Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ARGININE PEPTIDE ANALOGS USEFUL AS FIBROBLAST GROWTH FACTOR ANTAGONISTS
Document Type and Number:
WIPO Patent Application WO/1999/052936
Kind Code:
A2
Abstract:
Pharmaceutical compositions containing arginine amides, peptides or analogs and methods for using the pharmaceutical compositions for modulating the activity of the FGF family of peptides are provided. Methods for inhibiting the binding of an FGF peptide to an FGF receptor by contacting the receptor with arginine amides, peptides or analogs of formulae (I) or (II) are provided. Methods for treating FGF-mediated disorders by administering effective amounts of one or more of these compounds or pharmaceutically acceptable derivatives thereof that inhibit the activity of one or more FGF peptides are also provided.

Inventors:
YALAMOORI VENTKATACHALAPATHI
KALYANARAMAN RAMNARAYAN
SCHOVE LAURA
BALAJI VITUKUDI NARAYANAIYENGA
CHAN MING FAI
Application Number:
PCT/JP1999/001844
Publication Date:
October 21, 1999
Filing Date:
April 07, 1999
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
EISAI CO LTD (JP)
International Classes:
A61K31/15; A61K31/155; A61K31/16; C07C281/18; C07K5/065; C07K14/71; A61K38/00; (IPC1-7): C07K5/06; A61K31/15; A61K31/155; A61K31/16; A61K38/05; C07C281/18
Foreign References:
ES2039161A
GB774794A1957-05-15
GB927659A1963-05-29
US4665085A1987-05-12
Other References:
L BOURGEOIS ET AL.: "Serpin-derived peptide substrates for investigating the substrate specificity of human tissue kallikrein hK1 and hK2" JOURNAL OF BIOLOGICAL CHEMISTRY., vol. 272, no. 47, 21 November 1997 (1997-11-21), pages 29590-29595, XP002123201 AMERICAN SOCIETY OF BIOLOGICAL CHEMISTS, BALTIMORE, MD., US ISSN: 0021-9258
CHEMICAL ABSTRACTS, vol. 125, no. 17, 21 October 1996 (1996-10-21) Columbus, Ohio, US; abstract no. 215442, K AIBE ET AL.: "Substrate specificity of recombinant osteoclast-specific cathepsin K from rabbits" XP002123202 & BIOL. PHARM. BULL., vol. 19 , no. 8, 1996, pages 1026-1031,
CHEMICAL ABSTRACTS, vol. 109, no. 21, 21 November 1988 (1988-11-21) Columbus, Ohio, US; abstract no. 186027, R A HAYMAN ET AL.: "Characterization of an acid collagenase from human osteoclastomas" XP002123203 & BIOCH. SOC. TRANS., vol. 16, no. 6, 1988, pages 1064-1065,
CHEMICAL ABSTRACTS, vol. 97, no. 19, 8 November 1982 (1982-11-08) Columbus, Ohio, US; abstract no. 158495, K UCHIDA ET AL.: "Estimation of plasma and glandular kallikrein activity using fluorogenic substrates and its clinical applications" XP002123204 & AGENTS ACTIONS UPPL., vol. AAS9, 1982, pages 91-96,
Attorney, Agent or Firm:
Kawaguchi, Yoshio (Shinjuku 1-chome Shinjuku-ku Tokyo, JP)
Baldock, Sharon Claire (Wade & Tennant 27 Furnival Street London EC4A 1PQ, GB)
Download PDF:
Claims:
CLAIMS
1. A pharmaceutical composition, comprising, in a pharmaceutically acceptable vehicle, an effective amount of a compound of formula 1: or a pharmaceutically acceptable derivative thereof, wherein: R', R2, R3, R4, R5 and R6 are each independently selected from (i) or (ii) as follows: (i) R', R3 and R5 are each independently selected from alkyl, alkenyl, alkynyl, aryl, cycloalkyl, aralkyl and heteroaralkyl, and are unsubstituted or substituted with one or more substituents each independently selected from Z, and R2, R4 and R6 are each independently selected from hydrogen and alkyl; or (ii) one or more of R'and R2, or R3 and R4, or R5 and R6 form alkylene or alkenylene which is unsubstituted or substituted with one or more substituents each independently selected from Z, and the others are selected as in (i); A is hydrogen, alkoxycarbonyl, aryloxycarbonyl, heteroaryloxycarb onyl, aralkoxycarbonyl, heteroaralkoxycarbonyl, alkylcarbonyl, arylcar bonyl, heteroarylcarbonyl, aralkylcarbonyl or heteroaralkylcarbonyl; and D is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl, and is unsubstituted or substituted with one or more substituents each independently selected from Z; Z is halo, hydroxy, amino, azido, guanidino, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, alkoxycarbonyl, aryloxycarbonyl or heteroaryloxycarbonyl, or any two Z substituents may form alkylene, alkenylene, alkylenedioxy, alkylenethioxyoxy or alkylenedithioxy, and is unsubstituted or substituted with one or more substituents each independently selected from Y; and Y is halo, amino, alkylamino, dialkylamino, arylamino, diarylamino, acylamino, azido, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy or heteroaryloxy.
2. The pharmaceutical composition of claim 1, wherein: A is hydrogen, arylalkoxycarbonyl or arylcarbonyl; and D is aryl, heteroaryl, aralkyl or heteroaralkyl, and is unsubstituted or substituted with one or more substituents each independently selected from Z.
3. The pharmaceutical composition of claim 1 or claim 2, wherein: A is hydrogen, benzyloxycarbonyl (Cbz), 9fluorenylmethoxycar bonyl (Fmoc) or benzoyl (Bz).
4. The pharmaceutical composition of any of claims 13, wherein: D is benzyl (Bn), 4nitrophenyl, 1methoxy3naphthyl (4MbNA) or 4methyl7cumarinyl (AMC).
5. The pharmaceutical composition of any of claims 14, wherein: R', R2, R3, R4, R5 and R6 are selected as in (i) or (ii) as follows: (i) R', R3 and R5 are each independently selected from alkyl and aralkyl, and are unsubstituted or substituted with one or more substituents each independently selected from Z, and R2, R4 and R6 are each hydrogen; or (ii) one of R'and R, or R3 and R4, or R5 and R6 form alkylene, and the others are selected as in (i).
6. The pharmaceutical composition of any of claims 15, wherein: R', R2, R3, R4, R5 and R6 are selected as in (i) or (ii) as follows: (i) R', R3 and R5 are each independently selected from benzyl, 3 guanidinylpropyl, 4hydroxyphenylmethyl, methyl, 2methylpropyl, isopropyl and 4aminobutyl, and R2, R4 and R6 are each hydrogen; or (ii) R3 and R4, or R5 and R6 form propylene, and the others are selected as in (i).
7. The pharmaceutical composition of any of claims 16, wherein the compounds contain amino acids selected from phenylalanine, arginine, tyrosine, alanine, valine, leucine, lysine and proline.
8. The pharmaceutical composition of any of claims 17, wherein the compound is selected from the group consisting of (Fmoc)PheArg NHBn, (Fmoc)PheDArgNHBn, (Fmoc)DPheArgNHBn, (Fmoc)DPhe DArgNHBn, (Fmoc)TyrArgNHBn, (Fmoc)AlaArgNHBn, (Fmoc)Leu ArgNHBn, (Fmoc)ArgPheNHBn, (Fmoc)Arg (Pmc)PheN H Bn, (Fmoc)D ArgPheNHBn, (Fmoc)ArgDPheNHBn, (Fmoc)DArgDPheNHBn, (Fmoc)LysPheNHBn, CbzArgAMC, CbzArgArgAMC, CbzPhePro Arg4MbNA, CbzPheArgAMC, CbzDPheArgAMC, BzPheValArg NH (4nitrophenyl), NaBzArgAMC, HProPheArgAMC and HDPhe ProArg4MbNA.
9. A pharmaceutical composition, comprising, in a pharmaceutically acceptable vehicle, an effective amount of a compound of formula li: or pharmaceutically acceptable derivatives thereof, wherein: Ar'is higher alkylene, or is monocyclic or polycyclic arylene, alkenylarylene, arylalkenylene, heteroarylene, alkenylheteroarylene or heteroarylalkenylene, and is unsubstituted or substituted with one or more substituents each independently selected from Z; Z is halo, hydroxy, amino, azido, guanidino, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, alkoxycarbonyl, aryloxycarbonyl or heteroaryloxycarbonyl, or any two Z substitutents may form alkylene, alkenylene, alkylenedioxy, alkylenethioxyoxy or alkylenedithioxy, and is unsubstituted or substituted with one or more substituents each independently selected from Y; Y is halo, amino, alkylamino, dialkylamino, arylamino, diarylamino, acylamino, azido, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy or heteroaryloxy; R7, R8, R9, R10, R11 and R12 are each independently selected from (i) or (ii) as follows: (i) R7 is selected from hydrogen and alkyl, R8 and R9 are hydrogen, R'° is hydrogen, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aralkyl or heteroaralkyl, R"is hydrogen, and R12 is hydrogen or Z, or (ii) any two of R7 R8, R9, R°, R"and R12 together form alkylene, alkenylene, carbonylalkylene or carbonylalkenylene, and the remainder are selected as in (i); and m is 0 or 1 when Ar'is arylalkenylene or heteroarylalkenylene, or is 1.
10. The pharmaceutical composition of claim 9, wherein: R7, R8, R9, R10, R11 and R12 are each independently selected from (i) or (ii) as follows: (i) R7 is selected from hydrogen and alkyl, R8 and R9 are hydrogen, R10 is hydrogen, arylsulfonyl or aralkyl, R"is hydrogen, and R12 is hydrogen or Z; or (ii) any two of R7, R8, R9, R10, R11 and R12 together form alkylene or carbonylalkylene, and the remainder are selected as in (i).
11. The pharmaceutical composition of claim 9 or claim 10, wherein: R7, R8, R9, R10, R11 and R12 are each independently selected from (i) or (ii) as follows: (i) R7 is selected from hydrogen and alkyl, R8 and R9 are hydrogen, R10 is hydrogen, arylsulfonyl or aralkyl, R"is hydrogen, and R12 is hydrogen or Z; or (ii) R8, R9 and R10 are selected as in (i), and (a) R"is selected as in (i), and R7 and R12 together form alkylene or carbonylalkylene, or (b) R7 is selected as in (i), and R11 and R12 together form alkylene or carbonylalkylene.
12. The pharmaceutical composition of any of claims 911, wherein Ar'and R12 taken together are higher alkyl.
13. The pharmaceutical composition of any of claims 912, wherein: Ar1 and R12 taken together are monocyclic or bicyclic aryl, alkenylaryl, arylalkenylene or heteroarylalkenylene, and is unsubstituted or substituted with one or more substituents selected from Z.
14. The pharmaceutical composition of any of claims 913, wherein: Ar1 and R12 taken together are phenyl, 2phenylalkenyl, 2 thienylalkylene or naphthyl, and are substituted with one or more groups selected from chloro, methoxy, biphenyl, benzoyloxy, 4nitrophenyloxy carbonyl, 4acetylaminophenyloxycarbonyl, ethylene, methylene, fluoro, 2phenylethyloxy, 3,4dichlorobenzyloxy, 4chlorobenzyloxy, phenyloxy, pentyl, benzyloxy and 2,4dichlorobenzyloxy.
15. The pharmaceutical composition of any of claims 914, wherein: Ar1 and R12 taken together are phenyl, 2phenylethenyl, 2 thienylethylene or naphthyl.
16. The pharmaceutical composition of any of claims 915, wherein: R7, R8, R9, R10, R11 and R12 are each independently selected from (i), (ii) or (iii) as follows: (i) R7 is hydrogen or methyl, or forms ethylene or propylene with R12, R8 and R9 are each hydrogen, or together form ethylene or propylene, R10 is hydrogen, 4chlorophenylsulfonyl or benzyl, R"is hydrogen and R12 forms ethylene or propylene with R, or is hydrogen or Z; or (ii) R8, R9, R11 and R12 are selected as in (i), and R7 and R10 together form methylene; or (iii) R8, R9, R10 and R12 are selected as in (i), and R'and R" together form methylenecarbonyl.
17. The pharmaceutical composition of any of claims 916, wherein: Arl and R 12 taken together are selected from (2 phenylethyloxy) phenyl, 2,4dichlorophenylethen1yi, 3 (3,4 dichlorobenzyloxy) phenyl, 2 (4chlorobenzyloxy) phenyl, 4 phenyloxyphenyl, 1pentyl2phenylethenyl, 3,4dibenzyloxyphenyl, 2 (2,4dichlorobenzyloxy) phenyl, 2 (3, 4dichlorobenzyloxy) phenyl, 1 naphthyl, phenyl, 2,6dichlorophenyl, 2methoxyphenyl, (4 (3 phenyl) phenyl phenyi, 3benzoyloxyphenyl, 4nitrophenyloxycar bonylphenyl, 4acetylaminophenyloxycarbonylphenyl, 2ethylenylphenyl, 2methylenylphenyl, 3,4difluorophenyl, 2phenylethenyl and 2thienyl 1,2ethenylene.
18. The pharmaceutical composition of any of claims 917, wherein the compound is selected from the group consisting of 3 (2 phenylethoxy) acetophenone aminoimino hydrazone, 2,4dichloro benzylideneacetone aminoimino hydrazone, 3 (3, 4dichlorobenzyloxy) benzaldehyde aminoimino hydrazone, 2(4chlorobenzyloxy)benzaldehyde aminoimino hydrazone, 4phenyloxybenzaldehyde aminoimino hydrazone, 2pentylcinnamaldehyde aminoimino hydrazone, decanal aminoimino hydrazone, 3,4dibenzyloxybenzaldehyde aminoimino hydrazone, 2 (2,4 dichlorobenzyloxy) benzaldehyde aminoimino hydrazone, 2 (3, 4dichloro benzyloxy) benzaldehyde aminoimino hydrazone, 1naphthylcarbox aldehyde aminoimino hydrazone, benzaldehyde aminoimino hydrazone, 2,6dichlorobenzaldehyde aminoimino hydrazone, 2methoxybenzaldehyde aminoimino hydrazone, 4 (3phenylphenyl) acetophenone aminoimino hydrazone, 3benzoyloxyacetophenone aminoimino hydrazone, 4 (4nitro phenyloxycarbonyl) acetophenone aminoimino hydrazone, 4(4acetyl aminophenyloxycarbonyl) acetophenone aminoimino hydrazone, aceto phenone 4,5dihydro2imidazolyl hydrazone, acetophenone 3,4,5,6 tetrahydro2pyrimidinyl hydrazone, acetophenone 3benzyl4,5dihydro 2imidazolyl hydrazone, atetralone aminoimino hydrazone, aindanone aminoimino hydrazone, 6 (3, 4difluorophenyl)4,5dihydroimidazo [2,1c] 2,3,4,5tetrahydro1,2,4triazine, 5phenyl2 (4, 5dihydroimidazol2yl) 3,4dihydro3oxo2 (H)pyrazole, benzylideneacetone aminoimino hydrazone and 1 (1 (4chlorophenylsulfonyl)4, 5dihydroimidazol2yl)3 methyl5 (2thienyl) pyrazole.
19. The pharmaceutical composition of any of claims 118 that is formulated for topical or local application to the eye.
20. The pharmaceutical composition of any of claims 118 that is formulated for topical or local application to the skin.
21. The pharmaceutical composition of any of claims 118 that is formulated for intravenous, intramuscular or parenteral administration.
22. A method of treating an FGFmediated disorder, comprising administering to a mammal an effective amount of a composition of any of claims 118, wherein the effective amount is sufficient for the prevention or treatment of the FGFmediated disorder.
23. A method of treating FGFmediated ophthalmic disorders comprising contacting a portion of an eye with an effective amount of a pharmaceutical composition of any of claims 118, wherein the amount is sufficient for the prevention or treatment of the FGFmediated ophthalmic disorder.
24. A method of treating a mammal having an FGFmediated tumorigenic pathophysiological condition comprising administering a effective amount of a pharmaceutical composition of any of claims 118, wherein the amount is effective for the prevention or treatment of the FGFmediated tumorigenic pathophysiological condition.
25. A method of treating a mammal having FGFmediated rheumatoid arthritis comprising administering a effective amount of a pharmaceutical composition of any of claims 118, wherein the amount is effective for the prevention or treatment of the FGFmediated rheumatoid arthritis.
26. A method of treating a mammal having FGFmediated restenosis, InStent restenosis, or other vascular injury comprising administering a effective amount of a pharmaceutical composition of any of claims 118, wherein the amount is effective for the prevention or treatment of the FGFmediated restenosis, InStent restenosis, or other vascular injury.
27. An article of manufacture, comprising packaging material and a pharmaceutical composition of any of claims 118, contained within the packaging material, wherein the pharmaceutical composition is effective for treatment or prevention of an FGFmediated disorder, and the packaging material includes a label that indicates that the pharmaceutical composition is used for treatment or prevention of an FGFmediated disorder.
28. The method of claim 22, wherein the disease is selected from the group consisting of rheumatoid arthritis, Kaposi's sarcoma, restenosis, InStent restenosis, FGFmediated ophthalmic disorders, FGF mediated dermatological disorders, psoriasis, FGFmediated tumorigenic pathophysiological conditions, rheumatoid arthritis, proliferative diabetic retinopathies, diabetes and melanoma.
29. The method of claim 23, wherein the disease is selected from the group consisting of rheumatoid arthritis, Kaposi's sarcoma, restenosis, InStent restenosis, FGFmediated ophthalmic disorders, FGF mediated dermatoiogical disorders, psoriasis, FGFmediated tumorigenic pathophysiological conditions, rheumatoid arthritis, proliferative diabetic retinopathies, diabetes and melanoma.
30. The method of claim 24, wherein the disease is selected from the group consisting of rheumatoid arthritis, Kaposi's sarcoma, restenosis, InStent restenosis, FGFmediated ophthalmic disorders, FGF mediated dermatological disorders, psoriasis, FGFmediated tumorigenic pathophysiological conditions, rheumatoid arthritis, proliferative diabetic retinopathies, diabetes and melanoma.
31. A method for inhibiting the binding of an FGF peptide to an FGF receptor, comprising contacting the receptor with an FGF peptide and with an effective amount of a compound of formula I: or a pharmaceutically acceptable derivative thereof, wherein: R', R2, R3, R4, R5 and R6 are each independently selected from (i) or (ii) as follows: (i) R', R3 and R5 are each independently selected from alkyl, alkenyl, alkynyl, aryl, cycloalkyl, aralkyl and heteroaralkyl, and are unsubstituted or substituted with one or more substituents each independently selected from Z, and R2, R4 and R6 are each independently selected from hydrogen and alkyl; or (ii) one or more of R'and R2, or R3 and R4, or R5 and R6 form alkylene or alkenylene which is unsubstituted or substituted with one or more substituents each independently selected from Z, and the others are selected as in (i); A is hydrogen, alkoxycarbonyl, aryloxycarbonyl, heteroaryloxycarb onyl, aralkoxycarbonyl, heteroaralkoxycarbonyl, alkylcarbonyl, arylcar bonyl, heteroarylcarbonyl, aralkylcarbonyl or heteroaralkylcarbonyl; and D is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl, and is unsubstituted or substituted with one or more substituents each independently selected from Z; Z is halo, hydroxy, amino, azido, guanidino, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, alkoxycarbonyl, aryloxycarbonyl or heteroaryloxycarbonyl, or any two Z substituents may form alkylene, alkenylene, alkylenedioxy, alkylenethioxyoxy or alkylenedithioxy, and is unsubstituted or substituted with one or more substituents each independently selected from Y; Y is halo, amino, alkylamino, dialkylamino, arylamino, diarylamino, acylamino, azido, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy or heteroaryloxy; and wherein the contacting is effected prior to, simultaneously with or subsequent to contacting the receptor with the FGF peptide; and the amount is effective to decrease binding of the FGF peptide to the receptor.
32. A method for inhibiting the binding of an FGF peptide to an FGF receptor, comprising contacting the receptor with an FGF peptide and with an effective amount of a compound of formula ll: or pharmaceutically acceptable derivatives thereof, wherein: Ar1 is higher alkylene, or is monocyclic or polycyclic arylene, alkenylarylene, arylalkenylene, heteroarylene, alkenylheteroarylene or heteroarylalkenylene, and is unsubstituted or substituted with one or more substituents each independently selected from Z; Z is halo, hydroxy, amino, azido, guanidino, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, alkoxycarbonyl, aryloxycarbonyl or heteroaryloxycarbonyl, or any two Z substitutents may form alkylene, alkenylene, alkylenedioxy, alkylenethioxyoxy or alkylenedithioxy, and is unsubstituted or substituted with one or more substituents each independently selected from Y; Y is halo, amino, alkylamino, dialkylamino, arylamino, diarylamino, acylamino, azido, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy or heteroaryioxy; R7, R8, R9, R10, R1l and R12 are each independently selected from (i) or (ii) as follows: (i) R7 is selected from hydrogen and alkyl, R8 and R9 are hydrogen, R10 is hydrogen, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aralkyl or heteroaralkyl, R"is hydrogen, and R12 is hydrogen or Z, or (ii) any two of R7 R8, R9, R10, R"and R'2 together form alkylene, alkenylene, carbonylalkylene or carbonylalkenylene, and the remainder are selected as in (i); and m is 0 or 1 when Ar'is arylalkenylene or heteroarylalkenylene, or is 1; and wherein the contacting is effected prior to, simultaneously with or subsequent to contacting the receptor with the FGF peptide; and the amount is effective to decrease binding of the FGF peptide to the receptor.
33. A method for altering FGF receptormediated activity, comprising contacting FGF receptors with an effective amount of a compound of formula I: or a pharmaceutically acceptable derivative thereof, wherein: R', R2, R3, R4, R5 and R6 are each independently selected from (i) or (ii) as follows: (i) R', R3 and R5 are each independently selected from alkyl, alkenyl, alkynyl, aryl, cycloalkyl, aralkyl and heteroaralkyl, and are unsubstituted or substituted with one or more substituents each independently selected from Z, and R2, R4 and R6 are each independently selected from hydrogen and alkyl; or (ii) one or more of R'and R2, or R3 and R4, or R5 and R6 form alkylene or alkenylene which is unsubstituted or substituted with one or more substituents each independently selected from Z, and the others are selected as in (i); A is hydrogen, alkoxycarbonyl, aryloxycarbonyl, heteroaryloxycarb onyl, aralkoxycarbonyl, heteroaralkoxycarbonyl, alkylcarbonyl, arylcar bonyl, heteroarylcarbonyl, aralkylcarbonyl or heteroaralkylcarbonyl; and D is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl, and is unsubstituted or substituted with one or more substituents each independently selected from Z; Z is halo, hydroxy, amino, azido, guanidino, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, alkoxycarbonyl, aryloxycarbonyl or heteroaryloxycarbonyl, or any two Z substituents may form alkylene, alkenylene, alkylenedioxy, alkylenethioxyoxy or alkylenedithioxy, and is unsubstituted or substituted with one or more substituents each independently selected from Y; Y is halo, amino, alkylamino, dialkylamino, arylamino, diarylamino, acylamino, azido, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy or heteroaryloxy; and the mediated activity is altered compared to the activity in the absence of the compound.
34. A method for altering FGF receptormediated activity, comprising contacting FGF receptors with an effective amount of a compound of formula II: or pharmaceutically acceptable derivatives thereof, wherein: Ar'is higher alkylene, or is monocyclic or polycyclic arylene, alkenylarylene, arylalkenylene, heteroarylene, alkenylheteroarylene or heteroarylalkenylene, and is unsubstituted or substituted with one or more substituents each independently selected from Z; Z is halo, hydroxy, amino, azido, guanidino, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, alkoxycarbonyl, aryloxycarbonyl or heteroaryloxycarbonyl, or any two Z substitutents may form alkylene, alkenylene, alkylenedioxy, alkylenethioxyoxy or alkylenedithioxy, and is unsubstituted or substituted with one or more substituents each independently selected from Y; Y is halo, amino, alkylamino, dialkylamino, arylamino, diarylamino, acylamino, azido, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy or heteroaryloxy; R7, R8, R9, R10, R"and R12 are each independently selected from (i) or (ii) as follows: (i) R7 is selected from hydrogen and alkyl, R8 and R9 are hydrogen, R'° is hydrogen, alkylsulfonyl, aryisulfonyl, heteroarylsulfonyl, aralkyl or heteroaralkyl, R"is hydrogen, and R12 is hydrogen or Z, or (ii) any two of R7 R8, R9, Rlo, R"and R12 together form alkylene, alkenylene, carbonylalkylene or carbonylalkenylene, and the remainder are selected as in (i); and m is 0 or 1 when Ar1 is arylalkenylene or heteroarylalkenylene, or is 1; and the mediated activity is altered compared to the activity in the absence of the compound.
35. Use of a compound of formula I for the treatment of FGF mediated diseases, wherein formula I is: or a pharmaceutically acceptable derivative thereof, wherein: R', R2, R3, R4, R5 and R6 are each independently selected from (i) or (ii) as follows: (i) R', R3 and R5 are each independently selected from alkyl, alkenyl, alkynyl, aryl, cycloalkyl, aralkyl and heteroaralkyl, and are unsubstituted or substituted with one or more substituents each independently selected from Z, and R2, R4 and R6 are each independently selected from hydrogen and alkyl; or (ii) one or more of R'and R2, or R3 and R4, or R5 and R6 form alkylene or alkenylene which is unsubstituted or substituted with one or more substituents each independently selected from Z, and the others are selected as in (i); A is hydrogen, alkoxycarbonyl, aryloxycarbonyl, heteroaryloxycarb onyl, aralkoxycarbonyl, heteroaralkoxycarbonyl, alkylcarbonyl, arylcar bonyl, heteroarylcarbonyl, aralkylcarbonyl or heteroaralkylcarbonyl; and D is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl, and is unsubstituted or substituted with one or more substituents each independently selected from Z; Z is halo, hydroxy, amino, azido, guanidino, nitro, alkyl, aryl, <BR> <BR> <BR> heteroaryl, alkoxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, alkoxycarbonyl, aryloxycarbonyl or heteroaryloxycarbonyl, or any two Z substituents may form alkylene, alkenylene, alkylenedioxy, alkylenethioxyoxy or alkylenedithioxy, and is unsubstituted or substituted with one or more substituents each independently selected from Y; Y is halo, amino, alkylamino, dialkylamino, arylamino, diarylamino, acylamino, azido, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy or heteroaryloxy.
36. Use of a compound of formula II for the treatment of FGF mediated diseases, wherein formula II is: or pharmaceutically acceptable derivatives thereof, wherein: Ar'is higher alkylene, or is monocyclic or polycyclic arylene, alkenylarylene, arylalkenylene, heteroarylene, alkenylheteroarylene or heteroarylalkenylene, and is unsubstituted or substituted with one or more substituents each independently selected from Z; Z is halo, hydroxy, amino, azido, guanidino, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, alkoxycarbonyl, aryloxycarbonyl or heteroaryloxycarbonyt, or any two Z substitutents may form alkylene, alkenylene, alkylenedioxy, alkylenethioxyoxy or alkylenedithioxy, and is unsubstituted or substituted with one or more substituents each independently selected from Y ; Y is halo, amino, alkylamino, dialkylamino, arylamino, diarylamino, acylamino, azido, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy or heteroaryloxy; R7, R8, R9, R10, R11 and R12 are each independently selected from (i) or (ii) as follows: (i) R'is selected from hydrogen and alkyl, R8 and R9 are hydrogen, R'° is hydrogen, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aralkyl or heteroaralkyl, R"is hydrogen, and R12 is hydrogen or Z, or (ii) any two of R7 R8, R9, R'°, R"and R12 together form alkylene, alkenylene, carbonylalkylene or carbonylalkenylene, and the remainder are selected as in (i); and m is 0 or 1 when Ar'is arylalkenylene or heteroarylalkenylene, or is 1.
37. Use of a compound of formula I for the formulation of a medicament for the treatment of FGFmediated diseases, wherein formula I is: or a pharmaceutically acceptable derivative thereof, wherein: R', R2, R3, R4, R5 and R6 are each independently selected from (i) or (ii) as follows: (i) R', R3 and R5 are each independently selected from alkyl, alkenyl, alkynyl, aryl, cycloalkyl, aralkyl and heteroaralkyl, and are unsubstituted or substituted with one or more substituents each independently selected from Z, and R2, R4 and R6 are each independently selected from hydrogen and alkyl; or (ii) one or more of R'and R2, or R3 and R4, or R5 and R6 form alkylene or alkenylene which is unsubstituted or substituted with one or more substituents each independently selected from Z, and the others are selected as in (i); A is hydrogen, alkoxycarbonyl, aryloxycarbonyl, heteroaryloxycarb onyl, aralkoxycarbonyl, heteroaralkoxycarbonyl, alkylcarbonyl, arylcar bonyl; heteroarylcarbonyl, aralkylcarbonyl or heteroaralkylcarbonyl; and D is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl, and is unsubstituted or substituted with one or more substituents each independently selected from Z; Z is halo, hydroxy, amino, azido, guanidino, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, alkoxycarbonyl, aryloxycarbonyl or heteroaryloxycarbonyl, or any two Z substituents may form alkylene, alkenylene, alkylenedioxy, alkylenethioxyoxy or alkylenedithioxy, and is unsubstituted or substituted with one or more substituents each independently selected from Y; Y is halo, amino, alkylamino, dialkylamino, arylamino, diarylamino, acylamino, azido, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy or heteroaryloxy.
38. Use of a compound of formula 11 for the preparation of a medicament for treatment of FGFmediated diseases, wherein formula 11 is: or pharmaceutically acceptable derivatives thereof, wherein: Ar'is higher alkylene, or is monocyclic or polycyclic arylene, alkenylarylene, arylalkenylene, heteroarylene, alkenylheteroarylene or heteroarylalkenylene, and is unsubstituted or substituted with one or more substituents each independently selected from Z; Z is halo, hydroxy, amino, azido, guanidino, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, alkoxycarbonyl, aryloxycarbonyl or heteroaryloxycarbonyl, or any two Z substitutents may form alkylene, alkenylene, alkylenedioxy, alkylenethioxyoxy or alkylenedithioxy, and is unsubstituted or substituted with one or more substituents each independently selected from Y ; Y is halo, amino, alkylamino, dialkylamino, arylamino, diarylamino, acylamino, azido, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy or heteroaryloxy; R7, R8, R9, R10, R11 and R12 are each independently selected from (i) or (ii) as follows: (i) R7 is selected from hydrogen and alkyl, R8 and R9 are hydrogen, R'° is hydrogen, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aralkyl or heteroaralkyl, R"is hydrogen, and R12 is hydrogen or Z, or (ii) any two of R7 R5 R9, R'°, R"and R12 together form alkylene, alkenylene, carbonylalkylene or carbonylalkenylene, and the remainder are selected as in (i); and m is 0 or 1 when Ar'is arylalkenylene or heteroarylalkenylene, or is 1.
Description:
DESCRIPTION ARGININE PEPTIDE ANALOGS USEFUL AS FIBROBLAST GROWTH FACTOR ANTAGONISTS RELATEDAPPLICATIONS This application is related to U. S. application Serial No.

09/058,002, to Yalamoori et al., entitiled"ARGININE PEPTIDE ANALOGS USEFUL AS FIBROBLAST GROWTH FACTOR ANTAGONISTS", filed April 9,1998. Priority is claimed herein to the above application, the disclosure of which is incorporated herein by reference in its entirety.

FIELD OF THE INVENTION The present invention relates to compositions containing arginine amides, arginine peptides and analogs thereof, and methods using the compositions for treatment or prevention of fibroblast growth factor (FGF)-mediated diseases. In particular, compositions containing arginine amides, arginine peptides and analogs thereof and use of the compositions as FGF antagonists are provided.

BACKGROUND OF THE INVENTION Fibroblast growth factors (FGFs) are a family of polypeptide mitogens and are ubiquitous in mammals. FGFs and their corresponding receptors, FGFRs, are widely distributed in tissues throughout the body, i. e., the central and peripheral nervous system, retina, kidneys, and <BR> <BR> <BR> <BR> <BR> myocardium (see, e. g., Johnson etal. Adv. Cancer Res. 1993,60,1), and are expressed during embryogenesis (Kimelman et a/Science 1988, 242,1053). FGFs exhibit potent mitogenic activity in these areas (see, e. g., Gospodarowicz Nature 1974,249,123), are also mitogenic for mesenchymal, neuronal, and epithelial cells (see. e. g., Johnson et al.

Molecular and Cellular Bioloctv 1990,10,4728; Gospodarowicz et al.

Exp. Eve Res. 1977,25,631; Thomas FASEB J. 1987,1,434; Gospodarowicz et al. J. Cell Phvsiol. 1987, S5 15) and have been

implicated in the processes of cell differentiation and maintenance (see, e. g., Anderson Nature 1988,332,360).

The FGFs consist of a family of peptides, of which ten have been identified (FGF-1 through 10). The first two peptides of this family to be isolated and characterized were FGF-1 and FGF-2, more commonly referred to as acid FGF (aFGF) and basis FGF (bFGF), respectively, for their acidic and basic isoelectric points, respectively. aFGF and bFGF were initially isolated from the bovine pituitary (Gospodarowicz J. Biol. <BR> <BR> <BR> <BR> <P>Chem. 1975,250,2515), then from bovine brain (Gospodarowicz et al.

J. Biol. Chem. and later isolated from human brain (Gimenez-Gallego et al. Biochem. Biophys. Res. Comm. 1986,135, 541). aFGF and bFGF have common biological properties, including the ability to bind to one or more FGF receptors. They also exhibit 55% homology in their amino acid sequences and are highly conserved among species (i. e., human and bovine bFGF exhibit 98.7% identity (see, e. g., U. S. Patent No. 5,288,855; U. S. Patent No. 5,155,214)). Eight other FGFs have been identified based on these structures (FGF-3 through 10): int-2 (FGF-3) (Moore et al. EMBO J. Jakobovits et al.

Proc. Nat. Acad. Sci. USA 1986,83,7806), hst-1/KS-FGF (identified <BR> <BR> <BR> <BR> from Kaposi's sarcoma DNA) (FGF-4) (Delli-Bovi et al. Cell 1987,50,<BR> <BR> <BR> <BR> <BR> <BR> 729; Taira et al. Proc. Nat. Acad. Sci. USA Huang et<BR> <BR> <BR> <BR> <BR> <BR> <BR> a/. J. Clin. Invest. 1993,91,1191), FGF-5 (Zhan etal Mol. Cell Biol.<BR> <BR> <BR> <BR> <BR> <BR> <P>1988,8,3487), FGF-6/Hst-2 (Marics et al. Oncoaene 1989,4,335),<BR> <BR> <BR> <BR> <BR> <BR> keratinocyte growth factor (KGF) (FGF-7) (Finch et al. Science 1989, 245,752), FGF-8, FGF-9 and FGF-10 (PCT International Publication Number WO 95/24,414). The structures of aFGF and bFGF have also been determined through single-crystal x-ray diffraction (Eriksson Proc. <BR> <BR> <BR> <P>Nat. Acad. Sci. USA Zhang et al. Proc. Nat. Acad. Sci.<BR> <BR> <BR> <BR> <BR> <BR> <BR> <P>USA 1991,88,3446; Zhu et al. Science 1991,251,90).

Basic FGF is a 16kD, acid-and thermally-sensitive peptide. It is an angiogenic factor causing the migration, proliferation and differentiation of endothelial cells to form blood vessels (see, e. g., Montesano et al.

Proc. Nat. Acad. Sci. USA Folkman etal. Science 1987,235,442). This effect indicates possible therapeutic uses of bFGF for wound healing (Folkman Science 1987,235,442; Buntrock et a/. Exp. Pathol. 1982, 21,62), neovascularization, nerve regeneration, cartilage repair, and enhancing success of tissue transplantation and of bone graft healing (see, generally, PCT International Publication No. WO 92/12,245). FGFs have also been reported to be useful as hypotensive agents for reducing high blood pressure and preventing myocardial <BR> <BR> <BR> <BR> infarction and cerebral hemorrhages (Saltis etal. Atherosclerosis 1995, 118,77; PCT International Publication No. WO 92/08,473), for the treatment of ulcers (U. S. Patent No. 5,401,721; U. S. Patent No.

5,175,147), in protecting the retina by inhibiting the release of nitric <BR> <BR> <BR> <BR> oxide in retinal inflammatory disorders (Goureau et a/. Proc. Nat. Acad.

Sci. USA 1993,90,1) and as a saporin conjugate in treating other <BR> <BR> <BR> <BR> ocular pathologies (Lappi et a/. Biochem. Biophvs. Res. Commun. 1989, 160,919; Lappi J. Cell Phvsiol. 1991,147,17; PCT International Publication No. WO 93/16,734) and vascular injury due to balloon angioplasty, preventing restenosis (U. S. Patent No. 5,308,622; U. S.

Patent No. 4,378,347).

Basic FGF may, however, be harmful in some cases in that cell proliferation and angiogenesis are important aspects of tumor growth and tumor development, rheumatoid arthritis, restenosis, In-Stent restenosis, proliferative diabetic retinopathies and diabetes (see, e. g., Folkman Adv.

Cancer Res. Melnyk et a/. Arthritis Rheum. 1990,33, 493; Sivalingam Arch. Ophthalmol. 1990,108,869). Basic FGF also functions as an oncogene in melanoma.

FGF activities are mediated by high and low affinity receptors.

There are many diverse forms of aFGF and bFGF receptors (Hanneken et a/. Proc. Nat. Acad. Sci. USA Four FGF receptor genes have been identified of which at least two produce multiple mRNA transcripts through alternative splicing of the primary transcript. This splicing creates a large number of forms of the receptors, potentially ninety-six receptor isoforms, and leads to response of the cell to many FGF family members, i. e., one gene gives FGFR-2 and KGF receptors, and alternate FGFR-1 splicing gives a 50 fold decrease in bFGF binding with unchanged aFGF binding. Receptor expression is also altered by injury and pathological conditions (restenosis, tumors and proliferative diseases). For example, receptor mRNA and protein are present in <BR> <BR> <BR> <BR> melanoma cells (see, e. q., Becker et a/. Oncoqene the receptor message is not usually found in palmar fascia, but is found in the proliferative hand disease Dupuytren's contracture (see, e. g., <BR> <BR> <BR> <BR> Gonzales et al. Amer. J. Pathol. 1992,141,661), and smooth muscle cells (SMCs) have no response to bFGF, but proliferating SMCs (i. e., during restenosis after balloon angioplasty) strongly respond to bFGF (see, e. g., Casscells etal. Proc. Nat. Acad. Sci. USA 1992,89,7159).

There are also soluble forms of FGFs in blood, suggesting further activity (Venkateswaran et a/. Hvbridoma 1992,11,729).

The potentially harmful effects of bFGF have led to attempts to identify human bFGF antagonists to treat and/or prevent FGF-mediated diseases. While large peptide analogs, derivatives and truncated forms of bFGF have been reported as antagonists of bFGF (U. S. Patent No.

5,132,408; U. S. Patent No. 5,252,718; PCT International Publication No. WO 92/12245; U. S. Patent No. 5,491,220), there have been no reports of small peptide and small peptide analog antagonists.

Therefore, it is an object herein to provide small peptide and small

peptide analog antagonists of human FGF, particularly basic FGF, for treatment or prevention of FGF-mediated diseases.

SUMMARY OF THE INVENTION Pharmaceutical compositions containing arginine amides, arginine peptides and analogs thereof, or pharmaceutically acceptable derivatives, including, but not limited to, salts, esters, acids, bases, solvates, hydrates and prodrugs thereof, are provided. Methods for modulating the interaction of an FGF peptide with FGF receptors using such compositions are also provided. Methods of treating or preventing FGF- mediated diseases are also provided.

In particular, pharmaceutical compositions containing arginine amides, peptides or analogs, and methods for inhibiting the binding of an FGF peptide to FGF receptors using such compositions are provided.

The arginine amides, peptides and analogs are arginine amide derivatives, or C-amido di-or tripeptides, or are aminoimino hydrazones. Among the pharmaceutical compositions provided herein are those that are particularly active as bFGF antagonists, as evidenced by in vitro assays described herein.

In one embodiment, the pharmaceutical compositions contain arginine amides, peptides or analogs that have formula I: or pharmaceutically acceptable derivatives thereof, including salts, esters, acids, bases, solvates, hydrates and prodrugs thereof, where R',

R2, R3, R4, R5 and R6 are each independently selected from (i) or (ii) as follows: (i) R', R3 and R5 are each independently selected from alkyl, alkenyl, alkynyl, aryl, cycloalkyl, aralkyl and heteroaralkyl, and are unsubstituted or substituted with one or more substituents each independently selected from Z, and R2, R4 and R6 are each independently selected from hydrogen or alkyl; or (ii) one or more of R'and R2, or R3 and R4, or R5 and R6 form alkylene or alkenylene which is unsubstituted or substituted with one or more substituents each independently selected from Z, and the others are selected as in (i); A is hydrogen, alkoxycarbonyl, aryloxycarbonyl, heteroaryloxy- carbonyl, aralkoxycarbonyl, heteroaralkoxycarbonyl, alkylcarbonyl, aryl- carbonyl, heteroarylcarbonyl, aralkylcarbonyl or heteroaralkylcarbonyl; and D is alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, aralkyl or heteroaralkyl, and is unsubstituted or substituted with one or more substituents each independently selected from Z; Z is halo, hydroxy, amino, azido, guanidino, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, alkoxycarbonyl, aryloxycarbonyl or heteroaryloxycarbonyl, or any two Z substituents may form alkylene, alkenylene, alkylenedioxy, alkylenethioxyoxy or alkylenedithioxy, and is unsubstituted or substituted with one or more substituents each independently selected from Y; and Y is halo, amino, alkylamino, dialkylamino, arylamino, diarylamino, acylamino, azido, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy or heteroaryloxy.

Thus, the arginine amides, peptides and analogs of formula I are arginine amide derivatives or di-and tripeptide analogs which are carboxamides at the C-terminus. The amino acids are of either the D-or L-configuration, or are racemic.

In another embodiment, the pharmaceutical compositions contain arginine amides, peptides or analogs that have formula ll: or pharmaceutical acceptable derivatives thereof, including salts, esters, acids, bases, solvates, hydrates and prodrugs thereof, where: Ar'is higher alkylene, which, as defined herein, contains about 7- 30 carbon atoms, or is monocyclic or polycyclic arylene, alkenylarylene, arylalkenylene, heteroarylene, alkenylheteroarylene or heteroarylalkenylene, and is unsubstituted or substituted with one or more substituents each independently selected from Z; Z is halo, hydroxy, amino, azido, guanidino, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, alkoxycarbonyl, aryloxycarbonyl or heteroaryloxycarbonyl, or any two Z substituents may form alkylene, alkenylene, alkylenedioxy, alkylenethioxyoxy or alkylenedithioxy, and is unsubstituted or substituted with one or more substituents each independently selected from Y; Y is halo, amino, alkylamino, dialkylamino, arylamino, diarylamino, acylamino, azido, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy or heteroaryloxy;

R7, R8, R9, R10, R1l and R12 are each independently selected from (i) or (ii) as follows : (i) R7 is selected from hydrogen and alkyl, R8 and R9 are hydrogen, R10 is hydrogen, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, aralkyl or heteroaralkyl, R"is hydrogen, and R12 is hydrogen or Z, or (ii) any two of R7 R8, R9, Rlo, R11 and R12 together form alkylene, alkenylene, carbonylalkylene or carbonylalkenylene, and the remainder are selected as in (i); and m is 0 or 1 when Ar'is arylalkenylene or heteroarylalkenylene, or is 1.

Of the compounds described herein, those that inhibit an FGF- mediated activity by about 50% at concentrations of less than about 500 uM are preferred. More preferred are those that inhibit an FGF- mediated activity by about 50% at concentrations of less than about 330 µM, more preferably less than about 150 hum, and most preferably less than about 30, uM.

The pharmaceutical compositions formulated for administration by an appropriate route and means containing effective concentrations of one or more of the compounds described herein, or pharmaceutically acceptable salts, esters, acids, bases, solvates, hydrates or prodrugs thereof, that deliver amounts effective for the treatment of FGF-mediated disorders, and other conditions that are in some manner mediated by an FGF peptide or whose symptoms can be ameliorated by administration of a bFGF-specific FGF antagonist, are also provided. The effective amounts and concentrations are effective for ameliorating any of the symptoms of any of the disorders.

Also of interest for use in the pharmaceutical compositions are any pharmaceutically-acceptable derivatives, including salts, esters, acids, bases, solvates, hydrates and prodrugs of the arginine amides, peptides

or analogs. Pharmaceutically-acceptable salts, include, but are not limited to, amine salts, such as but not limited to N, N'- dibenzylethylenediamine, chloroprocaine, choline, ammonia, diethanolamine and other hydroxyalkylamines, ethylenediamine, N- methylglucamine, procaine, N-benzylphenethylamine, 1-para- chlorobenzyl-2-pyrrolidin-1'-ylmethylbenzimidazole, diethylamine and other alkylamines, piperazine and tris (hydroxymethyl) aminomethane; alkali metal salts, such as but not limited to lithium, potassium and sodium; alkali earth metal salts, such as but not limited to barium, calcium and magnesium; transition metal salts, such as but not limited to zinc; and other metal salts, such as but not limited to sodium hydrogen phosphate and disodium phosphate; and also including, but not limited to, salts of mineral acids, such as but not limited to hydrochlorides and sulfates; and salts of organic acids, such as but not limited to acetates, lactates, malates, tartrates, citrates, ascorbates, succinates, butyrates, valerates and fumarates.

Methods for treatment or prevention of FGF-mediated diseases, including, but not limited to, diabetes, cancer, including, but not limited to, melanoma and tumor growth and development, restenosis, In-Stent restenosis, rheumatoid arthritis, ophthalmic disorders, including, but not limited to, corneal clouding following excimer laser surgery, closure of trabeculectomies, hyperproliferation of lens epithelial cells following cataract surgery, the recurrence of pterygii and diabetic retinopathy, and other proliferative diseases, including, but not limited to, Dupuytren's contracture, conditions that are in some manner mediated by an FGF peptide that binds to FGF receptors, or that are ameliorated by administration of an FGF receptor bFGF antagonist are provided.

Methods for inhibiting binding of an FGF peptide to an FGF receptor are provided. These methods are practiced by contacting the

receptor with one or more of the compositions provided herein simultaneously, prior to, or subsequent to contacting the receptor with an FGF peptide.

In particular, methods of treating FGF-mediated disorders by administering effective amounts of the arginine amides, peptides or analogs, or salts, acids, bases, solvates, hydrates, prodrugs or other suitable derivatives thereof are provided. In particular, methods for treating FGF-mediated disorders, including, but not limited to, diabetes, cancer, including, but not limited to, melanoma and tumor growth and development, restenosis, In-Stent restenosis, rheumatoid arthritis, ophthalmic disorders, including, but not limited to, corneal clouding following excimer laser surgery, closure of trabeculectomies, hyperproliferation of lens epithelial cells following cataract surgery, the recurrence of pterygii and diabetic retinopathy, and other proliferative diseases, including, but not limited to, Dupuytren's contracture, and other proliferative diseases in which FGF receptor bFGF-mediated physiological responses are implicated, by administering effective amounts of one or more of the compositions provided herein in pharmaceutically acceptable carriers are provided.

The methods are effected by contacting FGF receptors with one or more of the arginine amides, peptides or analogs prior to, simultaneously with, or subsequent to contacting the receptors with an FGF peptide. In practicing the methods, effective amounts of compositions containing therapeutically effective concentrations of the compounds formulated for oral, intravenous, local and topical application for the treatment of FGF- mediated disorders, including, but not limited to, diabetes, cancer, including, but not limited to, melanoma and tumor growth and development, restenosis, In-Stent restenosis, rheumatoid arthritis, ophthalmic disorders, including, but not limited to, corneal clouding

following excimer laser surgery, closure of trabeculectomies, hyperproliferation of lens epithelial cells following cataract surgery, the recurrence of pterygii and diabetic retinopathy, and other proliferative diseases, including, but not limited to, Dupuytren's contracture, and other diseases in which FGF-mediated physiological responses are implicated are administered to an individual exhibiting the symptoms of one or more of these disorders. The amounts are effective to ameliorate or eliminate one or more symptoms of the disorders.

In addition, methods for identifying compounds that are suitable for use in treating particular diseases based on their preferential affinity for an FGF receptor are also provided.

Articles of manufacture containing packaging material, a composition, or salt, ester, acid, base, solvate, hydrate, or prodrug thereof, provided herein, which is effective for ameliorating the symptoms of an FGF-mediated disorder, antagonizing the effects of bFGF or inhibiting binding of an FGF peptide to an FGF receptor, within the packaging material, and a label that indicates that the composition, or salt, ester, acid, base, solvate, hydrate, or prodrug thereof, is used for antagonizing the effects of bFGF, treating an FGF-mediated disorder, or inhibiting the binding of an FGF peptide to an FGF receptor, are provided.

DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS Definitions Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which this invention belons. All patents and publications referred to herein are incorporated by reference.

As used herein, fibroblast growth factor (FGF) peptides include peptides that have substantially the amino acid sequence of any one of

FGF-1 through 10 and that act as potent endogenous proliferative peptides.

As used herein, an FGF-mediated condition is a condition that is caused by anormal FGF activity or one in which compounds that inhibit FGF activity have therapeutic use. Such diseases include, but are not limited to diabetes, cancer, including, but not limited to, melanoma and tumor growth and development, restenosis, In-Stent restenosis, rheumatoid arthritis, ophthalmic disorders, including, but not limited to, corneal clouding following excimer laser surgery, closure of trabeculectomies, hyperproliferation of lens epithelial cells following cataract surgery, the recurrence of pterygii and diabetic retinopathy, and other proliferative diseases, including, but not limited to, Dupuytren's contracture, and other diseases in which FGF-mediated physiological responses are implicated.

As used herein an effective amount of a compound for treating a particular disease is an amount that is sufficient to ameliorate, or in some manner reduce the symptoms associated with the disease. Such amount may be administered as a single dosage or may be administered according to a regimen, whereby it is effective. The amount may cure the disease but, typically, is administered in order to ameliorate the symptoms of the disease. Typically, repeated administration is required to achieve the desired amelioration of symptoms.

As used herein, an FGF antagonist is a compound, such as a drug or an antibody, that inhibits FGF-stimulated proliferation and other FGF- mediated physiological responses. The antagonist may act by interfering with the interaction of the FGF with an FGF-specific receptor or by interfering with the physiological response to or bioactivity of an FGF isopeptide, such as proliferation. Thus, as used herein, an FGF antagonist interferes with FGF-stimulated proliferation or other response

or interferes with the interaction of an FGF peptide with an FGF-specific receptor, such as bFGF receptors, as assessed by assays known to those of skill in the art.

The effectiveness of potential FGF antagonists can be assessed using methods known to those of skill in the art. For example, the effectiveness may be measured by inhibition of binding of t251-bFGF to a human extracellular-domain FGFR1-TPA fusion protein immobilized on a solid phase (hsRRA assay) (for the extracellular form of human FGFR, see U. S. Patent 5,288,855). Effectiveness may also be measured through the use of a membrane-bound competitive binding assay, quantifying inhibition of binding of'251-bFGF to FGF receptors on cultured smooth muscle cells (SMCs). Effectiveness may also be measured by determination of inhibition of 3H-thymidine incorporation into DNA, which is promoted by bFGF stimulation of SMC proliferation (see, generally; <BR> <BR> <BR> <BR> Nachtigal et al. In Vitro Cellular and Developmental Biology 1989,25,<BR> <BR> <BR> <BR> <BR> <BR> 892).

As used herein, the biological activity or bioactivity of an FGF peptide includes any activity induced, potentiated or influence by FGF in vivo. It also includes the ability to bind to particular receptors and to induce a functional response, such as proliferation. It may be assessed by in vivo assays or by in vitro assays, such as those exemplified herein.

The relevant activity includes, but is not limited to, proliferation. Any assay known to those of skill in the art to measure or detect such <BR> <BR> <BR> <BR> activity may be used to assess such activity (see, e-a., Nachtigal et a/. In Vitro Cellular and Developmental Biology 1989,25,892; and the Examples herein).

As used herein, the IC50 refers to an amount, concentration or dosage of a particular test compound that achieves a 50% inhibition of a

maximal response, such as binding of FGF to tissue receptors, in an assay that measures such response.

As used herein, EC50 refers to a dosage, concentration or amount of a particular test compound that elicits a dose-dependent response at 50% of maximal expression of a particular response that is induced, provoked or potentiated by the particular test compound.

As used herein, pharmaceutically acceptable derivatives of a compound include salts, esters, acids, bases, solvates, hydrates or prodrugs thereof that may be readily prepared by those of skill in this art using known methods for such derivatization and that produce compounds that may be administered to animals or humans without substantial toxic effects and that either are pharmaceutically active or are prodrugs. For example, acidic groups can be esterified or neutralized.

As used herein, pharmaceutically-acceptable salts, esters, hydrates, solvates or other derivatives of the compounds include any such salts, esters and other derivatives that may be prepared by those of skill in this art using known methods for such derivatization and that produce compounds that may be administered to animals or humans without substantial toxic effects and that either are pharmaceutically active or are prodrugs. Pharmaceutically-acceptable salts include, but are not limited to, salts of alkali metals and alkaline earth metals, including but not limited to sodium salts, potassium salts, lithium salts, calcium salts and magnesium salts; transition metal salts, such as zinc salts, copper salts and aluminum salts; polycationic counter ion salts, such as but not limited ammonium and substituted ammonium salts and organic amine salts, such as hydroxyalkylamines and alkylamines; salts of mineral acids, such as but not limited to hydrochlorides and sulfates, salts of organic acids, such as but not limited acetates, lactates,

malates, tartrates, citrates, ascorbates, succinates, butyrate, valerate and fumarates. Also contemplated herein are the corresponding esters.

Preferred pharmaceutically-acceptable salts include, but are not limited to, N, N'-dibenzylethylenediamine, chloroprocaine, choline, ammonia, diethanolamine and other hydroxyalkylamines, ethylenediamine, N-methylglucamine, procaine, N-benzylphenethylamine, <BR> <BR> <BR> <BR> 1-para-chlorobenzyl-2-pyrrolidin-1'-ylmethylbenzimidazole, diethylamine and other alkylamines, piperazine, tris (hydroxymethyl) aminomethane, aluminum, calcium, lithium, magnesium, potassium, sodium hydrogen phosphate, disodium phosphate, sodium, zinc, barium, gold, silver and bismuth salts. Sodium salts, particularly the sodium salt of each of the compound, are most preferred herein.

As used herein, treatment means any manner in which the symptoms of a conditions, disorder or disease are ameliorated or otherwise beneficially altered. Treatment also encompasses any pharmaceutical use of the compositions herein, such as use as contraceptive agents.

As used herein, amelioration of the symptoms of a particular disorder by administration of a particular pharmaceutical composition refers to any lessening, whether permanent or temporary, lasting or transient that can be attributed to or associated with administration of the composition.

As used herein, substantially pure means sufficiently homogeneous to appear free of readily detectable impurities as determined by standard methods of analysis, such as thin layer chromatography (TLC), gel electrophoresis and high performance liquid chromatography (HPLC), used by those of skill in the art to assess such purity, or sufficiently pure such that further purification would not detectably alter the physical and chemical properties, such as enzymatic

and biological activities, of the substance. Methods for purification of the compounds to produce substantially chemically pure compounds are known to those of skill in the art. A substantially chemically pure compound may, however, be a mixture of stereoisomers. In such instances, further purification might increase the specific activity of the compound.

As used herein, biological activity refers to the in vivo activities of a compound or physiological responses that result upon in vivo administration of a compound, composition or other mixture. Biological activity, thus, encompasses therapeutic effects and pharmaceutical activity of such compounds, compositions and mixtures.

As used herein, a prodrug is a compound that, upon in vivo administration, is metabolized or otherwise converted to the biologically, pharmaceutically or therapeutically active form of the compound. To produce a prodrug, the pharmaceutically active compound is modified such that the active compound will be regenerated by metabolic processes. The prodrug may be designed to alter the metabolic stability or the transport characteristics of a drug, to mask side effects or toxicity, to improve the flavor of a drug or to alter other characteristics or properties of a drug. By virtue of knowledge of pharmacodynamic processes and drug metabolism in vivo, those of skill in this art, once a pharmaceutically active compound is known, can design prodrugs of the compound (see, e. q., Nogrady (1985) Medicinal Chemistrv A Biochemical Approach, Oxford University Press, New York, pages 388-392). For example, succinyl-sulfathiazole is a prodrug of 4-amino-N- (2-thiazoyl) ben- zenesulfonamide (sulfathiazole) that exhibits altered transport characteristics.

As used herein, alkyl, alkenyl and alkynyl carbon chains, if not specified contain from 1 to 20 carbons, preferably 1 to 16 carbons, and

are straight or branched. Alkenyl carbon chains of from 1 to 20 carbons preferably contain 1 to 8 double bonds, and the alkenyl carbon chains of 1 to 16 carbons preferably contain 1 to 5 double bonds. Alkynyl carbon chains of from 1 to 20 carbons preferably contain 1 to 8 triple bonds, and the alkynyl carbon chains of 1 to 16 carbons preferably contain 1 to 5 triple bonds. The alkyl, alkenyl and alkynyl groups may be optionally substituted, with one or more groups, preferably alkyl group substituents that may be the same or different.

As used herein, lower alkyl, lower alkenyl, lower alkynyl, lower alkylene, lower alkenylene and lower alkynylene refer to carbon chains having up to about 6 carbons. As used herein, higher alkyl, higher alkenyl, higher alkynyl, higher alkylene, higher alkenylene and higher alkynylene refer to carbon chains having about 7-30 carbons.

As used herein, an alkyl group substituent includes halo, haloalkyl, preferably halo lower alkyl, aryl, hydroxy, alkoxy, aryloxy, alkyloxy, alkylthio, arylthio, aralkyloxy, aralkylthio, carboxy, alkoxycarbonyl, oxo and cycloalkyl.

As used herein,"aryl"refers to cyclic groups containing from 3 to 19 carbon atoms containing 1-5 rings, including 1-5 fused rings. Aryl groups include, but are not limited to groups, such as phenyl, substituted phenyl, naphthyl, substituted naphthyl, in which the substituent is lower alkyl, halogen, or lower alkoxy.

As used herein, an"aryl group substituent"includes alkyl, cycloalkyl, cycloalkylalkyl, aryl, heteroaryl optionally substituted with 1 or more, preferably 1 to 3, substituents selected from halo, halo alkyl and alkyl, arylalkyl, heteroarylalkyl, alkenyl containing 1 to 2 double bonds, alkynyl containing 1 to 2 triple bonds, halo, hydroxy, haloalkyl and polyhaloalkyl, preferably halo lower alkyl, especially trifluoromethyl, formyl, alkylcarbonyl, arylcarbonyl that is optionally substituted with 1 or

more, preferably 1 to 3, substituents selected from halo, halo alkyl and alkyl, heteroarylcarbonyl, carboxy, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, arylaminocar- bonyl, diarylaminocarbonyl, arylalkylaminocarbonyl, alkoxy, aryloxy, perfluoroalkoxy, alkenyloxy, alkynyloxy, arylalkoxy, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, arylaminoalkyl, amino, alkylamino, dialkylamino, arylamino, alkylarylamino, alkylcarbonylamino, arylcarbonyl- amino, azido, nitro, mercapto, alkylthio, arylthio, perfluoroalkylthio, thiocyano, isothiocyano, alkylsulfinyl, alkylsufonyl, arylsulfinyl, arylsulfonyl, aminosulfonyl, alkylaminosulfonyl, dialkylaminosulfonyl and arylaminosulfonyl. Exemplary aryl groups include optionally substituted phenyl and optionally substituted naphthyl.

As used herein,"cycloalkyl"refers to a saturated mono-or multi- cyclic ring system, preferably of 3 to 10 carbon atoms, more preferably 3 to 6 carbon atoms; cycloalkenyl and cycloalkynyl refer to mono-or multicyclic ring systems that respectively include at least one double bond and at least one triple bond. Cycloalkenyl and cycloalkynyl groups may preferably contain 3 to 10 carbon atoms, with cycloalkenyl groups more preferably containing 4 to 7 carbon atoms and cycloalkynyl groups more preferably containing 8 to 10 carbon atoms. The ring systems of the cycloalkyl, cycloalkenyl and cycloalkynyl groups may be composed of one ring or two or more rings which may be joined together in a fused, bridged or spiro-connected fashion, and may be optionally substituted with one or more alkyl group substituents.

As used herein,"heteroaryl"refers to a monocyclic or multicyclic ring system, preferably of about 5 to about 15 members where one or more, more preferably 1 to 3 of the atoms in the ring system is a heteroatom, that is, an element other than carbon, for example, nitrogen, oxygen and sulfur atoms. The heteroaryl may be optionally substituted

with one or more, preferably 1 to 3, aryl group substituents. Exemplary heteroaryl groups include, for example, furyl, thienyl, pyridyl, pyrrolyl, N- methylpyrrolyl, quinolinyl and isoquinolinyl, with pyridyl and quinolinyl being preferred.

As used herein,"heterocyclic"refers to a monocyclic or multicyclic ring system, preferably of 3 to 10 members, more preferably 4 to 7 members, even more preferably 5 to 6 members, where one or more, preferably 1 to 3 of the atoms in the ring system is a heteroatom, that is, an element other than carbon, for example, nitrogen, oxygen and sulfur atoms. The heterocycle may be optionally substituted with one or more, preferably 1 to 3 aryl group substituents. Preferred substituents of the heterocyclic group include hydroxy, alkoxy containing 1 to 4 car- bon atoms, halo lower alkyl, including trihalomethyl, such as trifluoromethyl, and halogen. As used herein, the term heterocycle may include reference to heteroaryl. Exemplary heterocycles include, for example, pyrrolidinyl, piperidinyl, alkylpiperidinyl, morpholinyl, oxadiazolyl or triazolyi.

As used herein, the nomenclature alkyl, alkoxy, carbonyl, etc. are used as is generally understood by those of skill in this art. For example, as used herein alkyl refers to saturated carbon chains that contain one or more carbons; the chains may be straight or branched or include cyclic portions or be cyclic. As used herein, alicyclic refers to aryl groups that are cyclic.

As used herein,"halogen"or"halide"refers to F, Cl, Br or 1.

As used herein, pseudohalides are compounds that behave substantially similar to halides. Such compounds can be used in the same manner and treated in the same manner as halides (X-, in which X is a halogen, such as Cl or Br). Pseudohalides include, but are not

limited to cyanide, cyanate, thiocyanate, selenocyanate, trifluoromethyl and azide.

As used herein,"haloalkyl"refers to a lower alkyl radical in which one or more of the hydrogen atoms are replace by halogen including, but not limited to, chloromethyl, trifluoromethyl, 1-chloro-2-fluoroethyl and the like.

As used herein,"haloalkoxy"refers to RO-in which R is a haloalkyl group.

As used herein,"sulfinyl"refers to-S (O)-. As used herein, "sulfonyl"refers to-S (0) 2-.

As used herein,"aminocarbonyl"refers to-C (O) NH2.

As used herein,"alkylaminocarbonyl"refers to-C (O) NHR in which R is hydrogen or alkyl, preferably lower alkyl. As used herein "dialkylaminocarbonyl"as used herein refers to-C (O) NR R in which R and R are independently selected from hydrogen or alkyl, preferably lower alkyl;"carboxamide"refers to groups of formula-NR COR.

As used herein,"diarylaminocarbonyl"refers to-C (O) NRR' in which R and R'are independently selected from aryl, preferably lower aryl, more preferably phenyl.

As used herein,"arylalkylaminocarbonyl"refers to-C (O) NRR' in which one of R and R'is aryl, preferably lower aryl, more preferably phenyl, and the other of R and R'is alkyl, preferably lower alkyl.

As used herein,"arylaminocarbonyl"refers to-C (O) NHR in which R is aryl, preferably lower aryl, more preferably phenyl.

As used herein,"alkoxycarbonyl"refers to-C (O) OR in which R is alkyl, preferably lower alkyl.

As used herein,"aryloxycarbonyl"refers to-C (O) OR in which R is aryl, preferably lower aryl, more preferably phenyl.

As used herein,"alkoxy"and"alkylthio"refer to RO-and RS-, in which R is alkyl, preferably lower alkyl.

As used herein,"aryloxy"and"arylthio"refer to RO-and RS-, in which R is aryl, preferably lower aryl, more preferably phenyl.

As used herein,"alkylene"refers to a straight, branched or cyclic, preferably straight or branched, bivalent aliphatic hydrocarbon group, preferably having from 1 to about 20 carbon atoms, more preferably 1 to 12 carbons, even more preferably lower alkylene. The alkylene group is optionally substituted with one or more"alkyl group substituents." There may be optionally inserted along the alkylene group one or more oxygen, sulphur or substituted or unsubstituted nitrogen atoms, where the nitrogen substituent is alkyl as previously described. Exemplary alkylene groups include methylene (-CH2-), ethylene (-CH2CH2-), propylene (- (CH2) 3-), cyclohexylene (-C6Ho-), methylenedioxy (-O-CH2- O-) and ethylenedioxy (-O-(CH2) 2-O-). The term"lower alkylene"refers to alkylene groups having 1 to 6 carbons. Preferred alkylene groups are lower alkylene, with alkylene of 1 to 3 carbon atoms being particularly preferred.

As used herein,"alkenylene"refers to a straight, branched or cyclic, preferably straight or branched, bivalent aliphatic hydrocarbon group, preferably having from 1 to about 20 carbon atoms and at least one double bond, more preferably 1 to 12 carbons, even more preferably lower alkenylene. The alkenylene group is optionally substituted with one or more"alkyl group substituents."There may be optionally inserted along the alkenylene group one or more oxygen, sulphur or substituted or unsubstituted nitrogen atoms, where the nitrogen substituent is alkyl as previously described. Exemplary alkenylene groups include -CH = CH-CH = CH-and-CH = CH-CH2-. The term"lower alkenylene" refers to alkenylene groups having 2 to 6 carbons. Preferred alkenylene

groups are lower alkenylene, with alkenylene of 3 to 4 carbon atoms being particularly preferred.

As used herein,"alkynylene"refers to a straight, branched or cyclic, preferably straight or branched, bivalent aliphatic hydrocarbon group, preferably having from 1 to about 20 carbon atoms and at least one triple bond, more preferably 1 to 12 carbons, even more preferably lower alkynylene. The alkynylene group is optionally substituted with one or more"alkyl group substituents."There may be optionally inserted along the alkynylene group one or more oxygen, sulphur or substituted or unsubstituted nitrogen atoms, where the nitrogen substituent is alkyl as previously described. Exemplary alkynylene groups include -C=C-C--_C-,-C---C-and-C-_-C-CH2-. The term"lower alkynylene" refers to alkynylene groups having 2 to 6 carbons. Preferred alkynylene groups are lower alkynylene, with alkynylene of 3 to 4 carbon atoms being particularly preferred.

As used herein,"arylene"refers to a monocyclic or polycyclic, preferably monocyclic, bivalent aromatic group, preferably having from 1 to about 20 carbon atoms and at least one aromatic ring, more preferably 1 to 12 carbons, even more preferably lower arylene. The arylene group is optionally substituted with one or more"alkyl group substituents."There may be optionally inserted around the arylene group one or more oxygen, sulphur or substituted or unsubstituted nitrogen atoms, where the nitrogen substituent is alkyl as previously described. Exemplary arylene groups include 1,2-, 1,3- and 1,4- phenylene. The term"lower arylene"refers to arylene groups having 5 or 6 carbons. Preferred arylene groups are lower arylene.

As used herein,"heteroarylene"refers to a bivalent monocyclic or multicyclic ring system, preferably of about 5 to about 15 members where one or more, more preferably 1 to 3 of the atoms in the ring

system is a heteroatom, that is, an element other than carbon, for example, nitrogen, oxygen and sulfur atoms. The heteroarylene group may be optionally substituted with one or more, preferably 1 to 3, aryl group substituents. Exemplary heteroaryiene groups include, for example, 1,4-imidazolylene.

As used herein,"alkylenedioxy"refers to-O-alkylene-O-; alkylenethioxyoxy refers to-S-alkylene-O- ; and"alkylenedithioxy"refers to-S-alkylene-S-.

As used herein,"alkylidene"refers to a bivalent group, such as =CR'R", which is attached to one atom of another group, forming a double bond. Exemplary alkylidene groups are methylidene (=CH2) and ethylidene (=CHCH3). As used herein,"arylalkylidene"refers to an alkylidene group in which either R'or R"is and aryl group.

As used herein,"amido"refers to a bivalent group, either-C (O) NH- or-HNC (O)-."Thioamido"refers to a bivalent group, either-C (S) CH- or- HNC (S)-."Oxyamido"refers to a bivalent group, either-OC (O) NH- or- <BR> <BR> <BR> <BR> HNC (O) O-."Thiaamido"refers to a bivalent group, either-SC (O) NH-or-<BR> <BR> <BR> <BR> <BR> HNC (O) S-."Dithiaamido"refers to a bivalent group, either-SC (S) NH- or<BR> <BR> <BR> <BR> <BR> <BR> -HNC (S) S-."Ureido"refers to the bivalent group-HNCONH-.

"Thioureido"refers to the bivalent group-HNCSNH-.

As used herein, the term"amino acid"refers to a-amino acids which are racemic, or of either the D-or L-configuration.

As used herein, when any particular group, such as phenyl or pyridyl, is specified, this means that the group is unsubstituted or is substituted. Preferred substituents where not specified are halo, halo lower alkyl, and lower alkyl.

As used herein, the abbreviations for any protective groups, amino acids and other compounds, are, unless indicated otherwise, in accord with their common usage, recognized abbreviations, or the IUPAC-IUB

Commission on Biochemical Nomenclature (see, Biochem. 1972,11, 942).

A. Compounds for use in the Pharmaceutical Compositions 1. Arginine Amides and Di-and Tripeptides In one embodiment, the pharmaceutical compositions contain arginine amides or C-amido arginine di-or tripeptide analogs. The amino acids are of either the D-or L-configuration, or are racemic. In particular, the pharmaceutical compositions contain compounds of formulae I: or pharmaceutically acceptable derivatives thereof, including salts, esters, acids, bases, solvates, hydrates and prodrugs thereof, where R', R2, R3, R4, R5 and R6 are each independently selected from (i) or (ii) as follows: (i) R', R3 and R5 are each independently selected from alkyl, alkenyl, alkynyl, aryl, cycloalkyl, aralkyl and heteroaralkyl, and are unsubstituted or substituted with one or more substituents each independently selected from Z, and R2, R4 and R6 are each independently selected from hydrogen or alkyl; or (ii) one or more of R'and R2, or R3 and R4, or R5 and R6 form alkylene or alkenylene which is unsubstituted or substituted with one or more substituents each independently selected from Z, and the others are selected as in (i);

A is hydrogen, arylalkoxycarbonyl or arylcarbonyl; and D is aryl, heteroaryl, aralkyl or heteroaralkyl, and is unsubstituted or substituted with one or more substituents each independently selected from Z; Z is halo, hydroxy, amino, azido, guanidino, nitro, alkyl, aryl, <BR> <BR> <BR> <BR> heteroaryl, alkoxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, alkoxycarbonyl, aryloxycarbonyl or heteroaryloxycarbonyl, or any two Z substituents may form alkylene, alkenylene, alkylenedioxy, alkylenethioxyoxy or alkylenedithioxy, and is unsubstituted or substituted with one or more substituents each independently selected from Y; and Y is halo, amino, alkylamino, dialkylamino, arylamino, diarylamino, acylamino, azido, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy or heteroaryloxy.

In preferred embodiments, A is hydrogen, benzyloxycarbonyl (Cbz), 9-fluorenylmethoxycarbonyl (Fmoc) or benzoyl (Bz). In other preferred embodiments, D is benzyl (Bn), 4-nitrophenyl, 1-methoxy-3- naphthyl (4MbNA) or 4-methyl-7-cumarinyl (AMC). In more preferred embodiments, A is hydrogen, benzyloxycarbonyl (Cbz), 9- fluorenylmethoxycarbonyl (Fmoc) or benzoyl (Bz) and D is benzyl (Bn), 4- nitrophenyl, 1-methoxy-3-naphthyl (4MbNA) or 4-methyl-7-cumarinyl (AMC).

In more preferred embodiments, R', R2, R3, R4, R5 and R6 are selected as in (i) or (ii) as follows: (i) R', R3 and R5 are each independently selected from alkyl and aralkyl, and are unsubstituted or substituted with one or more substituents each independently selected from Z, and R2, R4 and R6 are each hydrogen; or (ii) one of R'and R2, or R3 and R4, or R5 and R6 form alkylene, and the others are selected as in (i).

In other embodiments, R', R2, R3, R4, R5 and R6 are selected as in (i) or (ii) as follows: (i) R', R3 and R5 are each independently selected from benzyl, 3- guanidinylpropyl, 4-hydroxyphenylmethyl, methyl, 2-methylpropyl, isopropyl and 4-aminobutyl, and R2, R4 and R6 are each hydrogen; or (ii) R3 and R4, or R5 and R6 form propylene, and the others are selected as in (i).

In certain embodiments, the guanidinyl group is further substituted with 2,2,5,7,8-pentamethyl-6-chromylsulfonyl (Pmc).

Thus, in more preferred embodiments, the peptides contain amino acids selected from phenylalanine, arginine, tyrosine, alanine, valine, leucine, lysine and proline.

Presently preferred compounds of formula I include (Fmoc)-Phe- Arg-NHBn, (Fmoc)-Phe-D-Arg-NHBn, (Fmoc)-D-Phe-Arg-NHBn, (Fmoc)-D- Phe-D-Arg-NHBn, (Fmoc)-Tyr-Arg-NHBn, (Fmoc)-Ala-Arg-NHBn, (Fmoc)- Leu-Arg-NHBn, (Fmoc)-Arg-Phe-NHBn, (Fmoc)-Arg (Pmc)-Phe-NHBn, (Fmoc)-D-Arg-Phe-NHBn, (Fmoc)-Arg-D-Phe-NHBn, (Fmoc)-D-Arg-D-Phe- NHBn, (Fmoc)-Lys-Phe-NHBn, Cbz-Arg-AMC, Cbz-Arg-Arg-AMC, Cbz- Phe-Pro-Arg-4MbNA, Cbz-Phe-Arg-AMC, Cbz-D-Phe-Arg-AMC, Bz-Phe- Val-Arg-NH (4-nitrophenyl), N-a-Bz-Arg-AMC, H-Pro-Phe-Arg-AMC and H- D-Phe-Pro-Arg-4MbNA.

2. Aminoimino Hydrazones In another embodiment, the pharmaceutical compositions contain aminoimino hydrazones of formula ll:

or pharmaceutically acceptable derivatives thereof, including salts, esters, acids, bases, sovates, hydrates and prodrugs thereof, where: Ar'is higher alkylene, which, as defined herein, contains about 7- 30 carbon atoms, or is monocyclic or polycyclic arylene, alkenylarylene, arylalkenylene, heteroarylene, alkenylheteroarylene or heteroarylalkenylene, and is unsubstituted or substituted with one or more substituents each independently selected from Z; Z is halo, hydroxy, amino, azido, guanidino, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy, heteroaryloxy, arylalkoxy, heteroarylalkoxy, alkoxycarbonyl, aryloxycarbonyl or heteroaryloxycarbonyl, or any two Z substituents may form alkylene, alkenylene, alkylenedioxy, alkylenethioxyoxy or alkylenedithioxy, and is unsubstituted or substituted with one or more substituents each independently selected from Y; Y is halo, amino, alkylamino, dialkylamino, arylamino, diarylamino, acylamino, azido, nitro, alkyl, aryl, heteroaryl, alkoxy, aryloxy or heteroaryloxy; R7, R8, R9, R'°, R"and R12 are each independently selected from (i) or (ii) as follows: (i) R'is selected from hydrogen and alkyl, R8 and R9 are hydrogen, Rlo is hydrogen, arylsulfonyl or aralkyl, R"is hydrogen, and R'2 is hydrogen or Z, or (ii) any two of R', R8 R9, Rlo, R"and R12 together form alkylene or carbonylalkylene, and the remainder are selected as in (i); and m is 0 or 1 when Ar'is arylalkenylene or heteroarylalkenylene, or is 1.

In certain embodiments, R7, R8, R9, R'°, R"and R12 are each independently selected from (i) or (ii) as follows:

(i) R'is selected from hydrogen and alkyl, R8 and R9 are hydrogen, R'° is hydrogen, arylsulfonyl or aralkyl, R"is hydrogen, and R12 is hydrogen or Z, or (ii) R8, R9 and R'° are selected as in (i), and (a) R"is selected as in (i), and R'and R12 together form alkylene or carbonylalkylene, or (b) R7 is selected as in (i), and R"and R12 together form alkylene or carbonylalkylene.

In a preferred embodiment, Ar'and R"taken together are higher alkyl, preferably C20H41,morepreferablyC8H17toC15H31,mostto preferably C9H19.

In another preferred embodiment, Ar1 and R12 taken together are monocyclic or bicyclic aryl, alkenylaryl, arylalkenylene or heteroarylalkenylene, and is unsubstituted or substituted with one or more substituents selected from Z. Ar'and R12 taken together are more preferably phenyl, 2-phenylalkenyl, 2-thienylalkylene or naphthyl, most preferably phenyl, 2-phenylethenyl, 2-thienylethylene and naphthyl, and are substituted with one or more groups selected from chloro, methoxy, biphenyl, benzoyloxy, 4-nitrophenyloxycarbonyl, 4-acetylamino- phenyloxycarbonyl, ethylene, methylene, fluoro, 2-phenylethyloxy, 3,4- phenyloxy,pentyl,benzyloxyanddichlorobenzyloxy,4-chlorobenzy loxy, 2,4-dichlorobenzyloxy.

In more preferred embodiments, R', R8, R9, R'°, R"and R12 are each independently selected from (i), (ii) or (iii) as follows: (i) R7 is hydrogen or methyl, or forms ethylene or propylene with R'2, R8 and R9 are each hydrogen, or together form ethylene or propylene, R'° is hydrogen, 4-chlorophenylsulfonyl or benzyl, R"is hydrogen and R12 forms ethylene or propylene with R, or is hydrogen or Z; or

(ii) R8, R9, R"and R12 are selected as in (i), and R7 and R° together form methylene; or (iii) R8, R9, R10 and R12 are selected as in (i), and R7 and R" together form methylenecarbonyl.

In particularly preferred embodiments, Ar1 and R12 taken together are selected from (2-phenylethyloxy) phenyl, 2, 4-dichlorophenylethen-1- yl, 3-(3, 4-dichlorobenzyloxy) phenyl, 2-(4-chlorobenzyloxy) phenyl, 4- phenyloxyphenyl, 1-pentyl-2-phenylethenyl, 3,4-dibenzyloxyphenyl, 2- (2,4-dichlorobenzyloxy) phenyl, 2- (3, 4-dichlorobenzyloxy) phenyl, 1- naphthyl, phenyl, 2,6-dichlorophenyl, 2-methoxyphenyl, (4- (3- phenyl)phenyl) phenyl, 3-benzoyloxyphenyl, 4-nitrophenyloxy-car- bonylphenyl, 4-acetylaminophenyloxycarbonylphenyl, 2-ethylenylphenyl, 2-methylenylphenyl, 3,4-difluorophenyl, 2-phenylethenyl and 2-thienyl- 1,2-ethenylene.

Presently preferred compounds of formula 11 include 3- (2-phenyl- ethoxy) acetophenone aminoimino hydrazone, 2,4-dichlorobenzylidene- acetone aminoimino hydrazone, 3- (3, 4-dichlorobenzyloxy) benzaldehyde aminoimino hydrazone, 2-(4-chlorobenzyloxy) benzaldehyde aminoimino hydrazone, 4-phenyloxybenzaldehyde aminoimino hydrazone, 2- pentylcinnamaldehyde aminoimino hydrazone, decanal aminoimino hydrazone, 3,4-dibenzyloxybenzaldehyde aminoimino hydrazone, 2- (2,4- dichlorobenzyloxy) benzaldehyde aminoimino hydrazone, 2- (3, 4-dichloro- benzyloxy) benzaldehyde aminoimino hydrazone, 1-naphthaldehde amino- imino hydrazone, benzaldehyde aminoimino hydrazone, 2,6-dichloro- benzaldehyde aminoimino hydrazone, 2-methoxybenzaldehyde aminoi- mino hydrazone, 4- (3-phenylphenyl) acetophenone aminoimino hydrazone, 3-benzoyloxyacetophenone aminoimino hydrazone, 4-(4-nitrophenyloxy- carbonyl) acetophenone aminoimino hydrazone, 4- (4-acetylamino- phenyloxycarbonyl) acetophenone aminoimino hydrazone, acetophenone

4,5-dihydro-2-imidazolyl hydrazone, acetophenone 1,4,5,6-tetrahydro-2- pyrimidinyl hydrazone, acetophenone 3-benzyl-4,5-dihydro-2-imidazolyl hydrazone, a-tetralone aminoimino hydrazone, a-indanone aminoimino hydrazone, 6- (3, 4-difluorophenyl)-4,5-dihydroimidazo [2,1-c]-2,3,4,5- tetrahydro-1,2,4-triazine, 5-phenyl-2- (4, 5-dihydroimidazol-2-yl)-3,4- dihydro-3-oxo-2 (H)-pyrazole, benzylideneacetone aminoimino hydrazone and 1- (1- (4-chlorophenylsulfonyl)-4, 5-dihydroimidazol-2-yl)-3-methyl-5- (2-thienyl)pyrazole.

3. Arginine amide, peptide and analog derivatives Also of interest are any pharmaceutically-acceptable derivatives, including salts, esters, acids, bases, solvates, hydrates and prodrugs of the arginine amides, peptides and analogs. Such derivatives may be readily prepared by methods known to those of ordinary skill in the art.

Pharmaceutically-acceptable salts, include, but are not limited to, amine salts, such as but not limited to N, N'-dibenzylethylenediamine, chloroprocaine, choline, ammonia, diethanolamine and other hydroxyalkylamines, ethylenediamine, N-methylglucamine, procaine, N- benzylphenethylamine, 1-para-chlorobenzyl-2-pyrrolidin-1'-ylmethyl- benzimidazole, diethylamine and other alkylamines, piperazine and tris (hydroxymethyl) aminomethane; alkali metal salts, such as but not limited to lithium, potassium and sodium; alkali earth metal salts, such as but not limited to barium, calcium and magnesium; transition metal salts, such as but not limited to zinc; and other metal salts, such as but not limited to sodium hydrogen phosphate and disodium phosphate; and also including, but not limited to, salts of mineral acids, such as but not limited to hydrochlorides and sulfates; and salts of organic acids, such as but not limited to acetates, lactates, malates, tartrates, citrates, ascorbates, succinates, butyrates, valerates and fumarates. Preferred

salts include hydrochloride, nitrate, sulfate, acetate and hydrobromide salts.

B. Preparation of the compounds The preparation of the above compounds is described below. Any such compound or similar compound may be synthesized according to a method discussed in general below or by only minor modification of the methods by selecting appropriate starting materials. Additionally, many of the compounds described herein may be obtained from commercial sources known to those of skill in the art.

1. Arginine Amides and Di-and Tripeptides Arginine amides and di-and tripeptides of formulae I may be prepared according to methods known to those of skill in the art. Such methods may include solid phase synthesis. See, e-ct., Merrifield J. Am.

Chem. Soc. 1963,85,2149-2154. In general, the C-terminal amino acid may be N-protected as the corresponding Fmoc or Cbz derivative by reaction with the appropriate chloroformate, eq., 9-fluorenylmethyl chloroformate or benzyl chloroformate. The C-terminal amino acid may then be derivatized as the corresponding amide, such as a benzylamide, by reaction of the N-protected amino acid with, e. a., benzylamine, either HOBt (1-hydroxybenzotriazole) or HOAt (1-hydroxy-7-azabenzotriazole), and either DCC (dicyclohexylcarbodiimide) or DIC (diisopropylcarbo- diimide). Removal of the nitrogen protecting group by, e. g., treatment with piperidine or catalytic hydrogenation, provides the free amino acid amide.

Coupling of this amino acid amide with a N-protected amino acid, prepared as described above, provides a dipeptide. Coupling may be achieved by reaction in the presence of, e. q., HOBt or HOAt, and DCC or DIC. Removal of the amino protecting group provides the desired

dipeptides. Further coupling with a N-protected amino acid, followed by deprotection, affords the desired tripeptides.

2. Aminoimino hydrazones Aminoimino hydrazones of formula 11 may be prepared according to the method described below or by other methods known to those of ordinary skill in the art. See, generally, Buckingham Q. Rev., Chem. Soc. <BR> <BR> <BR> <BR> <P>1969,23,37-56; Newkome et al. Org. Svnth. 1976,50,102; McMurry J. Am. Chem. Soc. 1968,90,6821. Additionally, many of the compounds of formula II are commercially available from sources known to those of skill in the art (e-a., Aldrich Chemical Co., Milwaukee, WI).

In general, most of the aminoimino hydrazones of formula 11 may be prepared by reacting aminoguanidine (neural or as a salt, such as, but not limited to, the hydrochloride, bicarbonate or nitrate salt) with the appropriate aldehyde or ketone under conditions allowing for the removal of water. Such conditions include, but are not limited to, azeotropic removal of water, use of a dehydrating agent, such as, but not limited to, magnesium sulfate, molecular sieves or phosphorous pentoxide, or an organic/aqueous biphasic system where the organic solvent (i. e., dichloromethane or hexane) is highly immiscible with water. The product is then isolated by crystallization, chromatography or other suitable methods. In some instances, compounds of formula 11 may be prepared by heating an aqueous alcoholic solution of the aldehyde or ketone with aminoguanidine (neural or as a salt, such as, but not limited to, the hydrochloride, bicarbonate or nitrate salt). Evaporation of the solvent, followed by recrystallization of the crude product from, e. a., methanol or ethyl acetate/hexanes provides the desired compounds.

Certain compounds of formula 11 may be synthesized by substituting 2-hydrazino-2-imidazoline hydrobromide, 1-benzyl-2- hydrazino-2-imidazoline or 2-hydrazino-1,4,5,6-tetrahydropyrimidine for

aminoguanidine. Other compounds of formula 11 are prepared by condensing 2-hydrazino-2-imidazoline hydrobromide with benzoylacetic acid or 3,4-difluoro-a-bromoacetophenone. Reaction of 2-hydrazino-2- imidazoline hydrobromide with 1- (2-thienyl)-1,3-butandione, followed by reaction of the resultant 1- (2-imidazoline) pyrazole with 4-chloro- benzenesulfonyl chloride, provides other compounds of formula 11.

C. Formulation of Pharmaceutical Compositions containing Arginine Amides, Peptides or Analogs useful in the Prevention or Treatment of FGF-mediated diseases The pharmaceutical compositions provided herein contain therapeutically effective amounts of one or more of the arginine amides, peptides or analogs of formula I or II that are useful in the prevention or treatment of FGF-mediated diseases. The compositions contain arginine amides, C-amido di-and tripeptide arginine peptide analogs, or aminoi- mino hydrazones.

The compounds are preferably formulated into suitable pharmaceutical preparations such as solutions, suspensions, tablets, dispersible tablets, pills, capsules, powders, sustained release formulations or elixirs, for oral administration or in sterile solutions or suspensions for parenteral administration, as well as transdermal patch preparation and dry powder inhalers. Typically the compounds described above are formulated into pharmaceutical compositions using techniques and procedures well known in the art (see, e. a., Ansel Introduction to Pharmaceutical Dosage Forms, Fourth Edition 1985,126).

In the compositions, effective concentrations of one or more compounds or pharmaceutically acceptable derivatives is (are) mixed with a suitable pharmaceutical carrier or vehicle. The compounds may be derivatized as the corresponding salts, esters, acids, bases, solvates, hydrates or prodrugs prior to formulation, as described above. The concentrations of the compounds in the compositions are effective for

delivery of an amount, upon administration, that ameliorates the symptoms of the FGF-mediated disease. Typically, the compositions are formulated for single dosage administration. To formulate a composition, the weight fraction of compound is dissolved, suspended, dispersed or otherwise mixed in a selected vehicle at an effective concentration such that the treated condition is relieved or ameliorated. Pharmaceutical carriers or vehicles suitable for administration of the compounds provided herein include any such carriers known to those skilled in the art to be suitable for the particular mode of administration.

In addition, the compounds may be formulated as the sole pharmaceutically active ingredient in the composition or may be combined with other active ingredients. Liposomal suspensions, including tissue-targeted liposomes, may also be suitable as pharmaceutically acceptable carriers. These may be prepared according to methods known to those skilled in the art. For example, liposome formulations may be prepared as described in U. S. Patent No.

4,522,811.

The active compound is included in the pharmaceutically acceptable carrier in an amount sufficient to exert a therapeutically useful effect in the absence of undesirable side effects on the patient treated. The therapeutically effective concentration may be determined empirically by testing the compounds in known in vitro and in vivo systems (see, e-ct., Moscatelli et al. J. Cell. Physiol. 1987,131,123- 130; Gospodarowiczeta/. Proc. Natl. Acad. Sci. U. S. A. 1984,81, <BR> <BR> <BR> 6963-6967; Thomas et al. Proc. Natl. Acad. Sci. U. S. A. 1984,81,357; European Patent Application No. EP 645 451; International Application Publication No. WO 92/12245; Moscatelli eta/. Proc. Natl. Acad. Sci.

U. S. A. 1986,83,2091-2095; Phadke Biochem. Biophys. Res. Comm.

Togari et a/. Biochem. Biophvs. Res. Comm. 1983,

114, 1189-1193 ; and Wagner et al. J. Cell Biol. 1986,103,1363-1367) and then extrapolated therefrom for dosages for humans.

The concentration of active compound in the drug composition will depend on absorption, inactivation and excretion rates of the active compound, the physicochemical characteristics of the compound, the dosage schedule, and amount administered as well as other factors known to those of skill in the art. For example, the amount that is delivered is sufficient to treat the symptoms of diabetes.

Typically a therapeutically effective dosage should produce a serum concentration of active ingredient of from about 0.1 ng/ml to about 50-100, ug/ml. The pharmaceutical compositions typically should provide a dosage of from about 0.001 mg to about 2000 mg of compound per kilogram of body weight per day. Pharmaceutical dosage unit forms are prepared to provide from about 1 mg to about 1000 mg and preferably from about 10 to about 500 mg of the essential active ingredient or a combination of essential ingredients per dosage unit form.

The active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is understood that the precise dosage and duration of treatment is a function of the disease being treated and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data. It is to be noted that concentrations and dosage values may also vary with the severity of the condition to be alleviated.

It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions.

Preferred pharmaceutically acceptable derivatives include acids, bases, salts, esters, hydrates, solvates and prodrug forms. The derivative is selected such that its pharmacokinetic properties are superior to the corresponding neutral compound.

Thus, effective concentrations or amounts of one or more of the compounds described herein or pharmaceutically acceptable derivatives thereof are mixed with a suitable pharmaceutical carrier or vehicle for systemic, topical or local administration to form pharmaceutical compositions. Compounds are included in an amount effective for ameliorating or treating the FGF-mediated disorder for which treatment is contemplated. The concentration of active compound in the composition will depend on absorption, inactivation, excretion rates of the active compound, the dosage schedule, amount administered, particular formulation as well as other factors known to those of skill in the art.

The compositions are intended to be administered by a suitable route, which includes orally, parenterally, rectally and topically and locally depending upon the disorder being treated. For oral administration, capsules and tablets are presently preferred. The compositions are in liquid, semi-liquid or solid form and are formulated in a manner suitable for each route of administration. Preferred modes of administration include parenteral and oral modes of administration.

Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application can include any of the following components: a sterile diluent, such as water for injection, saline solution, fixed oil, polyethylene glycol, glycerine, propylene glycol or other synthetic solvent; antimicrobial agents, such as benzyl alcohol and methyl parabens; antioxidants, such as ascorbic acid and sodium bisulfite; chelating agents, such as ethylenediaminetetraacetic acid (EDTA); buffers, such as acetates, citrates and phosphates; and agents

for the adjustment of tonicity such as sodium chloride or dextrose.

Parenteral preparations can be enclose in ampules, disposable syringes or single or multiple dose vials made of glass, plastic or other suitable material.

In instances in which the compounds exhibit insufficient solubility, methods for solubilizing compounds may be used. Such methods are known to those of skill in this art, and include, but are not limited to, using cosolvents, such as dimethylsulfoxide (DMSO), using surfactants, such as TWEEN@, or dissolution in aqueous sodium bicarbonate.

Derivatives of the compounds, such as prodrugs of the compounds may also be used in formulating effective pharmaceutical compositions.

Upon mixing or addition of the compound (s), the resulting mixture may be a solution, suspension, mulsion or the like. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle. The effective concentration is sufficient for ameliorating the symptoms of the disease, disorder or condition treated and may be empirically determined.

The pharmaceutical compositions are provided for administration to humans and animals in unit dosage forms, such as tablets, capsules, pills, powders, granules, sterile parenteral solutions or suspensions, and oral solutions or suspensions, and oil-water mulsions containing suitable quantities of the compounds or pharmaceutically acceptable derivatives thereof. The pharmaceutically therapeutically active compounds and derivatives thereof are typically formulated and administered in unit-dosage forms or multiple-dosage forms. Unit-dose forms as used herein refers to physically discrete units suitable for human and animal subjects and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of the therapeutically active

compound sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carrier, vehicle or diluent.

Examples of unit-dose forms include ampoules and syringes and individually packaged tablets or capsules. Unit-dose forms may be administered in fractions or multiples thereof. A multiple-dose form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dose form. Examples of multiple-dose forms include vials, bottles of tablets or capsules or bottles of pints or gallons. Hence, multiple dose form is a multiple of unit-doses which are not segregated in packaging.

The composition can contain along with the active ingredient: a diluent such as lactose, sucrose, calcium phosphate, or carboxymethylcellulose; a lubricant, such as magnesium stearate, calcium stearate and talc; and a binder such as starch, natural gums, such as gum acaciagelatin, glucose, molasses, polvinylpyrrolidine, celluloses and derivatives thereof, povidone, crospovidones and other such binders known to those of skill in the art. Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, or otherwise mixing an active compound as defined above and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, glycols, ethanol, and the like, to thereby form a solution or suspension. If desired, the pharmaceutical composition to be administered may also contain minor amounts of nontoxic auxiliary substances such as wetting agents, emulsifying agents, or solubilizing agents, pH buffering agents and the like, for example, acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and other such agents. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see

Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 15th Edition, 1975. The composition or formulation to be administered will, in any event, contain a quantity of the active compound in an amount sufficient to alleviate the symptoms of the treated subject.

Dosage forms or compositions containing active ingredient in the range of 0.005% to 100% with the balance made up from non-toxic carrier may be prepared. For oral administration, a pharmaceutically acceptable non-toxic composition is formed by the incorporation of any of the normally employed excipients, such as, for example pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, talcum, cellulose derivatives, sodium crosscarmellose, glucose, sucrose, magnesium carbonate or sodium saccharin. Such compositions include solutions, suspensions, tablets, capsules, powders and sustained release formulations, such as, but not limited to, implants and microencapsulated delivery systems, and biodegradable, biocompatible polymers, such as collagen, ethylene vinyl acetate, polyanhydrides, polyglycolic acid, polyorthoesters, polylactic acid and others. Methods for preparation of these compositions are known to those skilled in the art. The contemplated compositions may contain 0.001%-100% active ingredient, preferably 0.1-85%, typically 75-95%.

The active compounds or pharmaceutically acceptable derivatives may be prepared with carriers that protect the compound against rapid elimination from the body, such as time release formulations or coatings.

The compositions may include other active compounds to obtain desired combinations of properties. The compounds of formulae I or 11, or pharmaceutically acceptable derivatives thereof as described herein, may also be advantageously administered for therapeutic or prophylactic

purposes together with another pharmacological agent known in the general art to be of value in treating one or more of the diseases or medical conditions referred to hereinabove, such as beta-adrenergic blocker (for example atenolol), a calcium channel blocker (for example nifedipine), an angiotensin converting enzyme (ACE) inhibitor (for example lisinopril), a diuretic (for example furosemide or hydrochlorothiazide), an endothelin converting enzyme (ECE) inhibitor (for example phosphoramidon), a neutral endopeptidase (NEP) inhibitor, an HMGCoA reductase inhibitor, a nitric oxide donor, an anti-oxidant, a vasodilator, a dopamine agonist, a neuroprotective agent, a steroid, a beta-agonist, an anti-coagulant, or a thrombolytic agent. It is to be understood that such combination therapy constitutes a further aspect of the compositions and methods of treatment provided herein.

1. Compositions for oral administration Oral pharmaceutical dosage forms are either solid, gel or liquid.

The solid dosage forms are tablets, capsules, granules, and bulk powders. Types of oral tablets include compressed, chewable lozenges and tablets which may be enteric-coated, sugar-coated or film-coated.

Capsules may be hard or soft gelatin capsules, while granules and powders may be provided in non-effervescent or effervescent form with the combination of other ingredients known to those skilled in the art.

In certain embodiments, the formulations are solid dosage forms, preferably capsules or tablets. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder; a diluent; a disintegrating agent; a lubricant; a glidant; a sweetening agent; and a flavoring agent.

Examples of binders include microcrystalline cellulose, gum tragacanth, glucose solution, acacia mucilage, gelatin solution, sucrose and starch paste. Lubricants include talc, starch, magnesium or calcium

stearate, lycopodium and stearic acid. Diluents include, for example, lactose, sucrose, starch, kaolin, salt, mannitol and calcium phosphate.

Glidants include, but are not limited to, colloidal silicon dioxide.

Disintegrating agents include crosscarmellose sodium, sodium starch glycolate, alginic acid, corn starch, potato starch, bentonite, methylcellulose, agar and carboxymethylcellulose. Coloring agents include, for example, any of the approved certified water soluble FD and C dyes, mixtures thereof; and water insoluble FD and C dyes suspended on alumina hydrate. Sweetening agents include sucrose, lactose, mannitol and artificial sweetening agents such as saccharin, and any number of spray dried flavors. Flavoring agents include natural flavors extracted from plants such as fruits and synthetic blends of compounds which produce a pleasant sensation, such as, but not limited to peppermint and methyl salicylate. Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene laural ether. Emetic-coatings include fatty acids, fats, waxes, shellac, ammoniated shellac and cellulose acetate phthalates. Film coatings include hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000 and cellulose acetate phthalate.

If oral administration is desired, the compound could be provided in a composition that protects it from the acidic environment of the stomach. For example, the composition can be formulated in an enteric coating that maintains its integrity in the stomach and releases the active compound in the intestine. The composition may also be formulated in combination with an antacid or other such ingredient.

When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil. In addition, dosage unit forms can contain various other materials which

modify the physical form of the dosage unit, for example, coatings of sugar and other enteric agents. The compounds can also be administered as a component of an elixir, suspension, syrup, wafer, sprinkle, chewing gum or the like. A syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.

The active materials can also be mixed with other active materials which do not impair the desired action, or with materials that supplement the desired action, such as antacids, H2 blockers, and diuretics. For example, if the compound is used for treating asthma or hypertension, it may be used with other bronchodilators and antihypertensive agents, respectively. The active ingredient is a compound or pharmaceutically acceptable derivative thereof as described herein. Higher concentrations, up to about 98% by weight of the active ingredient may be included.

Pharmaceutically acceptable carriers included in tablets are binders, lubricants, diluents, disintegrating agents, coloring agents, flavoring agents, and wetting agents. Enteric-coated tablets, because of the enteric-coating, resist the action of stomach acid and dissolve or disintegrate in the neutral or alkaline intestines. Sugar-coated tablets are compressed tablets to which different layers of pharmaceutically acceptable substances are applied. Film-coated tablets are compressed tablets which have been coated with a polymer or other suitable coating. Multiple compressed tablets are compressed tablets made by more than one compression cycle utilizing the pharmaceutically acceptable substances previously mentioned. Coloring agents may also be used in the above dosage forms. Flavoring and sweetening agents are used in compressed tablets, sugar-coated, multiple compressed and

chewable tablets. Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges.

Liquid oral dosage forms include aqueous solutions, mulsions, suspensions, solutions and/or suspensions reconstituted from non-effervescent granules and effervescent preparations reconstituted from effervescent granules. Aqueous solutions include, for example, elixirs and syrups. Emulsions are either oil-in-water or water-in-oil.

Elixirs are clear, sweetened, hydroalcoholic preparations.

Pharmaceutically acceptable carriers used in elixirs include solvents.

Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may contain a preservative. An mulsion is a two-phase system in which one liquid is dispersed in the form of small globules throughout another liquid. Pharmaceutically acceptable carriers used in mulsions are non-aqueous liquids, emulsifying agents and preservatives.

Suspensions use pharmaceutically acceptable suspending agents and preservatives. Pharmaceutically acceptable substances used in non-effervescent granules, to be reconstituted into a liquid oral dosage form, include diluents, sweeteners and wetting agents. Pharmaceutically acceptable substance used in effervescent granules, to be reconstituted into a liquid oral dosage form, include organic adds and a source of car- bon dioxide. Coloring and flavoring agents are used in all of the above dosage forms.

Solvents include glycerin, sorbitol, ethyl alcohol and syrup.

Examples of preservatives include glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcool. Examples of non-aqueous liquids utilized in mulsions include mineral oil and cottonseed oil.

Examples of emulsifying agents include gelatin, acacia, tragacanth, bentonite, and surfactants such as polyoxyethylene sorbitan monooleate.

Suspending agents include sodium carboxymethylcellulose, pectin,

tragacanth, Veegum and acacia. Diluents include lactose and sucrose.

Sweetening agents include sucrose, syrups, glycerin and artificial sweetening agents such as saccharin. Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether. Organic adds include citric and tartaric acid. Sources of carbon dioxide include sodium bicarbonate and sodium carbonate. Coloring agents include any of the approved certified water soluble FD and C dyes, and mixtures thereof. Flavoring agents include natural flavors extracted from plants such fruits, and synthetic blends of compounds which produce a pleasant taste sensation.

For a solid dosage form, the solution or suspension, in for example propylene carbonate, vegetable oils or triglycerides, is preferably encapsulated in a gelatin capsule. Such solutions, and the preparation and encapsulation thereof, are disclosed in U. S. Patent Nos 4,328,245; 4,409,239; and 4,410,545. For a liquid dosage form, the solution, e. a., for example, in a polyethylene glycol, may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e. a., water, to be easily measured for administration.

Alternatively, liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active compound or salt in vegetable oils, glycols, triglycerides, propylene glycol esters (e. g., propylene carbonate) and other such carriers, and encapsulating these solutions or suspensions in hard or soft gelatin capsule shells. Other useful formulations include those set forth in U. S. Patent Nos.

Re 28,819 and 4,358,603.

In all embodiments, tablets and capsules formulations may be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient. Thus, for example, they may be

coated with a conventional enterically digestible coating, such as phenylsalicylate, waxes and cellulose acetate phthalate.

2. Injectables, solutions and mulsions Parenteral administration, generally characterized by injection, either subcutaneousiy, intramuscularly or intravenously is also contemplated herein. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as mulsions. Suitable excipients are, for example, water, saline, dextrose, glycerol or ethanol.

In addition, if desired, the pharmaceutical compositions to be administered may also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate and cyclodextrins. Implantation of a slow-release or sustained-release system, such that a constant level of dosage is maintained (see, e. a., U. S. Patent No. 3,710,795) is also contemplated herein. The percentage of active compound contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the activity of the compound and the needs of the subject.

Parenteral administration of the compositions includes intravenous, subcutaneous and intramuscular administrations. Preparations for parenteral administration include sterile solutions ready for injection, sterile dry soluble products, such as lyophilized powders, ready to be combined with a solvent just prior to use, including hypodermic tablets, sterile suspensions ready for injection, sterile dry insoluble products ready to be combined with a vehicle just prior to use and sterile mulsions. The solutions may be either aqueous or nonaqueous.

If administered intravenously, suitable carriers include physiological saline or phosphate buffered saline (PBS), and solutions containing thickening and solubilizing agents, such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof.

Pharmaceutically acceptable carriers used in parenteral preparations include aqueous vehicles, nonaqueous vehicles, antimicrobial agents, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, emulsifying agents, sequestering or chelating agents and other pharmaceutically acceptable substances.

Examples of aqueous vehicles include Sodium Chloride Injection, Ringers Injection, Isotonic Dextrose Injection, Sterile Water Injection, Dextrose and Lactated Ringers Injection. Nonaqueous parenteral vehicles include fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil and peanut oil. Antimicrobial agents in bacteriostatic or fungistatic concentrations must be added to parenteral preparations packaged in multiple-dose containers which include phenols or cresols, mercurials, benzyl alcool, chlorobutanol, methyl and propyl p-hydroxybenzoic acid esters, thimerosal, benzalkonium chloride and benzethonium chloride.

Isotonic agents include sodium chloride and dextrose. Buffers include phosphate and citrate. Antioxidants include sodium bisulfate. Local anesthetics include procaine hydrochloride. Suspending and dispersing agents include sodium carboxymethylcelluose, hydroxypropyl methylcellulose and polyvinylpyrrolidone. Emulsifying agents include Polysorbate 80 (TWEENs 80). A sequestering or chelating agent of metal ions include EDTA. Pharmaceutical carriers also include ethyl alcool, polyethylene glycol and propylene glycol for water miscible vehicles and sodium hydroxide, hydrochloric acid, citric acid or lactic acid for pH adjustment.

The concentration of the pharmaceutically active compound is adjusted so that an injection provides an effective amount to produce the desired pharmacological effect. The exact dose depends on the age, weight and condition of the patient or animal as is known in the art.

The unit-dose parenteral preparations are packaged in an ampoule, a vial or a syringe with a needle. All preparations for parenteral administration must be sterile, as is known and practiced in the art.

Illustratively, intravenous or intraarterial infusion of a sterile aqueous solution containing an active compound is an effective mode of administration. Another embodiment is a sterile aqueous or oily solution or suspension containing an active material injected as necessary to produce the desired pharmacological effect.

Injectables are designed for local and systemic administration.

Typically a therapeutically effective dosage is formulated to contain a concentration of at least about 0.1 % w/w up to about 90% w/w or more, preferably more than 1 % w/w of the active compound to the treated tissue (s). The active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is understood that the precise dosage and duration of treatment is a function of the tissue being treated and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data. It is to be noted that concentrations and dosage values may also vary with the age of the individual treated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the formulations, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed formulations.

The compound may be suspended in micronized or other suitable form or may be derivatized to produce a more soluble active product or to produce a prodrug. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle. The effective concentration is sufficient for ameliorating the symptoms of the condition and may be empirically determined.

3. Lyophilized powders Of interest herein are also lyophilized powders, which can be reconstituted for administration as solutions, mulsions and other mixtures. They may also be reconstituted and formulated as solids or gels.

The sterile, lyophilized powder is prepared by dissolving a compound of formula (I) or (II) in a buffer solution. The buffer solution may contain an excipient which improves the stability or other pharmacological component of the powder or reconstituted solution, prepared from the powder. Subsequent sterile filtration of the solution followed by lyophilization under standard conditions known to those of skill in the art provides the desired formulation. Briefly, the lyophilized powder is prepared by dissolving an excipient, such as dextrose, sorbital, fructose, corn syrup, xylitol, glycerin, glucose, sucrose or other suitable agent, in a suitable buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, typically, about neutral pH. Then, a selected compound of formula (I) or (II) is added to the resulting mixture, and stirred until it dissolves. The resulting mixture is sterile filtered or treated to remove particulates and to insure sterility, and apportioned into vials for lyophilization. Each vial will contain a single dosage (100-500 mg, preferably 250 mg) or multiple

dosages of the compound. The lyophilized powder can be stored under appropriate conditions, such as at about 4 °C to room temperature.

Reconstitution of this lyophilized powder with water for injection provides a formulation for use in parenteral administration. For reconstitution about 1-50 mg, preferably 5-35, more preferably about 9- 30 is added per mL of sterile water or other suitable carrier. The precise amount depends upon the indication treated and selected compound.

Such amount can be empirically determined.

4. Topical administration Topical mixtures are prepared as described for the local and systemic administration. The resulting mixture may be a solution, suspension, mulsions or the like and are formulated as creams, gels, ointments, mulsions, solutions, elixirs, lotions, suspensions, tinctures, pastes, foams, aerosols, irrigations, sprays, suppositories, bandages, dermal patches or any other formulations suitable for topical administration.

The compounds or pharmaceutically acceptable derivatives thereof may be formulated as aerosols for topical application, such as by inhalation (see, e. q., U. S. Patent Nos. 4,044,126,4,414,209, and 4,364,923, which describe aerosols for delivery of a steroid useful for treatment inflammatory diseases, particularly asthma). These formulations for administration to the respiratory tract can be in the form of an aerosol or solution for a nebulizer, or as a microfine powder for insufflation, alone or in combination with an inert carrier such as lactose.

In such a case, the particles of the formulation will typically diameters of less than 50 microns, preferably less than 10 microns.

The compounds may be formulated for local or topical application, such as for topical application to the skin and mucous membranes, such as in the eye, in the form of gels, creams, and lotions and for application

to the eye or for intracisternal or intraspinal application. Topical administration is contemplated for transdermal delivery and also for administration to the eyes or mucosa, or for inhalation therapies. Nasal solutions of the active compound alone or in combination with other pharmaceutically acceptable excipients can also be administered.

These solutions, particularly those intended for ophthalmic use, may be formulated as 0.01 %-10% isotonic solutions, pH about 5-7, with appropriate salts.

5. Compositions for ophthalmic administration For ophthalmic indications, the compositions are formulated in an ophthalmically acceptable carrier. For the ophthalmic uses herein, local administration, either by topical administration or by injection is preferred. Time release formulations are also desirable. Typically, the compositions are formulated for single dosage administration, so that a single dose administers an effective amount.

Ophthalmologically effective concentrations or amounts of one or more of the compounds are mixed with a suitable pharmaceutical carrier or vehicle. The concentrations or amounts of the conjugates that are effective requires delivery of an amount, upon administration, that prevents or substantially reduces the effects of FGF-mediated ophthalmological conditions, including, but not limited to, diabetic retinopathy, corneal clouding following excimer laser surgery, closure of trabeculectomies, hyperproliferation of lens epithelial cells following cataract surgery and the recurrence of pterygii.

The compounds can also be mixed with other active materials, that do not impair the desired action, or with materials that supplement the desired action, including viscoelastic materials, such as hyaluronic acid, which is sold under the trademark HEALON (solution of a high molecular weight (MW of about 3 million) fraction of sodium

hyaiuronate; manufactured by Pharmacia, Inc. see, e a., U. S. Patent Nos.

5,292,362,5,282,851,5,273,056,5,229,127,4,517,295 and 4,328,803), VISCOAT (fluorine-containing (meth) acrylates, such as, <BR> <BR> <BR> <BR> 1 H, 1 H, 2H, 2H-heptadecafluorodecylmethacrylate; see, e. a., U. S. Patent Nos. 5,278,126,5,273,751 and 5,214,080; commercially available from Alcon Surgical, Inc.), ORCOLON (see, e. a., U. S. Patent Nos. 5,273,056; commercially available from Optical Radiation Corporation), methylcellulose, methyl hyaluronate, polyacrylamide and polymethacrylamide (see, e. a., U. S. Patent No. 5,273,751). The viscoelastic materials are present generally in amounts ranging from about 0.5 to 5.0%, preferably 1 to 3% by weight of the conjugate material and serve to coat and protect the treated tissues. The compositions may also include a dye, such as methylene blue or other inert dye, so that the composition can be seen when injected into the eye or contacted with the surgical site during surgery.

6. Compositions for other routes of administration Depending upon the condition treated other routes of administration, such as topical application, transdermal patches, an rectal administration are also contemplated herein.

For example, pharmaceutical dosage forms for rectal administration are rectal suppositories, capsules and tablets for systemic effect. Rectal suppositories are used herein mean solid bodies for insertion into the rectum which melt or soften at body temperature releasing one or more pharmacologically or therapeutically active ingredients. Pharmaceutically acceptable substances utilized in rectal suppositories are bases or vehicles and agents to raise the melting point.

Examples of bases include cocoa butter (theobroma oil), glycerin-gelatin, carbowax (polyoxyethylene glycol) and appropriate mixtures of mono-, di-and triglycerides of fatty acids. Combinations of the various bases

may be used. Agents to raise the melting point of suppositories include spermaceti and wax. Rectal suppositories may be prepared either by the compressed method or by molding. The typical weight of a rectal suppository is about 2 to 3 gm.

Tablets and capsules for rectal administration are manufactured using the same pharmaceutically acceptable substance and by the same methods as for formulations for oral administration.

7. Articles of manufacture The pharmaceutical compositions containing compounds or pharmaceutically acceptable derivatives may be packaged as articles of manufacture containing packaging material, a composition containing a compound or pharmaceutically acceptable derivative thereof provided herein, which is effective for antagonizing the effects of an FGF peptide, preferably bFGF, ameliorating the symptoms of an FGF-mediated disorder, or inhibiting binding of an FGF peptide to an FGF receptor with an ICSO of less than about 500, uM, within the packaging material, and a label that indicates that the composition containing the compound or derivative thereof is used for antagonizing the effects of FGF, treating FGF-mediated disorders or inhibiting the binding of an FGF peptide to an FGF receptor.

D. Evaluation of the bioactivity of the compounds Standard physiological, pharmacological and biochemical procedures are available for testing the compounds to identify those that possess biological activities of compounds that interfere with or inhibit or otherwise modulate the activity of FGF peptides. Numerous assays are known to those of skill in the art for evaluating the ability of compounds to modulate the activity of one or more FGF peptides. For example, the properties of a potential antagonist may be assessed as a function of its <BR> <BR> <BR> ability to inhibit FGF activity including the ability in vitro to compete for

binding to FGF receptors present on the surface of tissues or recombinant cell lines, cell-based competitive assays (see, e. g., Moscatelli eta/. J. Cell. Phvsiol. mitogenic assays (Gospodarowiczeta/. Proc. Natl. Acad. Sci. U. S. A. 1984,81,6963- 6967; Thomas eta/. Proc. Natl. Acad. Sci. U. S. A. 1984,81,357); stimulation of angiogenesis in vitro (see, e. q., European Patent Application No. EP 645 451); cell proliferation assays or heparin binding assays (see, e. g., International Application Publication No. WO 92/12245); assays measuring the release of cellular proteases (Moscatelli eta/. Proc. Natl. Acad. Sci. U. S. A. 1986,83,2091-2095; Phadke Biochem. Biophvs. Res. Comm. and, assays for the promotion of FGF-mediated neurite outgrowth and neuron <BR> <BR> <BR> <BR> survival (Togari eta/. Biochem. Biophys. Res. Comm. 1983,114, 1189- 1193; Wagnereta/. J. Cell Biol. 1986, 103,1363-1367).

In addition, FGF isotype specific antagonists may be identified by the ability of a test compound to interfere with one or more FGF peptide binding to different tissues or cells expressing different endothelin receptor subtypes, or to interfere with the biological effects of an FGF peptide (see, e. a., International Patent Application Publication No. WO 95/24414).

Using such assays, the relative affinities of the compounds for FGF receptors have been and can be assessed. Those that possess the desired in vitro properties, such as specific inhibition of the binding of bFGF, are selected. The selected compounds that exhibit desirable activities may be therapeutically useful in the methods described herein and are tested for such uses employing the above-described assays from which the in vivo effectiveness may be evaluated (Gospodarowicz et a/.

Endocrin. Rev. 1987,8,95-114; Buntrock et a/. Exp. Pathol. 1982,21, 62-67; International Patent Application Publication No WO 92/08473).

Compounds that exhibit the in vitro activities that correlate with the in vivo effectiveness will then be formulated in suitable pharmaceutical compositions and used as therapeutics.

E. Methods of treating of FGF-mediated disorders Methods using the compositions containing therapeutically effective concentrations of the compounds of formulae I or 11, or pharmaceutically acceptable derivatives thereof for treating disorders, particularly proliferative disorders, in which FGF causes or contributes to the pathology are provided herein. In particular, methods for using the compositions to prevent the undesired growth and proliferation of FGF- sensitive cells occurring in vascular disorders characterized by accelerated smooth muscle cell proliferation, such as rheumatoid arthritis, tumor angiogenesis, Kaposi's sarcoma, restenosis, In-Stent restenosis, certain ophthalmic disorders and dermatological disorders, such as psoriasis, are provided herein.

Preferably, the medicament containing the compound is administered intravenously (IV), although treatment by localized administration may be tolerated in some instances. Generally, the medicament containing the compound is injected into the circulatory system of a subject in order to deliver a dose to the targeted cells.

Targeting may be effected by linking the compound to a targeting agent specific for FGF receptors, particularly bFGF receptors. Dosages may be determined empirically, but will typically be in the range of about 0.01 mg to about 100 mg of the compound per kilogram of body weight as a daily dosage.

1. Restenosis and vascular injury Methods for treating vascular injury, particularly, restenosis or In- Stent restenosis by contacting the vascular wall with an effective amount of a composition containing compound (s) of formulae I or 11 are <BR> <BR> <BR> <BR> provided (see generally, Lindner et a/. Proc. Natl. Acad. Sci. USA 1991, 88,3739; Kearney et au Circulation 1997,95,1998).

Atherosclerosis, also referred to as arteriosclerosis, results from the development of an intimal lesion and the subsequent narrowing of the vessel lumen. Frequently, atherosclerosis originally appears as a result of the buildup of plaque which lines the interior of blood vessels, particularly the arteries. Whereas bypass surgery is sometimes employed to replace such clogged arteries, in recent years, a number of surgical procedures have been developed so as to interarterially remove such plaque, often by balloon catheterization or other such treatments in which the plaque is either compressed against or scraped away from the interior surface of the artery. This scraping of the interior wall removes endothelial cells, which constitute the lining of the blood vessel. As a result of this removal, the smooth muscle cells (SMCs), which are normally located exterior of the endothelial cells (ECs) and form the blood vessel structure, begin to grow and multiply causing a narrowing of the vessel lumem. Not infrequently, the patient so treated finds a recurrence of such narrowing of the vessel lumen in a relatively short period thereafter as a result of this proliferation, generally referred to as restenosis, requiring a repetition of the surgical procedure to again remove the increasing blockage.

Proliferating SMCs express functional FGF receptors and are responsive to bFGF. By inhibiting proliferation of migrating smooth muscle cells (SMCs), it is possible to prevent the undesirable growth and ultimate clogging which occurs following vascular injury, and which is

generally referred to as restenosis. Basic FGF appears to play a pivotal role in the subsequent responses of the vascular wall. Basic FGF is known to be synthesized by endothelial and smooth muscle cells (SMCs) and is thought to be stored in the subendothelial matrix, and in some instances, this growth factor is released from cells after injury.

Therefore, compounds that inhibit FGF-mediated proliferation of SMCs may be used in methods for treating restenosis by preventing the proliferation that causes the narrowing of the vessel lumem.

Treatment is effected by administering a therapeutically effective amount of a medicament containing the compound in a physiologically acceptable carrier or recipient, in a manner so that the compound reaches regions in a human or other mammal where the compound will inhibit the proliferation of the target cells. For restenosis, intraarterial infusion will be among the preferred methods. Although a single dose should inhibit neointimal proliferation, IV administration over a period of time is preferred.

The compounds for treating restenosis may be formulated for intravenous or local administration. Alternatively, compounds may be conjugated to an agent that specifically targets proliferating SMCs, such as antibodies, hormones, ligands or the like to improve delivery and uptake of the compound. The therapeutically effective concentration may be determined empirically by testing the compounds in known in <BR> <BR> <BR> <BR> vitro and in vivo systems (see, e. a., Moscatelli et al. J. Cell. Phvsiol.<BR> <BR> <BR> <BR> <BR> <BR> <P>1987,131,123-130); mitogenic assays (Gospodarowicz et a/. Proc.

Natl. Acad. Sci. U. S. A. 1984,81,6963-6967; Thomas etal. Proc. Natl.

Acad. Sci. U. S. A. 1984,81,357); stimulation of angiogenesis in vitro (see, e. a., European Patent Application No. EP 645 451); cell proliferation assays or heparin binding assays (see, e.q., International Application Publication No. WO 92/12245); assays measuring the

release of cellular proteases (Moscatelli et a/. Proc. Natl. Acad. Sci.

U. S. A. Phadke Biochem. Biophys. Res. Comm. and, assays for the promotion of FGF-mediated neurite outgrowth and neuron survival (Togari et a/. Biochem. Biophvs.

Res. Comm. 1189-1193 ; Wagnereta/. J. Cell Biol. 1986, 103,1363-1367) and then extrapolated therefrom for dosages for humans.

2. Rheumatoid arthritis Rheumatoid arthritis is a systemic, chronic inflammatory disease, that is characterized by the destruction of the joint cartilage and inflammation of the synovium. The hallmark feature of rheumatoid arthritis is the production circulating autoantibodies, also referred to as rheumatoid factors, which are reactive with the Fc portions of the patients own IgG molecules (e. a., see Abbas et a/., Cellular and Molecular Immunoloav, W. B. Saunders Co., Philadelphia, PA).

One of the systemic complications of rheumatoid arthritis is the formation of injurious immune complexes in the synovial fluid of the joints that initiates vascular inflammation by activation of the complement cascade. T-cells, activated B-cells, plasma cells and macrophages are often found in synovial fluid of affected joints as well as a variety of soluble proteins, such as cytokines (e.q., interleukin-1, IFN-y and tumor necrosis factor (TNF)) and growth factors, such as bFGF. It has been suggested that cytokines act in concert with the inflammatory mediators, e. a., bFGF, to cause local tissue destruction.

Chronically, cytokines and bFGF stimulate fibroblast and collagen proliferation resulting in angiogenesis, and prolonge exposure can result in hyperproliferation of epithelial cells that form fibrous tissue, referred to as fibrosis.

Thus, compounds that inhibit the FGF-mediated hyperproliferation of epithelial cells may be used to treat rheumatoid arthritis. The compounds for treating rheumatoid arthritis may be formulated for oral administration or intravenous injection and an effective concentration may be administered. The effective concentration is sufficient for ameliorating the symptoms of the disease, disorder or condition treated and may be empirically determined.

3. Tumor Angiogenesis Angiogenesis plays a critical role in embryonic development and in several physiologic and pathologic conditions, including wound healing, ovulation, diabetic retinopathy and malignancy. In particular, without the nutrients and oxygen provided via this neovascularization, solid tumors would be unable to grow beyond about 2 mm in diameter.

Evidence exists that several cancers, including melanomas, ovarian, pancreatic and some colon carcinomas, have receptors for bFGF. Testing with radioactive binding assays on a number of human carcinogenic cell lines isolated from human cancers demonstrated that many but not all of these cell lines bind 1251-FGF. Thus, compounds that inhibit the activity of FGF may be used to treat tumorigenic pathophysiological conditions caused by a proliferation of cells which are sensitive to FGF mitogenic stimulation. In addition, tumor growth can be inhibited by modulating FGF receptor activity in the components of blood vessels (e. g., vascular endothelial cells or vascular SMCs) (Halberman <BR> <BR> <BR> <BR> Anqioqenesis 1996,98-1, Colville-Nash et al. Molec. Med. Todav 1997,<BR> <BR> <BR> <BR> <BR> <BR> 14; Shawver et a/. Drug Discoverv Todav 1997,2,50).

The compounds may be specifically targeted to tumorigenic tissues by linking the compound to an agent that specifically binds to the surface of the tumorigenic cell, e.q., an anti-tumor antigen antibody, or linking the compound to an agent that is preferentially interacts with or

taken up by targeted tumor. In addition, compounds may be encapsulated in tissue-targeted liposomal suspensions for targeted delivery of the compound.

The compounds for treating tumor angiogenesis may be formulated for topical application and administered to the skin, e.q., for treatment of melanoma, or may be formulated for intravenous administration for treatment of solid tumors, such as carcinomas. The therapeutically effective concentration may be determined empirically by testing the compounds in known in vitro (e. q., inhibition of angiogenesis in vitro (see, e. q., European Patent Application No. EP 645 451)) and then extrapolated therefrom for dosages for humans.

4. Ophthalmic Disorders Pharmaceutical compositions provided herein may be used in methods of treating ophthalmic disorders resulting from FGF-mediated hyper-proliferation of lens epithelial cells, fibroblasts or keratinocytes. In particular, ophthalmic disorders that may be treated using the methods and compositions provided herein include, but are not limited to, corneal clouding following excimer laser surgery, closure of trabeculectomies, hyperproliferation of lens epithelial cells following cataract surgery, the recurrence of pterygii and diabetic retinopathy (see, Dell Druq Discoverv Todav 1996,1,221).

The compounds for treating ophthalmic disorders may be formulated for local or topical application and administered by topical application of an effective concentration to the skin and mucous membranes, such as in the eye. The compositions may also include a dye, such as methylene blue or other inert dye, so that the composition can be seen when injected into the eye or contacted with the surgical site during surgery. The effective concentration is sufficient for

ameliorating the symptoms of the disease, disorder or condition treated and may be empirically determined.

The following examples are included for illustrative purposes only and are not intended to limit the scope of the invention.

EXAMPLE 1 1-Naphthaldehyde aminoimino hydrazone hydrochloride 1-Naphthaldehyde (781 mg, 5.0 mmol) and aminoguanidine hydrochloride (553 mg, 5.0 mmol) were heated under reflux in a mixture of water (2 mL) and methanol (2 mL) for 20 min. The solvents were evaporated in vacuo to give a viscous oil. The crude product was recrystallized from ethyl acetate/hexanes to give 762 mg (61 % yield) of white crystals, mp 150-152 °C.

EXAMPLE 2 Benzaldehyde aminoimino hydrazone hydrochloride Using the method described in Example 1, the title compound was synthesized from benzaldehyde (5.0 mmol) and aminoguanidine hydrochloride (5.0 mmol). The crude product was recrystallized from hot methanol to give very hygroscopic white crystals, mp 65-70 °C.

EXAMPLE 3 Fmoc-Phe-Arg-Benzylamide A. Fmoc-Arg (Pmc)-Benzylamide Fmoc-Arg (Pmc)-OH (1.324 g) was dissolved in N-methylpiperidone (NMP). HOBt (1-hydroxybenzotriazole) (0.306 g) was added followed by DCC (dicyclohexylcarbodiimide) (0.412 g). The mixture was stirred for 25 min. Benzylamine (0.196 mL) was added slowly in six equal amounts. The mixture was stirred overnight. The mixture was diluted with water and extracted into ethyl acetate (EtOAc) (3 x 25 mL). The EtOAc extract was washed with 1 N HCI (1 x 10 mL) and 5% sodium bicarbonate solution (2 x 20 mL) and brine solution (2 x 20 mL) and then

dried over anhydrous sodium sulfate. The EtOAc on evaporation gave a solid. Yield: 1.2 g (89%).

B. Fmoc-Phe-Arg (Pmc)-benzylamide Fmoc-Arg (Pmc)-Benzylamide (1.1 g) was treated with 25% piperidine in DMF (dimethylformamide). The mixture was stirred for 1 hr, when TLC showed the absence of starting material, the solution was concentrated to a small volume and diluted with ether. The white solid precipitated was filtered, washed with ether and dried under vacuum.

Yield: 0.7 g.

Fmoc-Phe-OH (0.774 g) was dissolved in NMP. HOBt (0.306 g) and DCC (0.412 g) were added and the mixture was stirred for 25 min.

H-Arg (PMC)-Benzylamide (0.4 g) was added and the mixture was stirred overnight. The reaction mixture was diluted with water and extracted with EtOAc (3 x 20 mL). The EtOAc was washed with 1 N HCI (1 x 10 mL), 5% sodium bicarbonate solution (2 x 20 mL) and finally water (2 x 10 mL). The EtOAc solution was dried over anhydrous sodium sulfate <BR> <BR> <BR> <BR> and evaporated to a solid. Yield: 0.60 g (89%). The solid was purified by HPLC. HPLC: 99% pure. Mass Spectra Calc.: 898.4, Found: 899.4.

C. Fmoc-Phe-Arg-Benzylamide Fmoc-Phe-Arg (Pmc)-Benzylamide (0.2 g) was treated with 95% trifluoroacetic acid (TFA)/water. The mixture was stirred for 1 hr. TLC showed the absence of the starting material. TFA was evaporated to dryness and the residue on trituration with ether gave a solid. The solid was filtered, washed with ether and dried under vacuum. Yield: 0.12 g (85%). The solid was purified by HPLC. Purity (HPLC): 100%. Mass Spectra: Cale.: 632.8, Found: 633.4.

EXAMPLE 4 Fmoc-Phe-dArg-Benzylamide A. Fmoc-dArg (Pmc)-Benzylamide

Fmoc-dArg (Pmc)-OH (1.324 g) was dissolved in N- <BR> <BR> <BR> <BR> methylpiperidone (NMP). HOAt (1-hydroxy-7-azabenzotriazole) (0.272 g) was added followed by diisopropylcarbodiimide (DIC) (0.252 g). The mixture was stirred for 25 min. Benzylamine (0.196 mL) was added slowly in six equal amounts. The mixture was stirred overnight. The mixture was diluted with water and extracted into EtOAc (3 x 25 mL).

The EtOAc extract was washed with 1 N HCI (1 x 10 mL), 5% sodium bicarbonate solution (2 x 20 mL) and brine solution (2 x 20 mL), and then dried over anhydrous sodium sulfate. The EtOAc on evaporation gave a solid. Yield: 1.11 g (82%).

B. Fmoc-Phe-dArg (Pmc)-Benzylamide Fmoc-dArg (Pmc)-Benzylamide (1.1 g) was treated with 25% piperidine in DMF. The mixture was stirred for 1 hr, when TLC showed the absence of starting material, the solution was concentrated to a small volume and diluted wtih ether. The white solid which precipitated was filtered, washed with ether and dried under vacuum. This product used in the next reaction.

Fmoc-Phe-OH (0.587 g) was dissolved in NMP. HOAt (0.206 g) and DIC (0.19 g) were added and the mixture was stirred for 25 min. H- dArg (Pmc)-Benzylamide was added and the mixture was stirred overnight. The reaction mixture was diluted with water and extracted with EtOAc (3 x 20 mL). The EtOAc was washed with 1 N HCI (1 x 10 mL), 5% sodium bicarbonate solution (2 x 20 mL) and finally water (2 x 10 mL). The EtOAc solution was dried over anhydrous sodium sulfate and evaporated to get a solid. Yield: 0.335 g (62%). The solid was purified by HPLC. HPLC: 98% pure. Mass Spectra: Calc: 898.4, Found: 899.6.

C. Fmoc-Phe-dArg-Benzylamide

Fmoc-Phe-dArg (Pmc)-Benzylamide (0.3 g) was treated with 95% trifluoroacetic acid (TFA)/water. The mixture was stirred for 1 hr. TLC showed the absence of the starting material. TFA was evaporated to dryness and the residue on trituration with ether gave a solid. The solid was filtered by HPLC. Purity (HPLC): 100%. Mass Spectra: Calc.: 632.8, Found: 633.6.

EXAMPLE 5 Fmoc-dPhe-Arg-Benzylamide A. Fmoc-dPhe-Arg (Pmc)-Benzylamide The procedure was the same as Example 4. B. Fmoc-dPhe-OH (0.387 g), HOAt (0.136 g), DIC (0.126 g) and H-Arg (Pmc)-benzylamide (0.24 g) were used. The yield was 0.55 g (61 %). The solid was purified by HPLC. Purity: 100%. Mass Spectra: Calc.: 898.8, Found: 898.3.

B. Fmoc-dPhe-Arg-Benzylamide The procedure was the same as Example 4. C. Fmoc-dPhe- <BR> <BR> <BR> <BR> Arg (Pmc)-Benzylamide (0.3 g) was used. The yield was 0.15 g (71 %).

The solid was purified by HPLC. Purity: 100%. Mass Spectra: Cale.: 632.8, Found: 633.8.

EXAMPLE 6 Fmoc-dPhe-d-Arg-Benzylamide A. Fmoc-dPhe-dArg (Pmc)-Benzylamide The procedure was same as Example 4. B. Fmoc-dPhe-OH (0.58 g), HOAt (0.204 g), DIC (0.191 g) and H-Arg (Pmc)-benzylamide (0.32 g) were used. The yield was 0.37 g (67%). The solid was purified by HPLC. Purity: 100%. Mass Spectra: Calc. 898.8, Found: 898.3.

B. Fmoc-dPhe-dArg-Benzylamide The procedure was same as Example 4. C. Fmoc dPhe-Arg (Pmc)- <BR> <BR> <BR> Benzylamide (0.3 g) was used. The yield was 0.16 g (75%). The solid

was purified by HPLC. Purity: 100%. Mass Spectra: Calc.: 632.8, Found: 633.8.

EXAMPLE 7 Fmoc-Tyr-Arg-Benzylamide A. Fmoc- Tyr (O'Bu)-Arg (Pmc)-Benzylamide Fmoc-Tyr (OtBu)-OH (0.46 g) was dissolved in NMP (5 mL). With stirring HOAt (0.136 g) and DIC (0.126 g) were added followed by H- Arg (Pmc)-benzylamide (see Example 3. B.). The mixture was stirred overnight and worked up as in Example 4. B. The solid was purified by <BR> <BR> <BR> <BR> HPLC. Purity: 100%. Yield: 0.4 g (91%). Mass Spectra: Calc.: 970.5, Found: 971.4.

B. Fmoc-Tyr-Arg-Benzylamide Fmoc-Tyr (OtBu)-Arg (Pmc)-Benzylamide (0.3 g) was treated with a mixture of TFA/water/triisopropylsilane 10/0.5/0.5 and the mixture was stirred for 60 min. The reaction was worked as in Example 3. A. The compound was purified by HPLC. Purity: 100%. Mass Spectra: Calc.: 648.3, Found: 648.8.

EXAMPLE 8 The following compounds were prepared by minor modifications of the methods in Examples 1-7: Fmoc-Ala-Arg-benzylamide, Fmoc-Leu- Arg-benzylamide, Fmoc-Arg (Pmc)-Phe-benzylamide, Fmoc-Arg-Phe- benzylamide, Fmoc-dArg (Pmc)-Phe-benzylamide, Fmoc-dArg-Phe- benzylamide, Fmoc-Arg (Pmc)-dPhe-benzylamide, Fmoc-dArg-Phe- benzylamide, Fmoc-dArg-(Pmc)-dPhe-benzylamide,(Pmc)-dPhe-benzylamide, Fmoc-dArg-dPhe- benzylamide, Fmoc-Lys-Phe-benzylamide.

EXAMPLE 9 Assays for identifying compounds that exhibit FGF antagonistic activity A. Soluble FGF receptor assay Compounds of formulae I or 11 that exhibit FGF antagonist activity were and can be identified by testing their ability to compete with 1251- bFGF for binding to one or more FGF receptor or FGF-binding fragment thereof. In one embodiment, a recombinant FGF receptor fusion protein was used in which the extracellular domain of a human FGF receptor, FGFR1, was fused to the amino terminal fragment of tissue plasminogen activator (tPA) protein. This fusion protein retains the ability to bind FGF, such as bFGF (Zhu etal. J. Biol. Chem. 1995,270,21869-21874).

1. Isolation of DNA encoding the shorter form of human fibroblast growth factor receptor 1 (FGFR1) The nucleotide sequence of the DNA encoding the shorter form of human basic fibroblast growth factor receptor 1 (FGFR1) has been determined (e. a., Itoh etal. Biochem. Biophys. Res. Comm. 1990, 169: 680-685). This shorter form of FGFR1 is a 731 amino acid polypeptide that has a signal peptide, two extracellular immunoglobulin- like domains, a transmembrane domain and an intracellular tyrosine kinase domain.

Based on the reported sequence, two oligonucleotides complementary to sequences flanking the FGFR1 coding region were synthesized and used as primers in polymerase chain reactions (PCR) to isolate a DNA encoding a full-length human FGFR1 from a human aorta cDNA-library (Quickclone, Clontech, Palo Alto, CA). PCR amplification was performed using a commercially available PCR kit according to manufacturer's instructions (Perkin Elmer Cetus, Norwalk, CT). An oligonucleotide corresponding to nt-20 to + 5, relative to the A of the <BR> <BR> ATG initiation codon of FGFR1, (e. a., Itoh et a/. Biochem. Biophys. Res.

Comm. 1990,169,680-685) and an oligonucleotide complementary to nt 2218-2243 were used as primers to amplify a 2,243 bp PCR product encoding the entire FGRF1 coding region.

The full-length FGFR1-encoding DNA was used as a template for a subsequent PCR reaction, performed as described above, to amplify a 869 bp DNA fragment encoding only the FGFR1 extracellular domain.

Simultaneously, a Hindlll restriction endonuclease site was introduced upstream of the FGFR1 initiation codon and a Sall site was introduced downstream of the second immunoglobulin-like extracellular domain (Igil) to facilitate cloning of the amplifie product.

The Hindlll site was introduced at nt-8 to-3 during the PCR reaction by synthesizing an oligonucleotide primer corresponding to nt- 12 to + 22 that introduced nucleotide changes at three positions in the FGFR1 sequence: nt-3 (G to T), nt-6 (A to G) and nt-8 (G to A). The Sall site was introduced at nt 849 to nt 854 by synthesizing an oligonucleotide primer complementary to nt 823 to 857 containing nucleotide substitutions at three positions in the FGFR1 sequence: nt 849 (C to G), nt 851 (G to C) and nt 854 (G to C). The 857 bp PCR fragment was incubated with Hindlil and Sall and purified by agarose gel electrophoresis according to the standard procedures (Sambrook et al.

(1989) Molecular Cloning, 2nd ed., Cold Spring Harbor Laboratory Press, New York). The DNA was isolated from gel by electroelution and recovered by precipitation with ethanol.

Thus, the resulting Hindlll to Sall DNA fragment consists of nt-7 to nt 849 of the FGFR1 cDNA described by Itoh et a/and encodes amino acid residues 1 to 284 of the shorter form of the bFGF receptor.

2. Isolation of DNA encoding human tissue plasminogen activator

The nucleotide sequence of the DNA encoding human tissue <BR> <BR> <BR> <BR> plasminogen activator (tPA) has been determined (e. a., see Pennica et al.

Nature Human tPA is a 562 amino acid polypeptide which is processed during secretion to its mature form by cleavage of a 35 amino acid signal peptide. Several regions of the primary structure of mature tPA have a high degree of homology to known structural domains of other proteins, such as homology to the finger and growth factor domains, the Kringle 1 and Kringle 2 domains of plasminogen and prothrombin and the C-terminal serine protease domain (e.a., see Ny et aL Proc. Natl. Acad. Sci. USA 1984,81,5355).

Based on the reported sequence, oligonucleotides complementary to sequences flanking the tPA coding region were synthesized and used as primers in PCR reactions to isolate a full-length cDNA encoding human tPA from a human placenta cDNA library (Clontech, Palo Alto, CA). An oligonucleotide corresponding to nt-6 to + 21, relative to the A of the initiation codon of the of human tPA prepro polypeptide (e-q., see Pennica etal. Nature 1983,301,214-221) and an oligonucleotide complementary to nt 1558 to nt 1584 were used to amplify a 1591 bp DNA encoding the entire human tPA prepro polypeptide.

The full-length DNA was used as a template for a subsequent PCR reaction to amplify a 599 bp DNA encoding the a portion of the signal peptide-finger-growth factor-first Kringle domains of tPA, and which also to introduce an in-frame amber stop codon (i. e., UGA) at amino acid codon 180 of mature tPA sequence. Concurrently, a Sall restriction endonuclease site and a mutation substituting a Pro for an Arg at position-6 were introduced upstream of the first Ser codon of mature tPA and a BamHl site was introduced downstream of newly introduced translational stop codon to allow for convenient subcloning of the amplifie product. The substitution of Pro for Arg at amino acid residue

position-6 introduces a proteolytic cleavage site for thrombin in the linker sequence (i. e., Phe-Pro-Arg-Gly at positions-7 to-4).

The Sall site and the amino acid substitution were introduced at nt 76 to 81 and 91 and 92 (nt-30 to-25 and-15 and-14, respectively, relative to the first nucleotide of mature tPA) during the PCR reaction by synthesizing an oligonucleotide primer corresponding to nt 72 to nt 111 containing nucleotide substitutions at six positions in the tPA sequence: nt 76 (A to G), nt 79 (C to G), nt 81 (T to C), nt 91 (A to C) and nt 92 (G to C). The BamHl site at nt 652 to nt 657 and translational stop codon at amino acid codon 180 (nt 642-644) were introduced by synthesizing an oligonucleotide primer complementary to nt 623 to 661 containing nucleotide substitutions at three positions in the tPA sequence: nt 644 (C to A), nt 655 (A to T) and nt 657 (G to C).

The amplifie PCR fragment was incubated with Sall and BamHl and subjected to agarose gel electrophoresis according to the standard procedures (Sambrook et al. (1989) Molecular Cloning, 2nd ed., Cold Spring Harbor Laboratory Press, New York). The 585 bp DNA was isolated from gel by electroelution and recovered by precipitation with ethanol.

3. Construction of a vector for expressing human FGFR1-tPA fusion protein The isolated Sall to BamHl fragment encoding the portion of human tPA was ligated into the Sall and BamHl sites of pUC18 to generate plasmid HTPA3/4-pUC18. HTPA3/4-pUC18 was then digested with Hindlll and Sall into which the isolated Hindlll to Sall FGFR1- encoding fragment was inserted. The plasmid carrying the FGFR1-tPA chimeric DNA was digested with Hindlll and BamHl, subjected to agarose gel electrophoresis and the 1,426 bp DNA fragment was excised from the gel and isolated as described above. The resulting DNA encodes a

472 amino acid peptide comprised of amino acids 1-284 of human FGFR1, a 10 amino acid linker sequence VDARFPRGAR, derived from the human tPA signal peptide, and amino acids 1-178 from human tPA. The resulting DNA encoding the FGFR1-tPA fusion protein is shown in SEQ ID No: 1 and the deduced amino acid is shown in SEQ ID No: 2.

The DNA of SEQ ID No. 1 was digested with Hindlil to BamHl and the 1,434 bp fragment (nt 2-1435 of SEQ ID No: 1) was isolated and ligated into the mammalian expression vector pK4K for recombinant expression of the FGFR1-tPA fusion protein (Niidome et a/. Biochem.

Biophys. Res. Commun. The plasmid pK4K is a pBR322-based vector that has unique Hindlil and BamHl sites for directional cloning of heterologous DNAs whose expression is under the control of the SV40 early promoter. This plasmid also contains the?- lactamase and DHFR genes for use as selectable markers in prokaryotes and eukaryotic organisms, respectively.

4. Expression of FGFR1-tPA chimeric protein in mammalian cells Baby hamster kidney cells (BHK cells; Waechter, D. E., et a/. Proc.

Natl. Acad. Sci., USA 1982,79,1106) were transfected with 5 ug of the FGFR1-tPA-containing expression plasmid using the CellPhect calcium phosphate method according to manufacturer's instructions (Pharmacia, Sweden). Transfectants were selected for the presence of the DHFR gene by selecting resistance to methotrexate and maintained in Dulbecco's Eagle medium containing 10% fetal bovine serum and 250 nM methotrexate.

Upon expression, the recombinant FGFR1-tPA fusion protein is secreted into the surrounding culture medium. Recombinant FGFR1-tPA fusion protein expression in BHK cells was monitored by sandwich enzyme-linked immunosorbent assays (sandwich ELISAs). A mouse IgG

monoclonal antibody specific for human tPA, designated 14-6, was used as the capture antibody and a polyclonal, rabbit anti-IgG antibody conjugated to horseradish peroxidase was used as the secondary-labeled antibody.

5. Purification of FGFR1-tPA chimeric protein The recombinant FGFR1-tPA fusion protein was purified from conditioned medium of BHK-expressing cells by affinity chromatography.

Transfected cells were grown as described above and the condition medium was harvested. The osmolarity of the conditioned medium was adjusted to a final concentration of 0.5 M NaCI by the addition of solid NaCI. The sample was applied onto a column of Cellulofine (Seikagaku Kogyo, Tokyo, Japan) conjugated with anti-tPA 14-6 monoclonal antibody previously equilibrated in column buffer (50 mM Tris-HCI, pH 7.5, and 0.5 M NaCI). The column was then washed with 10 column volumes of column buffer and bound fusion protein was eluted from the column by the addition of 0.2 M glycine-HCI, pH 2.5. Fractions (0.5 ml) were collecte into a tube containing 0.5 ml of 1 M Tris-HCI, pH 8.0 to neutralize the acidic eluate. Eluted fractions were monitored for the presence of FGFR1-tPA protein by measuring the absorbance of each fraction at 280 nm. The FGFR1-tPA-containing fractions were dialyzed against PBS and concentrated to a final concentration of 1.5-2.0 mg/ml using Centriprep filters (AMICON).

6. Analysis of bFGF-FGFR1 interaction The soluble, recombinant FGFR1-tPA fusion protein was immobilized to a solid support by attachment to the surface of the wells of an enzyme-linked immunosorbent assay plate (High binding plates, COSTAR). A 0.1 ml aliquot of a 10Ng/ml solution of rFGFR1-tPA in PBS was added and the plate was incubated for approximately 16 hr at 4 °C.

Unbound fusion protein was removed by washing three times with an equal volume of cold PBS.

To each well, a 0.1 ml aliquot of blocking buffer (25 mM HEPES, pH 7.5,100 mM NaCI and 0.5% gelatin) was added, and the samples incubated for 1 hr at ambient temperature to prevent non-specific binding of reagents. The wells were washed three times with binding buffer (25 mM HEPES, pH 7.5,100 mM NaCI and 0.3% gelatin) followed by addition of 0.1 mi of binding buffer supplemented with 2 Ng/ml heparin and a range of 1-20ng/ml of labeled'251-bFGF (800-1200Ci/mmol; Amersham, Arlington Heights, IL) and incubated in the absence or presence of 2.5 ug/ml unlabeled bFGF or a test compound for 3 hr at ambient temperature. The buffer was removed by aspiration and the wells were washed twice each with PBS and a solution of 25 mM HEPES, pH 7.5, containing 2 M NaCI. Bound bFGF was dissociated from the immobilized fusion protein by the addition of two aliquots of a solution of 25 mM sodium acetate, pH 4.0, containing 2 M NaCI. The two sodium acetate washes were combined and the amount of radioactivity present was determined using a gamma counter.

The amount of bound radiolabeled bFGF in each well was calculated and the specificity of bFGF binding was analyzed according to Scatchard (Scatchard Ann. N. Y. Acad. Sci. From this <BR> <BR> <BR> <BR> analysis, a 280 pM dissociation constant (KD) for the binding of bFGF to the recombinant FGFR1-tPA fusion protein of was calculated. This value <BR> <BR> <BR> <BR> correlates well with 130 pM KD value reported for bFGF binding to native<BR> <BR> <BR> <BR> <BR> <BR> <BR> FGFR1 receptors expressed in smooth muscle cells (Saltis et al.

Atherosclerosis 1995,118,77-87).

B. Membrane-bound FGF receptor assays 1. Competitive inhibition of FGF binding The rabbit aortic smooth muscle cell line, Rb-1, expresses high and low affinity FGF receptors (e. q., see Nachtigal et a/. In Vitro Cell. & Develop. Bill. 1989,25,892-897). Compounds of formula I or II that have FGF antagonist activity were and can be identified by their ability to compete with'251-bFGF for binding to the FGF receptors expressed on <BR> <BR> <BR> <BR> cell surface of such cells (see eq., see, Moscatelli et al. J. Cell. Phvsiol.

1987,131,123-130).

Rb-1 cells were grown in 24-well plates to near-confluence in Dulbecco's modified Eagle's medium (DMEM; GIBCO BRL) supplemented with 10% fetal bovine serum, penicillin (100 unit/ml) and streptomycin (100 ug/ml). The culture medium was removed by aspiration and the cells were incubated in binding buffer (serum-free DMEM supplemented with 20 mM HEPES (pH 7.5) and 0.1 % BSA) containing 0.2 ng/ml recombinant human 1251-bFGF (800-1200Ci/mmol; Amersham, Arlington Heights, IL) and varying concentrations of test compound, for 2 hr at ambient temperature. The nonspecific binding of iodinated bFGF to Rb-1 cells was estimated in parallel reactions performed in the presence of an excess of unlabeled bFGF.

The cells were washed twice with cold phosphate-buffered saline (PBS) and the bFGF bound to low affinity heparin sulfate proteoglycan (HSPG) receptors was dissociated by the addition to each well of a 1 ml solution of 25 mM HEPES (pH 7.5) containing 2 M NaCI. Following removal of the low affinity sample, the bFGF bound to high affinity FGF receptors was dissociated by the addition to each well of a 1 ml solution of 25 mM sodium acetate (pH 4.0) containing 2 M NaCI. A 1 ml aliquot from each well was transferred to a polypropylene tube and the amount

of radioactivity present in the high affinity samples was determined using a gamma counter.

2. Competitive inhibition of EGF binding The specificity of identified FGF antagonists was examined by measuring the ability of compounds to inhibit the binding of epidermal growth factor (EGF) to the surface of Rb-1 cells. Rb-1 cells were grown as described above and incubated in binding buffer containing 2 ng/ml of '251-EGF (> 750Ci/mmol; Amersham) under similar conditions. Non- specific binding of radiolabeled EGF was estimated in parallel reactions performed in an excess of unlabeled EGF.

After washing the cells twice with cold PBS, specifically bound EGF was dissociated from the cells by addition of a solution of 0.1 % Triton-X-100 and 5 min incubation at ambient temperature. The amount of radioactivity in each supernatant was measured using a gamma counter.

C. Inhibition of 3H-thymidine incorporation The incorporation of radiolabeled nucleotides into newly synthesized cellular DNA may be used as an indicator of cell proliferation.

SMCs, such as rat aortic SMCs, incorporate tritiated thymidine into DNA upon stimulation with bFGF or PDGF.

The effectiveness of compounds of formulae I or II as FGF antagonists was and can be assessed by measuring the inhibition of tritiated thymidine incorporation into the DNA of cultured SMCs incubated in the presence of bFGF, PDGF or EGF. An inoculum of approximately 2 X 104 rat aortic SMCs was added to a plurality of wells and the cells cultured for three days as described in EXAMPLE 1 B (i). The cells were washed twice with serum-free medium (DMEM supplemented with 0.1 % BSA, 5, ug/ml transferrin, penicillin (100 unit/ml) and

streptomycin (100 ug/ml)) and cultured for an addition three days in serum-free DMEM medium.

After washing twice in serum-free DMEM medium, the follow was added to each well: 400 NI of serum-free DMEM, 50, ul of 3 ng/ml bFGF in DMEM and 50, ul of known concentration test compound in DMEM 1.0% DMSO for 23 hr at 37° C. To each well, 10, ul of tritiated thymidine (3H-thymidine, 50, uCi/ml) was added and cells were incubated for 1 hr (37 °C). The medium was removed and the cells were washed twice with cold PBS. An 500, ul aliquot of a cold 10% TCA solution was added to each well and the cells incubated at 4° C overnight. After washing three times in cold PBS, the cells were incubated in 500, ul of 0.5 N NaOH for 30 min and the pH of the sample was neutralized by the addition of an equal volume of 0.5 N HCI. The amount of radioactivity present the supernatant of each well was determined using a liquid scintillation counter.

D. Results The percent inhibition of bFGF for each of the compounds described in detail above has been measured. Almost all of the compounds exhibited some inhibition of bFGF at concentrations of less than 500//M. Many of the compounds exhibited some inhibition of bFGF at concentrations of less than 300, uM. Several of these compounds exhibited some inhibition of bFGF at concentrations of less than 30, uM, while a few had measured IC50 values of less than 30, uM.

Since modifications will be apparent to those of skill in this art, it is intended that this invention be limited only by the scope of the appended claims.

SEQUENCE LISTING (1) GENERAL INFORMATION (i) APPLICANT: (A) NAME: Eisai, Ltd.

(B) STREET: 112-88 4-6-10 Koishikawa (C) CITY: Bunkyo-ku Tokyo (D) STATE: (E) COUNTRY: Japan (F) POSTAL CODE (ZIP): (i) INVENTOR: (A) NAME: Ventkatachalapathi Yalamoori (B) STREET: 12246 Summer Breeze Lane (C) CITY: San Diego (D) STATE: California (E) COUNTRY: USA (F) POSTAL CODE (ZIP): 92128 (i) INVENTOR: (A) NAME: Kalyanaraman Ramnarayan (B) STREET: 11674 Springside Rd.

(C) CITY: San Diego (D) STATE: California (E) COUNTRY: USA (F) POSTAL CODE (ZIP): 92128 (i) INVENTOR: (A) NAME: Laura Schove (B) STREET: 1240 Greenlake Drive (C) CITY: Cardiff (D) STATE: California (E) COUNTRY: USA (F) POSTAL CODE (ZIP): 92007 (i) INVENTOR: (A) NAME: Vitukudi Narayanaiyengar Balaji (B) STREET: No. 3 Type 4 CPRI (C) CITY: Bangalore (D) STATE: (E) COUNTRY: India (F) POSTAL CODE (ZIP): 560 012 (i) INVENTOR: (A) NAME: Ming Fai Chan (B) STREET: 1642 Orchard Wood Road (C) CITY: Encinitas (D) STATE: California (E) COUNTRY: USA (F) POSTAL CODE (ZIP) : 92024 (ii) TITLE OF THE INVENTION: ARGININE PEPTIDE ANALOGS USEFUL AS FIBROBLAST GROWTH FACTOR ANTAGONISTS (iii) NUMBER OF SEQUENCES: 2 (iv) CORRESPONDENCE ADDRESS: (A) ADDRESSEE: Heller Ehrman White & McAuliffe (B) STREET: 4250 Executive Square, 7th Floor (C) CITY: La Jolla (D) STATE: California (E) COUNTRY: US (F) ZIP: 92037 (v) COMPUTER READABLE FORM: (A) MEDIUM TYPE: Diskette (B) COMPUTER: IBM Compatible (C) OPERATING SYSTEM: DOS (D) SOFTWARE: FastSEQ Version 1.5 (vi) CURRENT APPLICATION DATA: (A) APPLICATION NUMBER: (B) FILING DATE: herewith (C) CLASSIFICATION: (vii) PRIOR APPLICATION DATA: (A) APPLICATION NUMBER: 09/058,002 (B) FILING DATE: 09-APR-98 (viii) ATTORNEY/AGENT INFORMATION: (A) NAME: Seidman, Stephanie L.

(B) REGISTRATION NUMBER: 33,779 (C) REFERENCE/DOCKET NUMBER: 24732-1201PC (ix) TELECOMMUNICATION INFORMATION: (A) TELEPHONE: (619) 450-8400 (B) TELEFAX: (619) 587-5360 (2) INFORMATION FOR SEQ ID NO: 1: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 1440 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE: (ix) FEATURE: (A) NAME/KEY: Coding Sequence (B) LOCATION: 9... 1427 (D) OTHER INFORMATION: (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 1: AAGCTTGG ATG TGG AGC TGG AAG TGC CTC CTC TTC TGG GCT GTG CTG GTC 50 Met Trp Ser Trp Lys Cys Leu Leu Phe Trp Ala Val Leu Val 1 5 10 ACA GCA ACA CTC TGC ACC GCT AGG CCG TCC CCG ACC TTG CCT GAA CAA 98 Thr Ala Thr Leu Cys Thr Ala Arg Pro Ser Pro Thr Leu Pro Glu Gln 15 20 25 30 GAT GCT CTC CCC TCC TCG GAG GAT GAT GAT GAT GAT GAT GAC TCC TCT 146 Asp Ala Leu Pro Ser Ser Glu Asp Asp Asp Asp Asp Asp Asp Ser Ser 35 40 45 TCA GAG GAG AAA GAA ACA GAT AAC ACC AAA CCA AAC CCC GTA GCT CCA 194 Ser Glu Glu Lys Glu Thr Asp Asn Thr Lys Pro Asn Pro Val Ala Pro 50 55 60 TAT TGG ACA TCC CCA GAA AAG ATG GAA AAG AAA TTG CAT GCA GTG CCG 242 Tyr Trp Thr Ser Pro Glu Lys Met Glu Lys Lys Leu His Ala Val Pro 65 70 75 GCT GCC AAG ACA GTG AAG TTC AAA TGC CCT TCC AGT GGG ACC CCA AAC 290 Ala Ala Lys Thr Val Lys Phe Lys Cys Pro Ser Ser Gly Thr Pro Asn 80 85 90 CCC ACA CTG CGC TGG TTG AAA AAT GGC AAA GAA TTC AAA CCT GAC CAC 338 Pro Thr Leu Arg Trp Leu Lys Asn Gly Lys Glu Phe Lys Pro Asp His 95 100 105 110 AGA ATT GGA GGC TAC AAG GTC CGT TAT GCC ACC TGG AGC ATC ATA ATG 386 Arg Ile Gly Gly Tyr Lys Val Arg Tyr Ala Thr Trp Ser Ile Ile Met 115 120 125 GAC TCT GTG GTG CCC TCT GAC AAG GGC AAC TAC ACC TGC ATT GTG GAG 434 Asp Ser Val Val Pro Ser Asp Lys Gly Asn Tyr Thr Cys Ile Val Glu 130 135 140 AAT GAG TAC GGC AGC ATC AAC CAC ACA TAC CAG CTG GAT GTC GTG GAG 482 Asn Glu Tyr Gly Ser Ile Asn His Thr Tyr Gln Leu Asp Val Val Glu 145 150 155 CGG TCC CCT CAC CGG CCC ATC CTG CAA GCA GGG TTG CCC GCC AAC AAA 530 Arg Ser Pro His Arg Pro Ile Leu Gln Ala Gly Leu Pro Ala Asn Lys 160 165 170 ACA GTG GCC CTG GGT AGC AAC GTG GAG TTC ATG TGT AAG GTG TAC AGT 578 Thr Val Ala Leu Gly Ser Asn Val Glu Phe Met Cys Lys Val Tyr Ser 175 180 185 190 GAC CCG CAG CCG CAC ATC CAG TGG CTA AAG CAC ATC GAG GTG AAT GGG 626 Asp Pro Gln Pro His Ile Gln Trp Leu Lys His Ile Glu Val Asn Gly 195 200 205 AGC AAG ATT GGC CCA GAC AAC CTG CCT TAT GTC CAG ATC TTG AAG ACT 674 Ser Lys Ile Gly Pro Asp Asn Leu Pro Tyr Val Gln Ile Leu Lys Thr 210 215 220 GCT GGA GTT AAT ACC ACC GAC AAA GAG ATG GAC GTG CTT CAC TTA AGA 722 Ala Gly Val Asn Thr Thr Asp Lys Glu Met Asp Val Leu His Leu Arg 225 230 235 AAT GTC TCC TTT GAG GAC GCA GGG GAG TAT ACG TGC TTG GCG GGT AAC 770 Asn Val Ser Phe Glu Asp Ala Gly Glu Tyr Thr Cys Leu Ala Gly Asn 240 245 250 TCT ATC GGA CTC TCC CAT CAC TCT GCA TGG TTG ACC GTT CTG GAA GCC 818 Ser Ile Gly Leu Ser His His Ser Ala Trp Leu Thr Val Leu Glu Ala 255 260 265 270 CTG GAA GAG AGG CCG GCA GTG ATG ACC TCG CCC CTG TAC GTC GAC GCC 866 Leu Glu Glu Arg Pro Ala Val Met Thr Ser Pro Leu Tyr Val Asp Ala 275 280 285 CGA TTC CCA AGA GGA GCC AGA TCT TAC CAA GTG ATC TGC AGA GAT GAA 914 Arg Phe Pro Arg Gly Ala Arg Ser Tyr Gln Val Ile Cys Arg Asp Glu 290 295 300 AAA ACG CAG ATG ATA TAC CAG CAA CAT CAG TCA TGG CTG CGC CCT GTG 962 Lys Thr Gln Met Ile Tyr Gln Gln His Gln Ser Trp Leu Arg Pro Val 305 310 315 CTC AGA AGC AAC CGG GTG GAA TAT TGC TGG TGC AAC AGT GGC AGG GCA 1010 Leu Arg Ser Asn Arg Val Glu Tyr Cys Trp Cys Asn Ser Gly Arg Ala 320 325 330 CAG TGC CAC TCA GTG CCT GTC AAA AGT TGC AGC GAG CCA AGG TGT TTC 1058 Gln Cys His Ser Val Pro Val Lys Ser Cys Ser Glu Pro Arg Cys Phe 335 340 345 350 AAC GGG GGC ACC TGC CAG CAG GCC CTG TAC TTC TCA GAT TTC GTG TGC 1106 Asn Gly Gly Thr Cys Gln Gln Ala Leu Tyr Phe Ser Asp Phe Val Cys 355 360 365 CAG TGC CCC GAA GGA TTT GCT GGG AAG TGC TGT GAA ATA GAT ACC AGG 1154 Gln Cys Pro Glu Gly Phe Ala Gly Lys Cys Cys Glu Ile Asp Thr Arg 370 375 380 GCC ACG TGC TAC GAG GAC CAG GGC ATC AGC TAC AGG GGC ACG TGG AGC 1202 Ala Thr Cys Tyr Glu Asp Gln Gly Ile Ser Tyr Arg Gly Thr Trp Ser 385 390 395 ACA GCG GAG AGT GGC GCC GAG TGC ACC AAC TGG AAC AGC AGC GCG TTG 1250 Thr Ala Glu Ser Gly Ala Glu Cys Thr Asn Trp Asn Ser Ser Ala Leu 400 405 410 GCC CAG AAG CCC TAC AGC GGG CGG AGG CCA GAC GCC ATC AGG CTG GGC 1298 Ala Gln Lys Pro Tyr Ser Gly Arg Arg Pro Asp Ala Ile Arg Leu Gly 415 420 425 430 CTG GGG AAC CAC AAC TAC TGC AGA AAC CCA GAT CGA GAC TCA AAG CCC 1346 Leu Gly Asn His Asn Tyr Cys Arg Asn Pro Asp Arg Asp Ser Lys Pro 435 440 445 TGG TGC TAC GTC TTT AAG GCG GGG AAG TAC AGC TCA GAG TTC TGC AGC 1394 Trp Cys Tyr Val Phe Lys Ala Gly Lys Tyr Ser Ser Glu Phe Cys Ser 450 455 460 ACC CCT GCC TGC TCT GAG GGA AAC AGT GAC TGA TACTTTGGGA TCC 1440 Thr Pro Ala Cys Ser Glu Gly Asn Ser Asp * 465 470 (2) INFORMATION FOR SEQ ID NO: 2: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 472 amino acids (B) TYPE: amino acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: internal (vi) ORIGINAL SOURCE: (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 2: Met Trp Ser Trp Lys Cys Leu Leu Phe Trp Ala Val Leu Val Thr Ala 1 5 10 15 Thr Leu Cys Thr Ala Arg Pro Ser Pro Thr Leu Pro Glu Gln Asp Ala 20 25 30 Leu Pro Ser Ser Glu Asp Asp Asp Asp Asp Asp Asp Ser Ser Ser Glu 35 40 45 Glu Lys Glu Thr Asp Asn Thr Lys Pro Asn Pro Val Ala Pro Tyr Trp 50 55 60 Thr Ser Pro Glu Lys Met Glu Lys Lys Leu His Ala Val Pro Ala Ala 65 70 75 80 Lys Thr Val Lys Phe Lys Cys Pro Ser Ser Gly Thr Pro Asn Pro Thr 85 90 95 Leu Arg Trp Leu Lys Asn Gly Lys Glu Phe Lys Pro Asp His Arg Ile 100 105 110 Gly Gly Tyr Lys Val Arg Tyr Ala Thr Trp Ser Ile Ile Met Asp Ser 115 120 125 Val Val Pro Ser Asp Lys Gly Asn Tyr Thr Cys Ile Val Glu Asn Glu 130 135 140 Tyr Gly Ser Ile Asn His Thr Tyr Gln Leu Asp Val Val Glu Arg Ser 145 150 155 160 Pro His Arg Pro Ile Leu Gln Ala Gly Leu Pro Ala Asn Lys Thr Val 165 170 175 Ala Leu Gly Ser Asn Val Glu Phe Met Cys Lys Val Tyr Ser Asp Pro 180 185 190 Gln Pro His Ile Gln Trp Leu Lys His Ile Glu Val Asn Gly Ser Lys 195 200 205 Ile Gly Pro Asp Asn Leu Pro Tyr Val Gln Ile Leu Lys Thr Ala Gly 210 215 220 Val Asn Thr Thr Asp Lys Glu Met Asp Val Leu His Leu Arg Asn Val 225 230 235 240 Ser Phe Glu Asp Ala Gly Glu Tyr Thr Cys Leu Ala Gly Asn Ser Ile 245 250 255 Gly Leu Ser His His Ser Ala Trp Leu Thr Val Leu Glu Ala Leu Glu 260 265 270 Glu Arg Pro Ala Val Met Thr Ser Pro Leu Tyr Val Asp Ala Arg Phe 275 280 285 Pro Arg Gly Ala Arg Ser Tyr Gln Val Ile Cys Arg Asp Glu Lys Thr 290 295 300 Gln Met Ile Tyr Gln Gln His Gln Ser Trp Leu Arg Pro Val Leu Arg 305 310 315 320 Ser Asn Arg Val Glu Tyr Cys Trp Cys Asn Ser Gly Arg Ala Gln Cys 325 330 335 His Ser Val Pro Val Lys Ser Cys Ser Glu Pro Arg Cys Phe Asn Gly 340 345 350 Gly Thr Cys Gln Gln Ala Leu Tyr Phe Ser Asp Phe Val Cys Gln Cys 355 360 365 Pro Glu Gly Phe Ala Gly Lys Cys Cys Glu Ile Asp Thr Arg Ala Thr 370 375 380 Cys Tyr Glu Asp Gln Gly Ile Ser Tyr Arg Gly Thr Trp Ser Thr Ala 385 390 395 400 Glu Ser Gly Ala Glu Cys Thr Asn Trp Asn Ser Ser Ala Leu Ala Gln 405 410 415 Lys Pro Tyr Ser Gly Arg Arg Pro Asp Ala Ile Arg Leu Gly Leu Gly 420 425 430 Asn His Asn Tyr Cys Arg Asn Pro Asp Arg Asp Ser Lys Pro Trp Cys 435 440 445 Tyr Val Phe Lys Ala Gly Lys Tyr Ser Ser Glu Phe Cys Ser Thr Pro 450 455 460 Ala Cys Ser Glu Gly Asn Ser Asp 465 470