23 (d, J 11 2 Hz, 1 H, 4,08 (s3 H), 3.16-2.97 (m, il H), 2.30 (Idd, J= 14.8, 14.8 Hz, I H), 2.24 (ddd, J - 14.4, 5.2 2 8 5 Hz, I1 H), 1.75-1,69 (m, 2 H), 1,66 (ddd, J= 13.6, 13,6, 13.6 Hz, I H), 1.49-1.41 (m 2 1). 1,01 (ItJ 7 2 H z, 3 I) MS (ESi) wm'; 561.2 (M+Hi). Compound 14 F 61- CH3 H3Cs/H......0H H H H N <H H H O OH 0 0 10 14 H1 NMR (400 Ml 1z, CD30D) 6 8.21 (d, J= 11 2 Hz., 1 H), 1.08 (s, I IT), 4.06 (s, 2 1), 3 16-2.96 (n, I H), 2.28 (dd, J= 14.8, 14.8 lz, I H). 2.22 (ddd, 14.4, 52, 2.8 Hz, IH.), 1.77- 17 1 (m, 2 H), 1.66 (ddd, - 14.0, 14.0, 14.0 lHz, I 1.1), 15 1.43-1.35 (m, 6 H),0 93 (t,J- 7.2 Hz, 3 H); MS (ESI) m z 589.2 (M-H). Compound 15 F HsGc.N'CH3R H H H .- fr H . -. CHJ OH 0 OHU C 15 H1 NMR (400 M-Hz, CDOD) 8 8.23 (d, J= 10.8 Hz, I1), 4.09 (s, 2 H), 4.07 (s, I H), 3.15-2.95 (i, II H), 2.29 (dd, J= 14.4, 14.4 Hz, 1 H). 2.25 (ddd,, = 14.4, 5.2, 2.8 Hz, 11), 1.66 (ddd, J= 13.2, 13.2, 13.2 Hz, 111), 1.10 (s, 9 [1); MS (ESi) mz 575.2 (NIH).
- 56 Compound 16 F HC, DCH, OH ,H -c NN H3C N CH, OH 0 o o 16 5 i HNMR (400 Mz, CD 3 0D) S 8.24 (dtJ= 11.0 Hz 11H), 4.08 (. 2H), 4.01 3.89 (n, I H), 3.50-3.42 (m, 1Hf)7 3.20-2.84 (m 911), 2.30 (at,J= 1.4.7 Hz, IH), 2.23-2.] 5 (n, 1H), 1.70-1.58 (m, I [1), 137 (d, J = 6.7Hz, 6H); MS (ESI) m z 547,25 (M+H). 10 Compound 17 F HSC N-CH, H H N2 H [r <jT N OH 0 OH 0 O 17 H NMR (400 MHz, CDOD) 6 824 (d, J= 11 0 Hz, I F), 4.20 (s, 2 H), 15 4 09 (br , 1 H), 3.19-3. 13 (m, I H), 3.12-2.89 (m, 2 H), 2.89-2.38 (m 1 H), 3.04 (s 31H), 2.96(s, 3 1), 2 35-2.19 (m, 2 H) 1, 71-1, 59 (m, 1) 0.95 (br s, 2), 0.94 (br s, 2); MS (ESI) miz 545,37 (M+H) Compound 18 20 F Hf N CF 3 H..OH H Y! N NH2 OH C OH C O H NMR (400 MHz, CD 3 0D) 6 8.23 (d, J 10 8 Hz, I H), 4.09 (s, 3 .H), 3.68-3.61 (m, 1 1-1), 3.16-2.97 (m, 9 .), 2.29 (dd,J 1 . I 4.4 1 z, I H), 2.25 (ddd - 57 J= 14.4, 5.2, 2.8 Hz, I H), 2.20-2.12 (m 2 IT), 1.98-1.91 (in, 2 H1), 1.75-1.68 (in, 4 H), 1.66 (ddd, J- 13.6, 13.6, 13.6 1, 1 H), MS (ESi) mz 573. 1 (MtTI) Compound 19 F OC.
CH
[ H H H Q OH HO' NNH OH 0 OH % 19 'I NMR (400 MHz, CD 3 OD) 4 8.26 (d 1 11.0 Hz, 1 H), 4.09 (br s, 3 HT), 3. 19-2.93 (n, 511), 3.04 (s, 3 11), 2.96 (s, 3 H) 2.35-2 26 (mA, 1 H), 2.25-2.18 (m, 1 10 H), 2.14-2.02 (m , 1 H), .' 1-1.59 (n, I H), 1.07 (d, = 6.7, 6 EI); MS (ES) m/z 561.24 (M-H). Compound 20 F ' OH .NZ N NH2, OH 0 OH 0 o 15 20 I NNIR (400 MYTz, CD01)) 6 8 24 (d, J= 11. Hlzs 1 H), 4. 1 (s, 2 f), 4.08 (br s, 1 H), 3 22-2.92 (n, 5 H), 3.03 (s, 3 H), 2.95 (s, 3 H) 2.33-2,24 (m, 1 H.), 2.24-2.17 (n, FH), 1.69-1.58 (m ,1 1), 1.17-1.07 (m, 1 i), 0.77-0.71 (i, 2 H), 20 0.46-0.40 (m, 2 H); MS (ESI)n r 559.23 (M+H). Compound 21 Fo- H NU OH 0 oPW o 21 - 58 1 H NMR (400 MHz, CmOD) 5 8.2,5 (d=1.0 Hz. IH). 4.12 (s, 2H), 4.09 (s, 11), 3.72-3.67 (m, 2H), 3 43 (s, 3H), 3.1.9-2.92 (m, 1 M), 2.35-2.18 (m, 2H) 1.71-1.58 (m, IH); MS (ESI) m/z 563.23 (M+H), Compound 22 F HN CHI S HOH
F
3 CO. N gM~ 4 -y #.. ~ NIH 2 OH 0 OH 0 0 22 10 H11 NMR (400 M-fRz, CD;FOD) 5 8.25 (d, = I I.0 9z, 1I.), 4.22 (s, 2-H), 4. 4 4.05 (m 3 H), 3 18-2,84 (n, 911), 2.34-217 (m, 2H), 1 70-1.57 (], ilt) MS (ESI) mnz 587.28 (M 1 FT). Compound 23 15 F N HOH H H0 040 0 23 H NMR (400 MHz, CDOD) 6 8,24 (dJ 1. 0 Hz, ill), 4.24 (s, 21), 4.09 (s, 1), 3. 14-2.93 (m, 1SH), 2.24-2.18 (m, 2H), 1, 65 (dt, J = 13.4, 11.6 Hz, I); MS 20 (ES1) m/z 533.17 (M+H). Compound 24 F N-H 9H3 OH 0 OMO 0 24 - 59 1H NMR (400 MHz, CD-OD) 6 8.23 (d, ,.= 10.4 Hz, lH) 4.29 (d, .1- 16.5 Hz, I H), 4.18 (d J= 15.9 Hz, 1H), 4.09 (s, 1H), 3,19-2.89 (m, 14H), 2.36-2.17(rn 2.H), 1.70-1.58 (m. 1), 1,38 (tJ = 7.32 Hz, 31-): MS (ESI.) m/z 547.25 (M+H). Compound 25 F H3C CH, H-OH HIC N - 'k NH OH 0 O 4 H a 26 10 11 NMR (400 MHz, CD-,OD) 5 8.21 (d, J 10.8 Hz, I H), 4.25 (s, 2 H), 4.10 (s, 1 H), 3 35 (t, J= 7.2 Hz, 3 -), 3,34 (t.= 7 2 1Hz, 3 I), 3.13-2.99 (m. 9 h), 2 31 (dd. .1 = 14.8, 14.8 Iz, 1 H), 2 27 (ddd J 14 8, 5.2, 2.8 Hz, tH), 1.78-1.74 (m, 2 11), 1.68 (ddd. = 13.6,1 136,136 Hz, 1 ) 1.38 (t, - 72 Hz, 6 H); MS (E SI) 561.2 (M+- H). 1 5 Compound 26 F H H H HC N NH OH ON ) 0 28 20 1 .NMit (400 MHz, CD30D) 6 8.23 (d, / 11.2 Hz, 1 H), 4.10 (s, 3 H), 3.16-2.96 (m, I I H), 2.31 (dd, J= 14.4, 14.4 Hzi 11), 2 24 (ddd, .! = 14.4, 5,2, 2.8 17, 111), 1.78-1.71 (m, 2 HI), 1.66 (ddd, J= 14.9, 14,0, 14.0 1z1, 1[1), 1.45-1.38 (m, 4 H),0.98 (t,J = 72 Hz, 3 14) MS (ESI) mi'z 75 2 (M4H) 25 Compound 27 - 60 F Hoc C. C H3 H H1 H S'N N NH OH 0 OH 0 0 27 I I NMR.(400 MHz, Cl1OD) 5 8.24 (dJ 10.8 Hz, 1), 1.09 (s, 3 H), 3.59 (tJ= 5.6 Liz, 2 H), 3.40 (s 3 H), 3.23 (t. J= 5.6 Hz, 2 I), 3.15-2.94 (m. 9 ll) 5 232 (dd, J = 15.2, I 5.2 Hz. .1 H), 2.24 (ddd, J [4 0. 5 2, 2 8 Hz, 11), 2.08-2 02 (m, 2 11), 1 66 (ddd.J- 15.2 15.2, 15.2 Hz, 1H); MS (ESI) m/z 5772 (M+ II) Compound 28 F H3C. N-C; J H H YCHNH N1 NT 1 OH 0 OH O 0 10 23 'if NMR (400 MlHz, CD 3 OD) a 8.24 (d. J= 108 Hz, 1 H), 4.32 (., J- 8_ 0 Iz, I H), 4 21 (d, J-- 8.0 Hz, 1 H), 4.10 (s, 1 H), 3.18-2.99 (ma, 9 H), 3.01 (s, 3 H), 2.33 (dd, t4. 14.8 Hz, 11H), 229 (ddd, J= 15.2, 5.2, 2.81Hz, ]H), 178- L74 15 (m, 2 H1), 188- 181 (m 2 H), 1,68 (ddd,.J - 15.6, 15.6, 15.6 Hz, 1 H), 1.08 (t,.. 7.2 1 1z, 3 1); MS (ES) m 561,2 (M+H). Compound 29 F NCH N H? OH HsC N -.. N N H OH 20 29 'H NMR (400 MHlz, CDGOD) a 8.25 (3.d = 10.8 1, 1 1), 4 i0 (s, 3 H), 3.18-2.98 (m, 11 H), 2.31 (dd, J/l 148, 14.81-1z, 1 H1 2.26 (ddd, J- 14.4, 52,2 8 - 61 Hz, I I), 1.78- 1.74 (n, 2 1I), 1.66 (ddd, ,J = 13.6, 13.6, 13.6 Hz, I H), 1.42-1.30 (m, 8 H-),0.94 (t, J= 6,8 Hiz 3 TT); MS (ES1) mWZ 603.2 (Mt-H) Compound 30 5 F CH H OH 0Oh O 30 . H NMR. (400 MHz, CDsOD) 6 8.32 (d, J 10 4 Hz, I H), 7.38-7.34 (n, 2 l), 7. 1-7.06 (m, 3 11). 417 (s, 2 H4) 4,10( , I H) 3.18-2.99(m, t I H), 2.29 (dd, J 10 = 15.6, 15.6 Hz, I H), 2.25 (ddd, J= 14.8, 5.2, 2.8 Hz, Il), 1 66 (ddd,J= 14.8, 14.8, 14,8 Hz, L U): MS (E SI) mz 58 1 1 (M+H. Compound 31 F HN H H N ]ONH OHO HO O 15 31 1H NMR (400 MHz, CD 3 I)) 6 8.25 (d J 10. 8 Hz, 1 ), 4 36 (d,! = 8,0 I z, I Hi), 4.21 (d, J= 8.0 Hz, I H). 4.10 (s, I H), 3.68-3.61 (n, 1 1.1), 318-2.98 (mi, 9 H). 3 00 (s, 3 1H), 2.29 (dd., J= 14.4, 14.4 1 Lz, 1 H.), 2.20-2.10 (m, 3 11), 1.96-1 89 20 (m, 2 H), 1.78- 1.68 (m, 4 1), 1.66 (cddd, J= 14.4, 14.4, 14.4 Hz, 1 H): MS (ES!) m/z 587.2 (MiH). Compound 33 -62 H H HOH H O 0 C , OHa 0 33 -I NMR (400 MHz, CD 3 OD) 5 8.20 (d, J 11.0 Hz, 1 11), 5.51-5.33 (m, 2 H), 4.71-4.37 (i, 4 H), 4.40 (s, 2 H), 4.06 (br s, 111), 3.17-2.92 (mn, 3 1), 2,99 (s, 6 5 H), 233-2.24 (m, 1 H), 2.23-2.16 (m. 1 H), 1.70-1 58 ( ) ,1 H-) MS (EST) ma 563.20 (M+H). Compound 34 hi0 H . .... ' x j N H2 OH COH O 10 34 1H NMIR (400 MHz, CD;OD) 6 8.22 (d, J= 11,0 Hz, 1.11), 4 33 (s, 2H), 4. 10 (s, 1H), 3.83-3.72 (i, 2H) 3 25-2 89 (m, 121), 2.32-2.00 (i, 61H), 1.69-1.56 (n, 1iU); MS (ESI) m/FZ 559.39 (M+Hl) 15 Compound 35 C- OH NN HHH H CH 0 OH 0 0 35 20 H 1NMR (400 MHz CDOD) 8 8.25 (d, = 1L Hz, 111), 5_54-5 3(, I$), 4.39-4.20 (in, 2H), 4 09-4.01 (m, 11H), 3.40-330 (m, 211), 3,09-2,89 (mn, 1214), 2.50-2,34 (), 2, 2 34-2,25 (in, 1N), 2 24-2.16 (m, 11:), 1.71 -1.58 (m, I"H); MS (E Si) mn> 577.32 (M I 1H).
- 63 Compound 36 F
H
3 C CHo H QH OH 0 OH H 0 35 5 H NMR (400 MI-iz, CD 1 OD)5 8.23 (d.J= 10.4 Hz, 1), 5.57-5.37(m, 1H), 4 47-4.33 (m, 2H) 4 15-3.87 (m, 2 3.72-3 .40 (m, IH) 3.17-2.83 (m, 1211), 2.55-2.34 (m7 211), 2.33-2 18 (m. 211), 1 69-1.57 (m, Il) MS (ESi) nz 577.37 (Md+I). 10 Compound 37 H H
H
3 0C -- Q OH H3C 37 H NMR (400 M4Hz, CThOD) 5 8,28 (d, J= 10 1, I H), 4 08 (s, 11-1), 4.00 15 3.91 (m, 2H), 3.09-2.57 (m, 181H), 3,26-3.18 (m, 311), 2.49-2.34 (m, 2H), 2.35-2.06 (m,7 2H), 1.72-1.59 (m, I T) MS (ESI) m i 602 37 (M+-H). Coimpound 38 F H CH H H, OH 0 OH C 20 38 1 H NMvR (400 MHz, CD,0D) 6 8.24 (dJ - 1 0 1 z, 1H), 4.46-4.32 (n, 211), 4.26-4.16 (i, Il). 4.08 (s, 21), 4.00-3.72 (M, 21), 3.8 -2.91 (m, 1611), 2.68- - 64 2.56 (n, 1H), 2.51-2.39 im 111), 2.34-2.24 (m. 1H) 2 23-2.17 (mn, IH).170-i.57 (m, 111); MS (ESI) m/z 602 37 (M+ HI-I) Compound 39 F-N F N H HH HOyNH2 OH 0 OH 0 0 39 H NtR (400 MHz. CD 3 OD) 8 8,23 (d J= 11.0 HzI- IH), 4.62-4.54 (m, I [) 4,48-4,24 (m, 2H), 4.08 (s, ]11), 3,99-3,69 (i, 3H), 3.50-3.40 (i, H), 3.17 t0 2.90 (m, 9[l), 2 44-2,11 (m, 41-), 2.10-2.00 ( 1 H), 1 69-1.56 (m, I H); MS (ESI) mnz575.27 (M+H), Compound 40 F - HC. C-, HC H Ht
NH
2 15 40 H NNIR (400 MHz, CD 3 OD) 8 8.23 (d-J = I 0 Hz, I1H), 4,62-4.54 (m, I H), 4.50-4 38 (m, 1.), 4.37-4.27 (m, i H1), 4.10 (s, i), 3 99-3.70 (in, 311). 3.50 3.-0 (m 111), 3.24-2.84 (m, 911), 2.40-2,11 (m, 4H), 2.10-2.01 (m, 11H) 170-L57 20 (m, 1 H); MS (E SI) m/Z 575.33 (M +H). Compound 41 F N H H 'N H, 41 0 OH 41 - 65 II NIR (400 Mz, C D 1 OD) 3 8.25 (d,i - I LO Hz, I H). 4.54 (d, J= [6.5 Hlz, I H), 4.26 (d,. J 15,9 Hz, 1.1H), 4.09 (s, 1.H), 3.95-3.81 (m, 21), 3.81-3.75 (m, 5 11H), 369-3.62 (in. 114). 3.35 (s, 3H), 3.23-2.92 (in, 91), 2.35-2.04 (i, 611), I 91 1.80 (i, 1f), 1.71-1.59 (im, i1H); MS (EST) m;a2 603.35 (M+TH), Compound 42 F N H HO N -Z OH NH 2 H OHO Hc 0 10 42 'I] N.MR (400 MHz, CDOD) & 8.24 (d,J= 11.0 Hz, iMH), 4.55 (d,J= 16.5 Hz, 111), 4 27 (d. J -- 16.5 Hz, lid), 4. 10 (s, iH), 3-95-3.82 (m, 2H), 3.8 1-3.75 (in, 1H), 3.70-3.63 (n, [H), 3 38 (s 31-), 3.20-2,92 (m, 91), 2.35-2.02 (m, 61), 1.92 15 1 80 (m, 1 1, .70-1.58 (m, 1l); MS (ESI) m z 603.41 (M"'H). Compound 43 H H -" OH - N NH? H H'K. Oh0 01- 0 43 20 11 NMR (400 MHz, CD;OD) 6 8.22 (d, -= 1 1.0 Hz, 1H), 4.19 (s, 211), 4.09 (s, 1H), 3.65-3.58 (, 21-) 3 19-2.92 (m, 1014) 234-2. 18 (m, 21, 2.02-1 79(m. 61), L.69-1.50 (m, 2H); MS (EST) m4 573 35 (M+H). 25 Compound 44 -66 F H,, CHs H H N NH 2 H THI OH o O H0 44 H NMR. (400 M T CD)OD) 58.24 (d, J 11 0 Hz, H), '1.28 (s, 211) 4.03 4.00 (m, 211), 3.94-3.81 (m, 2H), 3.68-3.55 (i, 2HF) 3,20-2.88 (m, 1211), 2.36-2.18 5 (m, 2H), 1.71-1 57 (m, 1H) MS (ES!) nz 575.37 (M+H). Compound 45 H O r 0N ~ NH, CH 0 OH 0 0 45 10 'H NMIR (400 MHz, CDOD, 2:1 mixture of d astereomers) 3 8.25 (d + d, J S11 0 Hz, 11), 4.29, 4,24 (s + s, 2H), 4.08 (s s, I H), 4,01.-3.92 (in+m, 311), 3.20 2.62 (m I- m, 1311), 2.35-2.16 (m + m, 3 1); 183- 1.46 (m + n, 51); MS (ESl) Fyi 599.36 (M+Hf). 15 Compound 46 F HN H H .- Nr ~ N H 2 H OH 0 OH u C 46 20 'H NMR (400 MHz CD 3 OD) } 8.29 (d. = 11.0 1 1H), 7.41 (s, 5f). 4 50 4.37 (m, 21), 4 05 (s, 111), 3.95-3.81 (m, 2H), 3,40-3.37 (m, 111), 3,24-3,15 (i, 3H). 3,1 0-2.70 (m, 9H.), 2.36-2.25 (m, 11H), 2.25-2.16 (n, 111), 1.72-; .59 (m, IH), MS (E, ST) m/7 607.34 (M-1).
- 67 Compound 47 0 UH H C ,.. ,N NH, H OH O O OH 0 47 'H NMR (400 MI Iz, CD 3 0D) 6 8.15 (d, -= 10.8 Hz. I H), 4.00 (s, I H), 3.99 (s, 2 H), 3 10-2.87 (im, 11 H), 2 32-2.12 (m, 2 H), 1.59-1.51 (i, 1 II), 1.26 (t, J 5 -7.2 Hz 3 ); M S (ESI) mnz 533.1 (MI I). Compound 48 F N H H N NH2 0-1 0 OF1u 48 4H NNR (400 MlCDLOOD) 5 8.15 (d, J = 11.2 iz, I H), 4.00 (s, 1f-), 10 3,96 (s, 2 H), 3.08-2.87 (m, 11 H), 2 70-2. 61 (m, 1 H). 2,23-2.09 (,i. 4 H), 1.97-1.75 (i, 4 H). 1.59-1.51 (n ,i H); MS (ESI) nz 572.2 (MdR). (ompourd 49 F H-C.. CH, F H. H HO . NH 2 OH 0 O? . 49 15 'FH NMR (400 M-z, CD.OD) 6 8.26 (d, J= 10.8 Hz, 1 1), 4. 10 (s, 3 H), 3,21-2.97 (m, I1 H), 2.35-2.20 (m, 2 H)- 2,15-2.05 (m, 1 H.), 1.98-1.82 (m, 2 H), 1.77-1.61 (im, 5 14), 1.35-1,26 (m, 2 H); MS (ESI) m 587.1 (MH) Compound 50 F HNC 2CHa C 'OH 1, N NINH 2 OH o OF? o 20 60 - 68 HNM R(400 MIz, CD 5 0D)6 8.26 (d,. - 10.8 z, 11 H), 4.16 (s, I H), 4.14 (s, 2WH), 3.20-2.95 (m, I! H), 2.32-2.20 (m, 2 H), 1.88-1 59 (m, 6 i), 1 39-1.21 (i, 4 H), !. 12-1.02 (m, 2 H): MS (ESI) m/nz 6011 (M+). 5 Compound 51 OH Cf HOHO0 OH 0 51 'H NMR (400 MHz, CD 3 OD) 5 8.14 (d, J= 10 8Sz, I1 H), 4-00 (s, 1 11), 3.88 (s 2 H), 3.77-3.73 (i, 1 1), 3.09-2.87 (m, 9 H), 2 29-2.] 0 (in, 6 H), 1.88-1.81 (m, 2 H), 1.59- i50 (m, I H); MS (ES1) /z 559. 1 (M+H), 10 Compound 52 F H- N. GfHa3 Oi H
HNH
2 H OF O 52 'H NMR (400 MHz, CD:,OD) 5 8,24 (d, J = 10.8 Hz, I H).. 4,10 (s, 3 H). 3 17-2,97 (., 9 H1), 2.32-2.09 (n, 4 H), 1 92- .85 (m, 2 TI), 1.75-1.63 (m, 2 H), 15 1 43 -1.26 (m, 6 t-); MS (ESN) m/z 587 2 (M-+H). Compound 53 F HfCsN-H 3 Nazi 53 11 NMR (400 MHz, CD 3 O.D) 3 8.20 (d J = 1,2 Hz, I H). 8.16 (d, J=- 2.4 20 H z, I H), 8.06 (d, J= 5.2 U1z, 1 1 1) 7 85-7 78 (n, 2 H), 4 27 (s, 2 H), 4.11l (s, I H), 3 1 8-2.98 (m, 9 i), 2.32-2.23 (in, 2 H), 170-1.60 (im, 1 H); MS (ESi) m z 582.2 (M-H) CompouFnd 54 -69 H H C OA H
OH
3 H 0 IF OHm 0 54 HIJ NMR (400 MHz, C 5 3 D) S 8.24 (d, J = 11.0 .Hz, I H). 4.31 (s., 2 I), 4.1! (s I H), 3.22-2,88 (m, 9 H) 2.36-2. 16 (m 2 H), 1.70-1.56 (n I H), 1. 44 (s, 9 H); MS (ESI) m/z 577.41 (M H.). 5 Compound 55 F '3;,NCH3 OH F NH, H 55 H NMR (400 MI [z, CD 3 OD) 3 8. 1 (d J - 10.8 Hz, 1 Fl), 1 65 (tJ 4.8 liz, 2 1 ), 4.08 (s, 2 1), 4.00 (s, 1 H), 3.45 (t J - 4.4 Hz, 11 ), 3.38 (t., J- 5.6 Hz, I .0 H), 3.20-2.87 (, 9 11), 2 .2 5 -2,09 (n, 2 H), 1 59-1.50 (m, I H); MS (ES 1) m/z 551.0 (M+1), Compound 56 F HN'C CH H:H F t OHN~ H OF HZ OH 0 5 IH NMR (400 MIlz, CDOD) 6 8.25 (d, J= 11.2 Hz., 11 ), 6.39 (it, J= 53.6, 3.2 Hz, 1 ), 4.24 (s, 2 H) 4.13 (s, 1 ), 3.71 (td, J= 15.2, 2.8 Hz, 2 H ), 3.19-2.91 (m, 9 1), 2.33-2.24 (m, 2 H), 1 70-1.60 (m, I H); MS (ESI) z 569.0 (M- 1). Compound 57 F 1 '13 NC OH H1 2lc,. 0 H NNH - H O ot a 20 57 - 70 'H NMR (4100 Mz, CDOD) S 8 21 (d, j = 10.8 1 z, 1l), 4.01 (s, I [I), 3.85 (s, 2 H), 3.73 (s, 3 H), 3.59-3.51 (m, I H), 3.12-2.87 (m, 9 H), 2,23-2 12 (n, 2 H1), 1.88-1.50 (m, 9 H1); MS (ESI) mhz 559,1 (M+L). 5 Compound 58 F H C'N' HOHH Fe\/' OHNV H OHO C o 58 'H4 NMR (400 M H z, CD3OD) 6 8.24 (d, J= 10.8 Hz, I H), 4.48 (, 2 1H), 4,12 (s. 1 H), 4.10-4.07 (m, 2 H), 3.93-3.86 (m, 2 H), 3 19-2.90 (m, 9-.H), 2.79-2.67 (m, 2 H), 2.37-2.21 (m, 2 1), 1,59-1.51 (m, I H); MS (EST) mn 595-0 (M- L) 10 Example 2. Synthesis of Compokuds of Structural Formula (1), wherein R and R 2 taken together with the nitrogen to which they are bonded form a mnonocyclic or bicyclic heteroaryl. Schemc 2 15 F F F F C CH 3 Br;BrKI CH 3 OH: HNO300 a2
.O
2 Fh O2N COPh CO 3 Ph H 2 N 7 OH OH Cr OBn 3 2-1 2-2 2-3 allylbromide
K
2 C0 2 'KI O-n--H-, JonC (- F Ha NH F HN ~ H H H H o .... a a)LDNTMEDA CH3 N N CD2Ph Bno 0 HO Q O n onc Bno C HC O On 2-8 OTBS H oi 2-5 2Aenne Pd(PPh) 4 1)BrCH2COB'r - HOC OCH., H C, ,CHI H H Na HFHF H,. F 0 2H HC '
RI
BnO O HO C O OH 0 HO 0 a 2-7 OTBS - 71 The following compounds were prepared according to Scheme 2. Compound 2-1 F
--
CH (JkN GO 2 h OH 5 2-1 To a 250 mL round bottom flask was added compound 3 (14,47&, 56 30 ainol, 1.0 equiv, crude), tetrabutylammonium bromide (0.90 g, 2.80 nnol, 0.05 equiv), 1,2-dichloroethane (60 mL), and water (60 mL). The clew bi-layer was cooled in a 20 "C water bath. Nitric acid (7.2 mL, 70 wt%, 112.60 mmol, 2.0 equiv) 10 was added After the addition, the reaction temperature slowly rose to 26 C. The reaction was stirred at room temperature overnight (19 hrs). TLC (heptane/EtOAc = 9.5/0.5) showed the reaction was complete. The organic layer was separated, washed with water (60mL x 2) and brine, and dried over anhydrous sodium sulfate. The solvent was removed to give compound 2-1 as a brown oil, which solidified on 15 standing (17,71 g, quantitative). The crude product was used directly for the next step. Compound 2-2 F CH O N Co 2 Ph OBn 2-2 20 To a 250 nL round bottom flask was added compound 2-1 (17.7 g, 56.30 mmol 1.0 equiv), acetone (177 mL), anhydrous potassium carbonate (15.6 .113.00 mrnol, 2.0 equiv), and potassium iodide (0.47 g, 2.80 mnol, 0 05 equiv) To The stirred suspension at room temperature was added benzyl bromide (7.03 mL, 59.10 mrnol, L-O5 equiv). The suspension was then heated to 56 "C for 4 hrs. TLC 25 (heptane/EtOAc = 9/1) showed the reaction was complete. The solid was removed by filtration and w-vashed with acetone (30 mL). The filtrated was concentrated to give a paste. The paste was partitioned between nmethyl t-butyl ether (MITBE, 120 mL) and water (80 mL). The organic layer was washed with water (80 ML) and - 72 brinc, dried over anlwdrous sodium sulfate, and concentrated to give compound 2-2 as a brown oil (2 1,09 g, 98%) The crude product was used directly for the next step Compound 2-3 F CHi H:N COWPh OBn 5 2-3 To a I L round bottom flask was added compound 2-2 (21.08 g, 55.40 mmol, 1.0 equiv) and TI-IF (230 mIL) The solution was cooled in a cold water bath to 10 "C To another 500 ml. round bottom flask containing water (230 iL), sodium hydrosulfite (Na 2
S
2 O4, 56.7 g, 276.80 rmol, 5.0 equiv) was added slowly with 10 stirring. The aqueous solution of sodium hydrosulfite was added to the THF solution of compound 2-2. The temperature quickly rose from 10 C to 20.4 C after the addition The yellow suspension was stirred while the cold water bath slowly warmed up to room temperature ovemight to give an orange cloudy solution. The reaction temperature during this period was between 15 C to 19 C, 'TLC 15 (heptane/EtOAc = 9/1) showed the reaction was complete. The orange cloudy solution was diluted with EtOAc (460 mL). The organic layer was washed with water (150 nL x 2) and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give the crude product as a brown oil. The crude product was purified by flash silica gel column eluted with ieptane/EtOAc 9/1 20 to yield the desired product 2-3 (15.83 g, 80%, 3 steps) Compound 2-4 F 2-4 To an NMIP solution (50 niL) aniline 2-3 (10.02 g, 28.5 mnol, 1 equiv) was 25 added allyl bronide (7.65 niL, 85 5 mmol, 3 eouiv) and potassium carbonate (11 79 g, S5.5 n-ml, 3 equiv). Potassium iodide (9)4.8 tmg, 6 mmol, 0 2 equiv) was added and the reaction mixture was placed under nitrogen and heated to [00 C After 16 h, the reaction was cooled, diluted with water (60 mIL), and extracted with EtOAc - 73 (75 mL, then 2 x 50 miL). ihe combined organic extracts were washed with water (2 x 35 mL), were dried (Na 2 S04), filtered, and concentrated to yield the crude product, Purification via flash column chromatography on silica gel (RediSep, 125 g, gradient 1-6% EtOAc in hexanes) gave 10.97 g of pure 2-4 (89%): 'H NMR (400 MIz. 5 CDC3) 5 7,42-7.30 (nt 7 H1), 7,42-7,20 (n I JI), 7.00 (d, .= .5 lIz, 2 f), 6 72 (d, J = 11.0 [1Z, 13H), 5.77-5.70 (m, 2 H), 5.20-5.12 (n. 6 H), 3,81 (4 ,= 6.1 Hz, 4 H), 2.26 (s, 3 H); MS (ESI) m, z 432.34 (M+H). Compound 2-5 F H,C H, H ,N BnO OHO o OBn 10 2-6 oS A T ILF solution (6,5 mL) of 2-4 (875 mg, 2,03 nmol, 1.25 equiv) was added to a freshly-prepared solution of LDA in THF (0.051 M 2.03 mnol, 40 mL, 1.25 eqviy) and TMFDA (304 .L, 2.03 mmol, 1.25 equiv) at -78 DC. The reaction was stirred at -78 'C for 15 rain, A TI-F solution (6.5 rnL) of enone (784 mg, 1 62 nniol, 15 1.0 equiv) was added to the reaction mixture dropwise, followed by addition of LHM7DS solution (10 M in THY, 2 03 nmL, 2.03 mmol, I 25 equiv). The reaction ywas stirred from -78 'C to -10 C for I h, quenched with saturated NH 4 CI (6 mL), and warmed to 25 'C. The solution was poured into saturated NI4iCl (20 mL) and extracted with EtOAc (2 x 75 nL). ihe combined EtOAc extracts were dried 20 (Na2aSO 4 ), filtered, and concentrated to yield the crude product. Preparative reverse phase HPLC purification on a Waters Autopurification system using a Sunfire Prep Ci 8 OBD column [5 m, .9 - 50 mm; flow rate, 20 mnL/min; Solvent A: 120 vith 0(1% HCO 2 H; Solvent B: CH3CN with 0.1% HC0211; injection volume: 4x 3.6 4.2mL (CIHCN),; gradient: 88-+100% B over 12 min; mass-directed fraction 25 collection], Fractions with the desired MW, eluting at 6.6-10.6 min, were collected and iyophilized to give 552 mg of pure 2-5 (41%)l: 'H NMR (400 MITz, CDCI) 6 16.22 (s, 1 H), 7,49-7,47 (n, 4 H), 7.37-71 (m, 6 H), 6.80 (d, J = 11.0 liz, 1 H), 5,76-5,64 (ni, 2 I), 5.35 (s, 2 H), 517-5.11 (i, 4 H) 4.98 (d, J= 9 2, 1 IH). 4.87 (d, J = 9,8 lHz, 1 H1), 3.96 (m, J= 10.4 Hz, 1 14), 3.83-3.71 (m, 4 [1), 3.14 (dd, .J- 14.7, - 74 4.3 Hz, 1 H), 3.0-2 87 (m, 1. 1) 2.55-2.35 (i, 9 H), 2.1 1 (d, J= 14.7 Hz, I H), 0.82 (s, 9 f), 026 (s, 3 H), 0.13 (s, 311); MS (ESI) mrz 820.55 (M+H). Compound 2-6 F HC.. CH, H2N BnO 0 HO I OBr 2$ OTBS A solution of 2-5 (550 mg, 0 671 mmol, L 0 equiv) in degassed CH2lC 2 (2,5 mnL, with I and 1.5 mL rinse) was added under nitrogen via syringe to a flame-dried flask containing N,N-dimethylbarbituric aud (324 mg, 2 07 mmol, 3.0 equiv), and Terraki s(tri phenylphosphine)palladium (0) (56.9 mg, 0.0492 mmol, 0 07 equiv). The I 0 resuing solution was heated to 35 C for 4 h, then concentrated to remove the solvent. The resulting crude mixture was purified via preparative reverse phase HIPLC on a Waters Autopurification system using a Sunfire Prep C18 0BD column [5 pm, 19 -50 mm; flow rate, 20 mL/min; Solvent A: H 2 0 with 0. 1% HCOH; Solvent 13: CH 3 CN with 0.1% tlCO2I : injection volume: 3x 3.1 rT (CH,;CN); 15 gradient: 80->100% B over 17 min; mass-directed fraction collection] Fractions with the desired MW, eluting at 6 1-10 1 mini, were collected and freeze-dried to give 352 mug ofpure 2-6 (71%): 'H NMR (400 Mtz. CDCI 3 ) 5 16 10 (s, 1 H), 7.51 7.43 (m, 4 H), 7.9-7.29 (m , 6 H), 6.61 (d, J 9.8 F lz, I H). 5.35 (s, 2 1H), 4.87 (dd, J = 22 6, 104 Hz, 2 H), 3.96 (d. J= 10.4 Hz, 1B), 3.91 (s, 2 11), 3.12 (dd, J= 15.3, 20 10.1 Hz, [ H), 3.04-2.92 (m I H), 2.55-2.31 (m, 9 ), 2. 11 (d, J= 14.7 Hz, I H), (.82 (s, 9 11), 0.27 (s, 3 11), 0 12 (s, 3 H); MS (EST) n z 740.44 (M+H). Compound 59 F HI'CH H H NH2 OH 0 on 59 25 To a solution of aniline 2-6 (30 mg, 0.041 mnol, I equiv) in TFI (600 L) was added bromoacetylbromide (3 7 ItL, 0.043 mmol, 1.05 eqaiv) After 15 minutes, indazolc (53 mig, 0.45 mrnol 10 equiv) was added. After 15 I the reaction was - 75 heated to 80 C,. After an additional 26 h, another 20 mg indazole (0 17 mmol, 4 equiv was added and the reaction heated at 80 C. After 20 h, the solvent was removed in vacuo and the resulting crude mixture dned under vacuurn. The above crude intermediate was transferred to a plastic vial in dioxane (1 2 5 mL) and an aqueous solution of hydrogen fluoride (50%, 300 pL) was added. After five hours, the reaction solution was diluted with ar aqueous solution of K2H-P0 4 (3 6 g in 30 mL) and extracted with EtOAc (2 x 30 mL). The combined organic layers were dried (Na2SO 4 ), fihered, and concentrated to yield the crude product Palladium on carbon (10%, 10 mg) was added to solution of the above crude 10 intermediate in dioxane:methanol (1:1, 1 rnL). The flask was fitted with a septum and evacuated and back-filled three times with hydrogen gas. Hydrogen gas was bubbled through the reaction solution for two minutes, then the reaction was stirred under an atmosphere (balloon) of hydrogen gas for 1.5 h, 'The reaction mixture was filtered through celite to remove the palladium catalyst and concentrated under 15 reduced pressure Preparative reverse phase HPLC purification of the resulting oil was performed on a Waters Autopurification system using a Polymerx 10 L Ri> 100 R column [30 x 21 20 mm, 10 micron, solvent A: 0 05N HCI in water, solvent B: ClCN; injection volume: 4.8 mL (0.05N HC] in water); gradient elution with t0 60% 1 over 1.5 min, then held at 100% for 5 mit; mass-directed fraction 20 collection] Fractions with the desired MW, eluting at 14-14.65 min, were collected and freeze-dried to yield 3.6 mg of compound 59 (15%): 1 H NNfR (400 MHz,
CD
3 0D) 8 8,71 (s, 1 H), 8.19 (d, J - 11.0 H I H), 7.92-7.90 (na 1 1H), 7.72-7 57 (m, 2 H-), 7/35-729 (m, I T) 5,65 (s, 2 1) 4 08 (s, I H.), 3.16-2.92 (m, 9H), 2.3 1 2.18 (m, 2 A), 1,67-160 (-, 1H); MS (ESI) rnz 606.41 (M it) 25 Compound 60 F HC. NCH, H H H O F H 60 To a solution of aniline 2-6 (22 mg. 0,030 mmol, 1 equiv) ri THFE (500 L) was added hromnoacetylbronide (2.7 L 0.031 mmol, 1 .05 equiv). After 30 minutes. 30 pyrazole (36 mg,, 0.53 nmmol, 18 equiv) was added. After 20 min the reaction was - 76 heated to 80 *C for 1.5 i, cooled to room temperature for I 5 h, then heated at 80 "C for 4.5 h. The solvent was removed in vacuo and the resulting crude mixture dried under vacuum. The above crude intermediate was transferred to a plastic vial in acetonitrile 5 (1.0 mL) and an aqueous solution of lhydrogenfuoride (50%, 200 pt) was added. After 20 h, the reaction solution was diluted With an aqueous solution of K21PO (2.4 g in 20 mL) and extracted with EtOAc (2 x 25 nL). 1he combined organic layers were dried (Na 2 SO), fltered, and concentrated to yield the crude product. Palladium on carbon (10%, 10 mug) was added to solution of the above crude 10 intemediate in dioxanemethanol ([:1, 1 mL). The flask was fitted with a septum and evacuated and back-filled three times with hydrogen gas. Hydrogen gas was bubbled through the reaction solution for two minutes and the reaction was stirred under an atmosphere (balloon) of hydrogen gas for 1,5 h. The reaction mixture was filtered through celite to remove the palladium catalyst and concentrated under 15 reduced pressure Preparative reverse phase HPLC puriFication of the resulting oil was performed on a Waters Autopurification system using a Polymnerx 10 P RP! 100 R column [30 - 21.20 rim, 10 micron, solvent A. 0.05N HC in water, solvent B: CEHCN; injection voimc 3 0 ml. (10% CH3CN in 0 05N HCI in water); gi adient elution with 10-+60% B over 10 muin, then held at 100% for 5 rmin; mass 20 directed fraction collection]. Fractions with the desired MW, outing at 8.8-10.2r min were collected and freeze-dred The resulting yellow solid was purified a second time via preparative reverse phase HPEC purification uLsing the above procedure with a gradient over 20 minutes, with the fractions with the desired MW cIting at 10.7-12.4 min were collected and freeze-dried to give 8.2 mug of pure 60 (50%): 1f 25 NMR (400 MHz, CD 3 O)) 6 8 19 (d, J= 11.0 Hz, I H), 8,05-7,92 (m, 2 H), 6.62 6.57 (in 1 ), 5,33 (d, J= 4.9 Hz, 2 1-1), 4.08 (s, 1 H), 31 6-2,90 (m, 9 H), 2 31-2.17 (in, 2 It), 1 69 1.55 (in, 1 H); MS (ESI) m z 556,42 (M+H). Compound 61
H
3 C'N-CH. NH H HH CH OH AN0- NH? 30 81 - 77 To a solution of aniline 2-6 (23 mg, 0.032 mmcl, I ecuiv) in THF (500 L) was added bromoacetylbromide (2.9 pL., 0.034 mmol. 1.05 equiv). After 30 minutes, imidazole (32 ig, 0.47 mmol, 15 equiv) was added and the solution was heated to 80 C After 2 h, the solution was cooled and the solvent was removed in vaco. 5 The above crude intermediate was transferred to a plastic vial in dioxane (1.2 nL) and an aqueous solution of hydrogen fluoride (50%, 200 1 .L) was added. After 1.5 h, the reaction solution was diluted with an aqueous solution of K 2 1P0 4 (2.4 g in 30 Tl.) and extracted with. EtOAc (2 x 25 rnL). The combined organic layers were dried (Na 2 SO4) filtered, and concentrated to yield the crude product, 10 Palladium on carbon (10%, 8 mg) was added to solution of the above cruce intermediate in dioxane: methanol (1:1, 1 mL). The flask was fitted with a septum and evacuated and back-filled three tires with hydrogen gas I lydrogen gas was bubbled through the reaction solution and the reaction was stirred under an atmosphere (balloon) of hydrogen gas for 1.5 h More palladium catalyst was added 15 and the evacuation and backfilling with hydrogen was performed twice more at 1.5 h and 5 h. The reaction mixture was filtered through elite to remove the palladium catalyst and concentrated under reduced pressure. Preparative reverse phase HPL( puriication of the resulting oil was performed on a Waters Autopurification system using a Polymerx 10 RP-y 100 R column [30 21.20 mm, 10 mc.ron, selvent A: 20 0.05N [IC in water, solvent B: CHhCN; injection volume: 2.8 mL (0.05N IICl in water); radient elution with 1.0-+60% B over 15 min, ihen held at 100% for 5 minl: mass-directed fraction collection]. Fractions with the desired MW, el utg at 70-7.8 rmin were collected and freeze-dried to give 4 1 mg of pure 61 (23%): H NMR (400 Ml Hz, CD OD) 6 9.02 (s, 1 H), 8. 17 (d, J= 11.0 Hz, 1 H1), 7.67 (s, 1 T), 7.61 (s, 1 25 I), 534 (s, 2 H), 4.09 (s, Fl), 3.18-2.90 (m, 9 H), 2,34-21.7 (in, 2 II), 1.71-156 (m,. t A); MS (EFSI)m/ 556.45 (-M+1H). Co mpound 62 F H 3 XC CH, , N N H, H CHo o-? 0 62 - 78 To a solution of aniline 2-6 (20,2 mng, 0.027 nnol, I equiv) in 11TF (500 1L) was added bromoacetylbroiide (2.5 pL, 0.029 mmol, 1.05 equiv). After 30 minutes, I 1H- 1,2,3-triazole (31 L. 0.54 mmol, 20 equiv) was added and the solution was heated to 80 *C. After 17h, an additional 31 uL (20 equiv) of I H1-2,3-triazole was 5 added and the solution was heated for 22 h. The solution was cooled and the solvent was removed in vacuo. The above crude intermediate was transferred to a plastic vial in dioxane (1 0 rLP) and an aqueous solution of hydrogen fluoride (50%, 200 pL) was added. After 17 h1, the reaction solution was diluted with an aqueous solution ol' K 2 1-PO 4 (2.4 g in 10 20 mL) and extracted with EtOAc (2 x 25 mL), The combined organic layers were dried (Na2SO 4 ), filtered, and concentrated to yield the crude product. Palladium on carbon (10%, 7 mg) was added to solution of the above crude interm ediate in dioxane methanol (1:1, 1 miiL). The flask was fitted with a septum and evacuated and back-filled three rimes with hydrogen gas. Hydrogen gas was 15 bubbled through the reaction solution arid the reaction was stirred under an atmosphere (balloon) of hydrogen gas for 1.5 h. The reaction mixture was filtered through celite to remove the palladium catalyst and concentrated tinder reduced pressure. Preparatve reverse phase HPLC purification of the resulting oil was performed on a Waters AutopurieICation system using a Polymerx 10 g RP-y .100 R 20 column [30 x 21.20 mrn, 10 micron, solvent A: 0.05N HCl in water, solvent B: CHtCN; injection volume: 2.5 niL (0.05N HCl in water), gradient elation with 10 >60% B over 15 min, then held at 100% for 5 inn; mass-directed fraction collection] Fractions with the desired MW, eluting at 9.25-10.5 min were collected and freeze-dried. Purification was performed a second time as abovo and the 25 fractions with the desired MW, eluting at 9.75-10.25 min were collected and freeze dried to give 1 5 ng of pure 62 (10%): 1H NMR (400 MHz, CD 3 OD) 8 8.24 (s, 1I) 8.17 (d, J= I 1.0 Hz, 1 H), 8.00 (s, I LH), 5.57 (s, 2 lI), 4.09 (s, I H), 3.16292 (ni, 9 H), 2.34-2. 16 (m, 2 H), 1 71-1,67 (m, 1 Hi), MS (ESI) m,: 557.44 (M+ H1) 30 Compound 63 - 79 F H3CN- Hi H H N, NH Nt' OH O 0 63 To a solution of aniline 2-6 (16.7 ng, 0.023 inmol, I equiv) in THF (500 pL) was added brornoacetylbromide (2.0 pL, 0.024 mmol, 1.05 equiv). After 20 minutes, a tetrazole solution (0.45M in C1lCN, 500 pL, 0,23 mmol, 10 equiv) was added and 5 the solution was heated to 80 oC. After 4 b, potassium carbonate (35 ng, 0.25 mmol, 11 equiv) was added and the reaction healed for 35 minutes. The solution was cooled and filtered through colite, and the solvent was removed ini vacuo. The above crude intermediate was transferred to a plastic vial in dioxane (1. 0 niL) and an aqueous solution of hydrogen fluoride (50%, 200 L) was added After 10 18 h, the reaction solution was diluted with an aqueous solution of KjHPO (2.4 g in 20 ilL) and extracted with EtOAc (2 x 25 mL). The combined organic layers were dried (N2SO4), filtered, and concentrated to yield the crude product. Palladium on carbon (10%, 7 ig) was added to solution of the above crude intermediate in dioxane-methanol (1:1, I niL). The flask was fitted with a septum 15 and evacuated and back-filled three times with hydrogen gas. Hydrogen gas was bubbled through the reaction solution and the reaction was stirred under an atmosphere (balloon) of hydrogen gas for I h. The reaction mixture was filtered through celite to remove the palladium catalyst and concentrated under reduced pressure. Preparative reverse phase HPLC purification of the resulting oil was 20 performed on a Waters Autopurification system using a Polyrnerx 10 P RP-y 100 R column [30 x 21.20 m, 10 micron, solvent A: 0.05N HCi in water, solvent B. CT;CN; injection volume: 2-5 mL (10% CHI 3 CN in 0.05N HCI iII water); gradient elution with '10-60% B over 15 min, then held at L.00% for 5 min; mass-directed fraction collection]. Fractions with the desired MW, eluting at 11.2-12.1 min were 25 collected and freeze-dried. Purification was performed a second time as above with the gradient extended over 20 min. Fractions with the desired MW, eluting at 13.7 14.5 mnu were collected and freeze-dried to give 1.6 mg of pure 63 (13%): 'H NMR (400 MHz, CD 3 OD) 6 3.78 (s, I H), 8.14 (d, J- 1 LO Hz, 1 1), 5.78 (s, 2 H), 4.07 (s, I H), 3 17-281 (m, 9 1), 236-2,16 (n, 2 H), 1.70-1.52 (i, I H); MS (ESI) nol 30 558.43 (M+H), - 80 Example 3. Synthesis of Compounds of Structural Formula (A), wherein X is hydrogen and Y is -NI-C(O)-heterocyclvI, -NH-C(O)-heteroaryl -NH C(O)- [C(R")( RT)orN(R)(RB), -NH-C(O)-carhocyclyl, -NH-C(O)-aryl, 5 -NB-SOr((;C )aIyI, -NH-SO-carbocyclyl, -NII-SO 2 -aryl, -NIH-SOrhetercyclyI or -NH-SO 2 -beteroarvl. Scheme 3 F N R NNH 2 OH 0 HO 0 O R = R'QO, RNHCO. or RSC:>] 3-1 R'COOH/coupling reagent or R COCl :r R'NCO r 'SO2CI R H. CH. Cbr H HrC, .N' OH OH HATUEt x H 0 N NH2 PH,/Pd-C F:,' blbC' HN' ..~ R N NNa( N -(o '-sakn- carbocycHy7, -(AC aklneay -(r'jlyen-ee rl r -o- a N cr1 cpud m N: 3 NH G HO-OH H H HO H 3-4 5.,3 10 In Scheme '3, R' represents h ete rocyclyl, hetero arvl, carbocrycivi1, ry , (Qc Ct)alkiyl1, cr -[( (R 1
')(RJ
0
],
1 -N(R RA)(RB); an d K 2 re.pre sents hydroc-en, (C]-C6)alkvl,
-(C
0 - 5fllvlenocarbocycly ,C-C 5 )alkylune-nry], -(C 1 'I-C lalk~ylene-heteroa'A, or -( -C~aJkylen~-icteocc~y. For certain comipo unds ruacic by Schemei 3 and described below, Rz is hydrogen and Rx and R are taken together with the carbon 15 and nirogen atoms to which they are bound to form an optionally substituted 4-7 mernbered saturated heterocycly! It will be readily apparent to those of skill in the art., however, that this Scheme 3 will also be useful to synthesi ze compounds where R7 R and R are R' . R." and R. respectively, as defined in structural formula (A). The following compounds were prepared according to Scheme 3. 20 - 81 Compou id 64 F H 1-2- ,CHb, F H H H O N~J N' ~ NH, HHNH 64 To a solution of aniline 9 (170 mg, 0.038 nunol, I equiv) in DMF (200 IL) was added N-Benzyioxycarbonyl-L-proine acid chloride (1.0 N in toluene, 57 [LL, 5 1 .5 equiv). After 50 min the reaction mixture was diluted to 3 mL with 0 05 N 1C in H2O and filtered to remove any solids. Preparative reverse phase HIPLC purification of the resulting solution was performed on a Waters Autopurification ssten using a Poymerx 10 RP- 100 R column [30 x 21,20 mm, 10 micron, solvent A: 0.05TN 1(C in water, solvent B: CH 3 CN; injection volume 3 5 niL (0,05N 10 HCI in water): gradient elution .with 10-20% B over 25 min, then held at 100% for 5 nin; mass-directed fraction collection]. Fractions with the desired MW, Cluting at 27.1-28 4 min, were collected and freeze-dried. Palladium on carbon (1.0%, 10 mg) was added to a solution of tie above intermediate in dioxane:McOH (1:3, 2,3 rmL.). The flask was fitted with a septum 15 and evacuated and back-filled three tines with hydrogen gas. The reaction solution was stirred under an atmosphere (balloon) of hydrogen gas for 1.7 h, then was filtered through celite to remove the palladium catalyst and concentrated under reduced pressure. Half of the rcsultng residue was purified via preparative reverse phase HPLC purification on a Waters Autopurification system using a Polymerx 20 10 p. RP t 100 R column £30 x 21.20 mm, 10 micron, solvent A: 0.5N HC in water, solvent B: C.HCN; injection volume: 1.8 mL. (0 05 N H-CI in water): gradient clution with 0.35% B over 10 mii, then held at 100% for 5 min; mass-directed fracuon coliectionj Fractions with the desired MW, eluting at 7.8-S min, were collected and freeze-dried to yield 1 9 mg of compound 64 (30%): H NM' R (400 25 MHz CD 3 OD) S 8.16 (d, J 11.0 Hz, 11 H),4.59-4.56 (m, I H), 4,10 (s, 1 ), 3 48 3.33 (m, 2 H), 3.18-2,95 (m, 9 1.), 2 59-2,50 (m, I T), 2.34-2.05 (ni 5 H), [.70-1.60 (in, 1 H); MS (ESI) wit 545.38 (M-). Compound 65 -82 F HaX, HNHt as H To a solution of aniline 9 (15,7 mg, 0.035 nmol, I equiv) in DMF (200 pL) was added N-Benzyloxycarbonyl-D-proline acid chloride (1.0 M in toluene, 53 IL, 1.5 equiv). After 50 min, the reaction was complete. The reaction nuxture was 5 diluted to 3 miL with 0.05 N HC1 in HO and filtered to remove any solids. Preparative reverse phase iPLC purification of the resulting solution was performed on a Waters AItopurification system using a Polymerx 10 . RP-y 100 R column [30 x 21.20 mm, 10 micron, solvent A: 0.05 N HCI in water, solvent B CH 3 CN; injection volume: 3.5 nL (0.05 N HCl n vater); gradirnt elution with 15-+80% B 10 over 10 min, then held at 100% for 5 mr mass-directed fraction collection]. Fractions with the desired MW, eluting at 6.95-8.10 min, were collected and freeze dried Palladium on carbon (10%, 15 mg) was added to a solution of the above intlermediate in dio'ane:MeOH (1:3, 2.3 ruL..). [he flask was fitted with a sepIumr 15 and evacuated and back-filled three times with hydiogen gas, and the reaction was stirred under an atmosphere (balloon) of hydrogen gas for 1.5 h. 'Tlhe reaclion mixture was filtered through ceimte to remove the palladium catalyst and concentrated under reduced pressure Half of the resulting residue was purified via preparative reverse phase H'PLC purification on a Waters Autoporification system 20 using a Polymerx 10 p Ri-y 100 R column [30 x 21.20 mm, 10 micron, solvent A: 0,05 N IC in water, solvent B. CH 3 CN; injection volume: 1.8 niL (0.05 N HCI in water); gradient elution with 0- 35% B over 10 inu, their held at 100% for 5 min: mass-directed fraction collection]. Fractions with the desired MW, cluting at 8.35 8.85 rmn, were collected and freeze-dried to yield 0.93 mg of compound 65 (24%): 25 '14 NMR (400 MILz CD;OD) 6 8.17 (d,. = 11.0 Iz, I H), .159-4.53 (m, 1 11), 4.09 (s, I H), 3 48-3.37 (n, 2 H), 3,18-2,90 (i, 9 IT), 2.59-2. 50 (i. 1 H), 2.34-2.05 (m., 5 H), 1.70-1.59 (in, I H); MS (EST) mto 545.37 (M+ I) Compound 66 - 83 F N H NH2 OH 0 OH O 0 66 The second half of crude 64 (0.012 mmol, I0 equiv) was dissolved inl DMF (500 pL) and formaldehyde (37% aqueous solution, 5.3 pL, 0.072 mmoi, 6 equiv), triethylaiine (5 0 uL, 0.036 mmol, 3 equiv), and sodium triacetoxyborohydride (8.4 5 mg, 0.039 mmol, 3.2 equiv) were added sequentially. After 2 h1, the reaction mixture was diluted to 1.8 mL with 0.05N 14C in 1120 and purified via preparative reverse phase IHPLC on a Waters Autopurification system using a Polynerx 10 A RP-y 100 R column [30 x 21.20 mm, 10 micron, solvent A: 0 05 N HC1 in water, solvent B:
CH
1 CN; injection volume: 1.8 mnL (0.05 N [C1 In water); gradient: 0-- >30% B over 10 10 min, mass-directed fraction collection. Fractions with the desired MW, eluling at 8.6-9.35 min, were collected and freeze-dried to provide a mixture of the desired compound and an over-formylated product. The resulting compound mixture was dissolved in 4 N aqueous H0 solution (1.5 mL) and stirred for 50 h, then freeze dried to provide 1.0 ng of the desired compound 66 (15%) 'H NMR (400 MHz. 15 CDOD) 6 8]? (d, .= 104 Hz, 1 U), 4.36 (t, J= 8.6 Hz, 1 H), 4.08 (s, I H), 3.82 3,73 (m, I H), 3.20-2.90 (in, 12 11), 2.73-2.68 (in 1 H-), 2.35-2,10 (im, 5 H), 1.70 1.60 (m, 1 H); MS (EST) m z 559.38 (M+H). Compound 67 H O' *H O N
HNNH
2 "H Oil 0 OHM~ 0 20 67 Tho second half of crude 65 (C?007 inol, 1.0 equiv) was dissolved in D/F (500 pL), and formaldehyde (37% aqueous solution, 31 , 0.042 mmol, 6 equiv) and YEA (3.0 psL, 0.021 mmol, 3 equiv) and sodium triacetoxyborohydride (4 mg, 0.026 mmol, 2.6 equiv) were added sequentially. After 2.2 h, the reaction mixture 25 was diluted to 1.8 mL with 0.05N HC1 in 120 and purified via preparative reverse phase HPLC on a Waters Autopurificaltion system using a using a Polymerx 10 p. RP-y 100 R column [30 x 21.20 mm, 10 micron, solvent A: 0.05 N 11 in water, -84 solvent B: CHLCN injection volume. 2.0 niL (0.05 N HlCl in water), gradient: 0-30% B over 10 min; mass-directed fraction collection]. Fractions with the desired MW, eluting at 8.9-9.6 min, were collected and freeze-dried to provide a mixture of the desired compound and an over-formylated product. The resulting 5 compound mixture was dissolved in 6 N aqueous HC solution and stirred for 50 h, then freeze-dried to provide 1.5 mng of the desired compound 67 (38%) 'H NMR (400 Mz, CD 3 0D) 68.17 (d, J= 10.4 Hz, I 14), 4.45-4.34 (m, 1 H), 4,08 (s, I H), 3.84-3.74 (rn I H), 3.20-2.90 (m, 12 1H), 2 79-2.65 (rn, 1 H1), 2.33-2.05 (m, 5 H), L1.70-1.58 (m, 1 EH); MS (ESI) m 55940 (M+HI) 10 Compound 68 F N H Hr N HH OH 0 0 FO a 'l'o a solution of (.S)-()-1-Cbz-piperidinecarboxylic acid (34.2 rg, 0 13 mlol, 3 equiv), and (2-(7-aza-i-betzotrazole-i-yi)-1,1,3,3-tetramethyluronium 15 hexafluoro-phosphate) (50 0 mg, 0.13 mol, 3 equiv) in DMF (200 pL) was added triethylamine (18 pl, 0.13 mrnol, 3 equiv). After 30 miii, aniline 9 (17.5 mig, 0.039 inmol, I equiv) was added. After 16 h., the reaction mixture was diluted to 3 niL with 0.05 N HCI in H-120 and purified via preparative reverse phase HPLC on a Waters Autopunfication system using a Polyrmerx 10 t RP-'y 100 R column 20 [30 21.20 mm, 10 micron, solvent A: 0 05 N HCl in water, solvent B: CH;CN: injection volume: 3.5 mL (0.05 N -Ct in water); gradient elution with 15--70% B over 10 min, then held at 100% for 5rmmi; mass-directed fraction collection]. Fractions with the desired MW, eluting at 9.07-10.0 min, were collected and freeze dried. Palladium on carbon (10%, 4 rng) was added to a solution of this foam in 25 dioxane:MeOH (1:3, 1.2 nL). The flask was .ftted with a septum and evacuated and back-filled thr -e times with hydrogen gas. The reaction mixture was stirred under an atmosphere (balloon) of hydrogen gas for 1.5 h, then was filtered through elite to remove the palladium catalyst and concentrated under reduced pressure, Preparative reverse phase HPLC purification of the resulting oil was performed on a Waters 30 Autopurification system using a Polymerx 10 r RP--y 100 R column - 85 [30 / 20[20 mm, 10 micron, solvent A: 0.05 N .-Cl in water, solvent 13. CHLCN, injection volume: 2.0 rmL (0.05 N HC) in water); gradient elation with 0-35% B over i min, then held at 100% for 5 min: mass-directed fraction collection] Fractions with the desired MW, eluting at 8.15-8 58 nin, were collected and freeze 5 dried to yield 0.75 mg of compound 68 (4%). 1 H NMR (400 Mz, EDOD) 6 8.15 (d, J= I L0 Hz, I H), 4.12-406 (i, 2 H), 3.48-3.40 (m), 2 H), 3.20-2.90 (m, 9 IH), 2.36-218 (im, 3 1), 2.02-1.90 (m, 2 H), 1,82-1.60 (m, 4 H); MS (ESl) Mz 559.37 (M+H). 10 Compound 69 FHC,. -CH 9 N U H? IN OH H C- HD- 0 o 69 To a solution of (R)-(+)-1-Cbz-piperidinecarboxylic acid (35 0 mg, 0 13 mmol, 3 equiv), and (2-(7-aza-1I -benzotriazole- I -l)- 1, L,3,3 -tetramethyl roum urn hexaflioro-phosphate) (50.0 mg, 0,13 molt 3 equiv) in DMF (200 RL) was added 15 T EA (18 pL 0 13 minol, 3 equiv). After 30 min, aniline 9 (16.6 mg, 0.037 mmol, 1 equiv) was added. After t6 h, the reaction rnixture was diluted to 3 nL. with 0 05 N HO in 112O and purified via preparative cversa phase HPLC on a Waters Autopurification system. using a Polyrnerx 10It RP-y 100 R column [30 x 21.20 mm, 10 mricron, solvent A: 0.05 N HC in water, solvent B: CH3CN; 20 injection volume: 3.5 mL (0.05 N HC in water); gradient Ciution with 10-+50% B over 10 min, then held at 100% for 5 min: mass-directed fraction collection]. Fractions with the desired MW, elLting at 12.1 -12.9 uin, were collected and freeze dried Palladium on carbon (1 0%, 5 mg) was added to a solution of this foham in dioxane:MOOH (1:3, 800 pL). The flask was fitted with a septum and evacuated and 25 back-filled three times with hydrogen gas. The reaction mixture was stirred under an atmosphere (balloon) of hydrogen gas for 1,75 h, then was filtered through eclite to remove the palladium catalyst and concentrated under reduced pressure Preparative reverse phase HP! LC purification of the resulting on was performed on a Waters Autopurifi cation system using a Poiymerx 10 p RP-y 100 R column 30 [30 21.2) mm, 10 rmicron. solvent A: 0.05 N HCI in water, solvent B: C13CN; - 86 injection volume: 2.0 mL (0.05 N HCl in water); gradient caution with 0 -,35% B over 10 mim, then held at 100% for 5 min mass-directed fraction collection]. Fractions with the desired MW, eluting at 8.75-9.16 min, were collected and freeze dred to vield 0,55 m of compound 69 (3%): 'ii NMR (400 MIz, CD 3 0D) 6 8.16 5 (d, ./ = 11,0 Hz, 1 H), 4 .13-4 06 (in, 2 H), 3 50-3.43 (m, 2 H), 3 20-2.90 (m, 9 1i), 238-2 18 (in 3 H), 2.04-1.88 (n, 2 11), 1.83-1.60 (i, 4 IH) MS (EST) o/l 559.38 (M+H) Compound 70 F N I- H = HH to~i TO O To a solution of compound 68 (0.0138 ninol, I equiv) in D.MF (750 gL), were added formaldehyde (37% aqueous solution, 6.2 L, 0.083 mmol, 6 equiv), TEA (5.8 pL, 0.041 nmool, 3 equiv), and sodium triacetoxyborohydride (It mg, 0.051 mmol, 3.7 equiv) sequentially. After 17 h, the reaction mixture was 15 concentrated to remove amine and 6 N aqueous HCI (500 [iL) was added. Afier 19 days, the reaction solution was purified via preparative reverse phase I{PLC on a Waters Autopurification system using a Polymerx 10 a RP- 7 100 R column [30 x 2120 mm., 10 micron, solvent .A 0.05 N HCI in water, solvent B: CHCN; injection volume: 2 5 mL (0.05 N Hi in water);gradient: 15- )50% .B over 15 min; 20 mass-directed Craction collection]. Fractions with the desired MW, eluting at 5.75 6.2 min, were collected and freeze-dried to provide 2 4 ig of the desired compound 70 (31%): H NMR (400 MHz, CDOD) 3 8.16 (d, J 11.0 H1z, 1 H), 4.08-4.04 (m, I JU), 3 59-3.53 (i, I H), 3.20-3. 10 (m, 5 11), 3.06-2 96 (m, 5 H), 2.90m (s, 3 1), 2.36-2 25 (m, 2 H), 2.11-2.05 (M, 1 -), 2.02-1.94 (n, 2 H), 1.90-1.74 (m, 2 11), 25 1,71-1.58 (m, 2 F); MS (ESI) inz 573,33 (M+H), CompoInd 71 -87 F Yq CH H HH H~C C NOH Nit g 1 NH-, F 71 Compound 9 (20 mg, 0.045 mmio, 1.0 equiv) in THF was added Na 2
CO
3 (9.5 mg, 0.089 mmol, 2.0 equiv) (4R)-4-fluoro-l-methy-L-proline (9.8 mg, 0.067 mmiol 1 5 *quiv) and HATU (34.6 mg, 0.047 nmol, 2,0 equiv). The reaction 5 mixture was stirred at room temperature for 20 hour. LC-MS analysis indicated the starurng material was consumed completely. HCIMeOH (1 mL, 4 N) was added to the mixture at 0 'C and stirred for 2 min. The mixture was concentrated under vacuum, the residue was puried by reverse phase HPLC to afford product 71 (6.1 mg): 1 H NMR (400 MHz, CD 3 00) 6 8,18 (d, = 10.8 Hz, 1 H.), 5.51 (d, J = 51.6 10 Hz, 1 1), 4 76-4,72 (i, 1 H), 4.22-416 (in 1 IH), 4.10 (s, I H), 3.74-3.63 (n I I), 3.21-2.97 (in, 14 .H), 2.35-2.2t (n 2 H), 1.69-1.60 (ni, I H); MS (ESI) mi 577 I (M I H). Compounds 72 and 73 were prepared similarly to compound 71 using the 1 corresponding amino acids. Compound 72 F H..CH 3 H H HC NH2 H 0 IP'A N 72 Prepared simni larly to compound 71: '11 NMR (400 MIlz, CRO6) 8.16 (d, 20 J - 10.8-z, 111), 5.48 (d, J= 51.2 Hz, 1 4) 4.60-4.56 (mi 1 ), 4.11 (s, [ H), 4.05 3.98 (i I H). 3.67-3 54 (i, t T-), 3.24-2.96 (mr, 13 Hi), 2.55-2.44 (m, 1 [H), 2 34 2.22 (i, 2 1-), 1.70-1.66 (in, I H); MS (ESI) m/z 577 1 (M+i). Compound 73 -88 F N H H H H 'J O H O O F F 73 Prepared similarly to compound 71: 'H NMR (400 MHz, CD 3 OD) a 8.18 (d, J = 10.8 Hz, 1 H), 4.76-4.71 (m, I H), 4.17-4.12 (7 114)., 4 09 (s, 1 H), 3.96-3,86 (im l H), 3 67-3.53 (m I H), 3,55-3 53 (-n I H), 3,25-2.73 m, K H), 2 33-2 19 (m, 5 2 H), 1 68-1.59 (in, I H); MS (ES1) nz 59513 (M-H-) Compound 3-1-1 F
H
3 C' NCH 3 FC N BH4NH 2 3-1-1 1 -(BccamP io)cyclopropanecarboxylic acid (67.4 mg, 0.335 mmol), 0-(7 10 azab enztriazol-1-yl)-N.N,N N'-tetramethyluranium hcxafluorophosphato (127 mg, 0.335 mmol), and triethy lamine (0.078 muL. 0.56 mmol) were stirred in DM1F (I mL) for 30 minutes. Compound 9 (50 mg, 0 112 mmol) was added. After stirring overnight, the reaction mixture was purified directly by preparative reverse phase HPLC pur i fiction on a Waters Au[opurification system using a Polymnerx 10p RP-y 15 100 R column [3D x 21.20 mm, 10 micron, solvent A: 0.05N- HC, solvent B: CH3CN, gradient elution with 0-->50% IB over 10 min; mass-directed fraction collection]. Fractions with the desired MW were collected and freeze-dried. The material was repurified by preparative reverse phase 1]PLC purification on a Waters Autopurification system using a Polymerx 10 [ RP-y 100 R coluin 20 [30 x 2120 mm, I 10micron, solvent A: 0,05N IlCl, solvent B: CHCN, gradient elution with 0->[00% .B over 15 rnin; nass-directed fraction collection. Fractions with the desired MW were collected and freeze-dried, This gave 42 mg of compound 3-i-I (59%, ~80% pore) which was used without furher purification.: MS (ESI) n/ 631 41 (M+H L). 25 Compound 74 - 89 F H' N'CH, H H: H2N HN OHO O1 C 74 Compound 3-1-1 (42 mg. 0.067 mniol, -80% pure) was stirred in 4M 1RC in 1,4-dioxaue (5 niL) overnight. The reaction mixture was concentrated in der reduced pressure and was purified by preparative reverse phase HPLC purification on a 5 Waters Autopurification system using a Polymerx 10 p RP-y 100 R colunm [30 x 21.20 mm, 10 micron, solvent A: 0.05N HG, solvent B: CHaCN, gradient elution with 0- 50/ B over 10 min; mass-directed fraction collection], Fractions with the desired MW were collected and freeze-dried. The material was dissolved :n MeOHl (I ml.), and the solution was added dropwise to vigorously stirring diethyl 10 ether (200 mL). The resulting solid was collected by filtration on a pad of Celite. fhis was washed with diethyl etheir (3 tin es), and the solid was dissolved in MeOI and concentrated under reduced pressure. The material was freeze-dried, yielding 25.8 ng of compound 74: '11 NMR (400 MI-z, CD 3 OD with I drop DCl) 6 8.00 (d, J= 70 Hz, i ), 4.05 (s, 1 11), 3.20-2.85 (in, 9 H), 2.36-2.06 (m, 2 1), 1.70-152 (i. 15 3 H), [.35-1,22 (in, 2 H); MS fSl) rt 53 1.33 (M+H-) Compound 75 H H I H3 O
-
HEH N N C, NH2 H H 75 20 To a dichloroniethane (5 mL) suspension of compound 9 (0.260 p, 0.50 mmol, 1.0 equiv) at A was added triethylaime (0 139 nL, 1.00 mmiol, 2.0 equiv). The reaction was stirred at rt until form a clear solution. Methylisocyanate (89.4 IL, 1.50 mrno1, 3.0 equiv) was added to the reaction mixture dropwise. The reaction was allowed to stir at 25 'C for I h Additional methylisocyanate (45 piL, 0.75 mmol, 1.5 25 equiv) was added and stirred overnight. LCMS indicate there are still starting material preserri. The solvent was removed under vacuum to give the crude 75. The crude products was purified by HPLC on a Polymerx 10 p RP-7 100 R column - 90 [30 x 21.20 mm, 10 micron, solvent A: 0.05 N HCl. solvent B CH 3 CN, sample in 2.0 mL (0 05 N HCl), gradient elation with 15-+65 % B over 15 rnin, mass-directed fraction coileclioi] to yield te desired product 75 as a yellow solid (80 mg, 31.7 %): ' NNR (400 MHz, CTh OD) 5 8.12 (d, J= 11.4 iz, 1 14), 4.07 (s, 1I1), 3.04 (s, 5 3 H), 2.96 (s, 3 H), 3.13 -2.93 (in 3 H), 2.77 (s 3 1), 2.27-2,15 (m, 2 H), L69- 157 (I, I H); MS (ESI) m/z 505.41 (MNt H). Compound 76 HH H2 N N H 10 76 Compound 9 (20 mig, 0.045 Immol, LO equiv) in THT was added NC 2 CO, (9.5 mg, 0.089 iniol, 2.0 equiv) and 0.1 mL benzoyl chloride solution (54 U L in I mLf, TIF, 0.047 inmol, 1.05 equiv), The reaction mixture was stirred at room temperature for 1 hour. LC-MS dialysis indicated the strong material was 15 consumed completely. H{Cl/iMeOH (1 iL, 4 N) was added to the mixture at 0 C and stirred for 2 min. The mixture was concentrated Under vacuum, the residue was purified by reverse phase IIPLC to afford product 76 (5.5 mg): IH NMR (4100 MW, CD30D) 5 8.23 (d. J= 10,8 Hz, 11 ), 7.97 (d, J- 7.6 liz, 2 H), 7.66-7.54 (i, 3 HI), 4. 11 (s, 1 H), 3,21-2.90 (m, 9 H), 2 37-2.24 (m. 2 H), 1 72-1.66 (in, I FH'); MS (ESI) 20 mwz 552 1 (MIl). Compounds 77 - 83 were prepared similarly to compound 76 using the corresponding g acid chlorides. 25 Compound 77 H Hr FNC NNOH 77 - 91 II NMR (400 Mlz, CDaOD) 6 8.25 (s, I I), 8.21 (d, J= 8.0 1 z, 11), 8.14 (d, J= 10.4 Hz, I H), 7.92 (d, - 8.0 Hz, I H), 7.76 (t, J= 8.0 Hz, I H), 4 08 (s, H), 3.21-2.89 (in, 9 H), 2.35-2,22 (n, 2 H), 1.7 1- 161 (m, I l); MS (ESi) pny 620 1 (M+H)I. 5 Compound 78 F HCl 'NCH, F H3C0 0 0 78 A1 NMR (400 MHz, CD 3 OD) 6 8 10 (d, J= 10.8 Iz, 1 H), 7.41-7.33 (m, 3 H), 7.09-7 07 (m, I H), 4.00 (s I H). 3.78 (s, 3 H), 3.12-2.86 (m, 9 H), 2.23-2.13 10 (n, 2 H), 1 60-1.50 (in, 1 H); MS ([S1) m5i 582 1 (M+H). Compound 79 F IH '.C H 3
OHNH
HOH 0 02 0HO 79 ri NMR (400 MHz, CD-OD) 8 8,12 (d, J - 10.8 .Hz, 1 H), 7,89 (d, . = 3.2 15 Iz, I H), 7.78 (d, J 4.8 Hz, I H), 7.22 (t, J - 8.8 Hz, I H), 4 10 (s, 1 11), 3.20 2.98 (im, 9 H), 2.36-2.20 (m. 2 H), 1.68-1,61 (m, 1 H); MS (ESI) m/± 558 11(M+H). Compound 8.) F H'N' CH # H' O H HO O' o o so 20 'H NMIR (400 MHz, CD 4 OD) 5 9.34 (s, .1f), 9.04-9.00 (in, 2 11), 8.20-8.15 (m, 2 H), 4.07 (s, I H), 3.27-2 94 (m, 9 TI), 2.34-2.18 (n 2 H), 1.68-1.59 (n, I HI), MS (ESI) mlz 553. 1 (-M ru1) Cornpou)d 81 -92 F N 1H C H OH 0 4+ '1 NNIR (400 MHz, CD-OD) 6 8.13-8,06 (mn, 2 H'), 7.98 (d, J= 7.6 Hz, I H ),7.77 (d, J=7,2 Hz, I H ),767 (tJ= 8.0Hz. 1H-), 4 01 (s, 11H), 3,26 (s, 61-H), 3, 14-2 8 (m, 9 H), 2,27-2.13 (m 2 H), 1.64-1 52 (m, 1 H); MS (ESI) n/z 595.1 5 (M+H). Compound 82 F HsC N II HNC NH 2 II HOH c 2 0 cOH, 82 II \MR (400 MHz, CD0D) 6 8.08 (d, J = 108 I Iz, I II), 7.98 (d. J- 8.4 10 Hz. 2 14), 749 (d, J = 8.4 Hz, 2 11), 102 (s, I H), 3.19 (s, 6 H), 3 12-2.88 (m, 9 11 2.24-2,13 (m,2 H), 1.60-1.51 (n, 1 H); MS (ESI) mz 595.1 (N i1) Compound 83 F H 3 C.. .. 0H3 H0C N NN H 1- o ov? a 83 15 'H NMR (400 M tz, CD 3 OD) 6 8 19-8. 14 (, 2 14), 8.05 (d, J=8.4 I Iz, I H), 7 91-7.89 (m, 1 1H), 7.76-7.74 (m 1 II) 4 12 (s, I 11), 3.32 (s, 6 H), 3.21-2.96 (n, 9 H), 2.4 1-1 98 (mn, 2 1), 1.72-1.59 (m, 1 ); MS (ESI) m z 595.0 (M-+H). Conpo-nd 84 F H; 20N OH ~~NO, OH 0A oi[o 20 84 - 93 Compounc 9 (20 mg, 0.045 mmo1, 1.0 equiv) in THF was added DIEA (11.5 rug, 0 089 mmol, 2.0 equiv) and 2-thiophenesulfonyl chloride (12.2 mg, 0067 mumol, 1.5 equiv) The reaction mixture was stirred at room temperature for 20 hour. LC-MS analysis indicated the starting material was consumed completely 5 IIC/McOT (1 mL, 4' N) was added to the mxture at O" and stirred for 2 rnin. The rnixture was concenlrated under vacuum, the residLe was purified by reverse phase HPLC to afford compound 84 (2.0 mg): iH NMR (400 MHz, CD 3 OD) S 7.75 (dd, J = 5.2,1 1z,1 11-), 7.59 (d, J= 2.8 Hz, 1 Fl), 7.52 (d,J 10.4 Hz, I H), 709 (tJ= 4.4 Iz, I .H), 4.07 (s, I H), 3. 1 1-2.92 (m, 9 H), 2.30-2.18 (n 2 H), 1.68-1.58 (I, 1 10 1.); MS (ESI) m/z 593.9 (M+H). Compounds 85 - 87 were prepared similarly to compound 84 using' the corresponding sulfonyl chlorides. 15 Compound 85 F HsC. C~ OH Ha l N i NH? OHO Ohf 0 85 'H NNIR (400 \MHz, CDsiOD) 5 7.44 (d, J 10.0 Hz, I H), 4.10 (s, I f), 3.21-2.90 (m. 1 2 1), 2.34-2.22 (m, 2 F), 1.67-1.61 (m, 1 1) MS (ESt) m/z 526.1 20 Compound 86 F H.-C.. CH, H-, NH N'H, K~~JOH OH OFO H NMR (400 MHz, CD30D) S 7.82 (d, J= 7.6 z, 2 Fl), 7, 58-7,46 (i, 4 H), 4.07 (s, 1 H), 3.10-2.92 (m, 9 H), 2.35-2.25 (m, 2 Hl), 1 65-1.55 (in, I 1-H); MS 25 (ESI) mvz 552. 1 (M--H). Compound 87 - 94 clH S c N C
H
0 OH OH V 0HZ O O 0r 97 'HI NMR (400 Nlz, CD30D) 6 7.72( J = 5.6 Hz 1 H), 7.62 (d, J 7.6 Hz, I H), 7.50 (d. -- .4 Hz, I H), 7.41-738 (m, 2 H), 3,97 (S 1I), 3,C3-2.82 (nm, 9H), 5 2.19-2.06 (m, 2 11), 1 53-1 50 (m, I A); MS (ESI) m/z 622. (M+-H). Example 4. Synthesis of Compounds of Structra Formula (I), wherein X is hydrogen and Y is -NHI-C(O)-heterocvcly or -NI-C(O)-hetermaryl Scheme 4 0 F N N A A:. H H = X3' H H -N . 'N 0- - 1. 11|N .N H2NN or R^ rR N BOr 0110 ONo Y H nO 0 HO 0 On 2-6 OTB N 4-1 0T5,S depreotecin when RCOC I 2 R s a protecting F N" F H <H HH H a 0 NN R Nor2 H H O H0 -r"R H BnO 0 HO 0 O r 4-4 01063 4-2 CTBS 1) aq H: 1; aq IF 2) H/Pd-C 2) H,/Pd-C F N F "H.. ..GH3 R H (or R) H N OH 0 HC 0 RY RH OH 0 . 4-5 4-3 In Scheme 4 R represents heteroaryI and R2 is R' as deflned i n Structural Formula (A) For certain compounds made by Scheme 4 and described belov, Rz is hydrogen and R. and R are taken together with the carbon and nitrogen atoms to 15 which they are respectively bound to form an optionally substituted 4-7 rmnmbered - 95 saturated helerocyclyl. It will be readily apparent to those of skill in the art, however, that this Scheme 4 will also be useful to synthesize compounds where R,. R Y and R? are RH, R and RV respectively, as defined in structural formula (A). The following compounds were prepared according to Scheme 4 Compound 88 F N H H aOH HN NH, ior &JfrIH OHO0 OHP 0 To a suspension of 1-Fmoc-L-azetidine-2-carboxylic acid (135 mg, 042 mniol, 2.9 equiv), and (2-(7-aza- IH-benzotriazole-]-yl)-1,1,3.3-tetrauerhvluronium 10 hexafluoro-phosphate) (164 rng, 0.43 iml, 3 equiv) in TiF (1.5 mL.) was added trkiethylainie (60 gL, 0.43 mmol, 3 equiv). After 30 min, aniline 2-6 (106 mg, 0.14 mmol, I equiv) was added. After 18 h, the reaction mixture was concentrated under reduced pressure. Preparative reverse phase HPLC of the resulting oil was performed on a Waters Autopurification system using a Sunfire Prep C1.8 OBD 15 column [5 pm, 19 x 50 mnm; flow rate, 20 mL/muin: Solvent A: H20 with 0.1% HCOOI; Solyent B: CH3CN with 0.1% HCO2H; injection volume: 3 x 2.0 ml, (CH3CN); gradient: 80-100% B over 15 min; mass-dIrected fraction collection]. Fractions with the desired MW, eluting at 10 35-12.0 min, were collected and freeze-dried to provide 131 mg of a yellow powder. 20 To a solution of the above internediate in CH2C2 (2 ml) was added piperidine (500 L) After 30 min, the reaction solution was poured into aqueous pH 7 phosphate buffer and extracted with EtOAc (3 x 20 mL), The combined organic layers were dried (Na2SO.4), filtered, and concentrated under reduced pressure. 25 Purification of the resulting crude oil via flash column chromatography on silica gel (Silicycle, 5 g, 0 to 5 to 10 to 50 % EtOAc in hexane gradient) provided 47.6 rng of the intermediate Half of the above intermediate (24 mg) was dissolved in acetonitrile (1 nl), 30 and an aqueous solution of H F (50%, 200 L) was added. After 18.5 h, Lhe reaction -96 solution was poured into an aqueous Kj1HP0 4 solution (2.5 g in 2C mL) and extracted with FtOAc (2 x 25 mL), The combined organic layers were dried (NaSO4), filtered. and concentrated under reduced pressure, 5 Palladium on carbon (10%, 12.5 mu) was added to a sou tion of the above intermediate in dioxane:MeOH (1:1, 1 nL). The flask was fitted with a septum and evacuated and back-filled three times With hydrogen gas. Hydrogen gas was bubbled through the reaction solution for three minutes, and the reaction mixture was stirred under an atmosphere (balloon) of hydrogen gas For 4 5 h. The reaction mixture was 10 filtered through ectite to remove the palladium catalyst and concentrated under reduced pressure. Preparative reverse phase HPLC purification of the resulting oil was performed on a Waters Autopurification system using a Polvrmerx 10 RP-, 100 R column [30 x 21.20 nm, 10 rnicron., solvent A: 0 05N HCl in water, solvent B: CI-L;CN; injection volume: 3.0 mL (0,05N HCI in water); gradient elution 15 with 0--30% B over 10 min, then held at 100% fo- 5 min; mass-directed fraction collection] Fractions with the desired M W, eluting at 9.8-11.25 mi, were collected and freeze-dried. The resulting impure powder was purified via pireparative reverse phase HPLC as above wifti gradient elution with 15-+50% B over 12 min, then held at 100% for 3 min; mass-directed fraction collection. Fractions with the desired 20 MW, outing at 6 5-8 0 min, were collected and freeze-dried to yield 2.0 rng of compound 88 (5%): jH NMR (400 Nil z, CD 3 OD) 6 8.25 (d, J = 11.0 Hz, 1 H), 5.29-5.24 (n I H), 4.20-4.1 1 (in, I H), 4.09 (s, 1 H), 3.19-2.89 (m11, 10 H), 2.69-2.56 (m I H), 2.33-2 19 (m, 2 1), 1.68-1.56 (m, I H); MS (PSI) mp)/z 53.30 (M+1). 25 Ni-methyl-L- azet id ine-2-carboxilic acid HOc H To a suspension of L-azetidine-2-carboxylic acid (290 mg. 2.87 mmol, I equiv) in MeOH (3.6 mL), was added aqueous formaldehyde solution (37%, 235 L, 3.15 nimol, I1 equiv) and palladium on carbon (10%, 76 mg). The flask was fitted 30 with a septum and evacuated and back-filled three times with hydrogen gas The reaction was stirred under an atmosphere (balloon) of hydrogen gas for 19 h, and was filtered through elite to remove the palladium catalyst. The resulting solution - 97 was concentrated under reduced pressure, concentrated from toluene three Pines and dried under vacuum to afford N-methyl-L-azetidine-2-carboxilic acid: IH NMhiR (400 MHz, CD 3 OD) 3 4.50 (tJ- 9.5 Hz, I fH), 3.96 (dt,.J= 4.3, 9.8 Hz, I H), 3.81 (q,,J 9.8 z, 1 H), 2.86 (s, 3 1), 2.71-2.60 (m, I H), 2.50-2.38 (m, 1 11). 5 Cormpour d 4-1-1 F CN' H ;
H
3 Cs| N N 1 HH BnO 0 HO 0 O 4-1-1 To a suspension of aniline 2-6 (302 mg, 0.408 mrnol, 1 equiv) and A-methyl I -azelid ine- 2-carboxilic acid (148 mg, L28 mmol, 3. 1 equiv) in CH 2 Cl 2 (6 mL) 'was 10 added O-(Benzotriazol-1-vl)-N,N, NKN-tetramethyluroni umn tetralfluoroborate (395 mg,] 23 mmol, 3 equiv) and DIEA (285 yL, l,6A mmol, 4 equiv). After 16.5 h, the resulting orange solution was concentrated under reduced pressIe and. pLrified via preparauve reverse phase HPLC on a Waters Autopurification system using a Sunfire Prep C18 OBD column [5 pa 19 x 50 mm, flow rate, 20 nlmin; Solvent 15 A: H20 with 0.1% HlCO1-1H; Solvent 13: C[ 3 CN with 0.1% HCOlH, injection volume: 4 x 2.5-3.0 mL (CH,-CN); gradient: 50-90% B over 15 mn; mass-directed fraction collection]. Two sets of fractions with the desired MW, eluding at 4.6 6,5 min and 6,5-9.4 min, were collected separately and freeze-dried to provide 147 mg of 4-1-1 (43%): 'Hj N4R (400 MHz, CDCl) 8 16.04 (s, I H), 10.10 (s, I H), 20 8.48 (d, J= 11.0 Hz, I H), 7.54-7.48 (mr, 4 H.), 7.40-7.32 (m, 5 H), 5 36 (s, 2 HI), 4.99 (d,1J- 9,8 Hz, [ H), 4.90 (d,J = 9.8 H1z, I l), 3.96 (d, 1= 0.4 Hz, .1 13.54 (t J= 7.9 4z, 1 H), 3.39-3.34 (in, 1 11), 3.25-3.19 (in, I H). 3 05-2.92 (m, 2 It), 2.58-2.36 (i, 10 H), 2.23-2.06 (m, 4 H), 0.81 (s, 9 11), 0.28 (s, 3 H), 0.11 (s, 3 H); M S(1) mcz 837.37 M I I 1). 25 Compound 89 FH30'N'CHa H H3 H O H OH 89 - 98 To a solution of 4-1-1 (147 mg, 0. 175 mmol, 1 equiv) in dioxane (3.5 mL) was added an aqueous solution of F (50%, 750 IL), After 4 h, the reaction solution was poured into an aqueous K-HPO i solution (9 g in 90 mL) and extracted with EtOAc (2 x 50 nL). The combined organic layers were dried (Na 2 SO) filtered, and 5 concentrated uLnder reduced pressure to provide 128.4 ing of a crude yellow foam. The IF deprotectjon product (144 mng, 0.199 mmol, I equiv) was dissolved in dioxane:MeO-f (1:1, 4 mL), and palladium on carbon (10%, 43.5 mg) was added The flask was fitted with a septum and evacuated and back-filled three tines with 10 hydrogen gas. Hydrogen gas was bubbled through the reaction solution for three minutes, and the reaction mixture was stirred under an atmosphere (balloon) of hydrogen gas for 3,25 1 The reaction mixture was filtered through celite to remove the palladium catalyst and concentrated under reduced pressure. Preparative reverse phase HTPLC of this oil was performed on a Waters Autopurification system using a 15 Polymerx 10 g RP-y 100 R column [30 x 21 20 mm, I0 micron, solvent A: 0.05 N 11C1 in water, solvent B: CH 2 CN; injection volume.: 2 x 3.2 ruL (0.05 N hCl in water); gradient: 10->35% B over 20 in a; mass-directed fraction collection], Fractions with the desired MW, elutiing at 6.10-8.40 min and 6 9-9.4 rin, respectively for each run, were combined. The pH1 of the solution at 0 "C was 20 adjusted (p-H 1.8 to pH 7.4) via dropwise addition of 0 5 M aqueous NaOF solution (approximately 7.8 mL.) and careful monitorimgr with an electronic pH meter. The aqueoLIs solution was extracted with CH2-sCl2 (3 x 60 mE) and the combined organic layers were dried (Na2S0 4 ), filtered, and concentrated under reduced pressure to provide 79.7 nig of compound 89 as the free base (0,146 mmot 73%). This yellow 25 solid was dissolved in MeOH (3 mL), and MeSO 3 H (19 pL, 0.292 inmol, 2 equiv) was added The solution was concentrated under reduced pressure, dried under vacumin, and freeze-dried from water to provide 105 mg of 89 as the dimesylate salt. 'iH NMR (400 MI Iz, CD 3 OD) 6 8.22 (d, J= 11.0 Hz, I H), 5.16 (1, J = 8.6 Hz I H), 4 21-4.12 (in, I H), 4.09-4 02 (n, 2 H). 3.1 7-2.85 (m, 10 H), 2.68 (s, 6 F, mesylate 30 R), 2.64-2.59 (m. I R), 2.34-2.15 (in, 2 H), 1.70-1.58 (in, 1 H), MS (EST) 0m/z 545.18 (M IH). General Propcedures for the Preparation of Compounds 90 - 94 - 99 To a solution of aniline 2-6 (1 equiv) in THF (0.05-0.09M) was added an acid chloride (3 equiv). The reaction solution was filtered through celite and concentrated under reduced pressure. The resulting oil was dissolved in dioxane (1 5 niL) and an aqueous solution of HF (50%, 200 pL) was added. Upon comply etion. the reaction was poured into an aqueous 1K 2 HP04 solution (2,6 g in 30 mL) and extracteod with EtOAc (2 x 25 mL). The combined organic layers were dried (NazSO4), filtered, and concentrated under reduced pressure. Palladium on carbon (10%) was added to a solution of this crude oil in dioxaie:MN.eOI1 (1:1, 1 ml), The 10 flask was fitted with a septum and evacuated and back-filled three times with hydrogen gas, and then the solution was degassed with bubbling hydrogen for 2 minutes. The reaction was stirred under an atmosphere (balloon) of hydrogen gas for 2 h The reaction mixture was filtered through celite to remove the palladium catalyst and concentrated under reduced pressure. The crude products were purified 15 by preparalive reverse phase HfPLC. Compound 90 F H c . H2 T) H Prepared by above general procedure with the following reagents: aniline 2-6 20 (2 L. mg, 0.028 mmol, I equiv), picolinoyl chloride hydrochloride (15.8 mig, 0.088, 3 equiv), with triethylamine (I1 .7 iL, 0.084 rmmol, 3 ecquiv), and 10% Pd-C (10 mg). provided a crude oil. Preparative reverse phase 1PL.C of the crude product was performed on a Waters Autopurification system using a Polymerx 10 [t RP-/ 100 R column [30 x 21.20 mm, 10 micron, solvent A: 0.05 N HCI in water, solvent B: 25 CH 3 CN; injection volume: 2.5 nmL (0.05 N HC in water); gradient: I 0--60% B over 20 rin; mass-directed fraction collection], Fractions with the desired MW, eluting at 14.8-16.4 min, were collected and freez'e-dried to provide 5.8 m of the desired compound 90 (37%): 'H1 NMR (400 MHz, CDOD) 6 8.73-8.69 (m, i Ti), 8,58-8.52 (t, I H), 8.27-8.21 (in, ] 11), 8,08-8.00 ( , I H)., 7,66-7.60 (m, ] 11), 4 09 (s, 1 H.), -100 3.29-2,92 (m, 9 1), 2.38-2.18 (m, 2 H), 1.72-1.60 (m, I H); MS (ESI) m z553.27 (M+If). Compound 91
H
3 0c CH HH: I H OH H 0 5 91 Prepared by above general procedure with the following reagents: aniline 2-6 (31 .0 mg, 0.042 rmmo L1 equiv), I -methlwpyrrole-2-carbonyl chloride (22 mg, 0. I 5 mmol, 3 equiv), and 10% Pd-C (10 ng), Preparative reverse phase HPL.C of the crude product was performed on a Waters AutopuTification system using a Polymerx 10 10 LL RP- 100 R column [30 x 21,20 mm. 10 micron. solvent A: 0.05 N ICl n water, solvent B: CTCN; injection volurne: 2.0 nL (0.05 N 1Ci in waler), gradient: 20-70% B over 20 mi. mass-directed fraction collection]. Fractions with the desired MW were collected and freeze-dried and repurified via the same system with the gradient 10-+60% B over 20 mn, Fractions with the desired MW, eluting at 15 15 5-16.5 min, were collected and freeze-drited to provide 2.5 mg of the desired compound 91 (11%): 'H-1 NMR (400 MIHz CD 2 OD) 5 8.20 (d, J= 11,6 Hz, I F), 6.98-6.86 (n, 2 H), 6.17-6.10 (m1, 1 11), 4 08 (s, I H), 3,94 (s, 3 F), 3.19-2.90 (m, 9 [1), 2.33-2.18 (m, 2 H) :80-I 56 (m, 11); MS (ESI) m/z 555.32 (M+H). 20 Compound 92 F H'C .CRs OHH HIC 92 Prepared by above general procedure with the following reagents: aniline 2-6 (3 [.0 rng, 0.042 nimol, 1 equiv), 5-iethylisoxazole-3-carbonyl chloride (19.0 mg, 0 13 nmmol, 3 equiv), and 10% Pd-C (10 ing), Preparative reverse phase HIPL C of the 25 crude product was performed on a Waters Autopurification system tiLngl a Polymerx 10 p RP-y 100 R column [30 x 21.20 mm, 10 micron, solvent A: 005 N HCI in water- solvent B. CH 3 CN;- injection volume: 2.8 ml (0.05 N FICI in water), gradient: - 101 10-460% 13 over 20 min; mass-directed fraction collection]. Fractions with the desired MW, eluting at 14.5-15.5 min, were collected and freeze-dried to provide 4.0 ing of the desired compound 92 (17%): 1 HNMR (400 MHz, CD 3 0D) a 8 32 (d, J= 11.0 Hz, 1 H), 6.59 (s, 1 H), 4.09 (s, I H).. 3.19-2.90 (ni, 9 11), 2.52 (s, 3 H), 2.34 5 2.18 (n, 2 H), 1.-7 -1.58 (m, I H); MS (ESI) nz 557.26 (MA-H). Compound 93
H
2 C, r-H OH H3CtH HoN H 1 r.~ NH> N rI 041 0 Or 0 93 Prepared by above general procedure with the following reagents: aniline 2-6 10 (30,0 rng, 0.041 mmol, I equiv), 1imethyl-I H-pyrazole-3-carbonyl chloride (16.8 rmg, 0 12 nimol, 3 equiv), and 10% Pd-C (20 mg). Preparative reverse phase F-HILC of the crude product was performed on a Waters Autopurification system using a Polymerx 10 lr RP-y 100 R column [30 21 20 mm, I'1 micron, solvent A: 0.05 N HCl in water, solvent B: C1CN; injection volume: 3.2 mL (0.05 N HC in water) 15 gradient: 10-60% B over 20 min; mass-directed fraction collection] Fractions with the desired MW, l uting at 12.5--145 min, were collected and freeze-dried to provide 11.2 ing of the desired compound 93 (49%): 'H NMR (400 M-Hz, CDOD) 5 8.38 (d, .J = 11.0 liz, 1 11), 7.68 (s, 1 1-), 6.82-6 76 (in, I I), 4.09 (s, 1 ), 3 99 (s, 3 1-), 3.16-2.90 (m, 9 TI), 2.3 I-216 (m, 2 H), 1 70-1.56 (rr I H); MS (ESI) m z 556.31 20 (M+-H)3 Compound 94 F H3c
C..M
3 H CH N NH 94 Prepared by above general procedure with the following reacents aniline 2-6 25 (30.0 mg, 0.041 mmol, 1 equiv), 1,3-thiazole-2-carbonyl chloride (17.8 mag, 0, 12 mmol, 3 equiv), and 1.0% Pd-C (15 ig) Preparaive reverse phase HPI.C of the crude product was performed on a Waters Autopurification system using a Polymerx - 102 10 p RP- 100 R column [30 x 21.20 mm, 1.0 micron, solvent A: 0 05 N CI in water, solvent B: CHCN; injection volume: 3.2 mL (0.05 N HC in water); gradient: 1.0-60% B over 20 min; mass-directed fraction collection]. Fractions with the desired MW, eluting at 14.6-17.0 mi, were collected and freeze-dried to provide 5A4 5 mg of the desired compound 94 (23%): 'H NMR (400 MHz, C.D3OD) 6 8,38 (d, J 11.0 Hz., I H), 8.02 (d, J= 3.0 1lz, I 1), 7.95 (d, J= 2 4 Hz7, 1 ), 4.09 (s. I H), 3.20-2.90 (n, 9 1-1), 2.34-2.17 (in, 2 H), 1.70-1.56 (m, i H); MS (EST) mvz 559.23 (\I :H1). 10 Example 5. Synthesis of Compounds of Structural Formula (A), wherein Y is -N(R^)(RD), or -NIR-SO 2 r(C12')N(R A)(R"), Scheme 5 a [HO -CH, 1A 3 0. OrH, NF N' H aU H2 F HC,.CN HF lC-CN NB H!PdC R NH Bno H H 10 O H OHO HO 00 5-1 OTBS 5-2 RCHO/HOAc Na(O/c4aBH F HaC N' FC H, H H H H HN N rO 0 HO 0 8n H o 2 6 OTBS 5-5 OTOS SAr-X R RN F HN CH) F HJC'N'CHs H H H Hv N"N 0 N Ar N N N
OH
8 C BnO O HO 0 CR RV HO:C 5.3 OTS i OTBS 1) aq HF 1)qIF 2) H/Pd-C 2) H-VPd-C F H3C CH, F IHC'N 'CH H H H H' IOH .OH Ar-N NH 2 R NH2 H h - . h OH CH U HO ) v OH 0 HO 0 0 5-4 5-7 - 103 In Scheme 5. R represents -(C -Cs)alkyl, -(Cu-Cs)alkylene-carbocycyL -(CO-C5)aikylenc-aryI, -(Co-CalkLylene-heterocyclyl1, -(Ce-Cf)alkvlene-heteroarVl,
-(C;-C
3 )alkylene-N(Rj)(R); Ar represents an aryl or a he teroaryl group; and R and R are RA and RE, repectively, as defined in Structural Formula (B). 5 The following compounds were prepared according to Scheme 5. Compound 5-1-1 HH H U ~ N U N H-G N H B On arno 0 HO 0 U 5-1-1 OTPS Compound 2-6 (150 ig, C.203 rmol 1.0 equiv) was dissolved in 1,2 10 dichloroethane (3 mL). HOAc (58.1 pL, 1,01 mmol, 5 equiv.) and isovaleraldehyde (32.9 ., 0.304 nmol, L5 equiv) were added The mixture was stirred for I h. Na(OAc) 3 1Bl (129 mg, 0.609 rmol, 3.0 equiv) was added and the resulting mixture was snted for another hour. Ihe mixture was washed with Hi-0 (10 mL) and concentrated to give crude 5-1-1 (250 ng), which was used for the next step without 15 further purifEcaion: MS (ESI) m/Z 810.59 (M IH). Compound 95 F H F-N-H H3 H Hac NHz H H 0 Oh?) 95 Aqueous 1F (0.3 mL, 48-50%) was added to a CHICN solution (1.5 ml) of 20 5-1-1 (250 mg crude) in a plastic vial at 25 C. The reaction was stirred at 25 'C for 18 hrs. The resulting mixture was poured into an aqueous solution (10 mL-) of
K
2 H-PO4 (2 g). The solution was extracted with EtOAc (3 x 15 mL) The combined EtOAc extracts were dried over sodium sul 'ate and concentrated to give the crude intermediate (155 mg). 25 10% Pd-C (20 mg) was added to a di oxanefMeQH solution (4 nIL, L1) of the above crude intermediate. 1CI/MeOH (0.5 mL, 0.5 N) was also added. The reaction mixture was stirred under Hz{ (balloon) at 25 'C for 2hrs and filtered - 104 through a pad of Celite. 'lhe filtrate was concentrated to give the crude product 144 mg. The crude product was purified by HFLC on a Polymerx 10 p RP--{ 100 R column [30 x 21.20 mm, 10 micron, solvent A. 0.05 N IC], solvent B: C13C:N, sample in 2.0 iL (0.05 N HQ), gradient elution with 10--+100% 3 over 15 min, 5 mass-directed faction collection] to yield the desired product 95 as a yellow solid (82 ng, 78 M. 2 steps): HjI NMR (400 MI Iz, CD0D) 6 7.44 (d, J - 9.2 Hz, 1 i), 1.12 (s, 1H) 3.42-3.37 (n, 2 H), 3.05 (s, 3 11) 2.97 (s, 3 11), 3.2] -2,97 (n, 3 1), 2.39-2 30 (rm, I H), 2 29-2.22 (m, I H), L ,79-1.59 (n, 4 H), 0.98 (d, J = 6.4 Hz, 6 1H); MS (ESI) m/n 518 43 (M--1l). 10 Compounds 96 - 101 were prepared similarly to compound 95 using the corresponding aldehydes in the reductive alkylation step. Compound 96 F H 3 C N'CH OH H H HC N - NOH2 H OHO HOM 96 15 'H NMR (400 MHz, CDs 1 D) 6 7.39 (d, J= 9.2 Hz, 1 [H), 4.10 (s, I Fl), 3.34 (t, J= 7.8 Hz, 2 H), 3.04 (s, 3 1-) 2.96 (s, 3 1) 3,21 -2.95 (m, 3 T), 2,35 (t, f= 13 7 Hz, 1 H), 2.27-2.20 (in, 1. H), 1 82-1.72 (m, 2 HA), 1.71- L.60 (m, I Hl), 105 (t, J= 7.4 Hz, 3 .); MS (1SI) w 490.32 (M+1). 20 Compound 97 S H.. ON H H ,o H3CN . .NH 2 H OH 0 TAp 97 - NMR (4:10 MI-lz, CD 3 OD) 6 7.34 (d, J= 9.2 Hz, I H), 4, 10 (1, .), 3 34 S- 7.8 Hz, 2 11), 3.04 (s 3 l), 2.96 (s, 3 H1), 3.24 -2 95 (in, 11 H), 2.33 (t, J 13.7 Hz, I H), 2.27-220 (in, I H), 2.11-1.98 (m, 1 i), 1.71-1.60 (m 11H), 1.08 (d, J 25 = 9 Hz, 6 H); MS (ESI) riz 504.46 (M--H). CO(mpound 98 - 105 HC. .. N H H: N ' OH N "NH 2 OH 0 CO- O 98 -1 NMR (400 MHz, CD3OD) 37,43 (d, J= 87.Hz, I H1), 4.10 (s, I H), 3.34 (,J= 78 IHz. 2:H), 3.04 (s7 3 H). 2 96 (s, 3 H), 3.28 -2.95 (m. 11 H), 2.41-2.31 (m, 'I ), 2,27-2,20 (in, I H), 2.11-1.98 (in, I 1.), 1.72-1.60 (i, 1 H), 1 20-1 1 (m , 5 H), 0 74-0.68 (m, 2 H), 0.43-0.38 (m, 2 I1); MS (ESI) i/z 502 40 (M-H). Compound 99 F H H HNH H H 01 OFPt 99 1 1 NMR (400 MHz, CD 3 0D) 6 6 97-6.89 (n, I H), 4.07 (s, [ H), 3.34 (t, 'J 10 7.8 Hz, 2 H), 3 03 (s, 3 H): 2.95 (s, 3 H), 3 14-2.92 (m, 1 11), 2.30-2.15 (m, 2 H), 1.89-1 59 (i, 7 1), 1.38-1.20 (m, 3 H), 1,11-1.00 (m, 2 11), MS (ESI) m/z 544.50 Compound 100 N"' >rN NF1 2 HC H HCHe OH O OF O 15 100 H NMIR (400 MHz, CD 3 OD) 6 6.83 (d, J 10 5 Hz, I H), 4,06 (s, I l), 3.34 (t, J= 7.8 Hz, 2 14), 3,03 (s, 3 H), 2.95 (s, 3 H), 311 -2,93 (m, 5 H.), 2.27-2.14 (m, 2 II), 1,67-] 57 (m, I H), 1 04 (s, 9 H); MS (ESI) n 518.48 (1M-H1). 20 Compound 101 F -,3C, CH H HO H-C N
CH
3 OH 5 Oo a 101 - 106 'H NMR (400 MHz, CDhOD) 87.46-7 42 (m, I H), 4.15 (s, I H), 3 33 (s, 6 H), 3 04 (s, 3 HT), 2.96 (s, 3 H), 3 17 -2.95 (m 3 H), 244-2.34 (n, 1 H), 229-2.22 ([, H), 1.71 -. 1.60 (m, 1 H); MS (ESI) i z 476 29 (M+H). 5 Compound 102 F [N GOH 0 U HUT 01i N. H CH OH 0 0& 2 ' 102 Prepared similarly to 95 using tBuN(Cb.)CH,CHO: 'H NMR (400 MHz,
CD
3 OD) 6 6.72 (d, J- 11.0 Hz, I H), 4.07 (s, 1 14), 3.54-3.46 (n, 2 H) 3.26-3.19 (m, 2 H), 3.03 (s, 3 H.), 2.95 (s, 3 H), 3 14-2,92 (m, 3 i), 2.23-2 14 (m, 2 H), 1.67 10 1.55 (m, I H), 1.38 (s, 9 H); MS (ESI) m z 547.51 (M-i+H). Compound 103 r H OCN-CH 3 H Or1 0 0 103 Compound 103 was also isolated from the preparation of 102 'H NMRI (400 15 MHz, CD:OD) 6 6 71 (d, J= 1.0 Hz. 1 H), 4.07 (s, 1 H), 3.47 (t.,J 6.0 Iz, 2 I, 3.17 (1, J 6,0 Iz, 2 H), 3.03 (s, 3 H), 2,95 (s, 3 H), 3.13-2.92 (m, 3 ), 2.23-2. 12 (ni, 2 1), L 66- 1 .54 (m, I f); MS (ESI) tnm 491.42 (MiH). Compound 5-3-1 F H30',.H F N H H NN a N BnO 0 HO 0 O ri 20 3-1 OTBS A vessel containing aniline 2-6 (18,2 ng, 0.024 miol, J eq'iiv), Pd2dba (3.0 mg, 0.0033 mmol, 0.13 cquiv), Xautphos (3.4 ng, 0,0059 immol, 0.25 equiv), K 3 P0 4 (40 mg, 0.188 nnol, 7.8 equiv) and 4.6-clichloropyrimi dine (6 img, 0.044 mimol L8 -quiv) was evacuated and back-filled with nitrogen goas three times. Dioxane 25 (500 L) was added, and the reaction mixture stirred vigoorously and heated at 80 C - 107 for 4.5 h. The reaction mixture was filered through celite and concentrated under reduced pressure. Preparative reverse phase FPLC of the resulting yellow oil was performed on a Waters Autopurification system using a Sunfire Prep C18 OBD column 15 m, 19 x 50 mm; flow rate, 20 mL/mi; Solvent A: H2 0 with 0. 1% 5 H1CO4H; Solvent B: C1 3 CN with 0 1% THCO,-H: injection volume: 1.8 mL (Cl CN); gradient: 80 ->100% B over 15 min; mass-directed fraction collection]. Fractions with the desired MW, eluting at 9.2-9.8 min, were collected and freeze dried to provide 7.5 mg of compound 5-3-1 (37%), HFl NMR (400 M[-Iz, CDCh) 3 15 97 (s, I 11), 8.48 (s, I H), 8.33 (d,J - 5.5 Hz, 1 H1), 7.52-7.16 (m, 2 1), 7,40-7.28 10 (n, 8 H), 7 07 (s, I1 H), 6.11 (s, 1 H), 5.34 (s, 2 H), 4.97 (d, J= 11.6 Hz, 1 HO, 4.88 (d, J1 1,0 Hz, I H), 3.95 (d, J= 10.4 Hz. 1 H), 3.28-3 19 (m. I [-H), 3,09-2.98 (m, I H), 2.61-2.54 (i, I H), 2.54-2,39 (i, 8 11), 2.16 (d, .J- 14.6 Hz, I H), 0.83 (s, 9 i1), 0.28 (0, 3 H), 0 14 (s, 3 H); MS (ESI) m/z 852.57 (M\I 11). 15 Compound 104 F HGC'N.,CH, H H' N~N a.,,OH
-
N HN~ 104 To a solution of 5-3-1 (7.5 mg, 0.0088 rmnol, 1 equvl) in dioxane (1.4 mit) was added an aqueous solution of HF (50%, 200 pL). After 15,5 h, the reaction solution was poured into an aqueous K;2HPO 4 solution (2.4 g in 20 ml,) and 20 extracted with EtOAc (2 x 20 ml). The combined orgnirc layers were dried (Na2SO4), filtered, and concentrated under reduced pressure. Palladium on carbon (10%, 10 mg) was added to a solution of this oil in dioxane:MeOH. (1:1, 1 mL). The flask was fitted wth a septum and evacuated and back-filled three times with hydrogen gas. Hydrogen gas was bubbled through the reaction solution for three 25 minutes, and the reaction mixture was stirred under an atmosphere (balloon) of hydrogen gas for 2.5 h. 'he reaction mixture was filtered through celite to remove the palladium catalyst anid concentrated under reduced pressure Preparative reverse phase HPLC purification of the resulting oil was performed on a Waters Autop.ification system using a Polymerx '10 RP-y 100 R column 30 [30 x 21.20 mm, 10 micron, solvent A: 0.05 N 1C11 in water, solvent B: CH.CN; - 108 injection volume: 2-0 mL (0.05 N HC in water); gradient elation with 10-+50% B over 10 mii then held at 100% for 5 min; mass-directed fraction collection]. Fractions with the desired MW, Cl uting at 6.90-7.80 min, were collected and freeze dried to provide 2.2 mg of 104 (48%). '11 NMR (400 MHz, CD 3 OD) 6 8.83 (s, .11), 5 8.37-8.25 (m, 1 H), 8.18-8.05 (mn, 1 11), 73 0-7.20 (n, 11H), 4.10 (s., I I), 3.20-2,90 (n, 9 H), 2.40-2.29 (in, I 11), 2 27-2.19 (m, 1 H), 1 72-1 58 (m, 1 H); MS (ES1) nz 526.31 (M+H). Compound 105 F Htc 'Hz H H HN 100 1H To a solution of aniline 2-6 (30 0 mg, 0 041 imol, I equiv) in 1,2 dichloroethane (500 L) was added pyridine (16.3 L, 0.20 mol, 5 equiv) and 2 chloroethanesulfony chloride (21 LL, 0,20 mmol, 5 equiv). The reaction vessel was sealed and heated to 45 *C, After one hour, the reaction, was a solid yellow gel, and 15 another 500 pL 12-dichloroethane was added to form a suspension and the reaction was scaled and healed to 45 *C. After 18.5 h pyrrolidine (68 pL, 0.82 mmol, 20 equiv) was added and the reaction heated to 45 C for 2,5 hours. The solution was poured into aqueous pH 7 phosphate buffer (8 ml,) and extracted with EtOAc (2 x 25 niL), The combined organic layers were dried (Ni2SO4), filtered, and 20 concentrated under reduced pressure. To a solution of this crude oil in CH 3 CN (.8 rmL) was added an aqueous solution of HF (50%, 300 Ld,). After 15 I the reaction solution was poured into an aqueous K2B PO solut ion (3.6 g in. 30 iL) and extracted with EtOAc (3 x 15 mL). The combined organic layers were dried (Na 2 SO4), filtered, and concentrated under reduced pressure. Palladium on carbon 25 (10%, 8.4 mg) was added to a solution of this oil in dioxane:MeOH (1:1, 1.2 mL). The flask was fitted with a septum and evacuated and back-filled three times with hydrogen gas, and the reaction mixture was stirred under an atmosphere (balloon) of hydrogen gas for 1.5 I. Another 10 mg palladium catalyst was added and the reaction was evacuated and back-filled with hydrogen gas as before, After 6 h, the 30 reaction mixture was filtered through celite to remove the palladium catalyst and - 109 concentrated under reduced pressure, Preparative reverse phase HPLC purification of the resulting oil was performed on a Waters Autopurification system using a Polymerx 10 V RP-y 100 R column [30 x 2120 mn, 10 micron, solvent A: 0.05 N H-C in water, solvent B: C.'H 3 CN; injection volume: 3.5 mL (0.95 N HC in water); 5 gradient elulion with 0-100% B over 10 mn, then held at 100% for 5 rmi; mass directed fraction collection]. Two sets of fractions with the desired MW, eluding at 6,3-7 1 min and 8.7-9.3 min, were collected separaLely and freeze-dried to provide 9,7 rng of crude compounds 105. Purification via preparative reverse phase HPLC with gradient elAon with 20.-.-70% B over 20 min; mass-directed fraction 10 collection] provided 3.3 mg of pure 105 (13%): ' 4MR (400 MHz, CD 3 OD) 6 7_44 (d,J= 9.8 Hz, 1 H), 4.09 (s, I 11) 379-3.65 (m, 4 H), 3.63-3.56 (m, 2 1H), 3.18-2.90 (n. I l), 2 35-2.26 (i, I H), 2.26-2. 10 (i, 3 H), 2.10-1.96 (i, 2 H), 1.69-1 59 (m, I H), MS (ESi) m'iz 609.36 (M-H). 15 Compound 106 F HsC., 2Hs U Oil -? NH2 106 Compound L06 (1.7 ng, 7%) was also isolated from the preparation of compound 105: 'HI NM/fR (400 MH z CD 3 0D) 6 6.7 L (d, J= 11.0 Hz, 1 H), 4.06 (s, 1 H), 3.67-3.60 (m, 2 11), 3.38-333 (i, 4 1), 3.09-2.90 (m, 9 11), 2,24-2. 13 (vs 2 H), 20 1.95-1.9] (i, 5 Hl), 190-1.85 (i, 1 1-1), 1.68-1.55 (in, I H) MS (EST) in/z 609,36 Example 6. Synthesis of Compounds 107 and 108 Scheme 6 25 - 110 F F F C~a HCHO . : i o2 ~ 2N CO 2 Ph H2/Pd-C H 3 C C2[H' Ca HN 02hN OOQ 2 PhN OP OBn H OH BoU O&oc 2-3 6-1 6-2 a) LOA)TMEDA b)enn F. HaC N'C~ H H H H NN HN NNqH OH b HO u 01n Bo O O o OBn 6.4 -3 OTBS H/Pd-C F HaCON'a OH / -i HICNCHs OH H HNOH HOC NH2 HTAJ _ N N - 2 H OhO-10 2IC a OH O HO U OH 3 OH 0 O 107 108 The following compounds were prepared according to Scheme 6. 5 Compound 6-1 F H3C..N ''CO>Ph H OH 6-1 Compound 2-3 (5,0 g, 1.4.25 mmol, L,0 equiv) In MeOH (20 m-L) was added a aqueous solution of HCHO (2.3 g, 37%, 28,50 rTlnol, 2.0 equiv) and palladiur on carbon (0. 5 g, 10 wt%) The reaction was purged with hydrogen and stirred under .H 10 (bLloon) at room temperature for 2 hours. The reaction mixture was filtered through colte and concentrated to afford 1.3 g crude compound 6-1 as a yellow solid. Compound 6-2 F HNC CO 2 Ph BoC OBoc 6-2 To compound 6-1 (0.9 g, 3.27 umol, 1.0 equiiv) in DCM was added Boc 2 O (2.14 g. 9,81 mmol, 3.0 equiv) dropwise. DMAP (135 mg, 15 wt%) was added to the mixture ane the reaction was stirred at room temperature for 1 hour. Then the 5 reaction mixture was heated to reflux for I hour, The reaction mixture was concentrated. The crude compound was purified by column chromatography on silica gel eluted with (PF.:EA= 200:1-100:1) to yield compound 6-2 (1.16 g. 73.4%) as a light yellow solid. 'H NVR (400 MHz, DMSO) 3 7.60 (d, J= 10 0 H-z, 1 H). 7.53-7.44 (in, 2 1), 7.36-7.31 (i, I H), 7.28-7 22 (m, 2 1-), 3.06 (s, 3 R), 233 10 (d, J= 2.0 Hz, 3 H), 1,38 (s, 9 H), 1.34 (s, 9 H) MS (EST) m/z 476 2 (M-H). Compound 6-3 - H 3 C'NCts HsC N S H N/ Bo6.3a OTS To diisopropylamine (0,28 mL, 3,2 rnmol, 10.0 equiv) in THF at -78 'C was 15 added nBuLi (0 8 mL, 2.50 M/hexane, 3,2 mnol, 10.0 equiv) and TMEDA (0.40 rnL, 5.0 mmol, 10.0 equiv) at -78 C dropwise. The reaction was stirred at -78 C for 40 rnin, Compound 6-2 (480 mg, [.0 minol, 3.0 equiv) in THF was added to the reaction mixture dropwi se at -78 C The resulting deep-red solution was stirred at -78 C 60 min, the none (160 mug 0.33 nunol, .0 equiv) in '11-F was added to the 20 mixture dropwise at -78 C. The deep-red solution was gradually warned tip with stirring Ironi -78 C (o -20 'C over a period of I h. The resulting orange solution was brought to 0 C, and quenched with aqueous saturated ammonium chloride (100 mL). The yellow-green mixture was extracted with EtOAc two times. The combined EtOAc extracts were dried (Na2S4) and concentrated to yield the crude product. 25 Flash column chromatography on silica gel with 0%, 5%, 10%, EtOAc/hexane sequentialy yielded the desired product 6-3 as a light-yellow solid (42 rug, 14.8%). [H NMR 0100 MHz, CDCl;) 6 15.70 (s. 1 11), 7.52-7.50 (1, 2 .1), 7.42-7.33 (a, 3 H), 7. 1:6 (d, J = 8.4 iz, 1 H), 5 37 (s, 2 H), 3,95 (d, J- 10 .8 Hz, i H), 3.28-3,23 (in, - 11 1 I), 3.14 (s, 3 H), 3.10-3.05 (in, H), 2.58-2.47 (rn, 9 11), 2.16 (d, J = 14.0 Hz, I H), 1.53 (s. 9 H), 1.42 (s, 9 H), 0.89 (s, 9 H), 0.29 (s. 3 H), 0.15 (s, 3 1); MS (ESI) m/z 864,43 (M IT). 5 Compound 107 F
H
3 c- CH,, HN!1 H OH O HO O 107 Compound 6-3 (120 mg, 0 14 mmol) was dissolved in IIIF (5 iL) and aqueous HF (40%, 2 nL) was added dropwise. The yellow solution was stirred at room temperature overnight. The resulting deep-red solution was slowly added into 10 an aqUeous K 2
HPO
4 solution with stirring. The pH of the mixture was adjusted by aqueous K 2 l-fPO 4 solution to about 8 The yellow mixture was extracted with EtOAc two limes. The combined EtOAc extracts were dried (Na 2
SO
4 ) and concentrated to yield the crude product. The above crude compound (120 mg, crude, -0.14 mmol, 1.0 cquiv) was 15 dissolved in 1PLC grade MeOHI(10 rnL) and 10% Pd-C (25 mg, 0.03 mmio, 0.2 equiv) were added The mixture was purged with hydrogen by bubbling hydrogen through with gentle stirring for 5 min, The reaction was then vigorously stirred under hydrogen balloon at room temperature For 2 hr. LC-MS analysis indicated the reaction completed. The catalyst was filtered off and the mixture was concentrated, 20 the residue was purified by reverse phase IPLC to afford the desired compound 107 (50 mg, 78%) as a yellow solid. 1H NMR (400 N11z, CD 3 OD) 6 7.462 (d, .J 8.4 liz, l H), 4.14 (s, 1 IH). 3.21-2.93 (n, 9 H), 3.10 (s, 3 1), 2 38-2.25 (m, 2 H) t 68 1.62 (m, I 1), MS (ESi) Tm> 462.2 (M+I). 25 Compound 108 F N3YH H H:7 OO CN N N H CHs OH C Hvo u 102 - 113 Compound 107 (15 mg, 0.033 minol, 1.0 equiv) in THF (2 m1L) was added with pyrrolidin-I-yv-acetic acid (10.2 mg, 0.066 mmol, 2.0 equiv), NaC 3 (10 2 rmg, 0.066 mmol, 2.0 equiv) and HATU (25.5 mg. 0 066 mmol, 2.0 equiv). The reaction mixture was stirred at room temperature for 48 hours. LC-MS analysis 5 indicated the reaction completed The reaction mixture was conceirated under vacuum, the crude product was purified by reverse phase HPLC to afford the desired compound 108 (2. 1 mg) as a yellow solid. iH NMR (400 MHz, CD 3 OD) 5 7.44-7.10 (, I H), 4.02-3.97 (m. 2 H). 3.83-3.76 (i, 1 F-), 3.60-3.58 (m, 2 H), 3.15 (d, J= 6.4 Iz, 3 H1)- 3 03-2.83 (m, 11 H), 2.3 1-2.13 (m, 2 ) 2.03-1.85 (im, 4 H), 1.61-1.52 (M, 10 1ll), MS ([SI) m z 572.9 (M-H). Example 7. Synthesis of Compounds 109-112 Scheme 7 O O 1) CH3.K 2 COO 0 0 0 O 2) Boc 2 O;DMAP DIRAL-H GnO . OH ---------- *- Bno OCH3 BnO H NHBoc B~oc2 J NBOc2 7-1 7-2 7-3 anilire S Na eOAc) 3 BH F H CH 3 F H NH 3 H H OH OH! HtC HI ilH H 2 /Pd-C Bn N H. NHBoc OH 0 HO 0 0 NHBoc H H HO 0 0 7-5 7.4 HATU F H-C. - CH,5F HC, C H O H N HOH BnHN, ~ NNH, diecael H 2 N,. N H.. SOH O HO 00 OH 0 HO 0 7-6 109 R'CH]O/InCi3 Na(OAcrBH H H:>1= (or R) H 2 0 .
O (R R'CH) JH 0 HO 0 7-7 - 114 The following compounds were prepared according to Scheme 7. Compound 7-2 0 0 N B 7-2 5 To Boc-L-glutamic acid-i-benzyl ester (7-1) (3.00 g, 8.89 mmol, 1.0 eq) in DM1 (20 mL) at rt was added potassium carbonate (1.84 g., 13.33 nnol, 1.5 eq) and methyl iodide (0.67 mL, 10.74 mmot 1.2 eq). The mixture was diluted with EtOAc (200 MI, washed with water (200 mL), saturated aqueuis sodium bicarbonate (100 ml, x 2), and brine (100 ml x 1). The EtOAc solution was dried over sodium sulfate 10 and concentrated in. vacuo: Re 0.33 (20% EtOAc/hexane), Boc 2 O (2 91 g, 13,33 mmol, 1.5 eq), DMAP (54 mug, 0.44 nimol, 0.05 eq), and DILA (310 TnL, 17.80 mnrnol, 2 eq) were added to the above intermediate in acetonitrile (20 nL), The solution was stirred at rt for 60 hrs, added with saturated aqueous sodium bicarbonate (100 mL), and extracted with EtOAc (100 mt x 1, 50 15 mL x 2). The EtOAc extracts were cornbined, dried over sodium sulfate, and concentrated in vacio to yield the desired product 7-2 as a pale liquid (quantitative) Rf 0.15 (20% EtOAc/hexane) 1H NMR (400 MHz, CDCI) 6 725-7,35 (m, 5 H), 5,14 (s, 2 HI), 4.95 (dd, J= 4.9, 9.8 Hz, I H), 3.65 (s, 3 H), 2.43-2.52 (m, I H), 2.37 2 42 (m, 2 H), 2. 5-2.25 (m, 1 H), 1.42 ( 18 [H); MS (ESI) na 452.3 (M+H). 20 Compound 7-3 NBoo 2 7-3 To compound 7-2 (8.89 nmol, I eq) in anhydrous diethyl ether (40 mL) at 78 "C was added DIBAL-H (12.33 mL, I M/hexane, 12.33 mnmol, 1,25 eq) 25 dropwise. The reaction was stirred at -78 C for 2 hrs. Additional DIBA-I (1.20 mL, 1 M/iexane, 1.20 mmol) was added The reaction was stirred at -'8 C for another I hr and quenched with HOAc (2.80 mL) at -78 'C. The reaction was warmed to rt and added with 10% aqueous sodium carbonate (75 miL). The mixture was stirred for 15 min and extracted with inethylene chloride (200 nl x 1, 50 nmL x 30 2) The methyl ene chloride extracts were combined, dried over sodium slfato, and - 115 conetntrated in vacuo to yield the desired product 7-3 (quantitative) R 0.40 (20% EtOAc/hexane); iH NM7R (400 MHz CDC]) 6 9.75 (s, 1 H), 7.25-7.35 (n, 5 H), 5 14 (s, 2 -4), 4,87-4,92 (m, 11H), 2 45-2.65 (n, 3 1), 2.12-2.22 (m, I H), 1.42 (s, 18 H); MS (ESI) m/,_ 122.3 (MI+L). 5 Compound 7-5 F HC ,CH 3 H H HO JN NH, NHBoc OH 0 HO 0 0 7-5 To aniline 9 (90 mg, 0.20 mmol, bis--IC1 salt, I eq) in anhydrous DMF (2 mL) was added aldehvde 7-3 (101 mg, 0.24 miiol, 1.2 eq), triethylanme (0.028 ml, 10 0 20 minol, J eq), and Na(OAc)BH (64 mg, 0.30 mmol, 1,5 eq). Tc solution was stirred at rt for 1 hr and added slowly into diethyl ether (50 mL) with rapid stirring, The yellow solid was collected, washed with more diethyl ether (5 mL x 3), and dried under vacuum to afford the intermediate 7-4 Intermediate 7-4 was dissolved in dioxane/miethanol (5 ml, 1:4 v/v, 1.5 containing 0J N HC), 10% Pd-C (85 ng, 0.04 mol, 0.2 eq) was added, The mixture was purged with Idrogcn and stirred under 1 atm hydrogen at rt for i hr. The catalyst was filtered with a small Celite pad and washed with ethanol (2 m.L 3). The filtrate was concentrated in vacuo. The crude product was purified bV reverse phase preparative I 1PLC using methanol and 0.05 N HCl/water as mobile 20 phases. Free-drying yielded mostly the Boc-deprotected product as a brovn solid (25 mg, 22%, 2 steps), which was re-protected by treatment with Boc2O (11 mg, 0.050 mmol, 1.1 eq) and DIEA (0,039 ml, 0.22 nmol, 5 eq) in THF/water (5 mL 1:1 viv) at rt for i hr. Concentration yielded the desired product 7-5 as a yellow solid: MS (ESI) n/s 663 2 (M-H), which was used directly in the subsequent steps 25 without further puriication. Corpoind 7-6 -116 F HCCs SH OH BoiHN,
NH
2 OH 0 HO 0 0 7-6 To a suspension of compound 7-5 (0.044 mmol, I eq) and sodium carbonate (7 mg, 0.066 mmol, 1.5 eq) in THF at rt was added HATU (20 mgp, 0.053 mnol, 1. 2 eq). The mixture was rapidly sti-red at it for 2 hrs. Methanol (5 mL) was added. The 5 solds were filtered of. The filtrate was concentrated under reduced pressure to yield crude 7-6 as a yellow solid d; MS (ES [) m 545 1 (M4f), Compound 109 F H 0J, N 1AOH
H
2 N, .N H NH 2 OH o Ho O 6 109 10 Compound 7-6 (0.044 mmol) was treated with 4 N ICI/dioxane (5 ml.) at rt for overnight and concentrated in vacuo. The residue was re-dissolved in methanol (I mL) and added dropwise into diethyl ether (50 mL) with rapid stirring. The yellow precipitates were collected, washed with more diethyl ether (5 ml., x 3), and dried 1uder vacuo to afford crude 109 as a brown solid. 15 One fifth of the above crude product was purified by reverse phase HIPLC to yield pure 109 as a yellow solid (1.5 ng, 31%): H .NMR (400 MHz, CD 3 OD) 6 7,36 (d, 9.2 Hz, I H), 4.09-4. 15 (m, 1 H), 4.08 (s, I A), 3 70-3.80 (m, 11-1), 3.58-3.68 (m, .1 H), 2.90-3.50 (m, 12 11), 2.30-2.45 (m, 2 H), 2 10-2 25 (m, 3 H), .95-2.10 (m, I It), 1.58-1.70 (m. 11); MS (ESI) nz 545. 1 (M+1H). 20 Compound 110 F H 8 . CH' NN H H z HC NI 0H ' HO H C 110 To 2/5 of crude 109 (0.018 mmol, I eq) li DMF (I mL) was added aqueous formaldehyde (0.007 mLU, 36.5%/water, 0.094 mmol, 5 eq), InCh (0.4 mg, 0.002 - 117 mmol, 0.1 eq), and Na(OAc)3BH (15 rmg, 0.071 mmol, 4 eq). The reaction was stirred at rt for 2 hrs and quenched with 0.5 N HCI/methanol (1 mI) The solution was added dropwise into diethyl other (100 m1L) with rapid stirring. The precipitates were collected, washed with more dietivl ether (2 niL x 4), and purified by reverse 5 phase .HPLC to afford the desired compound 110 as a yellow solid (1.8 rmg, 18%): 'H NMR ('100 MIz, C.D 3 OD) 5 7.7.44 (d, J - 9.1 Hz, I H), 4.37 (dd, J 6.1, 11.6 Iz, 1l), 4.09 (s. 1 11), 3.60-3.75 (in, 2 1 ), 2.92-3.50 (m, 15 11). 2.86 (s. 3 1), 2.10 2.50 (m, 6 H), 1.60-1.72 (n, 1 H1): MS (ESI) m/z 5733 (M-H), 10 Compound 111 F HsCN 0H, NHNH J OH C HO 0 0 111 To 2/5 of crude 109 (0.018 innol, 1 eq) in DMF (1 rL) was added cvcopropanecarboxaldehyde (1.4 pL, 0.018 mniol, I eq), inCh (0.4 Mg, 0.002 mmoL, 0.1 eq), and Na(OAc)hBH (6 mg, 0.028 miol, L5 eq). 'The reaction was 15 stirred at rt for overnight and quenched. with 0 5 N [ICl/methaiol (1 ii). The solution was added dropwise into diethyl ether (100 mL) with rapid stirring. The precipitates were collected, washed with more di ethyl eth er (2 miL x 4), and purified by reverse phase HPLC to afford the desired compound 111 as a yellow sold (1.3 ig, 12%): 11[ NMR (400 MHz, CD OD) a 7.38 (d, J= 9.2 Hz, 1 11), 4.22 (dd, .1 20 6. I, l I.6 Hz: 1 ), 4.09 (d, I H), 3 60-3.78 (m.. 2 H) 2,85-3 50 (m1 1), 2 00-2.50 (in. 6. 11) 1.60-1.72 (n, ] H) 1. 10-1.20 (n, 1 H), 0.70-0.75 (m, 2 IT), 0.40-0 50 (in, 2 H); MS (ESI) m/z 599.4 (M+H). Compound 112 F H H O H N- NH 2 OH 0 H00 0 25 112 Dialkylated product 112 was also isolated from the preparation of compound 111 (1.0 mg, yellow solid, 9%) 'H NMR (400 MHz, CD 3 OD) 6 7.42 (d,J= 9 2Hz, - 118 1 1.1), 4.70-4.80 (m,. I Ii), 4.09 (s, 1 I), 3.55-3.80 (n 3 H), 2.95-3.50 (m, 13 11), 2 ]0-2.50 (m, 6H), 1.55-1.75 (mn, 1 H), 1.20-1.30 (m, 2 H), 0.68-0.90 (m, 4 H), 0.38-0.58 (m, 4 11); MS (EST) rnz 653.3 (M-H). 5 Example 8. Synthesis of Compoinds of Structrual Formula (A), wherein Y is -(C C4alc(C-C4klee-N(R )-C(0)-[C(R")(R E)]4 N(R^)(R It), Scheme 8 HjC,N F N-H3 U U OH OH(or R) IIQ 113H SN NHNH2 Oh OH0 HO 0O OH 0 NOO O O 10 0 -O H 3 In Scheme8, Ran R ect Nas) 2 d nH F HN., HON N OH HO O 113 F H H.. OHl F C HO Cr10 a o / ( m u co un 7 H H - OO O (HOHHO1i 0 OHH CC 101 Ii Szlm 8, iyit and iItv is crE a (9A reoetvlv, as5 defnol . eiv wLcuas 15 de tonoin 113F/C-o omom (1 g I-bNJ XLJ NL solutinW - 119 0.25 nimnol at 25 C The reaction was stirred at 25 *C for 30 min. Preparative reverse phase IUIPLC purification on a Waters Autopurification system using a Phenomenex Polymerx 10 [t RP-1 1 OOA colurnn [10 am, 150 x 21.20 mm; flow rate, 20 mL/min; Solvent A: 0.05 N HCl; Solvent B: CH1-CN; injection volume: 4.0 mL.. 5 (0.05 N HICwater); gradient: 0-30% B over 20 min; mass-directed fraction collectionj. Fractions w1ith the desired MW were collected and freeze-dried to yield 23 ing of pure 113. -IH NN{R (400 MHz, CD 3 OD) 6 7.47 (d, J= 9,2 Hz, I T), 4.16 (s, 2 H), 4.13 (s ItH), 3.21-2.94 (i, 3 H), 3.06 (s, 3 1H), 2 97 (S, 3), 2.37-2.22 (im 2 H), 1.7) 1.58 (ni 1. H); MS (EI) m/iz 162.20 (M-I1) 10 Compoound 114 CH . HC CHH H NN OH 0 HO 0 0 114 Et 3 N (2 L, 0 0136 mmio, 2.0 equiv) was added to a mixture of 1.13 (3 mg, 00065 mmol) and pivaldehyde (0.8 A, 0,00715 mmol, 1.1 equiv) in DMF (0. niL) 15 at 25 *C. The reaction was stirred at 25 'C for 15 inn. NaB-1(OAc)j (3 mg, 0.013 mmol) and I LOAc (2 pL) was added to the resulting mixture. The reaction was stirred at 25 'C for I h. Preparative reverse phase HPLC purification on a Waters Autopurificaton system using aPhenomenex Polymerx 10 p RP-1 100A column [10 um, 150 21.20 mm; flow rate, 20 nL/min; Solvent A: 0.05 N H-CL Solvent B: 20 CH 3 CN injection volume: 4.0 mL (0.05 N1 Hl/water); gradient: 0-100% B over 15 min, mass-directed fraction collection]. Fractions with the desired MW were collected and freeze-dried to yield I mg of 114: H N MR (400 MHz, CD 3 0D) S 7.52 (d, J= 9.1 Hz, I H), 1.30 (s, 2 H), 4.09 (s, 1 H), 3 23-293 (m , 15 ), 3.01 (s., 3 11), 2.95 (s, 3 11), 2.40-2.19 (, 2 14), 1.71-1.60 (i, I R), 1O (s, 9 -); MS (ESI) n z 25 532.27 (M-H). Compounds 115 - 118 were prepared similarly to coropoumd 114 using the corresponding aidehydes. 30 Compound 115 120 F H3C. .GH 3 H CH HNH a.- NH 2 OH 0 H0 O 0 -15 H1 NMR (400 MHz, CD30D) 5 7.51 (d. -= 8.8 Hz, I H.), 4.25 (s, 2 H), 4.10 (s, I H), 3.25-2.90 (m, 5 H), 3.05 (s, 3 H), 2.96 (s, 3 11), 2,40-2 21 (rn, 2 11), 1.90 1.60 (m. 7 H), 1.42-0.95 (m, 5 H): MS (ESI) nv± 558.31 (I J1). Comnpound 1.16 H OH H3 N ,- N.. NH 2 OH 0 HO 0 116 'H NMR. (400 M Hz, CD 3 OD) a 7.50 (d, J= 9.0 Hz, t H), 4.24 (s, 2 H), 4.09 (s, I H), 3.25-2.90 (m, 5 H) 3.07 (s, 3 H), 2.94 (s, 3 H), 2.40-2 21 (m, 2 H), 1.82 10 1.58 (m, 3 1), 1,01 J-- 6.7 z, 3 H.) MS (E SI) m/z 504.22 (M-H), Compound d 117 F
H
3 C' N-CHi H HIH H [tC NNH2 CHs OH 0 nO t 117 'H NMR (400 Miz, CD-OD) 6 7,51 (d, ,J 8,9 Hz, 1 1), 4,23 (s, 2 H), 4.09 15 (s, 1 H), 3 25-2.92 (m, 4), 3.02 (s, 3 H), 2.95 (s, 3 H), 2.40-2 I9 (m, 2 H) 1.71-1,60 (at 11H), 1.10 (d, J= 7.0 Hz, 6 H) MS (ESI) m z 504.23 (M+H). Compound 118 F H3 NN NH 2 OH C HO 0 11B 20 INMR (400 MHz, CD 3 0D) 67 54 (d,J= 9.1 H z, 1 H.), 4.37 (5,2 H), 4.10 (s 1 H), 3.20-2 85 (m, 3 Hf), 3.05 (s, 3 H). 2.97 (s, 3 11), 2.91 (s, 3 H), 2.90 (s, 3 H), 2.42-2,20 (m, 2 11), [.72-1,60 (i, 1 H), MS (ESI) n?, 490.]9 (M 1I-1).
- 121 Compound 119 F HN HIC CH 3 CHH HAc N
NH
2 OH 0 HO O 119 Prepared from compound 114 by reductive alkylation using formaldehyde 5 under similar conditons 'HI NMR (400 Mlz, CDOD) 5 7.57 (d, J 9.1 Hz, I H), 4.61 (d, J= 12.8 Hz, 1 H), 4.27 (dd, J= 12.8, 6.4 117, 1 H), 4.10 (s, 1 .11), 3.25-290 (m, 5), 3.03 (s, 3 l) 2,96 (s, 3 11), 2.95 (s, 3 T), 2.42-2.-33 (i, I H). 2.29-2.20 (im, I H), 1.72-1.61 (in, 1 H), 1 10 (d, j= 6 Hz, 9 H); MS (ES1) rez 546.30 (M+H). 10 Compound 120 F H3C'N-CH> H F-I ''N H1 3 C >1 N 1J OH O HO 0 0 120 Prepared similarly to 114 by reductive akylation of 113 wkh t-Bu -N(Cbz)
CH
2 CHO followed bw hydrogenation: 'H NMR (400 MHz, CD 3 OD) 3 7.59 (d, J= 8.6 Hz, 111), 4,38 (s, 2 ), 4.09 (s, I H), 3.60-2.95 (m, 7 H4), 3.03 (s, 3 11), 2.96 (s, 3 15 H), 2.41-2.30 (ni I H), 2.28-220 (i, I H), 172-1 60 (m, 1 -), 1.44 (s, 9 H.) MS (ESI) m/z 561.31 (M+H). Compound 121 F N-CHs FN CH H O 121 20 2-/-Butyiainoacetylchloride hydrochloride (5 8 mg, 0 3 i nmol, 1.2 equiv) was added to a DMF solution (0.2 mL) of 113 (12 mg, 0.026 mmol) at 25 'C. The reaction was stirred at 25 C for 30 min. The reaction mixture was diluted with 0.05 N H1-I (2 m) and injected into a Waters Autopurification system equipped with a Phenoimenox Polynerx 1 0 LL RI'-1 100A coLmm [10 pn, 150 x 21.20 mm; flow rate, - 122 20 mLhnin; Solvent A: 0 05 N HCl; Solvent B: CHCN, gradient: 0-+I00% B ovor 15 min; mass-directed fraction coliectionj. Fractions with the desired MW were collected and freeze-dried to yield 3.0 mg of pure 12L: 'H NMR (400 MH7, CDOD) 6 7,34 (d, J= 9.6 Hz, 1 H), 4.46 (s, 2 14), 4,08 (s, 1 H), 3.81 (s, 2 1H.), 3.18 5 2.92 (m, 3 H), 3 03 (s, 3 H1), 2.96 (s, 3 1), 2.32-2, 18 (in, 2 UT), 1.69-1.60 (m, t 1H), 1,38 (s, 9 N); MS (EST) m," 575,30 (M+4-). Compound 122 F H G H H: OH H'C HNH 2 CH, e OH 0 HO 0 122 10 Prepared similarly to COmpound 121: H1 NMIR (400 MHz, CDOD) 6 7.33 (d, J= 9.9 Hz, 1 I), 4.46 (s. 2 H), 4.08 (s, L H), 4.00 (s, 2 H), 3.23-2.91 (in, 3), 3.04 (, 3 1), 2,97 (s, 3 11), 2 95 (s, 6 H1), 2.32-2.18 (in, 2 H), 1.70-1.58 (in, 1 H); MS (FST) m z 547.23 (M+H). 15 Compound 123 - HOC.. 0H 1 H H .H HF H YNN .NH 0 OH O HO 0 0 123 Prepared similarly to 121 using n-propyl isocyanate: 1 H 1\MR. (400 MI-lz,
CD
2 OD) 5 7.24 (d, J= 9 8 Hz, I H), 4.31 (s., 2 H), 4.08 (s, 1H), 3.18-2,93 (in, 3 11), 3.10 (1, J= 6 7 1z, 2 1), 3.03 (s, 3 H), 2.96 (s, I H 2.32-2.18 (m. 211), 1.69-1.58 2 0 (m, I H), 1.55- 1.46 (m, 2 H), 0.92 (, J = 6.7 Hz, 3 H); IS (ES1) W 5 584.01 (M II). Example 9. Synthesis of Compomds of Structural Formula (A), wherein Y is -(C1 2 )-N(R^)(R"). SchemC 9 25 - 123 F F F C CH2 C H 3 Aly -OTBS CHS BrJHAo ~BnBr/KCO [ Pd cat,
CO
2 Ph Br 02Ph BI EC0 2 H OFI OH OBn OTBS OBr 3 9-1 9-2 9-3 a) LD/VTVEDA b) enona H3C, CH3F H3C 'H 3 HOA TBSO N BnO 0 HO 0 OBn n O HO - Bn OTBS 9 OTBS Dess-Marth F HC ,CH F H-, '-H H H RR'NHlHOAcH FN N(ON yl-7 Bnm 0 HO 0 Oln Bn0 0 HO 6 O1n 9-6 OTB 9-70TBS 1) ,q HF 2) IA/Pd-C F N H HR R N OH 0 aHO O 9-8 In Scheme 9, R and R' are R^ and R respectively, as definedd sn Structural Formula (A). The following compounds werer prepared according to Scheme 9. 5 Compound 9-1 F Br'^ CO 2 Ph OH 9-1 Br, (2.7 nL, 52.0 mmol, 1.2 equiv) was added to a solution of 3 (10.6 g, 43.3 mmol) in acetic acid (100 n1L) at 25 C The reaction was stirred at 25 rC for 10 12 h. The resulting mixture was added dropwise to ice-water (400 nL). The mixture - 124 was allowed 1 warm to 25 C over I i The resulting suspension was filtered through a pad of Celite. The solid was washed off with EtOAc. The combined organic layer was dried (Na 2 SO4) and concentrated to give 14 g of crude 9- I. 5 Compound 9-2 F Br CO 2 Ph CBn 9-2 Potassium carborate (8 9 g: 64.5 mmol, 1.5 equiv) and benzyl bromide (11.5 mL, 96.8 mol, 2 25 equiv) were added to an acetone solution (100 mL) of crude 9-1 (14g, 43 nunol) at 25 'C_ The reaction was stirred at 25 -C for 12 h and 10 concentrated. The resulting mixture was partitioned between 120 and FtOAc. The aqueous layer was extracted with EtOAc. The combined EtOAc extracts were dried (Na 2
SO
4 ) and concentrated to yield crude 9-2 Flash chromatography on silica gel (100:1 to 30: 1 hexanes/EtOAc) yielded 15.4 g of compound 9-2 (87% for 2 steps). 15 Compound 9-3 F CH3 TB50s,-cO2Ph 9-3 Pd(OAc)2 (227 mng, 1.0 mmrrol, 0,2 equiv) and P'(0:I ol) (462 mrg, 1.5 mm11-ol, 0.3 euiqv) were added to a DMF solution (10 rnL) of 9-2 (2.1 g, 5.,06 mminol).Th raonwa.s purg ,ed with N2. for 5 miin. Et3N (3,5 mL,, 25.3 mmnol, 5 equiv) and 20 allyloxy:-t-bu~tyldlimepthylsilanie (2.2 mtL, 10. 1 mmnol, 2 equiv) were added to te reaction. The re-action was heated to 88 'C and stirred at 88 C for 5 h. The reactio n was allowed to cool to 25 (C and quienchied withi H2,,0. The resulting miixture was extracted with EtOAc. The combined E.tOAc extracts were dried (Na,.SOI) an'd concentrated to give crude 9-3. Flashi chromiatography oni silica gel (l00:0 to .1.00:1 25 hexanes/H tOA c) yielded 1. 2 g of compound 9-3 (47%.). Compound 9-4 - 125 F Hqc'N-CHI H H -B oN BnO 6 HO - O DOn 94 OdTBS n-BuLi (1.3 niL, 2,07 nmol, .5 equiv) was added to a THF solution (5 mfL) of diisopropylarnine (0 3 ml, 2.07 nmol, 5 5 equiv) at 0 C. The reaction was stirred at 0 C for 30 min and cooled to -78 "C, TNIEDA (0.8 ml, 5.64 mmol, 15 5 eqUiV) was added to the mixture. To the resulting solution was added a THF solution (5 mL) of 9-3 (475 mg, 0.94 mmol, 2,5 equiv), The reaction was stirred at -78 C for 10 min. A TF solution (5 mL) of enone (181 mg, 0 376 mmol) was added to the reaction at -78 *C. The reaction was stirred at -78 C flor 30 min and allowed to warm to 25 *C over 1 h, quenched with saturated NI I:Cl, and extracted with EtOAc. 10 Th e conibined EtOAc extracts werm dried. (Na2SO4) and concentrated to yield th crude product. Preparative reverse phase HPLC purification on a Waters Autopurificrtion system using a. Sunfire Prep CI8 OBD column [5 pm.. 19 x 50 mm; flow rate, 20 mL/rnn; Solvent A: H20 with 0.1% HC0 2 1H; Solvent B: CHVCN with 0.1% HCO 2 H; injection volume: 4.0 mL (CH3CN); gradient. 100-100% B over 15 15 min; rnass-directed fraction collection], Fractions with the desired XMW were collected and concentrated on a RotaVap at 25 0 C to remove most of the acetoririle. The resulting miostly aqueous solution was extracted with E*OAc. The combined EtOAc extracts were dried (Na 2
SO
4 ) and concentrated to give 2.00 n of 9-4 (59%), 20 Conpound 9-5 F HcNs F - N0H HO BnO 0 HO O OBn S-5 TFA (0.5 mL) was added to a THF/H 2 O (2 niL/0.5 mL) soition of 9-4 at 25 "C The reaction was stirred at 25 "C for t h. Te reaction was quenched with sat. NaRCLEO solution. The reaction was extracted with EtOAc, The combined EtOAc 25 extracts were dried (Na 2
SO
4 ) and concentrated to give crude 9-5. Preparative reverse phase 1IPLC purification on a Waters Autopurification system using a Sunfire Prep C 18 OBD column [5 mnu, 19 - 50 mm; flow rate, 20 m/mi Solvent A: H20 with - 126 0.1% HCO 2 H; Solvent B: CH3CN with 0,1% HCO 2 H, injection volume: 4.0 niL (CH3CN); gradient: 80-100% 13 over 15 min; mass-directed fraction coliectionJ. Fractions with the desired MW were collected and concentrated on a RotaVap at 25 C to remove most of the acetonitrile. The resulting mostly aqueous solution was 5 extracted with EtOAc. The combined EtOAc extracts weree dried (Na2SO4) and concentrated to give 80 mg of 9-5 (46%) Compound 9-6 F. H H FnO o) HO r, OBn OTBS 10 Dess-Martin periodinane (IS mg, 0.043 mmol, 1.2 equiv) was added to a CH2CN solution ( i mL) of 9-5 (28 mg, 0.036 mmol) at 25 "C. The reaction was stirred at 25 0 C for 30 miii an.d diluted with H 2 0. The resultmg mixture was extracted with C 2 . The combined CLCI extracts were dried (Na 2 SO4) and concentrated to give crude 9-6. 15 Compound 9-7-1 FNN N N BnO 0 HO o OBn OTBS 9-7-1 Pyrrolidine (15 p.L, 0. 1 9 mmol, 5 equiv) wvas added to a dichloroethane solution (1 in) of crude 9-6 (0.036 muol) at 25 'C. The reaction was stirred at 20 25 'C for 10 min. HOAc (15 [IL) and NaBHi(OA c) (15 mg, 0.072 mrnol, 2 eCquiv) were added to the reaction. The reaction mixture was stirred at 25 'C for I h and quenched with H 2 O. The resulting mixture was extracted with CH 2 C1]. The combined CH2C 2 extracts were dried (Na2S0 4 ) and concentrated to give crude 9-7 1 Preparative reverse phase HIPLC purification on a Waters Autopurification system 25 using a Sunfire Prep C1 8 OBD column [5 [im, 19 x 50 mm; flow rate, 20 mUmin; Solvent A: H.O with 0.1% HCO2H; Solvent B: CHICN With 0.1% HCOKH: injection volume: 4.0 mnL (CH3CN); gradient: 0 -100% B over I. mn; mass- - 127 directed fraction collection]. Fractions with the desired MW were collected and concentrated on a RotaVap at 25 0 C to remove most of the acetonitrile. The resulting mostly aqueous solution was extracted with EtOAc. The combined FtOAc extracts were dried (Na2SO) and concentrated to give 6 mg of 9-7-1 (20% for 2 steps). Compound 124
H
3 CN 'CH, F N d H OH N N H OH O HO 0 810 Aqueous I-F (0.3 mL 48%) was added to a CH 3 CN solution (2 mL) of 9-7-1 (6 mg, 0.007 mmol) in a polypropylene tube at 25 'C, The reaction was Stirred at 10 25 'C ]or 18 h. The resulting mixture was poured into an aqueous solution of K2HPO4 (2 g., dis solved in 15 niL water). The mixture was extracted with EtOAc. The combined EtOAc extracts were dried (Na2S0 4 ) and concentrated to yield crude desilyl product. 15 Palladium on carbon (2 mg, 10 wt%) was added to a HCl/.MeOH solution (. 5N, 2 ml.) of tie crude deslyl product, The reactIon was purged with hydrogen and stirred under H2 (balloon) at 25 C for 4 h. The reaction mixture was filtered through a small Celite plug. The filtrate was concentrated to yield the crude product. Preparative reverse phase HPLC purification on a Waters Autopurification system 20 using a Phenoinenex Polymerx ' 0 1. RP- I I OA column [10 pm, 150 x 2 .20 nim; flow rate, 20 mL/min; Solvent A: 0 05 N HC/water, Solvent B- CHLCN; injection volume: 1.0 ml, (0.05 N Hl/water); gradient: 0-50% B over 7 nt, 50-1 00% over 3 min, and 100% over 5 m;in mass-directed fraction collection]. Fractions with the desired MW, eluting at 6.4-8.2 min, were collected and freeze-dried to vield 25 1 5 mg of compound 124: 'I NMR (400 MI-z, CD/OD) S 7.28 (d, J= 9.7 liz, I I), 4.08 (s, ] H), 3.71-3.63 (in, 2 H), 3.32-2.95 (m, 7 H), 3.04 (s, 3 I), 2.96 (s, 3 Hi), 281-2.73 (m, 2 11), 2.32-1.98 (n 8 11), 1.70-1 59 (in, 1 H); MS (ESI) n/z 544.18 (M+1). Compounds 125 - 127 were prepared similarly to compound 124 using the 30 corresponding aMines in the reductive arniation step.
- 128 Compound 125 F N CH H HC. N
NH
2 OH 0 H0 c 125 'H NMR (400 MHz, CD 3 OD) 8 7.28 (d, J= 9 7 Hz, H-), 4.08 (s, I 11), 3.25 5 2.94 (m, 5 H), 3.04 (s, 3 -.), 2.96 (s, 3 H), 2.89 (s, 6 1), 2.80-2.70 (i, 2 H), 2.32 2.18 (m, 2 H), 2.10-2.00 (mn. 2 H1), 1.70- 158 (m, 1 H), MS (ESI) mn 518.26 (M+AH). Compound 126 F HHC 'NCH C NH H2C OH 2 OH 0 0 126 10 1 1 NMR (400 MHz, C DOD) 6 7.27 (d J - 9.6 H1z, 1 H), 4.08 (s, i H), 3.20 2.93 (m, 5 H), 3.04 (s, 3 11), 2.96 (s, 3 H), 2.82-2.72 (in, 2 H-), 2.33-2,19 (m., 2 H-), 2.04-1.94 (m, 2 11), 1.70-1.58 (i, 2T H) 1.37 (s, 9 14) MS (ESI) tt 546.20 (M+H). Compound 127 F jG, N I H H OH 0 HO OH 0 1 5 127 1 H NMR (400 MHz, CD-0OD) 6 7 28 (d. J= 9.7 Hz, 1 H), 4.09 (s, I H), 4.04 (q, J= 9,0 liz, 2 H), 3.25-2.95 (m, 5 H), 3.04 (s, 3 H-l), 2.97 (s, 3 H). 2.84-2.75 (m, 2 IF), 2.32-2.20 (m, 2 H), 2.13-2.03 (m, 2 H), 1.70-1.58 (m, I H); MS (ESI) m/z 5 72.22 (M+H). 20 Compound 128 - 129 F HNC H H OH HO
NH
2 CH 0 H o 0 128 Compound 128 was prepared from compound 9-5 by HF treatment following by hydrogenation under similar conditions: '1H- NMR (400 M-. CD 3 QD) 5 7.21 (d, J = 9.87 Hz, 1 1), 4.07 (s, I H), 3.63-3.57 (m, 2 1-1), 3 20-2.90 (in 5 11): 3.04 (s, 3 5 H), 2.96 (s, 3 H), 2.75-2.68 (m, 2 .H), 2.32-2.17 (m, 2 H), 1.89-1.79 (m, 2 1H), ,70 1.57 (m, I H), L.25 (d,.J- 7.2 Hz, L H); MS (ESF)tnz 491. 18 (N+-H) ExamIple 10. Synthesis of Compound 129 Scheme 10 10 F F C'N'CHs Ci 1 9) ifrMgCU-CO 3 H ) LDA/TMEDA H H )CHb) eno ne Sr COrPh- HC COHPh HC OBn O~n SnO o Ho o 08r 9-2 10-1 10-2 jaq HF H 1C CHH-&FH-H
HNH
2 HPd-C OH 0 HO 0 BrO 0 HO O OBn 129 10-3 The cFo lowing compounds were prepared according to Scheme 10. 15 Compound 10-1 HaC COPh On 10-1 i.PrigsCl-LiC1 (0.68 niL, 1.2 N, 0.82 mmol, 2 equiv) was added to a Tff solution (5 nL) ofI9-2 (L70 mg, 0.41 mmol) at 0 C. The reaction was stirred at 0 C for 30 min. Mel (0.2 mL, I 64 mmol, 4 cquiv) was added to the reaction mixture - 130 The reaction was stirred at 0 *C for 30 min and allowed to warm to 25 C over I h. The reaction was quenched with N 1 C1 solution ard extracted with EtQAc. To combined EtOAc extracts were dried (Na 2
SO
4 ) and conceltrated to give crude 10-1. Flash chromatography on silica gel (30:1 hexanes/EtOAc) yielded 3-1 rg of 5Y compound d 10-1. (22%) Compoun d 10-2 H H? HC 10-2 OT3S A TIF solution (1 ml) of 10-1 (31 mg, 0.088 mmol, 1.7 equiv) was add to a 10 TI IF solution (I iL) of LDA (0.13 niL, 1.3 M, 0. 176 mnol, 3.3 equiv) and TMEDA (39 u.L, 0.26 mmol, 4.9 equiv). The reaction was stirred at -78 C for 10 min, A TIF solution (1 niL) of none (26 mg, 0.053 mmol) was added to the reaction at -78 'C. The reaction was stirred at -78 C for 30 Min and allowed to warm to 25 'C over I h, quenched with saturated NH 1 C solution, and extracted with 15 EtOAc, The combined EtOAc extracts were dried (Na 2 SOI) and concentrated to yield the crude 10-2. Preparative reverse phase IHPILC purification onl a Waters Autopurification system using a Simfire Prep CI 8 OBD column [5 pin, 19 x 50 mm, flow rate, 20 mL/min; Solvent A: 1120 with 0.1% 4 HCO 2 H; Solvent B: CH 3 CN with 0.1% HCO 2 H; injection volume: 4.0 mL (CHCN); gradient: 80->100% B over 15 20 min; mass-directed fraction collection] Fractions with the desired MW were collected and concentrated on a RotaVap at 25 ' to remove most of the acetonitrile. The resulting mostly aqueous solution was extracted with EtOAc. The combined EtOAc extracts were dried (Na2SO 4 ) and concentrated to give 10 mg of 10-2 (26%). 25 Compound 10-3
F
3 C N H H HC BnO O HO OBn 10-3 - 131 Aqueous HF (0.3 rmL, 48%) was added to a CIIJCN solution (2 mL) of 10-2 (6 .mg, 0.008 mmol) in. a polypropylene tube at 25 'C The reaction was stirred at 25 "C For 18 h The resulting mixture was poured into an aqueous solution of
K
2
HPO
4 (2 g, dissolved in 15 mL water). The mixture was extracted with EtOAc 5 The combined .EtOA.c extracts were dried (Na 2 SO4) and concentrated to yield crude 10-3. Compound 129 F HIc CHt - OH OH, H C H OH 0 HO 0 0 129 10 Palladium on carbon (2 mg, 10 wt%) was added to a HClIMeOHi solution (0 5N, 2 mL) of the crude 10-3. The reaction was purged with hydrogen and stirred under 1112 (balloon) at 25 'C for 4h. The reaction mixture was filtered through a snall Celite plig. The filtrate was concentrated to yield the crude product. Preparative reverse phase HPLC purification on a Waters Autopurification system 15 using a Phenomenex Polymerx 10 p RP-1 IOA column [10 pm. 150 x 21.20 mirn; flow rate, 20 mL/min; Solvent A: 0 05 N ICI/waIer Solvent B. CIHCN; inection volume: 4.0 mL (0.05 N HCI/water); gradient: 0--->70% B over 7 rin, 70--* 100% over 3 min, and 100% over 5 min; mass-directed Fraction collection], Fractions with the desired MW were collected and freeze-dried to yield 1.5 mg of compound 129: 20 'H NMR (400 Mz, CD 3 01)) 8 7.19 (dJ = 9,7 1Hz, 1 H), 4.07 (s, 1 -), 3,20-2193 (i, 3 11), 3.03 (s, 3 I), 2.96 (s, 3 H), 2.3 1-2.17 (m, 2 H), 2.22 (s, 3 H), 1.69-1.58 (m, 1 H1); MS (PSI) mt 447.23 (M+H). Example 11. Synthesis of Compounds 130-132 25 Scheme I I -132 IF F R 6 F R' F R 6 a, sOuLi/TMEDA a) (COCI)2 b) R' 3 CH2tr or R ICH21 b) PhOH;DMA 2 P BCr3 C<H 00 2 H CO2Ph C2Ph OCH, OCH, OCH3 OH 11.1 11-2 11-3 Hac 2 O F RE HC 'N.CHs F RE H 3 C N'CH, F R HNH2) OH H)d- NF LDAFMEDA C NHH 1)0' b) Oore cncP OH-O HO O 0 BocO 0 HO O OBn OBoc 11-6 11-5 OTBS 11-4 1) HNO, 2) H-/Pd F R HC, N'CH, F R' N TH N H 3 0 H H I ~OH R)r0 2 0B F0 - OH -N"2
H
2 N HR- H H OH 0 HOHO 0 OH 0 HOHO 0 11-7 11-8 In Scheme 11, R', R', and R' is X, R{'U and RA, respectively, as defined in Structural Formula (A). 5 Compounds 130 - 132 were prepared according to Scheme 11. Compound 130 F ' H3'N...CH OH NHZ OH 0 HO 0 0 10 130 Compound 131 mFEt HN-s - H :> OH- C HO D O 131 15 Compound 132 - 133 F Et HCN J H H:= 3 OH H-H NH ~CH-. 0 OHOPO 00 132 Example 1-2. Synthesis of Compounds 133-.135 Scheme 12 F CH3 r 0h, F CH 3 F CH3 a) LDNTMEDA Br 1Br3 Br Bra/HOAr:1 b) C 7 Br C 2) Boc:O AC2OPh Br' C 2 PhI Br CO2Ph Br CO 2 Ph OCH3 OCH OCH 3 OBoc 11-2-1 12-1 12-2 12-3 (Prepar6 according to Scheme 11 using Et In the first step) a) LDNTMEDA b) none F HC HXN'CH 3 CH3C, -CH tH H = iq HF F-G N,
NH
2 Nr N OH 0 11 0 BocO 0 H0;O OBn 13 124 OTBS F HC HC' N'CH, F H>C HC.. CH, H:;N NH1 C~ "Rl- KN i NH 3 OH 01H) 0 O 01 H HOHO O 12-5 1246 In Scheme 1.2. R 9 and R is Rn, and R, respectiv'ely, as defined in Structural Flormnula (A). Compounds 133 - 135 were prepared according to Scheme 12. Compound 133 F HO H3CsN'CH2 :rI HH 6 II OH 0 HO H O 15 133 Compound 134 - 134 F H-IC H ,C.CH, FHH Hd H O ,jC N HC CH HO 134 Compound 135 F H 9 C H-Cs O-H4 S H H H H CN N ... NH H NH C HO C 5 135 Example 13 The antibacterial activities for the compounds of the invention were studied according to the following protocols. 10 Minimum Inhibitory Concentration Assay Frozen bacteral strains were thawed and subcultu red onto Mueller Hiiiriton Broth (Ml [D) or other appropriate media (Sureprococcus requires blood and Haemophilus requires hiemin aid NAD). Following incubation overnight, the strains were subCultured ato Mueller .Hinon Agar and again incubated overnight. 15 Colonies were observed for appropriate colony morphology and lack of contamination Isolated colonies were selected to prepare a starting inoculum equivalent to a 0.5 McFarlan d standard. The starting inoculurm was diluted 1:125 using Mi1i for further use. Test compounds were prepared by dilution in sterile water to a final concentration of 5.1i28g/nL Antibiotics (stored frozen, thawed 20 and used within 3 hours of thawing) and compounds were further diluted to the desired working concentrations. The assays were run as follows, Fifty iL' of MIB was added to wells 2 -- -2 of a 96-well plate. One h hundred L of appropriately diluted antibiotics was added to well . Fifty pL of antibiotics was removed front well 1 and added to well 2 and the 25 contents of well 2 mixed by pipetting up and down five times. Fifty L of the mixture in well 2 was removed and added to well 3 and mixed as above. Serial dilutions were continued in the same manner through wail 12. Fifty pL was removed from well 12 so that all contained 50 il. Fifty Lui. of the working inoculum was then added to all test wells. A growth control well was prepared by adding 50 LII. of -135 working inoculum and 50 L of M.HB to an empty well. The plates were then incubated at 37 "C overnight, removed from the incubator and each well was read on a plate reading mirror. The lowest concentration (MIC) of test compound that inhibited the growth of the bacteria was recorded. 5 Example: 1 2 3 5 6 7 8 9 10 11 12 |AbtJ 32 16 8 4 2 10.5 0,125, 0,0 0.03 0015 grow - - - -+ + + lI) I = mu1I Lioti Ion1centin in the well Grow = bactcial growth coudie) Interpretation: MIC -- 2 pg/mL 10 Protocol for Determining Inoculun Concentration (Viable Count) Ninety IL of sterile 0.9% NaC! was pipetted into wells 2-6 of a 96-well ri crotitor plate. Fifty 50pl of the inoculum was pipetted into well I Ten 1 .L was removed from well I and added It to well 2 followed by mixing. f en L was 15 removed from well two and mixed with the contents of well 3 and so on creating senal dilutions through well 6. Ten pL was removed from each well and spotted onto an appropriate agar plate. The plate was placed into a CO 2 incubator overnight, The colonies in spots that contain distinct colonies wore counted. Viable count was calculated by multiplying the number of colonies by the dil ution factor. 20 Spot from 1 2 4 6 Well Dilution 102 103 104 0 0 F actor Bacterial Strains Fifteen bacterial strains, listed below, were examined in minimum inhibitory concentration (MC) assays. ID Organiism Source Resistance Comments Grain Rx Smith strain (aniimal SAl 00 8 aurcus ATCC 13709 MSSA model) positive SA101. 3, aturus ATCC 29213 MSSA control pstive tet resistant: tet. SA 158 S aureus MR, SK75( positive Micromyx, tet resistant: tet(M) SA161 S. aureus LLC ribosomal protection positive EFI03 E faecalis CATC 292i2 tet control positive -136 intermedi ate/resistant - mchanism not specified tot resistant:ttM (ri b 1 positive EF159 E fccailis MR, 1)S160 protect) cip-R, ery SP106 S pnemnoniae A'CC 49619 wt control positive tot resistant:tet M positive :SP160 S pneurmoniae MR, 54 (rib protect) pen-R, ery-R EC 07 E coli ATCC 25922 wt control negative tet resistant: teLA zel esisa:AA,,LAnegative EC 55 E co/i MR, 10 (efflux) K KPI09 Imnegative KQP109 pncumoniae ATCC 13883 wt K tet resistant:tetA negative KP153 pneumoniae MR, I effluxx) cip-R, gen-R: EC08 E cloacae ATCC 13047 Wt negative AD 110 A .aumni ATCC 19606 wt negative PAl11 P aeruginosa ATCC 27853 wt control negative MSS = methicillin susceptible S, nursus wt = wild type ATCC American Type Culture Collection MR - Marilyn Roborts, University of Washington tot = tctracyvcinoi cip = ciprofloxacin R = resistant gen =genlamicin cry =erytiheromyin pen =piclill Re~sults Values of mllimum inhibition concentration (MIC) lr the compounds of the invention ar provided in Tables 1 a, l b 2a , 2b and I Compound MICs (ug/mL) ID SA101 SA100 SA161 3A158 EF103 EF159 SP106 SPI6 0.125 0.25 0.25 0.0625 0.0625 0.125 <0.0156 O.Ct6 12 0.0156 <0.0156 0.125 0.5 t0D156 0.0625 0.01 56 0.0156 13 5z0.0i156 0.0156 00625 0,25 50.0156 0.0625 50.0156 0.0156 14 0.5 0.25 u.5 0.25 0.125 0.25 0.03 0,125 15 0.5 05 0. 025 Q.5 0.5 0.016 00CIE 16 0.125 0.25 0 25 0.5F 0.625 a.0625 o0.0156 s0.0156 0.25 0.5 2 025 1 0.0625 0.0625 0.0625 50.0156 0,0625 C 125 0>.156 0.0313 <0.0156 <0.0156 - 137 1 0.125 325 0.25 0.25 0.0156 0 25 0.0156 0.0156 20 50.0156 0.25 0.25 0.5 0.0156 0.0025 -0.0150 so.0156 0.25 0.25 0.5 4 0.125 1 20.0156 0.0625 22 1 1 2 4 4 25 1 23 <0 0156 0.5 c.125 0.25 <0.0156 0.0313 0 0156 <0.0156 24 0.25 0.25 C.125 0.125 0.0156 <.0156 0 0156 0 0156 25 0.25 0.125 0.125 0.0313 0.0156 0.0625 0.0155 s0 0156 26 1 1 1 0.25 0.5 50.0156 50 0156 27 2 2 4 16 2 4 0.125 0.5 28 2 2 1 1 1 0.125 0.0325 29 4 4 4 2 2 2 1 1 30 1 4 2 2 2 2 4 31 2 2 1 0.5 1 0.25 0.25 Table Ila Compound MICs (uglmL) ID CiU7 EC15 AB110 PA1l1 EC108 KP209 KP153 1 0.25 2 0.3 16 1 2 12 0.25 8 1 16 1 4 13 0.125 4 0.25 16 1 0 5 14 0.5 4 0.25 26 2 1 1 4 0.125 16 4 2 4 1 16 0.5 8 1 16 1 2 4 17 2 32 05 32 8 4 32 1 0.125 4 0.25 16 0.5 0.5 0.25 4 0.25 16 2 1 4 20 0.25 8 1 8 1 1 4 21 1 16 2 16 4 2 22 16 >32 2 >32 >32 32 >32 23 0.125 0.125 6 1 05 6 24 ~ 0 8 00313 16 2 8 25 105 6 02.25 32 2 2 8 26 0.5 4 0.25 16 2 2 4 27 4 32 1e >32 16 8 32 2 2 10 0.5 32 a 4 16 29 8 8 a >32 8 8 U 30 >32 >32 8 >32 >32 >32 >32 31 2 8 0 >32 8 4 8 Table Ib 5 - 138 Compound MICs (ug/mL) ID SAD 3101 cA100 SA161 SA15 EF103 EF19 SP106 SP160 32 2.125 0.5 0.25 0.5 0.0625 0125 <0.01560. 33 0.25 0.5 1 2 C.25 <. 0.016 0 125 34 <0.0156 .D625 0.0625 .125 <0 0156 <0,0156 50.0156 >0.0156 0.25 025 0.5 0.5 0.25 0.25 0016 0.016 36 0.25 0.5 0.5 1 0.25 0.25 0,0156 s0.0156 37 8 >32 >32 16 15 2 4 38 5 6 16 32 6 13 2 8 39 2 2 16 2 2 0.25 0.5 40 1 1 16 1 1 0,0625 0.5 41 21 2 0.125 0.125 42 0.5 1 2 1 0.5 1 1 0.125 0.DB25 0.5 0.5 1 0.5 0.25 0.5 0156 0.0156 44 4 4 8 8 6 9 0,5 1 450 0.5 1 0.5 0.25 0.5 D 125 0.06 0,25 0.25 0.5 025 0.25 0.5 2125 0.125 Table 2a Compound MICs (ugimL) ID EC107 EC155 Al10 PA111 EC108 KP109 KP'52 32 025 8 2 P 1 1 4 2 >32 4 >32 16 4 '32 34 0.25 2 0.125 16 1 0.5 2 .. 16 0.25 -32 8 4 6 36 2 32 0,125 '32 4 4 32 >32 >32 522 >32 '32 332 >32 38 >2 >32 >32 >32 '32 '32 >32 39 4 >32 32 >32 16 16 >32 40 4 >32 8 32 4 P 32 41 4 16 0,5 32 16 8 16 42 4 16 0,5 >32 16 8 16 1 0125 32 1 4 2 4 32 '32 8 >32 >32 >32 '2 46 1 2 0.06 >32 4 2 4 46 2 4 0125 '32 32 1 32 Table 2b -139 cf l I CICC C C C t- n * I - -f - - - m m mm ~D22 < 22 m u fno< m x0 jzm <m M mm <m mmlomm 22 m Cz -H CQ C- CQ frQ r C rCC << CQ x X f22 C C u 4 C 2 nC C - C (3 '~ C mC mm m mm <22Cm C 0 il r U>. 22 22 22 22 22 < C CQ - __ _ (3 O2 -- C.
o _~ -~Vu::~uzz2v ------- C C i CCC l 1~ -- --. lemmm.C<22.- - <<<e cc|- ee m maQ ara m gm m 7. '- s09 C M09c2 CIe r n h Or c-c - 140 Li C T- m -1 2 Q .I - - 'O - x x m - -- CC [f, ODWq UT4 M0 r J:C4 '-4y4Mm oc CC Q ~4 Q Iq M Q Q QQUCCQQQ 4tP2 X~ 1i ) -Q, CC Co m 4 CQ E M CLQ e 0 -'M C """ Elf - C "- -( - - "- "" - -J- -- > -'J --- - -- r -, -C. : t - Ci>'":-1. V.YC -. D ' -C ia- C '0 C(' ' 0 o~ %c 024 ccriJ)I 0r oQ0000c0 Oum)O3000 M mm mc 2oooooo22 M0M00o o<2coo0000000000 z% mE r r ou~oo O O O O m uu O u ~mCm f12 mm2ommmzmmm2mm 7j- L 0flU~flflm~2m~ mmmncc or cck -r -142 U~rjlc)~0000r< u1 uj 0u I- K) m u Um uC m IL 2 n In. Q 7 O Q Q Q Q Q Q Q CQ , Q QQQQQ < < rcj ; c 000000mmQQ QQ QQQ omm QQCQ p 2 n 0 000- mmm emmom-Nmm1n e-am omo m+ninn < 7- O O 3 o O .- .- - -. - - -- - ce CN 4 c C ca C4 C r - .
- 143 F I ~ I ~ &1, K Cr 4'l C r Cl -I _ ~ K) K) zI Cr' 'r ('I <r~ A fl (1 C - tt-~ Ifl -z ~r'1 In I/i -, - ' Cl fl ~J rf~ Cl (~l cC Cl 33 K) ~zCr' ~ Cl ""C4 7' ~ Cl Cy K) d -j 'C> - C,-, fl Cl IS '~2 o ~ - 144 EDx1V study Compounds of the present invention were evaluated for efficacy in a mouse systemic infection model against a susceptible S. am-eus isolate, ATCC 13709. 5 Mice were infected via intraperitoneal injection with a bacterial load that would result in 0% survival within 48 hours after infection. Mice received the test compounds 60 m inaes post infection via intravenous injection. Infection control grou ups did not receive treatment Survival was assessed over a 48 hour period. Percent survival was calculated and PD,: values determined using Probit analysis. 10 The protocol for tetracycline-resistance sepsis study was simiar to the protocol for rFVo IV study described above except the infection model used S HMreuS SA1 61. a tetracycline-resistant strain. 15 GN sepsis The protocol for GN sepsis study was smiar to the protocol for EVso IV study described above except the infection model used I coih ATCC 25922. 20 Metabolic stability A stock solution of the test compounds were prepared by dissolving the compound in DMSO to a final concentration of 1.0 mg/(mL The analyses and internal standard were diluted arid infused imo the LC/MS system to determine optimal ionization, polarity and MS'MS fragmentation for 25 selection of specific MR M multiplee reaction monitoring) transitions Generic chromatographic conditions were developed with a less than 5 minute cycle tine Pooled human microsome assays were conducted at 0.2 ng/mL protein with an NADPH generating cofactor system (1.3mM NADP+, 3.3mM gLucose-6 phosphate. 0.4Ui/mL glucose-6-phosphate dehydrogenase, and 3.3mM magnesium 30 chloride). lest compounds were diluted from DMSO stocks to I pM reaction concentrations. Time points were removed at 0 and 60 minutes. After a protein crash - 14-5 with acetonitrile, samples are analyzed by LC/MS/MS. Peak areas are compared to the time 0 sample, and the percent analyte remaining is calculated. A reaction containing no cofactor was used to control for analyte loss due to non-specific binding, thermal degradation and solubility. 5 SA MIC90 Iwenty randomly selected clinical isolates of S aurcus were used to determine the minimal inhibitory concentration (I1C) of test compounds for 90% of the isolates (MIC'a). MICs w'ere performed by rnicrotiter broth dilution in a 96-well 10 format according to Clinical Laboratory Standards Institute (CLS[) guidelines, as described above. Viable counts were determined by I 0-fold serial dilution. Dilutions were prepared in sterile 0.9% NaCI Ten micioliters of the inoculum and of each of 5 dilutions were plated onto blood or Mueller Hinton agar plates, incubated overnight 15 at 37 "C with 5% CO., and counted. TeiR MIC90 Ten isolates selected based on resistance to tetracvcline were used to determine the M]Cgc as described above. 20 UC MIC90 Twenty randomly sciected clinical isolates of E coli were used to determine the ICo as described above. 25 Protein bin din 'est compounds were prepared as 1 0 ng/mL stock solutions in DISO The analytes and internal standard were dilited and infused into the LC/MS system to detennine optimal ionization, polarity and MS/MS fragmentation for selection of specific MRM (multiple reaction monitoring) transitions. Generic chromatographic 30 conditions were developed with a Icss than 5 minute cycle time.
- 146 The DMSO stocks were diluted to I and 10 ig/mL in human plasma and incubated in RED devices for 4 hours at 37 "C. The time point was removed at the end of the incubation period. After a protein crash with acetonitrile, the samples were analyzed by LC/MS/MS. Peak areas for the buffer (receiver) and sample 5 (donor) chambers were compared and the protein bound fraction is calculated, Analysis was conducted in duplicate. Thigh burden Female CD-1 mice were pre-treated with cyclophosphamide to render the 10 ice neutropenic. Mice were infected with S. aureus ATCC 13709 via injection into the right thigh muscle of 0 1 ml per mouse. One and a half hours post infection mice were treated IV with test compounds in doses ranging from 0.3 to 30 mg/kg or C.3 to 20 mg/kg. Four mice were relatedd with each drug conceritration. Iwent-four hours post treatment, moice were euthanized by C02 inhalation. The right thighs of 15 the mice were aseptically removed, weighed, homogenized, serially diluted, and plated on TSA medium The plates were incubated overnight at 37 C in 5% CO, Colony forming units per gram of thigh was calculated by enumerating the plated colonies then adjusting for serial dilutions and the weight of the thigh 20 The results for the biological activity studies described above are listed in Table 4.
- 147 0.i ~~Ln j," rL D co T- cc i cLc 0 0 00 0O(V i ccco U) I- Lo 0 6 6: CJ - j 7CA *os.~ Cr Z,. Ms -2 V -149 While this invention has been particularly shown and described with references to example embodiments thereof, it will be understood by those skilled in 5 the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appenoed claims.

Claims (30)

1. A compound represented by Sturctural Formula (A): H 3 C, -CH 3 F X N H H= OH NH 2 0 OH 0 HOHO 0 (A), 5 or a pharmaceutically acceptable salt thereof, wherein: X is selected from hydrogen, -(CI-C 7 )alkyl, carbocyclyl, aryl and heteroaryl; Y is selected from hydrogen, -(C 1 -C 7 )alkyl, carbocyclyl, -(C C 4 )alkylene-N(RA)(R B 10 -(CC4)alkylene-N(.R. )-C(O)-[C(R")(RE o)1-4-N(R^ )(RS), -CHJ=N-OR^ -N(R^A)(R B), -N(RF')-C(O)-[C(R")(RE')]I-s4N(RA)(RB), -N(RF)-C(O)-N(R.")(R'), -N(RF CO)(C-6)ak -N(R )-C(O)-heterocyclyl, -N(R')-C(O)-heteroaryl, -N(R)-C(O)-carbocyclyl, -N(RF)-C(O)-aryl, 15 -N(RF)-S(O) 1 -(C 1 -C 4 )akylene-N(R^)(RB), -N(RF)-S(O),,-(C 1 -C 4 )alkylene-carbocyclyl, and -N(R')-S(O)T-(CI-C4)alkylene-aryI wherein: at least one of X and Y is not hydrogen; each R' and R" are independently selected from hydrogen, 20 (C 1 -C 7 )alkyl, -O-(C -C7)alkyl, -(Co-C 6 )alkylene-carbocyclyl, -(CO-C 6 )aikylene-aryl, -(Co-C6)alkylene-heterocyclyl, -(Co-C)alkylene-heteroaryl, -(C 1 -C6)alkylene-O-carbocyclyl, -(C -C 6 )alkylene-O-aryl, -(C] -C 6 )alkylene-O-heterocyclyl, -(C -C 6 )alkylene-O-heteroaryl, -S(O)m-(C -C 6 )alkyl, 25 -(Co-C 4 )alkylen e-S(O)m,-carbocyclyl, -(CO-C 4 )alkylene-S(O),, aryl, - 151 -(Co-C4)alkylene-S(O),-heterocycly and -(Co-C 4 )alkylene-S(O),-heteroaryl; or R^ and RB taken together with the nitrogen atom to which they are bound form a heterocyclyl or heteroaryl, wherein the heterocycle or 5 heteroaryl optionally comprises 1 to 4 additional heteroatoms independently selected from N, S and 0; each R') and each Re is independently selected from hydrogen, (C 1 -C 6 )alkyl, carbocyclyl, aryl, heterocyclyl, heteroaryl, or a naturally occurring amino acid side chain moiety, or 10 RD and RE taken together with the carbon atom to which they are bound form a 3-7 membered carbocyclyl, or a 4-7 membered heterocyclyl, wherein the heterocyclyl formed by R and RE optionally comprises one to two additional heteroatoms independently selected from N, S and 0; RF is selected from hydrogen, (C] -C 7 )alkyl, carbocyclyl, aryl and 15 heteroaryl; and m is 1 or 2, wherein: each carbocyclyl, aryl, heterocyclyl or heteroaryl is optionally and independently substituted with one or more substituents independently selected from halo, -(CI-C 4 )alkyl, -OH, =0, -O-(C 1 -C4)alkyl, 20 -(C-C 4 )alky]-O-(CI-C 4 )alkyl, halo-substituted -(C-C4)alkyl, halo substituted -O-(CI-C 4 )alkyl, -C(O)-(C C.4)alkyl, -C(0)-(fluoro-substituted-(C'iC4)alkl,:,), -S(0),,,(C" [C4)alkyl, -N(R'-)(R""), and CN; each alkyl in the group represented by R, R', RD and R is 25 optionally and independently substituted with one or more substituents independently selected from halo, -(C -C4)alkyl, -OH, -0-(C-CC7)alkyl, -(Ce-C4)alkyl-O-(CI-C 4 )alkyl, fluoro-substituted-(C -C4)alkyl, -S(O)m-(CI-C4)alkyl, and -N(R)(R), wherein each RD is hydrogen or (C 1 -C4)alkyl, wherein each alkyl in the group 30 represented by R( is optionally and independently substituted with one or more substituents independently selected from -(CI-C 4 )alkyl, (C3 C 6 )cycloalkyl, halo, -01, -O-(C -C4)alkyl, and (C-C 4 )alkyl-O-(C-C4)alkyl. - 152
2. The compound of Claim I, wherein the compound is represented by the following Stuctural Formula: H 3 C, -CH 3 F X N H H7 OH R N NH 2 2N6 1F OH O HO HO 0 5R (), or a pharmaceutically acceptable salt thereof, wherein: R' and R2 are each independently selected from hydrogen, (CI C7)alkyl, (C 3 -C 6 )cycloalkyl(Ci -C4)alkyl, (CI-C 7 )alkoxy(C -C 4 )alkyl, (Cr C 6 )cVcloalkoxy(C-C 4 )alkyl, (C 3 -C6)cycloalkyl, aryl, aryl(C-C4)alkvl, 10 arvloxy(CpC4)alkyl, arylthio(C -C4)alkyl, arylsufinyl(C 1 -C 4 )alkyl, arylsulfonyl(CI -C4)alkyl, and -O-(C-C7)alkyl, or R I and R2 taken together with the nitrogen atom to which they are bonded form a monocyclic or bicyclic heteroaryl, or a monocyclic, fused bicyclic, bridged bicyclic or spiro bicyclic heterocycle, wherein the 15 heteroaryl or heterocycle optionally contains one or two additional heteroatoms independently selected from N, 0 and S; and wherein each alkyl, cycloalkyl, alkoxy and cycloalkoxy moiety in the groups represented by R' and R 2 and each heterocycle represented by NR R2 taken together is optionally substituted with one or more substituents 20 independently selected from the group consisting of (C]-C 4 )alkyl, halo, -OH, (CI-C 4 )alkoxy, (CI -C4alkylthio, (CI -C4)alkylsulfinyl, (C] -C4)alkylsulfonyl, (C-C4)alkoxy(Ci-C4 1 )alkyl, and -N(R)(R 4 ); and each aryl, aryloxy, arylthio, arylsufinyl and arylsulfonyl moiety in the groups represented by R' and R 2 and each heteroaryl represented by NRI R2 25 taken together is optionally substituted with one or more substituents independently selected from the group consisting of (C]-C4)alkyl, halo, -OH1, (C -C 4 )alkoxy, -S-(C -C 4 )alkyl, -S(0)(CI-C 4 )alkyl, -S(O)2(CI-C 4 )alkyl, - 153 (C 1 -C4)alkoxy(C -C 4 )alkyl, -N(R 3 )(R 4 ); -CN, halo(C -C4)alkyl, and halo(Cp C4)alkoxy, and R' and R' are each independently selected from the group consisting of -H and (CI-C4)alkyl, wherein the (C 1 -C 4 )alkyl represented by R 3 and R 4 is 5 optionally substituted with one or more substituents independently selected from the group consisting of(C-C4)alkyl, halo, -OH, (C 1 -C 4 )alkoxy, and (C -C 4 )alkoxy(C-C 4 )alkyl.
3. The compound of Claim 2, wherein the compound is represented by the 10 following Structural Formula: H 3 C, -CH 3 F N H H =: RH H OH 2 -NH 2 OH O HOHO 0 (I) or a pharmaceutically acceptable salt thereof, wherein. RI and R 2 are each independently selected from hydrogen, (Ci-C 7 )alkyl, (C 3 -C 6 )cycloalkyl(CI-C 4 )alkyl, (C-C 7 )alkoxy(CI-C 4 )alkyl, 15 (C--C6)cycloalkoxy(C 1 -C4)ailkyl, (C 3 -Cs)cycloalkyl, aryl, aryl(C-C4)alkyl, aryloxy(CI-C 4 )alkyl, arylthio(Ci-C4)alkyl, arylsufinyl(CI-C 4 )alkyl, arylsulfonyl(Cj-C4)alkyl; or R' and R 2 taken together with the nitrogen atom to which they are bonded form a monocyclic or bicyclic heteroaryl, or a monocyclic, fused 20 bicyclic, bridged bicyclic or spiro bicyclic heterocycle, wherein the heteroaryl or heterocycle optionally contains one or two additional heteroatoms independently selected from N, 0 and S.
4. The compound of Claim 3, wherein R' is hydrogen or a (CiC4)alkyl. 25
5. The compound of Claims 3 or 4, wherein R 2 is selected from (Ci-C 7 )alkyl, (C-C 6 )cycloalkyl(C -C 4 )alkyl, (C-C 7 )alkoxy(C-C4)alkyl, phenyl, phenyl(C 1 -C4)alkyl, (C 3 -C 6 )cycloalkyl and halo(C-C 4 )alkyl, wherein each - 154 alkyl, alkoxy and cycloalkyl moiety in the groups represented by R 2 is optionally substituted with one or more substituents independently selected from the group consisting of (C -C 4 )alkyl and halo; and each phenyl moiety in the groups represented by R 2 is optionally substituted with one or more 5 substituents independently selected from the group consisting of (Cl-C 4 )alkyl, halo, (CI-C 4 )alkoxy, (C 1 -C 4 )alkoxy(C-C4)alkyl, -CN, halo(CI-C 4 )alkyl, and halo(C 1 -C 4 )alkoxy.
6. The compound of any one of Claims 3-5, wherein R' is selected from 10) hydrogen, methyl and ethyl.
7. The compound of Claim 6, wherein R 2 is selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, cyclobutylmethyl, phenyl, benzyl, -(CH 2 ) 2 -0-CH 3 , -(CIH 2 ) 3 -OCf3, 15 -C(CH 3 ) 3 , -CI(C-l 3 ) 2 , -CI 2 C(CH,)-, -CH1 2 CH{(CH 3 ) 2 , -C-1 2 -CF 3 , -(C1 2 ) 2 -CH 2 F, and -(CHI2)aC1 3 ; n is 0, 1, 2, 3, 4, 5 or 6; and wherein the phenyl or benzyl group represented by R2 is optionally substituted with one or two substituents independently selected from the group consisting of (C-C4)alkyl, halogen, (CI-C 4 )alkoxy, (C-C4)alkoxy(Cr -C 4 )alkyl, -CN, 20 halo(CI -C4)alkyl, and halo(CI-C4)alkoxy.
8. The compound of Claim 7, wherein R2 is selected from cyclopropyl, cyclopropyl methyl, cyclobutyl, cyclopentyl, cyclohexyl, -(CH 2 ) 2 -O-CHI, -C(CH.;)3, -CH(CH 3 ) 2 , -CH-CF,, -CIH2CH(CHi) 2 , -CH 3 and -CH 2 CH 3 . 25
9. The compound of Claim 3, wherein R' and R 2 taken together with the nitrogen atom to which they are bonded form a monocyclic or bicyclic heteroaryl, or a monocyclic, fused bicyclic, bridged bicyclic or spiro bicyclic heterocycle, wherein the heteroaryl or heterocycle optionally contains one 30 additional heteroatom selected from N, 0 and S; and the heterocycle is optionally substituted with one or more substituents independently selected front the group consisting of (Cr-C 4 )akyl, halo, -OH, (CI-C.I)alkoxy, - 155 (C 1 -C 4 )alkylthio, (C-C 4 )alkylsulfinyl, (C 1 -C 4 )alkylsulfonyl, (C 1 -C 4 )alkoxy(C1-C 4 )alkyl, and -N(R )(R'); and the heteroaryl is optionally substituted with one or more substituents independently selected from the groLip consisting of (C 1 -C 4 )alkyl, halo, -OH, (Cr C-4t)alkoxy, -S-(C -C4)alkyl, 5 -S(O)(Ce-C 4 )alkyl, -S(0) 2 (CI -C 4 )alkyl, (C -C 4 )alkoxy(Cj-C 4 )alkyl, -N(R)(R 4 ), -CN, halo(C-C 4 )alkyl, and halo(Ci-C4)alkoxy.
10. The compound of Claim 9, wherein R' and R taken together with the nitrogen atom to which they are bonded form a heterocycle selected from the 10 group consisting of azetidine, pyrrolidine, morpholine, piperidine, octahydrocyclopenta[c]pyrrol, isoindoline, and azabicyclo[3. 1.0]hexane, wherein the heterocycle is optionally substituted with one or more substituents independently selected from the group consisting of (C -C4)alkyl, halogen, -011, (C-C 4 )alkoxy, -S-(Cj-C.4)alkyl, 15 -S(O)(Ce-C 4 )alkyl, -S(O) 2 (CI-C 4 )alkyl, (C 1 -C 4 )alkoxy(C]-C 4 )alkyl, and
11. The compound of Claim 10, wherein the heterocycle is optionally substituted with halogen, methoxy, hydroxy, methoxymethyl or dimethylamino group. 20
12, The compound of Claim 3, wherein: a) R' is methyl, and R 2 is cyclopropyl; b) R 1 is hydrogen, and R 2 is cyclopropyl; c) R' is hydrogen, and R 2 is cy clobutyl; 25 d) R' is methyl, and R 2 is cyclobutyl; e) R' is hydrogen, and R 2 is cyclopropylmethyl; f) R' is hydrogen, and R 2 is cyclobutylmethyl; g) R' is hydrogen, and .R 2 is benzyl; h) R' is hydrogen, and R 2 is methoxypropyl; 30 i) R 1 is hydrogen, and R2 is methoxyethyl; j) R' is hydrogen, and R 2 is phenyl; k) R' is methyl, and R 2 is t-butyl; - 156 1) R' is hydrogen, and R 2 is t-butyl; m) R 1 is hydrogen, and R 2 is methyl; n) R' is hydrogen, and R 2 is ethyl; o) R' is hydrogen, and R2 is propyl; 5 p) R' is hydrogen, and R2 is butyl; q) R1 is hydrogen, and R 2 is pentyl; r) R 1 is hydrogen, and R2 is hexyl; s) R1 is hydrogen, and R 2 is heptyl; t) R 1 is methyl, and R2 is methyl; 10 u) R' is hydrogen, and R' is isopropyl; v) R1 is hydrogen, and R.2 is 2,2-dimethylpropyl; w) R1 is hydrogen, and R2 is trifluoroethyl; x) R1 is hydrogen, and R2 is 2-methylpropyl; y) R' is hydrogen, and R2 is 3-fluoropropyl, 15 z) R' is ethyl, and R2 is ethyl; al) R' is methyl, and R 2 is methyl; bI) R. is hydrogen, and R 2 is hydrogen; c1) R is hydrogen, and R 2 is cyclopentyl; dl) R' is methyl, and R2 is cyclopentyl; or 20 e1) R' is methyl, and R2 is propel, or a pharmaceutically acceptable salt of any of the foregoing,
13, The compound of Clairm 3, wherein R and 12 taken together with the nitrogen atom to which they are bonded form a group selected from: 25 a) azetidin-I-yl; b) 3-fluoroazetidin- I -yl; c) 3 -methylazetidi n-I -yl; d) 3-methoxyazetidin-.1 -yl; e) pyrrolidin-1-yl; 30 f) morpholin-4-yJ; g) 3-fluoropyrrolidin-1-yl; h) 3-hydroxypyrrolidin-i-yl; -157 i) -N,N-dimethylaminopyrrolidin-1-yl; j) 2-methoxymethylpyrrolidin-1-yl-; k) piper di n-1 -y.; 1) octahydrocyclopenta[c]pyrrol-2-yl; 5 in) isoindolin-2-yl; and n) 3-azabicyclo[3. 1.O]hexan-3-yl, or a pharmaceutically acceptable salt of any of the foregoing.
14. The compound of claim 3, wherein 10 R 1 is hydrogen or a (CI -C 4 )alkyl; and R 2 is selected from (CC -7)alkyl, (C3-Cs)cycloalkyl(C -C 4 )alkyl, (C-C 7 )alkoxy(C-C4)alkyl, phenyl, phenyl(Ci-C4)alkyl, (C 3 -Q)cycloalkyl and halo(C 1 -C 4 )alkyl, wherein each alkyl, alkoxy and cycloalkyl moiety in the groups represented by R2 is optionally substituted with one or more 15 substituents independently selected from the group consisting of (Cl-C 4 )alkyl and halo; and each phenyl moiety in the groups represented by R 2 is optionally substituted with one or more substituents independently selected from the group consisting of (C-C4)alkyl, halo, (C] -C4)alkoxy, (C-C 4 )alkoxy(CL-C 4 )alkyl, -CN, halo(Cj-C 4 )alkyl, and halo(CI-C4I)alkoxy; 20 or R' and R 2 taken together with the nitrogen atom to which they are bonded form a monocyclic or bicyclic heteroaryl, or a monocyclic, fused bicyclic, bridged bicyclic or spiro bicyclic heterocycle, wherein the heteroaryl or heterocycle optionally contains one additional heteroatom 25 selected from N, 0 and S; and the heterocycle is optionally substituted with one or more substituents independently selected from the group consisting of (CI-C 4 )alkyl, halo, -011, (C 1 -C.i.)alkoxy, (-C)alkylthio, (C 1 -C 4 )alkylsulfinyl, (C-4)alkylsulfonyl, (C 1 .C4)alkoxy(C-C 4 )alkyl, and -N(R )(R 4 ); and the heteroaryl is optionally substituted with one or more 30 substituents independently selected from the group consisting of (CI-C 4 )alkyl, halo, -OH, (C -C 4 )alkoxy, -S-(CI-C 4 )alkyl, -S(O)(C-C4)alkyl, -S(O) 2 (C-C4)alkyl, (C-C4)alkoxy(Cj-C4)alkyl, -N(R3)(R 4 ), -CN, - 158 halo(C-C4)alkyl, and halo(C-C4)alkoxy.
15. The compound of Claim 2, wherein R 1 is hydrogen, methyl, ethyl, methoxy or tert-butoxy; 5 R2 is selected from (CI-C 7 )alkyl, (C 3 -C 6 )cycloalkyi(C-C 4 )alkyl, (C 1 -C 7 )alkoxy(C 1 -C 4 )alkyl, phenyl, (C 3 -C 6 )cycloalkyl, and fluoro(CI C 4 )alkyl; or R' and R2 taken together with the nitrogen atom to which they are bonded form a ring selected from pyrrolidinyl, morpholinyl, azetidinyl, 10 piperidinyl, octahydrocyclopenta[c]pyrrolyl, isoindolinyl, indazolyl, imidazolyl, pyrazolyl, triazolyl, and tetrazolyl, wherein the ring formed by R 1 and R2 taken together with the nitrogen atom to which they are bonded is optionally substituted with fluoro, -OH, -OCH 3 , or N(C:H 3 ) 2 . 15
16, The compound of Claim 3, wherein: R hydrogen, methyl, or ethyl R 2 is selected from methyl, ethyl, n-propyl, isopropyl, n-butyl, 2,2 dimethylpropyl, t-butyl, isobutyl, n-pentyl, (C 4 -Cs)cycloalkyl, (C,, Ci)cycloalkylimethyl, methoxyethyl, and 2-fluoroethyl; or 20 !R 1 and R2 taken together with the nitrogen atom to which they are bonded form a ring selected from azetidinyl, pyrrolidinyl, piperidinyl, tetrazolyl, or octahydrocyclopenta[c]pyrrolyl, and wherein the ring formed by R' and R2 taken together with the nitrogen atom to which they are bonded is optionally substituted with fluoro, 25
1 7. A pharmaceutical composition comprising a pharmaceutically acceptable carrier or diluent and a compound of any one of Claims 1-16 or a salt thereof. 30
18. A method for treating or preventing an infection or colonization in a subject comprising administering to the subject an effective amount of a compound - 159 of any one of Claims 1-16 or a salt thereof.
19. The method of Claim 18, wherein the infection is caused by a Gram-positive organism. 5
20. The method of Claim 19, wherein the Gram-positive organism is selected from the group consisting of Staphylococcus spp, Streptococcus spp-, Propionibaclerium spp., Enterococcus spp., Bacillus spp., (orynebacteriui spp., Nocardia spp,, Clostridium spp., Actinobacteria spp., and Listeria spp. 10
21. The method of Claim 18, wherein the infection is caused by a Gram-negative organism.
22. The method of Claim 21, wherein the Gram-negative organism is selected 15 form the group consisting of Enterobactericeac, Bacteroidaceace, Vibrionaceae, Pasteurellae, Pseudomonadaceace, Neisseriaceae, Rickettsiae, Moraxellaceae, any species of Proteeae, Acinetobacter spp., Helicobacter spp., and (ampylobacter spp. 20
23. The method of Claim 18, wherein the infection is caused by an organism selected from the group consisting of rickettsiae, chlamydiae, Legionella spp, My)coplasna spp., and any other intracellular pathogens.
24. The method of Claim 18, wherein the infection is caused by more than one 25 organism.
25. The method of Claim 18, wherein the infection is caused by an organism resistant to one or more antibiotics. 30
26. The method of Claim 25, wherein the infection is caused by an organism resistant to tetracycline or any member of first and second generation of - 160 tetracycline antibiotics ,
27. The method of Claim 25, wherein the infection is caused by an organism resistant to methicillin or any antibiotic in the -lactam class. 5
28. The method of Claim 25, wherein the infection is caused by an organism resistant to vancomycin.
29. The method of Claim 25 wherein the infection is caused by an organism 10 resistant to a quinolone or fluoroquinolone.
30. The method of Claim 25 wherein the infection is caused by an organism resistant to tigecycline.
AU2015203286A 2008-08-08 2015-06-16 C7-fluoro substituted tetracycline compounds Active AU2015203286B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2015203286A AU2015203286B2 (en) 2008-08-08 2015-06-16 C7-fluoro substituted tetracycline compounds
AU2016259418A AU2016259418A1 (en) 2008-08-08 2016-11-18 C7-fluoro substituted tetracycline compounds

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US61/188,307 2008-08-08
AU2013200565A AU2013200565B2 (en) 2008-08-08 2013-02-04 C7-fluoro substituted tetracycline compounds
AU2015203286A AU2015203286B2 (en) 2008-08-08 2015-06-16 C7-fluoro substituted tetracycline compounds

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2013200565A Division AU2013200565B2 (en) 2008-08-08 2013-02-04 C7-fluoro substituted tetracycline compounds

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2016259418A Division AU2016259418A1 (en) 2008-08-08 2016-11-18 C7-fluoro substituted tetracycline compounds

Publications (2)

Publication Number Publication Date
AU2015203286A1 true AU2015203286A1 (en) 2015-07-09
AU2015203286B2 AU2015203286B2 (en) 2016-12-15

Family

ID=53627545

Family Applications (3)

Application Number Title Priority Date Filing Date
AU2013200565A Ceased AU2013200565B2 (en) 2008-08-08 2013-02-04 C7-fluoro substituted tetracycline compounds
AU2015203286A Active AU2015203286B2 (en) 2008-08-08 2015-06-16 C7-fluoro substituted tetracycline compounds
AU2016259418A Abandoned AU2016259418A1 (en) 2008-08-08 2016-11-18 C7-fluoro substituted tetracycline compounds

Family Applications Before (1)

Application Number Title Priority Date Filing Date
AU2013200565A Ceased AU2013200565B2 (en) 2008-08-08 2013-02-04 C7-fluoro substituted tetracycline compounds

Family Applications After (1)

Application Number Title Priority Date Filing Date
AU2016259418A Abandoned AU2016259418A1 (en) 2008-08-08 2016-11-18 C7-fluoro substituted tetracycline compounds

Country Status (1)

Country Link
AU (3) AU2013200565B2 (en)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB935384A (en) * 1959-03-30 1963-08-28 Pfizer & Co C Tetracycline derivatives and preparation thereof

Also Published As

Publication number Publication date
AU2013200565A1 (en) 2013-02-21
AU2013200565B2 (en) 2015-07-09
AU2015203286B2 (en) 2016-12-15
AU2016259418A1 (en) 2016-12-08

Similar Documents

Publication Publication Date Title
AU2009279473B2 (en) C7-fluoro substituted tetracycline compounds
US10072007B2 (en) Tetracycline compounds
EP2470500B1 (en) Tetracycline compounds
AU2015203286A1 (en) C7-fluoro substituted tetracycline compounds

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)