Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
FUSED RING ANALOGUES OF ANTI-FIBROTIC AGENTS
Document Type and Number:
WIPO Patent Application WO/2011/047432
Kind Code:
A1
Abstract:
The present invention relates to arylcarbonyl and heteroarylcarbonyl anthranilate compounds that may be useful as anti-fibrotic agents. The present invention also relates to methods for their preparation, pharmaceutical compositions containing these compounds and uses of these compounds in the treatment disorders.

Inventors:
WILLIAMS SPENCER JOHN (AU)
ZAMMIT STEVEN (AU)
KELLY DARREN JAMES (AU)
Application Number:
PCT/AU2010/001398
Publication Date:
April 28, 2011
Filing Date:
October 21, 2010
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
FIBROTECH THERAPEUTICS PTY LTD (AU)
WILLIAMS SPENCER JOHN (AU)
ZAMMIT STEVEN (AU)
KELLY DARREN JAMES (AU)
International Classes:
C07D413/04; A61K31/343; A61K31/352; A61K31/381; A61K31/382; A61K31/403; A61K31/4035; A61K31/404; A61K31/416; A61K31/4184; A61K31/424; A61K31/428; A61K31/429; A61K31/435; A61K31/4355; A61K31/4365; A61K31/437; A61K31/498; A61K31/506; A61K31/517; A61K31/536; A61K31/5365; A61K31/538; A61K31/5383; A61K31/5415; A61P9/00; A61P11/00; A61P13/12; A61P17/02; A61P29/00; A61P35/00; C07C233/81; C07D209/12; C07D209/34; C07D209/46; C07D211/18; C07D215/233; C07D215/58; C07D235/06; C07D235/12; C07D235/16; C07D235/26; C07D235/28; C07D239/88; C07D239/91; C07D241/42; C07D241/44; C07D261/20; C07D263/56; C07D263/58; C07D265/22; C07D265/24; C07D265/36; C07D275/04; C07D277/62; C07D277/64; C07D277/68; C07D277/70; C07D279/16; C07D291/08; C07D307/87; C07D311/04; C07D311/08; C07D311/76; C07D317/70; C07D333/62; C07D333/64; C07D335/06; C07D401/04; C07D401/12; C07D403/04; C07D403/12; C07D405/12; C07D409/12; C07D417/04
Domestic Patent References:
WO2005030705A12005-04-07
WO2008003141A12008-01-10
Foreign References:
GB2372986A2002-09-11
US4769384A1988-09-06
Other References:
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 2004:198513, XP008156462
SATO, A ET AL.: "A.Novel Class of in Vivo Active Anticancer Agents: Achiral seco-Amino- and seco=Hydroxycyclopropyllienz[e]indolone (seco-CBI) Analogues of the Duocarmycins and-CC-1065", JOURNAL OF MEDICINAL CHEMISTRY, vol. 48, no. 11, 2005, pages 3903 - 3918, XP008155007
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 2009:875995, XP008156461
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 1994:244706, XP008156460
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 1999:420913, XP008156463
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 1999:420913, XP008156463
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 1987:4861, XP008156465
MESSIHA, N. N. ET AL.: "Synthesis of Some Benzoxazin-4-ones, Quinazolin-4-ones and the Related Products", INDIAN JOURNAL OF CHEMISTRY, vol. 13, 1975, pages 326 - 328, XP008155008
GAZIT, A. ET AL.: "Tyrphostins. 2. Heterocyclic and a-Substituted Benzylidenemalononnitrile Tyrphostins as Potent Inhibitors of EGF Receptor and ErbB2/neu Tyrosine Kinases", JOURNAL OF MEDICINAL CHEMISTRY, vol. 32, no. 17, 1991, pages 1896 - 1907, XP008155011
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 1999:182701, XP008156466
GREENE, T. W.; WUTS, P. G. M.: "Protective Groups in Organic Synthesis", 1991, WILEY-INTERSCIENCE
"Protective Groups in Organic Chemistry", 1973, PLENUM PRESS
KOCIENSKI, P. J.: "Protecting Groups", 2000, THEIME MEDICAL PUB.
"Remington's Pharmaceutical Sciences", 1995, MACK PUBLISHING CO.
REDFORD MM, JAMA, vol. 289, 2003, pages 194 - 203
"Remingtons Pharmaceutical Sciences", 1995, MACK PUBLISHING CO.
"T.W. Greene's Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS
JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY, vol. 51, no. 3, 2003, pages 594 - 600
KAKIZAKI Y; KRAFT N; ATKINS RC: "Differential control of mesangial cell proliferation by interferon-gamma", CLIN EXP IMMUNOL, vol. 85, 1991, pages 157 - 163
See also references of EP 2491030A4
Attorney, Agent or Firm:
PIZZEYS PATENT & TRADE MARK ATTORNEYS (324 Queen StreetBrisbane, QLD 4000, AU)
Download PDF:
Claims:
What is claimed is:

1. A compound of Formula (I)

Formula (I) or a pharmaceutically acceptable salt or prodrug thereof, wherein:

- W is selected from the group consisting of: CR7 and N;

- A is selected from the group consisting of: -(CR8RV(Y)q-(C(O)MCR10R1 V and -(CR8RV(C(O))r(Y)q-(CR10R1V, wherein Y is selected from the group consisting of: O, S, NR12, each p and s are an integer independently selected from the group consisting of 0, 1 , and 2, each q and r are an integer independently selected from the group consisting of 0 and 1 , and p + q + r + s is an integer selected from the group consisting of 1 , 2, and 3;

- Z -Z2 is selected from the group consisting of N-C= and C=C;

- X1 selected from the group consisting of: C=0, CF2 or S02, P02;

- X2 is selected from the group consisting of: NR13 and (CH2)t wherein t is an integer selected from the group consisting of: 0 and 1 ;

- D is selected from the group consisting of: a cycloalkyi, heterocycloalkyi, cycloalkenyl, heterocycloalkenyl, aryl or heteroaryl ring;

- R1, R4, R5, R®, R7, RB, R9, R10, and R 1 are each independently selected from the group consisting of: H, halogen, OH, N02, CN, NH2, optionally substituted C C 2 afkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted C Ci0 heteroalkyl, optionally substituted C3-d2 cycloalkyi, optionally substituted C3-C12 cycloalkenyl, optionally substituted C2-C 2 heterocycloalkyi, optionally substituted C2-C12 heterocycloalkenyl, optionally substituted Ce-C18 aryl, optionally substituted ( Cie heteroaryl, optionally substituted d-C12 alkyloxy, optionally substituted C2-C12 alkenyloxy, optionally substituted C2- C12 alkynyloxy, optionally substituted f- do heteroalkyloxy, optionally substituted C3-C12 cycloalkyloxy, optionally substituted C3-C12 cycloalkenyloxy, optionally substituted C C12 heterocycloalkyloxy, optionally substituted Ci-C 2 heterocycloalkenyloxy, optionally substituted C8-C18 aryloxy, optionally substituted Ci-C18 heteroaryloxy, optionally substituted Ci-dz alkylamino, SR14, S03H, S02NR15Rie, S02R14, SONR15Rie, SOR 4, COR14, COOH, COOR14, CONR15Rie, NR15COR14, NR15COOR14, NR15S02R14, NR15CONR15Rie, NR15R18, and acyl;

R2 and R3, are each independently selected from the group consisting of. H, OH, N02, CN, NH2l optionally substituted Ci-C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted CrCi2 alkynyl, optionally substituted Ci-C10 heteroalkyl, optionally substituted C3-C12 cycloalkyi, optionally substituted C3-C 2 cycloalkenyl, optionally substituted C2-C12 heterocycloalkyi, optionally substituted C2-Ci2 heterocycloalkenyl, optionally substituted Ce-C1B aryl, optionally substituted CrC18 heteroaryl, optionally substituted C C12 alkyloxy, optionally substituted C2-C 2 alkenyloxy, optionally substituted C2-C12 alkynyloxy, optionally substituted C1-C10 heteroalkyloxy, optionally substituted C3-C12 cycloalkyloxy, optionally substituted C3-C12 cycloalkenyloxy, optionally substituted Ci-C12 heterocycloalkyloxy, optionally substituted Ci-C12 heterocycloalkenyloxy, optionally substituted Ce-C18 aryloxy, optionally substituted C^C^ heteroaryloxy, optionally substituted Ci-C12 alkylamino, SR14, S03H, S02NR15R18, S02R14, SONR15Rie, SOR14, COR14, COOH, COOR14, CONR15Rie, NR15COR14, NR 5COOR14, NR15S02R14, NR15CONR1BR17, NR15Rie, and acyl; or R2 and R3 may be fused to form a 5 or 6 membered cycloalkyi, heterocycloalkyi, aryl or heteroaryl ring- each of which may be optionally substituted;

R12, R13, R1S, R18, and R17 are each independently selected from the group consisting of: H, an N-protecting group, optionally substituted C C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted Ci-C10 heteroalkyl, optionally substituted C3-C12 cycloalkyi, optionally substituted C3-C12 cycloalkenyl, optionally substituted Ci-C12 heterocycloalkyi, optionally substituted C C12 heterocycloalkenyl, optionally substituted Ce-C1B aryl, and optionally substituted CrC18 heteroaryl; - R14 is selected from the group consisting of H, optionally substituted C^C^ alkyl, optionally substituted C2-C 2 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted Ci-C-,0 heteroalkyl, optionally substituted C3- C12 cycloalkyl, optionally substituted C3-C12cycloalkenyl, optionally substituted C C12 heterocycloalkyl, optionally substituted Ci-C12 heterocycloalkenyl, optionally substituted Ce-C18 aryl, and optionally substituted d-C™ heteroaryl;

- m is an integer selected from the group consisting of 0, 1 , 2, 3, and 4;

- n is an integer selected from the group consisting of 1, 2, 3, 4, and 5; and

- m + n is an integer selected from the group consisting of 1 , 2, 3, 4, and 5.

2. A compound as claimed in claim 1, wherein R2 is -X3-R18 and R3 is -X4-R19, wherein:

- R18 and R19 are the same or different and are selected from the group consisting of: H, halogen, C1-10 alkyl, C3-C10 cyclokalkyl, C3-C10 cycloalkylmethyl, C3-C10 alkene, C3-C10 alkyne, aryl, C5-C20 alkaryl, fused C5- C20 aryl or alkaryl and a hydrocarbon chain containing a heterocyclic or fused ring, any of which may be optionally substituted;

- X3 and X4 are the same or different and are selected from the group consisting of: a bond CR20R21, O, NR22 and S;

- R20 and R21 are the same or different and are selected from the group consisting of: H, halogen, OH, N02, CN, NH2, optionally substituted d-Ci2 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted d-do heteroalkyl, optionally substituted C3- C12 cycloalkyl, optionally substituted C3-C12 cycloalkenyl, optionally substituted C2-C 2 heterocycloalkyl, optionally substituted C2-C12 heterocycloalkenyl, optionally substituted Ce-Cie aryl, optionally substituted d-de heteroaryl, optionally substituted -Ci2 alkyloxy, optionally substituted C2-C12 alkenyloxy, optionally substituted C2-C12 alkynyloxy, optionally substituted d-do heteroalkyloxy, optionally substituted C3-C12 cycloalkyloxy, optionally substituted C3-C12 cycloalkenyloxy, optionally substituted d-C½ heterocycloalkyloxy, optionally substituted d-C2 heterocycloalkenyloxy, optionally substituted Ce-C B aryloxy, optionally substituted Ci-C18 heteroaryloxy, optionally substituted d-Ci2 alkylamino, SR14, SO3H, S02NR15Rie, S02R14, SONR15R1B, SOR14, COR14, COOH, COOR 4, CONR15Rie, NR15COR14, NR15COOR14, NR1SS02R14, NR15CONR18R17, NR15R16, and acyl; and - R22 is selected from the group consisting of: H, an N-protecting group, optionally substituted Ci-C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted C,-C,0 heteroalkyl, optionally substituted C3-C12 cycloalkyi, optionally substituted C3-C12 cycloalkenyl, optionally substituted C C12 heterocycloalkyi, optionally substituted Ci-C12 heterocycloalkenyl, optionally substituted Ce-Ci8 aryl, and optionally substituted C C^ heteroaryl.

3. A compound as claimed in claim 2, wherein R18 and R19 are fused to form a 5 or 6 membered cycloalkyi, heterocycloalkyi, cycloalkenyl, heterocycloalkenyl, aryl or heteroaryl ring, each of which may be optionally substituted.

4. A compound as claimed in any one of claims 1 to 3, wherein Z -Z2 is C=C. 5. A compound as claimed in any one of claims 1 to 4, wherein D is selected from the group consisting of: phenyl, C3-C6 cycloalkyi, C2-C5 heterocycloalkyi, C2-C5 heteroaryl.

6. A compound as claimed in any one of claims 1 to 5, wherein Rs is selected from the group consisting of: COOR 4, COR14, CONR15R1B, NR15R18, S02R14 and

SONR15R16.

7. A compound as claimed in claim 6, wherein R5 is selected from the group r consisting of: COOH, CONH2, CONHCH3, CONHOH, NH2, S02CH3> S02NH2, SONHCH3, and SON(CH3)2.

8. A compound as claimed in any one of claims 1 to 7, wherein D is selected from the group consisting of phenyl and C2-C5 heteroaryl. 9. A compound as claimed in claim 8, wherein the R5 substituent is ortho to X2.

10. A compound as claimed in any one of claims 1 to 7, wherein D is selected from the group consisting of Cs-Ce cycloalkyi and C2-C5 heterocycloalkyi. 11. A compound as claimed in claim 10, wherein the Rs substituent is either geminal or vicinal to X2.

12. A compound as claimed in any one of claims 1 to 11, wherein R6 is selected from the group consisting of: H and halogen.

13. A compound as claimed in any one of claims 1 to 12, wherein X1 is selected from the group consisting of: C=0 and S02.

14. A compound as claimed in any one of claims 1 to 13, wherein X2 is NR13.

15. A compound as claimed in claim 14, wherein R 3 is selected from the group consisting of: H and optionally substituted C C12 alkyl.

16. A compound as claimed in any one of claims 1 to 15, wherein W is CR7. .

17. A compound as claimed in claim 16, wherein R7 is H.

18. A compound as claimed in any one of claims 1 to 17, wherein A is selected from the group consisting of: CH, CH2CH2, CH=CH, O, OC(O), OC(S), OCH2, C(0)0, C(S)0, SCH2, SC(O), SC(S), S, S02CH2, S02, C(0)S, C(0)NH, C(S)S, NH, NH(CO), N, N=CH, and NHCH2.

19. A compound as claimed in any one of claims 1 to 18, wherein R2 and R3 are each independently selected from the group consisting of: optionally substituted (- -C12 alkyloxy and optionally substituted C2-C12alkynyloxy. 20. A compound as claimed in claim 19, wherein the optionally substituted C^C^ alkyloxy is C C12fluoroalkyloxy.

21. A compound as claimed in claim 19, wherein the optionally substituted C2-C12 alkynyloxy is C^C^ alkynylalkyloxy.

22. A compound as claimed in any one of claims 1 to 21, wherein R1 is selected from the group consisting of: H and optionally substituted d-Ce alkoxy.

23. A compound as claimed in any one of claims 1 to 22, wherein R2 is selected from the group consisting of: H3CO, F2HCF2CO, F2HCO, F3CO, and CHCCH20.

24. A compound as claimed in any one of claims 1 to 23, wherein R3 is selected from the group consisting of: H3CO, F2HCF2CO, F2HCO, F3CO, and CHCCH20.

25. A compound as claimed in any one of claims 1 to 24, wherein R4 is selected from the group consisting of: H and optionally substituted C Ce alkoxy.

26. A compound as claimed in any one of claims 1 to 25, wherein R2 and R3 together are selected from the group consisting of: OCF2CF2CO, OCH2CH20, OCF20, and OCH20.

A compound selected from the group consisting of:

R = S02Me, S02NH2, SONHMe, SON ea R = NH2, CONH2, CONHMe, CONHOH

R = S0 Me, S02NH2l SONHMe, SONMe2 R = NH2l CONH2, CONHMe, CONHOH

or a pharmaceutically acceptable salt or prodrug thereof.

28. A pharmaceutical composition including a compound as claimed in any one of claims 1 to 27, and a pharmaceutically acceptable diluent, excipient or carrier.

29. A method of treating a disease or condition associated with fibrosis, the method including administering to a subject in need of such treatment a therapeutically effective amount of a compound as claimed in any one of claims 1 to 27.

30. A method as claimed in claim 29, wherein the disease or condition is selected from the group consisting of fibrotic skin disorders, lung disease, heart disease and kidney disease. 31. A method of treating a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease, the method including administering to a subject in need of such treatment a therapeutically effective amount of a compound as claimed in any one of claims 1 to 27. 32. A method of inhibiting fibrosis in a subject, the method including administering to the subject a therapeutically effective amount of a compound as claimed in any one of claims 1 to 27.

33. Use of a compound as claimed in any one of claims 1 to 27 to inhibit fibrosis.

34. Use of a compound as claimed in any one of claims 1 to 27 in the treatment of a disease or condition associated with fibrosis.

35. Use of a compound as claimed in any one of claims 1 to 27 in the treatment of a disease or condition characterised by inflammation and or a benign or malignant neoplastic disease.

36. Use of a compound as claimed in any one of claims 1 to 27 in the preparation of a medicament for treating a disease or condition associated with fibrosis.

37. Use of a compound as claimed in any one of claims 1 to 27 in the preparation of a medicament for treating a disease or condition characterised by inflammation and or a benign or malignant neoplastic disease. ' 38. A compound of Formula (II)

Formula (II) pharmaceutically acceptable salt or prodrug thereof, wherein:

- A is selected from the group consisting of: -(CR8R9)p-(Y)q-(C(O))r(CR10R11)8- and -(CReRV(C(O))r(Y)q-(CR10R1V, wherein Y is selected from the group consisting of: O, S, and NR12, each p and s are an integer independently selected from the group consisting of: 0, 1, and 2, each q and r are an integer independently selected from the group consisting of: 0 and 1 , and p + q + r + s is an integer selected from the group consisting of: 1, 2, and 3;

- X1 is selected from the group consisting of: C=0, CF2, and S02, P02; X2 is selected from the group consisting of: NR13 or (CH2)t wherein t is an integer selected from the group consisting of: 0 and 1 ;

O is selected from the group consisting of: a cycloalkyi, heterocycloalkyi, cycloalkenyl, heterocycloalkenyl, aryl or heteroaryl ring;

R\ Rz and R3, are each independently selected from the group consisting of: H, OH, N02, CN, NH2, optionally substituted CrC12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted CrC10 heteroalkyl, optionally substituted C3-C12 cycloalkyi, optionally substituted C3-C12 cycloalkenyl, optionally substituted C2-C 2 heterocycloalkyi, optionally substituted C2-C12 heterocycloalkenyl, optionally substituted Ce-C18 aryl, optionally substituted C ds heteroaryl, optionally substituted C Ci2 alkyloxy, optionally substituted C2-C12 alkenyloxy, optionally substituted C2-C12 alkynyloxy, optionally substituted C1-C10 heteroalkyloxy, optionally substituted C3-C 2 cycloalkyloxy, optionally substituted C3-C12 cycloalkenyloxy, optionally substituted C,-C 2 heterocycloalkyloxy, optionally substituted Ci-C12 heterocycloalkenyloxy, optionally substituted Ce-C1B aryloxy, optionally substituted heteroaryloxy, optionally substituted Ci-C12 alkylamino, SR14, S03H, S02NR 5Rie, S02R14, SONR 5R1e, SOR 4, COR14, COOH, COOR14, CONR15R18, NR15COR14, NR15COOR14, NR 5S02R14, NR15CONR18R17, NR15R e, and acyl; or R2 and R3 may be fused to form a 5 or 6 membered cycloalkyi, heterocycloalkyi, aryl or heteroaryl ring- each of which may be optionally substituted;

R4, R5, Re, R7, R8, R9, R10 and R11 are each independently selected from the group consisting of: H, halogen, OH, N02, CN, NH2, optionally substituted C C12 alkyl, optionally substituted C^C^ alkenyl, optionally substituted Cr C12 alkynyl, optionally substituted C|-C10 heteroalkyl, optionally substituted C3-Ci2 cycloalkyi, optionally substituted C3-C12 cycloalkenyl, optionally substituted C2-C12 heterocycloalkyi, optionally substituted C2-C12 heterocycloalkenyl, optionally substituted Ce-C18 aryl, optionally substituted C CIB heteroaryl, optionally substituted Ci-C12 alkyloxy, optionally substituted C2-C12 alkenyloxy, optionally substituted C2-C12 alkynyloxy, optionally substituted Ct-C 0 heteroalkyloxy, optionally substituted C3-C12 cycloalkyloxy, optionally substituted C3-C12 cycloalkenyloxy, optionally substituted C C 2 heterocycloalkyloxy, optionally substituted C Ci2 heterocycloalkenyloxy, optionally substituted Ce-C1B aryloxy, optionally substituted C^Cm heteroaryloxy! optionally substituted Ci-C12 alkylamino, SR14, S03H, S02NR15Rie, S02R14, SONR15Rie, SOR14, COR14, COOH, COOR14, CONR15Rie, NR15COR14, NR 5COOR14, NR15S02R14, NR15CONR16R17, NR15R16, and acyl;

- R12, R13, R15, Rie, and R17 are each independently selected from the group consisting of: H, a N-protecting group, optionally substituted C C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted CrC12 alkynyl, optionally substituted CrC 0 heteroalkyl, optionally substituted C3-C 2 cycloalkyi, optionally substituted C3-C12 cycloalkenyl, optionally substituted C1-C12 heterocycloalkyl, optionally substituted Ci-C12 heterocycloalkenyl, optionally substituted Ce-C18 aryl, and optionally substituted C^-C^ heteroaryl;

- R14 is selected from the group consisting of H, optionally substituted C^-C^ alkyl, optionally substituted C2-C 2 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted C C10 heteroalkyl, optionally substituted C3- C12 cycloalkyi, optionally substituted C3-C12cycloalkenyl, optionally substituted C C12 heterocycloalkyl, optionally substituted Ci-C12 heterocycloalkenyl, optionally substituted C6-C1e aryl, and optionally substituted CrC 8 heteroaryl;

- m is an integer selected from the group consisting of: 0, 1, 2, 3, and 4;

- n is an integer selected from the group consisting of: 1, 2, 3, 4, and 5; and

- m + n is an integer selected from the group consisting of: 1 , 2, 3, 4, and 5.

39. A compound as claimed in claim 38, wherein R1 is -X3-R18 and R2 is -X4-R19, wherein:

- R18 and R 9 are the same or different and are selected from the group consisting of: H, halogen, d-10 alkyl, C3-C10 cyclokalkyl, C3-C10 cycloalkylmethyl, C3-C10 alkene, C3-C 0 alkyne, aryl, C6-C alkaryl, fused Cs-C20 aryl or alkaryl and a hydrocarbon chain containing a heterocyclic or fused ring, any of which may be optionally substituted;

- X3 and X4 are the same or different and are selected from the group consisting of: a bond CR^R21, O, NR22, and S;

- R20 and R21 are the same or different and are selected from the group consisting of: H, halogen, OH, N02, CN, NH2| optionally substituted C C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted C C10 heteroalkyl, optionally substituted C3-C12 cycloalkyi, optionally substituted C3-C12 cycloalkenyl, optionally substituted C2-C 2 heterocycloalkyl, optionally substituted C2-C12 heterocycloalkenyl, optionally substituted C6-C18 aryl, optionally substituted C C18 heteroaryl, optionally substituted f- C^ alkyloxy, optionally substituted C2-C12 alkenyloxy, optionally substituted C2-Ci2 alkynyloxy, optionally substituted C C10 heteroalkyloxy, optionally substituted C3-C12 cycloalkyloxy, optionally substituted C3-C12 cycloalkenyloxy, optionally substituted Ci-C12 heterocycloalkyloxy, optionally substituted Ci-Ci2 heterocycloalkenyloxy, optionally, substituted Ce-C18 aryloxy, optionally substituted C C18 heteroaryloxy, optionally substituted C -Cn alkylamino, SR14, S03H, S02NR15Rie, S02R14, SONR15Rie, SOR14, COR14, COOH, COOR14, CONR15Rie, NR15COR14, NR1sCOOR14, NR16S02R14, NR 5CONR eR17, NR15R18, and acyl; and

- R22 is selected from the group consisting of: H, an N-protecting group, optionally substituted C1-C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted Ci-Cw heteroalkyl, optionally substituted C3-C12 cycloalkyl, optionally substituted C3-C12 cycloalkenyl, optionally substituted C Ci2 heterocycloalkyl, optionally substituted Ci-C12 heterocycloalkenyl, optionally substituted Ce-C18 aryl, and optionally substituted C1-C18 heteroaryl.

40. A compound as claimed in claim 38, wherein R2 is -X3-R1B and R3 is -X4-Rie, wherein:

- R1B and R 9 are the same or different and are selected from the group consisting of: H, halogen, Ci-10 alkyl, C3-C10 cyclokalkyl, C3-C10 cycloalkylmethyl, C3-C10alkene, C3-C 0 alkyne, aryl, C5-C20 alkaryl, fused C5- C20 aryl or alkaryl and a hydrocarbon chain containing a heterocyclic or fused ring, any of which may be optionally substituted;

- X3 and X4 are the same or different and are selected from the group consisting of: a bond CR20R21, O, NR22, and S;

- R20 and R21 are the same or different and are selected from the group consisting of: H, halogen, OH, N02, CN, NH2, optionally substituted Ci-C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted C C10 heteroalkyl, optionally substituted C3- C 2 cycloalkyl, optionally substituted C3-C 2 cycloalkenyl, optionally substituted Cr-C12 heterocycloalkyl, optionally substituted C2-C12 heterocycloalkenyl, optionally substituted Ce-C 8 aryl, optionally substituted Ci-C18 heteroaryl, optionally substituted C,-d2 alkyloxy, optionally substituted C2-C12 alkenyloxy, optionally substituted C2-C12 alkynyloxy, optionally substituted C1-C10 heteroalkyloxy, optionally substituted C3-C12 cycloalkyloxy, optionally substituted C3-C12 cycloalkenyloxy, optionally substituted C C12 heterocycloalkyloxy, optionally substituted C1-C12 heterocycloalkenyloxy, optionally substituted Ce-C18 aryloxy, optionally substituted CrC18 heteroaryloxy, optionally substituted Ci-C12 alkylamino, SRU, SO3H, S02NR15Rie, S02R14, SONR15Rie, SOR14, COR14, COOH,

COOR14, CONR15Rie, NR 5COR14, NR 5COOR14, NR15S02R14, NR15CONRieR17, NR SR16, and acyl; and

• - R22 is selected from the group consisting of: H, an N-protecting group, optionally substituted C1-C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted C -C,0 heteroalkyl, optionally substituted C3-C12 cycloalkyi, optionally substituted C3-C12 cycloalkenyl, optionally substituted Ci-Ci2 heterocycloalkyi, optionally substituted C^-C^ heterocycloalkenyl, optionally substituted Ce-C1ftaryl, and optionally substituted CrC1B heteroaryl.

41. A compound as claimed in either claim 39 or claim 40, wherein R 8 and R19 are fused to form a 5 or 6 membered cycloalkyi, heterocycloalkyi, aryl or heteroaryl ring, each of which may be optionally substituted. 42. A compound as claimed in any one of claims 38 to 41 , wherein D is selected from the group consisting of: phenyl, C3-Ce cycloalkyi, C2-C5 heteroaryl.

43. A compound as claimed in any one of claims 38 to 42, wherein Rs is selected from the group consisting of: H, CN, N02l and C^-Cealkyl.

44. A compound as claimed any one of claims 38 to 43, wherein Re is selected from the group consisting of: COOR14, COR14, CONR16Rie, NR15Rie, and S02R14 and SONR15Rie. 45. A compound as claimed in claim 44, wherein R6 is selected from the group consisting of: COOH, CONH2l CONHOH, CONHCH3, NH2, S02CH3, S02NH2, SONHCHs, and SON(CH3)2.

46. A compound as claimed in any one of claims 38 to 45, wherein D is selected from the group consisting of phenyl and C^Cs heteroaryl.

47. A compound as claimed in claim 46, wherein the R8 substituerit is ortho to X2.

48. A compound as claimed in any one of claims 38 to 45, wherein D is selected from the group consisting of C3-Ce cycloalkyl and C2-C5 heterocycloalkyl. 49. A compound as claimed in claim 48, wherein the Rs substituent is either geminal or vicinal to X2.

50. A compound as claimed in any one of claims 38 to 49, wherein R7 is selected from the group consisting of H and halogen.

51. A compound as claimed in any one of claims 38 to 49, wherein X1 is selected from the group consisting of: C=0 and S02.

52. A compound as claimed in any one of claims 38 to 51 , wherein X2 is NR13.

53. A compound as claimed in claim 52, wherein R13 is selected from the group consisting of: H and optionally substituted C1-C12 alkyl.

54. A compound as claimed in any one of claims 38 to 53, wherein A is selected from the group consisting of: CH2CH2> CH=CH, C(0)0, C(0)S, C(0)NR12, OC(O),

SC(O), NR12C(0), CH2CH2CH2, CH=CHCH2, CH2CH=CH, OCH2CH2l SCH2CH2, NR12CH2CH2, CH2CH20, CH2CH2S, CH2CH2NR12, C(0)OCH2, C(0)SCH2, C(0)NR12CH2, CH2OC(0), CH2SC(0), and GH2NR12C(0). 55. A compound as claimed in any one of claims 38 to 54, wherein R1, R2 and R3 are each independently selected from the group consisting of: optionally substituted d- C12 alkyloxy and optionally substituted C2-C12 alkynyloxy.

56. A compound as claimed in claim 55, wherein the optionally substituted Ci-C12 alkyloxy is Ci-C12fluoroalkyloxy.

57. A compound as claimed in claim 55, wherein the optionally substituted C2-C12 alkynyloxy is C2-C 2 alkynylalkyloxy. 58. A compound as claimed in any one of claims 38 to 57, wherein R1 is selected from the group consisting of: H3CO, F2HCF2CO, F2HCO, F3CO, and CHCCH20.

59. A compound as claimed in any one of claims 38 to 58, wherein R2 is selected from the group consisting of: H3CO, F2HCF2CO, F2HCO, F3CO, and CHCCH20.

60. A compound as claimed in any one of claims 38 to 59, wherein R3 is selected from the group consisting of: H3CO, F2HCF2CO, F2HCO, F3CO, and CHCCH20.

61. A compound as claimed in any one of claims 38 to 60, wherein R1 and R2 together are selected from the group consisting of: OCF2CF2CO, OCH2CH20, OCF20, and OCH20.

62. A compound as claimed in any one of claims 38 to 60, wherein R2 and R3 together are selected from the group consisting of: OCF2CF2CO, OCH2CH20, OCF20, and OCH20. 63. A compound selected from the group consisting of:

or a pharmaceutically acceptable salt or prodrug thereof.

64. A pharmaceutical composition including a compound as claimed in any one of claims 38 to 63, and a pharmaceutically acceptable diluent, excipient or carrier.

65. A method of treating a disease or condition associated with fibrosis, the method including administering to a subject in need of such treatment a therapeutically effective amount of a compound as claimed in any one of claims 38 to 63.

66. A method as claimed in claim 65, wherein the disease or condition is selected from the group consisting of fibrotic skin disorders, lung disease, heart disease and kidney disease. 67. A method of treating a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease, the method including administering to a subject in need of such treatment a therapeutically effective amount of a compound as claimed in any one of claims 38 to 63. 68. A method of inhibiting fibrosis in a subject, the method including administering to the subject a therapeutically effective amount of a compound as claimed in any one of claims 38 to 63.

69. Use of a compound as claimed in any one of claims 38 to 63 to inhibit fibrosis.

70. Use of a compound as claimed in any one of claims 38 to 63 in the treatment of a disease or condition associated with fibrosis.

71. Use of a compound as claimed in any one of claims 38 to 63 in the treatment of a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease.

72. Use of a compound as claimed in any one of claims 38 to 63 in the preparation of a medicament for treating a disease or condition associated with fibrosis.

73. Use of a compound as claimed in any one of claims 38 to 63 in the preparation of a medicament for treating a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease. ,

74. A compound of Formula (III)

Formula (III) or a pharmaceutically acceptable salt or prodrug thereof, wherein:

- A is selected from the group consisting of: -(CR9 10)P-(Y)q-(C(O))r (CR11R12)8- and -(CReR10)p-(C(O))r(Y)q-(CR11R12)8-, wherein Y is selected from the group consisting of: O, S, NR13, each p and s are an integer independently selected from the group consisting of: 0, 1, and 2, each q and r are an integer independently selected from the group consisting of: 0 and 1, and p + q + r + s is an integer selected from the group consisting of: 1, 2, and 3;

- T is selected from the group consisting of: a single bond, a double bond, a

triple bond and

X1 is selected from the group consisting of: C=0, CF2 or S02, P02;

X2 is selected from the group consisting of: CR17 and N;

Z\ Z2, Z3, and Z4 are each independently selected from the group consisting of CR8 and N; R\ R4, R6, R8, R9, R10, R1 , R12, R1S and R18 are each independently selected from the group consisting of: H, halogen, OH, N02, CN, NH2, optionally substituted C C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted C,-Ci0 heteroalkyl, optionally substituted C3-C12 cycloalkyi, optionally substituted C3-C 2cycloalkenyl, optionally substituted C2-C12 heterocycloalkyi, optionally substituted CrCi2 heterocycloalkenyl, optionally substituted Ce-Ci8 aryl, optionally substituted d-C™ heteroaryl, optionally substituted C C12 alkyloxy, optionally substituted C2-C12 alkenyloxy, optionally substituted (_¼- C12 alkynyloxy, optionally substituted 'Ci-C 0 heteroalkyloxy, optionally substituted C3-C12 cycloalkyloxy, optionally substituted C3-C12 cycloalkenyloxy, optionally substituted d-C^ heterocycloalkyloxy, optionally substituted d-C^ heterocycloalkenyloxy, optionally substituted Ce-C1B aryloxy, optionally substituted C C^ heteroaryloxy, optionally substituted Ci-C12 alkylamino, SR18, S03H, S02NRieR2°, S02R18, SONR19R20, SOR18, COR18, COOH, COOR18, CONRieR20, NR19COR1B, NR19COOR18, NR19S02R18, NR19CONR20R21, NR19R20, and acyl;

R2 and R3, are each independently selected from the group consisting of: H, OH, N02, CN, NH2, optionally substituted d-C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C-C12 alkynyl, optionally substituted d-C10 heteroalkyl, optionally substituted C3-C 2 cycloalkyi, optionally substituted C3-C12 cycloalkenyl, optionally substituted C2-C12 heterocycloalkyi, optionally substituted C2-C 2 heterocycloalkenyl, optionally substituted Ce-C18 aryl, optionally substituted Ci-CiB heteroaryl, optionally substituted C C 2 alkyloxy, optionally substituted C2-C12 alkenyloxy, optionally substituted C2-C12 alkynyloxy, optionally substituted C C 0 heteroalkyloxy, optionally substituted C3-C 2 cycloalkyloxy, optionally substituted C3-C 2 cycloalkenyloxy, optionally substituted Ci-C 2 heterocycloalkyloxy, optionally substituted d-C^ heterocycloalkenyloxy, optionally substituted Ce-C18 aryloxy, optionally substituted Ci-C 8 heteroaryloxy, optionally substituted C C12 alkylamino, SR18, SO3H, S02R18, SONR19R20, SOR18, COR18, COOH, COOR18, CONR19R20, NR19COR18, NRieCOOR18, NR19S02R18, NR19CONR20R21, NR19R20, and acyl; or R2 and R3 may be fused to form a 5 or 6 membered cycloalkyi, heterocycloalkyi, aryl or heteroaryl ring, each of which may be optionally substituted; and R7 are present when T is a single bond, a double bond

but not when T is a triple bond, each Re and R7 being independently selected from the group consisting of: H, N02, CN, optionally substituted Ci-C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted C^-do heteroalkyl, optionally substituted C3-C12 cycloalkyl, optionally substituted C3-C12cycloalkenyl, optionally substituted C2-C12 heterocycloalkyl, optionally substituted C2-Ci2 heterocycloalkenyl, optionally substituted Ce-Ci8 aryl, optionaljy substituted C C1e heteroaryl, optionally substituted Ci-C12 alkyloxy, optionally substituted C2-C12 alkenyloxy, optionally substituted C2- C 2 alkynyloxy, optionally substituted d-Cio heteroalkyloxy, optionally substituted C3-C12 cycloalkyloxy, optionally substituted C3-C12 cycloalkenyloxy, optionally substituted Ci-Ci2 heterocycloalkyloxy, optionally substituted Ci-C 2 heterocycloalkenyloxy, optionally substituted C6-C18 aryloxy, optionally substituted Ci-Cie heteroaryloxy, optionally substituted d-Ci2 alkylamino, SR18, S03H, S02NR 9R20, S02R18, SONR^R20, SOR18, COR18, COOH, COOR18, CONR^R20, NR19COR18, NR19COOR18, NR19S02R18, NR19CONR20R21, NR^R20, and acyl;

R13, R19, R20 and R21 are each independently selected from the group consisting of: H: a N-protecting group, optionally substituted Ci-Ci2 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted C Ci0 heteroalkyl, optionally substituted C3-C12 cycloalkyl, optionally substituted C3-C12 cycloalkenyl, optionally substituted C C12 heterocycloalkyl, optionally substituted C C12 heterocycloalkenyl, optionally substituted C8-C18 aryl, and optionally substituted d-C™ heteroaryl;

R 8 is selected from the group consisting of H, optionally substituted C C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted CrC 2 alkynyl, optionally substituted Ci-C10 heteroalkyl, optionally substituted d- Ci2 cycloalkyl, optionally substituted C3-C12cycloalkenyl, optionally substituted Ci-C12 heterocycloalkyl, optionally substituted Ci-C12 heterocycloalkenyl, optionally substituted Ce-C18 aryl, and optionally substituted C^de heteroaryl; and

n is an integer selected from the group consisting of: 0, 1, 2, 3, and 4. 75; A compound as claimed in claim 74, wherein R2 is -X'-R22 and R3 is -X^R23, wherein:

- R22 and R23 are the same or different and are selected from the group consisting of: H, halogen, Ci-10 alkyl, C3-Ci0 cyclokalkyl, C3-Ci0 cycloalkylmethyl, C3-Ci0alkene, C3 C10 alkyne, aryl, C5-C2o alkaryl, fused Cs- C20 aryl or alkaryl and a hydrocarbon chain containing a heterocyclic or fused ring, any of which may be optionally substituted;

- X3 and X4 are the same or different and are selected from the group consisting of: a bond CR24R25,0, NR26, and S;

- R24 and R25 are the same or different and are selected from the group consisting of: H, halogen, OH, N02, CN, NH2> optionally substituted C,-C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C;>-C 2 alkynyl, optionally substituted C C10 heteroalkyl, optionally substituted C3- C12 cycloalkyl, optionally substituted C3-Ci2 cycloalkenyl, optionally substituted Cj Ci2 heterocycloalkyl, optionally substituted C2-C 2 heterocycloalkenyl, optionally substituted Ce-C18 aryl, optionally substituted Ci-de heteroaryl, optionally substituted C Ci2 alkyloxy, optionally substituted CrC12 alkenyloxy, optionally substituted G2-C12 aikynyloxy, optionally substituted Ci-C10 heteroalkyloxy, optionally substituted C3-C12 cycloalkyloxy, optionally substituted C3-Ci2 cycloalkenyloxy, optionally substituted Ci-C 2~ heterocycloalkyloxy, optionally substituted C -C12 heterocycloalkenyloxy, optionally substituted Ce-C18 aryloxy, optionally substituted C C 8 heteroaryloxy, optionally substituted C C12 alkylamino, SR 4, S03H, S02NR15R e, S02R14, SONR15R16, SOR14, COR14, COOH, COOR14, CONR1sRie, NR15COR14, NR15COOR14, NR15S02R14, NR15C0NR1eR17, NR 5R18, and acyl; and

- R26 is selected from the group consisting of: H, an N-protecting group, optionally substituted C C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted CrC 0 heteroalkyl, optionally substituted C3-C12 cycloalkyl, optionally substituted C3-C12 cycloalkenyl, optionally substituted C C12 heterocycloalkyl, optionally substituted Ci-C12 heterocycloalkenyl, optionally substituted Ce-Cie aryl, and optionally substituted CrC18 heteroaryl.

76. A compound as claimed in claim 75, wherein R22 and R23 are fused to form a 5 or 6 membered cycloalkyi, cycloalKenyl, heterocycloalkyi, heterocycloatkenyl, aryl or heteroaryl ring, each of which may be optionally substituted. 77. A compound as claimed in any one of claims 74 to 76, wherein X1 is selected from the group consisting of: C=0 and S02.

78. A compound as claimed in any one of claims 74 to 77, wherein X2 is N. 79. A compound as claimed in any one of claims 74 to 78, wherein A is selected from the group consisting of: CH2CH2CH2, CH2CH20, CH2CH2S, CH2CH2NR13, CH2CH=CH, CH=CHCH2> CH=CHC(0), C(0)CH=CH, C=NC(0), CH2OC(0), C(0)OC(0), CH2SC(0), C(0)SC(0), C(0)OCH2, C(0)SCH2l C(0)CH2NR13, C(0)CH2S, C(O)CH20, C(S)CH20, OC(O), CH20, C(0)0, CHzS, CH2NR13, CH2CH2, SC(O), C(S)0, C(0)S, C(S)S, C(0)NR13, C(S)NR13.

80. A compound as claimed in any one of claims 74 to 79, wherein R2 and R3 are each independently selected from the group consisting of: optionally substituted C^C^ alkyloxy and optionally substituted C2-Ci2 alkynyloxy.

81. A compound as claimed in claim 80, wherein the optionally substituted Cr-C12 alkyloxy is Ci-C12fluoroalkyloxy.

82. A compound as claimed in claim 80, wherein the optionally substituted C^C^ alkynyloxy is CrC12 alkynylalkyloxy.

83. A compound as claimed in any one of claims 74 to 82, wherein R2 is selected from the group consisting of: H3CO, F2HCF2CO, F2HCO, F3CO, and CHCCH20.

84. A compound as claimed in any one of claims 74 to 83, wherein R3 is selected from the group consisting of: H3CO, F2HCF2CO, F2HCO, F3CO, and CHCCH20.

85. A compound as claimed in any one of claims 74 to 84, wherein R2 and R3 together are selected from the group consisting of: OCF2CF2CO, OCH2CH20, OCF20, and OCH20. A compound as claimed in any one of claims 74 to 85, wherein T is selected

from the group consisting of: a double bond and

87. A compound as claimed in any one of claims 74 to 86, wherein Rs is selected from the group consisting of: H and Ci-Cealkyl.

88. A compound as claimed in any one of claims 74 to 87, wherein R7 is selected from the group consisting of: H, CN, N02 and d-C6alkyl.

89. A compound as claimed in any one of claims 74 to 88, wherein R1 is selected from the group consisting of: H and optionally substituted Ci-C6 alkoxy.

90. A compound as claimed in any one of claims 74 to 89, wherein R4 is selected from the group consisting of: H and optionally substituted Ci-CB alkoxy.

91. A compound as claimed in any one of claims 74 to 90, wherein R5 is H.

92. A compound as claimed in any one of claims 74 to 91 , wherein R8 is selected from the group consisting of: H. halogen, COOR18, COR18, CONR^R20, R19R20, S02R1B, and SONR^R20.

93. A compound as claimed in claim 92, wherein R8 is selected from the group consisting of: H, halogen, COOH, CONH2, CONHCH3, CONHOH, NH2l S02CH3, S02NH2l SONHCH3, and SON(CH3)2.

A compound as claimed in either claim 92 or claim 93, wherein n is 1.

95. A compound selected from the group consisting of: 136

' or a pharmaceutically acceptable salt or prodrug thereof.

96. A pharmaceutical composition including a compound as claimed in any one of claims 74 to 95, and a pharmaceutically acceptable diluent, excipient or carrier.

97. A method of treating a disease or condition associated with fibrosis, the method including, administering to a subject in need of such treatment a therapeutically effective amount of a compound as claimed in any one of claims 74 to 95.

98. A method as claimed in claim 97, wherein the disease or condition is selected from the group consisting of fibrotic skin disorders, lung disease, heart disease and kidney disease. 99. A method of treating a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease, the method including administering to a subject in need of such treatment a therapeutically effective amount of a compound as claimed in any one of claims 74 to 95. 100. A method of inhibiting fibrosis in a subject, the method including administering to the subject a therapeutically effective amount of a compound as claimed in any one of claims 74 to 95.

101. Use of a compound as claimed in any one of claims 74 to 95 to inhibit fibrosis.

102. Use of a compound as claimed in any one of claims 74 to 95 in the treatment of a disease or condition associated with fibrosis.

103. Use of a compound as claimed in any one of claims 74 to 95 in the treatment of a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease.

104. Use of a compound as claimed in any one of claims 74 to 95 in the preparation of a medicament for treating a disease or condition associated with fibrosis.

105. Use of a compound as claimed in any one of claims 74 to 95 in the preparation of a medicament for treating a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease.

A compound of Formula (IV)

Formula (IV)

c

or a pharmaceutically acceptable salt or prodrug thereof, wherein: ' - Z-X-Y is selected from the group consisting of: N=C-N, N-C=N, 0-C=N, S-

C=N, N=C-0, N=C-S, C=C-NH, OC-O, C=C-S, and C(0)-C=N;

- A is selected from the group consisting of: a bond, S02l C, C=S, C=0, C=NR9, and NR9;

- T is selected from the group consisting of: a single bond, a double bond, a

triple bond, ; Z1, Z2, Z3, and Z4 are each independently selected from the group consisting of CR8 and ;

R\ R4, R5, R8, R10, and R1 are each independently selected from the group consisting of: H, halogen, OH, N02, CN, NH2, optionally substituted Ci-C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted C^Cm heteroalkyl, optionally substituted C3- C12 cycloalkyl, optionally substituted C3-C12 cycloalkenyl, optionally substituted C^C^ heterocycloalkyl, optionally substituted Cr-C12 heterocycloalkenyl, optionally substituted Ce-C18 aryl, optionally substituted Ci-C18 heteroaryl, optionally substituted C C12 alkyloxy, optionally substituted C2-C12 alkenyloxy, optionally substituted C2-C12 alkynyloxy, optionally substituted CrC,0 heteroalkyloxy, optionally substituted C3-C 2 cycloalkyloxy, optionally substituted C3-C 2 cycloalkenyloxy, optionally substituted C Ci2 heterocycloalkyloxy, optionally substituted Ci-C2 heterocycloalkenyloxy, optionally substituted Ce-C18 aryloxy, optionally substituted Ci-C18 heteroaryloxy, optionally substituted Ci-C12 alkylamino, SR12, S03H, S02NR13R14, S02R12, SONR 3R14, SOR12, COR12, COOH, COOR12, CONR13R14, NR13COR12, NR13COOR12, NR13S02R12, NR13CONR14R15, NR14R1S, and acyl;

R2 and R3, are each independently selected from the group consisting of; H, OH, N02, CN, NH2, optionally substituted C C12 alkyl, optionally substituted C2-Ci2 alkenyl, optionally substituted C2-Ci2 alkynyl, optionally substituted d-C10 heteroalkyl, optionally substituted C3-C12 cycloalkyl, optionally substituted C3-C12 cycloalkenyl, optionally substituted C2-d2 heterocycloalkyl, optionally substituted C2-C12 heterocycloalkenyl, optionally substituted Ce-Ci8 aryl, optionally substituted heteroaryl, optionally substituted C C12 alkyloxy, optionally substituted C2-C12 alkenyloxy, optionally substituted CrC12 alkynyloxy, optionally substituted C C10 heteroalkyloxy, optionally substituted C3-C12 cycloalkyloxy, optionally substituted C3-C12 cycloalkenyloxy, optionally substituted C C12 heterocycloalkyloxy, optionally substituted C,-C,2 heterocycloalkenyloxy, optionally substituted C8-C18 aryloxy, optionally substituted C-C18 heteroaryloxy, optionally substituted C C12 alkylamino, SR12, SO3H, S02NR13R14, S02R12, SONR 3R14, SOR12, COR12, COOH, COOR12, CONR13R14, NR13COR12, NR13COOR12, NR 3S02R12, NR13CONR14R15, NR14R15, and acyl; or R2 and R3 may be fused to form a 5 or 6 membered cycloalkyi, heterocycloalkyi, aryl or heteroaryl ring' each of which may be optionally substituted;

Re and R7 are present when T is a single bond, a double bond or

but not when T is a triple bond, each R6 and R7 being independently selected from the group consisting of: H, N02, CN, optionally substituted d-C 2 alkyl, optionally substituted C2-Ci2 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted d-do heteroalkyl, optionally substituted C3-C 2 cycloalkyi, optionally substituted C3-C12 cycloalkenyl, optionally substituted C2-C12 heterocycloalkyi, optionally substituted C2-C12 heterocycloalkenyl, optionally substituted Ce-Ci8 · aryl, optionally substituted d-Ci8 heteroaryl, optionally substituted Ci-Ci2 alkyloxy, optionally substituted C2-C12 alkenyloxy, optionally substituted Cy C12 alkynyloxy, optionally substituted d-C10 heteroalkyloxy, optionally substituted C3-C12 cycloalkyloxy, optionally substituted C3-C cycloalkenyloxy, optionally substituted C,-Ci2 heterocycloalkyioxy, optionally substituted Ci-Ci2 heterocycloalkenyloxy, optionally substituted Ce-Ci8 aryloxy, optionally substituted d-C1e heteroaryloxy, optionally substituted C,-C12 alkylamino, SR12, S03H, S02NR13R14, S02R12, SONR13R14, SOR12, COR12, COOH, COOR12, CONR13R14, NR13COR12, NR13COOR12, NR13S02R12, NR13CONR14R15, NR14R15, and acyl;

R9, R13, R14, and R15 are each independently selected from the group consisting of: H, an N-protecting group, optionally substituted C,-C12 alkyl, optionally substituted d-C^ alkenyl, optionally substituted d-djj alkynyl, optionally substituted Ci-C10 heteroalkyl, optionally substituted C3-C 2 cycloalkyi, optionally substituted C3-C12 cycloalkenyl, optionally substituted C,-C12 heterocycloalkyi, optionally substituted d-C12 heterocycloalkenyl, optionally substituted Ce-C1B aryl, and optionally substituted C C18 heteroaryl;

R12 is selected from the group consisting of H, optionally substituted Ci-Ci2 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted C,-C10 heteroalkyl, optionally substituted C3- d2 cycloalkyi, optionally substituted C3-C12cycloalkenyl, optionally substituted C Ci2 heterocycloalkyi, optionally substituted d-C12 heterocycloalkenyl, optionally substituted Ce-C18 aryl, and optionally substituted Ci-C 8 heteroaryl; and

- n is an integer selected from the group consisting of: 0, 1, 2, 3, and 4.

107. A compound as claimed in claim 106, wherein R2 is -X2-R18 and R3 is -X3-R17, wherein:

- R18 and R17 are the same or different and are selected from the group consisting of: H, halogen, Ci- 0 alkyl, C3-C10 cyclokalkyl, C3-C10 cycloalkylmethyl, C3-C10alkene, C3-Ci0 alkyne, aryl, C5-C20 alkaryl, fused C5- C20 aryl or alkaryl and a hydrocarbon chain containing a heterocyclic or fused ring, any of which may be optionally substituted;

- X2 and X3 are the same or different and are selected from the group consisting of: a bond CR1BR19, O, NR20, and S;

- R18 and R18 are the same or different and are selected from the group consisting of: H, halogen, OH, N02, CN, NH2, optionally substituted Ci-C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C2-C12 alkynyl, optionally substituted C^do heteroalkyl. optionally substituted C3- C12 cycloalkyl, optionally substituted C3-C 2 cycloalkenyl, optionally substituted C2-C 2 heterocycloalkyl, optionally substituted Cr-C12 heterocycloalkenyl, optionally substituted Ce-C18 aryl, optionally substituted C de heteroaryl, optionally substituted Ci_Ci2 alkyloxy, optionally substituted C2-C12 alkenyloxy, optionally substituted C2-C12 alkynyloxy, optionally substituted C C10 heteroalkyloxy, optionally substituted C3-C 2 cycloalkyloxy, optionally substituted C3-C12 cycloalkenyloxy, optionally substituted C C 2 heterocycloalkyloxy, optionally substituted d-C^ heterocycloalkenyloxy, optionally substituted C8-C18 aryloxy, optionally substituted Ci-C1B heteroaryloxy, optionally substituted C1-C12 alkylamino, SR12, SO3H, S02NR13R14, S02R12, SONR13R14, SOR12, COR12, COOH, COOR12, CONR13R14, NR13COR12, NR13COOR12, NR13S02R12, NR13CONR1 R15, NR1 R15, and acyl; and

- R20 is selected from the group consisting of: H, an N-protecting group, optionally substituted C C12 alkyl, optionally substituted C2-C12 alkenyl, optionally substituted C^C^ alkynyl, optionally substituted Ci-C 0 heteroalkyl, optionally substituted C3-C12 cycloalkyl, optionally substituted C3-Ci2 cycloalkenyl, optionally substituted Ci-Ci2 heterocycloalkyl, optionally substituted Ci-C12 heterocycloalkenyl, optionally substituted Cg-Cie aryl, and optionally substituted Ci-C1B heteroaryl.

108. A compound as claimed in claim 107, wherein R18 and R 7 are fused to form a 5 or 6 membered cycloalkyi, heterocycloalkyi, cycloalkenyl, heterocycioalkenyl, aryl or heteroaryl ring, each of which may be optionally substituted.

109. A compound as claimed in any one of claims 106 to 108, wherein R2 and R3 are each independently selected from the group consisting of: optionally substituted Ci-C 2 alkyloxy and optionally substituted C2-C12alkynyloxy. 110. A compound as claimed in claim 109, wherein the optionally substituted Cr-C12 alkyloxy is CrC 2fluoroalkyloxy.

111. A compound as claimed in claim 109, wherein the optionally substituted C2-C 2 alkynyloxy is alkynylalkyloxy.

112. A compound as claimed in claim 109, wherein R2 is selected from the group consisting of: H3CO, F2HCF2CO, F2HCO, F3CO, and CHCCH20.

1 3. A compound as claimed in claim 109, wherein R3 is selected from the group consisting of: H3CO, F2HCF2CO, F2HCO, F3CO, and CHCCH20.

114. A compound as claimed in claim 108, wherein R2 and R3 together are selected from the group consisting of: OCF2CF2CO, OCH2CH20, OCF20, and OCH20.

115. A compound as claimed in any one of claims 106 to 114, wherein T is selected

from the group consisting of: a double bond and

116. A compound as claimed in any one of claims 106 to 115, wherein Re is selected from the group consisting of: H and C^Cealkyl.

117. A compound as claimed in any one of claims 106 to 116, wherein R7 is selected from the group consisting of: H, CN, N02l and CrC6alkyl.

118. A compound as claimed in any one of claims 106 to 117, wherein R1 is selected from the group consisting of: H and optionally substituted C^-Ce alkoxy.

119. A compound as claimed in any one of claims 106 to 118, wherein R4 is selected from the group consisting of: H and optionally substituted C-i-Ce alkoxy.

120. A compound as claimed in any one of claims 106 to 119, wherein Rs is H.

121. A compound as claimed in any one of claims 106 to 120, wherein R8 is selected from the group consisting of: H. halogen, COOR12, COR12, CONR13R14, NR13R14,

S02R12, and SONR13R14.

122. A compound as claimed in claim 121, wherein RB is selected from the group consisting of: H, halogen, COOH, CONH2, CONHOH, CONHCH,, NH2, S02CH3, S02NH2, SONHCH3, and SON(CH3)2.

123. A compound as claimed in either claim 121 or claim 122, wherein n is 1.

124. A compound as claimed in claim 123, wherein the R8 group is in the position ortho to the Y group.

125. A compound selected from the group consisting of:

or a pharmaceutically acceptable salt or prodrug thereof.

126. A pharmaceutical composition including a compound as claimed in any one of claims 106 to 125, and a pharmaceutically acceptable diluent, excipient or carrier.

127. A method of treating a disease or condition associated with fibrosis, the method including administering to a subject in need of such treatment a therapeutically effective amount of a compound as claimed in any one of claims 106 to 125. 128. A method as claimed in claim 127, wherein the disease or condition is selected from the group consisting of fibrotic skin disorders, lung disease, heart disease and kidney disease.

129. A method of treating a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease, the method including administering to a subject in need of such treatment a therapeutically effective amount of a compound as claimed in any one of claims 106 to 125.

130. A method of inhibiting fibrosis in a subject, the method including administering to the subject a therapeutically effective amount of a compound as claimed in any one of claims 106 to 125.

131. Use of a compound as claimed in any one of claims 106 to 125 to inhibit \ fibrosis.

132. Use of a compound as claimed in any one of claims 106 to 125 in the treatment of a disease or condition associated with fibrosis. 33. Use of a compound as claimed in any one of claims 106 to 125 in the treatment of a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease.

134. Use of a compound as claimed in any one of claims 106 to 125 in the preparation of a medicament for treating a disease or condition associated with fibrosis.

135. Use of a compound as claimed in any one of claims 106 to 125 in the preparation of a medicament for treating a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease.

Description:
FUSED RING ANALOGUES OF ANTI-FIBROTIQ AGENTS Field of the Invention The present invention relates to compounds that may be useful in the treatment of medical conditions associated with tissue fibrosis. More particularly, the present invention relates to fused ring derivatives of 2-{[(2£)-3-(3,4-dimethoxyphenyl)prop-2- enoyl]amino}benzoic acid, pharmaceutical compositions containing these derivative compounds, and uses of the derivative compounds in the treatment of certain conditions associated with tissue fibrosis.

Background of the Invention

Fibrosis occurs when excess fibrous connective tissue forms or develops in an organ or tissue. Fibrosis can occur as a part of the wound-healing process following tissue damage resulting from physical injury, inflammation, infection, exposure to toxins, and other causes. Examples of conditions associated with tissue fibrosis include dermal scar formation, keloids, liver fibrosis, kidney fibrosis (including diabetic nephropathy), hepatic cirrhosis, pulmonary interstitial fibrosis, glomerulonephritis, heart failure (ischaemic and non-ischaemic), diabetic nephropathy, scleroderma, excessive scar tissue post surgery or device insertion, progressive kidney disease, hypertension, heart failure due to ischaemic heart disease, valvular heart disease, hypertensive heart disease, and hypertrophic scars. The elaboration of extracellular matrix also has a role in fibroproliferative tumor progression and metastasis. Accordingly, strategies that reduce the accumulation of extracellular matrix have been advocated as potential therapies for the treatment and prevention of heart failure in both diabetic and nondiabetic states. At present the pathogenic mechanism of fibrosis is not completely understood. In general, the proliferation and function of fibroblasts are closely controlled in normal conditions. However, in pathological states in which inflammation or tissue injury is serious or sustained, the tissue repair mechanism goes into overdrive and the control mechanism is abrogated. Excessive tissue repair is caused by over-production of connective tissue protein probably due to abnormal proliferation of fibroblasts and extracellular matrix dysbolism. The cytokines causing such a phenomenon include, fibroblast growth factor (FGF family), transforming growth factor (TGF-β), platelet derived growth factor (PDGF), etc.

Anti-inflammatory agents have been used to treat fibrosis with the aim of suppressing chronic inflammation, but such treatments can be unsatisfactory in terms of efficacy and side effects. Numerous studies have been performed to obtain substances that inhibit the production or the activity of the cytokines thought to be involved in fibrosis. Tranilast (n-[3,4-dimethoxycinnamoyl] anthranilic acid) is an anti-fibrotic agent used in Japan for the treatment of fibrotic skin disorders such as keloids and scleroderma. Although the precise mechanisms and mode of action of tranilast are incompletely understood, its ability to. inhibit ER phosphorylation, a major intermediate in the TGF-β signalling pathway, may underlie its antifibrotic effects, with known actions of tranilast including the inhibition of TGF-(3-induced extracellular matrix production in a range of cell types. Tranilast has also been shown to attenuate TGF- -induced collagen synthesis in cardiac fibroblasts, using an experimental model of diabetic cardiac disease, and to reduce inflammation in allergic diseases, such as allergic rhinitis and bronchial asthma, etc. In addition, tranilast has been shown to have anti-proliferative activity.

However, it has recently been shown that genetic factors in certain patients may confer susceptibility to tranilast-induced hyperbilirubinemia. One possibility for how this may arise is the presence of Gilbert's syndrome polymorphisms of the glucuronosyltransferase UGT1A1, which leads to increased susceptibility to tranilast- induced hyperbilirubinemia. Such hyperbilirubinemia may result from. the low level of UGT1A1 glucuronosyltransferase present in individuals with this syndrome. Tranilast itself, and its major metabolite N3 (4-desmethyl-trani!ast), have been shown to be inhibitors of UGT1A1 , potentially leading to aberrant metabolism of bilirubin and its accumulation.

Accordingly, compounds that are based on tranilast have the potential to provide further biologically active compounds that would be expected to have useful, improved pharmaceutical properties with potential anti-fibrotic, anti-inflammatory, and antiproliferative or anti-neoplastic activity for the treatment or prevention of diseases associated with fibrosis, diseases characterised by inflammation or neoplastic disease (both benign and malignant), and as alternatives/adjuncts to tranilast. Summary of the Invention

In one aspect, the present invention provides a compound of Formula (I)

Formula (I) or a pharmaceutically acceptable salt or prodrug thereof, wherein:

- W is selected from the group consisting of: CR 7 and N;

- A is selected from the group consisting of: -(CR 8 RV(Y) q -(C(O))r-(CR 10 R 11 ) 8 - and

-(CR 8 R 9 )p-(C(O))r(Y) q -(CR 10 R 11 ) 8 -, wherein Y is selected from the group consisting of: O, S, NR 12 , each p and s are an integer independently selected from the group consisting of 0, 1 , and 2, each q and r are an integer independently selected from the group consisting of 0 and 1 , and p + q + r + s is an integer selected from the group consisting of 1 , 2, and 3;

- Z 1 -Z 2 is selected from the group consisting of N-C= and C=C;

- X 1 is selected from the group consisting of: C=0, CF 2 or S0 2 , P0 2 ;

- X 2 is selected from the group consisting of: NR 13 and (CH 2 ) t wherein t is an integer selected from the group consisting of: 0 and 1 ;

- D is selected from the group consisting of: a cycloalkyi, heterocycloalkyi, cycloalkenyl, heterocycloalkenyl, aryl or heteroaryl ring;

- R\ R 4 , R 5 , R e , R 7 , R 8 , R 9 , R 0 , and R 1 are each independently selected from the group consisting of: H, halogen, OH, NQ 2 , CN, NH 2 , optionally substituted d- C 2 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted d-Cio heteroalkyl, optionally substituted C 3 -d 2 cycloalkyi, optionally substituted C 3 -C 2 cycloalkenyl, optionally substituted C 2 - C 12 heterocycloalkyi, optionally substituted C 2 -C 12 heterocycloalkenyl, optionally substituted C e -C 16 aryl, optionally substituted Ci-C 18 heteroaryl, optionally substituted CVC 12 alkyloxy, optionally substituted C 2 -C 2 alkenyloxy, optionally substituted C 2 -C 12 alkynyloxy, optionally substituted d-do heteroalkyloxy, optionally substituted C 3 -Ci 2 cycloalkyloxy, optionally substituted C 3 -C 12 cycloalkenyloxy, optionally substituted Ci-C 12 heterocycloalkyloxy, optionally substituted C 1 -C 12 heterocycloalkenyloxy, optionally substituted C e -C 1B aryloxy, optionally substituted Ci-C 18 heteroaryloxy, optionally substituted C,-C 12 alkylamino, SR 14 , S0 3 H, S0 2 NR 15 R 16 , S0 2 R 14 , SONR 15 R 16 , SOR 14 , COR 14 , COOH, COOR 14 , CONR 15 R 1B , NR 5 COR 14 , NR 15 COOR 14 , NR 15 S0 2 R 14 , NR 15 CONR R 16 , NR 15 R 16 , and acyl;

R 2 and R 3 , are each independently selected from the group consisting of: H, OH, N0 2 , CN, NH 2 , optionally substituted C t -C 2 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted C1-C10 heteroalkyl, optionally substituted C 3 -Ci 2 cycloalkyl, optionally substituted C 3 -C 12 cycloalkenyl, optionally substituted C 2 -C 12 heterocycloalkyi, optionally substituted C 2 -C 2 heterocycloalkenyl, optionally substituted C e -C 1B aryl, optionally substituted C de heteroaryl, optionally substituted C C 2 alkyloxy, optionally substituted C 2 -C 12 alkenyloxy, optionally substituted C r C 2 alkynyloxy, optionally substituted C -C, 0 heteroalkyloxy, optionally substituted C 3 -C 12 cycloalkyloxy, optionally substituted C 3 -C 12 cycloalkenyloxy, optionally substituted C,-C 12 heterocycloalkyloxy, optionally substituted Ci-Ci 2 heterocycloalkenyloxy, optionally substituted Ce-Cie aryloxy, optionally substituted C -C a heteroaryloxy, optionally substituted d-C 12 alkylamino, SR 14 , S0 3 H, S0 2 NR 5 R 16 , S0 2 R 14 . SONR 15 R 18 , SOR 14 , COR 14 , COOH, COOR 14 , CONR 15 R ie , NR 15 COR 14 , NR 15 COOR 14 , NR 5 S0 2 R 14 , NR 15 CONR 18 R 17 , NR 5 R 18 , and acyl; or R 2 and R 3 may be fused to form a 5 or 6 membered cycloalkyl, heterocycloalkyi, aryl or heteroaryl ring' each of which may be optionally substituted;

R 12 , R 13 , R 15 , R ie , and R 17 are each independently selected from the group consisting of: H, an N-protecting group, optionally substituted C C 12 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 2 alkynyl, optionally substituted C C 10 heteroalkyl, optionally substituted C 3 -C 2 cycloalkyl, optionally substituted C 3 -C 2 cycloalkenyl, optionally substituted Ci-C 12 heterocycloalkyi, optionally substituted Ci-C 12 heterocycloalkenyl, optionally substituted C 6 -C 8 aryl, and optionally substituted C C ie heteroaryl;

R 14 is selected from the group consisting of H, optionally substituted C Ci 2 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted C C 10 heteroalkyl, optionally substituted C 3 -C 12 cycloalkyl, optionally substituted C 3 -Ci 2 cycloalkenyl, optionally substituted CrC i2 heterocycloalkyl, optionally substituted Ci-C 12 heterocycloalkenyl, optionally substituted C e -C 1e aryl, and optionally substituted d-Ci 8 heteroaryl;

- m is an integer selected from the group consisting of 0, 1 , 2, 3, and 4;

- n is an integer selected from the group consisting of 1 , 2, 3, 4, and 5; and

- m + n is an integer selected from the group consisting of 1 , 2, 3, 4, and 5.

As with any group of structurally related compounds which possess a particular utility, certain embodiments of variables of the compounds of the Formula (I), may be particularly useful in their end use application.

In some embodiments of the first aspect of the invention, R 2 is -X 3 TR 18 and R 3 is -X 4 - R 19 , wherein:

- R 1B and R B are the same or different and are selected from the group consisting of: H, halogen, C n o alkyl, C 3 -Ci 0 cyclokalkyl, C3-C10 cycloalky I methyl, C3-C10 alkene, d-do alkyne, aryl, C 5 -C 20 alkaryl, fused C 5 -C 20 aryl or alkaryl and a hydrocarbon chain containing a heterocyclic or fused ring, any of which may be optionally substituted;

- X 3 and X 4 are the same or different and are selected from the group consisting of: a bond CR 20 R 21 , O, NR 22 and S;

- R 20 and R 21 are the same or different and are selected from the group consisting of: H, halogen, OH, N0 2 , CN, NH 2 , optionally substituted C r d 2 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 2 alkynyl, optionally substituted d-do heteroalkyl, optionally substituted C 3 -C 12 cycloalkyl, optionally substituted C 3 -C 12 cycloalkenyl, optionally substituted C 2 -C 12 heterocycloalkyl, optionally substituted C 2 -C 12 heterocycloalkenyl, optionally substituted C e -C 18 aryl, optionally substituted Ci-C 18 heteroaryl, optionally substituted C Ci 2 alkyloxy, optionally substituted C 2 -C 12 alkenyloxy, optionally substituted C 2 -C 12 alkynyloxy, optionally substituted d-do heteroalkyloxy, optionally substituted C 3 -C 12 cycloalkyloxy, optionally substituted C 3 -C 12 cycloalkenyloxy, optionally substituted d-d 2 heterocycloalkyloxy, optionally substituted C r C 12 heterocycloalkenyloxy, optionally substituted C e -C 8 aryloxy, optionally substituted d-d 8 heteroaryloxy, optionally substituted d-d 2 alkylamino, SR 14 , S0 3 H, S0 2 NR 15 R IE , S0 2 R 14 , SONR 15 R 16 , SOR 4 , COR 14 , COOH, COOR 14 , CONR 15 R IE , NR 15 COR 14 , NR 15 COOR 14 , NR 1 S0 2 R 14 , NR 1 CONR 18 R 17 , NR 15 R 16 , and acyl; and

- R 22 is selected from the group consisting of: H, an N-protecting group, optionally substituted d-C 12 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted C^do heteroalkyl, optionally substituted C 3 -C 12 cycloalkyl, optionally substituted C 3 -C 12 cycloalkenyl, optionally substituted Ci-Ci 2 heterocycloalkyl, optionally substituted Ci-C 12 heterocycloalkenyl, optionally substituted C 6 -C 18 aryl, and optionally substituted C1-C18 heteroaryl.

This provides compounds of Formula (la):

In some embodiments of the first aspect of the invention, R 18 and R 9 are fused to form a 5 or 6 membered cycloalkyl, heterocycloalkyl, cycloalkenyl, heterocycloalkenyl, aryl or heteroaryl ring, each of which may be optionally substituted.

In some embodiments of the first aspect of the invention, D is selected from the group consisting of: phenyl, C 3 -C 6 cycloalkyl, C2-C5 heterocycloalkyl, C 2 -C 5 heteroaryl.

In some embodiments of the first aspect of the invention, R 5 is selected from the group consisting of: COOR 14 , COR 14 , CONR 15 R 18 , NR 15 R ie , SO z R 14 and SONR 15 R 18 . For example, R 5 may be selected from the group consisting of: COOH, CONH 2 , CONHCH 3 , CONHOH, NH 2 , S0 2 CH 3 , S0 2 NH 2l SONHCH 3 , and SON(CH 3 ) 2 .

In some embodiments of the first aspect of the invention, D is selected from the group consisting of phenyl and C 2 -C 5 heteroaryl and the R 5 substituent is ortho to X 2 . In some other embodiments, D is selected from the group consisting of C 3 -C e cycloalkyl and C 2 - C 5 heterocycloalkyl and the R 5 substituent is either geminal or vicinal to X 2 .

In some embodiments of the first aspect of the invention, R e is selected from the group consisting of: H and halogen. In some embodiments of the first aspect of the invention, X 1 is selected from the group consisting of: C=0 and S0 2 .

In some embodiments of the first aspect of the invention, X 2 is NR 13 .

In some embodiments of the first aspect of the invention, R 13 is selected from the group consisting of: H and optionally substituted d-C^alkyl.

In some embodiments of the first aspect of the invention, W is CR 7 .

In some embodiments of the first aspect of the invention, R 7 is H.

In some embodiments of the first aspect of the invention, A is selected from the group consisting of: CH, CH 2 CH 2 , CH=CH, O, OC(O), OC(S), OCH 2 , C(0)0, C(S)0, SCH 2 , SC(O), SC(S), S, S0 2 CH 2 , S0 2l C(0)S, C(0)NH, C(S)S, NH, NH(CO), N, N=CH, and IMHCH 2 .

In some embodiments of the first aspect of the invention, R 2 and R 3 are each independently selected from the group consisting of: optionally substituted Ci-C 12 alkyloxy and optionally substituted C 2 -C 12 alkynyloxy.

In some embodiments of the first aspect of the invention, the optionally substituted C C 12 alkyloxy is C ! -C^fluoroalkyloxy. In some embodiments of the first aspect of the invention, the optionally substituted C 2 - C 12 alkynyloxy is C 2 -Ci 2 alkynylalkyloxy.

In some embodiments of the first aspect of the invention, R 1 is selected from the group consisting of: H and optionally substituted Ci-C e alkoxy.

In some embodiments of the first aspect of the invention, R 2 is selected from the group consisting of: H 3 CO, F 2 HCF 2 CO, F 2 HCO, F 3 CO, and CHCCH 2 0.

In some embodiments of the first aspect of the invention, R 3 is selected from the group consisting of. H 3 CO, F 2 HCF 2 CO, F 2 HCO, F 3 CO, and CHCCH 2 0. In some embodiments of the first aspect of the invention, R 4 is selected from the group consisting of: H and optionally substituted Ci-C 6 alkoxy.

In some embodiments of the first aspect of the invention, R 2 and R 3 together are selected from the group consisting of: OCF 2 CF 2 CO, OCH 2 CH 2 0, OCF 2 0, and OCH 2 0.

In some embodiments of the first aspect of the invention, the compound of Formula (I) is selected from the group consisting of:

R = S0 2 Me, S0 2 NH 2 , SONHMe, SONMe 2 R = NH 2l CONH 2l CONHMe, CONHOH

H

or a pharmaceutically acceptable salt or prodrug thereof.

In a second aspect, the present invention provides a compound of Formula (II)

Formula (li) or a pharmaceutically acceptable salt or prodrug thereof, wherein:

- A is selected from the group consisting of: -(CR 8 R 9 ) p -(Y) q -(C(O)) r -(CR 10 R 11 ) s - and

-(CR 8 R 8 )p-(C(O))r(Y)q-(CR 10 R 1 V, wherein Y is selected from the group consisting of: O, S, and NR 12 , each p and s are an integer independently selected from the group consisting of: 0, 1, and 2, each g and r are an integer independently selected from the group consisting of: 0 and 1, and p + q + r + s is an integer selected from the group consisting bf: 1 , 2, and 3;

- X 1 is selected from the group consisting of: C=0, CF 2l and S0 2l P0 2 ;

- X 2 is selected from the group consisting of: NR 13 or (CH 2 ) t wherein t is an integer selected from the group consisting of: 0 and 1 ; D is selected from the group consisting of: a cycloalkyi, heterocycioalkyi, cycloalkenyl, heterocycloalkenyl, aryl or heteroaryl ring;

R 1 , R 2 and R 3 , are each independently selected from the group consisting of: H, OH, N0 2 , CN, NH 2 , optionally substituted C C 12 alkyl, optionally substituted C 2 - C 12 alkenyl, optionally substituted C 2 -C 2 alkynyl, optionally substituted C C i0 heteroalkyl, optionally substituted C 3 -C 12 cycloalkyi, optionally substituted C 3 -C 12 cycloalkenyl, optionally substituted C 2 -C 12 heterocycioalkyi, optionally substituted C 2 -Ci 2 heterocycloalkenyl, optionally substituted Ce-Ci 8 aryl, optionally substituted Ci-C 18 heteroaryl, optionally substituted C^C^ alkyloxy, optionally substituted C 2 -C 12 alkenyloxy, optionally substituted C 2 -C 12 alkynyloxy, optionally substituted C 1 -C 10 heteroalkyloxy, optionally substituted C 3 -C 2 cycloalkyloxy, optionally substituted C 3 -C 12 cycloalkenyloxy, optionall substituted C r C 12 heterocycloalkyloxy, optionally substituted C1-C 1 2 heterocycloalkenyloxy, optionally substituted C e -C 18 aryloxy, optionally substituted C Ci B heteroaryloxy, optionally substituted 0,-0,2 alkylamino, SR 14 , SO 3 H, S0 2 NR 15 R ie , S0 2 R 14 , SONR 15 R ie , SOR 14 , COR 14 , COOH, COOR 14 , CONR 15 R 18 , NR 15 COR 14 , NR 15 COOR 14 , NR 15 S0 2 R 14 , NR 15 CONR 18 R 17 , NR 5 R 18 , and acyl; or R 2 and R 3 may be fused to form a 5 or 6 membered cycloalkyi, heterocycioalkyi, aryl or heteroaryl ring' each of which may be optionally substituted;

R 4 , R 5 , R e , R 7 , R 8 , R 8 , R 10 and R 1 are each independently selected from the group consisting of: H, halogen, OH, N0 2 , CN, NH 2 , optionally substituted 0,-0,2 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C, 2 alkynyl, optionally substituted C C 10 heteroalkyl, optionally substituted C 3 -C 12 cycloalkyi, optionally substituted C 3 -C, 2 cycloalkenyl, optionally substituted C 2 -C 2 heterocycioalkyi, optionally substituted C 2 -C 12 heterocycloalkenyl, optionally substituted C e -C, e aryl, optionally substituted C,-C, 8 heteroaryl, optionally substituted C,-C, 2 alkyloxy, optionally substituted C 2 -C 12 alkenyloxy, optionally substituted C 2 -C 12 alkynyloxy, optionally substituted C,-C 10 heteroalkyloxy, optionally substituted C 3 -C 12 cycloalkyloxy, optionally substituted C 3 -C, 2 cycloalkenyloxy, optionally substituted C,-C 12 heterocycloalkyloxy, optionally substituted Ci-C 12 heterocycloalkenyloxy, optionally substituted C 6 -C B aryloxy, optionally substituted Ci-Ci 8 heteroaryloxy, optionally substituted C,-Ci 2 alkylamino, SR 14 , S0 3 H, S0 2 NR 5 R 16 , S0 2 R 14 , SONR 15 R ie , SOR 14 , COR 14 , COOH, COOR 14 , CONR 1 R ie , NR 5 COR 14 , NR 15 COOR 14 , NR 15 S0 2 R 14 , NR 5 CONR 16 R 17 , NR 15 R 16 , and acyl; - R 12 , R 3 , R 15 , R 16 , and R 7 are each independently selected from the group consisting of: H, a ^protecting group, optionally substituted CrC 2 alkyl, optionally substituted C 2 -C 2 alkenyl, optionally substituted C 2 -Ci 2 alkynyl, optionally substituted Ci-C 0 heteroalkyl, optionally substituted C 3 -C 12 cycloalkyl, optionally substituted C 3 -C 12 cycloalkenyl, optionally substituted Ci-C 12 heterocycloalkyl, optionally substituted Ci-Ci 2 heterocycloalkenyl, optionally substituted C 6 -C 8 aryl, and optionally substituted C C 18 heteroaryl;

- R 14 is selected from the group consisting of H, optionally substituted C C 2 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -G 12 alkynyl, optionally substituted C^do heteroalkyl, optionally substituted C 3 -C 12 cycloalkyl, optionally substituted C 3 -C 2 cycloalkenyl, optionally substituted C C 12 heterocycloalkyl, optionally substituted Ci-C 12 heterocycloalkenyl, optionally substituted Ce-Ci 8 aryl, and optionally substituted C1-C18 heteroaryl;

- m is an integer selected from the group consisting of: 0, 1 , 2, 3, and 4;

- n is an integer selected from the group consisting of: 1 , 2, 3, 4, and 5; and

- m + n is an integer selected from the group consisting of: 1 , 2, 3, 4, and 5.

In some embodiments of the second aspect of the invention, R is -X 3 -R 18 and R 2 is - X 4 -R 19 , wherein:

- R ia and R 19 are the same or different and are selected from the group consisting of: H, halogen, C no alkyl, C 3 -Ci 0 cyclokalkyl, C 3 -C 10 cycloalkylmethyl, C 3 -Ci 0 alkene, C 3 -C 10 alkyne, aryl, C 5 -C 20 alkaryl, fused C 5 -C 20 aryl or alkaryl and a hydrocarbon chain containing a heterocyclic or fused ring, any of which may be optionally substituted;

- X 3 and X 4 are the same.or different and are selected from the group consisting of: a bond CR 20 R 21 , O, NR 22 , and S;

- R 20 and R 21 are the same or different and are selected from the group consisting of: H, halogen, OH, N0 2l CN, NH 2 , optionally substituted C r C 12 alkyl, optionally substituted C 2 -C 2 alkenyl, optionally substituted C 2 -C 2 alkynyl, optionally substituted Ci-do heteroalkyl, optionally substituted C 3 -C 12 cycloalkyl, optionally substituted C 3 -C 2 cycloalkenyl, optionally substituted C 2 -Ci 2 heterocycloalkyl, optionally substituted C 2 -C 2 heterocycloalkenyl, optionally substituted C 6 -C 18 aryl, optionally substituted Ci-C 18 heteroaryl, optionally substituted Ci-C 12 alkyloxy, optionally substituted C 2 -C 12 alkenyloxy, optionally substituted C 2 -C 2 alkynyloxy, optionally substituted C do heteroalkyloxy, optionally substituted

C 3 -Ci 2 cycloalkyloxy, optionally substituted C 3 -C 12 cycloalkenyloxy, optionally substituted C C 12 heterocycloalkyloxy, optionally substituted C C 12 heterocycloalkenyloxy, optionally substituted C 6 -C 18 aryloxy, optionally substituted d-C 18 heteroaryloxy, optionally substituted C r C 12 alkylamino, SR 14 , S0 3 H, S0 2 NR 15 R ie , S0 2 R 14 , SONR 15 R 16 , SOR 14 , COR 14 , COOH, COOR 14 , CONR 15 R e , NR 15 COR 14 , NR 15 COOR 14 , NR 15 S0 2 R 14 , NR 1s CONR 16 R 17 , NR 5 R 18 , and acyl; and

- R 22 is selected from the group consisting of: H, an N-protecting group, optionally substituted d-C^ alky I, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 2 alkynyl, optionally substituted d-Cn^heteroalkyl, optionally substituted C 3 -C 12 cycloalkyl, optionally substituted C 3 -C 12 cycloalkenyl, optionally substituted Ci-Ci 2 heterocycloalkyl, optionally substituted Ci-C 12 heterocycloalkenyl, optionally substituted C e -C 18 aryl, and optionally substituted heteroaryl.

This provides compounds of Formula (lla):

Formula (lla)

In some embodiments of the second aspect of the invention, R 2 is -X 3 -R 18 and R 3 is - X -R 19 , wherein:

- R 18 and R 19 are the same or different and are selected from the group consisting of: H, halogen, d-1 0 alkyl, C 3 -C 0 cyclokalkyl, C 3 -C 10 cycloalkylmethyl, C3-C10 alkene, C 3 -C 10 alkyne, aryl, Cs-C^ alkaryl, fused C 3 -C 20 aryl or alkaryl and a hydrocarbon chain containing a heterocyclic or fused ring, any of which may be optionally substituted;

- X 3 and X 4 are the same or different and are selected from the group consisting of: a bond CR 20 R 21 , O, NR 22 , and S;

. - R 20 and R 21 are the same or different and are selected from the group consisting of: H, halogen, OH, N0 2 , CN, NH 2 , optionally substituted d-d2 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted d-do heteroalkyl, optionally substituted C 3 -C 12 cycloalkyl, optionally substituted C 3 -d 2 cycloaikenyl, optionally substituted C 2 -Ci 2 heterocycloalkyi, optionally substituted C 2 -C 12 heterocycloalkenyl, optionally substituted C 6 -C 18 aryl, optionally substituted Ci-C 18 heteroaryl, optionally substituted C C 12 alkyloxy, optionally substituted C 2 -Ci 2 alkenyloxy, optionally substituted C 2 -C 2 alkynyloxy, optionally substituted Ci-C 0 heteroalkyloxy, optionally substituted C 3 -C 2 cycloalkyloxy, optionally substituted C 3 -C 2 cycloalkenyloxy, optionally substituted Ci-C 12 , heterocycloalkyloxy, optionally substituted C C 2 heterocycloalkenyloxy, optionally substituted C 9 -C 18 aryloxy, optionally substituted Ci-C 18 heteroaryloxy, optionally substituted C C 2 alkylamino, SR 14 , S0 3 H, S0 2 NR 15 R ie , S0 2 R 14 , SONR 15 R 18 , SOR 14 , COR 14 , COOH, COOR 14 , CONR 15 R ie , NR 15 COR 14 , NR 15 COOR 14 , NR 5 S0 2 R 14 , NR 15 CONR ie R 17 , NR 15 R 16 , and acyl; and

- R 22 is selected from the group consisting of: H, an N-protecting group, optionally substituted Ci-C 12 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted C C 10 heteroalkyl, optionally substituted C 3 -C 12 cycloalkyi, optionally substituted C 3 -C 12 cycloaikenyl, optionally substituted C C 12 heterocycloalkyi, optionally substituted C C 2 heterocycloalkenyl, optionally substituted C 6 -C 18 aryl, and optionally substituted C1-C18 heteroaryl.

This provides compounds of Formula (lib):

In some embodiments of the second aspect of the invention, R 8 and R 19 are fused to form a 5 or 6 membered cycloalkyi, heterocycloalkyi, aryl or heteroaryl ring, each of which may be optionally substituted. In some embodiments of the second aspect of the invention, D is selected from the group consisting of: phenyl, C 3 -C e cycloaikyi, C 2 -C 5 heterparyl.

In some embodiments of the second aspect of the invention, R 5 is selected from the group consisting of: H, CN, N0 2l and d-Cealkyl.

In some embodiments of the second aspect of the invention, R 6 is selected from the group consisting of: COOR 14 , COR 14 , CONR 15 R 16 , NR 15 R ie , and S0 2 R 14 and SONR 15 R 1B .

In some embodiments of the second aspect of the invention, R 6 is selected from the group consisting of: COOH, CONH 2 , CONHOH, CONHCH 3 , NH 2 , S0 2 CH 3 , S0 2 NH 2 , SONHCH3, and SON(CH 3 ) 2 . In some embodiments of the second aspect of the invention, D is selected from the group consisting of phenyl and C 2 -C 5 heteroaryl and the R 6 substituent is ortho to X 2 .

In some embodiments of the second aspect of the invention, D is selected from the group consisting of C 3 -C e cycloaikyi and C 2 -C 5 heterocycloalkyl and the R 6 substituent is either geminal or vicinal to X 2 .

In some embodiments of the second aspect of the invention, R 7 is selected from the group consisting of H and halogen. In some embodiments of the second aspect of the invention, X 1 is selected from the group consisting of: C=0 and S0 2 .

In some embodiments of the second aspect of the invention, X 2 is NR 13 . In some embodiments of the second aspect of the invention, R 13 is selected from the group consisting of: H and optionally substituted Ci-C 2 alkyl.

In some embodiments of the second aspect of the invention, A is selected from the group consisting of: CH 2 CH 2 , CH=CH, C(0)0, C(0)S, C(0)NR 12 , OC(O), SC(O), NR 12 C(0), CH 2 CH 2 CH 2 , CH=CHCH 2 , CH 2 CH=CH, OCH 2 CH 2 , SCH 2 CH 2l NR 12 CH 2 CH 2 , CH 2 CH 2 0, CHzCHzS, CH 2 CH 2 NR 12 , C(0)OCH 2l C(0)SCH 2 , C(0)NR 2 CH 2 , CH 2 OC(0), CH 2 SC(0), and CH 2 NR 12 C(0). In some embodiments of the second aspect of the invention, R 1 , R 2 and R 3 are each independently selected from the group consisting of: optionally substituted (- C 12 alkyloxy and optionally substituted C 2 -C 12 alkynyloxy.

In some embodiments of the second aspect of the invention, the optionally substituted C C 12 alkyloxy is CrC^fluoroalkyloxy.

In some embodiments of the second aspect . of the invention, the optionally substituted C 2 -C 12 alkynyloxy is C 2 -C 12 alkynylalkyloxy.

In some embodiments of the second aspect of the invention, R 1 is selected from the group consisting of: H 3 CO, F 2 HCF 2 CO, F 2 HCO, F 3 CO, and CHCCH 2 0. In some embodiments of the second aspect of the invention, R 2 is selected from the group consisting of: H 3 CO, F 2 HCF 2 CO, F 2 HCO, F 3 CO, and CHCCH 2 0.

In some embodiments of the second aspect of the invention, R 3 is selected from the group consisting of: H 3 CO, F 2 HCF 2 CO, F 2 HCO, F 3 CO, and CHCCH z O.

In some embodiments of the second aspect of the invention, R 1 and R 2 together are selected from the group consisting of: OCF 2 CF 2 CO, OCH 2 GH 2 0, OCF 2 0, and OCH 2 0.

In some embodiments of the second aspect of the invention, R 2 and R 3 together are selected from the group consisting of: OCF 2 CF 2 CO, OCH 2 CH 2 0, OCF 2 0, and OCH 2 0.

In some embodiments of the second aspect of the invention, the compound of Formula (II) is selected from the group consisting of:

or a pharmaceutically acceptable salt or prodrug thereof. In a third aspect, the present invention provides a compound of Formula (III)

Formula (III) or a pharmaceutically acceptable salt or prodrug thereof, wherein:

- A is selected from the group consisting of: -(CR 9 R 10 ) p -(Y) q -(C(O)) r -(CR 11 R 12 ) s - and

-(CR 9 R 10 )p-(C(O)) r (Y) q -(CR 11 R 12 ) 8 -, wherein Y is selected from the group consisting of. O, S, NR 13 , each p and s are an integer independently selected from the group consisting of: 0, 1, and 2, each q and r are an integer independently selected from the group consisting of: 0 and 1, and p + q + r + s is an integer selected from the group consisting of: 1, 2, and 3;

- T is selected from the group consisting of: a single bond, a double bond, a triple

X 1 is selected from the group consisting of. C=0, CF 2 or S0 2 , P0 2 ; X 2 is selected from the group consisting of: CR 17 and N;

Z 1 , Z 2 , Z 3 , and Z 4 are each independently selected from the group consisting of CR B and N;

R\ R 4 , R 5 , R 8 , R 9 , R 10 , R 11 , R 12 , R 15 , R 16 and R 17 are each independently selected from the group consisting of: H, halogen, OH, N0 2 , CN, NH 2 , optionally substituted Ci-C 12 alky I, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted C C w heteroalkyl, optionally substituted C 3 -C 12 cycloalkyi, optionally substituted C 3 -C 12 cycloalkenyl, optionally substituted C 2 -C 12 heterocycloalkyl, optionally substituted C 2 -C 12 heterocycloalkenyl, optionally substituted C e -Ci 8 aryl, optionally substituted d- Ci8 heteroaryl, optionally substituted C r -C 12 alkyloxy, optionally substituted C 2 - C 12 alkenyloxy, optionally substituted C 2 -C 12 alkynyloxy, optionally substituted C1-C10 heteroalkyloxy, optionally substituted C 3 -Ci 2 cycloalkyloxy, optionally

. substituted C 3 -C 12 cycloalkenyloxy, optionally substituted C- -C 12 heterocycloalkyloxy, optionally substituted C C 12 heterocycloalkenyloxy, optionally substituted C e -Ci 8 aryloxy, optionally substituted Ci-C 18 heteroaryloxy, optionally substituted C C 12 alkylamino, SR 18 , SO 3 H, S0 2 NR 19 R 20 , S0 2 R 18 , SONR 19 R 20 , SOR 18 , COR 18 , COOH, COOR 18 , CONR 19 R 20 , NR 19 COR 18 , NR 19 COOR 18 , NR 19 S0 2 R 18 , NR 19 CONR 20 R 21 , NR 1fl R 20 , and acyl;

R 2 and R 3 , are each independently selected from the group consisting of: H, OH, N0 2 , CN, NH 2 , optionally substituted Ci-Ci 2 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted Ci-C 10 heteroalkyl, optionally substituted C 3 -C 12 cycloalkyi, optionally substituted C 3 -C 12 cycloalkenyl, optionally substituted C 2 -C 12 heterocycloalkyl, optionally substituted C 2 -C 12 heterocycloalkenyl, optionally substituted e~Ci 8 aryl, optionally substituted C C 18 heteroaryl, optionally substituted C Ci 2 alkyloxy, optionally substituted C 2 -C 12 alkenyloxy, optionally substituted C 2 -C 12 alkynyloxy, optionally substituted Ci-C 10 heteroalkyloxy, optionally substituted C 3 -C 12 cycloalkyloxy, optionally substituted C 3 -C 12 cycloalkenyloxy, optionally substituted C,-C 12 heterocycloalkyloxy, optionally substituted C Ci 2 heterocycloalkenyloxy, optionally substituted C 6 -C 1B aryloxy, optionally substituted C1-C18 heteroaryloxy, optionally substituted C Ci 2 alkylamino, SR 18 , SO 3 H, S0 2 NR 9 R 20 , S0 2 R 1B , SONR 19 R 20 , SOR 18 , COR 18 , COOH, COOR 18 , CONR 19 R 20 , NR 19 COR 18 , NR 19 COOR 18 , NR 19 S0 2 R 18 , NR 19 CONR 0 R 21 , NR^R 20 , and acyl; or R 2 and R 3 may be fused to form a 5 or 6 membered cycloalkyi, heterocycloalkyl, aryl or heteroaryl ring, each of which may be optionally substituted; and R 7 are present when T is a single bond, a double bond

but not when T is a triple bond, each R e and R 7 being independently selected from the group consisting of: H, N0 2 , CN, optionally substituted Ci-C 12 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 2 alkynyl, optionally substituted C do heteroalkyi, optionally substituted C 3 -C 12 cycloalkyi, optionally substituted C 3 -C 12 cycloalkenyl, optionally substituted C 2 -C 12 heterocycloalkyl, optionally substituted C 2 -C 12 heterocycloalkenyl, optionally substituted C e -C B aryl, optionally substituted C r C 1B heteroaryl, optionally substituted C Ci 2 alkyloxy, optionally substituted C 2 - C 12 alkenyloxy, optionally substituted C 2 -C 12 alkynyloxy, optionally substituted

( C heteroalkyloxy, optionally substituted C 3 -C 2 cycloalkyloxy, optionally substituted C 3 -C 12 cycloalkenyloxy, optionally substituted C C 12 heterocycloalkyloxy, optionally substituted C C 12 heterocycloalkenyloxy, optionally substituted C 6 -Ci 8 aryloxy, optionally substituted Ci-C 1B heteroaryloxy, optionally substituted C r Ci 2 alkylamino, SR B , S0 3 H, S0 2 R 18 ,

SONR 19 R 20 , SOR 18 , COR 1B , COOH, COOR 18 , CONR 1 R 20 , NR 19 COR 18 , . NR 9 COOR 18 , NR 19 CONR 20 R 21 , NR 19 R 20 , and acyl;

- R 3 , R 19 , R 20 and R 21 are each independently selected from the group consisting of: H: a N-protecting group, optionally substituted C C 12 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted d-do heteroalkyi, optionally substituted C 3 -C 2 cycloalkyi, optionally substituted C 3 -C 12 cycloalkenyl, optionally substituted -d? heterocycloalkyl, optionally substituted d-Ci 2 heterocycloalkenyl, optionally substituted C e -C 18 aryl, and optionally substituted d-de heteroaryl;

- R 18 is selected from the group consisting of H, optionally substituted Ci-C 12 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted d-do heteroalkyi, optionally substituted C 3 -Ci 2 cycloalkyi, optionally substituted C 3 -C 2 cycloalkenyl, optionally substituted d-d heterocycloalkyl, optionally substituted C C 12 heterocycloalkenyl, optionally substituted C e -C 18 aryl, and optionally substituted C 1 -C18 heteroaryl; and .

- n is an integer selected from the group consisting of: 0, 1 , 2, 3, and 4.

In some embodiments of the third aspect of the invention, R 2 is -X 3 -R 22 and R 3 is -X 4 - R 23 , wherein: - R 22 and R 23 are the same or different and are selected from the group consisting of: H, halogen, C no alkyl, C 3 -C 10 cyclokalkyl, C 3 -Ci 0 cycloalkylmethyl, C 3 -C 10 alkene, C 3 -C 10 alkyne, aryl, C 5 -C 20 alkaryl, fused C 5 -C 20 aryl or alkaryl and a hydrocarbon chain containing a heterocyclic or fused ring, any of which may be optionally substituted;

- X 3 and X 4 are the same or different and are selected from the group consisting of: a bond CR 24 R 25 , O, NR 28 , and S;

- R 24 and R 25 are the same or different and are selected from the group consisting of: H, halogen, OH, N0 2> CN, NH 2 , optionally substituted C C 12 alkyl, optionally substituted ,C 2 -Ci 2 alkenyl, optionally substituted C 2 -Ci 2 alkynyl, optionally substituted C C™ heteroalkyl, optionally substituted C 3 -C 12 cycloalkyi, optionally substituted C 3 -C 12 cycloalkenyi, optionally substituted C 2 -C 12 heterocycloalkyi, optionally substituted C 2 -C 12 heterocycloalkenyl, optionally substituted C e -C 18 aryl, optionally substituted C C ie heteroaryl, optionally substituted C C 2 alkyloxy, optionally substituted C 2 -C 12 alkenyloxy, optionally substituted C 2 -Ci 2 alkynyloxy, optionally substituted C^C^ heteroalkyloxy, optionally substituted C 3 -C 12 cycloalkyloxy, optionally substituted C 3 -C 12 cycloalkenyloxy, optionally substituted C C 12 heterocycloalkyloxy, optionally substituted C C 12 heterocycloalkenyloxy, optionally substituted C 6 -C B aryloxy, optionally substituted d-C 18 heteroaryloxy, optionally substituted C^C^ alkylamino, SR 14 , SQ 3 H, S0 2 NR 15 R 18 , S0 2 R 14 , SONR 15 R 18 , SOR 4 , COR 14 , COOH, COOR 14 , CONR 15 R 18 , NR 15 COR 14 , NR 16 COOR 14 , NR 15 S0 2 R 14 , NR 15 CONR 18 R 17 , NR 15 R 18 , and acyl; and

- R 28 is selected from the group consisting of: H, an N-protecting group, optionally substituted Ci-C 12 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted C -C \0 heteroalkyl, optionally substituted C 3 -C 12 cycloalkyi, optionally substituted C 3 -C 12 cycloalkenyi, optionally substituted C C 12 heterocycloalkyi, optionally substituted CrC 12 heterocycloalkenyl, optionally substituted Ce _ Ci 8 aryl, and optionally substituted Ci-Cie heteroaryl.

In some embodiments of the third aspect of the invention, R 22 and R 23 are fused to form a 5 or 6 membered cycloalkyi, cycloalkenyi, heterocycloalkyi, heterocycloalkenyl, aryl or heteroaryl ring, each of which may be optionally substituted.

In some embodiments of the third aspect of the invention, X 1 is selected from the group consisting of: C=0 and S0 2 . In some embodiments of the third aspect of the invention, X 2 is N.

In some embodiments of the third aspect of the invention, A is selected from the group consisting of: CH 2 CH 2 CH 2 , CH 2 CH 2 0, CH 2 CH 2 S, CH 2 CH 2 NR 13 , CH 2 CH=CH, CH=CHCH 2 , CH=CHC(0), C(0)CH=CH, C=NC(0), CH 2 OC(0), C(0)OC(0), CHzSC ), C(0)SC(0), C(0)OCH 2l C(0)SCH 2 , C(0)CH 2 NR 13 , C(0)CH 2 S, C(0)CH 2 0, C(S)CH 2 0, OC(O), CH 2 0, C(0)0, CH 2 S, CH 2 NR 13 , CH 2 CH 2 , SC(O), C(S)0, C(0)S, C(S)S, C(0)NR 13 , C(S)NR 13 .

In some embodiments of the third aspect of the invention, R 2 and R 3 are each independently selected from the group consisting of: optionally substituted C C 12 alkyloxy and optionally substituted C 2 -C 12 alkynyloxy. In some embodiments of the third aspect of the invention, the optionally substituted C Ci2 alkyloxy is Ci-C 12 fluoroalkyloxy.

In some embodiments of the third aspect of the invention, the optionally substituted C 2 - C 12 alkynyloxy is C2-Ci 2 alkynylalkyloxy.

In some embodiments of the third aspect of the invention, R 2 is selected from the group consisting of: H 3 CO, F 2 HCF 2 CO, F 2 HCO, F 3 CO, and CHCCH 2 0.

In some embodiments of the third aspect of the invention, R 3 is selected from the group consisting of: H 3 CO, F 2 HCF 2 CO, F 2 HCO, F 3 CO, and CHCCH 2 0.

In some embodiments of the third aspect of the invention, R 2 and R 3 together are selected from the group consisting of: OCF 2 CF 2 CO, OCH 2 CH 2 0, OCF 2 0, and OCH 2 0. In some embodiments of the third aspect of the invention, T is selected from the group

consisting of: a double bond .

In some embodiments of the third aspect of the invention, R 8 is selected from the group consisting of: H and C,-C B alkyl. In some embodiments of the third aspect of the invention, R 7 is selected from the group consisting of: H, CN, N0 2 and C C e alkyl. In some embodiments of the third aspect of the invention, R 1 is selected from the group consisting of: H and optionally substituted C t -C 6 alkoxy.

In some embodiments of the third aspect of the invention, R 4 is selected from the group consisting of: H and optionally substituted C -C e alkoxy.

In some embodiments of the third aspect of the invention, R 5 is H.

In some embodiments of the third aspect of the invention, R 8 is selected from the group consisting of: H. halogen, COOR 18 , COR 18 , CONR 19 R 20 , R^R 20 , S0 2 R 18 , and SONR 19 R 20 .

In some embodiments of the third aspect of the invention, R 8 is selected from the group consisting of: H, halogen, COOH, CONH 2) CONHCH 3 , CONHOH, NH 2 , S0 2 CH 3 , S0 2 NH 2 , SONHCH 3 , and SON(CH 3 ) 2 .

In some embodiments of the third aspect of the invention, n is 1.

In some embodiments of the third aspect of the invention the compound of Formula (III) is selected from the group consisting of:

or a pharmaceutically acceptable salt or prodrug thereof.

In a fourth aspect, the present invention provides a compound of Formula (IV)

Formula (IV) or a pharmaceutically acceptable salt or prodrug thereof, wherein:

- Z-X-Y is selected from the group consisting of: N=C-N, N-C=N, 0-C=N, S-C=N,

N=C-0, N=C-S, C=C-NH, C=C-0, C=C-S, and C(0)-C=N;

- A is selected from the group consisting of: a bond, S0 2 , C, C=S, C=0, C=NR B , and NR 9 ;

- T is sele nsisting of: a single bond, a double bond, a triple

bond Z 1 , Z 2 , Z 3 , and Z are each independently selected from the group consisting of CR 8 and ;

R\ R 4 , R 5 , R 8 , R 0 , and R 11 are each independently selected from the group consisting of: H, halogen, OH, N0 2 , CN, NH 2 , optionally substituted CrC 12 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted C C 10 heteroalkyl, optionally substituted C 3 -C 2 cycloalkyi, optionally substituted C 3 -C 2 cycloalkenyl, optionally substituted (_½- C12 heterocycloalkyi, optionally substituted C 2 -C Z heterocycloalkenyl, optionally substituted C 3 -C 1a aryl, optionally substituted C -C18 heteroaryl, optionally substituted Ci-C 2 alkyloxy, optionally substituted C 2 -C 2 alkenyloxy, optionally substituted C 2 -C 12 alkynyloxy, optionally substituted C C 10 heteroalkyloxy, optionally substituted C 3 -C 12 cycloalkyloxy, optionally substituted C 3 -C 12 cycloalkenyloxy, optionally substituted C1-C12 heterocycloalkyloxy, optionally substituted C^C^ heterocycloalkenyloxy, optionally substituted C 6 -C 18 aryloxy, optionally substituted C C^ heteroaryloxy, optionally substituted Ci-C 2 alkylamino, SR 12 , S0 3 H, S0 2 NR 13 R 14 , S0 2 R 12 , SONR 13 R 14 , SOR 2 , COR 12 , COOH, COOR 12 , CONR 13 R 14 , NR 13 COR 12 , NR 13 COOR 12 , NR 13 S0 2 R 12 , NR 13 CONR 14 R 15 , NR 14 R 15 , and acyl;

R 2 and R 3 , are each independently selected from the group consisting of: H, OH, N0 2 , CN, NH 2 , optionally substituted C C 12 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 2 alkynyl, optionally substituted Ci-C 10 heteroalkyl, optionally substituted C 3 -d 2 cycloalkyi, optionally substituted C 3 - C 2 cycloalkenyl, optionally substituted C2-C 12 heterocycloalkyi, optionally substituted C 2 -C 12 heterocycloalkenyl, optionally substituted

optionally substituted C Ci 8 heteroaryl, optionally substituted C C 12 alkyloxy, optionally substituted C 2 -C 12 alkenyloxy, optionally substituted C 2 -C 12 alkynyloxy, optionally substituted Ci-C 10 heteroalkyloxy, optionally substituted C 3 -Ci 2 cycloalkyloxy, optionally substituted C 3 -C 12 cycloalkenyloxy, optionally substituted C C 12 heterocycloalkyloxy, optionally substituted C C 12 heterocycloalkenyloxy, optionally substituted C 6 -C 18 aryloxy, optionally substituted C,-Ci 8 heteroaryloxy, optionally substituted Ci-C 12 alkylamino, SR 12 , SO 3 H, S0 2 NR 13 R 14 , S0 2 R 12 , SONR 3 R 14 , SOR 12 , COR 12 , COOH, COOR 12 , CONR 13 R 14 , NR ,3 COR 12 , NR 13 COOR 12 , NR 13 S0 2 R 12 , NR 13 CONR 14 R 15 , NR 14 R 15 , and acyl; or R 2 and R 3 may be fused to form a 5 or 6 membered cycloalkyi, heterocycloalkyi, aryl or heteroaryl ring' each of which may be optionally substituted; 7 are present when T is a single bond, a double bond

but not when T is a triple bond, each R s and R 7 being independently selected from the group consisting of: H, N0 2 , CN, optionally substituted Ci-C 12 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted Ci-C 10 heteroalkyi, optionally substituted C 3 -C 12 cycloalkyl, optionally substituted C3-C12 cycloalkenyl, optionally substituted C 2 -Ci 2 heterocycloalkyl, optionally substituted C2-C 12 heterocycloalkenyl, optionally substituted Ce-C 8 aryl, optionally substituted C,- C18 heteroaryl, optionally substituted C C 12 alkyloxy, optionally substituted C 2 - C 2 alkenyloxy, optionally substituted C 2 -C 2 alkynyloxy, optionally substituted Ci-C 10 heteroafkyloxy, optionally substituted C 3 -C 12 cycloalkyloxy, optionally substituted C 3 -C 12 cycloalkenyloxy, optionally substituted C C 12 heterocycloalkyloxy, optionally substituted CrC 12 heterocycloalkenyloxy, optionally substituted C 6 -Ci B aryloxy, optionally substituted Ci-C 8 heteroaryloxy, optionally substituted C r C 2 alkylamino, SR 12 , S0 3 H, S0 2 NR 13 R 14 , S0 2 R 12 , SONR 13 R 14 , SOR 12 , COR 12 , COOH, COOR 2 , CONR 13 R 14 , NR i3 COR 12 , NR 13 COOR 12 , NR 13 S0 2 R 12 , NR 13 CONR 14 R 15 , NR 1 R 15 , and acyl;

R 9 , R 13 , R 14 , and R 15 are each independently selected from the group consisting of: H, an N-protecting group, optionally substituted (-VC 12 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted C Ci 0 heteroalkyi, optionally substituted C 3 -Ci 2 cycloalkyl, optionally substituted C 3 -C 12 cycloalkenyl, optionally substituted C C 12 heterocycloalkyl, optionally substituted C1-C12 heterocycloalkenyl, optionally substituted C e -C 18 aryl, and optionally substituted Ci-C 1B heteroaryl;

R Z is selected from the group consisting of H, optionally substituted Cn-C^ alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted C,-C 0 heteroalkyi, optionally substituted C 3 -C 12 cycloalkyl, optionally substituted C 3 -C 12 cycloalkenyl, optionally substituted C,- C12 heterocycloalkyl, optionally substituted C -d 2 heterocycloalkenyl, optionally substituted C a -C 18 aryl, and optionally substituted d-C B heteroaryl; and

n is an integer selected from the group consisting of: 0, 1, 2, 3, and 4. In some embodiments of the fourth aspect of the invention, R 2 is -X 2 -R 16 and R 3 is -X 3 - R 17 , wherein:

- R 16 and R 17 are the same or different and are selected from the group consisting of: H, halogen, C no alkyl, C3-C 10 cyclokalkyl, C 3 -C 10 cycloalkylmethyl, C 3 -C 10 alkene, C 3 -C 10 alkyne, aryl, C 5 -C 2 o alkaryl, fused C 5 -C 2 o aryl or alkaryl and a hydrocarbon chain containing a heterocyclic or fused ring, any of which may be optionally substituted;

- X 2 and X 3 are the same or different and are selected from the group consisting of: a bond CR 18 R 19 , O, NR 20 , and S;

- R 18 and R 19 are the same or different and are selected from the group consisting of: H, halogen, OH, N0 2 , CN, NH 2 , optionally substituted d-C 12 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -C 12 alkynyl, optionally substituted C,-Ci 0 heteroalkyl, optionally substituted C 3 -C 12 cycloalkyi, optionally substituted C 3 -C 2 cycloalkenyl, optionally substituted C 2 - C 12 heterocycloalkyl, optionally substituted C 2 -C 12 heterocycloalkenyl, optionally substituted C e -C 18 aryl, optionally substituted Qi-C 18 heteroaryl, optionally substituted CVC 12 alkyloxy, optionally substituted C 2 -C 12 alkenyloxy, optionally substituted C 2 -C 12 alkynyloxy, optionally substituted C^C^ heteroalkyloxy, optionally substituted C 3 -C 12 cycloalkyloxy, optionally substituted C 3 -C 12 cycloalkenyloxy, optionally substituted C,-C 12 heterocycloalkyloxy, optionally substituted C C 12 heterocycloalkenyloxy, optionally substituted C e -C 8 aryloxy, optionally substituted C Ci e heteroaryloxy, optionally substituted C C 12 alkylamino, SR 12 , S0 3 H, S0 2 NR 13 R 14 , S0 2 R 12 , SONR 13 R 14 , SOR 12 , COR 12 , COOH, COOR 12 , CONR 13 R 14 , NR 13 COR 12 , NR 13 COOR 12 , NR 13 S0 2 R 12 , NR 13 CONR 14 R 15 , NR 1 R 15 , and acyl; and

- R 20 is selected from the group consisting of: H, an N-protecting group, optionally substituted C r C 2 alkyl, optionally substituted C 2 -C 12 alkenyl, optionally substituted C 2 -Ci 2 alkynyl, optionally substituted Ci-C 10 heteroalkyl, optionally substituted C 3 -Ci 2 cycloalkyi, optionally substituted C 3 -C 12 cycloalkenyl, optionally substituted C Ci 2 heterocycloalkyl, optionally substituted C C 2 heterocycloalkenyl, optionally substituted C 6 -Ci 8 aryl, and optionally substituted C C 18 heteroaryl.

In some embodiments of the fourth aspect of the invention, R and R are fused to form a 5 or 6 membered cycloalkyi, heterocycloalkyl, cycloalkenyl, heterocycloalkenyl, aryl or heteroaryl ring, each of which may be optionally substituted. In some embodiments of the fourth aspect of the invention, R 2 and R 3 are each independently selected from the group consisting of: optionally substituted C 1 -C 12 alkyloxy and optionally substituted C 2 -C 12 alkynyloxy.

In some embodiments of the fourth aspect of the invention, the optionally substituted <- Ci2 alkyloxy is ( C^fluoroalkyloxy.

In some embodiments of the fourth aspect of the invention, the optionally substituted C 2 -C 12 alkynyloxy is C 2 -C 12 alkynylalkyloxy.

In some embodiments of the fourth aspect of the invention, R 2 is selected from the group consisting of: H 3 CO, F 2 HCF 2 CO, F 2 HCO, F 3 CO, and CHCCH 2 0. In some embodiments of the fourth aspect of the invention, R 3 is selected from the group consisting of. H 3 CO, F 2 HCF 2 CO, F 2 HCO, F 3 CO, and CHCCH 2 0.

In some embodiments of the fourth aspect of the invention, R 2 and R 3 together are selected from the group consisting of: OCF 2 CF 2 CO, OCH 2 CH 2 0, OCF 2 0, and OCH 2 0.

In some embodiments of the fourth aspect of the invention, T is selected from the

group consisting of: a double bond and

In some embodiments of the fourth aspect of the invention, R e is selected from the group consisting of: H and Ci-C e alkyl.

In some embodiments of the fourth aspect of the invention, R 7 is selected from the group consisting of: H, CN, N0 2 , and Ci-C e a'kyl.

In some embodiments of the fourth aspect of the invention, R is selected from the group consisting of: H and optionally substituted Ci-C e alkoxy. In some embodiments of the fourth aspect of the invention, R 4 is selected from the group consisting of: H and optionally substituted C C 6 alkoxy.

In some embodiments of the fourth aspect of the invention, R 5 is H.

In some embodiments of the fourth aspect of the invention, R 8 is selected from the group consisting of: H. halogen, COOR 12 , COR 12 , CONR 13 R 14 , NR 3 R 14 , S0 2 R 12 , and SONR 13 R 14 . In some embodiments of the fourth aspect of the invention, R 8 is selected from the group consisting of: H, halogen, COOH, CONH 2 , CONHOH, CONHCH 3 , NH 2 , S0 2 CH 3 , S0 2 NH 2 , SONHCH 3 , and SON(CH 3 ) 2 .

In some embodiments of the fourth aspect of the invention, n is 1.

In some embodiments of the fourth aspect of the invention, the R 8 group is in the position ortho to the Y group.

In some embodiments of the fourth aspect of the invention the compound of formula (IV) is selected from the group consisting of:

or a pharmaceutically acceptable salt or prodrug thereof. The present invention also provides a pharmaceutical composition including a compound of any of the aforementioned first, second, third or fourth aspects of the invention, and a pharmaceutically acceptable diluent, excipient or carrier. The present invention also provides a method of treating a disease or condition associated with fibrosis, the method including administering to a subject in need of such treatment a therapeutically effective amount of a compound of any of the aforementioned first, second, third or fourth aspects of the invention. The disease or condition may be selected from the group consisting of fibrotic skin disorders, lung disease, heart disease and kidney disease.

The present invention also provides a method of treating a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease, the method including administering to a subject in need of such treatment a therapeutically effective amount of a compound of any of the aforementioned first, second, third or fourth aspects of the invention.

The present invention also provides a method of inhibiting fibrosis in a subject, the method including administering to the subject a therapeutically effective amount of . a compound of any of the aforementioned first, second, third or fourth aspects of the invention.

The present invention also provides a use of a compound of any of the aforementioned first, second, third or fourth aspects of the invention to inhibit fibrosis.

The present invention also provides a use of a compound of any of the aforementioned first, second, third or fourth aspects of the invention in the treatment of a disease or condition associated with fibrosis. The present invention also provides a use of a compound of any of the aforementioned first, second, third or fourth aspects of the invention in the treatment of a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease. The present invention also provides a use of a compound of any of the aforementioned first, second, third or fourth aspects of the invention in the preparation of a medicament for treating a disease or condition associated with fibrosis. The present invention also provides a use of a compound of any of the aforementioned first, second, third or fourth aspects of the invention in the preparation of a medicament for treating a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease.

Detailed Description of the Invention

In this specification a number of terms are used which are well known to a skilled addressee. Nevertheless for the purposes of clarity a number of terms will be defined.

As used herein, the term "unsubstituted" means that there is no substituent or that the only substituents are hydrogen. The term "optionally substituted" as used throughout the specification denotes that the group may or may hot be further substituted or fused (so as to form a condensed polycyclic system), with one or more non-hydrogen substituent groups. In certain embodiments the substituent groups are one or more groups independently selected from the group consisting of halogen, =0, =S, -CN, -N0 2 , -CF 3 , -OCF 3 , alkyl, alkenyl, alkynyl, haloalkyl, haloalkenyl, haloalkynyl, heteroalkyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl, aryl, heteroaryl, cycloalkylalkyl, heterocycloalkylalkyl, heteroarylalkyl, arylalkyl, cycloalkylalkenyl, heterocycloalkylalkenyl, arylalkenyl, heteroarylalkenyl, cycloalkylheteroalkyl, heterocycloalkylheteroalkyl, arylheteroalkyl, heteroarylheteroalkyl, hydroxy, hydroxyalkyl, alkoxy, alkoxyalkyl, alkoxycycloaikyl, alkoxyheterocycloalkyl, alkoxyaryl, alkoxyheteroaryl, alkoxycarbonyl, alkylaminocarbonyl, alkenyloxy, alkynyloxy, cycloalkyloxy, cycloalkenyloxy, heterocycloalkyloxy, heterocycloalkenyloxy, aryloxy, phenoxy, benzyloxy, heteroaryloxy, arylalkyloxy, arylalkyl, heteroarylalkyl, cycloalkylalkyl, heterocycloalkylalkyl, arylalkyloxy, amino, alkylamino, acylamino, aminoalkyl, arylamino, sulfonylamino, sulfinylamino, sulfonyl, alkylsulfonyl, arylsulfonyl, aminosulfonyl, sulfinyl, alkylsulfmyl, arylsulfinyl, aminosulfinylaminoalkyl, -COOH, - COR 11 , -C(0)OR 11 , CONHR 1 , NHCOR 1 , NHCOOR 11 , NHCONHR 11 , C(=NOH)R 11 , -SH, -SR 1 , -OR 11 and acyl. "Alkyl" as a group or part of a group refers to a straight or branched aliphatic hydrocarbon group, preferably a C -C u alkyl, more preferably Ci-C 0 alkyl, most preferably Ci-C 6 unless otherwise noted. Examples of suitable straight and branched Ci-Ce alkyl substituents include methyl, ethyl, n-propyl, 2-propyl, n-butyl, sec-butyl, t- butyl, hexyl, and the like. The group may be a terminal group or a bridging group.

"Alkylamino" includes both mono-alkylamino and dialkylamino, unless specified. "Mono-alkylamino" means a -NH-Alkyl group, in which alkyl is as defined above. "Dialkylamino" means a -N(alkyl) 2 group, in which each alkyl may be the same or different and are each as defined herein for alkyl. The alkyl group is preferably a C C 6 alkyl group. The group may be a terminal group or a bridging group. "Arylamino" includes both mono-arylamino and di-arylamino unless specified. Mono-arylamino means a group of formula arylNH-, in which aryl is as defined herein, di-arylamino means a group of formula (aryl) 2 N- where each aryl may be the same or different and are each as defined herein for aryl. The group may be a terminal group or a bridging group.

"Acyl" means an alkyl-CO- group in which the alkyl group is as described herein. Examples of acyl include acetyl and benzoyl. The alkyl group is preferably a d-Ce alkyl group. The group may be a terminal group or a bridging group. "Alkenyl" as a group or part of a group denotes an aliphatic hydrocarbon group containing at least one carbon-carbon double bond and which may be straight or branched preferably having 2-14 carbon atoms, more preferably 2-12 carbon atoms, most preferably 2-6 carbon atoms, in the normal chain. The group may contain a plurality of double bonds in the normal chain and the orientation about each is independently E or Z. Exemplary alkenyl groups include, but are not limited to, ethenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl and nonenyl. The group may be a terminal group or a bridging group.

"Alkoxy" refers to an -O-alkyl group in which alkyl is defined herein. Preferably the alkoxy is a d-Ce alkoxy. Examples include, but are not limited to, methoxy and ethoxy. The group may be a terminal group or a bridging group.

"Alkenyloxy" refers to an -O- alkenyl group in which alkenyl is as defined herein. Preferred alkenyloxy groups are Ci-Ce alkenyloxy groups. The group may be a terminal group or a bridging group. "Alkynyloxy" refers to an -O-alkynyl group in which alkynyl is as defined herein. Preferred alkynyloxy groups are C -C 6 alkynyloxy groups. The group may be a terminal group or a bridging group. "Alkoxycarbonyl" refers to an -C(0)-0alkyl group in which alkyl is as defined herein. The alkyl group is preferably a C-,-C e alkyl group. Examples include, but not limited to, methoxycarbonyl and ethoxycarbonyl. The group may be a terminal group or a bridging group. "Alkylsulfinyl" means a -S(0)-alkyl group in which alkyl is as defined above. The alkyl group is preferably a C Ce alkyl group. Exemplary alkylsulfinyl groups include, but not limited to, methylsulfinyl and ethylsulfinyl. The group may be a terminal group or a bridging group. "Alkylsulfonyl" refers to a -S(0) 2 -alkyl group in which alkyl is as defined above. The alkyl group is preferably a C C e alkyl group. Examples include, but not limited to methylsulfonyl and ethylsulfonyl. The group may be a terminal group or a bridging group. "Alkynyl" as a group or part of a group means an aliphatic hydrocarbon group containing a carbon-carbon triple bond and which may be straight or branched preferably having from 2-14 carbon atoms, more preferably 2-12 carbon atoms, more preferably 2-6 carbon atoms in the normal chain. Exemplary structures include, but are not limited .to, ethynyl and propynyl. The group may be a terminal group or a bridging group.

"Alkylaminocarbonyl" refers to an alkylamino-carbonyl group in which alkylamino is as defined above. The group may be a terminal group or a bridging group. "Cycloalkyl" refers to a saturated or partially saturated, monocyclic or fused or spiro polycyclic, carbocycie preferably containing from 3 to 9 carbons per ring, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like, unless otherwise specified. It includes monocyclic systems such as cyclopropyl and cyclohexyl, bicyclic systems such as decalin, and polycyclic systems such as adamantane. The group may be a terminal group or a bridging group. "Cycloalkenyl" means a non-aromatic monocyclic or multicyclic ring system containing at least one carbon-carbon double bond and preferably having from 5-10 carbon atoms per ring. Exemplary monocyclic cycloalkenyl rings include cyclopentenyl, cyclohexenyl or cycloheptenyl. The cycloalkenyl group may be substituted by one or more substituent groups. The group may be a terminal group or a bridging group.

The above discussion of alkyi and cycloalkyi substituents also applies to the alkyi portions of other substituents, such as without limitation, alkoxy, alkyi amines, alkyi ketones, arylalkyl, heteroarylalkyl, alkylsulfonyl and alkyi ester substituents and the like.

"Cycloalkylalkyl" means a cycloalkyl-alkyl- group in which the cycloalkyi and alkyi moieties are as previously described. Exemplary monocycloalkylalkyl groups include cyclopropylmethyl, cyclopentylmethyl, cyclohexylmethyl and cycloheptyl methyl. The group may be a terminal group or a bridging group.

"Halogen" represents fluorine, chlorine, bromine or iodine.

"Heterocycloalkyl" refers to a saturated or partially saturated monocyclic, bicyclic, or polycyclic ring containing at least one heteroatom selected from nitrogen, sulfur, oxygen, preferably from 1 to 3 heteroatoms in at least one ring. Each ring is preferably from 3 to 10 membered, more preferably 4 to 7 membered. Examples of suitable heterocycloalkyl substituents include pyrrolidyl, tetrahydrofuryl, tetrahydrothiofuranyl, piperidyl, piperazyl, tetrahydropyranyl, morphilino, 1 ,3-diazapane, 1,4-diazapane, 1 ,4- oxazepane, and 1 ,4-oxathiapane. The group may be a terminal group or a bridging group.

"Heterocycloalkenyl" refers to a heterocycloalkyl as described above but containing at least one double bond. The group may be a terminal group or a bridging group. "Heterocycloalkylalkyl" refers to a heterocycloalkyl-alkyl group in which the heterocycloalkyl and alkyi moieties are as previously described. Exemplary heterocycloalkylalkyl groups include (2-tetrahydrofuryl)methyl, (2-tetrahydrothiofuranyl) methyl. The group may be a terminal group or a bridging group. "Heteroalkyl" refers to a straight- or branched-chain alkyi group preferably having from 2 to 14 carbons, more preferably 2 to 10 carbons in the chain, one or more of which has been replaced by a heteroatom selected from S, O, P and N. Exemplary heteroalkyls include alkyl ethers, secondary and tertiary alkyl amines, amides, alkyl sulfides, and the like. The group may be a terminal group or a bridging group. As used herein reference to the normal chain when used in the context of a bridging group refers to the direct chain of atoms linking the two terminal positions of the bridging group.

"Aryl" as a group or part of a group denotes (i) an optionally substituted monocyclic, or fused polycyclic, aromatic carbocycle (ring structure having ring atoms that are all carbon) preferably having from 5 to 12 atoms per ring. Examples of aryl groups include phenyl, naphthyl, and the like; (ii) an optionally substituted partially saturated bicyclic aromatic carbocyclic moiety in which a phenyl and a C5.7 cycloalkyl or Cs-7 cycloalkenyl group are fused together to form a cyclic structure, such as tetrahydronaphthyl, indenyl or indanyl. The group may be a terminal group or a bridging group. "Arylalkenyl" means an aryl-alkenyl- group in which the aryl and alkenyl are as previously described. Exemplary arylalkenyl groups include phenylallyl. The group may be a terminal group or a bridging group.

"Arylalkyl" means an aryl-alkyl- group in which the aryl and alkyl moieties are as previously described. Preferred arylalkyl groups contain a Ci -5 alkyl moiety. Exemplary arylalkyl groups include benzyl, phenethyl and naphthelenemethyl. The group may be a terminal group or a bridging group.

"Heteroaryl" either alone or part of a group refers to groups containing an aromatic ring (preferably a 5 or 6 rnembered aromatic ring) having one or more heteroatoms as ring atoms in the aromatic ring with the remainder of the ring atoms being carbon atoms. Suitable heteroatoms include nitrogen, oxygen and sulphur. Examples of heteroaryl include thiophene, benzothiophene, benzofuran, benzimidazole, benzoxazole, benzothiazole, benzisothiazole, naphtho[2,3-b]thiophene, furan, isoindolizine, xantholene, phenoxatine, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indole, isoindole, 1H-indazole, purine, quinoline, isoquinoline, phthalazine, naphthyridine, quinoxaline, cinnoline, carbazole, phenanthridine, acridine, phenazine, thiazole, isothiazole, phenothiazine, oxazole, isooxazole, furazane, phenoxazine, 2-, 3- or 4- pyridyl, 2-, 3-, 4-, 5-, or 8- quinolyl, 1-, 3-, 4-, or 5- isoquinolinyl 1-, 2-, or 3- indolyl, arid 2-, or 3-thienyl. The group may be a terminal group or a bridging group. "Heteroarylalkyl" means a heteroaryl-alkyl group in which the heteroaryl and alkyl moieties are as previously described. Preferred heteroarylalkyl groups contain a lower alkyl moiety. Exemplary heteroarylalkyl groups include pyridylmethyl. The group may be a terminal group or a bridging group.

"Lower alkyl" as a group means unless otherwise specified, an aliphatic hydrocarbon group which may be straight or branched having 1 to 6 carbon atoms in the chain, more preferably 1 to 4 carbons such as methyl, ethyl, propyl (n-propyl or isopropyl) or butyl (n-butyl, isobutyl or tertiary-butyl). The group may be a terminal group or a bridging group.

As would be understood by the skilled person, throughout the synthesis of the compounds of Formulae (I), (II), (III) and (IV) it may be necessary to employ a protecting group on the amino group and/or on the carboxyl group in order to reversibly preserve a reactive amino or carboxyl functionality while reacting other functional groups on the compound. In such a case, the free amino group and/or the free carboxyl groups of the compounds of Formulae (I), (II), (III) and (IV) can be liberated either by deprotection of the amino group followed by deprotection of the acid moieties or vice versa.

Examples of suitable amino protecting groups that may be used include formyl, trityl, phthalimido, trichloroacetyl, chloroacetyl, bromoacetyl, iodoacetyl, and urethane-type blocking groups such as benzyloxycarbonyl ('CBz'), 4-phenylbenzyloxycarbonyl, 2- methylbenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 4-fluorobenzyloxycarbonyl, 4- chlorobenzyloxycarbonyl, 3-chlorobenzyloxycarbonyl, 2-chlorobenzyloxycarbonyl, 2,4- dichlorobenzyloxycarbonyl, 4-bromobenzyloxycarbonyl, 3-bromobenzyloxycarbonyl, 4- nitrobenzyloxycarbonyl, 4cyanobenzyloxycarbonyl, t-butoxycarbonyl ( Boc'), 2-(4- xenyl)-isopropoxycarbonyl, 1,1 -diphenyleth-1 -yloxycarbonyl, 1 , 1 -diphenylprop-1 - yloxycarbonyl, 2-phenylprop-2-yloxycarbonyl, 2-(p-toluyl)-prop-2-yloxycarbonyl, cyclopentanyloxy-carbonyl, 1-methylcyclopentanyloxycarbonyl, cyclohexanyloxycarbonyl, 1-methylcyclohexanyloxycarbonyl, 2- methylcyclohexanyloxycarbonyl, 2-(4-toluylsulfono)-ethoxycarbonyl, 2-

(methylsulfono)ethoxycarbonyl, 2-(triphenylphosphino)-ethoxycarbonyl, fluorenylmethoxycarbonyl ("FMOC"), 2-(trimethylsilyl)ethoxycarbonyl, allyloxycarbonyl, 1-(trimethylsilylmethyl)prop-1-enyloxycarbonyl, 5-benzisoxalylmethoxycarbonyl, 4- acetoxybenzyloxycarbonyl, 2,2,2-trichloroethoxycarbbnyl, 2-ethynyl-2-propoxycarbonyl, cyclopropylmethoxycarbonyl, 4-(decycloxy)benzyloxycarbonyl, isobornyloxycarbonyl, 1- piperidyloxycarbonlyl and the like; benzoylmethylsulfono group, 2-nitrophenylsulfenyl, diphenylphosphine oxide, and the like. The actual amino protecting group employed is not critical so long as the derivatised amino group is stable to the condition of subsequent reaction(s) and can be selectively removed as required without substantially disrupting the remainder of the molecule including any other amino protecting group(s). Preferred amino-protecting groups are t-butoxycarbonyl (Boc), and benzyloxycarbonyl (Cbz). Further examples of these groups are found in: Greene, T. W. and Wuts, P. G. M., Protective Groups in Organic Synthesis, Second edition; Wiley-lnterscience: 1991 ; Chapter 7; McOmie, J. F. W. (ed.), Protective Groups in Organic Chemistry, Plenum Press, 1973; and Kocienski, P. J., Protecting Groups, Second Edition, Theime Medical Pub., 2000.

Examples of carboxyl protecting groups that may be used include methyl, ethyl, n- propyl, i-propyl, p-nitrobenzyl, p-methylbenzyl, p-methoxybenzyl, 3,4-dimethoxybenzyl, 2,4-dimethoxybenzyl, 2,4,6-trimethoxybenzyl, 2,4,6-trimethylbenzyl, pentamethylbenzyl, 3,4-methylenedioxybenzyl, benzhydryl, 4,4'-dimethoxybenzhydryl, 2,2'4,4 , -tetramethoxybenzhydryl, t-butyl, t-amyl, trityl, 4-methoxytrityl, 4,4'- dimethoxytrityl, 4,4,'4"-trimethoxytrityl, 2-phenylprop-2-yl, trimethylsilyl, t- butyldimethylsilyl, phenacyl, 2,2,2-trichloroethyl, -(di(n-butyl)methylsilyl)ethyl, p- toluenesulfonoethyl, 4-nitrobenzylsulfonoethyl, allyl, cinnamyl, 1- (trimethylsilylmethyl)prop-1-en-3-yl, and the like. Preferred carboxyl protecting groups are methyl and t-butyl. Further examples of these groups are found in: Greene, T. W. and Wuts, P. G. M., Protective Groups in Organic Synthesis, Second edition; Wiley- lnterscience: 1991 ; McOmie, J. F. W. (ed.), Protective Groups in Organic Chemistry, Plenum Press, 1973; and Kocienski, P. J., Protecting Groups, Second Edition, Theime Medical Pub., 2000.

It is understood that included in the family of compounds of Formulae (I), (II), (ill) and (IV) are isomeric forms including diastereoisomers, enantiomers, taUtomers, and geometrical isomers in "E" or "Z" configurational isomer or a mixture of E and Z isomers. It is also understood that some isomeric forms such as diastereomers, enantiomers, and geometrical isomers can be separated by physical and/or chemical methods and by those skilled in the art. Some of the compounds of the disclosed embodiments may exist as single stereoisomers, racemates, and/or mixtures of enantiomers and /or diastereomers. All such single stereoisomers, racemates and mixtures thereof, are intended to be within the scope of the subject matter described and claimed.

Some of the compounds of the disclosed embodiments are substituted cyclopropanes

having the general The structure shown is intended to include isomeric forms of the cyclopropanes including diastereoisomers and enantiomers.

Additionally, Formulae (I) to (IV) are intended to cover, where applicable, solvated as well as unsolvated forms of the compounds. Thus, each formula includes compounds having the indicated structure, including the hydrated as well as the non-hydrated forms.

In addition to compounds of Formulae (I) to (IV), the compounds of the various embodiments include pharmaceutically acceptable salts, prodrugs, N-oxides and active metabolites of such compounds, and pharmaceutically acceptable salts of such metabolites.

The term "pharmaceutically acceptable salts" refers to salts that retain the desired biological activity of the above-identified compounds, and include pharmaceutically acceptable acid addition salts and base addition salts. Suitable pharmaceutically acceptable acid addition salts of compounds of Formulae (I) to (IV) may be prepared from an inorganic acid or from an organic acid. Examples of such inorganic acids are hydrochloric, sulfuric, and phosphoric acid. Appropriate organic acids may be selected from aliphatic, cycle-aliphatic, aromatic, heterocyclic carboxylic and sulfonic classes of organic acids, examples of which are formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, fumaric, maleic, alkyl sulfonic, arylsulfonic. Suitable pharmaceutically acceptable base addition salts of compounds of Formulae (I) to (IV) include metallic salts made from lithium, sodium, potassium, magnesium, calcium, aluminium, and zinc, and organic salts made from organic bases such as choline, diethanolamine, morpholine. Other examples of organic salts are: ammonium salts, quaternary salts such as tetramethylammonium salt; amino acid addition salts such as salts with glycine and arginine. Additional information on pharmaceutically acceptable salts can be found in Remington's Pharmaceutical Sciences, 19th Edition, Mack Publishing Co., Easton, PA 1995. In the case of agents that are solids, it is understood by those skilled in the art that the inventive compounds, agents and salts may exist in different crystalline or polymorphic forms, all of which are intended to be within the scope of the present invention and specified formulae.

"Prodrug" means a compound which is convertible in vivo by metabolic means (e.g. by hydrolysis, reduction or oxidation) to a compound of Formulae (I) to (IV). For example an ester prodrug of a compound of Formulae (I) to (IV) containing a hydroxyl group may be convertible by hydrolysis in vivo to the parent molecule. Suitable esters of compounds of Formulae (I) to (IV) containing a hydroxyl group, are for example acetates, citrates, lactates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis-(3-hydroxynaphthoates, gestisates, isethionates, di-p-toluoyltartrates, methanesulphonates, ethanesulphonates, benzenesulphonates, p-toluenesulphonates, cyclohexylsulphamates and quinates. As another example an ester prodrug of a compound of Formulae (I) to (IV) containing a carboxy group may be convertible by hydrolysis in vivo to the parent molecule. (Examples of ester prodrugs are those described by F.J. Leinweber, Drug Metab. Res., 18:379, 1987). The terms "treating", "treat", or "treatment" refer generally to amelioration or elimination of a named condition once the condition has been established. The term "prophylaxis" refers generally to treatment to prevent the onset of a named condition or of a process that can lead to the condition ("primary" prophylaxis), or the recurrence of symptoms of a condition.

The term "subject" refers generally to any warm blooded animal such as; but not limited to, a mouse, guinea pig, dog, horse, or human. In an embodiment, the subject is human. The term "therapeutically effective amount" or "effective amount" is an amount sufficient to effect beneficial or desired clinical results. An effective amount can be administered in one or more administrations. An effective amount is typically sufficient to palliate, ameliorate, stabilize, reverse, slow or delay the progression of the disease state. The term "pharmaceutically acceptable" refers generally to a substance or composition that is compatible chemically and/or toxicologically with the other ingredients including a formulation, and/or the subject being treated.

The term "compounds of the present invention" (unless specifically identified otherwise) refers generally to compounds, prodrugs thereof, pharmaceutically acceptable salts of the compounds and/or prodrugs, and hydrates or solvates of the compounds, salts, and/or prodrugs, as well as all stereoisomers (including diastereoisomers and enantiomers), tautomers and isotopically labelled compounds. The compounds of the present invention may exist, in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms.

The term "derivative thereof when used in reference to compounds of the present invention refers generally to prodrugs, pharmaceutically acceptable salts of the compounds and or prodrugs, and hydrates or solvates of the compounds, salts, and/or prodrugs.

Compounds of the present invention include compounds of Formula (I)

Formula (I) wherein W, A, Y, Z 1 -Z 2 , p, s, q, r, X 1 , X 2 , t, D, R 1 , R 2 , R 3 , R 4 , R 5 , R 8 , R 7 , R 8 , R 9 , R 10 , R 11 R 12 , R 13 , R 14 R 15 , R 8 , R 17 , m, and n are as previously defined in relation to Formula (I).

Compounds of the present invention also include compounds of Formula (II)

Formula (II) wherein A, Y, p, s, q, r, X 1 , X 2 , D, R\ R 2 , R 3 , R 4 , R 5 , R e , R 7 , R 8 , R 9 , R 10 , R 11 , R R 14 , R 5 , R 16 , R 17 , m, and n are as previously defined in relation to Formula (II).

Compounds of the present invention also include compounds of Formula (III)

wherein A, Y, p, s, T, X 1 , X 2 , Z\ Z 2 , Z 3 , Z 4 , R\ R 2 , R 3 , R 4 , R s , R e , R 7 , R 8 , R B , R 10 , R 11 , R 12 , R 13 , R 15 , R 18 , R 17 , R 18 , R 19 , R 20 and R 21 and n are as previously defined in relation to Formula (III).

Compounds of the present invention also include compounds of Formula (IV)

Formula (IV) wherein: Z-X-Y, A, T, Z\ Z 2 , Z 3 , Z 4 , R 1 , R 2 , R 3 , R 4 , R 5 , R 8 , R 7 , R 8 , R 9 , R 10 , R", R 12 , R 13 , R 14 , R 15 and n are as previously defined in relation to Formula (IV).

Specific compounds of the invention include the following:

R = S0 2 Me, S0 2 NH 2 , SONHMe, SONM¾ R = NH 2 , CONH 2 , CONHMe, CONHOH

U2010/001398

T U2010/001398

or a pharmaceutically acceptable salt or prodrug thereof.

It will be evident from the foregoing description that compounds of the present invention are fused ring analogues of tranilast (2-{[(2E)-3-(3,4-dimethoxyphenyl)prop-2- enoyl]amino}benzoic acid). As such, the compounds of the invention may have the ability to inhibit ERK phosphorylation and, therefore, act as an antifibrotic agent when administered to a subject. However, the compounds of the present invention are not limited to antifibrotic agents that act via inhibition of ERK phosporylation and it is possible for them to have antifibrotic activity via a mechanism that does not involve inhibition of ERK phosphorylation.

Accordingly the compounds of the invention may find a multiple number of applications in which their ability to prevent, ameliorate or inhibit fibrosis can be utilised. For example compounds of Formulae (I), (II), (III) and (IV) may be used to treat a disease or condition associated with fibrosis or characterised by inflammation and/or a benign or malignant neoplastic disease;

The disease or condition associated with fibrosis may be selected from fibrotic skin disorders, such as keloids, hypertrophic scars and scleroderma; lung disease, such as pulmonary fibrosis; heart disease, such as heart failure due to ischaemic heart disease, valvular heart disease and hypertensive heart disease, diabetic cardiomyopathy and hypertension; and kidney disease, such as progressive kidney disease, cirrhosis of the liver, glomerulonephritis and diabetic nephropathy. In specific embodiments, the disease or condition is diabetic heart disease, diabetic kidney disease, or diabetic cardiomyopathy.

Diabetic cardiomyopathy refers to any one or more cardiac pathology and/or dysfunction in a subject, which is a complication of either Type I or Type II diabetes in the subject. The diabetes may be symptomatic or asymptomatic. Cardiac pathology which is characteristic of diabetic cardiomyopathy includes myocellular hypertrophy, myocardial fibrosis, and in some cases left ventricular hypertrophy. The pathologies which are contemplated arise independently from complications arising from coronary artery disease, although both diabetic complications and coronary artery complications may be present in the same subject. Diastolic dysfunction, such as an impairment in early diastolic filling, a prolongation of isovolumetric relaxation and increased atrial filling is also characteristic of diabetic cardiomyopathy, and may be identified using Doppler methods such as Doppler 2-dimensional echocardiography (for example Redford MM et a/., JAMA (2003) 289:194-203) or radionuclide imaging for early or mild dysfunction and by standard echocardiograph testing for more severe dysfunction.

Cardiac fibrosis refers to the formation of fibrous tissue, including cellular and extracellular components, in the lining and muscle of the heart. If present in sufficient quantities, the fibrous tissue will result in a decrease in the contractility of one or more regions of the heart, resulting in functional deficit in cardiac output.

Alternatively, or in addition, it is also anticipated that the compounds will be useful in the treatment of a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease. The disease or condition characterised by inflammation may be selected from allergic rhinitis, bronchial asthma, rheumatoid arthritis, multiple sclerosis, type I and type II diabetes, systemic lupus, erythematosis, transplant rejection and inflammatory bowel disease. The benign or malignant neoplastic disease may be any such disease known to the skilled person.

The compounds may be used in the preparation of a medicament for treating a disease or condition associated with fibrosis. Alternatively, the compounds of the invention may be used in the preparation of a medicament for treating a disease or condition characterised by inflammation and/or a benign or malignant neoplastic disease.

Administration of compounds of Formulae (I), (II), (III) and (Ν)Λο humans can be by any of the accepted modes for enteral administration such as oral or rectal, or by parenteral administration such as subcutaneous, intramuscular, intravenous and intradermal routes. Injection can be bolus or via constant or intermittent infusion. The active compound is typically included in a pharmaceutically acceptable carrier or diluent and in an amount sufficient to deliver to the patient a therapeutically effective dose. In using the compounds they can be administered in any form or mode which makes the compound bioavailable. One skilled in the art of preparing formulations can readily select the proper form and mode of administration depending upon the particular characteristics of the compound selected, the condition to be treated, the stage of the condition to be. treated and other relevant circumstances. See Remingtons Pharmaceutical Sciences, 19 th edition, Mack Publishing Co. (1995) for further information. The compounds can be administered alone or in the form of a pharmaceutical composition in combination with a pharmaceutically acceptable carrier, diluent or excipient. The compounds may be administered as the compounds themselves or in the form of their pharmaceutically acceptable salts or derivatives.

The compounds are, however, typically used in the form of pharmaceutical compositions which are formulated depending on the desired mode of administration. As such in a further embodiment the present invention provides a pharmaceutical composition including a compound of Formula (I), (II), (III) or (IV) and a pharmaceutically acceptable carrier, diluent or excipient. The compositions are prepared in manners well known in the art.

The invention in other embodiments provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. In such a pack or kit can be found a container having a unit dosage of the agent(s). The kits can include a composition comprising an effective agent either as concentrates (including lyophilized compositions), which can be diluted further prior to use or they can be provided at the concentration of use, where the vials may include one or more dosages. Conveniently, in the kits, single dosages can be provided in sterile vials so that the physician can employ the vials directly, where the vials will have the desired amount and concentration of agent(s). Associated with such container(s) can be various written materials such as instructions for use, or a notice in the form prescribed by a govemmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.

The compounds may be used or administered in combination with one or more additional drug(s) for the treatment of the disorder/diseases mentioned. The components can be administered in the same formulation or in separate formulations. If administered in separate formulations the compounds of the invention may be administered sequentially or simultaneously with the other drug(s).

In addition to being able to be administered in combination with one or more additional drugs, the compounds may be used in a combination therapy. When this is done the compounds are typically administered in combination with each other. Thus one or more of the compounds of the invention may be administered either simultaneously (as a combined preparation) or sequentially in order to achieve a desired effect. This is especially desirable where the therapeutic profile of each compound is different such that the combined effect of the two drugs provides an improved therapeutic result.

Pharmaceutical compositions of this invention for parenteral injection comprise pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils (such as olive oil), and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. These compositions may also contain adjuvants such as preservative, wetting agents, emulsifying agents, and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents such as sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents that delay absorption such as aluminium monostearate and gelatin.

If desired, and for more effective distribution, the compounds can be incorporated into slow release or targeted delivery systems such as polymer matrices, liposomes, and microspheres.

The injectable formulations can be sterilised, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilising agents in the form of sterile solid compositions that can be dissolved or dispersed in sterile water or other sterile injectable medium just prior to use.

Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.

Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes.

The active compounds can also be in microencapsulated form, if appropriate, with one or more of the above-mentioned excipients.

Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the , art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethyl formamide, oils (in particular, cottonseed, groundnut, com, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavouring, and perfuming agents. Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminium metahydroxide, bentonite, agar-agar, and tragacanth, and mixtures thereof. Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at room temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.

Dosage forms for topical administration of a compound of this invention include powders, patches, sprays, ointments and inhalants. The active compound is mixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives, buffers, or propellants which may be required.

The amount of compound administered will preferably treat and reduce or alleviate the condition; A therapeutically effective amount can be readily determined by an attending diagnostician by the use of conventional techniques and by observing results obtained under analogous circumstances. In determining the therapeutically effective amount a number of factors are to be considered including but not limited to, the species of animal, its size, age and general health, the specific condition involved, the severity of the condition, the response of the patient to treatment, the particular compound administered, the mode of administration, the bioavailability of the preparation administered, the dose regime selected, the use of other medications and other relevant circumstances.

A typical dosage will be a range from about 0.01 to 1000 mg per kilogram of body weight per day. Small doses (0.01-1 mg/kg per day) may be administered initially, followed by increasing doses up to about 1000 mg/kg per day. In the event that the response in a subject is insufficient at such doses, even higher doses (or effective higher doses by a different, more localised delivery route) may be employed to the extent patient tolerance permits. A more preferred dosage will be in the range from 0.1 to 300 mg per kilogram of body weight per day, more preferably from 0.1 to 100 mg per kilogram of body weight per day. A suitable dose can be administered in multiple sub- doses per day. The compounds may be prepared using the reaction routes and synthesis schemes as described below, employing the techniques available in the art using starting materials that are commercially available or can be synthesised using known procedures or adaptations thereof. Whilst the preparation of particular compounds is outlined below, the skilled person will also recognise that the chemical reactions described may be readily adapted to prepare a number of other agents of the various embodiments. For example, the synthesis of non-exemplified compounds may be successfully performed by modifications apparent to those skilled in the art, e.g. by appropriately protecting interfering groups, by changing to other suitable reagents known in the art, or by making routine modifications of reaction conditions. A list of suitable protecting groups in organic synthesis can be found in T.W. Greene's Protective Groups in Organic Synthesis, 3 rd Edition, John Wiley & Sons, 1991. Alternatively, other reactions disclosed herein or known in the art will be recognized as having applicability for preparing other compounds of the various embodiments.

Reagents useful for synthesizing compounds may be obtained or prepared according to techniques known in the art.

Compounds of Formula (I) may be synthesized by N-acylation of aminobenzenes to acid chlorides which can be derived from the carboxylic acid.

carboxylic acid tranilast

precursor derivative

The route used to synthesise the carboxylic acid precursors will depend on whether the precursor is an 0-, S-, C- or N-based derivative.

C-based: Carboxylic acid, ester and aldehyde precursor syntheses are shown below. The ester or aldehyde precursors produced can be readily converted to the corresponding carboxylic acid using standard hydrolysis or oxidation reactions, respectively.

Synth. Comm. 39 (3), 506-515, 2009 J. Org. Chem. 69 (14), 4781-4787, 2004.

Bioorg. Med. Chem. 12 (16), 4313-4336, 2004.

Tet. Lett. 46 (33), 5483-5486, 2005

O-based: Carboxylic acid, ester and aldehyde precursor syntheses are shown below. The ester or aldehyde precursors produced can be readily converted to the corresponding carboxylic acid using standard hydrolysis or oxidation reactions, respectively.

WO2007/118137 page 180

Bioorg. Med. Chem. Lett. 18 (20), 5591-5593, 2008.

Synth Comm 31 (8), 1233-1235, 2001.

Tet. Lett 44 (9), 1755-1758, 2003

J. Med. Chem. 45 (10), 2040-2055, 2002.

Org Lett. 4 (14), 2317-2320, 2002.

S-based: Ester syntheses are shown below. These methyl and ethyl ester precursors can be readily converted to the corresponding carboxylic acid in one step.

J. Het. Chem. 45 (1), 235-240, 2008

J. HeL Chem. 25 (3), 711-714, 1988 (French).

Synthesis (1 ), 56-58, 1978.

WO2007/118137 page 204

(German)

N-based Ester syntheses are shown below. These methyl and ethyl ester precursors can be readily converted to the corresponding carboxylic acid in one step.

J. Med. Chem. 51 (6), 1849-1860, 2008.

WO2005/030704 page 248

POBr, K 2 C0 3

Tet. Lett 48 (40), 713707139, 2007

PMHS Pd(OH)2/C

Tet. Lett 48 (15), 2765-2768, 2007.

OBC 4 (21), 3960-3965, 2006.

Compounds of Formula (II) may be synthesized by N-acylation of aminobenzenes with acid chlorides which can be derived from the carboxylic acid.

carboxylic acid tranilast

precursor derivative

Synthetic schemes to synthesize 5,6-heterocyclic carboxylic acid precursors are shown below:

Perkin Transactions 1 , 3073-3079, 2001.

Letters in Organic Chemistry 4, 56-57, 2006.

Synth. Comm. 31 (5), 741-748, 2001. WO 2002/059108 page 292

Synthetic schemes to synthesize 6,6-heterocyclic carboxylic acid precursors are shown below:

P 4 0 io toluene

Chemistry: A European Journal 10 (11), 2722-2731, 2004.

Compounds of Formula (III) may be synthesized by N-acylation of commercially available aryl amines/amides to the corresponding cinnamoyi chloride, derived from the cinnamic acid:

A list of commercially available precursors and their corresponding CAS numbers is shown below. Each of these can be N-acylated by reacting with a corresponding cinnamoyi chloride.

615-16-7 149-3C 2382-96-9 583-39-1

Compounds of Formula (IV) may be synthesized by condensing 3-methyl-1H- quinoxalin-2-one, 2-methyl-4H-3, 1 -benzoxazin-4-one or 2-methylr3H-quinazolin-4-one with a substituted benzaldehyde to provide fused heterocyclic tranilast derivatives. The intermediates can also be synthesized as shown below:

Molecules 5 (6), 864-873, 2000.

Journal of Agricultural and Food Chemistry 51 (3), 594-600, 2003.

1769-24-0

Bioorg Med Chem 17 (1), 119-132, 2009.

Acta Poloniae Pharmaceutica 60 (4), 275-279, 2003.

Substituted benzoxazoles, benzothiazoles, and benzimidazoles can be obtained by reaction of aldehydes with 2-aminophenol, 2-aminothiophenol and o- phenylenediamine, with 4-methoxy-TEMPO radical as the catalyst:

The compounds of Formulae (I) to (IV) and intermediates in their synthesis can be isolated from a reaction mixture using standard work-up and purification procedures. Suitable procedures include solvent extraction, chromatography (thin or thick layer chromatography, HPLC, flash chromatography, MPLC, etc.), recrystallisation etc.

The present invention includes salts of the compounds of Formulae (I) to (IV). The salts may serve as intermediates in the purification of compounds or in the preparation of other, for example pharmaceutically acceptable, acid addition salts, or they may be useful for identification, characterisation or purification. The salts can exist in conjunction with the acidic or basic portion of the molecule and can exist as acid addition, primary, secondary, tertiary, or quaternary ammonium, alkali metal, or alkaline earth metal salts. Generally, acid addition salts are prepared by the reaction of an acid with compounds of Formulae (I) to (IV). The alkali metal and alkaline earth metal salts are generally prepared by the reaction of the hydroxide form of the desired metal salt with compounds of Formulae (I) to (IV). Acid addition salts are preferably the pharmaceutically acceptable, non-toxic addition salts with suitable acids, such as those with inorganic acids, for example hydrochloric, hydrobromic, nitric, sulphuric or phosphoric acids, or with organic acids, such as organic carboxylic acids, for example, glycollic, maleic, hydroxymaleic, fumaric, malic, tartaric, citric, salicyclic, o-acetoxybenzoic, or organic sulphonic, 2-hydroxyethane sulphonic, toluene-p-sulphonic, or naphthalene-2-sulphonic acid.

The present invention also includes esters of the compounds of Formulae (I) to (IV), such esters being for example aliphatic esters such as alkyl esters. The esters of the compounds of Formulae (I) to (IV) may be pharmaceutically acceptable metabolically labile esters. These are ester derivatives of compounds of Formulae (I) to (IV) that are hydrolysed in vivo to afford the compound of Formulae (I) to (IV) and a pharmaceutically acceptable alcohol. Examples of metabolically labile esters include esters formed with alkanols in which the alkanol moiety may be optionally substituted by an alkoxy group, for example methanol, ethanol, propanol and methoxyethanol.

The compounds of the various embodiments may be prepared using the reaction routes and synthesis schemes as described above, employing the techniques available in the art using starting materials that are readily available. The person skilled in the art will recognise that the chemical reactions described may be readily adapted to prepare a number of other compounds. For example, the synthesis of non-exemplified compounds may be successfully performed by modifications apparent to those skilled in the art, e.g. by appropriately protecting interfering groups, by changing to other suitable reagents known in the art, or by making routine modifications of reaction conditions. A list of suitable protecting groups in organic synthesis can be found in T.W. Greene's Protective Groups in Organic Synthesis, 3 rd Edition, John Wiley & Sons, 1991. Reagents useful for synthesizing compounds may be obtained or prepared according to techniques known in the art. The anti-fibrotic effects of compounds of Formulae (I) to (IV) can be tested using any of the following methods:

(i) In a renal cell line by measuring proline incorporation after transforming growth factor-β stimulation;

(ii) Matrix synthesis may be stimulated by platelet derived growth factor (PDGF). Accordingly, mesangial cells incubated with PDGF can be used to demonstrate proline incorporation, which is an indicator of matrix synthesis and thereby a model for fibrosis; or (iii) Matrix synthesis may be stimulated by both angiotensin II or transforming growth factor beta (TGF-β). Accordingly, neonatal cardiac fibroblasts incubated with angiotensin II or TGF-β can be used to demonstrate proline incorporation, which is an indicator of matrix synthesis and thereby a model for fibrosis.

Brief Description of the Drawings

Figure 1: Average % inhibition of TGF-β stimulated proline incorporation by compound FT98 at 10μΜ, 30μΜ and 100μΜ (SEM).

Figure 2: Average % inhibition of TGF-β stimulated proline incorporation by compound FT108 at 10μΜ, 30μΜ and 100μΜ (SEM). Figure 3: Average % inhibition of TGF-β stimulated proline ίηοοφθΓβΰοη by compound FT109 at 10μΜ, 30μΜ and 100μΜ (SEM).

Figure 4: Average % inhibition of TGF-β stimulated proline incorporation by compound FT113 at 10μΜ, 30μΜ and 100μΜ (SEM).

Figure 5: Average % inhibition of TGF-β stimulated proline incx^oration by compound FT121 at 10μΜ, 30μΜ and 10ΟμΜ (SEM).

Figure 6: Average % inhibition of TGF-β stimulated proline incorporation by compound FT122 at 10μΜ, 30μ and 100μΜ (SEM).

Figure 7: Average % inhibition of TGF-β stimulated proline ΐηοοφθΓβΑοη by compound FT126 at 10μΜ, 30μΜ and 100μΜ (SEM). Figure 8: Average % inhibition of TGF-β stimulated proline ΐηοοφθΓθ οη by compound FT128 at 10μΜ, 30μΜ and 100μΜ (SEM).

Figure 9: Average % inhibition of TGF-β stimulated proline ϊηοοφθΓβϋοη by compound FT129 at 10μΜ, 30μΜ and 100μΜ (SEM).

Figure 10: Average % inhibition of TGF-β stimulated proline incorporation by compound FT130 at 10μΜ, 30μΜ and 100μΜ (SEM). Figure 11: Average % inhibition of TGF-β stimulated proline incorporation by compound FT132 at 10μΜ, 30μΜ and 100μΜ (SEM). An average % inhibition of greater than 100% indicates cell stress or death in figures 5, 6 and 9.

Examples of materials and methods for use with the compounds of the present invention will now be provided. In providing these examples, it is to be understood that the specific nature of the following description is not to limit the generality of the above description.

Examples

Experimental Electrospray ionization (ESI) high resolution mass spectra (HRMS) were obtained on a Finnigan hybrid LTQ-FT mass spectrometer (Thermo Electron Corp.). Proton nuclear magnetic resonance ( 1 H NMR) and proton decoupled carbon nuclear magnetic resonance ( 13 C NMR) spectra were obtained on Unity 400, Innova 400 or Innova 500 instruments (Melbourne, Australia) operating at 400 or 500 MHz for 1 H and at 100 or 125 MHz for 13 C. All signals were referenced to solvent peaks (CDCI 3 :.7.26 ppm for and 77.0 ppm for 13 C; DMSO-d 6 : 2.49 ppm for 1 H and 39.5 ppm for 13 C). Infrared (IR) spectra were obtained using a PerkinElmer Spectrum One FT-IR spectrometer with zinc selenide/diamond Universal ATR Sampling Accessory. Melting points were obtained using a Reichert-Jung hot stage apparatus and are corrected. Analytical thin layer chromatography (TLC) was conducted on 2 mm thick silica gel GF 2 54- Compounds were visualised with solutions of 20% w/w phosphomolybdic acid in ethanol, 20% w w potassium permanganate in water or under UV (365 nm). Flash chromatography was performed with Merck Silica Gel 60. Petrol refers to the fraction boiling at 40-60 °C. All other reagents were used as received. Synthesis of Compounds of Formulae (\) to (IV) (E)-3-[2-(3,4-Dimethoxyphenyl)ethenyl]-4H-1,2,4-benzothiadia z ^

2-Aminobenzenesulfonamide (0.25 g, 1.4 mmol) was added to a suspension of (£)-3-(3,4-dimethoxypheny0acrylic acid (0.25 g, 1.2 mmol), HBTU (0.55 g, 1.4 mmol) and NEU (0.67 mL, 4.8 mmol) in MeCN (5 mL) and the reaction was stirred at rt for 16 h. Extra NEt 3 (0.67 mL, 4.8 mmol) in MeCN (5 mL) was added and the reaction was stirred at rt for 64 h. The solution was acidified and the precipitate was collected by filtration. Hot EtOH (15 mL) was added to the crude product and the undissolved solid was collected by hot filtration, providing (E)-3- [2-(3,4-dimethoxyphenyl)ethenyl]-4H-1 ,2,4-benzothiadiazine-1 , 1 -dioxide (58 mg, 14%) as a colourless crystalline solid; mp 276-279 °C; δ Η (500 MHz, DMSO-de) 3.81 (s, 3H, OCH 3 ), 3.83 (s, 3H, OCH 3 ), 6.75 (d, J = 15.6 Hz, 1H, CH=CHCO), 7.05 (d, ν7 5 ·, 6 · = 8.0 Hz, Η, H5'), 7.29 (d, J 5 ., 6 - = 8.0 Hz, 1H, HQ'), 7.31 (s, 1H, H2'), 7.38 (d, 5 .6 = 8.0 Hz, 1H, H5), 7.45 (t, J 5 , 6 = J 6 = 8.0 Hz, 1H, HQ), 7.68 (t, J 6 ,7 = = 8.0 Hz, 1H, 7), 7.80 (d, J 7 .e = 8.0 Hz, 1H, HS), 7.82 (d, J = 15.6 Hz, 1H, CH=CHCO), 12.13 (s, 1H, NH); 6 C (125 MHz, DMSO-d 6 ) 55.4, 55.6, 110.3, 111.8, 116.6, 117.6, 122.0, 122.7, 123.3, 126.3, 126.9, 133.0, 135.2, 142.2, 149.0, 151.1, 153.5; HRMS (ESI) calculated for C 17 H 16 N 2 0 S [M+Hf 345.0904, found 345.0900; v max 756, 1134, 1263, 1513, 1568, 1600, 3024, 3117, 3163, 3200 cm- 1 .

(E)-2-(3, 4-Dimethoxystyryl)-4H-pyrido[2, 3-d][1, 3]pxazin-4-one (FT102)

FT102 A suspension of (£)-3-(3,4-dimethoxyphenyl)acrylic acid (0.25 g, 1.2 mmol) in CH2CI2 (5 ml_) was treated with oxalyl chloride (0.41 mL, 4.8 mmol). The solution was stirred at rt for 1 h and the solvent was removed under reduced pressure to give the acid chloride as a yellow solid. 3-Aminonicotinic acid (0.25 g, 1.4 mmol) was added to a solution of the acid chloride (1.2 mmol) in pyridine (2.0 mL) and the suspension was stirred at rt for 7 d. The solution was diluted with water and the precipitate was collected by filtration providing (£)- 2-(3,4- dimethoxystyryl)-4H-pyrido[2,3-d][1,3]oxazin-4-one (0.16 g, 41%) as a green- yellow solid; mp 228-231 °C; δ Η (500 MHz, DMSO-cfe) 3.81 (s, 3H, OCH 3 ), 3.84 (s, 3H, OCH3), 6.99 (d, J = 16.5 Hz, 1H, CH=CHCO), 7.02 (d, J 5 \6- = 8.0 Hz, 1H, H5'), 7.37 (d, J 5 - ff = 8.0 Hz, 1H, H6'), 7.49 (s, 1 H, H2'), 7.55 (dd, J Afi = 8.0, Js,6 = 4.0 Hz, 1 H, H5), 7.82 (d, J = 16.5 Hz, 1H, CH=CHCO), 7.47 (d, J4.5 = 8.0 Hz, 1H, H4), 8.95 (d, J5.6 = 4.0 Hz, 1H, H6); 5 C (125 MHz, DMSO-d 6 ) 55.6, 55.7, 110:5, 111.6, 112.9, 116.5, 123.4, 127.2, 137.3, 143.0, 149.1 , 151.3, 157.1, 157.5, 159.4, 160.1 ; HRMS (ESI) calculated for C^H^C^ [M+Hf 311.1026, found 311,1025; v max 798, 1024, 1417, 1563, 1757, 2836, 2958 cm -1 .

(E)-2-(3, 4-Dimethoxystyryl)-4H-benzo[d][1, 3]oxazin-4-one (FT106) 2-[(Carboxyacetyl)amino]benzoic acid

Anthranilic acid (300 g, 2.08 mol) was added to a solution of Meldrum's acid (272 g, 1.98 mol) in toluene (2.0 L). The reaction flask was fitted with a Dean- Stark apparatus and the suspension was heated to reflux for 3 h. The suspension was cooled, filtered, washed with toluene and dried. 2- [(Carboxyacetyl)amino]benzoic acid (381 g, 86%) was obtained as a colourless solid; mp 171-173 °C; δ Η (500 MHz, DMSO-d 6 ) 3.45 (br s, 2H, CH 2 ), 7.16 (t, J3.4 = J 4 ,5 = 8.0 Hz, 1H, HA), 7.59 (td, J4.5 = Js,e = 8.0, J 3 ,s = 1.5 Hz, 1H, H5), 7.97 (dd, J3.4 = 8.0, J 3l5 = 1.5 Hz, 1H..H3), 8.44 (d, J 5 ,6 = 8.0 Hz, 1H, H6), 11.27 (s, 1H, NH), 12.83 (br s, 1H, C0 2 H), 13.57 (br s, 1H, C0 2 H); 5 C (125 MHz, DMSO- d 6 ) 45.0, 117.0, 120.3, 123.1 , 131.2, 134.1, 140.4, 164.9, 169.1 , 169.3; v max 760, 1234, 1385, 1544, 1684, 1712, 2653, 2964, 3119 cm -1 . (E)'2-[[3-(3 -Dimethoxyphenyl)-1-oxo-2-propenyl]amino]benzoic acid (tranilast)

Piperidine (0.96 mL, 9.7 mmol) was added to a suspension of 3,4- dimethoxybenzaldehyde (1.6 g, 9.7 mmol) and 2-[(carboxyacetyi)amino]benzoic acid (1.9 g, 8.6 mmol) in toluene (5.0 mL). The reaction flask was fitted with a Dean-Stark apparatus and heated to reflux for 4 h, then cooled to rt and the resulting suspension was filtered and washed with toluene. The piperidinium salt was dissolved in MeOH (5.0 mL) and water (2.0 mL) at 40 °C and the solution was acidified with concentrated HCI. The precipitate was filtered, providing (£)-2-[[3-(3,4-dimethoxyphenyl)-1 -oxo-2-propenyl]amino]benzoic acid (tranilast) (2.1 g, 74%) as a yellow crystalline solid; mp 208-209 °C, lit. 2 206 °C; δ Η (500 MHz, DMSO-d 6 ) 3.79 (s, 3H, OCH 3 ), 3.82 (s, 3H, OCH 3 ), 6.79 (d, J = 15.5 Hz, 1H, CH=CHCO), 6.99 (d, J S ;& = 8.5 Hz, 1H, H5 * ), 7.16 (t, 3 , 4 = JA,S = 7.9 Hz, 1H, H4), 7.25 (d, J 5 ,6 . = 8.5 Hz, 1H, H6'), 7.38 (s, 1 H, H2'), 7.56 (d, J = 15.5 Hz, 1 H, CW=CHCO), 7.61 (t, J , 5 = J&,s = 7.9 Hz, 1H, H5), 8.00 (d, J 3 ,4 = 7.9 Hz, 1H, H3), 8.62 (d, J s ,e = 7.9 Hz, 1H, H6), 11.30 (s, 1H, NW), 13.61 (br s, 1H, C0 2 H).

(E)-2-(3,4-Dimethoxystyryl)-4H-benzo[d][1,3]oxazin-4-one (FT106)

FT106

A solution of of (E)-2-[[3-(3,4-dimethoxyphenyl)-1 -oxo-2-propenyl]amino]benzoic acid (0.5 g, 1.5 mmol) in acetic anhydride (3 mL) was heated to reflux and stirred for 3 h. The reaction was cooled to rt and the resulting suspension was diluted with water. The suspension was stirred at rt for 1 h and the precipitate was collected by filtration, providing (£)-2-(3,4-dimethoxystyryl)-4H- benzo[d][1,3]oxazin-4-one (0.41 g, 88 %) as a yellow crystalline solid; mp 175- 179 °C; δ Η (500 MHz, DMSO-cfe) 3.82 (s, 3H, OCH 3 ), 3.85 (s, 3H, OCW 3 ), 6.94 (d, J = 16.0 Hz, 1H, CH=CHCO), 7.02 (d, J 6 \e- = 8.0 Hz, 1H, H5'), 7.35 (dd, J S ;G- = 8.0, J z # = 1.5 Hz, 1H, H&), 7.47 (d, J 2j6 - = 1.5 Hz, 1H, HZ'), 7.58 (t, J 3 , 4 = J 4 , 5 = 8.0 Hz, 1H, HA), 7.61 (d, 3 , 4 = 8.0 Hz, 1 H, HZ), 7.73 (d, J = 16.0 Hz, 1H, CH=CHCO), 7.92 (t, J 4 , 5 = = 8.0 Hz, 1H, H5), 8.12 (d, J 5 ,6 = 8.0 Hz, 1H, HQ); 6c (125 MHz, DMSO-cfe) 55.5, 55.6, 110.3, 111.6, 116.6, 122.9, 126.5, 127.4, 128.0, 128.0, 128.1 , 136.8, 141.4, 146.8, 149.0, 150.9, 157.3, 158.8; HRMS (ESI) calculated for C ie Hi 5 N0 4 [M+H] + 310.1074, found 310.1073; v max 1020, 1254, 1515, 1634, 1755, 2835, 2940 cm -1 .

(E)-2-(3-Methoxy-4-pmpargyloxystyryl)^H-benzo[d][1,3]oxaz in-4-one (FT107) 3-Methoxy-4-propargyloxybenzaldehyde

Propargyl bromide (293 mL, 80% w/v, 1.97 mol) was added to a suspension of vanillin (250 g, 1.64 mol) and potassium carbonate (681 g, 4.93 mol) in MeCN (2.0 L). The suspension was heated to reflux for 6 h and the solvent was removed under reduced pressure. Water was added and the aqueous phase was extracted with EtOAc, washed with water, brine and dried. The solvent was removed under reduced pressure to give 3-methoxy-4- propargyloxybenzaldehyde (302 g, 97%) as yellow crystalline solid; mp 95 °C; δ Η (400 MHz, CDCI 3 ) 2.56 (t, J = 2.5 Hz, 1H, C≡CH), 3.95 (s, 3H, OCH 3 ), 4.86 (d, J = 2.5 Hz, 2H, OCHz), 7.14 (d, J 5>6 = 6.8 Hz, 1H, H5), 7.44 (d, J 2 ,e = 1.4 Hz, 1H, H2), 7.47 (dd, J 5,6 = 6.8, J 2 ,6 = 1.4 Hz, 1H, H6), 9.87 (s, 1H, CHO); 5 C (100 MHz, CDCI 3 ) 56.0, 56.6, 77.2, 77.4, 109.4, 112.5, 126.3, 130.9, 150.0, 152.1 , 190.9; HRMS (ESI) Calculated for C Hi 0 O 3 [M+H]\ 191.0703 found 191.0706; v max 1006, 1130, 1259, 1586, 1677, 2119, 2845, 2932, 3266 cm -1 . (E)-2-[[3-(3-Methoxy-4-propargyloxyphenyl)-1-oxo-2-

piperidine, toluene ^f^CT

Piperidine (158 ml_, 1.59 mol) was added to a suspension of 3-methoxy-4- propargyloxybenzaldehyde (302 g, 1.59 mol) and 2- [(carboxyacetyl)amino]benzoic acid (322 g, 1.44 mol) in toluene (1.5 L). The reaction flask was fitted with a Dean-Stark apparatus and heated to reflux for 30 min. The reaction was then cooled to rt and the resulting suspension was filtered and washed with toluene. The pipendinium salt was dissolved in MeOH (4 L) and water (1 L) at 50 °C and the solution was acidified with 50% aqueous AcOH. The precipitate was filtered and the crude product was recrystallised from EtOH (35 mlVg), filtered and washed with cooled EtOH to afford (E)-2-[[3- (3-methoxy-4-propargyloxyphenyl)-1-oxo-2-propenyl]amino]benz oic acid (309 g, 61%) as a yellow crystalline solid; mp 201-203 °C; δ Η (400 MHz, DMSO-d 6 ) 3.59 (t, J = 2.4 Hz, 1H, C≡CH), 3.84 (s, 3H, OCH 3 ), 4.84 (d, J = 2.4 Hz, 2H, OCH 2 ), 6.81 (d, J = 15.6 Hz, 1H, CH=CHCO), 7.05 (d, J&,& = 8.4 Hz, 1H, H5 1 ), 7.16 (t, 3.4 = s = 8.0 Hz, 1H, HA), 7.25 (d, J 5 \e- .= 8.4 Hz, 1H, H5'), 7.41 (s, 1H, H2'), 7.56 (d, J = 15.6 Hz, 1H, CW=CHCO), 7.61 (t, J 4 , 5 = Js,e = 8.0 Hz, 1H, H5), 8.00 (d, J 3 ,4 = 8.0 Hz, 1 H, HZ), 8.62 (d, J 5 ,6 = 8.0 Hz, 1H, H6), 11.31 (s, 1H, HH), 13.57 (br s, 1H, C0 2 H); 6 C (100 MHz, DMSO-o * 6 ) 55.6, 55.9, 78.6, 79.1, 110.8, 113.5, 116.6, 120.4, 120.4, 122.2, 122.7, 128.2, 131.2, 134.0, 141.0, 141.5, 148.3, 149.3, 164.1, 169.5; HRMS (ESI) calculated for C 2 oHi 7 N0 5 [M+H] + 352.1179, found 352.1187; v max 755, 1010, 1140, 1253, 1502, 1582, 1657, 3278, 3522 cm -1 .

(E)-2-(3-Methoxy-4-propargyloxystyryl)-4H-benzo[d][1,3]ox azin-4-one (FT107)

A solution of of (£)-2-[l3-(3-methoxy-4-propargyloxyphenyl)-1-oxo-2- propenyl]aminojbenzoic acid (0.5 g, 1.4 mmol) in acetic anhydride (3 ml_) was heated to reflux and stirred for 3 h. The reaction was cooled to rt and the resulting suspension was diluted with water. The suspension was stirred at rt for 1 h and the precipitate was collected by filtration, providing (£)-2-(3-methoxy-4- propargyloxystyryl)-4H-benzo[d][1,3]oxazin-4-one (0.44 g, 93 %) as a yellow crystalline solid; mp 177-178 °C; δ Η (500 MHz, DMSO-d 6 ) 3.59 (m, 1H, C≡CH), 3.85 (s, 3H, OCH 3 ), 4.85 (s, 2H, OCH 2 ), 6.96 (d, J = 16.0 Hz, 1H, CH=CHCO), 7.07 (d, J 5 ,6' = 8.0 Hz, 1H, H5'), 7.34 (d, J s>v = 8.0, 1H, H6'), 7.49 (s, 1H, H2'), 7.57 (t, J3.4 = JA,S = 8.0 Hz, 1H, H4), 7.60 (d, J 3 ,4 = 8.0 Hz, 1H, H3), 7.72 (d, J = 16.0 Hz, 1H, CH=CHCO), 7.91 (t, J A ,s = Js.e = 8.0 Hz, 1H, H5), 8.11 (d, 5 ,6.= 8.0 Hz, 1H, /- e); 6c (125 MHz, DMSO-d 6 ) 55.7, 55.9, 78.5, 79.0, 110.8, 113.6, 117.0, 117.2, 122.3, 126.5, 128.0, 128.1 , 128.3, 136.8, 141.2, 146.8, 148.5, 149.4, 157.2, 158.8; HRMS (ESI) calculated for C20H15NO4 [M+H] + 334.1074, found 334.1074; v max 970, 1136, 1270, 1471 , 1743, 2135, 3268 cm -1 .

(E)-2-(3,4-Bis(difluommethoxy)styryl)^H-benzo[d][1,3]oxaz ^ (FT108)

(E)-2-[[3,4-Bis(difluommethoxy)phenyl)-1-ox< 2^mpenyl]am

Piperidine (100 pL, 1.01 mmol) was added to a suspension of 3,4- bis(difluoromethoxy)benzaldehyde (240 mg, 1.01 mmol) and 2- [(carboxyacetyl)amino]benzoic acid (204 mg, 0.92 mmol) in toluene (5.0 mL). The reaction flask was fitted with a Dean-Stark apparatus and heated to reflux for 30 min. The reaction was then cooled to rt and the resulting suspension was filtered and washed with toluene. The piperidinium salt was dissolved in MeOH (5 mL) and water (2 ml.) and the solution was acidified with 50% aqueous AcOH. The crude product was collected by filtration and recrystallised from EtOH water, filtered and washed with water to afford (E)-2-[[3,4- bis(difluoromethoxy)phenyl)-1-oxo-2-propenyl]amino]benzoic acid (259 mg, 71%) as a colourless crystalline solid; mp 190-193 °C; δ Η (400 MHz, DMSO-d 6 ) 6.96 (d, J = 15.6 Hz, 1 H, CH=CHCO), 7.18 (t, 3 , 4 = JA.S = 8.0 Hz, 1H, H4), 7.27 (t, J = 73 Hz, 2H, OCHF 2 ), 7.38 (d, J 5 , 6 . = 8.0 Hz, 1 H, H5'), 7.61 (d, J = 15:6 Hz, 1 H, CH=CHCO), 7.62 (t, J 4 , 5 = Js,e = 8.0 Hz, 1 H, H5), 7.78 (d, J 2 = 1.6 Hz, 1 H, H2 7.68 (dd, J S - = 8.0, J ZI6 - = 1.6 Hz, 1 H, H6'), 8.00 (d, J 3 , 4 = 8.0 Hz, 1H, H3), 8.69 (d, J 5( 6 = 8.0 Hz, 1H, H6), 11.35 (s, 1H, H), 13.56 (br s, 1H, C0 2 H); 6 C (100 MHz, DMSQ-o* 6 ) 116.3 (t, J = 258 Hz), 116.5 (t, J = 258 Hz), 117.0, 120.1 , 120.5, 120.8, 123.0, 123.8, 126.7, 131.1 , 132.8, 133.9, 139.3, 140.7, 141.9, 142.7, 163.5, 169.4; HRMS (ESI) calculated for Ci e H 13 F 4 N0 5 [Μ-ΗΓ 398.0646, found 398.0652; 1034, 1217, 1513, 1604, 1683, 2892, 3466 cm -1 .

A solution of of (E)-2-[[3,4-bis(difluoromethoxy)phenyl)-1-oxo-2- propenyl]amino]benzoic acid (0.5 g, 1.2 mmol) in acetic anhydride (3 mL) was heated to reflux and stirred for 3 h. The reaction was cooled to and the resulting suspension was diluted with water. The suspension was stirred at rt for 1 h and the precipitate was collected by filtration, providing (£)-2-(3,4- bis(difluoromethoxy)styryl)-4H-benzo[d][1 ,3]oxazin-4-one (0.41 g, 86 %) as a colourless crystalline solid; mp 1 13-115 °C; δ Η (500 MHz, DMSO-d 6 ) 7.07 (d, J = 16.0 Hz, 1 H, CH=CHCO), 7.29 (t, J = 74 Hz, 1 H, OCHF 2 ), 7.28 (t, J = 74 Hz, 1 H, OCHF 2 ), 7.39 (d, 5 \e- = 8.0 Hz, 1 H, H5'), 7.60 (t, J 3 , 4 = As = 8.0 Hz, 1 H, H4), 7.62 (d, J3.4 = 8.0 Hz, 1H, HZ), 7.75 (d, Jy.e- = 8.0, 1 H, H6'), 7.76 ; ' (d, J = 16.0 Hz, 1H, CH=CHCO), 7.87 (s, 1H, H2'), 7.92 (t, J 4 , 5 = Jss = 8.0 Hz, 1H, H5), 8.12 (d, J 5 .6 = 8.0 Hz, 1H, H6); 5 C (125 MHz, DMSO-d 6 ) 116.3 (t, J = 258 Hz), 116.4 (t, J = 258 Hz), 116.9, 119.9, 120.6, 120.8, 126.5, 126.7, 128.1 , 128.6, 132.9, 136.8, 139.0, 141.2, 142.0, 146.5, 156.5, 158.7; HRMS (ESI) calculated for C18H11F 4 NO4 [M+H] + 382.0697, found 382.0696; v max 969, 1042, 1087, 1123, 1279, 1383, 1472, 1595, 1747 cm -1 .

(E)-2-(4-Acetoxy-3-methoxystyry -4H-benzo[d][1,3]oxazin-4-one (FT109) (E)-2-[[3-(4^ydmxy-3-methoxyphenyl)-1^xo-2-pmpenyl]amino]ben zoic

Piperidine (0.50 mL, 5.1 mmol) was added to a suspension of 4-hydroxy-3- methoxybenzaldehyde (0.77 g, 5.1 mmol) and 2-[(carboxyacetyl)amino]benzoic acid (1.0 g, 4.5 mmol) in toluene (5.0 mL). The reaction flask was fitted with a Dean-Stark apparatus and heated to reflux for 3 h, then cooled to rt and the resulting suspension was filtered and washed with toluene. The piperidinium salt was dissolved in MeOH (12.0 mL) and water (12.0 mL) at 40 °C and the solution was acidified with concentrated HCI. The precipitate was filtered, providing (£)-2-[[3-(4-hydroxy-3-methoxyphenyl)-1-oxo-2- propenyl]amino]benzoic acid (1.1 g, 78%) as a yellow crystalline solid; mp 207.5-208.5 °C, lit. 5 230-233 °C; δ Η (500 MHz, DMSO-cfe) 3.83 (s, 3H, OCH 3 ), 6.71 (d, J = 15.5 Hz, 1 H, CH=CHCO), 6.80 (d, J 5 -,6- = 8.5 Hz, 1 H, H5'), 7.13 (dd, J 5 .6- = 8.5, J 2 ;v = 1.5 Hz, 1 H, Ηβ'), 7.15 (t, J 3 .4 = As = 8.0 Hz, 1H, HA), 7.34 (d, J 2 \6- = 1.5 Hz, 1H, H2 7.52 (d, J = 15.5 Hz, 1H, CW=CHCO), 7.60 (td, J 4l s = J 5 ,6 = 8.0, J3.5 = 2.0 Hz, 1H, H5), 8.00 (dd, J 3A = 8.0, J 3 ,s = 2.0 Hz, 1H, HZ), 8.62 (d, J5.6 = 8.0 Hz, 1H, H6), 9.57 (s, 1H, OH), 11.27 (s, 1H, NH), 13.61 (br s, 1H, C0 2 H). (E)-2-(4-Acetoxy-3-methoxystyryl)-4H-benzo[d][1,3]oxazin-4-o ne (FT109)

FT109

A solution of of (£)-2-[[3-(4-hydroxy-3-methoxyphenyl)-1-oxo-2- propenyl]amino]benzoic acid (0.5 g, 1.6 mmol) in acetic anhydride (3 mL) was heated to reflux and stirred for 3 h. The reaction was cooled to rt and the resulting suspension was diluted with water. The suspension was stirred at rt for 1 h and the precipitate was collected by filtration, providing (£)-2-(4-acetoxy-3- methoxystyryl)-4H-benzo[d][1 ,3]oxazin-4-one (0.52 g, 97 %) as a pale brown solid; mp 185-186 °C; δ Η (400 MHz, DMSO-d 6 ) 2.63 (s, 3H, COCH 3 ), 3.85 (s, 3H, OCH 3 ), 7.08 (d, J = 16.0 Hz, 1H, CH=CHCO), 7.17 (d, 5 -, 6 ' = 8.0 Hz, 1H, Η5·), 7.38 (d, 5 .,6- = 8.0, 1H, HQ'), 7.59 (t, J 3 , 4 = J 4l s = 8.0 Hz, 1H, H4), 7.61 (d,/3,4 = 8.0 Hz, 1H, H3), 7.63 (s, 1H, H2'), 7.76 (d, J = 16.0 Hz, 1H, CH=CHCO), 7.92 (t, J 4> 5 = sfi = 8.0 Hz, 1H, H5), 8.11 (d, J 5i6 = 8.0 Hz, 1H, H6); 6 C (100 MHz, DMSO-de) 20.4, 56.0, 111.9, 116.9, 119.5, 121.4, 123.3, 126.6, 128.1, 128.4, 133.5, 136.8, 140.5, 140.9, 146.6, 151.2, 156.8, 168.4; HRMS (ESI) calculated for Οι 9 Η 15 Ν0 5 [M+H] + 338.1023, found 338.1023; v max 1197, 1214,

1593, 1635, 1748 cm -1 (E)-2-(3-Acetoxy-4-methoxystyryl)-4H-benzo[d][1,3]oxazin-4-o ne (FT110)

(E)-2-[[3-(3-Hydmxy-4-methoxyphenyl)-1^xo-2^mpenyl]aminoJ benzoic acid

Piperidine (0.25 mL, 2.5 mmol) was added to a suspension of 3-hydroxy-4- methoxybenzaldehyde (0.39 g, 2.5 mmol) and 2-[(carboxyacetyl)amino]benzoic acid (0.50 g, 2.2 mmol) in toluene (5.0 mL). The reaction flask was fitted with a Dean-Stark apparatus and heated to reflux for 3 h, then cooled to rt and the resulting suspension was filtered and washed with toluene. The piperidinium salt was dissolved in MeOH (5.0 ml.) and water (2.0 mL) at 40 °C and the solution was acidified with concentrated HCI. The precipitate was filtered, providing (£)-2-[[3-(3-hydroxy-4-methoxyphenyl)-1-oxo-2- propenyl]amino]benzoic acid (0.53 g, 76%) as a yellow crystalline solid; mp 215-216 °C, lit. 5 219-222 °C; δ Η (500 MHz, DMSO-d 6 ) 3.81 (s, 3H, OCtf 3 ), 6.59 (d, J = 15.5 Hz, 1H, CH=CtfCO), 6.80 (d, J 5 i6 - = 8.5 Hz, 1H, H5'), 7.10-7.13 (m, 2H, HZ, HQ'), 7.15 (t, J 3 , 4 = s = 8.0 Hz, 1H, HA), 7.47 (d, J = 15.5 Hz, 1H, CW=CHCO), 7.60 (td, J 4 , 5 = J 5 ,6 = 8.0, J 3 , 5 = 1.5 Hz, 1 H, H5), 7.99 (dd, J Z = 8.0, J 3 ,5 = 1 5 Hz, 1 H, H3), 8.58 (d, J 5 , 6 = 8.0 Hz, 1H, H6), 11.25 (s, 1H, NH), 13.56 (br s, 1H, C0 2 H).

(E)-2-(3-Acetoxy-4-methoxystyryl)-4H-benzo[d][1,3]oxazin- 4-one (FT110)

FT110

A solution of of (E)-2-[[3-(3-hydroxy-4-methoxyphenyl)-1-oxo-2- propenyl]amino]benzoic acid (0.5 g, 1.6 mmol) in acetic anhydride (3 mL) was heated to reflux and stirred for 3 h. The reaction was cooled to rt and the resulting suspension was diluted with water. The suspension was stirred at rt for 1 h and the precipitate was collected by filtration, providing (£)-2-(3-acetoxy-4- methoxystyryl)-4H-benzo[d][1,3]oxazin-4-one (0.50 g, 93 %) as a yellow solid; mp 187-190 °C; δ Η (400 MHz, DMSO-cfe) 2.80 (s, 3H, COCH 3 ), 3.83 (s, 3H, OCH 3 ), 6.87 (d, J = 16.0 Hz, 1H, CH=CHCO), 7.19 (d, J 5 >, G = 8.0 Hz, 1H, H5'), 7.68 (d, J 5 I 6- = 8.0, 1H, H6') t 7.57 (t, J 3 .4 = J4.5 = 8.0 Hz, 1H, HA), 7.60 (d, J 3 , 4 = 8.0 Hz, 1H, HZ), 7.66 (s, 1H, H2'), 7.71 (d, J = 16.0 Hz, 1H, CH=CHCO), 7.89 (t, JA,S = Js,6 = 8.0 Hz, 1H, H5), 8.10 (d, J 5 .B = 8.0 Hz, 1H, H6); 6 C (100 MHz, DMSO-d 6 ) 20.4, 56.1, 112.9, 116.7, 117.6, 122.1, 127.5, 128.0, 128.1, 128.2, 136.8, 139.6, 140.2, 146.7, 152.7, 157.0, 158.8, 168.4; HRMS (ESI) calculated for C 19 H 15 N0 5 [M+H] + 338.1023, found 338.1023; v max 1197, 1268, 1588, 1631 , 1752 cm ~ (E)-3-(3,4 limethoxyphenyl)-1-(indolin-1-yl)prop-2-en-1-one (FT113)

A suspension of (£)-3-(3,4-dimethoxyphenyl)acrylic acid (0.50 g, 2.4 mmol) in CH 2 CI 2 (5 mL) was treated with oxalyl chloride (0.80 mL, 9.6 mmol). The solution was stirred at rt for 2 h and the solvent was removed under reduced pressure to give the acid chloride. Indoline (0.27 g, 2.4 mmol) was added to a solution of the acid chloride (2.4 mmol) in CH 2 CI 2 (5 mL) and NEfe (0.67 mL, 4.8 mmol). The mixture was stirred at rt for 16 h and concentrated under reduced pressure. The residue was diluted with water and the resulting precipitate was collected by filtration and recrystallised from EtOAc providing (£)-3-(3,4- dimethoxyphenyl)-1-(indolin-1-yl)prop-2-en-1-one (0.15 g, 20%) as a colourless crystalline solid; mp 118-120 °C; δ Η (500 MHz, DMSO-d 6 ) 3.18 (t, J = 8.0 Hz, 2H, CH 2 ), 3.79 (s, 3H, OCH 3 ), 3.83 (s, 3H, OCH 3 ), 4.33 (t, J = 8.0 Hz, 2H, NCH 2 ), 6.98-7.02 (m, 3H, CH=CHCO, H5, H6), 7.16 (t, J 3A = J4.5 = 8.0 Hz, 1H, H4), 7.25 (d, J 5 ,6- = 8.0 Hz, 1H, H5'), 7.26 (dd, J 56 " = 8 0 Hz, J 2 - & = 2.0 Hz, 1H, H6'), 7.38 (d, J 2 > & = 2.0 Hz, 1H, HZ), 7.58 (d, J - 15.5 Hz, 1 H, CH=CHCO), 8.19 (m, 1H, HZ); 5 C (125 MHz, DMSO-d 6 ) 27.3, 47.7, 55.5, 55.6, 110.6, 111.6, 116.4, 117.5, 122.6, 123.3, 124.8, 126.9, 127.7, 132.2, 142.3, 143.1, 148.9, 150.6, 163.9; HRMS (ESI) calculated for C 19 H 19 N0 3 [M+Hf 310.1438, found 310.1437; v max 1025, 1145, 1262, 1398, 1510, 1646 cm "1 .

(E)- -(3,4-Dimethoxystyryl)-1H-benzo[d]imidazole (FT121)

FT121

A mixture of (E)-3-(3,4-dimethoxyphenyl)acrylic acid (416 mg, 2.0 mmol) and o- phenylenediamine.sulfuric acid (412 mg, 2.0 mmol) in ethylene glycol (10 mL) was heated at reflux for three hours. The mixture was then cooled to room temperature and poured into water (50 mL). The pH of the solution was adjusted to >7 with NaHC0 3 and extracted with DCM. The organic layer was washed with saturated aqueous NaCI solution and dried over sodium sulfate. The solvent was concentrated under a reduced pressure and the residue was purified by flash chromatography on silica gel using petroleum ether/ethyl acetate 2:1 (v:v) as eluent to give (£)-2-(3,4-dimethoxystyryl)-1H- benzo[d]imidazole (130 mg, 23%). 1 H-NMR (400 MHz, CDCI 3 ) δ 3.78 (s, 3H), 3.88 (s, 3H), 6.80 (d, J = 8.0 Hz, 1H), 6.96-7.00 (m, 2H), 7.06 (d, J = 16.0 Hz, 1H), 7.25-7.29 (m, 2H), 7.59 (d, J = 16.0 Hz, 1H), 7.64 (m, 2H); LC-MS (ES- API); rt 7.55 min; m/z calculated for [M+Hf 281.1 , found 281.1. (E)-1-(2, 3-Dihydrobenzo[b][1,4]oxazin-4-yl)-3-(3, 4-dimethoxyphenyl)prop-2-en- 1-one (FT122)

3,4-Dihydro-2H-benzo[b][1,

To a suspension of 2-aminophenol (1.0 g, 9.2 mmol) and potassium carbonate (6.36 g, 46 mmol) in dry DMF (10 mL) was added 1, 2-dibromoethane (2.59 g, 13.8 mmol). The mixture was then heated at 125'C for 15 hours. After cooling, the mixture was treated with crushed ice and extracted with ethyl acetate. The organic extracts were dried over sodium sulfate and the solvent was removed under reduced pressure. The residue was purified by flash chromatography on silica gel using ethyl acetate/petroleum ether 1:10 (v:v) as eluent to give 3,4- dihydro-2H-benzo[b][1,4]oxazine as reddish oil (0.81 g, 65%). LC-MS (ES-API); rt 7.51 min; m/z calculated for C 8 H 9 NO [M+Hf 136.1, found 136.1. (E)-1-(2, 3-Dihydrobenzo[b][ 1, 4]oxazin-4-yl)~3-(3, 4-dimethoxyphenyl)prop-2- en-1-one (FT122)

FT122

To a solution of (E)-3-(3,4-dimethoxyphenyl)acrylic acid (0.46 g, 2.22 mmol) and 3,4-dihydro-2H-benzo[b][1,4]oxazine (0.20 g, 1.48 mmol) in DCM (15 mL) was added EDC.HCI (0.71 g, 3.70 mmol), HOBt (0.60 g, 3.70 mmol) and EfeN (0.37 g, 3.70 mmol). The mixture was stirred at room temperature overnight then diluted with water and extracted with DCM. The organic layer was dried over sodium sulfate and the solvent was removed under reduced pressure. The residue was purified by column chromatography on silica gel using petroleum ether/ethyl acetate 5:1 (v:v) as eluent to give (E)-1-(2,3- dihydrobenzo[b][1,4]oxazin-4-yl)-3-(3,4-dimethoxyphenyl)prop -2-en-1-one (230mg, 32%). 1 H-NMR (400 MHz, CDCI 3 ) δ 3.88(s, 3H), 3.91 (s, 3H), 4.08 (m, 2H), 4.37 (m, 2H), 6.86 (d, J = 8.4 Hz, 1 H), 6.90 (m, 1H), 6.92 (d, J = 15.2 Hz, 1H), 6.96 (dd, J = 8.0, 1.6 Hz, 1H), 6.99 (d, J = 2.0 Hz, 1H), 7.08-7.13 (m, 2H), 7.20 (br d, J = 7.6 Hz, 1H), 7.02 (d, J = 15.6 Hz, 1H); LC-MS (ES-API); rt 8.72 min; m/z calculated for C19H19NO4 [M+H] + 326.1 , found 326.1.

(E)-1-(3-(3,4-Dimethoxyphenyl)acry1oyO-3-methyl-1H-ben^

one (FT123)

(E)-3-(3, 4-Dimethoxyphenyl)acryloyl chloride

To a suspension of 3-(3,4-dimethoxyphenyl)acrylic acid (416 mg 2 mmol) in toluene (10 mL) was added thionyl chloride (1.45 mL, 20 mmol). The solution was heated at 50°C for 1 hour and then the solvent was removed under reduced pressure to give (E)-3-(3,4-dimethoxyphenyl)acryloyl chloride , which was used directly in the next step without purification. -1-(3-(3,4-Dimethoxyphenyl)acryloy -1H-benzo[d]imidazol-2(3H)^^

To a solution of 1H-benzo[d]imidazol-2(3H)-one (268 mg, 2 mmol) in DMF (10 mL) at 0°C was added 60% oily sodium hydride (88 mg, 2.2 mmol). When hydrogen evolution had ceased, a solution of (E)-3-(3,4- dimethoxyphenyl)acryloyl chloride (240 mg, 0.75mmol) in DMF (3 mL) was added and the reaction mixture allowed to warm to room temperature and stirred for 1 hour. The reaction was quenched by addition of 1 N hydrochloric acid, and extracted with DCM. The combined organic phases were washed with brine and dried over sodium sulfate. After concentration under reduced pressure, the residue was purified by flash column (hexane-EtOAc, 25: 10) to give (£)-1 -(3-(3,4-dimethoxyphenyl)acryloyl)-1 H-benzo[d]imidazol-2(3H)-one (240 mg, 37%). LC-MS (ES-API); rt 9.02 , min; m/z calculated for C ie H 16 N 2 04 , [M+Hf 325.1 , found 325.0 and 347.0 ([M+Naf).

(E)- 1-(3-(3, 4-Dimethoxyphenyl)acryloyl)-3-methyl- 1H-benzo[d]imidazol- -one (FT123)

FT123

A mixture of (£)-1-(3-(3,4-dimethoxyphenyl)acryloyl)-1H-benzo[d]imidazol - 2(3A7)-one (240 mg, 0.75 mmol), iodomethane (68 uL, 1.1 mmol) and K 2 C0 3 (204 mg, 1.5 mmol) in DMF (5 mL) was stirred at room temperature overnight. The reaction was quenched with water and extracted with DCM. The combined organic phases were washed with brine, dried over sodium sulfate and the solvent . removed under reduced pressure. The residue was purified by recrystallisation from EtOH to afford (E)-1-(3-(3,4-dimethoxyphenyl)acryloyl)-3- methyl-1 H-benzo[d]imidazol-2(3H)-one (200 mg, 79%). 'H-NMR (400 MHz,CDCI 3 ) δ 3.46 (s, 3H), 3.96 (s, 3H), 3.98 (s, 3H), 6.91 (d, J = 8.4 Hz, 1H), 7.01 (dd, J = 7.6, 0.8 Hz, 1H), 7.19-7.28 (m, 4H), 7.98 (d, J = 15.6 Hz, 1H), 8.15 (d, J = 15.6 Hz, 1H) 8.32 (dd, J = 8.0, 0.8 Hz, 1 H); LC-MS (ES-API); rt 9.05 min; m/z calculated for Ci 9 H 18 2 0 [M+H] + 339.1 , found 339.0 and 361.0 ([M+Na] + ).

(E)-3-(3-(3,4-Dimethoxyphenyl)acryloyl)benzo[d]oxazol-2(3 H)-one (FT124)

To a solution of (-E)-3-(3,4-dimethoxyphenyl)acrylic acid (208 mg, 1.0 mmol) and benzo[d]oxazol-2(3H)-one (162mg, 1.2mmol) in DCM (10 mL) was added EDC.HCI (230 mg, 1.2 mmol), HOBt (196 mg, 1.2 mmol) and EfeN (304 mg, 3.0 mmol). The mixture was stirred at room temperature overnight then diluted with water and extracted with DCM. The organic layer was dried over sodium sulfate and the solvent was removed under reduced pressure. The residue was purified by flash chromatography using petroleum ether/ethyl acetate 3:2 (v:v) to give (E)-3-(3-(3,4-d)acryloyl)benzo[d]oxazol-2(3H)-one (225 mg, 70%). 1 H-NMR (400 MHz, DMSO-d 6 ) δ 3.84 (s, 6H), 7.09 (d, J = 8.4 Hz, 1H), 7.31-7.38 (m, 4H), 7.44-7.46 (m, 1 H), 7.76 (d, J = 15.6Hz, 1H), 7.95 (d, J = 16Hz, 1 H), 8.03-8.06 (m, 1H); LC-MS (ES-API); rt 9.08 min; m z calculated for CieH 15 N0 5 [M+ Na] + 348.1, found 348.1. (E)-4-(3-(3, 4-Dimethoxyphenyl)acryloyl)-3, 4-dihydnoquinoxalin-2(1 H)-one

(FT125)

3, 4-Dihydroquinoxalin-2( 1 H)-one

To a solution of o-phenylenediamine (1.08 g, 10 mmol) in DMF (50 mL) was added E¾N (2.9 mL, 21 mmol), and ethyl 2-bromoacetate (1.2 mL, 11 mmol). The reaction mixture was stirred at room temperature for 16 h, then at 80°C for 3 h. The DMF was evaporated under reduced pressure and the reaction mixture partitioned between H 2 0 and EtOAc. The EtOAc layer was washed with sat. NaHCC>3, brine, and dried over Na 2 S0 4 . The solvent was evaporated under reduced pressure and the crude residue purified by flash chromatography eluting with petroleum ether/EtOAc (4/1) to give 3,4-dihydroquinoxalin-2(1 H)- one (560 mg, 38%) as yellow solid. LC-MS (ES-API); rt 7.40 min; m/z calculated for C 8 HeN 2 0 [M+Hf 149.0, found 149.0.

(E)-4-(3-(3,4-Dimethoxyphenyl)acryloyl)-3,4 lihydrOquino

(FT125)

FT125

To a stirred solution of 3,4-dihydroquinoxalin-2(1H)-one (142 mg, 0.96 mmol) and triethylamine (0.16 mL, 1.14 mmol) in anhydrous THF (15 ml_) at 0°C was added a solution of (E)-3-(3,4-dimethoxyphenyl)acryloyl chloride in THF (2 mL) dropwise. The reaction mixture was allowed to warm to room temperature and stirred for 2 hours. The reaction was quenched by addition of 1 N hydrochloric acid, and extracted with DCM. The combined organic phases were washed with brine, dried over sodium sulfate and concentrated under reduced pressure to afford the crude product which was purified by flash chromatography (hexane- EtOAc, 25:10) to give (£)-4-(3-(3,4-dimethoxyphenyl)acryloyl)-3,4- dihydroquinoxalin-2(1 H)-one (160 mg, 47%). 1 H-NMR (400 MHz,CDCI 3 ) δ 3.86 (s, 3H), 3.90 (s, 3H),' 4.63 (br s, 2H), 6.71 (d, J = 15.2 Hz, 1 H), 6.85 (d, J = 8.4 Hz, 1H), 6.94 (d, J = 2.0 Hz, 1H), 7.02 (dd, J = 7.6, 1.2 Hz, 1H), 7.07-7.13 (m, 2H), 7.22 (dd, J = 7.6, 1.2 Hz, 1H), 7.28 (br d, J = 8.0 Hz, 1H), 7.76 (d, J = 15.6 Hz, 1H), 8.89 (s, 1H); LC-MS (ES-API); rt 8.14 min; m/z calculated for Ci 9 H ie N 2 04 [M+H] + 339.1 , found 339.0.

2-(5,6-Dimethoxy-1H-indene-2-cart>oxamido)benzoic acid (FT126)

2-lodo-4, 5-dimethoxybenzaldehyde

To a solution of 3,4-dimethoxybenzaldehyde (2.00 g, 12.0 mmol) in EtOH (100 mL) under nitrogen was added iodine (3.65 g, 14.4 mmol) and silver sulfate (4.49 g, 14.4 mmol) and the mixture was stirred for 3 hours at room temperature. The solvent was removed, water was added and the mixture was extracted with DCM. The organic, extracts were washed with brine, dried over sodium sulfate and the solvent was removed under reduce pressure. The residue was purified by flash chromatography using petroleum ether/ethyl acetate 2:1 (v:v) as eluent to give 2-iodo-4,5-dimethoxybenzaldehyde (2.81g, 80%). LC-MS (ES-API); rt 8.52 min; m/z calculated for C9H9IO3 [M+H] + 293.0, found 293.0.

2-lodo-4,5-dimethoxybenzaldehyde (292 mg, 1.0 mmol), CoCI 2 (dppe) complex (26.4 mg, 0.05 mmol), dppe (19.9 mg, 0.05 mmol) and zinc power (179.8 mg, 2.75 mmol) were placed in a vial, which was sealed with a septum and flushed several times with nitrogen. Acetonitrile (3 mL) and methyl acrylate (172 mg, 2.0 mmol) were then sequentially added and the mixture was heated at 80°C overnight. Water was then added and the mixture was extracted with ethyl acetate. The organic extracts were concentrated under reduced pressure and the residue was purified by flash chromatography using petroleum ether/ethyl acetate 8:1 (v:v) as eluent to give methyl 5,6-dimethoxy-1 H-indene-2- carboxylate (50 mg, 21%). LC-MS (ES-API); rt 7.89 min; m/z calculated for C 13 H 14 04 [M+Hf 235.1 , found 235.1. -Dimethoxy-1hMndene-2-carboxylic acid

To a stirred solution of methyl 5,6-dimethoxy-1H-indene-2-cart)oxylate (330 mg, 1.41 mmol) in THF (8 mL) and MeOH (4 mL) was added 2 N NaOH solution (3.5 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for 2 hours and the reaction was quenched with 1 N HCI solution. Then mixture was diluted with water and the resulting suspension was filtered and the solid was dried under vacuum to give 5,6-dimethoxy-1H-indene- 2-carboxylic acid (150 mg, 48%). LC-MS (ES-API); rt 8.44 min; m z calculated for C12H12O4 [M-HT 219.1 , found 219.1. Methyl 2-(5, 6-dimethoxy-1H-indene-2-carboxamido)benzoate

To a stirred mixture of 5,6-dimethoxy-1 - -indene-2-carboxylic acid (230 mg, 1.05 mmol) and DMF (20 μί) in DCM ( 0 mL) was added oxalyl chloride (267 mg, 2.10 mmol) dropwise at room temperature. The resulting mixture was stirred for 2 hours then concentrated and dried under high vacuum. The resulting acid chloride was then taken up into DCM (10 mL) and added dropwise to a stirred mixture of methyl 2-aminobenzoate (174 mg, 1.16 mmol) and triethylamine (159.1 mg, 1.58 mmol) in DCM (10 mL) O'C. The mixture was then stirred overnight at room temperature before adding water and extracting with DCM. The organic extracts were dried over sodium sulfate and the solvent was removed under reduced pressure. The residue was purified by silica gel chromatography with petroleum ether/ethyl acetate 5:1 (v.v) as eluent to give methyl 2-(5,6-dimethoxy-1H-indene-2-carboxamido)benzoate (140 mg, 38%). LC-MS (ES-API); rt 9.35 min; m/z calculated for C20H19NO5 [M+H]* 354.1 , found 354.1. -(5,6-Dimethoxy-1H-indene-2-carboxamido)benzoic acid (FT126)

FT126

To a stirred solution of methyl 2-(5,6-dimethoxy-1H-indene-2- carboxamido)benzoate (120 mg, 0.34 mmol)in THF (6 mL) and MeOH (3 mL) was added 2 N NaOH solution (0.85 mL) dropwise. The resulting mixture was stirred at room temperature for 2 h, then quenched with 1N HCI solution and extracted with DCM. The organic extracts were washed with a saturated aqueous NaCI solution and dried over sodium sulfate. The solvent was then removed under reduced pressure to give 2-(5,6-dimethoxy-1 H-indene-2- carboxamido)benzoic acid (100 mg, 87%). Ή-NMR (400 MHz, DMSO-d 6 ) δ 3.72 (br s, 2H), 3.79 (s, 3H), 3.81 (s, 3H), 7.15 (m, 1H), 7.24 (s, 1H), 7.26 (s, 1H), 7.55-7.66 (m, 2H), 8.04 (dd, J = 8.0, 1.6 Hz, 1H), 8.70 (dd, J - 8.4, 1.2 Hz, 1 H), 11.91 (s, 1H); LC- S (ES-API); rt 9.23 min; mh calculated for 0 19 Η 17 Ν0 5 [M-HV 338.1, found 338.1.

2-(5,6-Dimethoxy-1H-benzo[d]imidazole-2-carboxamido)benzo ic acid (FT127) Methyl 2-aminobenzoate

To a stirred solution of 2-aminobenzoic acid (2.0 g, 14.59 mmol) in MeOH (70 ml.) at 0°C was added SOCI 2 (7.4 g, 145.9 mmol) dropwise. The mixture was heated at reflux overnight and was then concentrated under reduced pressure. DCM and saturated aqueous aHC0 3 were added and the aqueous phase extracted with DCM. The combined organic layers were washed with brine, dried over sodium sulfate and concentrated to afford methyl 2-aminobenzoate (2.05 g, 93%). LC-MS (ES-API); rt 8.46 min; m/z calculated for CeHgNOa [M+H] + 152.0, found 152.1.

Methyl 2-(2-chloroacetamido)benzoate

To a stirred solution of methyl 2-aminobenzoate (906 mg, 6.0 mmol) and NaHCC-3 (554.4 mg, 6.6 mmol) in THF (12 mL) and water (12 mL) at 0°C was added 2-chloroacetyl chloride (806 mg, 7.2 mmol) dropwise. After 2 hours, the mixture was diluted with water and extracted with ethyl acetate. The organic layer was washed with brine, dried over sodium sulfate and concentrated under reduced pressure to afford the crude product. The residue was purified by flash chromatography to give methyl 2-(2-chloroacetamido)benzoate (1.31 g, 96%). LC-MS (ES-API); rt 8.65 min; m/z calculated for doHioCINOa [M+Na] + 250.0, found 250.0.

A mixture of 4,5-dimethoxybenzene-1,2-diamine (1.08 g, 4.5 mmol), NaHC0 3 (756 mg, 9.0 mmol) methyl 2-(2-chloroacetamido)benzoate (1.03 g, 4.5 mmol) and sulfur (216 mg, 6.75 mmol) in CH 3 CN (27 mL) was stirred in a microwave reactor for 2 hours at 140°C. The mixture was concentrated and purified by flash column chromatography to give the desired methyl 2-(5,6-dimethoxy-1 H- benzo[d]imidazole-2-carboxamido)benzoate (190 mg, 12%). LC-MS (ES-API); rt 9.12 min; m/z calculated for CieH 7 N 3 05 [M+Na] + 378.1, found 378.1.

2-(5,6-Dimethoxy-1H-bBnzo[d]imidazole-2^arboxamido)benzoi c acid

To a stirred solution of methyl 2-(5,6-dimethoxy-1H-benzo[d]imidazole-2- carboxamido)benzoate (200 mg, 0.56 mmol) in THF (10 mL) and MeOH (5 mL) was added 2N NaOH solution (1.4 mL) dropwise. The resulting mixture was stirred at room temperature for 2 hours. The reaction was quenched by addition of 1 HCI solution and extracted with DCM. The organic layer was washed with brine, dried over sodium sulfate and concentrated to give 2-(5,6-dimethoxy-1 H- benzo[d]imidazole-2-carboxamido)benzoic acid (90 mg, 47.1%). 1 H-NMR (400 MHz, DMSO-de) δ 3.82 (s, 6H), 6.97 (s, 1H), 7.13-7.34 (m, 2H), 7.64 (m, 1H), 8.08 (d, J = 9.6 Hz, 1H), 8.78 (d, J = 7.6 Hz 1H), 12.90 (s, 1H), 13.27 (s, 1H); LC-MS (ES-API); rt 8.74 min; m/z calculated for CuH^NaOs [M-H] ' 340.1 , found 340:1. 2-( 1, 1-Dioxo-5, 6^imethoxybenzo[b]thiophene-2-carboxamido)benzoic acid (FT128)

Methyl 5, 6-dimethoxybenzo[b]thiophene-2-carboxylate

To a stirred solution of 2-fluoro-4,5-dimethoxybenzaldehyde (1.10 g, 6 mmol) in DMF (50 mL) was added methyl 2-mercaptoacetate (0.58 ml_, 6.6 mmol) and potassium carbonate (2.48 g, 18 mmol). The resulting mixture was then heated at 60'C for 15 hours. The DMF was removed via rotary evaporation and the residue was diluted with water and extracted with DCM. The organic phase was dried over sodium sulfate and the solvent was removed under reduced pressure to afford methyl 5,6-dimethoxybenzo[b]thiophene-2-carboxylate as a light yellow solid (1.0 g, 67%). LC-MS (ES-API); rt 8.48 min; m/z calculated for C 12 H 12 0 S [M+Naf 275.0, found 275.0.

5, 6-Dimethoxybenzo[b]thiophene-2-carboxylic acid

To a stirred solution of methyl 5,6-dimethoxybenzo[6]thiophene-2-carboxylate (1.0 g, 4 mmol) in MeOH (20 mL) and THF (10 mL) was added 2N NaOH (10 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for 2 hours before adjusting the pH value to 3 with 1 N HCI and diluting with water. The resulting suspension was filtered and the solid was dried under vacuum to afford 5,6-dimethoxybenzo[b]thiophene-2-carboxylic acid as a white solid (890 mg, 94%). LC-MS (ES-API); rt 8.17 min; m/z calculated for CiiH 10 O 4 S [M+Na] + 261.0, found 260.9.

Methyl 2-(5, 6-dimethoxybenzo[b]thiophene-2-carboxamido)benzoate

To a stirred solution of 5,6-dimethoxybenzo[b]thiophene-2-carboxylic acid (714 mg, 3 mmol) and DMF (20 μΙ) in anhydrous DCM (20 mL) was added oxalyl chloride (0.5 mL, 6 mmol) dropwise at 0°C. The mixture was then allowed to warm to room temperature and stirring was continued until the acid was consumed completely (about 1 h). The solvent was removed under reduced pressure and the crude 5,6-dimethoxybenzo[b]thiophene-2-carbonyl chloride was used directly in the next step.To a stirred solution of methyl 2- aminobenzoate (500 mg, 3.3 mmol) and Ε¾Ν (1.25 mL, 9 mmol) in anhydrous DCM (20 mL) was added 5,6-dimethoxybenzo[b]thiophene-2-carbonyl chloride in anhydrous DCM (2 mL) dropwise at 0°C. The resulting mixture was stirred at room temperature overnight. The reaction was quenched with water and extracted with DCM. The organic extract was dried over sodium sulfate, the solvent was removed under reduced pressure and the crude product was purified by re-crystallization from EtOH to give methyl 2-(5,6- dimethoxybenzo[b]thiophene-2-carboxamido)benzoate as a white solid (650 mg, 55%). LC-MS (ES-API); rt 9.38 min; m/z calculated for CigHiyNOsS [M+H] + 372.1, found 372.1. -(5, 6-Dimethoxybenzo[b]thiophene-2-carboxamido)benzoic acid

To a stirred solution of methyl 2-(5,6-dimethoxybenzo[to]thiophene-2- carboxamido)benzoate (650 mg, 1.75 mmol) in MeOH (9 mL) and THF (20 mL) was added 2N NaOH (9 mL) dropwise at room temperature. The resulting solution was stirred at room temperature for 2 hours. The reaction was quenched with 1N HCI and diluted with water. The resulting suspension was filtered and the solid was dried under vacuum to afford 2-(5,6- dimethoxybenzo[b]thiophene-2-carboxamido)benzoic acid (550 mg, 88%). LC- MS (ES-API); rt 9.22 min; m z calculated for Ci 8 H 15 N0 5 S [M+Naf 380.1 , found 380.0.

2-(1 , 1-Dioxo-5, 6 limethoxybenzo[b]thiophene-2-carboxamido)benzoic acid

FT128

To a stirred solution of 2-(5,6-dimethoxybenzo[b]thiophene-2- carboxamido)benzoic acid (500 mg, 1.4 mmol) in CH 3 COOH (100 mL), CH 3 CN (50 mL) and DCM (50 mL) was added m-Chloroperoxybenzoic acid (1.2 g, 7 mmol) at room temperature. The resulting solution was stirred at room temperature for 48 hours until the starting material was consumed completely by TLC analysis. The reaction was quenched with water and extracted with DCM. The combined organic phase was washed with water, brine and dried over sodium sulfate. The solvent was removed under reduced pressure and the residue was purified by flash column using DCM/MeOH (from 30 : 1 to 10 : 1) as eluent. Further purification by re-crystallization from CH 3 CN provided 2-(1,1- dioxo-5,6-dimethoxybenzo[b]thiophene-2-carboxamido)benzoic acid (220 mg, 40%). H-NMR (400 MHz, DMSO-d 6 ) δ 3.89 (s, 3H), 3.92 (s, 3H), 7.26 (app t, J = 7.6 Hz, 1H), 7.48 (s, 1H), 7.61 (s, 1H), 7.67 (m, 1H), 8.05 (dd, J = 8.0, 1.2 Hz, 1 H), 8.14 (s, 1H), 8.47 (d, J = 8.4 Hz, 1H), 11.87 (s, 1 H); LC-MS (ES-API); rt 8.91 min; m/z calculated for C 18 H 15 N0 7 S [M+H] + 390.1 , found 390.1. 2-(6, 7-Dimethoxy-1,2-dihydronaphthalene-3-carboxamido)benzoic acid (FT129)

6, 7-Dimethoxy-1,2,3,4-tetrahydmnaphthalen-1-ol To a stirred solution of 6,7-dimethoxy-3,4-dihydronaphthalen-1(2H)-one (3.09 g, 15 mmol) in MeOH (50 mL) was added NaBH (680 mg, 18 mmol) portion wise over 0.5 hour. The resulting mixture was stirred at room temperature overnight. The reaction was quenched with 1N HCI, diluted with water and extracted with DCM. The combined organic phase was washed with brine, dried over sodium sulfate and the solvent was removed under reduced pressure to afford 6,7- dimethoxy-1,2,3,4-tetrahydronaphthalen-1-ol as a yellow oil (2.9 g, 93%). LC- MS (ES-API); rt 7.96 min; m/z calculated for C 12 H 16 0 3 [M+Na] + 231.1, found 231.1.

6, 7-Dimethoxy-3, 4-dihydronaphthalene-2-carbaldehyde

A solution of 6,7-dimethoxy-1 ,2,3,4-tetrahydronaphthalen-1-ol (208 mg, 1 mmol) in dry DMF (5 mL) was cooled in an ice bath and phosphoryl chloride (0.2 mL, 2.3 mmol) was added dropwise. The reaction mixture was heated at 90°C for 3 hours, then cooled and treated with a cold saturated aqueous solution of sodium acetate (2 mL) and stirred at room temperature for 10 min. Water was added and the mixture was extracted with DCM. The combined organic phase was washed with brine and dried over sodium sulfate. The solvent was then removed under reduced pressure to afford 6,7-dimethoxy-3,4- dihydronaphthalene-2-carbaldehydeas white solid (160 mg, 46%). LC-MS (ES- API); rt 8.23 min; m/z calculated for C 3 H 14 0 3 [M+H] + 219.1 , found 219.0.

6, 7-Dimethoxy-3,4-dihydmnaphthalene-2-carboxylic acid

A mixture of 6,7-dimethoxy-3,4-dihydronaphthalene-2-carbaldehyde (2.2 g, 10 mmol) in EtOH (30 mL) was treated with a solution of silver nitrate (5.0 g, 30 mmol) in water (16 mL). A solution of NaOH (6.0 g, 150 mmol) in water (50 mL) was then added with continuous stirring and the resulting mixture was stirred at room temperature overnight. The reaction mixture was then extracted with DCM and combined organic phase was washed with brine and dried over sodium sulfate. The solvent was removed under reduced pressure and the residue was purified by flash column using petroleum. ether/ethyl acetate (from 6:1 to 3: 1, v:v) as the eluent to give 6,7-dimethoxy-3,4-dihydronaphthalene-2-carboxylic acid (1.8 g, 77%). LC-MS (ES-API); rt 8.23 min; m/z calculated for Ci 3 H 14 0 4 [M+H] + 235.1, found 235.0.

Methyl 2-(6, 7-dimethoxy-1,2-dihydronaphthalene-3-carboxamido)benzoate

A mixture of 6 I 7-dimethoxy-3 I 4-dihydronaphthalene-2-cari30xylic acid (476 mg, 2 mmol) and SOCI 2 (1.5 mL, 20 mmol) in toluene (20 mL) was heated at 50°C for 1 hour. The solvent was then removed under reduced pressure to give 6,7- dimethoxy-3,4-dihydronaphthalene-2-carbonyl chloride which was used directly in the next step without further purification: To a stirred solution of methyl 2- aminobenzoate (332 mg, 2.2 mmol) and Ε¾Ν (0.8 mL, 6 mmol) in anhydrous DCM (20 mL) was added 6,7-dimethoxy-3,4-dihydronaphthalene-2-carbonyl chloride in anhydrous DCM (2 mL) dropwise at 0°C. The resulting mixture was stirred at room temperature overnight. The reaction was quenched with water and extracted with DCM. The combined organic phase was dried over sodium sulfate, the solvent was removed under reduced pressure and the residue was purified by flash column, eluting with petroleum ether/ethyl acetate (10: 1) to give methyl 2-(6,7-dimethoxy-1 ,2-dihydronaphthalene-3-carboxamido)benzoate (330 mg, 45%). LC-MS (ES-API); rt 9.27 min; m/z calculated for C 2 iH 2 iN0 5 [M+Naf 390.1 , found 390.1.

2-(6, 7-Dimethoxy- 1, 2-dihydronaphthalene-3-carboxamido)benzoic acid (FT129)

FT129 To a stirred solution of methyl 2-(6,7-dimethoxy-3,4-dihydronaphthalene-2- carboxamido) benzoate (440 mg, 1.2 mmol) in MeOH (6 mL) and THF (3 mL) was added 2N NaOH (3 mL) dropwise at room temperature. The resulting solution was stirred at room temperature for 2 hours, then quenched with 1 N HCI and diluted with water. The resulting suspension was filtered and the solid was dried under vacuum to give 2-(6,7-dimethoxy-1,2-dihydronaphthalene-3- carboxamido)benzoic acid (350 mg, 83%). H-NMR (400 MHz, DMSO-d 6 ) δ 2.59 (t, J = 8 4 Hz, 2H), 2.83 (t, = 8.4 Hz, 2H), 3.75 (s, 3H), 3.79 (s, 3H), 6.90 (s, 1H), 6.97 (s, 1 H), 7.15 (app t, J = 7.6 Hz, 1H), 7.39 (s, 1H), 7.62 (m, 1 H), 8.03 (dd, J = 8.0, 1.6 Hz, 1H), 8.69 (d, J - 8.4 Hz, 1H), 11.89 (s, 1H); LC-MS (ES-API); rt 9.22 min; m z calculated for C20H19NO5 [M+Na] + 376.1, found 376.1.

2-(6, 7-Dimethoxy-1 ,2, 3, 4-tetrahydronaphthalene-3-carboxamido)benzoic acid (FT130)

FT130

A mixture of 2-(6,7-dimethoxy-3,4-dihydronaphthalene-2-carboxamido)benzoi c acid (350 mg, 1 mmol) and 10% Pd/C (35 mg) in MeOH (50 mL) and THF (50 mL) was stirred at room temperature under H 2 overnight. The mixture was filtered and the filtrate was concentrated under reduced pressure to afford 2- (6,7-dimethoxy-1 ,2,3,4-tetrahydronaphthalene-3-carboxamido)benzoic acid (250 mg, 71%). H-NMR (400 MHz, DMSO-d 6 ) δ 1.70-1.85 (m, 1H), 2.10-2.21 (m, 1H), 2.64-2.82 ,(m, 3H), 2.82-3.00 (m, 2H), 3.71 (s, 6H), 6.66 (s, 1H), 6.68 (s, 1 H), 7.16 (app t, J = 7.6 Hz, 1H), 7.60 (app t, J = 8.0 Hz, 1H), 8.00 (d, J = 7.2 Hz, 1H), 8.53 (d, J = 8.4 Hz, 1H), 11.32 (s, 1H); LC-MS (ES-API); rt 8.96 min; m/z calculated for C20H21NO5 [M+H] + 356.1 , found 356.1.

(Z)-2-(2-(6 -Dimethoxy-1-oxo-2,3-dihydmisoquinolin-4(1H)- ylidene)acetamido)benzoic acid (FT131)

2-lodo-4,5-dimethoxybenzoic acid

To a stirred solution of 2-iodo-4,5-dimethoxybenzaldehyde (1.0 g, 3.42 mmol) in

CH 3 CN (25 mL) was added a solution of KMn0 4 (0.76 g, 4.79 mmol) in water

(15 mL) dropwise at 0°C. The resulting mixture was stirred at room temperature overnight. After the reaction was complete, the pH was adjusted to <7 with 1 N

HCI. The mixture was extracted with DCM and the organic layers combined, washed with brine, dried over sodium sulfate, and evaporated to give 2-lodo-

4,5-dimethoxybenzoic acid (0.89 g, 85%). Ethyl 2-(2-iodo-4,5-dimethoxybenzamido)acetate

o '

Meo A, EDC, HOBt NEt 3 ■ J , " O

To a stirred solution of 2-iodo-4,5-dimethoxybenzoic acid (24.6 g, 80 mmol), ethyl 2-aminoacetate hydrochloride (11 g, 88 mmol), EDCI (17 g, 88 mmol) and HOBt (14 g, 88 mmol) in dry DCM (500 mL) was added Et 3 N (55 mL, 400 mmol) dropwise at rt. The resulting mixture was stirred at room temperature overnight then the reaction was quenched with water, and thoroughly extracted with DCM. The combined organic phases were washed with brine and dried over sodium sulfate. The solvent was removed under reduced pressure and the residue was purified by flash chromatography using petroleum ether/ethyl acetate 3:1 (v:v) as eluent to give ethyl 2-(2-iodo-4,5-dimethoxybenzamido)acetate (29 g, 95%).

N-(2-Hydroxyethyl)-2-iodo-4,5-dimethoxybenzamide

UBH 4 (63 mg, 3 mmol) was added to a stirred solution of ethyl 2-(2-iodo-4,5- dimethoxybenzamido)acetate (379 mg, 1 mmol) in THF (2 mL) at -10°C. The reaction was stirred for 5 min then methanol (0.22 mL) was added dropwise. The reaction mixture was warmed to room temperature, stirred for 30 minutes and quenched by addition of water. The THF was removed under reduced pressure and the aqueous residue was thoroughly extracted with DCM. The combined organic phases were washed with brine, dried over sodium sulphate and evaporated to give the crude product which was purified by re- crystallization from EtOAc and hexane to give N-(2-hydroxyethyl)-2-iodo-4,5- dimethoxybenzamide (280 mg, 80%) LC-MS (ES-API); rt 7.60 min; m/z calculated for C H 14 IN0 [M+H] + 352.0, found 352.0.

(Z)-Ethyl 4-(2-iodo-4, 5-dimethoxybenzamido)but-2-enoate

A solution of dry DMSO (0.453 mL, 6.4 mmol) in DCM (2 mL) was added dropwise to a solution of oxalyl chloride (0.338 mL, 4 mmol) in DCM (5 mL) at - 78°C. The reaction was stirred for 30 min at -78°C then a solution of N-(2- hydroxyethyl)-2-iodo-4,5-dimethoxybenzamide (702 mg, 2 mmol) in DCM (3 mL) was added via cannula. The mixture was stirred at -78°C for 1 hour then dry Ε¾Ν (1.7 mL, 12 mmol) was added slowly. Stirring was continued at -78°C for 1 hour by which time the formation of the aldehyde was complete (monitored by TLC). Solid Ph 3 P=CHCOOEt (1.0 g, 3 mmol) was then added and the reaction was allowed to warm slowly to room temperature overnight. The solvent was removed under reduced pressure and the crude product was purified by flash column using petroleum ether/ethyl acetate (from 6:1 to 3:1) as eluent to give (Z)-ethyl 4-(2-iodo-4,5-dimethoxybenzamido)but-2-enoate (280 mg, 34%). LC-MS (ES-API); rt 8.46 min; m/z calculated for C 15 Hi8l 0 5 [M+H]* 420.0, found 420.0. (Z)-Ethyl 2-(6 -dimethoxy-1-oxo-2,3-dihydroisoquinolin-4(1H)-

/ A stirred solution of Pd(OAc) 2 (16 mg, 0.07 mmol), PPh 3 (61 mg, 0.23 mmol), Ε¾Ν (0.58 mL, 4.2 mmol) and (Z)-ethyl 4-(2-iodo-4,5-dimethoxybenzamido)but- 2-enoate (860 mg, 2.1 mmol) in dry CH 3 CN (20 mL) was heated at 70°C ovemight under an argon atmosphere. The reaction was allowed to cool to room temperature and then concentrated under reduced pressure. The residue was purified by flash chromatography using DCM/MeOH 100:1 (v:v) as eluent to give (Z)-ethyl 2-(6 I 7-dimethoxy-1-oxo-2,3-dihydroisoquinolin-4(1H)- ylidene)acetate (510 mg, 83%). LC-MS (ES-API); rt 7.56 min; m/z calculated for Ci 5 H 17 N0 5 [M+H] + 292.1, found 292.1.

(Z)-2-(6 -Dimethoxy-1^xo-2,3 lihyd isoquinolin-4(1H)-yliden

To a stirred solution of (Z)-ethyl 2-(6,7-dimethoxy-1-oxo-2,3-dihydroisoquinolin - 4(1H)-ylidene)acetate (1.45 g, 5 mmol) in MeOH (20 mL) and THF (40 mL) was added 2N LiOH (5 mL) dropwise at room temperature and the resulting mixture was stirred at room temperature ovemight. The reaction was quenched by addition of 1N HCI and the solvents were removed under reduced pressure. The mixture was diluted with water and cooled and the. precipitate collected by filtration to afford (Z)-2-(6,7-dimethoxy-1-oxo-2,3-dihydroisoquinolin-4(1H)- ylidene)acetic acid (1.0 g, 77%). LC-MS (ES-API); rt 7.21 min; m/z calculated for Ci 3 H 13 N0 5 [M-H]- 262.0, found 262.1.

(Z)-Methyl 2-(2-(6, 7-dimethoxy-1-oxo-2,3-dihydroisoquinolin-4(1H)- ylidene)acetamido)benzoate

To a stirred solution of (2)-2-(6,7-dimethoxy-1-oxo-2,3-dihydroisoquinolin-4(1H)- ylidene)acetic acid (789 mg, 3 mmol), methyl 2-aminobenzoate (544 mg, 3.6 mmol), HOBt (587 mg, 3.6 mmol), EDCI (688 mg, 3.6 mmol) and DMAP (439 mg, 3.6 mmol) in dry DMF (30 ml_) was added Et 3 N (2 mL, 15 mmol) dropwise at room temperature. The resulting mixture was stirred at room temperature ovemight then the reaction was quenched with water and extracted with DCM. The combined organic phases were washed with brine, dried over sodium sulfate and evaporated to give the crude product which was purified by flash chromatography using DCM/MeOH 40:1 (v:v) as eluent to give (Z)-methyl 2-(2- (6,7-dimethoxy-1-oxo-2,3-dihydroisoquinolin-4(1H)-ylidene)ac etamido)benzoate (280 mg, 80%) LC-MS (ES-API); rt 8.39 min; m/z calculated for C 2 iH2oN 2 0 6 [M+Naf 419.1 , found 419.1.

(Z)-2-(2-(6, 7-Dimethoxy-1-oxo-2, 3-dihydmisoquinolin-4(1H)-

FT131

To a stirred solution of (Z)-methyl 2-(2-(6,7-dimethoxy-1-oxo-2,3- dihydroisoquinolin - (1 -V)-ylidene)acetamido)benzoate (140 mg, 0.35 mmol) in MeOH (4 mL) and THF (8 mL) was added 2N LiOH (0.7 mL) dropwise at room temperature. The resulting mixture was stirred at room temperature ovemight then the reaction was quenched by addition of 1N HCI and the solvent was removed under reduced pressure. The mixture was diluted with water, cooled and the precipitate collected by filtration to afford (Z)-2-(2-(6,7-dimethoxy-1-oxo- 2,3-dihydroisoquinolin-4(1H)-ylidene)acetamido)benzoic acid (125 mg, 93%). H -NMR (400 MHz, DMSO-d e ) δ 3.80, (s, 3H), 3.84 (s, 2H), 3.85 (s, 3H), 7.00 (s, 1H), 7.10 (m, 1H), 7.22 (d, J = 6.0 Hz, 1 H), 7.56 (m, 1H), 7.61 (s, 1H), 7.90 (dd, J = 8.0, 1.6 Hz, 1H), 8.57 (dd, J = 8.4, 0.8 Hz, 1H), 11.38 (d, J = 5.6 Hz, 1H), 13.54 (br s, 1H); LC-MS (ES-API); rt 8.27 min; m/z calculated for C 2 oHi 8 N 2 0 6 [M+H] + 383.1, found 383.1. (E)-2-(3,4-Dimethoxystyryl)benzo[d]oxazole-4-carboxylic acid (FT132) Methyl 2-amino-3-hydroxybenzoate

To a stirred solution of 2-amino-3-hydroxybenzoic acid (1.22 g, 8.0 mmol) in MeOH (80 mL) was added SOCI 2 (9.52g, 80.0 mmol) dropwise at 0°C. The mixture was then heated at reflux overnight. Most of the methanol was removed and the residue was partitioned between DCM and saturated aqueous NaHC0 3 . The organic phase was washed with brine, dried over sodium sulfate and the solvent was removed under reduced pressure to give methyl 2-amino- 3-hydroxybenzoate (1.27 g, 95%). LC-MS (ES-API); rt 8.09 min; m/z calculated for C 8 H 9 N0 3 [M+H] + 168.1, found 168.1.

Methyl

A solution of methyl 2-amino-3-hydroxybenzoate (167 mg, 1.0 mmol), acetyl chloride (86 mg, 1.1 mmol) and triethylamine (101 mg, 1.1 mmol) in xylene (10 mL) was stirred at 0°C for 2 h. Pyridine (20 mg, 0.25 mmol) and TsOH (43 mg, 0.25 mmol) were then added and the mixture was heated at reflux overnight. The mixture was then cooled to room temperature, diluted with water and extracted with ethyl acetate. The combined organic extracts were washed with water, brine, then dried over sodium sulfate and the solvent was removed under reduced pressure. The residue was purified by flash chromatography using petroleum ether/ethyl acetate 5:1 (v:v) as eluent to give methyl 2- methylbenzo[d]oxazole-4-carboxylate (151 mg, 79%). LC-MS (ES-API); rt 8.08 min; m/z calculated for C10H9NO3 [M+H] + 192.1 , found 192.1.

(E)-Methyl 2-(3, 4-dimethoxystyryl)benzo[d]oxazole-4-carboxylate

A solution of methyl 2-methylbenzo[d]oxazole-4-carboxylate (0.63 g, 4.12 mmol) and 3,4-dimethoxybenzaldehyde (0.68 g, 4.12 mmol) in THF (25 mL) and t- BuOH (5 mL) was cooled to -50°C under nitrogen and treated with a solution of t-BuOK in THF (1.0 mol/L, 4.94 mL, 4.94 mmol) at such a rate that the internal reaction temperature did not exceed -46C After 2 hours at -50°C, the cooling bath was removed and the mixture was stirred at room temperature for 12 h in the dark. Water was added and the mixture was made slightly acidic by addition of a 1N HCI solution then extracted with DCM. The organic layers were combined, washed with water and brine then dried over sodium sulfate and the solvent was removed under reduced pressure. The residue was purified by flash chromatography using petroleum ether: ethyl acetate 2:1 (v:v) as eluent to give (E)-methyl 2-(3,4-dimethoxystyryl)benzo[d]oxazole-4-carboxylate (0.30g, 27%). LC-MS (ES-API); rt 9.25 min; m/z calculated for C 19 H 17 N0 5 [M+H] + 340.1, found 340.1. -2- 3, 4-Dimethox st r l benzo[d]oxazole- -carboxylic acid (FT132)

FT132

To a stirred solution of (E)-methyl 2-(3,4-dimethoxystyryl)benzo[dloxazole-4- carboxylate (240mg, 0.71 mmol) in THF (10mL) and MeOH (5mL) was added 2 N NaOH solution (1.77mL) dropwise at room temperature. The resulting mixture was stirred at room temperature for two hours then quenched with a 1 N HCI solution. The mixture was then extracted with DCM and the organic phase was washed with saturated aqueous NaCI solution and dried over sodium sulfate. The solvent was then removed under reduced pressure to give (E)-2-(3,4- Dimethoxystyryl)benzo[d]oxazole-4-carboxylic acid (170 mg, 74%). H-NMR (400MHz, DMSO-d 6 ) δ 3.81 (s, 3H), 3.85 (s, 3H), 7.03 (d, J = 8.0 Hz, 1H), 7.33- 7 39 (m, 2H), 7.46 (app t, J = 8.0Hz, 1H), 7.50 (d, J = 2.0Hz, 1H), 7.80 (d, J = 16.0Hz, 1H), 7.87 (dd, = 8.0, 1.2Hz, 1H), 7.93 (dd, J = 8.0, 1.2Hz, 1H); LC-MS (ES-API): rt 8.88 min; m/z calculated for CieHi 5 N0 5 [M-Hf 324.1 , found 324.1. 097 rat mesanaial cells A well-characterised cloned rat mesangial cell line (Kakizaki Y, Kraft N, Atkins RC: Differential control of mesangial cell proliferation by interferon-gamma. Clin Exp Immunol 85:157-163, 1991) was cultured in DMEM (5mM glucose) with 5% FBS, 100U/mL penicillin, and 100ug/mL streptomycin in humidified 5% C02 atmosphere at 37C. Cells were used up to passage 40.

Proline incorporation Assay

Cells were plated into 24-well culture dishes in DMEM/5%FBS and allowed to adhere overnight. The subconfluent cells were then starved overnight in DMEM/0.1%FBS and 150uM L-ascorbic acid, prior to pre-treatment with or without FibroTech compounds for 4 hours at 10, 30 or 100uM in 0.1% DMSO, final concentration. TGF-beta 1 at 5ng/mL (Peprotech) and tritiated proline (Perkin-Elmer, (2,3,4,5- 3 H)-proline) at 1 uCi/mLwere added and the incubation continued for a further 44 hours. Treatments were performed in triplicate.

After the incubation, cells were placed on ice and washed three times with ice cold PBS. They were then incubated on ice with 10% TCA for 30 minutes, followed by a final wash in cold 10% TCA. The cells were solubilized in 0.75m|_ of 1M NaOH at 37C for 45 minutes, or overnight at 4C. Aliquots of the solubilized cells were neutralized in an equal amount of 1M HCI and counted in scintillant in a beta counter. Aliquots of neutralized solubilized cells were assayed for protein using the BioRad Bradford Protein Assay. Proline incorporation was normalized for protein content (cpm 3 H-proline incorporated per ug protein).

In order to compare the results of experiments performed on different days, the percentage inhibition of proline incorporation was calculated for each treatment by setting the TGF treatment at 0% inhibition and the control (incubation with media alone) at 100% inhibition. Mesangial cells

ppt indicates compound precipitated during assay. The details of specific embodiments described in this invention are not to be construed as limitations. Various equivalents and modifications may be made without departing from the essence and scope of this invention, and it is understood that such equivalent embodiments are part of this invention.