Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NOVEL ANGIOTENSIN RECEPTOR MODULATORS AND THEIR USES
Document Type and Number:
WIPO Patent Application WO/1999/063930
Kind Code:
A2
Abstract:
This invention relates to novel multibinding compounds that bind to angiotensin (AT) receptors and modulate their activity. The compounds of this invention comprise 2-10 AT receptor ligands covalently connected by a linker or linkers, wherein the ligands in their monovalent (i.e., unlinked) state bind to one or more types of AT receptors. The manner of linking the ligands together is such that the multibinding agents thus formed demonstrate an increased biologic and/or therapeutic effect as compared to the same number of unlinked ligands made available for binding to AT receptors. The invention also relates to methods of using such compounds and to methods of preparing them. The compounds of this invention are particularly useful for treating diseases and conditions of mammals that are mediated by AT receptors. Accordingly, this invention also relates to pharmaceutical compositions comprising a pharmaceutically acceptable excipient and an effective amount of a compound of this invention.

Inventors:
MARQUESS DANIEL (US)
THOMAS G ROGER (US)
GRIFFIN JOHN H (US)
Application Number:
PCT/US1999/011805
Publication Date:
December 16, 1999
Filing Date:
June 07, 1999
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ADVANCED MEDICINE INC (US)
MARQUESS DANIEL (US)
THOMAS G ROGER (US)
GRIFFIN JOHN H (US)
International Classes:
A61K31/13; A61K31/136; G01N33/50; A61K31/137; A61K31/155; A61K31/16; A61K31/18; A61K31/27; A61K31/40; A61K31/4015; A61K31/403; A61K31/404; A61K31/41; A61K31/4164; A61K31/4178; A61K31/4184; A61K31/435; A61K31/437; A61K31/44; A61K31/4402; A61K31/4427; A61K31/4439; A61K31/445; A61K31/454; A61K31/4545; A61K31/47; A61K31/4706; A61K31/4709; A61K31/485; A61K31/495; A61K31/496; A61K31/498; A61K31/506; A61K31/517; A61K31/519; A61K31/55; A61K31/551; A61K31/5517; A61K31/662; A61K38/00; A61K45/00; A61K47/48; A61P9/02; A61P9/04; A61P9/10; A61P9/12; A61P11/00; A61P11/06; A61P13/02; A61P13/08; A61P15/06; A61P17/04; A61P21/00; A61P25/00; A61P25/02; A61P25/04; A61P25/06; A61P25/08; A61P25/14; A61P25/18; A61P25/20; A61P25/22; A61P25/24; A61P25/28; A61P27/02; A61P27/16; A61P43/00; C07B61/00; C07C211/00; C07C213/00; C07C215/00; C07C215/34; C07C215/60; C07C217/00; C07C217/08; C07C217/10; C07C217/48; C07C217/58; C07C217/90; C07C237/08; C07C271/12; C07C311/37; C07C323/44; C07C323/62; C07D207/06; C07D207/08; C07D207/26; C07D209/08; C07D211/14; C07D211/18; C07D211/42; C07D211/52; C07D211/56; C07D211/60; C07D211/72; C07D211/84; C07D213/30; C07D213/38; C07D213/56; C07D213/74; C07D213/75; C07D213/80; C07D215/22; C07D215/48; C07D221/26; C07D221/28; C07D223/04; C07D235/18; C07D235/30; C07D239/95; C07D241/44; C07D249/12; C07D257/04; C07D263/32; C07D263/34; C07D265/32; C07D277/24; C07D277/28; C07D277/34; C07D295/02; C07D295/06; C07D295/08; C07D295/18; C07D295/20; C07D401/04; C07D401/12; C07D401/14; C07D403/02; C07D403/14; C07D413/06; C07D413/14; C07D417/12; C07D471/04; C07D471/08; C07D487/04; C07D495/04; C07D519/00; C07K1/04; C07K2/00; C07K4/00; C07K7/14; C07K16/28; C40B20/00; G01N33/15; G01N33/53; G01N33/543; G01N33/566; G01N33/94; G01N37/00; (IPC1-7): A61K/
Domestic Patent References:
WO1997035195A11997-09-25
Foreign References:
US4587046A1986-05-06
US4228237A1980-10-14
US5958703A1999-09-28
Other References:
WEXLER et al., "Nonpeptide Angiotensin II Receptor Antagonists: The Next Generation in Antihypertensive Therapy", J. MED. CHEM., 02 February 1996, Vol. 39, No. 3, pages 625-656, XP002922745.
SIEMENS et al., "Solubilization and Partial Characterization of Angiotensin II Receptors from Rat Brain", J. NEUROCHEMISTRY, August 1991, Vol. 57, No. 2, pages 690-700, XP002922744.
MIDDLEMISS et al., "Benzofuran Based Angiotensin II Antagonists Related to GR117289: Enhancement of Potency In Vitro and Oral Activity, BIOORG. MED. CHEM. LETT., April 1993, Vol. 3, No. 4, pages 589-594, XP002922743.
SHUKER et al., "Discovering High-Affinity Ligands for Proteins: SAR by NMR", SCIENCE, 29 November 1996, Vol. 274, pages 1531-1534, XP002922742.
See also references of EP 1085887A2
Attorney, Agent or Firm:
Swiss, Gerald F. (Doane Swecker & Mathi, L.L.P. P.O. Box 1404 Alexandria VA, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:
1. A multibinding compound comprising 2 to 10 ligands which may be the same or different and which are covalently attached to a linker or linkers, which may be the same or different, each of said ligands comprising a ligand domain capable of binding to an angiotensin receptor.
2. The multibinding compound of Claim 1 wherein said ligand is selected from the group consisting of Losartan, E3174, Eprosartan, Candesartan, L158,809, Irbesartan, Valsartan, Tasosartan, Ripisartan, Telmesartan, Zolasartan, PD 123,319, L163,958, EXP801, L162,313 andPD123,177.
3. The multibinding compound of Claim 1 which has 2 ligands.
4. A multibinding compound represented by Formula I: (L)p(X)q I where each L is a ligand that may be the same or different at each occurrence; X is a linker that may be the same or different at each occurrence; p is an integer of from 2 to 10; and q is an integer of from 1 to 20; wherein each of said ligands comprises a ligand domain capable of binding to an angiotensin receptor.
5. The multibinding compound of Claim 4, wherein q is less than p.
6. The multibinding compound of Claim 4 wherein said ligand is selected from the group consisting of Losartan, E3174, Eprosartan, Candesartan, L158,809, Irbesartan, Valsartan, Tasosartan, Ripisartan, Telmesartan, Zolasartan, PD 123, 319, L163,958, EXP801, L162,313 and PD123,177.
7. The multibinding compound of Claim 4 where p is 2 and q is 1.
8. A pharmaceutical composition comprising a phannaceutically acceptable excipient and a therapeutically effective amount of one or more multibinding compounds, or pharmaceutically acceptable salts thereof, comprising 2 to 10 ligands which may be the same or different and which are covalently attached to a linker or linkers, which may be the same or different, each of said ligands comprising a ligand domain capable of binding to an angiotensin receptor of a cell mediating mammalian diseases or conditions, thereby modulating the diseases or conditions.
9. The pharmaceutical composition of Claim 8 wherein said ligand is selected from the group consisting of Losartan, E3174, Eprosartan, Candesartan, L158,809, Irbesartan, Valsartan, Tasosartan, Ripisartan, Telmesartan, Zolasartan, PD 123,319, L 163,958, EXP801, L162,313 and PD123,177.
10. The pharmaceutical composition of Claim 8 which has 2 ligands.
11. A pharmaceutical composition comprising a pharmaceutically acceptable excipient and a therapeutically effective amount of one or more multibinding compounds represented by Formula I: (L)p(X)qI and pharmaceutically acceptable salts thereof, where each L is a ligand that may be the same or different at each occurrence; X is a linker that may be the same or different at each occurrence; p is an integer of from 2 to 10; and q is an integer of from 1 to 20; wherein each of said ligands comprises a ligand domain capable of binding to an angiotensin receptor of a cell mediating mammalian diseases or conditions, thereby modulating the diseases or conditions.
12. The pharmaceutical composition of Claim 11 wherein said ligand is selected from the group consisting of Losartan, E3174, Eprosartan, Candesartan, L158,809, Irbesartan, Valsartan, Tasosartan, Ripisartan, Telmesartan, Zolasartan, PD 123,319, L 163,958, EXP801, L162,313 and PD123,177.
13. The pharmaceutical composition of Claim 11 which has 2 ligands.
14. A method for modulating the activity of an angiotensin receptor in a biologic tissue, which method comprises contacting a tissue having an angiotensin receptor with a multibinding compound, or a pharmaceutically acceptable salt thereof, under conditions sufficient to produce a change in the activity of the channel in said tissue, wherein the multibinding compound comprises 2 to 10 ligands which may be the same or different and which are covalently attached to a linker or linkers, which may be the same or different, each of said ligands comprising a ligand domain capable of binding to an angiotensin receptor.
15. The method of Claim 14 wherein said ligand is selected from the group consisting of Losartan, E3174, Eprosartan, Candesartan, L158,809, Irbesartan, Valsartan, Tasosartan, Ripisartan, Telmesartan, Zolasartan, PD 123,319, L163,958, EXP801, L162,313 and PD123,177.
16. The method compound of Claim 14 wherien the multibinding compound has 2 ligands.
17. A method for treating a disease or condition in a mammal resulting from an activity of an angiotensin receptor, which method comprises administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising a pharmaceutically acceptable excipient and one or more multibinding compounds, or pharmaceutically acceptable salts thereof, comprising 2 to 10 ligands which may be the same or different and which are covalently attached to a linker or linkers, which may be the same or different, each of said ligands comprising a ligand domain capable of binding to an angiotensin receptor of a cell mediating mammalian diseases or conditions.
18. The method of Claim 17 wherein said ligand is selected from the group consisting of Losartan, E3174, Eprosartan, Candesartan, L158,809, Irbesartan, Valsartan, Tasosartan, Ripisartan, Telmesartan, Zolasartan, PD 123,319, L163,958, EXP801, L162,313 and PD123,177.
19. The method of Claim 17 wherein the multibinding compound has 2 ligands.
20. A method for treating a disease or condition in a mammal resulting from an activity of an angiotensin receptor, which method comprises administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising a pharmaceutically acceptable excipient and one or more multibinding compounds represented by formula I: (L) P (X) i and pharmaceutically acceptable salts thereof, where each L is a ligand that may be the same or different at each occurrence; X is a linker that may be the same or different at each occurrence; p is an integer of from 2 to 10; and q is an integer of from 1 to 20; wherein each of said ligands comprises a ligand domain capable of binding to an angiotensin receptor of a cell mediating mammalian diseases or conditions.
21. The method of Claim 20 wherein said ligand is selected from the group consisting of Losartan, E3174, Eprosartan, Candesartan, L158,809, Irbesartan, Valsartan, Tasosartan, Ripisartan, Telmesartan, Zolasartan, PD 123,319, L163,958, EXP801, L162,313 and PD123,177.
22. The method of Claim 20 wherein the multibinding compound has 2 ligands.
23. A method for identifying multimeric ligand compounds possessing multibinding properties for angiotensin receptors, which method comprises: (a) identifying a ligand or a mixture of ligands wherein each ligand contains at least one reactive functionality; (b) identifying a library of linkers wherein each linker in said library comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand; (c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the ligand or mixture of ligands identified in (a) with the library of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands; and (d) assaying the multimeric ligand compounds produced in the library prepared in (c) above to identify multimeric ligand compounds possessing multibinding properties.
24. A method for identifying multimeric ligand compounds possessing muldbinding properties for angiotensin receptors, which method comprises: (a) identifying a library of ligands wherein each ligand contains at least one reactive functionality; (b) identifying a linker or mixture of linkers wherein each linker comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand; (c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the library of ligands identified in (a) with the linker or mixture of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands; and (d) assaying the multimeric ligand compounds produced in the library prepared in (c) above to identify multimeric ligand compounds possessing multibinding properties.
25. The method according to Claim 23 or 24 wherein the preparation of the multimeric ligand compound library is achieved by either the sequential or concurrent combination of the two or more stoichiometric equivalents of the ligands identified in (a) with the linkers identified in (b).
26. The method according to Claim 25 wherein the multimeric ligand compounds comprising the multimeric ligand compound library are dimeric.
27. The method according to Claim 26 wherein the dimeric ligand compounds comprising the dimeric ligand compound library are heterodimeric.
28. The method according to Claim 27 wherein the heterodimeric ligand compound library is prepared by sequential addition of a first and second ligand.
29. The method according to Claim 23 or 24 wherein, prior to procedure (d), each member of the multimeric ligand compound library is isolated from the library.
30. The method according to Claim 29 wherein each member of the library is isolated by preparative liquid chromatography mass spectrometry (LCMS).
31. The method according to Claim 23 or 24 wherein the linker or linkers employed are selected from the group comprising flexible linkers, rigid linkers, hydrophobic linkers, hydrophilic linkers, linkers of different geometry, acidic linkers, basic linkers, linkers of different polarization and amphiphilic linkers.
32. The method according to Claim 31 wherein the linkers comprise linkers of different chain length and/or having different complementary reactive groups.
33. The method according to Claim 32 wherein the linkers are selected to have different linker lengths ranging from about 2 to lOOÅ.
34. The method according to Claim 23 or 24 wherein the ligand or mixture of ligands is selected to have reactive functionality at different sites on said ligands.
35. The method according to Claim 34 wherein said reactive functionality is selected from the group consisting of carboxylic acids, carboxylic acid halides, carboxyl esters, amines, halides, pseudohalides, isocyanates, vinyl unsaturation, ketones, aldehydes, thiols, alcohols, anhydrides, boronates, and precursors thereof wherein the reactive functionality on the ligand is selected to be complementary to at least one of the reactive groups on the linker so that a covalent linkage can be formed between the linker and the ligand.
36. The method according to Claim 23 or Claim 24 wherein the multimeric ligand compound library comprises homomeric ligand compounds.
37. The method according to Claim 23 or Claim 24 wherein the multimeric ligand compound library comprises heteromeric ligand compounds.
38. A library of multimeric ligand compounds which may possess multivalent properties for angiotensin receptors, which library is prepared by the method comprising: (a) identifying a ligand or a mixture of ligands wherein each ligand contains at least one reactive functionality; (b) identifying a library of linkers wherein each linker in said library comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand; and (c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the ligand or mixture of ligands identified in (a) with the library of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands.
39. A library of multimeric ligand compounds which may possess multivalent properties for angiotensin receptors, which library is prepared by the method comprising: (a) identifying a library of ligands wherein each ligand contains at least one reactive functionality; (b) identifying a linker or mixture of linkers wherein each linker comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand; and (c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the library of ligands identified in (a) with the linker or mixture of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands.
40. The library according to Claim 38 or Claim 39 wherein the linker or linkers employed are selected from the group comprising flexible linkers, rigid linkers, hydrophobic linkers, hydrophilic linkers, linkers of different geometry, acidic linkers, basic linkers, linkers of different polarization and amphiphilic linkers.
41. The library according to Claim 40 wherein the linkers comprise linkers of different chain length and/or having different complementary reactive groups.
42. The library according to Claim 41 wherein the linkers are selected to have different linker lengths ranging from about 2 to 100A.
43. The library according to Claim 38 or 39 wherein the ligand or mixture of ligands is selected to have reactive functionality at different sites on said ligands.
44. The library according to Claim 43 wherein said reactive functionality is selected from the group consisting of carboxylic acids, carboxylic acid halides, carboxyl esters, amines, halides, pseudohalides, isocyanates, vinyl unsaturation, ketones, aldehydes, thiols, alcohols, anhydrides, boronates, and precursors thereof wherein the reactive functionality on the ligand is selected to be complementary to at least one of the reactive groups on the linker so that a covalent linkage can be formed between the linker and the ligand.
45. The library according to Claim 38 or Claim 39 wherein the multimeric ligand compound library comprises homomeric ligand compounds.
46. The library according to Claim 38 or Claim 39 wherein the multimeric ligand compound library comprises heteromeric ligand compounds.
47. An interative method for identifying multimeric ligand compounds possessing multibinding properties for angiotensin receptors, which method comprises: (a) preparing a first collection or iteration of multimeric compounds which is prepared by contacting at least two stoichiometric equivalents of the ligand or mixture of ligands which target a receptor with a linker or mixture of linkers wherein said ligand or mixture of ligands comprises at least one reactive functionality and said linker or mixture of linkers comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand wherein said contacting is conducted under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands; (b) assaying said first collection or iteration of multimeric compounds to assess which if any of said multimeric compounds possess multibinding properties; (c) repeating the process of (a) and (b) above until at least one multimeric compound is found to possess multibinding properties; (d) evaluating what molecular constraints imparted or are consistent with imparting multibinding properties to the multimeric compound or compounds found in the first iteration recited in (a) (c) above; (e) creating a second collection or iteration of multimeric compounds which elaborates upon the particular molecular constraints imparting multibinding properties to the multimeric compound or compounds found in said first iteration; evaluating what molecular constraints imparted or are consistent with imparting enhanced multibinding properties to the multimeric compound or compounds found in the second collection or iteration recited in (e) above; (g) optionally repeating steps (e) and (f) to further elaborate upon said molecular constraints.
48. The method according to Claim 47 wherein steps (e) and (f) are repeated from 250 times.
49. The method according to Claim 47 wherein steps (e) and (f) are repeated from 550 times.
50. The method of Claim 17 wherein said disease or condition is selected from the group consisting of hypertension, heart failure and renal disease.
Description:
NOVEL ANGIOTENSIN RECEPTOR MODULATORS AND THEIR USES CROSS-REFERENCE TO RELATED APPLICATIONS This application claims priority to U. S. Applications Serial Nos. 60/088,466 filed June 8,1998 and 60/092,938 filed July 15,1998, the entire contents of which are incorporated herein by reference.

BACKGROUND Field of-the Invention This invention relates to novel multibinding compounds that bind to angiotensin (AT) receptors and modulate their activity. The compounds of this invention comprise 2-10 AT receptor ligands covalently connected by a linker or linkers, wherein the ligands in their monovalent (i. e., unlinked) state bind to one or more types of AT receptors. The manner of linking the ligands together is such that the multibinding agents thus formed demonstrate an increased biologic and/or therapeutic effect as compared to the same number of unlinked ligands made available for binding to AT receptors. The invention also relates to methods of using such compounds and to methods of preparing them.

The compounds of this invention are particularly useful for treating diseases and conditions of mammals that are mediated by AT receptors. Accordingly, this invention also relates to pharmaceutical compositions comprising a pharmaceutically acceptable excipient and an effective amount of a compound of this invention.

References The following publications are cited in this application as superscript numbers:

References The following publications are cited in this application as superscript numbers: etal.,J.HumanHypertension,12,311-318,1998.1Csikos,T.

2Balmforth, A. J., et al., Biochem. Society Transactions, 25, 1041-1046, 1997.

3Probst, W. C., et al., DNA Cell Biol., 11,1-20,1992.

4Strader, C. D., et al., Ann. Rev. Biochem., 63,101-132,1994.

5Savarese, T. M., et al., Biochem. J., 283,1-19,1992.

'Stoll, M., et al., J. Clin. Invest., 95,651-657,1995.

7Meffert, S., et al., Mol. Cell Endocrinol., 122 (1), 59-67,1996.

'Gallinat, S., et al., Neurosci. Lett., 227 (1), 29-32,1997.

9Duncia, J. V., et al., J. Med. Chem., 33,1312-1329,1990.

°Duncia, J. V., et al., Med. Res. Rev., 12,149-191,1992.

Canni, D. J., et al., J. Med Chem., 34,2525-2547,1991. l2Carini, D. J., et al., J. Med. Chem., 33,1330-1336,1990.

"Burnier, M., et al., Fxp. Opin. Invest. Drugs, 6 (5), 489-500,1997.

'4Merlos, M., et al., Drugs of the Future, 22 (8), 850-855,1997.

15Markham, A., et al., Drugs, 54 (2), 299-311,1997.

10Gillis, J. C., et al., Drugs, 54 (6), 885-902,1997.

"Merles, M., et al., Drugs of tSie Future, 22 (10), 1079-1085,1997. t8McCleLlan, K. J., et al., Drugs, 55 (5) 713-718,1998.

19Casa, A., et al., Drugs of the Future, 22 (5), 481-491,1997.

20Goa, K., et al., Drugs, 51 (5), 820-845,1996.

2lChristen, Y., et al., Circulation, 83,1333-1342,1991.

22Dzau, V. J., J. Hypertension, 12, S1-5, 1994.

23Bergsa, D. A., et al., Cloning and Characterization of the Human Angiotensin II Type 1 Receptor, Biochem. Biophys. Res. Commun., 183,989-935,1992.

24Mukoyama, M., et al., Expression Cloning of a Type 2 Angiotensin II Receptor Reveals a Unique Class of Seven Transmembrane Receptors, J. Biol. Chem., 268 (33), 24539-24542,1993.

25Azizi, M., et al., Additive Effects of Combined Angiotensin Converting Enzyme Inhibition and Angiotensin II Antagonists on Blood Pressure and Renin Release in Sodium Depleted Normotensives, Circulation, 92,825-834,1995.

26Gansevort, R. T., et al., Is the Antiproteinuric Effect of Angiotensin Converting Enzyme Inhibitors Mediated by Interference in the Renin-Angiotensin System, Kidney Int., 45,861-867,1994.

27Inoue, Y., et al., A Review of Mutagenesis Studies on Angiotensin II Type 1 Receptor, the Three-Dimensional Receptor Model in Seacrch of the Agonist and Antagonist Binding Site, J. Hypertension, 15,703-714,1997.

28Hunyady, L., et al., The Ligand Binding Site of the AT1 Receptor, Trends in Pharmacol. Sci., 17,135-140,1996.

29Robertson, M. J., et al., Agonist-Antagonist Interactions at Angiotensin <BR> <BR> <BR> <BR> Receptors: Application of the Two State Receptor Model, Trends in Pharmacol. Sci., 15, 364-396,1994.

30Chiu, A. T., et al., Non-Peptide Angiotensin II Receptor Antagonists. VII.

Cellular and Biochemical Pharmacology of DuP 753, an OraUy Active Hypertensive Agent, J. Pharm. Exp. Ther., 252 (2) 711-718,1990.

3tWong, P. C., et al., Non-Peptide Angiotensin II Receptor Antagonists. IX.

Antihypertensive Activity in the Rat of DuP 753, an Orally Active Antihypertensive Agent., J. Pharm. Exp. Ther., 252 (2) 719-725,1990.

32Wong, P. C., et al., Non-Peptide Angiotensin II Receptor Antagonists. IX.

Antihypertensive Activity in the Rat of DuP 753, an Orally Active Antihypertensive Agent., J. Pharm. Exp. Ther., 252 (2) 726-732,1990.

33Rivero, R. A., et al., The synthesis of [3H]-losartan, [3H]-L-158,641 and [3H]- L-158,809., Bioorg. Med. Chem. Lett., 3 (4), 557-60,1993. <BR> <BR> <BR> <BR> <P> 34Whitebread, S., et al., Biochem. Biophys. Res. Comm., 163,284-291,1989. <BR> <BR> <BR> <BR> <BR> <BR> <P> 35Timmermans, P. B. M. W. M., et al., Pharmacol. Rev., 45,205-251,1993.

36Smith R. D., et al., Ann. Rev. Pharmacol. Toxicol., 32,135-165,1992. <BR> <BR> <BR> <BR> <P> 37Morton, J. J., et al., J. Vasc. Res., 29,264-269,1992. <BR> <BR> <BR> <BR> <BR> <BR> <P> 38Shen, Y. T., et al., Cardiovas. Res., 39,413-422,1998. <BR> <BR> <BR> <BR> <BR> <BR> <P> 39Krombach, R. S., et al., Cardiovas. Res., 38,631-645,1998.

40Shen, Y. T., Circulation, 94,1-139,1996.

41Poilock, D. M., et al., J. Pharmacol. Exp. Ther., 267,657-63,1993.

42Lafayette, R. A., et al., J. Clin. Invest., 90 (3), 766-771,1992.

43 Rernuzzi, A., et al., Exp. Nephrol., 4 (1), 19-25,1996.

44Maxfield, E. K., et al., Diabetologia, 36 (12), 1230-1237,1993.

45Wexler, R. R., et al., J. Med. Chem., 39 (3), 625-656,1996.

'Monnot, C., Biol. Chem., 271 (3), 1507-13,1996.

47DeGasparo, J., Recept. Res., 11 (1-4), 247-57,1991.

48Dzielak, D. J., Comparative Pharmacology of the Angiotensin II Receptor Antagonists, Exp. Opin. Invest. Drues., 7 (5), 741-51,1998.

49Merlos, M., et al., Irbesartan, Drugs of the Future, 22 (5): 481-491,1997.

50Miyazawa, et al., Atherosclerosis, 121 (2), 167-173,1996.

The disclosure of each of the above patents, patent applications and publications is incorporated herein by reference in its entirety to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference in its entirety.

State of the Art The renin-angiotensin system (RAS) is an enzymatic cascade that plays a major role in blood pressure regulation, renal function, fluid volume homeostasis, and electrolyte balance. Figure 1, graphically describes this system. Angiotensinogen, the parent compound of the RAS, is produced and secreted by the liver. Circulating angiotensinogen is cleaved by the protease renin to release an intermediate product, the decapeptide angiotensin I, which is subsequently processed by angiotensin converting enzyme (ACE) to the bioactive octapeptide angiotensin II.

Angiotensin II is the bioactive peptide which regulates blood pressure, fluid volume homeostasis. It also has pituitary hormone release effects. These effects are mediated by binding to and activating receptors located on various target organs including the brain, heart, vascular wall, adrenal gland, kidney, liver and reproductive organsl. It exerts its effects on blood pressure through mechanisms that include increasing salt and water absorption from the kidney. The amino acid sequence of angiotensin II is: [NlAspl-Arg2-Val3-Tyr4-I Vals I-His6-Pro7-PheS |Ilc5| There are two naturally occurring forms of angiotensin II, depending on the amino acid found at residue 5. In cattle and sheep, valine is at this position, while isoleucine is at residue 5 in humans, pigs and horses. There is only a slight difference in potency between these two forms of angiotensin II. Peptide agonists and antagonists of angiotensin II are known45.

At least two distinct subtypes of angiotensin II receptors, designated AT, and AT2, have been distinguished, both of which are members of the seven transmembrane superfamily of cell surface receptors and are of the G-protein-coupled receptor family2'S) w The ATl receptor, which has 359 amino acids, accounts for most of the known actions of angiotensin II, including vasoconstriction, enhanced noradrenergic transmission, sodium reabsorption, aldosterone release and vascular hypertrophy. The function of the AT2 receptor, which has 363 amino acids and only 32% homology with AT, receptor, is not fully understood, but recent studies show it to be involved in antiproliferative effects, apoptosis, tissue regeneration, foetal development and neuronal differentiation6-8. The AT2 receptor is upregulated after myocardial infarction, vascular injury or cardiac failure.

Although not as widely distributed as AT, receptors, which are extensively expressed in the majority of tissues, AT2 receptors are present in the adrenal gland, brain, ovary and uterus.

The AT1 receptor possesses multiple proximal binding domain which include N- terminal and extracellular loops involved in peptide binding as well as interhelix binding for small molecule antagonists. There is evidence that suggests that AT, receptors may be dimeric, since co-expression of two deficient mutants restored the original binding profile.

The effect of covalent dimer conjugates of angiotensin II on receptor affinity and activity has been studied in vitre. Results of these studies showed that there was not an increase in biological activity for the specific types of dimers synthesized in the rabbit aortic ring assay.

Blockade of the RAS is now recognized as an effective approach for the treatment of a number of disease states, including hypertension, congestive heart failure and renal disease. In the last 15 years the standard treatment of these diseases has consisted of blocking the RAS with small molecule ACE inhibitors. However, despite their clinical success, the use of ACE inhibitors is limited by their side effects, including dry cough (due to interference with kinin metabolism) and angioedema. This has led to the more specific approach of blocking the terminal step of the RAS-namely the angiotensin II receptors.

Combined therapy with ATI, receptor antagonists and ACE inhibitors has been explored in the clinical setting. Limited clinical studies have been carried out with combined losartan-captopril therapy in healthy, normotensive volunteers with mild sodium depletion. This combination had a significant additive effect in blood pressure reduction. 25 26 However, this may have the effect of introducing the ACE inhibitor cough profile to the ATl antagonist ligands.

Due to the clear link between the ATl receptor and the control of blood pressure, numerous AT, selective antagonists have been developed for clinical use, of which the biphenyl tetrazole losartan is the prototype 12,". These sartan drugs are also being evaluated for clinical efficacy against congestive heart failure and progressive renal disease in patients with diabetes or renal deficiency.

As shown in Table 1, AT receptor modulators may show activity with both AT and AT2 receptors, while some bind specifically to one or the other. In terms of their

pharmacological binding profiles these compounds may generally be classified as follows: the AT, receptor has a high affinity for the sartan drugs and a low affinity for PD 123177.

The AT2 receptor has a high affinity for PD 123177 but a low affinity for losartan. It-is of interest to note that angiotensin II shows no selectivity for the AT receptor subtypes. The clinical doses of some AT, antagonists in clinical use, along with their affinity, bioavailability and half-life are presented in Table 2.

As a class, the ATl receptor antagonists appear to be as effective as the ACE inhibitors in treating hypertension, without the dry-cough side effect. However, the currently known drugs which are AT, receptor antagonists have many disadvantages, including slow onset of action, high plasma protein binding, low to moderate oral <BR> <BR> <BR> bioavailability, dizziness, headache, and fatigue"-'O,'9. In addition, increased circulating levels of angiotensin II after AT, receptor blockade have concerned clinicians because of the possible consequences of concurrently stimulating the angiotensin AT2 receptor subtype <BR> <BR> and other unblocked angiotensin binding sites2t 22, Accordingly, it would be advantageous to discover novel compounds with a more favorable pharmacokinetic profile, greater receptor specificity and reduced side effects.

SIJMMARY OF 1lIE INVENTION This invention is directed to novel multibinding compounds that bind to angiotensin receptors in mammalian tissues and can be used to treat diseases and conditions mediated by such receptors.

This invention is also directed to general synthetic methods for generating large libraries of diverse multimeric compounds which multimeric compounds are candidates for possessing multibinding properties for angiotensin receptors. The diverse multimeric compound libraries provided by this invention are synthesized by combining a linker or

linkers with a ligand or ligands to provide for a library of multimeric compounds wherein the linker and ligand each have complementary functional groups permitting covalent linkage. The library of linkers is preferably selected to have diverse properties such as valency, linker length, linker geometry and rigidity, hydrophilicity or hydrophobicity, amphiphilicity, acidity, basicity and polarization. The library of ligands is preferably selected to have diverse attachment points on the same ligand, different functional groups at the same site of otherwise the same ligand, and the like.

This invention is also directed to libraries of diverse multimeric compounds which multimeric compounds are candidates for possessing multibinding properties. These libraries are prepared via the methods described above and permit the rapid and efficient evaluation of what molecular constraints impart multibinding properties to a ligand or a class of ligands targeting an angiotensin receptor.

Accordingly, in one of its composition aspects, this invention is directed to a multibinding compound and salts thereof comprising 2 to 10 ligands which may be the same or different and which are covalently attached to a linker or linkers, which may be the same or different, each of said ligands comprising a ligand domain capable of binding to an AT receptor.

The multibinding compounds of this invention are preferably represented by Formula I: (L) p (X) q I where each L is a ligand that may be the same or different at each occurrence; X is a linker that may be the same or different at each occurrence; p is an integer of from 2 to 10; and q

is an integer of from 1 to 20; wherein each of said ligands comprises a ligand domain capable of binding to an AT receptor or receptors. Preferably q is less than p.

Preferably, the binding of the multibinding compound to an AT receptor or receptors in a mammal modulates diseases and conditions mediated by the AT receptor or receptors.

In another of its composition aspects, this invention is directed to a pharmaceutical composition comprising a pharmaceutically acceptable excipient and a therapeutically effective amount of one or more multibinding compounds (or pharmaceutically acceptable salts thereof) comprising 2 to 10 ligands which may be the same or different and which are covalently attached to a linker or linkers, which may be the same or different, each of said ligands comprising a ligand domain capable of binding to an AT receptor of a cell mediating mammalian diseases or conditions, thereby modulating the diseases or conditions.

In still another of its composition aspects, this invention is directed to a pharmaceutical composition comprising a pharmaceutically acceptable excipient and a therapeutically effective amount of one or more multibinding compounds represented by formula I: (L)p(X)qI or pharmaceutically acceptable salts thereof, where each L is a ligand that may be the same or different at each occurrence; X is a linker that may be the same or different at each occurrence; p is an integer of from 2 to 10; and q is an integer of from 1 to 20; wherein each of said ligands comprises a ligand domain capable of binding to an AT receptor of a

cell mediating mammaiian diseases or conditions, thereby modulating the diseases or conditions. Preferably q is less than p.

In one of its method aspects, this invention is directed to a method for modulating the activity of an AT receptor in a biologic tissue, which method comprises contacting a tissue having an AT receptor with a multibinding compound (or pharmaceutically acceptable salts thereof) under conditions sufficient to produce a change in the activity of the receptor in said tissue, wherein the multibinding compound comprises 2 to 10 ligands which may be the same or different and which are covalently attached to a linker or linkers, which may be the same or different, each of said ligands comprising a ligand domain capable of binding to an AT receptor.

In another of its method aspects, this invention is directed to a method for treating a disease or condition in a mammal resulting from an activity of an AT receptor, which method comprises administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising a pharmaceutically acceptable excipient and one or more multibinding compounds (or pharmaceutically acceptable salts thereof) comprising 2 to 10 ligands which may be the same or different and which are covalently attached to a linker or linkers, which may be the same or different, each of said ligands comprising a ligand domain capable of binding to an AT receptor of a cell mediating mammalian diseases or conditions.

In yet another of its method aspects, this invention is directed to a method for treating a disease or condition in a mammal resulting from an activity of an AT receptor, which method comprises administering to said mammal a therapeutically effective amount of a pharmaceutical composition comprising a pharmaceutically acceptable excipient and one or more multibinding compounds represented by Formula I:

(L)p(X)qI and pharmaceutically acceptable salts thereof, where each L is a ligand that may be the same or different at each occurrence; X is a linker that may be the same or different at each occurrence; p is an integer of from 2 to 10; and q is an integer of from 1 to 20; wherein each of said ligands comprises a ligand domain capable of binding to an AT receptor of a cell mediating mammalian diseases or conditions. Preferably q is less thanp.

In a further aspect, this invention provides processes for preparing the multibinding agents of Formula I. This can be accomplished by combining p appropriately functionalized ligands with q complementary functionalized linkers under conditions where covalent bond formation between the ligands and linkers occur. Alternatively linking portions of p appropriately functionalized ligands to q complementary functionalized linkers and then completing the synthesis of the ligands in a subsequent step may be performed to prepare these compounds. Another method which may be used involves linldng p appropriately functionalized ligands to portions of the linker (s) and then completing the synthesis of the linker (s) in a subsequent step.

Coupling one or more of the appropriately functionalized ligand (s) to a complementary functionalized linker, and subsequently coupling one or more addition ligands to said linker or linkers may be done to prepare the claimed compounds. Coupling as described in the preceding sentence, wherein coupling of different appropriately functionalized linkers occurs simultaneously may also be used.

In one of its method aspects, this invention is directed to a method for identifying multimeric ligand compounds possessing multibinding properties for angiotensin receptors, which method comprises:

(a) identifying a ligand or a mixture of ligands wherein each ligand contains at least one reactive functionality; (b) identifying a library of linkers wherein each linker in said library comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand; (c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the ligand or mixture of ligands identified in (a) with the library of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands; and (d) assaying the multimeric ligand compounds produced in (c) above to identify multimeric ligand compounds possessing multibinding properties.

In another of its method aspects, this invention is directed to a method for identifying multimeric ligand compounds possessing multibinding properties for angiotensin receptors, which method comprises: (a) identifying a library of ligands wherein each ligand contains at least one reactive functionality; (b) identifying a linker or mixture of linkers wherein each linker comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand; (c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the library of ligands identified in (a) with the linker or mixture of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands; and (d) assaying the multimeric ligand compounds produced in (c) above to identify multimeric ligand compounds possessing multibinding properties.

The preparation of the multimeric ligand compound library is achieved by either the sequential or concurrent combination of the two or more stoichiometric equivalents of the ligands identified in (a) with the linkers identified in (b). Sequential addition is preferred when a mixture of different ligands is employed to ensure heterodimeric or multimeric compounds are prepared. Concurrent addition of the ligands occurs when at least a portion of the multimer comounds prepared are homomultimeric compounds.

The assay protocols recited in (d) can be conducted on the multimeric ligand compound library produced in (c) above, or preferably, each member of the library is isolated by preparative liquid chromatography mass spectrometry (LCMS).

In one of its composition aspects, this invention is directed to a library of multimeric ligand compounds which may possess multivalent properties for angiotensin receptors, which library is prepared by the method comprising: (a) identifying a ligand or a mixture of ligands wherein each ligand contains at least one reactive functionality; (b) identifying a library of linkers wherein each linker in said library comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand; and (c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the ligand or mixture of ligands identified in (a) with the library of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands.

In another of its composition aspects, this invention is directed to a library of multimeric ligand compounds which may possess multivalent properties for angiotensin receptors, which library is prepared by the method comprising:

(a) identifying a library of ligands wherein each ligand contains at least one reactive functionality; (b) identifying a linker or mixture of linkers wherein each linker comprises-at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand; and (c) preparing a multimeric ligand compound library by combining at least two stoichiometric equivalents of the library of ligands identified in (a) with the linker or mixture of linkers identified in (b) under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands.

In a preferred embodiment, the library of linkers employed in either the methods or the library aspects of this invention is selected from the group comprising flexible linkers, rigid linkers, hydrophobic linkers, hydrophilic linkers, linkers of different geometry, acidic linkers, basic linkers, linkers of different polarization and amphiphilic linkers. For example, in one embodiment, each of the linkers in the linker library may comprise linkers of different chain length and/or having different complementary reactive groups. Such linker lengths can preferably range from about 2 to 100A.

In another preferred embodiment, the angiotensin receptor ligand or mixture of ligands is selected to have reactive functionality at different sites on said ligands in order to provide for a range of orientations of said ligand on said multimeric ligand compounds.

Such reactive functionality includes, by way of example, carboxylic acids, carboxylic acid halides, carboxyl esters, amines, halides, isocyanates, vinyl unsaturation, ketones, aldehydes, thiols, alcohols, anhydrides, and precursors thereof. It is understood, of course, that the reactive functionality on the ligand is selected to be complementary to at least one of the reactive groups on the linker so that a covalent linkage can be formed between the linker and the ligand.

In other embodiments, the multimeric ligand compound is homomeric (i. e., each of the ligands is the same, although it may be attached at different points) or heterodimeric (i. e., at least one of the ligands is different from the other ligands).

In addition to the combinatorial methods described herein, this invention provides for an interative process for rationally evaluating what molecular constraints impart multibinding properties to a class of multimeric compounds or ligands targeting a receptor.

Specifically, this method aspect is directed to a method for identifying multimeric ligand compounds possessing multibinding properties for angiotensin receptors which method comprises: (a) preparing a first collection or iteration of multimeric compounds which is prepared by contacting at least two stoichiometric equivalents of the ligand or mixture of ligands which target a receptor with a linker or mixture of linkers wherein said ligand or mixture of ligands comprises at least one reactive functionality and said linker or mixture of linkers comprises at least two functional groups having complementary reactivity to at least one of the reactive functional groups of the ligand wherein said contacting is conducted under conditions wherein the complementary functional groups react to form a covalent linkage between said linker and at least two of said ligands; (b) assaying said first collection or iteration of multimeric compounds to assess which if any of said multimeric compounds possess multibinding properties; (c) repeating the process of (a) and (b) above until at least one multimeric compound is found to possess multibinding properties; (d) evaluating what molecular constraints imparted multibinding properties to the multimeric compound or compounds found in the first iteration recited in (a)- (c) above; (e) creating a second collection or iteration of multimeric compounds which elaborates upon the particular molecular constraints imparting multibinding properties to the multimeric compound or compounds found in said first iteration;

(f) evaluating what molecular constraints imparted enhanced multibinding properties to the multimeric compound or compounds found in the second collection or iteration recited in (e) above; (g) optionally repeating steps (e) and (f) to further elaborate upon said molecular constraints.

Preferably, steps (e) and (f) are repeated at least two times, more preferably at from 2-50 times, even more preferably from 3 to 50 times, and still more preferably at least 5-50 times.

BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 graphically describes the renin-angiotensin system (RAS).

Figure 2 illustrates a method for optimizing the linker geometry for presentation of ligands (filled circles) in bivalent compounds: A. phenyldiacetylene core structure B. cyclohexane dicarboxylic acid core structure Figure 3 shows exemplary linker"core"structures.

Figure 4 illustrates examples of multi-binding compounds comprising (A) 2 ligands, (B) 3 ligands, (C) 4 ligands, and (D) > 4 ligands attached in different formats to a linker.

Figures 5A through 5C illustrate the ligand losartan, which may be used in preparing multi-binding compounds. Potentially modifiable positions are indicated by arrows (A). Examples of these sites are set forth (B), as well as multivalomers using

functionality within the molecule and constructed multivalomers by introducing a point of attachment (C).

Figures 6A through 6C illustrate convenient methods for preparing the multibinding compounds of this invention.

Figure 7 illustrates the differing valency of multivalomers (dimeric, trimeric and tetrameric), as exemplified using tetrazole linked multivalomers.

Figure 8 illustrates differing framework building blocks for losartin multivalomers.

Figure 9 illustrates different relative connectivity for losartan multivalomers.

Figure 10 illustrates exemplary heterovalomers of losartan and valsartan.

DETAILED DESCRIPTION OF 1lIE INVENTION Biological systems in general are controlled by molecular interactions between bioactive ligands and their receptors, in which the receptor"recognizes"a molecule or a portion thereof (i. e., a ligand domain) to produce a biological effect. The result of this interaction can be either to initiate the desired biological effect of the receptor, or alternatively to inhibit or alter (i. e., to modulate) the normal function of the receptor.

Accordingly, diseases or conditions that involve, or are mediated by, AT receptors can be treated with pharmacologically active ligands that interact with such receptors to initiate, modulate or abrogate angiotensin receptor activity.

The interaction of an AT receptor and an AT receptor-binding ligand may be described in terms of affinity"and"specificity". The"affinity"and"specificity"of any

given ligand-AT receptor interaction is dependent upon the complementarity of molecular binding surfaces and the energetic costs of complexation (i. e., the net difference in free energy between bound and free states). Affinity may be quantified by the equilibrium- constant of complex formation, the ratio of on/off rate constants, and/or by the free energy of complex formation. Specificity relates to the difference in binding affinity of a ligand for different receptors.

The net free energy of interaction of such ligand with an AT receptor is the difference between energetic gains (enthalpy gained through molecular complementarity and entropy gained through the hydrophobic effect) and energetic costs (enthalpy lost through decreased solvation and entropy lost through reduced translational, rotational and conformational degrees of freedom).

The compounds of this invention comprise 2 to 10 AT receptor-binding ligands covalently linked together and capable of acting as multibinding agents. Without wishing to be bound by theory, the enhanced activity of these compounds is believed to arise at least in part from their ability to bind in a multivalent manner with multiple ligand binding sites on an AT receptor or receptors, which gives rise to a more favorable net free energy of binding. Multivalent interactions differ from collections of individual monovalent (univalent) interactions by being capable of providing enhanced biologic and/or therapeutic effect. Multivalent binding can amplify binding affinities and differences in binding affinities, resulting in enhanced binding specificity as well as affinity.

Definitions As used herein: The term"alkyl"refers to a monoradical branched or unbranched saturated hydrocarbon chain, preferably having from 1 to 40 carbon atoms, preferably 1-10 carbon

atoms, more preferably 1-6 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n- butyl, secondary butyl, tert-butyl, n-hexyl, n-octyl, n-decyl, n-dodecyl, 2-ethyldodecyl, tetradecyl, and the like, unless otherwise indicated.

The term"substituted alkyl"refers to an alkyl group as defined above having from 1 to 5 substituents selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, nitro,-SO- alkyl,-SO-aryl,-SO-heteroaryl,-SO2-alkyl,-SO2-aryl,-SO2-hete roaryl, and-NRaRb, wherein Ra and Rb may be the same or different and and are chosen from hydrogen, optionally substituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic.

The term"alkylene"refers to a diradical of a branched or unbranched saturated hydrocarbon chain, preferably having from 1 to 40 carbon atoms, preferably 1-10 carbon atoms, more preferably 1-6 carbon atoms. This term is exemplified by groups such as methylene (-CH2-), ethylene (-CH2CH2-), the propylene isomers (e. g.,-CH2CH2CH2-and -CH (CH3) CH2-) and the like.

The term"substituted alkylene"refers to: (1) an alkylene group as defined above having from 1 to 5 substituents selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, aminoacyl, aminoacyloxy, oxyacylamino, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, thioaryloxy, heteroaryl, heteroaryloxy, thioheteroaryloxy,

heterocyclic, heterocyclooxy, thioheterocyclooxy, nitro, and-NRaRb, wherein Ra and Rb may be the same or different and are chosen from hydrogen, optionally substituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic. Additionally, such substituted alkylene groups include those where 2 substituents on the alkylene group are fused to form one or more cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heterocyclic or heteroaryl groups fused to the alkylene group; (2) an alkylene group as defined above that is interrupted by 1-20 atoms independently chosen from oxygen, sulfur and Nua-, where Ra is chosen from hydrogen, optionally substituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkenyl, cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic, or groups selected from carbonyl, carboxyester, carboxyamide and sulfonyl; and (3) an alkylene group as defined above that has both from 1 to 5 substituents as defined above and is also interrupted by 1-20 atoms as defined above. Examples of substituted alkylenes are chloromethylene (-CH (Cl)-), aminoethylene (-CH (N) CH2-), 2- carboxypropylene isomers (-CH2CH (CO2H) CH2-), ethoxyethyl (-CH2CH2O-CH2CH2-), ethylmethylaminoethyl (-CH2CH2N (CH3) CH2CH2-), 1-ethoxy-2-(2-ethoxy-ethoxy) ethane (-CH2CH2O-CH2CH2-OCH2CH2-OCH2CH2-), and the like.

The term"alkaryl"or"aralkyl"refers to the groups-alkylene-aryl and -substituted alkylene-aryl in which alkylene and aryl are as defined herein. Such alkaryl groups are exemplified by benzyl, phenethyl and the like.

The term"alkoxy"refers to the groups alkyl-O-, alkenyl-0-, cycloalkyl-O-, cycloalkenyl-O-, and alkynyl-0-, where alkyl, alkenyl, cycloalkyl, cycloalkenyl, and alkynyl are as defined herein. Preferred alkoxy groups are alkyl-O-and include, by way of example, methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n- pentoxy, n-hexoxy, 1,2-dimethylbutoxy, and the like.

The term"substituted alkoxy"refers to the groups substituted alkyl-O-, substituted alkenyl-O-, substituted cycloalkyl-O-, substituted cycloalkenyl-O-, and substituted alkynyl- O-where substituted alkyl, substituted alkenyl, substituted cycloalkyl, substituted cycloalkenyl and substituted alkynyl are as defined herein.

The term"alkylalkoxy"refers to the groups-alkylene-O-alkyl, alkylene-0- substituted alkyl, substituted alkylene-0-alkyl and substituted alkylene-O-substituted alkyl wherein alkyl, substituted alkyl, alkylene and substituted alkylene are as defined herein.

Examples of such groups are methylenemethoxy (-CH2OCH3), ethylenemethoxy (-CH2CH20CH3), n-propylene-iso-propoxy (-CH2CH2CH20CH (CH3) 2), methylene-t-butoxy (-CH2-O-C (CH3) 3) and the like.

The term"alkylthioalkoxy"refers to the group-alkylene-S-alkyl, alkylene-S- substituted alkyl, substituted alkylene-S-alkyl and substituted alkylene-S-substituted alkyl wherein alkyl, substituted alkyl, alkylene and substituted alkylene are as defined herein.

Preferred alkylthioalkoxy groups are alkylene-S-alkyl and include, by way of example, methylenethiomethoxy (-CH2SCH3), ethylenethiomethoxy (-CH2CH2SCH3), n-propylene- iso-thiopropoxy (-CH2CH2CH2SCH (CH3) 2), methylene-t-thiobutoxy (-CH2SC (CH3) 3) and the like.

"Alkenyl"refers to a monoradical of a branched or unbranched unsaturated hydrocarbon preferably having from 2 to 40 carbon atoms, preferably 2-10 carbon atoms, more preferably 2-6 carbon atoms, and preferably having 1-6 double bonds. This term is further exemplified by such radicals as vinyl, prop-2-enyl, pent-3-enyl, hex-5-enyl, 5- ethyldodec-3,6-dienyl, and the like.

The term"substituted alkenyl"refers to an alkenyl group as defined above having from 1 to 5 substituents selected from the group consisting of alkoxy, substituted alkoxy,

acyl, acylamino, acyloxy, amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thiol, thioalkoxy, substituted thioalkoxy, aryl, heteroaryl, heterocyclic, aryloxy, thioaryloxy, heteroaryloxy, thioheteroaryloxy, heterocyclooxy, thioheterocyclooxy, nitro,-SO-alkyl,-SO-substituted alkyl,-SO-aryl,-SO-heteroaryl,-SO2-alkyl,-SOZ-substituted alkyl,-SO2-aryl,-SO2- heteroaryl, and,-NRaRb, wherein Ra and Rb may be the same or different and are chosen from hydrogen, optionally substituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic.

"Alkenylene"refers to a diradical of an unsaturated hydrocarbon, preferably having from 2 to 40 carbon atoms, preferably 2-10 carbon atoms, more preferably 2-6 carbon atoms, and preferably having 1-6 double bonds. This term is further exemplified by such radicals as 1,2-ethenyl, 1,3-prop-2-enyl, 1,5-pent-3-enyl, 1,4-hex-5-enyl, 5-ethyl-1,12- dodec-3,6-dienyl, and the like.

The term"substituted alkenylene"refers to an alkenylene group as defined above having from 1 to 5 substituents, selected from the group consisting of alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, amino, aminoacyl, aminoacyloxy, oxyacylamino, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, thioaryloxy, heteroaryl, heteroaryloxy, thioheteroaryloxy, heterocyclic, heterocyclooxy, thioheterocyclooxy, nitro, and NRaRb, wherein Ra and Rb may be the same or different and are chosen from hydrogen, optionally substituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic. Additionally, such substituted alkenylene groups include those where 2 substituents on the alkenylene group are fused to form one or more cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heterocyclic or heteroaryl groups fused to the alkenylene group.

"Alkynyl"refers to a monoradical of an unsaturated hydrocarbon, preferably having from 2 to 40 carbon atoms, preferably 2-10 carbon atoms, more preferably 2-6 carbon atoms, and preferably having 1-6 triple bonds. This term is further exemplified by such radicals as acetylenyl, prop-2-ynyl, pent-3-ynyl, hex-5-ynyl, 5-ethyldodec-3,6-diynyl, and the like.

The term"substituted alkynyl"refers to an alkynyl group as defined above having from 1 to 5 substituents, selected from the group consisting of alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, amino, aminoacyl, aminoacyloxy, oxyacylamino, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, thioaryloxy, heteroaryl, heteroaryloxy, thioheteroaryloxy, heterocyclic, heterocyclooxy, thioheterocycloxy, nitro,-SO-alkyl,-SO-substituted alkyl, -SO-aryl,-SO-heteroaryl,-SO2-alkyl,-SO2-substituted alkyl,-SO2-aryl,-SO2-heteroaryl, SO2-heterocyclic, NRaRb, wherein Ra and Rb may be the same or different and are chosen from hydrogen, optionally substituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic.

"Alkynylene"refers to a diradical of an unsaturated hydrocarbon radical, preferably having from 2 to 40 carbon atoms, preferably 2-10 carbon atoms, more preferably 2-6 carbon atoms, and preferably having 1-6 triple bonds. This term is further exemplified by such radicals as 1,3-prop-2-ynyl, 1,5-pent-3-ynyl, 1,4-hex-5-ynyl, 5-ethyl-1,12-dodec-3,6- diynyl, and the like.

The term"acyl"refers to the groups-CHO, alkyl-C (O)-, substituted alkyl-C (O)-, cycloalkyl-C (O)-, substituted cycloalkyl-C (O)-, cycloalkenyl-C (O)-, substituted cycloalkenyl-C (O)-, aryl-C (O)-, heteroaryl-C (O)- and heterocyclic-C (O)- where alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic are as defined herein.

The term"acylamino"refers to the group-C (O) NRR where each R is independently hydrogen, alkyl, substituted alkyl, aryl, heteroaryl, heterocyclic or where both R groups are joined to form a heterocyclic group (e. g., morpholine) wherein alkyl, substituted alkyl, aryl, heteroaryl and heterocyclic are as defined herein.

The term"aminoacyl"refers to the group-NRC (O) R where each R is independently hydrogen, alkyl, substituted alkyl, aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, aryl, heteroaryl and heterocyclic are as defined herein.

The term"aminoacyloxy"refers to the group-NRC (0) OR where each R is independently hydrogen, alkyl, substituted alkyl, aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, aryl, heteroaryl and heterocyclic are as defined herein.

The term"acyloxy"refers to the groups alkyl-C (O) O-, substituted alkyl-C (O) O-, cycloalkyl-C (O) O-, substituted cycloalkyl-C (O) O-, aryl-C (O) O-, heteroaryl-C (O) O-, and heterocyclic-C (O) O- wherein alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, heteroaryl, and heterocyclic are as defined herein.

The term"aryl"refers to an unsaturated aromatic carbocyclic group of from 6 to 20 carbon atoms having a single ring (e. g., phenyl) or multiple condensed (fused) rings (e. g., naphthyl or anthryl).

Unless otherwise constrained by the definition for the aryl substituent, such aryl groups can optionally be substituted with from 1 to 5 substituents selected from the group consisting of acyloxy, hydroxy, thiol, acyl, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, substituted alkyl, substituted alkoxy, substituted alkenyl, substituted alkynyl, substituted cycloalkyl, substituted cycloalkenyl, amino, aminoacyl, acylamino, alkaryl, aryl, aryloxy, azido, carboxyl, carboxylalkyl, cyano, halo, nitro, heteroaryl, heteroaryloxy,

heterocyclic, heterocyclooxy, aminoacyloxy, oxyacylamino, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioheteroaryloxy,-SO-alkyl,-SO-substituted alkyl,-SO-aryl, -SO-heteroaryl,-SO2-alkyl,-SO2-substituted alkyl,-SO2-aryl,-SO2-heteroaryl, trihalomethyl, NRDRb, wherein Ra and Rb may be the same or different and are chosen from hydrogen, optionally substituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic. Preferred aryl substituents include alkyl, alkoxy, halo, cyano, nitro, trihalomethyl, and thioalkoxy.

The term"aryloxy"refers to the group aryl-O-wherein the aryl group is as defined above including optionally substituted aryl groups as also defined above.

The term"arylene"refers to a diradical derived from aryl or substituted aryl as defined above, and is exemplified by 1,2-phenylene, 1,3-phenylene, 1,4-phenylene, 1,2- naphthylene and the like.

The term"amino"refers to the group-NH2 The term"substituted amino"refers to the group-NRR where each R is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl and heterocyclic provided that both R's are not hydrogen.

The term"carboxyalkyl"refers to the group"-C (O) O-alkyl","-C (O) O-substituted alkyl","-C (O) O-cycloalkyl","-C (O) O-substituted cycloalkyl","-C (O) 0-alkenyl","-C (O) O- substituted alkenyl","-C (O) O-alkynyl" and"-C (0) O-substituted alkynyl"where alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl and substituted alkynyl where alkynyl are as defined herein.

The term"cycloalkyl"refers to cyclic alkyl groups of from 3 to 20 carbon atoms having a single cyclic ring or multiple condensed rings. Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl, and the like, or multiple ring structures such as adamantanyl, and the like.

The term"substituted cycloalkyl"refers to cycloalkyl groups having from 1 to 5 substituents selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylakyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, nitro,-SO-alkyl,-SO-substituted alkyl,-SO- aryl,-SO-heteroaryl,-SO2-alkyl,-SO2-substituted alkyl,-SO2-aryl,-SO2-heteroaryl, and NRaRb, wherein Ra and Ru mamy be the same or different and are chosen from hydrogen, optionally substituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic.

The term"cycloalkenyl"refers to cyclic alkenyl groups of from 4 to 20 carbon atoms having a single cyclic ring or fused rings and at least one point of internal unsaturation. Examples of suitable cycloalkenyl groups include, for instance, cyclobut-2- enyl, cyclopent-3-enyl, cyclooct-3-enyl and the like.

The term"substituted cycloalkenyl"refers to cycloalkenyl groups having from 1 to 5 substituents selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, aminoacyl, aminoacyloxy, oxyaminoacyl, azido, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy,

heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, nitro,-SO-alkyl,-SO- substituted alkyl,-SO-aryl,.-SO-heteroaryl,-SO2-alkyl,-SO2-substituted alkyl,-SO2-aryl, -SO2-heteroaryl, and NRR, wherein Ra and Rb may be the same or different and are chosen from hydrogen, optionally substituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic.

The term"halo"or"halogen"refers to fluoro, chloro, bromo and iodo.

"Haloalkyl"refers to alkyl as defined above substituted by 1-4 halo groups as defined above, which may be the same or different, such as 3-fluorododecyl, 12, 12, 12- trifluorododecyl, 2-bromooctyl,-3-bromo-6-chloroheptyl, and the like.

The term"heteroaryl"refers to an aromatic group of from 1 to 15 carbon atoms and 1 to 4 heteroatoms selected from oxygen, nitrogen and sulfur within at least one ring (if there is more than one ring).

Unless otherwise constrained by the definition for the heteroaryl substituent, such heteroaryl groups can be optionally substituted with 1 to 5 substituents selected from the group consisting of acyloxy, hydroxy, thiol, acyl, alkyl, alkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, substituted alkyl, substituted alkoxy, substituted alkenyl, substituted alkynyl, substituted cycloalkyl, substituted cycloalkenyl, amino, aminoacyl, acylamino, alkaryl, aryl, aryloxy, azido, carboxyl, carboxylalkyl, cyano, halo, nitro, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, aminoacyloxy, oxyacylamino, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioheteroaryloxy,-SO-alkyl,-SO- substituted alkyl,-SO-aryl,-SO-heteroaryl,-S02-alkyl,-SOZ-substituted alkyl,-SO2-aryl, -SO2-heteroaryl, trihalomethyl, mono-and di-alkylamino, mono-and NRaRb, wherein Ra and Rb may be the same or different and are chosen from hydrogen, optionally substituted alkyl,

cycloalkyl, alkenyl, cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic. Preferred heteroaryls include pyridyl, pyrrolyl and furyl.

The term aheteroaryloxyX refers to the group heteroaryl-O-.

The term"heteroarylene"refers to the diradical group derived from heteroaryl or substituted heteroaryl as defined above, and is exemplified by the groups 2,6-pyridylene, 2,4-pyridiylene, 1,2-quinolinylene, 1,8-quinolinylene, 1,4-benzofuranylene, 2,5- pyridinylene, 1,3-morpholinylene, 2,5-indolenyl, and the like.

The term"heterocycle"or"heterocyclic"refers to a monoradical saturated or unsaturated group having a single ring or multiple condensed rings, from 1 to 40 carbon atoms and from 1 to 10 hetero atoms, preferably 1 to 4 heteroatoms, selected from nitrogen, sulfur, phosphorus, and/or oxygen within the ring.

Unless otherwise constrained by the definition for the heterocyclic substituent, such heterocyclic groups can be optionally substituted with 1 to 5, and preferably 1 to 3 substituents, selected from the group consisting of alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, acyl, acylamino, acyloxy, amino, aminoacyl, aminoacyloxy, oxyaminoacyl, cyano, halogen, hydroxyl, keto, thioketo, carboxyl, carboxylalkyl, thioaryloxy, thioheteroaryloxy, thioheterocyclooxy, thiol, thioalkoxy, substituted thioalkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, heterocyclic, heterocyclooxy, hydroxyamino, alkoxyamino, nitro,-SO-alkyl,-SO-substituted alkyl,-SO- aryl,-SO-heteroaryl,-SO2-alkyl,-SOZ-substituted alkyl,-SO2-aryl,-SO2-heteroaryl, and NRaRb, wherein W and R may be the same or different and are chosen from hydrogen, optionally substituted alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, aryl, heteroaryl and heterocyclic. Such heterocyclic groups can have a single ring or multiple condensed rings.

Examples of nitrogen heterocycles and heteroaryls include, but are not limited to, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, phenanthroline, isothiazole, phenazine, isoxazole, phenoxazine, phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline, morpholino, piperidinyl, tetrahydrofuranyl, and the like as well as N-alkoxy-nitrogen containing heterocycles.

A preferred class of heterocyclics include"crown compounds"which refers to a specific class of heterocyclic compounds having one or more repeating units of the formula [-(CH2-) =Y-] where m is equal to or greater than 2, and Y at each separate occurrence can be O, N, S or P. Examples of crown compounds include, by way of example only, [-(CH2)3-NH-]3, [-((CH2)2-O)4-((CH2)2-NH)2] and the like. Typically such crown compounds can have from 4 to 10 heteroatoms and 8 to 40 carbon atoms.

The term"heterocyclooxy"refers to the group heterocyclic-O-.

The term"thioheterocyclooxy"refers to the group heterocyclic-S-.

The term"heterocyclene"refers to the diradical group derived from a heterocycle as defined herein, and is exemplified by the groups 2,6-morpholino, 2,5-morpholino and the like.

The term"oxyacylamino"refers to the group-OC (O) NRR where each R is independently hydrogen, alkyl, substituted alkyl, aryl, heteroaryl, or heterocyclic wherein alkyl, substituted alkyl, aryl, heteroaryl and heterocyclic are as defined herein.

The term"thiol"refers to the group-SH.

The term"thioalkoxy"refers to the group-S-alkyl.

The term"substituted thioalkoxy"refers to the group-S-substituted alkyl.

The term"thioaryloxy"refers to the group aryl-S-wherein the aryl group is as defined above including optionally substituted aryl groups also defined above.

The term"thioheteroaryloxy"refers to the group heteroaryl-S-wherein the heteroaryl group is as defined above including optionally substituted aryl groups as also defined above.

As to any of the above groups which contain one or more substituents, it is understood, of course, that such groups do not contain any substitution or substitution patterns which are sterically impractical and/or synthetically non-feasible. In addition, the compounds of this invention include all stereochemical isomers arising from the substitution of these compounds.

"Alkyl optionally interrupted by 1-5 atoms chosen from O, S, or N"refers to alkyl as defined above in which the carbon chain is interrupted by O, S, or N. Within the scope are ethers, sulfides, and amines, for example 1-methoxydecyl, 1-pentyloxynonane, 1- (2- isopropoxyethoxy)-4-methylnonane, 1- (2-ethoxyethoxy) dodecyl, 2- (t-butoxy) heptyl, 1- pentylsulfanylnonane, nonylpentylamine, and the like.

"Heteroarylalkyl"refers to heteroaryl as defined above linked to alkyl as defined above, for example pyrid-2-ylmethyl, 8-quinolinylpropyl, and the like.

"Optional"or"optionally"means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, optionally substituted alkyl means that alkyl may or may not be substituted by those groups enumerated in the definition of substituted alkyl.

The term"pharmaceutically acceptable salt"refers to salts which retain the biological effectiveness and properties of the multibinding compounds of this invention and which are not biologically or otherwise undesirable. In many cases, the multibinding compounds of this invention are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto.

Pharmaceutically acceptable base addition salts can be prepared from inorganic and organic bases. Salts derived from inorganic bases, include by way of example only, sodium, potassium, lithium, ammonium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary and tertiary amines, such as alkyl amines, dialkyl amines, trialkyl amines, substituted alkyl amines, di (substituted alkyl) amines, tri (substituted alkyl) amines, alkenyl amines, dialkenyl amines, trialkenyl amines, substituted alkenyl amines, di (substituted alkenyl) amines, tri (substituted alkenyl) amines, cycloalkyl amines, di (cycloalkyl) amines, tri (cycloalkyl) amines, substituted cycloalkyl amines, disubstituted cycloalkyl amine, trisubstituted cycloalkyl amines, cycloalkenyl amines, di (cycloalkenyl) amines, tri (cycloalkenyl) amines, substituted cycloalkenyl amines, disubstituted cycloalkenyl amine, trisubstituted cycloalkenyl amines, aryl amines, diaryl amines, triaryl amines, heteroaryl amines, diheteroaryl amines, triheteroaryl amines, heterocyclic amines, diheterocyclic amines, triheterocyclic amines, mixed di-and tri-amines where at least two of the substituents on the amine are different and are selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl,

heteroaryl, heterocyclic, and the like. Also included are amines where the two or three substituents, together with the amino nitrogen, form a heterocyclic or heteroaryl group.

Examples of suitable amines include, by way of example only, isopropylamine, trimethyl amine, diethyl amine, tri (iso-propyl) amine, tri (n-propyl) amine, ethanolamine, 2- dimethylaminoethanol, tromethamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, N-alkylglucamines, theobromine, purines, piperazine, piperidine, morpholine, N-ethylpiperidine, and the like.

It should also be understood that other carboxylic acid derivatives would be useful in the practice of this invention, for example, carboxylic acid amides, including carboxamides, lower alkyl carboxamides, dialkyl carboxamides, and the like.

Pharmaceutically acceptable acid addition salts may be prepared from inorganic and organic acids. Salts derived from inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Salts derived from organic acids include acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p- toluene-sulfonic acid, salicylic acid, and the like.

The term"protecting group"or"blocking group"refers to any group which when bound to one or more hydroxyl, thiol, amino or carboxyl groups of the compounds prevents reactions from occurring at these groups and which protecting group can be removed by conventional chemical or enzymatic steps to reestablish the hydroxyl, thiol, amino or carboxyl group. See, generally, T. W. Greene & P. G. M. Wuts, Protective Groups in Organic Synthesis, 2nd Ed., 1991, John Wiley and Sons, N. Y.

The particular removable blocking group employed is not critical and preferred removable hydroxyl blocking groups include conventional substituents such as allyl, benzyl, acetyl, chloroacetyl, thiobenzyl, benzylidine, phenacyl, t-butyl-diphenylsilyl and any other group that can be introduced chemically onto a hydroxyl functionality and later selectively removed either by chemical or enzymatic methods in mild conditions compatible with the nature of the product.

Preferred removable amino blocking groups include conventional substituents such as t-butyoxycarbonyl (t-BOC), benzyloxycarbonyl (CBZ), fluorenylmethoxycarbonyl (FMOC), allyloxycarbonyl (ALOC) and the like, which can be removed by conventional conditions compatible with the nature of the product.

Preferred carboxyl protecting groups include esters such as methyl, ethyl, propyl, t- butyl, etc. which can be removed by mild hydrolysis conditions compatible with the nature of the product.

As used herein, the terms"inert organic solventß or'$inert solvent"mean a solvent inert under the conditions of the reaction being described in conjunction therewith [including, for example, benzene, toluene, acetonitrile, tetrahydrofuran ("THF"), dimethylformamide ("DMF"), chloroform ("CHCI3"), methylene chloride (or dichloromethane or"CH2Cl2"), diethyl ether, ethyl acetate, acetone, methylethyl ketone, methanol, ethanol, propanol, isopropanol, tert-butanol, dioxane, pyridine, and the like].

Unless specified to the contrary, the solvents used in the reactions of the present invention are inert solvents.

The terms"angiotensin receptor"or"AT receptor"refer to membrane bound proteins that function to bind the vasoactive octapeptide hormone angiotensin II. The terms include both AT, receptor and AT2 receptor.

"Ligands used herein denotes a compound that is a binding partner for an angiotensin receptor, and is bound thereto, for example, by complementarity. The specific region or regions of the ligand molecule recognized by the ligand binding site of an angiotensin receptor is designated as the"ligand domain". A ligand may be either capable of binding to a receptor by itself, or may require the presence of one or more non-ligand components for binding (e. g., ions, a lipid molecule, a solvent molecule, and the like).

Ligands useful in this invention comprise AT receptor modulators such as Losartan13,20,Candesartan13,L-158,80945,Irbesartan16,19,Epro sartan17,18, Vals3, 15, Tasosartan, Ripisartan, Telmesartanl3, Zolasartan, PD 123,31945, L-163, 958, EXP-801, L-162,313 and PD123,177. See Table 3 for structures of various angiotensin receptor ligands. Preferred ligands are non-peptidyl, although peptidomimetics may be used.

All of the known non-peptide ATl receptor antagonists conform readily to the pharmacophore described below. Specifically, a proposed model demonstrates that there is significant overlap between the non-peptide antagonists and the endogenous peptide agonists.-This overlap is proposed to take place between the antagonist binding site and the binding site of the C-terminus of the endogenous peptide agonist. This model demonstrates an overlap between the propylimidazole and biaryl tetrazole motifs of the non- peptide antagonists with the side chains of the Ili-His'-Pr6-PhO and the terminal carboxylate in the N-terminus of the angiotensin II peptide''29, 3s. The non-peptide antagonists exemplified generally contain a biaryl with an acidic group such as carboxylate, a sulphonamide or isosteric tetrazole. Eprosartan is-the exception in that it does not contain this motif. They also contain a heterocyclic scaffold at the top of the molecule. This heterocyclic scaffold is generally substituted with a lipophilic alkyl group (e. g., the propyl group of losartan) and a hydrogen bond acceptor (e. g., carboxamide, carboxaldehyde, carboxylate or hydroxymethyl). At the C2 position of the imidazole, a linear alkyl or

alkenyl group is preferred, optimally 3-4 carbon atoms in length. At the C4 position of the imidazole, there is tolerance for large substituents. There are a number of potent analogues in the literature that introduce significant steric bulk and polar functionality into this region.

At the C5 position of the imidazole, a variety of substituents are acceptable. The general trend appears to be that a hydrogen bond acceptor is preferred (e. g., carboxamide, carboxaldehyde, carboxylate or hydroxymethyl).

As discussed above, the angiotensin receptors appear to have an endogenous peptide ligand (agonist) binding site and a different (or partially overlapping) small molecule antagonist (or possibly agonist) binding site. Accordingly, multivalent binding compounds which contain a fragment of the AT, endogenous ligand (or, preferably, a peptidomimetic thereof) linked to all or part of a small molecule antagonist (e. g., losartan) may be particularly useful.

While it is contemplated that many angiotensin receptor ligands that are currently known can be used in the preparation of multibinding compounds of this invention, it should be understood that portions of the ligand structure that are not essential for molecular recognition and binding activity (i. e., that are not part of the ligand domain) may be varied substantially, replaced with unrelated structures and, in some cases, omitted entirely without affecting the binding interaction. Accordingly, it should be understood that the term"ligand"is not intended to be limited to compounds known to be useful as angiotensin receptor-binding compounds (e. g., known drugs), in that ligands that exhibit marginal activity or lack useful activity as monomers can be highly active as multibinding compounds, because of the biological benefit conferred by multivalency. The primary requirement for a ligand as defined herein is that it has a ligand domain, as defined above, which is available for binding to a recognition site on an angiotensin receptor.

For purposes of the present invention, the term"ligand"or"ligands"is intended to include the racemic ligands as well as the individual stereoisomers of the ligands, including pure enantiomers and non-racemic mixtures thereof. The scope of the invention as described and claimed encompasses the racemic forms of the ligands as well as the individual enantiomers and non-racemic mixtures thereof.

The term"ligand binding site"as used herein denotes a site on an angiotensin receptor that recognizes a ligand domain and provides a binding partner for the ligand. The ligand binding site may be defined by monomeric or multimeric structures. This interaction may be capable of producing a unique biological effect, for example agonism, antagonism, modulation, or may maintain an ongoing biological event, and the like.

It should be recognized that the ligand binding sites of angiotensin receptors that participate in biological multivalent binding interactions are constrained to varying degrees by their intra-and intermolecular associations. For example, angiotensin receptor ligand binding sites may be covalently joined in a single structure, noncovalently associated in one or more multimeric structures, embedded in a membrane or biopolymer matrix, and so on, and therefore have less translational and rotational freedom than if the same sites were present as monomers in solution.

The terms"agonism"and"antagonism"are well known in the art. As used herein, the term"agonist"refers to a ligand that when bound to an angiotensin receptor stimulates its activity. The term"antagonist"refers to a ligand that when bound to an angiotensin receptor inhibits its activity. Receptor block or activation may result from allosteric effects of ligand binding to the receptor rather than occupancy of the receptor. These allosteric effects may produce changes in protein conformation that affect angiotensin binding sites.

The term"modulatory effect"is intended to refer to the ability of a ligand to change the biological activity of an agonist or antagonist through binding to a receptor.

"Multibinding agent"or"multibinding compound"refers herein to a compound that has from 2 to 10 ligands as defined herein, (which may be the same or different) covalently bound to one or more linkers (which may be the same or different), and is capable of multivalency, as defined below.

A multibinding compound provides. an improved biologic and/or therapeutic effect compared to that of the same number of unlinked ligands available for binding to the ligand binding sites on an angiotensin receptor or receptors. Examples of improved"biologic and/or therapeutic effect"include increased ligand-receptor binding interactions (e. g., increased affinity, increased ability to elicit a functional change in the target, improved kinetics), increased selectivity for the target, increased potency,. increased efficacy, decreased toxicity, increased therapeutic index, improved duration of action, improved bioavailability, improved pharmacokinetics, improved activity spectrum, and the like. The multibinding compounds of this invention will exhibit at least one, and preferably more than one, of the above-mentioned effects.

"Univalency"as used herein refers to a single binding interaction between one ligand with one ligand binding site as defined herein. It should be noted that a compound having multiple copies of a ligand (or ligands) exhibits univalency when only one ligand of that t compound interacts with a ligand binding site. Examples of univalent interactions are depicted below. univalent interaction

"Multivalency"as used herein refers to the concurrent binding of from 2 to 10 linked ligands, which may be the same or different, and two or more corresponding ligand binding sites, which may be the same or different. An example of trivalent binding is depicted below for illustrative purposes. trivalent interaction It should be understood that not all compounds that contain multiple copies of a ligand attached to a linker necessarily exhibit the phenomena of multivalency, i. e., that the biologic and/or therapeutic effect of the multibinding agent is greater than that of the same number of unlinked ligands made available for binding to the ligand binding sites. For multivalency to occur, the ligand domains of the ligands that are linked together must be presented to their cognate ligand binding sites by the linker or linkers in a specific manner in order to bring about the desired ligand-orienting result, and thus produce a multibinding interaction.

The term"library"refers to at least 3, preferably from 102 to 109 and more preferably from 102 to 104 multimeric compounds. Preferably, these compounds are prepared as a multiplicity of compounds in a single solution or reaction mixture which permits facile synthesis thereof. In one embodiment, the library of multimeric compounds can be directly assayed for multibinding properties. In another embodiment, each member of the library of multimeric compounds is first isolated and, optionally, characterized. This member is then assayed for multibinding properties.

The term"collection"refers to a set of multimeric compounds which are prepared either sequentially or concurrently (e. g., combinatorially). The collection comprises at least 2 members; preferably from 2 to 109 members and still more preferably from 10 to 104 members.

The term"multimeric compound"refers to compounds comprising from 2 to 10 ligands covalently connected through at least one linker which compounds may or may not possess multibinding properties (as defined herein).

The term"pseudohalide"refers to functional groups which react in displacement reactions in a manner similar to a halogen. Such functional groups include, by way of example, mesyl, tosyl, azido and cyano groups.

The term"linker", identified where appropriate by the symbol X, refers to a group or groups that covalently links from 2 to 10 ligands (as defined above) in a manner that provides a compound capable of multivalency. The linker is a ligand-orienting entity that permits attachment of multiple copies of a ligand (which may be the same or different) thereto.

The term"linker"includes everything that is not considered to be part of the ligand, e. g., ancillary groups such as solubilizing groups, lipophilic groups, groups that alter pharmacodynamics or pharmacokinetics, groups that modify the diffusability of the multibinding compound, spacers that attach the ligand to the linker, groups that aid the ligand-orienting function of the linker, for example, by imparting flexibility or rigidity to the linker as a whole, or to a portion thereof, and so on. The term"linker"does not, however, cover solid inert supports such as beads, glass particles, rods, and the like, but it is to be understood that the multibinding compounds of this invention can be attached to a

solid support if desired, for example, for use in separation and purification processes and for similar applications.

The extent to which the previously discussed enhanced activity of multibinding compounds is realized in this invention depends upon the efficiency with which the linker or linkers that joins the ligands presents them to their array of ligand binding sites. Beyond presenting these ligands for multivalent interactions with ligand binding sites, the linker spatially constrains these interactions to occur within dimensions defined by the linker.

The linkers used in this invention are selected to allow multivalent binding of ligands to any desired ligand binding sites of an angiotensin receptor, whether such sites are located within the cell membrane, on the surface of the cell membrane, extracellularly, or intracellularly, or at any intermediate position thereof. The preferred linker length will vary depending on the distance between adjacent ligand binding sites, and the geometry, flexibility and composition of the linker. The length of the linker will preferably be in the range of about 2A to about 100A, more preferably from about 2A to about 50A and even more preferably from about 5A to about 20A.

The ligands are covalently attached to the linker or linkers using conventional chemical techniques. The reaction chemistries resulting in such linkage are well known in the art and involve the use of reactive functional groups present on the linker and ligand.

Preferably, the reactive functional groups on the linker are selected relative to the functional groups available on the ligand for coupling, or which can be introduced onto the ligand for this purpose. Again, such reactive functional groups are well known in the art.

For example, reaction between a carboxylic acid of either the linker or the ligand and a primary or secondary amine of the ligand or the linker in the presence of suitable well- known activating agents results in formation of an amide bond covalently linking the ligand to the linker; reaction between an amine group of either the linker or the ligand and a

sulfonyl halide of the ligand or the linker results in formation of a sulfonamide bond covalently linking the ligand to the linker; and reaction between an alcohol or phenol group of either the linker or the ligand and an alkyl or aryl halide of the ligand or the linker results in formation of an ether bond covalently linking the ligand to the linker. The table below illustrates numerous reactive functional groups and the resulting bonds formed by reaction therebetween. Where functional groups are lacking, they can be created by suitable chemistries that are described in standard organic chemistry texts such as J. March, Advanced Organic Chem., 4 Ed., (Wiley-Interscience, N. Y., 1992).

Complementary Binding Chemistries First Reactive Group Second Reactive Group Linkage hydroxyl isocyanate urethane amine epoxide p-hydroxyamine sulfonyl halide amine sulfonamide carboxyl amine amide hydroxyl alkyl/aryl halide ether The linker is attached to the ligand at a position that retains ligand domain-ligand binding site interaction and specifically which permits the ligand domain of the ligand to orient itself to bind to the ligand binding site. Such positions and synthetic protocols for linkage are well known in the art. The term linker embraces everything that is not considered to be part of the ligand.

The relative orientation in which the ligand domains are displayed depends both on the particular point or points of attachment of the ligands to the linker, and on the framework geometry. The determination of where acceptable substitutions can be made on a ligand is typically based on prior knowledge of structure-activity relationships of the ligand and/or congeners and/or structural information about ligand-receptor complexes (e. g., X-ray crystallography, NMR, and the like). Such positions and synthetic protocols

for linkage are well known in the art and can be determined by those with ordinary skill in the art (see METHODS OF PREPARATION, Examples 1-3 and Figures 5-10).

Following attachment of a ligand to the linker or linkers, or to a significant portion thereof (e. g., 2-10 atoms of linker), the linker-ligand conjugate may be tested for retention of activity in a relevant assay system (see Utility and Testing below for representative assays).

At present, it is preferred that the multibinding compound is a bivalent compound in which two ligands are covalently linked, or a trivalent compound, in which three ligands are covalently linked. Linker design is further discussed under METHODS OF PREPARATION.

"Potency"as used herein refers to the minimum concentration at which a ligand is able to achieve a desirable biological or therapeutic effect. The potency of a ligand is typically proportional to its affinity for its receptor. In some cases, the potency may be non-linearly correlated with its affinity. In comparing the potency of two drugs, e. g., a multibinding agent and the aggregate of its unlinked ligand, the dose-response curve of each is determined under identical test conditions (e. g., in an in vitro or in vivo assay, in an appropriate animal model (such as a human patient)). The finding that the multibinding agent produces an equivalent biologic or therapeutic effect at a lower concentration than the aggregate unlinked ligand (e. g., on a per weight, per mole or per ligand basis) is indicative of enhanced potency.

"Selectivity"or"specificity"is a measure of the binding preferences of a ligand for different receptors. The selectivity of a ligand with respect to its target receptor relative to another receptor is given by the ratio of the respective values of K (i. e., the dissociation constants for each ligand-receptor complex) or, in cases where a biological effect is observed below the Kd, the ratio of the respective Ex, s or IC50s (i. e., the concentrations

that produce 50 % of the maximum response for the ligand interacting with the two distinct receptors).

The term"treatment"refers to any treatment of a disease or condition in a mammal, particularly a human, and includes: (i) preventing the disease or condition from occurring in a subject which may be predisposed to the condition but has not yet been diagnosed with the condition and, accordingly, the treatment constitutes prophylactic treatment for the pathologic condition; (ii) inhibiting the disease or condition, i. e., arresting its development; (iii) relieving the disease or condition, i. e., causing regression of the disease or condition; or (iv) relieving the symptoms resulting from the disease or condition without addressing the underlying disease or condition, e. g., relieving symptoms of angina pectoris and other conditions of ischemia but not an underlying cause such as, for example, atherosclerotic disease or hypertension.

The phrase"disease or condition which is modulated by treatment with a multibinding angiotensin receptor ligand"covers all disease states and/or conditions that are generally acknowledged in the art to be usefully treated with a ligand for an angiotensin receptor in general, and those disease states and/or conditions that have been found to be usefully treated by a specific multibinding compound of our invention, i. e., the compounds of Formula I. Such disease states include, by way of example only, hypertension, congestive heart failure, renal insufficiency, diabetic neuropathy, and the like.

The term"therapeutically effective amount"refers to that amount of multibinding compound that is sufficient to effect treatment, as defined above, when administered to a mammal in need of such treatment. The therapeutically effective amount will vary depending upon the subject and disease condition being treated, the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.

The term"pharmaceutically acceptable excipient"is intended ta include vehicles and carriers capable of being coadministered with a multibinding compound to facilitate the performance of its intended function. The use of such media for pharmaceutically active substances is well known in the art. Examples of such vehicles and carriers include solutions, solvents, dispersion media, delay agents, emulsions and the like. Any other conventional carrier suitable for use with the multibinding compounds also falls within the scope of the present invention.

METHODS OF PREPARATION Linkers The linker or linkers, when covalently attached to multiple copies of the ligands, provides a biocompatible, substantially non-immunogenic multibindi. ng compound. The biological activity of the multibinding angiotensin receptor compound is highly sensitive to the geometry, composition, size, length, flexibility or rigidity, the presence or absence of anionic or cationic charge, the relative hydrophobicity/hydrophilicity, and similar properties of the linker. Accordingly, the linker is preferably chosen to maximize the biological activity of the compound. The linker may be biologically"neutral,"i. e., not itself contribute any additional biological activity to the multibinding compound, or it may be chosen to further enhance the biological activity of the compound. In general, the linker may be chosen from any organic molecule construct that orients two or more ligands for binding to the receptors to permit multivalency. In this regard, the linker can be considered as a"framework"on which the ligands are arranged in order to bring about the desired ligand-orienting result, and thus produce a multibinding compound.

For example, different orientations of ligands can be achieved by varying the geometry of the framework (linker) by use of mono-or polycyclic groups, such as aryl and/or heteroaryl groups, or structures incorporating one or more carbon-carbon multiple bonds (alkenyl, alkenylene, alkynyl or alkynylene groups). The optimal geometry and composition of frameworks (linkers) used in the multibinding compounds of this invention are based upon the properties of their intended receptors. For example, it is preferred to use rigid cyclic groups (e. g., aryl, heteroaryl), or non-rigid cyclic groups (e. g., cycloalkyl or crown groups) to reduce conformational entropy when such may be necessary to achieve energetically coupled binding.

Different hydrophobic/hydrophilic characteristics of the linker as well as the presence or absence of charged moieties can readily be controlled by the skilled artisan.

For example, the hydrophobic nature of a linker derived from hexamethylene diamine (H2N (CH2) 6NH2) or related polyamines can be modified to be substantially more hydrophilic by replacing the alkylene group with a poly (oxyalkylene) group such as found in the commercially available"Jeffamines" (class of surfactants).

Different frameworks can be designed to provide preferred orientations of the ligands. The identification of an appropriate framework geometry for ligand domain presentation is an important first step in the construction of a multi binding agent with enhanced activity. Systematic spatial searching strategies can be used to aid in the identification of preferred frameworks through an iterative process. Figure 2 illustrates a useful strategy for determining an optimal framework display orientation for ligand domains and can be used for preparing the bivalent compounds of this invention. Various alternative strategies known to those skilled in the art of molecular design can be substituted for the one described here.

As shown in Figure 2, the ligands (shown as filled circles) are attached to a central core structure such as phenyldiacetylene (Panel A) or cyclohexane dicarboxylic acid (Panel B). The ligands are spaced apart from the core by an attaching moiety of variable lengths m and n. If the ligand possesses multiple attachment sites (see discussion below), the orientation of the ligand on the attaching moiety may be varied as well. The positions of the display vectors around the central core structures are varied, thereby generating a collection of compounds. Assay of each of the individual compounds of a collection generated as described will lead to a subset of compounds with the desired enhanced activities (e. g., potency, selectivity). The analysis of this subset using a technique such as Ensemble Molecular Dynamics will suggest a framework orientation that favors the properties desired.

The process may require the use of multiple copies of the same central core structure or combinations of different types of display cores. It is to be noted that core structures other than those shown here can be used for determining the optimal framework display orientation of the ligands. The above-described technique can be extended to trivalent compounds and compounds of higher-order valency.

A wide variety of linkers is commercially available (Chem Sources USA and Chem Sources International; the ACD electronic database; and Chemical Abstracts). Many of the linkers that are suitable for use in this invention fall into this category. Others can be readily synthesized by methods known in the art, and as described below. Examples of linkers include aliphatic moieties, aromatic moieties, steroidal moieties, peptides, and the like. Specific examples are peptides or polyamides, hydrocarbons, aromatics, heterocyclics, ethers, lipids, cationic or anionic groups, or a combination thereof.

Examples are given below and in Figure 3, but it should be understood that various changes may be made and equivalents may be substituted without departing from the true

spirit and scope of the invention. For example, properties of the linker can be modified by the addition or insertion of ancillary groups into the linker, for example, to change the solubility of the multibinding compound (in water, fats, lipids, biological fluids, etc.), hydrophobicity, hydrophilicity, linker flexibility, antigenicity, stability, and the like. For example, the introduction of one or more poly (ethylene glycol) (PEG) groups onto the linker enhances the hydrophilicity and water solubility of the multibinding compound, increases both molecular weight and molecular size and, depending on the nature of the unPEGylated linker, may increase the in vivo retention time. Further, PEG may decrease antigenicity and potentially enhances the overall rigidity of the linker.

Ancillary groups that enhance the water solubility/hydrophilicity of the linker, and accordingly, the resulting multibinding compounds, are useful in practicing this invention.

Thus, it is within the scope of the present invention to use ancillary groups such as, for example, small repeating units of ethylene glycols, alcohols, polyols, (e. g., glycerin, glycerol propoxylate, saccharides, including mono-, oligosaccharides, etc.) carboxylates (e. g., small repeating units of glutamic acid, acrylic acid, etc.), amines (e. g., tetraethylenepentamine), and the like to enhance the water solubility and/or hydrophilicity of the multibinding compounds of this invention. In preferred embodiments, the ancillary group used to improve water solubility/hydrophilicity will be a polyether. In particularly preferred embodiments, the ancillary group will contain a small number of repeating ethylene oxide (-CH2CH20-) units.

The incorporation of lipophilic ancillary groups within the structure of the linker to enhance the lipophilicity and/or hydrophobicity of the compounds of Formula I is also within the scope of this invention. Lipophilic groups useful with the linkers of this invention include, but are not limited to, lower alkyl, aromatic groups and polycyclic aromatic groups. The aromatic groups may be either unsubstituted or substituted with other groups, but are at least substituted with a group which allows their covalent attachment to

the linker. As used herein the term"aromatic groups"incorporates both aromatic hydrocarbons and heterocyclic aromatics. Other lipophilic groups useful with the linkers of this invention include fatty acid derivatives which may or may not form micelles in aqueous medium and other specific lipophilic groups which modulate interactions between the multibinding compound and biological membranes.

Also within the scope of this invention is the use of ancillary groups which result in the compound of Formula I being incorporated into a vesicle, such as a liposome, or a micelle. The term"lipid"refers to any fatty acid derivative that is capable of forming a bilayer or micelle such that a hydrophobic portion of the lipid material orients toward the bilayer while a hydrophilic portion orients toward the aqueous phase. Hydrophilic characteristics derive from the presence of phosphato, carboxylic, sulfato, amino, sulfhydryl, nitro and other like groups well known in the art. Hydrophobicity could be conferred by the inclusion of groups that include, but are not limited to, long chain saturated and unsaturated aliphatic hydrocarbon groups of up to 20 carbon atoms and such groups substituted by one or more aryl, heteroaryl, cycloalkyl, and/or heterocyclic group (s). Preferred lipids are phosphoglycerides and sphingolipids, representative examples of which include phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, phosphatidic acid, palmitoyleoyl phosphatidylcholine, lysophosphatidylcholine, lysophosphatidyl-ethanolamine, dipalmitoylphosphatidylcholine, dioleoylphosphatidyl-choline, distearoyl- phosphatidylcholine and dilinoleoylphosphatidylcholine. Other compounds lacking phosphorus, such as sphingolipid and glycosphingolipid families, are also within the group designated as lipid. Additionally, the amphipathic lipids described above may be mixed with other lipids including triglycerides and sterols.

The flexibility of the linker can be manipulated by the inclusion of ancillary groups which are bulky and/or rigid. The presence of bulky or rigid groups can hinder free

rotation about bonds in the linker, or bonds between the linker and the ancillary group (s), or bonds between the linker and the functional groups. Rigid groups can include, for example, those groups whose conformational freedom is restrained by the presence of rings and/or n-bonds, for example, aryl, heteroaryl and heterocyclic groups. Other groups which can impart rigidity include polypeptide groups such as oligo-or polyproline chains.

Rigidity can also be imparted electrostatically. Thus, if the ancillary groups are either positively or negatively charged, the similarly charged ancillary groups will force the linker into a configuration affording the maximum distance between each of the like charges. The energetic cost of bringing the like-charged groups closer to each other, which is inversely related to the square of the distance between the groups, will tend to hold the linker in a configuration that maintains the separation between the like-charged ancillary groups. Further, ancillary groups bearing opposite charges will tend to be attracted to their oppositely charged counterparts and potentially may enter into both inter-and intramolecular ionic bonds. This non-covalent mechanism will tend to hold the linker in a conformation which allows bonding between the oppositely charged groups. The addition of ancillary groups which are charged, or alternatively, protected groups that bear a latent charge which is unmasked, following addition to the linker, by deprotection, a change in pH, oxidation, reduction or other mechanisms known to those skilled in the art, is within the scope of this invention.

Bulky groups can include, for example, large atoms, ions (e. g., iodine, sulfur, metal ions, etc.) or groups containing large atoms, polycyclic groups, including aromatic groups, non-aromatic groups and structures incorporating one or more carbon-carbon-bonds (i. e., alkenes and alkynes). Bulky groups can also include oligomers and polymers which are branched-or straight-chain species. Species that are branched are expected to increase the rigidity of the structure more per unit molecular weight gain than are straight-chain species.

In preferred embodiments, rigidity (entropic control) is imparted by the presence of alicyclic (e. g., cycloalkyl),. aromatic and heterocyclic groups. In other preferred embodiments, this comprises one or more six-membered rings. In still further preferred embodiments, the ring is an aryl group such as, for example, phenyl or naphthyl, or a macrocyclic ring such as, for example, a crown compound.

In view of the above, it is apparent that the appropriate selection of a linker group providing suitable orientation, entropy and physico-chemical properties is well within the skill of the art.

Eliminating or reducing antigenicity of the multibinding compounds described herein is also within the scope of this invention. In certain cases, the antigenicity of a multibinding compound may be eliminated or reduced by use of groups such as, for example, poly (ethylene glycol).

The Compounds of Formula I As explained above, the multibinding compounds described herein comprise 2-10 ligands attached covalently to a linker that links the ligands in a manner that allows their multivalent binding to ligand binding sites of angiotensin receptors. The linker spatially constrains these interactions to occur within dimensions defined by the linker. This and other factors increases the biologic and/or therapeutic effect of the multibinding compound as compared to the same number of ligands used in monobinding form.

The compounds of this invention are preferably represented by the empirical formula (L) p (X) q where L, X, p and q are as defined above. This is intended to include the several ways in which the ligands can be linked together in order to achieve the objective of multivalency, and a more detailed explanation is provided below.

As noted previously, the linker may be considered as a framework to which ligands are attached. Thus, it should be recognized that the ligands can be attached at any suitable position on this framework, for example, at the termini of a linear chain or at any intermediate position thereof.

The simplest and most preferred multibinding compound is a bivalent compound which can be represented as L-X-L, where L is a ligand and is the same or different and X is the linker. A trivalent compound could also be represented in a linear fashion, i. e., as a sequence of repeated units L-X-L-X-L, in which L is a ligand and is the same or different at each occurrence, as is X. However, a trivalent compound can also comprise three ligands attached to a central core, and thus be represented as (LX, where the linker X could include, for example, an aryl or cycloalkyl group. Tetravalent compounds can be represented in a linear array: L-X-L-X-L-X-L, or a branched array: L-X-L-X-L, i. e., a branched construct analogous to the isomers of butane (n-butyl, iso-butyl, sec-butyl, and t-butyl). Alternatively, it could be represented as an aryl or cycloalkyl derivative as described above with four (4) ligands attached to the core linker.

The same considerations apply to higher multibinding compounds of this invention containing from 5-10 ligands. However, for multibinding agents attached to a central linker such as an aryl, cycloalkyl or heterocyclyl group, or a crown compound, there is a self- evident constraint that there must be sufficient attachment sites on the linker to

accommodate the number of ligands present; for example, a benzene ring could not accommodate more than 6 ligands, whereas a multi-ring linker (e. g., biphenyl) could accommodate a larger number of ligands.

The formula (L) p (X) q is also intended to represent a cyclic compound of formula (-L-X-) n, where n is 2-10.

All of the above variations are intended to be within the scope of the invention defined by the formula (L) p (X) q. Examples of bivalent and higher-order valency compounds of this invention are provided in Figures 4A to 4D.

With the foregoing in mind, a preferred linker may be represented by the following formula: -X'-Z-(Y'-Z)m-Y"-Z-X'- in which: m is an integer of from 0 to 20; X'at each separate occurrence is-O-,-S-, -S (O)-,-S (0) 2-,-NR-,-N+ R R-,-C (O)-,-C (O) O-,-C (O) NH-,-C (S),-C (S) O-,-C (S) NH- or a covalent bond, where R and R at each separate occurrence are as defined below for R' and R" ; Z is at each separate occurrence selected from alkylene, substituted alkylene, alkylalkoxy, cycloalkylene, substituted cycloalkylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, cycloalkenylene, substituted alkenylene, arylene, substituted arylene, heteroarylene, heterocyclene, substituted heterocyclene, crown compounds, or a covalent bond; Y'and Y"at each separate occurrence are selected from the group consisting of

-S-S-or a covalent bond; in which: n is 0,1 or 2; and R'and R"at each separate occurrence are selected from hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl or heterocyclic.

Additionally, the linker moiety can be optionally substituted at any atom therein by one or more alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl and heterocyclic group.

As indicated above, the simples (and preferred) construct is a bivalent compound which can be represented as L-X-L, where L is an angiotensin receptor ligand that is the same or different at each occurrence, and X is the linker. Accordingly, examples of the preparation of a bivalent ligand are given below as an illustration of the manner in which multibinding compounds of Formula I are obtained.

The reaction schemes that follow illustrate preferred linking strategies for linking biphenyltetrazole derivatives (e. g., losartan, valsartan, irbesartan candesartan, tasosartan, and ripisartan) classes of angiotensin receptor modulators. These strategies are intended to apply as well to any angiotensin receptor ligand that includes, or can be functionalized with groups compatible with the chosen linker (e. g., eprosartan, telmisartan or peptide receptor antagonists such as Saralasin (Sarl, Ala8-angiotensin Il)).

As was previously discussed, the linker or linkers can be attached to different positions on the ligand molecule to achieve different orientations of the ligand domains and thereby facilitate multivalency. For example, the positions that are potentially available for linking a biphenyltetrazole derivative such as losartan are indicated in Figures 5A and 5B.

Representative multivalomers using these positions are shown in Figures 5C and 5D.

Certain angiotensin receptor ligands may be chiral and exhibit stereoselectivity.

The most active enantiomers are preferably used as ligands in the multibinding compounds of this invention. The chiral resolution of enantiomers is accomplished by well known procedures that result in the formation of diastereomeric derivatives or salts, followed by conventional separation by chromatographic procedures or by fractional crystallization (see,<BR> e. g., Bossert, et al., Angew. Chem. Int. Ed., 20,762-769,1981 and U. S. Patent No.

5,571,827 and references cited therein). Single stereoisomers can also be obtained by stereoselective synthesis.

The ligands are covalently attached to the linker using conventional chemical techniques. The reaction chemistries resulting in such linkage are well known in the art and involve the coupling of reactive functional groups present on the linker and ligand. In some cases, it may be necessary to protect portions of the ligand that are not involved in linking reactions. Protecting groups for this purpose are well known in the art and are indicated generally in the reaction schemes by the symbols PG and PG'.

Preferably, the reactive functional groups on the linker are selected relative to the functional groups on the ligand that are available for coupling, or can be introduced onto the ligand for this purpose. In some embodiments, the linker is coupled to ligand precursors, with the completion of ligand synthesis being carried out in a subsequent step.

Where functional groups are lacking, they can be created by suitable chemistries that are described in standard organic chemistry texts such as J. March, Advanced Organic Chem., 4* Ed. (Wiley-Interscience, N. Y., 1992). Examples of the chemistry for connecting ligands by a linker are shown in Figure 6, where Rt and W represent a ligand and/or the linking group. One skilled in the art will appreciate that synthetically equivalent coupling reactions can be substituted for the reactions illustrated herein.

The linker to which the ligands or ligand precursors are attached comprises a"core" molecule having two or more functional groups with reactivity that is complementary to that of the functional groups on the ligand. Figure 3 illustrates the diversity of"cores"that are useful for varying the linker size, shape, length, orientation, rigidity, acidity/basicity, hydrophobicity/hydrophilicity, hydrogen bonding characteristics and number of ligands connected. This pictorial representation is intended only to illustrate the invention, and not to limit its scope to the structures shown. In the Figures and reaction schemes that follow, a solid circle is used to generically represent a core molecule. The solid circle is equivalent to a linker as defined above after reaction.

The preferred compounds of Formula I are bivalent. Accordingly, and for the purpose of simplicity, the figures and reaction schemes. below illustrate the synthesis of bivalent angiotensin receptor modulators. It should be noted, however, that the same techniques can be used to generate higher order multibinding compounds, i. e., the comnpounds of the invention where p is 3-10.

Reactions performed under standard amide coupling conditions are carried out in an inert polar solvent (e. g., DMF, DMA) in the presence of a hindered base (e. g., TEA, DIPEA) and standard amide coupling reagents (e. g., DPPA, PyBOP, HATU, DCC).

Several methods for preparing bivalent BPT compounds, as exemplified here for losartan and structurally analogous molecules, are illustrated in the reaction schemes shown in Figures 6A-6C and, described in detail in Examples 1-3.

Functional groups are those groups within the drug (pharmacophoric building blocks) that may be exploited for multivalomer formation. For example, the tetrazole NH or the primary hydroxyl of losartan may be used. Similarly, a functional group may be introduced to facilitate multivalomer construction. For example, such a functional group may be introduced into the aryl ring of the biaryl functionality of losartan or the Cl of the imidazole may be replaced with a functional group that could be used for the construction of multivalomers. The structures in Figure 7 exemplify different valencies of the multivalomers of losartan that may be used. Tetrazole linked dimers, trimers and tetramers are exemplified. These are all homovalomers using the same point of attachment within the ligand.

Figure 8 presents a grouping of framework cores which may be used in the losartan multivalomers of the present invention. The framework core may play an important role in governing the spatial, physicochemical and pharmacological profiles of these multivalomers. A similar approach would be used to expand to higher valency multivalomers, different points of attachment, different pharmacophores and alternative relative pharmacophore orientation within the multivalomer.

Figure 9 relates to possible differing orientations of binding elements within the multivalomer, and exemplifies the use of different points of attachment for one or more of the pharmacophoric building blocks within the multivalomer. Figure 10 illustrates heterovalomers that may be used as AT, antagonists. The combination presented has focused on the losartan and valsartan, but other ligands may be used. As in Figure 9, a similar approach would be used to expand to higher valency multivalomers, different points of attachment, different ligands and alternative relative ligand orientation within the multivalomer. Also, framework cores with differing physical and pharmacological profiles could be used.

The strategies for preparing compounds of Formula I discussed above involve coupling the ligand directly to a homobifunctional core. Another strategy that can be used with all ligands, and for the preparation of both bivalent and higher order multibinding compounds, is to introduce a'spacer'before coupling to a central core. Such a spacer can itself be selected from the same set as the possible core compounds. This linking strategy would use starting materials prepared as described above.

Compounds of Formula I of higher order valency, i. e., p > 2, can be prepared by simple extension of the above strategies. Compounds are prepared by coupling ligands to a central core bearing multiple functional groups. The reaction conditions are the same as described above for the preparation of bivalent compounds, with appropriate adjustments made in the molar quantities of ligand and reagents.

Ligands may also be coupled to a polypeptide core with a sidechain spacer. Solid phase peptide synthesis can be used to produce a wide variety of peptidic core molecules.

Techniques well-known to those skilled in the art (including combinatorial methods) are used to vary the distance between ligand attachment sites on the core molecule, the number of attachment sites available for coupling, and the chemical properties of the core molecule.

Orthogonal protecting groups are used to selectively protect functional groups on the core <BR> <BR> molecule, thus allowing ancillary groups to be inserted into the linker of the multibinding compound and/or the preparation of"heterovalomers" (i. e., multibinding compounds with nonidentical ligands).

All of the synthetic strategies described above employ a step in which the ligand, attached to spacers or not, is symmetrically linked to functionally equivalent positions on a central core. Compounds of Formula I can also be synthesized using an asymmetric linear approach. This strategy is preferred when linking two or more ligands at different points of connectivity or when preparing heterovalomers.

Isolation and Punfication of the Compounds Isolation and purification of the compounds and intermediates described herein can be effected, if desired, by any suitable separation or purification such as, for example, filtration, extraction, crystallization, column chromatography, thin-layer chromatography, thick-layer chromatography, preparative low or high-pressure liquid chromatography or a combination of these procedures. Characterization of preferably by NMR and mass spectroscopy.

Utility and Testmg The multibinding compounds of this invention can be used to modulate angiotensin receptors in various tissues including heart, kidney, and brain. They will typically be used for the treatment of diseases and conditions in mammals that involve or are mediated by

angiotensin receptors, such as hypertension, congestive heart failure, renal insufficiency, diabetic neuropathy and the like.

The multibinding compounds of this invention are tested in well-known and reliable assays and their activities are compared with those of the corresponding unlinked (i. e., monovalent) ligands.

Binding affimty to receptors The binding affinity is determined by a radioligand competitive inhibition assay.

The ability of the present compounds to compete with rlfllosartan or a similar radioactive ligand in binding to high-and low-affinity binding sites in isolated adrenal membrane preparations or in human uterus or rat vascular smooth muscle cells in culture11, 23, 24 34, is measured in vitro. The binding affinity, calculated from competitive curves, is compared with that of the monovalent ligand and/or monovalent linker-ligand conjugate.

In vitro function Functional activity is measured by the ability of the present compounds to inhibit the Angiotensin II-induced 45Ca2+ efflux from rat or human vascular smooth muscle cells30>0.

Ex vivo Ex vivo functional activity is determined in the rabbit aorta and is measured by the ability to antagonise the functional contractile response to angiotensin II in a dose- dependent manner to provide a pA2 value for the angiotensin II antagonis30,30.

Antihypertensiveeffect Antihypertensive effect of compounds of this invention may be determined in vivo in annal models of renin-dependent hypertension 35t36 including conscious renal artery ligated hypertensive rats9, or in spontaneously hypertensive rats37. A reduction in mean arterial

pressure is measured. The lack of effect of angiotensin II on blood pressure in conscious normotensive rats may also be measured, since antagonists such as losartan prevent pressor response to angiotensin II in this model31.

Effect on congestive heart failure The effect of compounds of this invention on congestive heart failure may be determined in vivo in pigs3S 39 where heart failure is induced by serial myocardial infarctions followed by rapid ventricular pacing40. The effects of the compounds on cardiac output, systemic vascular resistance, pulmonary vascular resistance, neurohormonal system activity and myocardial blood flow distribution are measured both at rest and under exercise.

Effect on renal insufficiency The effects of compounds of this invention on renal insufficiency may be determined in vivo in rats with either reduced renal mass41 or reduced nephron number42, or in MWF/Ztm rats43. Changes in urinary protein excretion, urine osmolality, systemic blood pressure, and kidney morphology are monitored.

Effect on diabetic neuropathy The effect of compounds of this invention on diabetic neuropathy may be determined in vivo using either streptozotocin-diabetic rats or Sprague-Dawley diabetic rats.

Changes in nerve function, capillary density, and blood flow is measured in treated and non-treated animals44.

Combmatonal Libraries The methods described above lend themselves to combinatorial approaches for identifying multimeric compounds which possess multibinding properties for angiotensin receptors.

Specifically, factors such as the proper juxtaposition of the individual ligands of a multibinding compound with respect to the relevant array of binding sites on a target or targets is important in optimizing the interaction of the multibinding compound with its target (s) and to maximize the biological advantage through multivalency. One approach is to identify a library of candidate multibinding compounds with properties spanning the multibinding parameters that are relevant for a particular target. These parameters include: (1) the identity of ligand (s), (2) the orientation of ligands, (3) the valency of the construct, (4) linker length, (5) linker geometry, (6) linker physical properties, and (7) linker chemical functional groups.

Libraries of multimeric compounds potentially possessing multibinding properties (i. e., candidate multibinding compounds) and comprising a multiplicity of such variables are prepared and these libraries are then evaluated via conventional assays corresponding to the ligand selected and the multibinding parameters desired. Considerations relevant to each of these variables are set forth below: Selection of ligand (s) A single ligand or set of ligands is (are) selected for incorporation into the libraries of candidate multibinding compounds which library is directed against a particular biological target or targets. The only requirement for the ligands chosen is that they are capable of interacting with the selected target (s). Thus, ligands may be known drugs, modified forms of known drugs, substructures of known drugs or substrates of modified forms of known drugs (which are competent to interact with the target), or other compounds. Ligands are preferably chosen based on known favorable properties that may be projected to be carried over to or amplifie in multibinding forms. Favorable properties include demonstrated safety and efficacy in human patients, appropriate PK/ADME profiles, synthetic accessibility, and desirable physical properties such as solubility, logP, etc. However, it is crucial to note that ligands which display an unfavorable property from

among the previous list may obtain a more favorable property through the process of multibinding compound formation; i. e., ligands should not necessarily be excluded on such a basis. For example, a ligand that is not sufficiently potent at a particular target so as to be efficacious in a human patient may become highly potent and efficacious when presented in multibinding form. A ligand that is potent and efficacious but not of utility because of a non-mechanism-related toxic side effect may have increased therapeutic index (increased potency relative to toxicity) as a multibinding compound. Compounds that exhibit short in vivo half-lives may have extended half-lives as multibinding compounds. Physical properties of ligands that limit their usefulness (e. g., poor bioavailability due to low solubility, hydrophobicity, hydrophilicity) may be rationally modulated in multibinding forms, providing compounds with physical properties consistent with the desired utility.

Orientation: selection of ligand attachment pomts and linking chemistry Several points are chosen on each ligand at which to attach the ligand to the linker.

The selected points on the ligand/linker for attachment are functionalized to contain complementary reactive functional groups. This permits probing the effects of presenting g the ligands to their receptor (s) in multiple relative orientations, an important multibinding design parameter. The only requirement for choosing attachment points is that attaching to at least one of these points does not abrogate activity of the ligand. Such points for attachment can be identified by structural information when available. For example, inspection of a co-crystal structure of a protease inhibitor bound to its target allows one to identify one or more sites where linker attachment will not preclude the enzyme: inhibitor interaction. Alternatively, evaluation of ligand/target binding by nuclear magnetic resonance will permit the identification of sites non-essential for ligand/target binding. See, for example, Fesik, et al., U. S. Patent No. 5,891,643. When such structural information is not available, utilization of structure-activity relationships (SAR) for ligands will suggest positions where substantial structural variations are and are not allowed. In the absence of both structural and SAR information, a library is merely selected with multiple points of

attachment to allow presentation of the ligand in multiple distinct orientations. Subsequent evaluation of this library will indicate what positions are suitable for attachment.

It is important to emphasize that positions of attachment that do abrogate the activity of the monomeric ligand may also be advantageously included in candidate multibinding compounds in the library provided that such compounds bear at least one ligand attached in a manner which does not abrogate intrinsic activity. This selection derives from, for example, heterobivalent interactions within the context of a single target molecule. For example, consider a receptor antagonist ligand bound to its target receptor, and then consider modifying this ligand by attaching to it a second copy of the same ligand with a linker which allows the second ligand to interact with the same receptor molecule at sites proximal to the antagonist binding site, which include elements of the receptor that are not part of the formal antagonist binding site and/or elements of the matrix surrounding the receptor such as the membrane. Here, the most favorable orientation for interaction of the second ligand molecule with the receptor/matrix may be achieved by attaching it to the linker at a position which abrogates activity of the ligand at the formal antagonist binding site. Another way to consider this is that the SAR of individual ligands within the context of a multibinding structure is often different from the SAR of those same ligands in momomeric form.

The foregoing discussion focused on bivalent interactions of dimeric compounds bearing two copies of the same ligand joined to a single linker through different attachment points, one of which may abrogate the bindinglactivity of the monomeric ligand. It should also be understood that bivalent advantage may also be attained with heterodimeric constructs bearing two different ligands that bind to common or different targets. For <BR> <BR> <BR> example, a 5HT4 receptor antagonist and a bladder-selective muscarinic M3 antagonist may be joined to a linker through attachment points which do not abrogate the binding affinity of the monomeric ligands for their respective receptor sites. The dimeric compound may

achieve enhanced affinity for both receptors due to favorable interactions between the 5HT4 ligand and elements of the M3 receptor proximal to the formal Mg antagonist binding site and between the M3 ligand and elements of the SHT4 receptor proximal to the formal 5HT4 antagonist binding site. Thus, the dimeric compound may be more potent and selective antagonist of overactive bladder and a superior therapy for urinary urge incontinence.

Once the ligand attachment points have been chosen, one identifies the types of chemical linkages that are possible at those points. The most preferred types of chemical linkages are those that are compatible with the overall structure of the ligand (or protected forms of the ligand) readily and generally formed, stable and intrinsically inocuous under typical chemical and physiological conditions, and compatible with a large number of available linkers. Amide bonds, ethers, amines, carbamates, ureas, and sulfonamides are but a few examples of preferred linkages.

Linkers: spammrelevantmultihmdmparameters through selection of valency. linker length, linker geometry. rigidity, physical properties, and chemical functional groups In the library of linkers employed to generate the library of candidate multibinding compounds, the selection of linkers employed in this library of linkers takes into consideration the following factors: Valencv. In most instances the library of linkers is initiated with divalent linkers.

The choice of ligands and proper juxtaposition of two ligands relative to their binding sites permits such molecules to exhibit target binding affinities and specificities more than sufficient to confer biological advantage. Furthermore, divalent linkers or constructs are also typically of modest size such that they retain the desirable biodistribution properties of small molecules.

Linker length. Linkers are chosen in a range of lengths to allow the spanning of a range of inter-ligand distances that encompass the distance preferable for a given divalent interaction. In some instances the preferred distance can be estimated rather precisely from high-resolution structural information of targets, typically enzymes and soluble receptor targets. In other instances where high-resolution structural information is not available (such as 7TM G-protein coupled receptors), one can make use of simple models to estimate the maximum distance between binding sites either on adjacent receptors or at different locations on the same receptor. In situations where two binding sites are present on the same target (or target subunit for multisubunit targets), preferred linker distances are 2-20 A, with more preferred linker distances of 3-12 A. In situations where two binding sites reside on separate (e. g., protein) target sites, preferred linker distances are 20-100 A, with more preferred distances of 30-70 A.

Linker geometry and rigidity. The combination of ligand attachment site, linker length, linker geometry, and linker rigidity determine the possible ways in which the ligands of candidate multibinding compounds may be displayed in three dimensions and thereby presented to their binding sites. Linker geometry and rigidity are nominally determined by chemical composition and bonding pattern, which may be controlled and are systematically varied as another spanning function in a multibinding array. For example, linker geometry is varied by attaching two ligands to the ortho, meta, and para positions of a benzene ring, or in cis-or trans-arrangements at the 1,1- vs. 1,2- vs. 1,3- vs. 1,4- <BR> <BR> <BR> <BR> positions around a cyclohexane core or in cis-or trans-arrangements at a point of ethylene unsaturation. Linker rigidity is varied by controlling the number and relative energies of different conformational states possible for the linker. For example, a divalent compound bearing two ligands joined by 1,8-octyl linker has many more degrees of freedom, and is therefore less rigid than a compound in which the two ligands are attached to the 4,4' positions of a biphenyl linker.

Linker physical properties. The physical properties of linkers are nominally determined by the chemical constitution and bonding patterns of the linker, and linker physical properties impact the overall physical properties of the candidate multibinding compounds in which they are included. A range of linker compositions is typically selected to provide a range of physical properties (hydrophobicity, hydrophilicity, amphiphilicity, polarization, acidity, and basicity) in the candidate multibinding compounds. The particular choice of linker physical properties is made within the context of the physical properties of the ligands they join and preferably the goal is to generate molecules with <BR> <BR> <BR> favorable PK/ADME properties. For example, linkers can be selected to avoid those that are too hydrophilic or too hydrophobic to be readily absorbed and/or distributed in vivo.

Lmker chemical functional groups. Linker chemical functional groups are selected to be compatible with the chemistry chosen to connect linkers to the ligands and to impart the range of physical properties sufficient to span initial examination of this parameter.

Combinatonal synthesis Having chosen a set of n ligands (n being determined by the sum of the number of different attachment points for each ligand chosen) and m linkers by the process outlined above, a library of (n!) m candidate divalent multibinding compounds is prepared which spans the relevant multibinding design parameters for a particular target. For example, an <BR> <BR> <BR> array generated from two ligands, one which has two attachment points (Al, A2) and one which has three attachment points (B1, B2, B3) joined in all possible combinations provide for at least 15 possible combinations of multibinding compounds: Al-Al A1-A2 Al-Bl A1-B2 A1-B3 A2-A2 A2-B1 A2-B A2-B3 B1-B1 B1-B2 B1-B3 B2-B2 B2-B3 B3-B3

When each of these combinations is joined by 10 different linkers, a library of 150 candidate multibinding compounds results.

Given the combinatorial nature of the library, common chemistries are preferably used to join the reactive functionalies on the ligands with complementary reactive functionalities on the linkers. The library therefore lends itself to efficient parallel synthetic methods. The combinatorial library can employ solid phase chemistries well known in the art wherein the ligand and/or linker is attached to a solid support. Alternatively and preferably, the combinatorial libary is prepared in the solution phase. After synthesis, candidate multibinding compounds are optionally purified before assaying for activity by, for example, chromatographic (e.g., HPLC). af'caIe Various methods are used to characterize the properties and activities of the candidate multibinding compounds in the library to determine which compounds possess multibinding properties. Physical constants such as solubility under various solvent conditions and logD/clogD values can be determined. A combination of NMR spectroscopy and computational methods is used to determine low-energy conformations of the candidate multibinding compounds in fluid media. The ability of the members of the library to bind to the desired target and other targets is determined by various standard methods, which include radioligand displacement assays for receptor and ion channel targets, and kinetic inhibition analysis for many enzyme targets. In vitro efficacy, such as for receptor agonists and antagonists, ion channel blocks, and antimicrobial activity, can also be determined. Pharmacological data, including oral absorption, everted gut penetration, other pharmacokinetic parameters and efficacy data can be determined in appropriate models. In this way, key structure-activity relationships are obtained for multibinding design parameters which are then used to direct future work.

The members of the library which exhibit multibinding properties, as defined herein, can be readily determined by conventional methods. First those members which exhibit multibinding properties are identified by conventional methods as described above including conventional assays (both in vitro and in vivo).

Second, ascertaining the structure of those compounds which exhibit multibinding properties can be accomplished via art recognized procedures. For example, each member of the library can be encrypted or tagged with appropriate information allowing determination of the structure of relevant members at a later time. See, for example, Dower, et al., International Patent Application Publication No. WO 93/06121; Brenner, et al., Proc. Natl. Acad. Sci., USA, 89: 5181 (1992); Gallop, et al., U. S. Patent No.

5,846,839; each of which are incorporated herein by reference in its entirety.

Alternatively, the structure of relevant multivalent compounds can also be determined from soluble and untagged libaries of candidate multivalent compounds by methods known in the art such as those described by Hindsgaul, et al., Canadian Patent Application No.

2,240,325 which was published on July 11,1998. Such methods couple frontal affinity chromatography with mass spectroscopy to determine both the structure and relative binding affinities of candidate multibinding compounds to receptors.

The process set forth above for dimeric candidate multibinding compounds can, of course, be extended to trimeric candidate compounds and higher analogs thereof.

Follow-up synthesis and analysis of additional array (s) Based on the information obtained through analysis of the initial library, an optional component of the process is to ascertain one or more promising multibinding"lead" compounds as defined by particular relative ligand orientations, linker lengths, linker geometries, etc. Additional libraries can then be generated around these leads to provide for further information regarding structure to activity relationships. These arrays typically

bear more focused variations in linker structure in an effort to further optimize target affinity and/or activity at the target (antagonism, partial agonism, etc.), and/or alter physical properties. By iterative redesign/analysis using the novel principles of multibinding design along with classical medicinal chemistry, biochemistry, and pharmacology approaches, one is able to prepare and identify optimal multibinding compounds that exhibit biological advantage towards their targets and as therapeutic agents.

To further elaborate upon this procedure, suitable divalent linkers include, by way of example only, those derived from dicarboxylic acids, disulfonylhalides, dialdehydes, diketones, dihalides, diisocyanates, diamines, diols, mixtures of carboxylic acids, sulfonylhalides, aldehydes, ketones, halides, isocyanates, amines and diols. In each case, the carboxylic acid, sulfonylhalide, aldehyde, ketone, halide, isocyanate, amine and diol functional group is reacted with a complementary functionality on the ligand to form a covalent linkage. Such complementary functionality is well known in the art as illustrated in the following table: COMPLEMENTARY BINDING CHEMISTRIES First Reactive Group Second Reactive Group Linkage hydroxyl isocyanate urethane amine epoxide P-hydroxy sulfonyl halide amine sulfonamide carboxyl acid amine amide hydroxyl alkyl/aryl halide ether aldehyde amine/NaCNBH4 ketone amine/NaCNBH4 amine amine isocyanate carbamate Exemplary linkers include the following linkers identified as X-1 through X-418 as set forth below:

Diacide Ho 0 OH HO HO p ( -S S 0 X-21 HO H X-22 0 O 0 HO OH 0 0 OH OU X-2J X-24 0 0 0 ho oh -2J O X-25 OH 0 D ,. -0 OH X-26 OH ° °\4 0-- Choral X-27 S o HO 0 0 HO 0 OH l l0 OH 0 CH 0 2 0 X-29 OH X-28 X-30 ou 0 i HO X31 HO o 0 0 HO N f OH Chirol -74-X-J2 . 74- -J2 \ OH 0 0 0 ho HO N HO H 0 X-34 X-35 Chirol Chral HO 0 0- 0 -0 0 0 0 HO F F X_ 3 OH X-38 X-36 OH 0 0 OU HO DCH3 S-N HO iH3 0 0 i i chiral ""fT 0 \\ CHj OH X-39 0 X-40N\ X-41 H3C CH3 0 tk < OH 0 OH HO CHj HA OH X 43 H3C QH X-42 X-44 p ou HO 0 HD',, 0 0 HO HO I 0 0 oh 0 0 S pOH X-45 X-46 Ch H3C OH 9 X-47 X-48 HO 0 0 CHj OH X-49 F HOA X52 OU OU ;-"° Choral Ho- F X-51 Chirol F F X-52 X-50 H3N HN 0 5,//S 0 HN 0-1-1-HO s 0 OH OH OH X-55 HO Chiral HO Chiral X-53 X-54 0 oh oh OH OH oh ho ou- 0 3 OH Chiral n X-58 H3b X-56 0 0 '-N OH HO HA ho 0 0 Chiral X-60 X-59 0 o, O OH eOH n HO 0 choral X-61 X-62 H3C CH3 H3C Q 0 N ' H CH3 H3 C OH o,, HO S S 0 HO 0 X-63 Chiral HO X-65 o X-64 0 0 OH 0 HOt = Ho HO X-67X66 OH HO 05 0 0 0 0 N HO'OH 0 0 HOH \ l S 0 X-68 Lu N Ho chiral X-69 X-70 0 OH 0 0 HO HO OH 0 FFFFFFFF p D l'''pH Chirol Sl HO FFFFFFFF X-71 X-72 X-73 HO HO Q 0H HO 0 0 N JJs < Co > OH X75 X76 X-74 0 0 OH OH 0 0 H3 C HO HO X-77 X-78 0 CH HO OH 0 X-79 Hj C 0 0 H OH 0 oNX N )) CH3 0H p, 3 0 OH X-80 X-81 O 0 OH OH 0 <9. HO NEZ OH HO OH HO O 0 0 Chirol X-83 X-84 X-82 o 0 HO X-85 OH CH3 OH H . /p/V,,,) N OH O 0 0 HO-..OH ..,, H H Chrol X-88 HO 0 X-86 H X-87 HO, 0 OH 0 OH 0 N H,,,"nNN 0 ' OH, HO\ 0 X-90 w o X-89 0 0 oh oh ou N,,..S S0 I OH HD X-93 S X-94 CH3 OH Chiral X-92 0 O FF FF FF FF FF 0 0 OH HO JJk JLo AAAAA" O OH g OH FF FF FF FF FF ° 0 OH X-97 X-95 x-96 0 0 OH JW N OH OH 0 f l OH CHj CHj Chiral I 3 ChiralBr X-111 Chiral X-110 X-112 0 0 OH OH 0 0 HO pH 0H 0 ; 0 OH 0 HO 0 OH 'OH 0 0. . X-114 HO'OH HQ OH X-113 Chiral X-115 o ß 0 HO N N, N OH 0 pf-/OH -y o"' OH HO OH HD p 0 OH 0,,,0 OH HO 0 s OH OH U X-120 X-119 0 0 OH 0 II N HO HO N-1 OH X-121 0 X-122 HO p I OH HO 0 S-S OH OH pH X 1 4 0 H2N X-125 X-126 Chirol X-123 CIIRCTITI ITC CLIGCT/QI11 F 7R OH 0 OH litHO t Ó > O X-128 /ON N ON OH 0/v 0 9 O <0 < 0 Chiral 0 X-129 X-127 HO 0 0 0 0 0OH OH OH l OH ? HO 0 HO OH HO 0 X-131 JL 'Y 0 -7J/ X-130 X-132 Disulfonyl Halides OZ=S S-C/ 0 X-l34 p o 0 QS-F 0k ( ; 1 0 0 lu4 X-133'o X-135 fez x-111 0 x-lj5 F 0 CH3 CI p 0 Cl 0 Ft X-lJ7 C/0/ X-136 N N S pS 0S. CI \0 cri N NaS's S' X-139 X-140 0 cri X-141 F ouzo H3 CHj 0 c 0 Sv rrV4/- ". zip I oSF CH3 0 X-144 H2 C 0//X-14J X-142 0 FS 0 Cl 0 F,// il s tir w0 N N 0. 0 X-146 X-145 3 0 i p 0 0 CI CI ii Os ii SCISSC S w Sv 0-S50 0 C/0 0 0 0 0 H3C CH, 3 w 0 CH3 X-148 HO 0 X-147 X-149 X-150 w o c. T 0 0 Y-79 !' 0 0 7 X-152 0 0 X-) 51 o Dioldehydes, I 4 X-15 x-151 u 0 C3 i 0 w X-156 O X155 0 < 1 3 0 o, I I N I X157 X158 vo ° 0 O X160 X 0 0 0 1 CH rr X-159 3 0 1 N 0 o nu 1-- I 1 0 X-) 64 X-161p X-162 X_ 3 CH3 r i I 0 0 w w X-166 0 OH X-165 o X-167 X-Z68 X X °@° 0 HO X-168 5 , w ( 0 X-169 X170 OH H3C X-171 lUl 9040 /S X-172 HX-'7 X-174 i I C C N C CH3 X-177 Dihalides CI 0""\o X-176 X-175 BrBr Br Br OH OH X-178 X-179 X-180 Diisocyonates 0 o/« N x N »/sNXN z 0 N N -215 0 X-216 0 \\ wNf N<N N ,, oxo / Hic-0 0-CHj X218 0 4 X-217 OFF 0===N F F °N N 0 0* X-219 X-220 o X-221 0 0 0 N 0 \ l/ 0 Br CHj0 N N CHj X-222 H3C CH3 X-224 0 X-223, ° N II i o N X-227 I 4 X-226 0 ,, CH3 CH3 0 /i N p I I D 0 \ i X-225 X-229 X-J? J? 5 c/ X-228 Cl CH3 N 0 N, C/ Cl) : :N, CI CH3 N I X-230 0 X-231 X-232 j 0 0 N 0 0 hic 0 N HJC Nit N H C N 0 0/X X-246 I X-247 X-248 Diamines XN~OmO XN » X-249 IvN N X-251 H2N 2 cl N N N X-251 CH, 3 , X-250 2 N t0 X252 CH3H2Ns l l2/lNH2 H2N w H N3 N H2N 3N X-255 X-257 0 NH2 H2NN/ X-255 X-257 /I I H2N NH2 H2N NH 2 H2N NH2 X-258 X-259 X-260 H3CN NCH H2NONH2 3 Nu N 2 HZN NH2 nu // X-26JnX-264 0 l \ p l S l \ p l H2N NH2 H N NH2 X-304 2 X-303 CH m 2 oX sNH2 C H2N NH2 Nu 2 X305 < N, CH3 X307 Chirol X-306 NH2 NH2 NH han HAN CHj 2 j CH3 CH3 X-309 X-308 X-ill 'o o H2N NH2 X-311 X-310 NH2 CH3 H2NH3/w NH2 w I/ CH3 CH3 NU Chirol X-313 X-312 X-314 H2N NH2 CH3 X-15 H3NN X-J 16 Hj C--CHj sN<\NtOH X 3 OU N N OH H3 C OH X-317 I = N Chirol CH3 X-J18 H2 N H2 N x-jl9 Hj C Cl2 3 3 X-320 HC N H2 N X-321 X-322 Hj CN N"% CHj HjC"I N-'- NCHj H2N NH2 -J27 - X-323 X-324 X-325 Diols CH3 HO v_ Br Br HO p 0 X-327 Br Br OH X-326 nN wOH , OH H0 eN ß ° X-328 S X-329 N -j2<9 rj -jj? -78n-OH Dithiols HS-o HS SH HS 0 1 SH, X-393 HS X-392 X-394 SH SH SH SH H e SH X-395 I I H3C CH3" X-396 SH X-397 0 SH H. 5 VS OH HS., a SH X-398 X-400 CHj , CH3 HS- HSSH HS w S---SH o X-403 SH X-402 X-401 0 0 HS SH HS SH HSNSH \ l X-404 X-405 X-406 HSs"5, ...,. SH HS SH HS SH X-407 X-408 X-409 OH SH OH OH SH SU OH OH OH SH X-412 X-413 X-4) 0 X 411 SH OH HS SH = SU OH SH Ch iral OH X-414 X-415 X-416 HS SH HS SH X-418 X-418 -/7-78s-

Representative ligands for use in this invention include, by way of example, sartan ligands (e. g., losartan compounds and valsartan compounds), designated L-1.

Combinations of ligands (L) and linkers (X) per this invention include, by way example only, homo-and hetero-dimers wherein a first ligand is selected from L-1 and the second ligand and linker is selected from the following: <BR> <BR> <BR> <BR> <BR> <BR> L-1/X-1-L-1/X-2-L-1/X-3-L-1/X-4-L-1/X-5-L-1/X-6-<BR> <BR> <BR> <BR> <BR> L-1/X-7-L-1/X-8-L-1/X-9-L-1/X-10-L-1/X-11-L-1/X-12-<BR> ; <BR> <BR> <BR> <BR> L-1/X-13-L-1/X-14-L-l/X-15-L-1/X-16-L-1/X-17-L-1/X-18-<BR > <BR> <BR> <BR> <BR> L-1/X-19-L-1/X-20-L-1/X-21-L-1/X-22-L-1/X-23-L-1/X-24-<BR > <BR> <BR> <BR> <BR> L-1/X-25-L-1/X-26-L-1/X-27-L-1/X-28-L-1/X-29-L-1/X-30-<BR > <BR> <BR> <BR> <BR> L-1/X-31-L-1/X-32-L-1/X-33-L-1/X-34-L-1/X-35-L-1/X-36- L-1/X-39-L-1/X-40-L-1/X-41-L-1/X-42-L-1/X-37-L-1/X-38- L-1/X-45-L-1/X-46-L-1/X-47-L-1/X-48-L-1/X-43-L-1/X-44- <BR> <BR> <BR> <BR> L-1/X49-L-1/X-50-L-1/X-51-L-1/X-52-L-1/X-53-L-1/X-54-<BR& gt; <BR> <BR> <BR> <BR> L-1/X-55-L-1/X-56-L-1/X-57-L-1/X-58-L-1/X-59-L-1/X-60- L-1/X-63-L-1/X-64-L-1/X-65-L-1/X-66-L-1/X-61-L-1/X-62- L-1/X-69-L-1/X-70-L-1/X-71-L-1/X-72-L-1/X-67-L-1/X-68- <BR> <BR> <BR> <BR> L-1/X-73-L-1/X-74-L-1/X-75-L-1/X-76-L-1/X-77-L-1/X-78-<BR > <BR> <BR> <BR> <BR> <BR> L-1/X-79-L-1/X-80-L-1/X-81-L-1/X-82-L-1/X-83-L-1/X-84-<BR > <BR> <BR> <BR> <BR> L-1/X-85-L-1/X-86-L-1/X-87-L-1/X-88-L-1/X-89-L-1/X-90-<BR > <BR> <BR> <BR> <BR> L-1/X-91-L-1/X-92-L-1/X-93-L-1/X-94-L-1/X-95-L-1/X-96- L-1/X-97-L-1/X-98-L-1/X-99-L-1/X-100-L-1/X-101-L-1/X-102- <BR> <BR> <BR> L-1/X-103-L-1/X-104-L-1/X-105-L-1/X-106-L-1/X-107-L-1/X-108- <BR> <BR> <BR> <BR> <BR> L-1/X-109-L-1/X-110-L-1/X-111-L-l/X-112-L-1/X-113-L-1/X-114- <BR> <BR> <BR> <BR> <BR> L-1/X-115-L-1/X-116-L-1/X-117-L-1/X-118-L-1/X-119-L-1/X-120- <BR> <BR> <BR> <BR> <BR> L-1/X-121-L-1/X-122-L-I/X-123-L-1/X-124-L-1/X-125-L-1/X-126-

L-1/X-127-L-1/X-128-L-1/X-129-L-1/X-130-L-1/X-131-L-1/X-132- <BR> <BR> <BR> <BR> <BR> L-1/X-133-L-1/X-134-L-1/X-135-L-1/X-136-L-1/X-137-L-1/X-138- <BR> <BR> <BR> <BR> <BR> L-1/X-139-L-1/X-140-L-1/X-141-L-1/X-142-L-1/X-143-L-1/X-144- <BR> <BR> <BR> <BR> <BR> L-1/X-145-L-1/X-146-L-1/X-147-L-1/X-148-L-1/X-149-L-1/X-150- <BR> <BR> <BR> <BR> <BR> L-1/X-151-L-1/X-152-L-1/X-153-L-1/X-154-L-1/X-155-L-1/X-156- <BR> <BR> <BR> <BR> <BR> L-l/X-157-L-l/X-158-L-1/X-159-L-1/X-160-L-1/X-161-L-1/X-162- <BR> <BR> <BR> <BR> <BR> L-l/X-163-L-l/X-164-L-l/X-165-L-l/X-166-L-l/X-167-L-l/X-168- <BR> <BR> <BR> <BR> <BR> L-1/X-169-L-1/X-170-L-1/X-171-L-1/X-172-L-1/X-173-L-1/X-174- L-1/X-177-L-1/X-178-L-1/X-179-L-1/X-180-L-1/X-175-L-1/X-176- L-l/X-181-L-1/X-182-L-1/X-183-L-1/X-184-L-1/X-185-L-1/X-186- L-1/X-187-L-l/X-188-L-1/X-189-L-1/X-190-L-1/X-191-L-1/X-192- <BR> <BR> <BR> L-1/X-193-L-1/X-194-L-1/X-195-L-1/X-196-L-llX-197-L-1IX-198- L-1/X-199-L-1/X-200-L-1/X-201-L-1/X-202-L-1/X-203-L-1/X-204- <BR> <BR> <BR> <BR> L-1/X-205-L-l/X-206-L-1/X-207-L-1/X-208-L-1/X-209-L-1/X-210- <BR> <BR> <BR> <BR> <BR> L-1/X-211-L-1/X-212-L-1/X-213-L-1/X-214-L-1/X-215-L-1/X-216- <BR> <BR> <BR> <BR> <BR> L-1/X-217-L-1/X-218-L-1/X-219-L-1/X-220-L-1/X-221-L-1/X-222- <BR> <BR> <BR> <BR> <BR> L-1/X-223-L-1/X-224-L-1/X-225-L-1/X-226-L-1/X-227-L-1/X-228- L-1/X-229-L-1/X-230-L-1/X-231-L-1/X-232-L-1/X-233-L-1/X-234- <BR> <BR> <BR> <BR> L-1/X-235-L-1/X-236-L-1/X-237-L-1/X-238-L-1/X-239-L-1/X-240- <BR> <BR> <BR> <BR> <BR> L-1/X-241-L-1lX-242-L-1/X-243-L-1/X-244-L-1/X-245-L-1/X-246- L-1/X-249-L-1/X-250-L-1/X-251-L-1/X-252-L-1/X-247-L-1/X-248- <BR> <BR> <BR> <BR> L-1/X-253-L-1/X-254-L-1/X-255-L-1/X-256-L-1/X-257-L-l/X-258- <BR> <BR> <BR> <BR> <BR> L-1/X-259-L-1IX-260-L-1/X-261-L-1/X-262-L-1/X-263-L-1lX-264- L-1/X-265-L-1/X-266-L-1/X-267-L-1/X-268-L-1/X-269-L-1/X-270- <BR> <BR> <BR> <BR> L-1/X-271-L-1/X-272-L-1/X-273-L-1/X-274-L-1/X-275-L-1/X-276- <BR> <BR> <BR> <BR> <BR> L-1/X-277-L-1/X-278-L-1/X-279-L-1/X-280-L-1/X-281-L-1/X-282- L-1/X-283-L-1/X-284-L-1/X-285-L-1/X-286-L-1/X-287-L-1/X-288- L-1/X-289-L-1/X-290-L-1/X-291-L-1/X-292-L-1/X-293-L-1/X-294-

L-1/X-295-L-1/X-296-L-1/X-297-L-1/X-298-L-1/X-299-L-1/X-300- <BR> <BR> <BR> <BR> <BR> <BR> L-1/X-301-L-1/X-302-L-I/X-303-L-1/X-304-L-I/X-305-L-1/X-306- L-l/X-307-L-1/X-308-L-1/X-309-L-1/X-310-L-1/X-311-L-1/X-312- <BR> <BR> <BR> <BR> L-l/X-313-L-l/X-314-L-l/X-315-L-l/X-316-L-l/X-317-L-l/X-318- <BR> <BR> <BR> <BR> <BR> <BR> L-l/X-319-L-l/X-320-L-l/X-321-L-l/X-322-L-l/X-323-L-l/X-324- <BR> <BR> <BR> <BR> <BR> L-1lX-325-L-1/X-326-L-1/X-327-L-1/X-328-L-I/X-329-L-1/X-330- L-1/X-331-L-1/X-332-L-1/X-333-L-1/X-334-L-1/X-335-L-1/X-336- L-1/X-337-L-1/X-338-L-1/X-339-L-1/X-340-L-1/X-341-L-1/X-342- <BR> <BR> <BR> <BR> L-1/X-343-L-1/X-3. 44- L-1/X-345-L-1/X-346-L-1/X-347-L-1/X-348-<BR> <BR> <BR> <BR> <BR> <BR> L-l/X-349-L-l/X-350-L-l/X-351-L-l/X-352-L-l/X-353-L-l/X-354- <BR> <BR> <BR> <BR> <BR> L-1/X-355-L-1/X-356-L-1/X-357-L-1/X-358-L-1/X-359-L-1/X-360- <BR> <BR> <BR> <BR> <BR> <BR> L-1/X-361-L-1/X-362-L-1/X-363-L-I/X-364-L-1/X-365-L-1/X-366- <BR> <BR> <BR> <BR> <BR> L-1/X-367-L-11X-368-L-1IX-369-L-11X-370-L-11X-371-L-1IX-372- L-1/X-373-L-1/X-374-L-1/X-375-L-1/X-376-L-1/X-377-L-1/X-378- <BR> <BR> <BR> <BR> L-1/X-379-L-1/X-380-L-1/X-381-L-1/X-382-L-1/X-383-L-1/X-384- <BR> <BR> <BR> <BR> <BR> <BR> L-1/X-385-L-1/X-386-L-1/X-387-L-1/X-388-L-1/X-389-L-1/X-390- L-1/X-391-L-1/X-392-L-1/X-393-L-1/X-394-L-1/X-395-L-1/X-396- L-1/X-399-L-1/X-400-L-1/X-401-L-1/X-402-L-1/X-397-L-1/X-398- L-1/X-405-L-1/X-406-L-1/X-407-L-1/X-408-L-1/X-403-L-1/X-404- L-1/X-409-L-1/X-410-L-I/X-41 1-L-1/X-412-L-1/X-413-L-1/X-414- L-1/X-417-L-1/X-418-.L-1/X-415-L-1/X-416- Pharmaceutical Fomulations When employed as pharmaceuticals, the compounds of Formula I are usually administered in the form of pharmaceutical compositions. This invention therefore provides pharmaceutical compositions which contain, as the active ingredient, one or more of the compounds of Formula I above or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients, carriers, diluents, permeation enhancers,

solubilizers and adjuvants. The compounds may be administered alone or in combination with other therapeutic agents (e. g., other antihypertensive drugs, diuretics and the like). <BR> <BR> <BR> <P>Such compositions are prepared in a manner well known in the pharmaceutical art (see,<BR> <BR> <BR> <BR> <BR> <BR> <BR> <BR> e. g., Rernington's Phann. Sci., Mack Publishing Co., Philadelphia, PA, 17'b Ed., 1985 and<BR> <BR> <BR> <BR> <BR> <BR> <BR> Modern Pham., Marcel Dekker, Inc., 3td Ed. (G. S. Banker & C. T. Rhodes, Eds.).

The compounds of Formula I may be administered by any of the accepte modes of administration of agents having similar utilities, for example, by oral, parenteral, rectal, buccal, intranasal or transdermal routes. The most suitable route will depend on the nature and severity of the condition being treated. Oral administration is a preferred route for the compounds of this invention. In making the compositions of this invention, the active ingredient is usually diluted by an excipient or enclosed within such a carrier which can be in the form of a capsule, sachet, paper or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders. Pharmaceutically acceptable salts of the active agents may be prepared using standard procedures known to those skilled in the art of synthetic organic chemistry and described, e. g., by J. March, Advanced <BR> <BR> <BR> <BR> Organic Chem. Reactions, Mechanisms and Structure, 4dl Ed. (N. Y.: Wiley-Interscience, 1992).

Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methyl cellulose. The formulations can additionally include: lubricating agents such as

talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl-and propylhydroxy-benzoates; sweetening agents; and flavoring agents.

The compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art. Controlled release drug delivery systems for oral administration include osmotic pump systems and dissolutional systems containing polymer- coated reservoirs or drug-polymer matrix formulations. Examples of controlled release systems are given in U. S. Patent Nos. 3,845,770; 4,326,525; 4,902,514; and 5,616,345.

Another preferred formulation for use in the methods of the present invention employs transdermal delivery devices ("patches"). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e. g., U. S. Patent Nos. 5,023,252; 4,992,445 and 5,001,139. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.

The compositions are preferably formulated in a unit dosage form. The term"unit dosage forms"refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient (e. g., a tablet, capsule, ampoule). The active compound is effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. Preferably, for oral administration, each dosage unit contains from 1- 1000 mg of a compound of Formula I, and for parenteral administration, preferably from 0.1 to 600 mg of a compound of Formula I or a pharmaceutically acceptable salt thereof.

It will be understood, however, that the amount of the compound actually administered will

be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered and its relative activity, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.

For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulaLton composition containing a homogeneous mixture of a compound of the present invention. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.

The tablets or pills of the present invention may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.

The liquid forms in which the novel compositions of the present invention may be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as corn oil, cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.

Compositions for inhalation or insufflation include solutions and suspensions in phannacsutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. Preferably the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions in preferably pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a face mask tent, or intermittent positive pressure breathing machine.

Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner.

The following formulation examples illustrate representative pharmaceutical compositions of the present invention.

Formulation Example 1 Hard gelatin capsules containing the following ingredients are prepared: Quantity Ingredient(mg/capsule) Active Ingredient 30.0 Starch 305.0 Magnesium stearate 5.0 The above ingredients are mixed and filled into hard gelatin capsules in 340 mg quantities.

Formulation Example 2 A tablet formula is prepared using the ingredients below:

Ingredient(mg/tablet) Active Ingredient 25.0 Cellulose, microcrystalline 200.0 Colloidal silicon dioxide 10.0 Stearic acid 5. 0 The components are blended and compressed to form tablets, each weighing 240 mg.

Formulation Example 3 A dry powder inhaler formulation is prepared containing the following components: I Active Ingredient 5 Lactose 95 The active ingredient is mixed with the lactose and the mixture is added to a dry powder inhaling appliance.

Formulation Example 4 Tablets, each containing 30 mg of active ingredient, are prepared as follows: Quantity Ingredient (mg/tablet) Active Ingredient 30.0 Starch 45. 0 Microcrystalline cellulose 35.0

Polyvinylpyrrolidone (as 10% solution in sterile water) 4.0 Sodium carboxymethyl starch 4.5 0.5Magnesiumstearate Talc 1.0 Total 120.0 The active ingredient, starch and cellulose are passed through a No. 20 mesh U. S. sieve and mixed thoroughly. The solution of polyvinylpyrrolidone is mixed with the resultant powders, which are then passed through a 16 mesh U. S. sieve. The granules so produced are dried at 50°C to 60°C and passed through a 16 mesh U. S. sieve. The sodium carboxymethyl starch, magnesium stearate, and talc, previously passed through a No. 30 mesh U. S. sieve, are then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets each weighing 120 mg.

Formulation Example 5 Capsules, each containing 40 mg of medicament are made as follows: <BR> <BR> <BR> <BR> <BR> <BR> Quantity<BR> <BR> <BR> Ingredient Imglcapsule Active Ingredient 40.0 Starch 109.0 Magnesium stearate 1.0 Total 150.0 The active ingredient, starch, and magnesium stearate are blended, passed through a No. 20 mesh U. S. sieve, and filled into hard gelatin capsules in 150 mg quantities.

Formulation Example 6 Suppositories, each containing 25 mg of active ingredient are made as follows:

Active Ingredient 25.0 mg Saturated fatty acid glycerides to 2,000.0 mg The active ingredient is passed through a No. 60 mesh U. S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimum heat necessary.

The mixture is then poured into a suppository mold of nominal 2.0 g capacity and allowed- to cool.

Formulation Example 7 Suspensions, each containing 50 mg of medicament per 5.0 mL dose are made as followsAmount Active Ingredient 50.0 mg Xanthan gum 4.0 mg Sodium carboxymethyl cellulose (11%) Microcrystalline cellulose (89%) 50.0 mg Sucrose 1.75 g Sodium benzoate 10.0 mg Flavor and Color q. v.

Purified water to 5.0 ml The active ingredient, sucrose and xanthan gum are blended, passed through a No.

10 mesh U. S. sieve, and then mixed with a previously made solution of the microcrystalline cellulose and sodium carboxymethyl cellulose in water. The sodium benzoate, flavor, and color are diluted with some of the water and added with stirring. Sufficient water is then added to produce the required volume.

Formulation Example 8<BR> <BR> <BR> <BR> <BR> <BR> <BR> <BR> <BR> Quantity<BR> <BR> <BR> hrgredient (MgLmmw Active Ingredient 15.0 mg Starch 407.0 mg Magnesium stearate 3.0 mg Total 425.0 mg The active ingredient, starch, and magnesium stearate are blended, passed through a No. 20 mesh U. S. sieve, and filled into hard gelatin capsules in 425.0 mg quantities.

Formulation Example 9 A subcutaneous formulation may be prepared as follows: in Ouantltv Active Ingredient 5.0 mg Corn Oil 1.0 mL Frequently, it will be desirable or necessary to introduce the pharmaceutical composition to the brain, either directly or indirectly. Direct techniques usually involve placement of a drug delivery catheter into the host's ventricular system to bypass the blood-brain barrier. One such implantable delivery system used for the transport of biological factors to specific anatomical regions of the body is described in U. S. Patent 5,011,472 which is herein incorporated by reference.

Indirect techniques, which are generally preferred, usually involve formulating the compositions to provide for drug latentiation by the conversion of hydrophilic drugs into lipid-soluble drugs. Latentiation is generally achieved through blocking of the hydroxy,

carbonyl, sulfate, and primary amine groups present on the drug to render the drug more lipid soluble and amenable to transportation across the blood-brain barrier. Alternatively, the delivery of hydrophilic drugs may be enhanced by intra-arterial infusion of hypertonic solutions which can transiently open the blood-brain barrier.

Synthesis Examples Example 1. (Figure 6A) Synthesis of a hydroxy linked losartan dimeric multivalomer.

A. Alkylation of the imidazole 1 to provide 2 <BR> <BR> The TBS protected imidazole 1 is prepared as in Greenlee, W. J., Biorg. Med.<BR> <BR> <BR> <BR> <BR> <P>Chem. Lett., 1993,3 (4), 557-660. The imidazole 1 (604mgs, 2mmol) is dissolved in DMF (lOmls, c. 0.2 and is then treated with NaH (48mgs, 2mmol.) and the reaction is stirred at RT for 30 minutes. The biaryl bromide (300mgs, lmmol.) is then added as a solution in DMF (5mls) and the reaction stirred for a further 60 minutes. The reaction is concentrated in vacuo. The crude reaction mixture is partitioned between ethyl acetate (25mls) and water (25mls). The organic layer is dried (MgSO4), filtered and concentrated in vacuo.

Flash chromatography provides the desired material 2.

B. Deprotection of the TBS ether 2 to provide the alcohol 3 The silyl ether 2 (1.02g, 2mmol.) is dissolved in THF (lOmls, c. 0.2M) and 1M TBAF in THF (3mls, 3mmol.) is added and the reaction is allowed to stir at room <BR> <BR> <BR> <BR> temperature for 2 hours. The reaction is concentrated in vacuo, and partitioned between ethyl acetate (25mls) and water (25mls). The organic layer is dried (MgSO4), filtered and concentrated in vacuo. The crude reaction mixture is purified by flash chromatography to provide the desired material 3.

C. Dimerization via the primary hydroxyl of 3 to provide the multivalomer 4 The primary alcohol 3 is dissolved in DMF (10mls, c. 0.2M) and cooled to OC, NaH (48mgs, 2mmol.) is added and the reaction is stirred at this temperature for 30 minutes. The dibromide (260mgs, lmmol.) is dissolved in DMF (lOmls) and is added to the alkoxide solution via syringe pump over 60 minutes. The reaction is concentrated in vacuo, and partitioned between ethyl acetate (25mls) and water (25mls). The organic layer is dried (MgSO4), filtered and concentrated m vacuo. This crude reaction mixture is purified by flash chromatography to provide the desired multivalomer 4.

D. Deprotection of the dimeric tetrazole 4 to the losartan multivalomer 5.

The dimeric protected tetrazole (1.17g, lmmol.) is dissolved in methanol (5mls, c.

0.2X) and is treated with 1M HC1 in methanol (3mls, 3mmol.) and the reaction is stirred at room temperature for 60 minutes. After this time, the reaction is concentrated in vacuo.

The reaction is partitioned between ethyl acetate (25mls) and water (25mls). The organic layer is separated, dried (MgSO, filtered and concentrated in vacuo. The crude reaction mixture is purified by flash chromatography to provide the losartan multivalomer 5.

Example 2 (Figure 6B) Synthesis of a dimeric losartan multivalomer linked through the primary hydroxyl which may be used to prepare either hydroxl linked or tetrazole linked multivalomers.

A. Alkylation of imidazole 1 The imidazole 1, (276mgs, 2mmol.) is added to a stirred solution of sodium methoxide (2mmol) in methanol (10mls) (2mmol, 46mgs of sodium dissolved in methanol) at CC. The solvent is removed in vacuo and the thus formed sodium salt of the imidazole is dissolved in DMF (10mls). The biaryl bromide (542mgs, 2mmol.) is added and the

reaction is stirred at room temperature for 12 hours. The solvent is then removed in vacuo, and the reaction partitioned between ethyl acetate (25mls) and water (25mls). The organic layer is combined and the organic layer is dried with MgSO4, the solvent in this removed in vacuo. Flash chromatography of the crude reaction mixture provides the alkylated imidazole 2.

B. AIkylation of alcohol 2 to provide dimer 3 Sodium hydride (48mgs, 2mmol.) is dissolved in DUT (lOmls) and the alcohol 2 (760mgs, 2mmol.) is added with stirring. This reaction is allowed to stir at room temperature. The benzylic dibromide (261mgs, lmmol.) in DMF is added dropwise via syringe pump over two hours. The reaction is allowed to stir at room temperature for a further two hours. The reaction is treated with aqueous NH4C1 solution and partitioned between ethyl acetate (25mls) and water (25mls). The organic layer is separated, dried with MgSO4, filtered and concentrated in vacuo. This crude reaction mixture is purified by flash chromatography to provide the pure dimer 3.

C. Conversion of the dimeric nitrile 3 to dimeric tetrazole 4 The dimer 3 (430mgs, lmmol) is dissolved in xylene (20mls) and the trimethylstannyl azide is added (615mg, 3mmol) and the reaction is heated to reflux in xylene (20mls) for 24 hrs. The solvent is removed in vacuo and the crude reaction mixture is treated with 2N NaOH in methanol (20mls) to remove the N-stannyl group. The solvent is removed in vacuo and the reaction is dissolved in water and the solution neutralized (pH=7). The product is extracted with ethyl acetate (25mls x 3). The organic layer is dried with MgS04, filtered and concentrated in vacuo. The crude reaction mixture is purified by flash chromatography. to provide the desired dimeric tetrazole 4.

Example 3. (Figure 6C) Synthesis of a losartan dimeric multivalomer linked through N3 of the tetrazole functionality using selective alkylation of the tetrazole in the presence of the primary hydroxyl.

A. Conversion of the nitrile 1 to the tetrazole 2 The biaryl nitrile 1 (380mgs, 2mmol.) prepared as above is dissolved in xylenes (lOmls) and the trimethylstannyl azide (820mgs, 4mmol.) is added. The reaction is heated at reflux for 24 hrs. The reaction is allowed to cool and the solvent is removed in vacuo.

The crude reaction mixture is treated with 1DI NaOH in methanol (20mls) to hydrolyse the N-stannyl bond. The methanol is removed in vacuo, the crude reaction mixture is dissolved in water and neutralized with 1M Hcl. The product is extracted from the aqueous phase with ethyl acetate (3 x 25mls). The organic layer is dried with MgSO4, the drying agent is filtered, and the solvent is removed in vacuo. The crude reaction mixture can be purified by flash chromatography to provide the desired tetrazole 2.

B. Conversion of the tetrazole 2 to the dimer 3 The tetrazole 2 (844mgs, 2mmol) is dissolved in DMF (5mls) and is treated with NaH (48mgs, 2mmol) and the reaction is stirred at RT for 20 minutes. The dibromide alkylating agent (260mgs, lmmol.) in DMF (10mls) is added to tetrazole solution via syringe pump over one hour. The reaction is allowed to stir at room temperature for a further hour. The reaction is concentrated in vacuo, and is partitioned between ethyl acetate (25mls) and water (25mls). The organic layer is separated, dried (MgSO4), filtered and concentrated in vacuo. The crude reaction mixture is purified by flash chromatography to provide the desired dimer 3.

While the present invention has been described with reference to the specific embodiments thereof, it should be understood by those skilled in the art that various

changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, many modifications may be made to adapt a particular situation, material, composition of matter, process, process step or steps, to the objective spirit and scope of the present invention. All such modifications are intended to be within the scope of the claims appended hereto.

All of the publications, patent applications and patents cited in this application are herein incorporated by reference in their entirety to the same extent as if each individual publication, patent application or patent was specifically and individually indicated to be incorporated by reference in its entirety.

Table 1: Anglotensin Receptors Currently Accepted Name AT1* AT2 Receptor Agonists Angiotensin II> Angiotensin III Anglotensin II=Angiotensin III CGP-42112A Receptor Antagonists (Peptide) [Sar1, Ala8]-Angiotensin II (Saralasin) [Sar1, ala8]-Anglotensin II (Saralasin) Receptor antagonists (Non-peptide) DUP-753 (Losartan) PD 123,319 E 3174 SKF-108566 (Eprosartan) TCV-116 Candesartan) L-158,809 SR 47436 (Irbesartan) CGP-48933 (Valsartan) BIBR277 (Telmesartan) GR 117239 (Zolasartan) able 2: AT1 Antagonists in Clinical Trial and/or Use<BR> Clinical Dose AT1 Affinity Bioavallability (F) Half-Life<BR> Losartan 50 mg 10.3 nM (mixed) 36% 7.6 hrs<BR> Valsartan 80 mg 50.5 nM (mixed) 23% 7.0 hrs<BR> KIrbesartan 150-300 mg 6.7 nM (mixed) 70% 6.0 hrs<BR> Candesartan 8-16 mg 2.4 nM (non-competitive) 42% 9.0 hrs<BR> Eprosartan 400-800 mg 50.5 nM (competitive) 15% 4.5 hrs Table 3: Structures of Various Angiotensin Receptor Ligands LOSARTAN (Cozaar) VALSARTAN (Diovan) IRBESARTAN CANDESARTAN (Ahcand) EPROSARTAN (Tevetan) TASOSARTAN (Verdia) TELMISARTAN RIPISARTAN Table 3: Structures of Various Angiotensin Receptor Ligands<BR> (Continued) DuP 753 Losartan L-162,313 L-163, 958 EXP-801 EXP3174