AU8585098A - Dipeptide and related compounds which inhibit leukocyte adhesion mediated by vla-4 - Google Patents

Dipeptide and related compounds which inhibit leukocyte adhesion mediated by vla-4 Download PDF

Info

Publication number
AU8585098A
AU8585098A AU85850/98A AU8585098A AU8585098A AU 8585098 A AU8585098 A AU 8585098A AU 85850/98 A AU85850/98 A AU 85850/98A AU 8585098 A AU8585098 A AU 8585098A AU 8585098 A AU8585098 A AU 8585098A
Authority
AU
Australia
Prior art keywords
substituted
heterocyclic
alkyl
aryl
sulfonyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU85850/98A
Inventor
Michael S. Dappen
Darren B. Dressen
Francine S. Grant
Louis John Lombardo
Michael A. Pleiss
Christopher M. Semko
Eugene D. Thorsett
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wyeth LLC
Elan Pharmaceuticals LLC
Original Assignee
Elan Pharmaceuticals LLC
American Home Products Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Elan Pharmaceuticals LLC, American Home Products Corp filed Critical Elan Pharmaceuticals LLC
Publication of AU8585098A publication Critical patent/AU8585098A/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06026Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atom, i.e. Gly or Ala
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06104Dipeptides with the first amino acid being acidic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06139Dipeptides with the first amino acid being heterocyclic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06139Dipeptides with the first amino acid being heterocyclic
    • C07K5/06165Dipeptides with the first amino acid being heterocyclic and Pro-amino acid; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Description

WO 99/06432 PCT/US98/15325 DIPEPTIDE AND RELATED COMPOUNDS WHICH INHIBIT LEUKOCYTE ADHESION MEDIATED BY VLA-4 CROSS-REFERENCE TO RELATED APPLICATIONS This application claims the benefit of U.S. Provisional Application No. 60/ , , which was converted pursuant to 37 C.F.R. § 1.53(c)(2)(i) from U.S. Patent Application No.08/904,417, filed July 31, 1997, the disclosure of which is incorporated herein by reference in its entirety. BACKGROUND OF THE INVENTION Field of the Invention This invention relates to compounds which inhibit leukocyte adhesion and, in particular, leukocyte adhesion mediated by VLA-4. 5 References The following publications, patents and patent applications are cited in this application as superscript numbers: 1 Hemler and Takada, European Patent Application Publication 10 No. 330,506, published August 30, 1989 2 Elices, et al., Cell, 60:577-584 (1990) 3 Springer, Nature, 346:425-434 (1990) 15 4 Osborn, Cell, 62:3-6 (1990) Vedder, et al., Surgery, 106:509 (1989) 20 6 Pretolani, et al., J. Exp. Med., 180:795 (1994) Abraham, et al., J. Clin. Invest., 93:776 (1994) WO 99/06432 PCT/US98/15325 -- 2 - 8 Mulligan, et al., J. Immunology, 150:2407 (1993) 9 Cybulsky, et al., Science, 251:788 (1991) 5 o10 Li, et al., Arterioscler. Thromb., 13:197 (1993) 11 Sasseville, et al., Am. J. Path., 144:27 (1994) 12 Yang, et al., Proc. Nat. Acad. Science (USA), 90:10494 10 (1993) 13 Burkly, et al., Diabetes, 43:529 (1994) 14 Baron, et al., J. Clin. Invest., 93:1700 (1994) 15 15 Hamann, et al., J. Immunology, 152:3238 (1994) 16 Yednock, et al., Nature, 356:63 (1992) 20 17 Baron, et al., J. Exp. Med., 177:57 (1993) 18 van Dinther-Janssen, et al., J. Immunology, 147:4207 (1991) 19 van Dinther-Janssen, et al., Annals. Rheumatic Dis., 52:672 25 (1993) 20 Elices, et al., J. Clin. Invest., 93:405 (1994) 21 Postigo, et al., J. Clin. Invest., 89:1445 (1991) 30 22 Paul, et al., Transpl. Proceed., 25:813 (1993) 23 Okarhara, et al., Can. Res., 54:3233 (1994) 35 24 Paavonen, et al., Int. J. Can., 58:298 (1994) 25 Schadendorf, et al., J. Path., 170:429 (1993) 26 Bao, et al., Diff., 52:239 (1993) 40 27 Lauri, et al., British J. Cancer, 68:862 (1993) 28 Kawaguchi, et al., Japanese J. Cancer Res., 83:1304 (1992) WO 99/06432 PCT/US98/15325 -- 3 - 29 Kogan, et al., U.S. Patent No. 5,510,332, issued April 23, 1996 30 International Patent Appl. Publication No. WO 96/01644 5 All of the above publications, patents and patent applications are herein incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety. 10 State of the Art VLA-4 (also referred to as a4pl integrin and CD49d/CD29), first identified by Hemler and Takada 1 is a member of the P1 integrin family of cell surface receptors, each of which comprises two subunits, an a chain and 15 a P3 chain. VLA-4 contains an a4 chain and a 131 chain. There are at least nine Pl31 integrins, all sharing the same P31 chain and each having a distinct a chain. These nine receptors all bind a different complement of the various cell matrix molecules, such as fibronectin, laminin, and collagen. VLA-4, for example, binds to fibronectin. VLA-4 is unique among P3, integrins in 20 that it also binds non-matrix molecules that are expressed by endothelial and other cells. These non-matrix molecules include VCAM-1, which is expressed on cytokine-activated human umbilical vein endothelial cells in culture. Distinct epitopes of VLA-4 are responsible for the fibronectin and VCAM-1 binding activities and each activity has been shown to be inhibited 25 independently Intercellular adhesion mediated by VLA-4 and other cell surface receptors is associated with a number of inflammatory responses. At the site of an injury or other inflammatory stimulus, activated vascular endothelial 30 cells express molecules that are adhesive for leukocytes. The mechanics of leukocyte adhesion to endothelial cells involves, in part, the recognition and binding of cell surface receptors on leukocytes to the corresponding cell WO 99/06432 PCT/US98/15325 -- 4surface molecules on endothelial cells. Once bound, the leukocytes migrate across the blood vessel wall to enter the injured site and release chemical mediators to combat infection. For reviews of adhesion receptors of the immune system, see, for example, Springer and Osborn 4 . 5 Inflammatory brain disorders, such as experimental autoimmune encephalomyelitis (EAE), multiple sclerosis (MS) and meningitis, are examples of central nervous system disorders in which the endothelium/leukocyte adhesion mechanism results in destruction to 10 otherwise healthy brain tissue. Large numbers of leukocytes migrate across the blood brain barrier (BBB) in subjects with these inflammatory diseases. The leukocytes release toxic mediators that cause extensive tissue damage resulting in impaired nerve conduction and paralysis. 15 In other organ systems, tissue damage also occurs via an adhesion mechanism resulting in migration or activation of leukocytes. For example, it has been shown that the initial insult following myocardial ischemia to heart tissue can be further complicated by leukocyte entry to the injured tissue causing still further insult (Vedder et al .5). Other inflammatory 20 conditions mediated by an adhesion mechanism include, by way of example, asthma" 8 , Alzheimer's disease, atherosclerosis 9-1 0 , AIDS dementia", diabetes 12- 1 4 (including acute juvenile onset diabetis), inflammatory bowel disease 15 (including ulcerative colitis and Crohn's disease), multiple sclerosis1 6-17 , rheumatoid arthritis' 8-2 1, tissue transplantation 2 2 , tumor 25 metastasis 2 3- 28, meningitis, encephalitis, stroke, and other cerebral traumas, nephritis, retinitis, atopic dermatitis, psoriasis, myocardial ischemia and acute leukocyte-mediated lung injury such as that which occurs in adult respiratory distress syndrome.
WO 99/06432 PCT/US98/15325 -- 5 - In view of the above, assays for determining the level VLA-4 in a biological sample containing VLA-4 would be useful, for example, to diagnosis VLA-4 mediated conditions. Additionally, despite these advances in the understanding of leukocyte adhesion, the art has only recently 5 addressed the use of inhibitors of adhesion in the treatment of inflammatory brain diseases and other inflammatory conditions 29
,
3 0 . The present invention addresses these and other needs. SUMMARY OF THE INVENTION 10 This invention provides compounds which bind to VLA-4. Such compounds can be used, for example, to assay for the presence of VLA-4 in a sample and, in pharmaceutical compositions to inhibit cellular adhesion mediated by VLA-4, for example, binding of VCAM-1 to VLA-4. The compounds of this invention have a binding affinity to VLA-4 as expressed 15 by an IC 5 0 of about 15 4M or less (as measured by Example 79 below) which compounds are defined by formula I below: 0
R
3 O 20 R 1
-S
O
2
-N(R
2 )-C-Q-CH-C-OH I I I H R 5 25 where R' is selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocylic, heteroaryl and substituted heteroaryl;
R
2 is selected from the group consisting of hydrogen, alkyl, 30 substituted alkyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, and R 1 and R 2 together with the nitrogen WO 99/06432 PCT/US98/15325 -- 6 atom bound to R 2 and the SO 2 group bound to R 1 can form a heterocyclic or a substituted heterocyclic group;
R
3 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, 5 heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic and, when R 2 does not form a heterocyclic group with R 1 , R 2 and R 3 together with the nitrogen atom bound to R 2 and the carbon atom bound to R 3 can form a heterocyclic or a substituted heterocyclic group;
R
5 is -ALK-X or = CH-Y where ALK is an alkyl group of from 1 to 10 10 carbon atoms attached via a methylene group (-CH 2 -) to the carbon atom to which it is attached; X is selected from the group consisting of substituted alkylcarbonylamino, substituted alkenylcarbonylamino, substituted alkynylcarbonylamino, heterocyclylcarbonylamino, substituted heterocyclylcarbonylamino, acyl, acyloxy, aminocarbonyloxy, acylamino, 15 oxycarbonylamino, alkoxycarbonyl, substituted alkoxycarbonyl, aryloxycarbonyl, substituted aryloxycarbonyl, cycloalkoxycarbonyl, substituted cycloalkoxycarbonyl, heteroaryloxycarbonyl, substituted heteroaryloxycarbonyl, heterocyclyloxycarbonyl, substituted heterocyclyloxycarbonyl, cycloalkyl, substituted cycloalkyl, saturated 20 heterocyclic, substituted saturated heterocyclic, substituted alkoxy, substituted alkenoxy, substituted alkynoxy, heterocyclyloxy, substituted heterocycloxy, substituted thioalkyl, substituted thioalkenyl, substituted thioalkynyl, aminocarbonylamino, aminothiocarbonylamino, guanidino, amidino, alkylamidino, thioamidino, halogen, cyano, nitro, -OS(O) 2 -alkyl, 25 -OS(O) 2 -substituted alkyl, -OS(O) 2 -cycloalkyl, -OS(O) 2 -substituted cycloalkyl, -OS(O) 2 -aryl, -OS(O) 2 -substituted aryl, -OS(O) 2 -heteroaryl,
-OS(O)
2 -substituted heteroaryl, -OS(O) 2 -heterocyclic, -OS(O) 2 -substituted heterocyclic, -OSO 2 -NRR where R is hydrogen or alkyl, -NRS(O) 2 -alkyl,
-NRS(O)
2 -substituted alkyl, -NRS(O) 2 -cycloalkyl, -NRS(O) 2 -substituted 30 cycloalkyl, -NRS(O) 2 -aryl, -NRS(O) 2 -substituted aryl, -NRS(O) 2 -heteroaryl, WO 99/06432 PCT/US98/15325 -- 7 -
-NRS(O)
2 -substituted heteroaryl, -NRS(O) 2 -heterocyclic, -NRS(O) 2 substituted heterocyclic, -NRS(O) 2 -NR-alkyl, -NRS(O) 2 -NR-substituted alkyl, -NRS(O) 2 -NR-cycloalkyl, -NRS(O) 2 -NR-substituted cycloalkyl,
-NRS(O)
2 -NR-aryl, -NRS(O) 2 -NR-substituted aryl, -NRS(O) 2 -NR-heteroaryl, 5 -NRS(O) 2 -NR-substituted heteroaryl, -NRS(O) 2 -NR-heterocyclic, -NRS(O) 2 NR-substituted heterocyclic where R is hydrogen or alkyl, -S(O)-alkyl, S(O) 2 -substituted alkyl, -S(O) 2 -aryl,
-S(O)
2 -substituted aryl, -S(O) 2 -substituted heteroaryl, -S(O) 2 -substituted heteroaryl, -S(O) 2 -heterocyclic, -S(O) 2 -substituted heterocyclic, mono- and 10 di-(substituted alkyl)amino, N,N-(alkyl, substituted alkyl)amino, N,N-(aryl, substituted alkyl)amino, N,N-(substituted aryl, substituted alkyl)amino, N,N (heteroaryl, substituted alkyl)amino, N,N-(substituted heteroaryl, substituted alkyl)amino, N,N-(heterocyclic, substituted alkyl)amino, N,N-N,N (substituted heterocyclic, substituted alkyl)amino, mono- and di 15 (heterocyclic)amino, mono- and di-(substituted heterocyclic)amino, N,N (alkyl, heterocyclic)amino, N,N-(alkyl, substituted heterocyclic)amino, N,N (aryl, heterocyclic)amino, N,N-(substituted aryl, heterocyclic)amino, N,N (aryl, substituted heterocyclic)amino, N,N-(substituted aryl, substituted heterocyclic)amino, N,N-(heteroaryl, heterocyclic)amino, N,N-(heteroaryl, 20 substituted heterocyclic)amino, N,N-(substituted heteroaryl, heterocyclic)amino, and N,N-(substituted heteroaryl, substituted heterocyclic)amino; and Y is selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic; 25 Q is -C(X)NR 7 - wherein R 7 is selected from the group consisting of hydrogen and alkyl; and X is selected from the group consisting of oxygen and sulfur; and pharmaceutically acceptable salts thereof with the provisos that WO 99/06432 PCT/US98/15325 -- 8 - A. when R 1 is p-methylphenyl, R 2 and R 3 are joined together with the nitrogen atom bound to R 2 and the carbon atom bound to R 3 to form a pyrrolidinyl ring and Q is -C(O)NH-, then R 5 is not -CH 2 C(O)-O-t-butyl or CH 2
CH
2 C(O)-O-t-butyl; and 5 B. when R 1 is p-methylphenyl, R 2 is methyl, R 3 is hydrogen and Q is -C(O)NH-, then R 5 is not -CH 2 (N-benzylpiperin-4-yl). In another embodiment, the compounds of this invention can also be provided as prodrugs which convert (e.g., hydrolyze, metabolize, etc.) in 10 vivo to a compound of formula I above. In a preferred example of such an embodiment, the carboxylic acid of the compound of formula I is modified into a group which, in vivo, will convert to a carboxylic acid (including salts thereof). In a particularly preferred embodiment, such prodrugs are represented by compounds of formula IA: 15
R
3 O III
R
1
-S
O
2
-N(R
2
)-C-Q-CH-C-R
6 IA 20 H I where
R
1 is selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heterocyclic, 25 substituted heterocylic, heteroaryl and substituted heteroaryl;
R
2 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, and R' and R 2 together with the nitrogen 30 atom bound to R 2 and the SO 2 group bound to R' can form a heterocyclic or a substituted heterocyclic group;
R
3 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, WO 99/06432 PCT/US98/15325 -- 9heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic and, when R 2 does not form a heterocyclic group with R', R 2 and R 3 together with the nitrogen atom bound to R 2 and the carbon atom bound to R 3 can form a heterocyclic or a substituted heterocyclic group; 5 R 5 is -ALK-X or = CH-Y where ALK is an alkyl group of from 1 to 10 carbon atoms attached via a methylene group (-CH 2 -) to the carbon atom to which it is attached; X is selected from the group consisting of substituted alkylcarbonylamino, substituted alkenylcarbonylamino, substituted alkynylcarbonylamino, heterocyclylcarbonylamino, substituted 10 heterocyclylcarbonylamino, acyl, acyloxy, aminocarbonyloxy, acylamino, oxycarbonylamino, alkoxycarbonyl, substituted alkoxycarbonyl, aryloxycarbonyl, substituted aryloxycarbonyl, cycloalkoxycarbonyl, substituted cycloalkoxycarbonyl, heteroaryloxycarbonyl, substituted heteroaryloxycarbonyl, heterocyclyloxycarbonyl, substituted 15 heterocyclyloxycarbonyl, cycloalkyl, substituted cycloalkyl, saturated heterocyclic, substituted saturated heterocyclic, substituted alkoxy, substituted alkenoxy, substituted alkynoxy, heterocyclyloxy, substituted heterocycloxy, substituted thioalkyl, substituted thioalkenyl, substituted thioalkynyl, aminocarbonylamino, aminothiocarbonylamino, guanidino, 20 amidino, alkylamidino, thioamidino, halogen, cyano, nitro, -OS(O) 2 -alkyl,
-OS(O)
2 -substituted alkyl, -OS(O) 2 -cycloalkyl, -OS(O) 2 -substituted cycloalkyl, -OS(O) 2 -aryl, -OS(O) 2 -substituted aryl, -OS(O) 2 -heteroaryl,
-OS(O)
2 -substituted heteroaryl, -OS(O) 2 -heterocyclic, -OS(O) 2 -substituted heterocyclic, -OSO 2 -NRR where R is hydrogen or alkyl, -NRS(O)-alkyl, 25 -NRS(O) 2 -substituted alkyl, -NRS(O) 2 -cycloalkyl, -NRS(O) 2 -substituted cycloalkyl, -NRS(O) 2 -aryl, -NRS(O) 2 -substituted aryl, -NRS(O) 2 -heteroaryl,
-NRS(O)
2 -substituted heteroaryl, -NRS(O)2-heterocyclic, -NRS(O) 2 substituted heterocyclic, -NRS(O) 2 -NR-alkyl, -NRS(O) 2 -NR-substituted alkyl, -NRS(O) 2 -NR-cycloalkyl, -NRS(O) 2 -NR-substituted cycloalkyl, WO 99/06432 PCT/US98/15325 -- 10-
-NRS(O)
2 -NR-aryl, -NRS(O) 2 -NR-substituted aryl, -NRS(O) 2 -NR-heteroaryl,
-NRS(O)
2 -NR-substituted heteroaryl, -NRS(O) 2 -NR-heterocyclic, -NRS(O) 2 NR-substituted heterocyclic where R is hydrogen or alkyl, -S(O)-alkyl, S(O) 2 -substituted alkyl, -S(O) 2 -aryl, 5 -S(O) 2 -substituted aryl, -S(O) 2 -substituted heteroaryl, -S(O) 2 -substituted heteroaryl, -S(O) 2 -heterocyclic, -S(O) 2 -substituted heterocyclic, mono- and di-(substituted alkyl)amino, N,N-(alkyl, substituted alkyl)amino, N,N-(aryl, substituted alkyl)amino, N,N-(substituted aryl, substituted alkyl)amino, N,N (heteroaryl, substituted alkyl)amino, N,N-(substituted heteroaryl, substituted 10 alkyl)amino, N,N-(heterocyclic, substituted alkyl)amino, N,N-N,N (substituted heterocyclic, substituted alkyl)amino, mono- and di (heterocyclic)amino, mono- and di-(substituted heterocyclic)amino, N,N (alkyl, heterocyclic)amino, N,N-(alkyl, substituted heterocyclic)amino, N,N (aryl, heterocyclic)amino, N,N-(substituted aryl, heterocyclic)amino, N,N 15 (aryl, substituted heterocyclic)amino, N,N-(substituted aryl, substituted heterocyclic)amino, N,N-(heteroaryl, heterocyclic)amino, N,N-(heteroaryl, substituted heterocyclic)amino, N,N-(substituted heteroaryl, heterocyclic)amino, and N,N-(substituted heteroaryl, substituted heterocyclic)amino; and Y is selected from the group consisting of alkyl, 20 substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic;
R
6 is selected from the group consisting of 2,4-dioxo-tetrahydrofuran 3-yl (3,4-enol), amino, alkoxy, substituted alkoxy, cycloalkoxy, substituted cycloalkoxy, -O-(N-succinimidyl), -NH-adamantyl, -O-cholest-5-en-3-3-yl, 25 NHOY where Y is hydrogen, alkyl, substituted alkyl, aryl, and substituted aryl, -NH(CH2)pCOOY where p is an integer of from 1 to 8 and Y is as defined above, -OCH 2
NR
9 R'o where R 9 is selected from the group consisting of -C(O)-aryl and -C(O)-substituted aryl and R' is selected from the group consisting of hydrogen and -CH 2
COOR
n where R" is alkyl, and -NHSO 2 Z 30 where Z is alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, WO 99/06432 PCT/US98/15325 -- 11 substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic; Q is -C(X)NR 7 - wherein R 7 is selected from the group consisting of hydrogen and alkyl; and X is selected from the group consisting of oxygen 5 and sulfur; and pharmaceutically acceptable salts thereof with the proviso that A. when R 1 is p-methylphenyl, R 2 and R 3 are joined together with the nitrogen atom bound to R 2 and the carbon atom bound to R 3 to form a 10 pyrrolidinyl ring, R 6 is methoxy, and Q is -C(O)NH-, then R is not CH 2
CH
2 COO-t-butyl or -CH 2 COO-t-butyl. 15 Preferably, in the compounds of formula I and IA above, R' is selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, heterocyclic, substituted heterocylic, heteroaryl and substituted heteroaryl. Even more preferably R' is selected from the group 20 consisting of 4-methylphenyl, methyl, benzyl, n-butyl, 4-chlorophenyl, 1 naphthyl, 2-naphthyl, 4-methoxyphenyl, phenyl, 2,4,6-trimethylphenyl, 2 (methoxycarbonyl)phenyl, 2-carboxyphenyl, 3,5-dichlorophenyl, 4 trifluoromethylphenyl, 3,4-dichlorophenyl, 3,4-dimethoxyphenyl, 4
(CH
3 C(O)NH-)phenyl, 4-trifluoromethoxyphenyl, 4-cyanophenyl, isopropyl, 25 3,5-di-(trifluoromethyl)phenyl, 4-t-butylphenyl, 4-t-butoxyphenyl, 4 nitrophenyl, 2-thienyl, 1-N-methyl-3-methyl-5-chloropyrazol-4-yl, phenethyl, 1-N-methylimidazol-4-yl, 4-bromophenyl, 4-amidinophenyl, 4 methylamidinophenyl, 4-[CH 3 SC(=NH)]phenyl, 5-chloro-2-thienyl, 2,5 dichloro-4-thienyl, 1-N-methyl-4-pyrazolyl, 2-thiazolyl, 5-methyl-1,3,4 30 thiadiazol-2-yl, 4-[H 2 NC(S)]phenyl, 4-aminophenyl, 4-fluorophenyl, 2- WO 99/06432 PCT/US98/15325 -- 12fluorophenyl, 3-fluorophenyl, 3,5-difluorophenyl, pyridin-3-yl, pyrimidin-2 yl, 4-(3'-dimethylamino-n-propoxy)-phenyl, and 1-methylpyrazol-4-yl. Preferably, in the compounds of formula I and IA above, R 2 is 5 hydrogen, methyl, phenyl, benzyl, -(CH 2
)
2 -2-thienyl, and -(CH1 2
)
2 -4 In one embodiment, R' and R 2 together with the nitrogen atom bound to R 2 and the SO 2 group bound to R 1 are joined to form a heterocyclic group or substituted heterocyclic group. Preferred heterocyclic and substituted 10 heterocyclic groups include those having from 5 to 7 ring atoms having 2 to 3 heteroatoms in the ring selected from nitrogen, oxygen and sulfur which ring is optionally fused to another ring such as a phenyl or cyclohexyl ring to provide for a fused ring heterocycle of from 10 to 14 ring atoms having 2 to 4 heteroatoms in the ring selected from nitrogen, oxygen and sulfur. 15 Specifically preferred R'/R 2 joined groups include, by way of example, benzisothiazolonyl (saccharin-2-yl). In one preferred embodiment, R 2 and R 3 together with the nitrogen atom bound to R 2 substituent and the carbon bound to the R 3 substituent form 20 a heterocyclic group or a substituted heterocyclic group of 4 to 6 ring atoms having 1 to 2 heteroatoms in the ring selected from nitrogen, oxygen and sulfur which ring is optionally substituted with 1 to 2 substituents selected from fluoro, methyl, hydroxy, amino, phenyl, thiophenyl, thiobenzyl, oxo or can be fused to another ring such as a phenyl or cycloalkyl ring to provide 25 for a fused ring heterocycle of from 10 to 14 ring atoms having 1 to 2 heteroatoms in the ring selected from nitrogen, oxygen and sulfur. Such heterocyclic rings include azetidinyl (e.g., L-azetidinyl), thiazolidinyl (e.g., L-thiazolidinyl), piperidinyl (e.g., L-piperidinyl), piperizinyl (e.g., L piperizinyl), dihydroindolyl (e.g., L-2,3-dihydroindol-2-yl), 30 tetrahydroquinolinyl (e.g., L-1,2,3,4-tetrahydroquinolin-2-yl), WO 99/06432 PCT/US98/15325 -- 13 thiomorpholinyl (e.g., L-thiomorpholin-3-yl), pyrrolidinyl (e.g., L pyrrolidinyl), substituted pyrrolidinyl such as 4-hydroxypyrrolidinyl (e.g., 4-a-(or P-)hydroxy-L-pyrrolidinyl), 4-fluoropyrrolidinyl (e.g., 4-ca-(or P-)fluoro-L-pyrrolidinyl), 5 3-phenylpyrrolidinyl (e.g., 3-a-(or P-)phenyl-L-pyrrolidinyl), 3-thiophenylpyrrolidinyl (e.g., 3-a-(or P-)thiophenyl-L-pyrrolidinyl), 4-aminopyrrolidinyl (e.g., 4-c-(or P-)amino-L-pyrrolidinyl), 3-methoxypyrrolidinyl (e.g., 3-a-(or P-)methoxy-L-pyrrolidinyl), 4,4-dimethylpyrrolidinyl, substituted piperizinyl such as 4-N-Cbz 10 piperizinyl, 5-oxopyrrolidinyl (e.g., 5-oxo-L-pyrolinyl), substituted thiazolidinyl such as 5,5-dimethylthiazolindin-4-yl, 1,1-dioxo-thiazolidinyl (e.g., L-1,1-dioxo-thiazolidin-2-yl), substituted 1,1-dioxo-thiazolidinyl such as L-1,1-dioxo-5,5-dimethylthiazolidin-2-yl, 1,1-dioxothiomorpholinyl (e.g., L-1,1-dioxo-thiomorpholin-3-yl) and the 15 like. Preferably, in the compounds of formula I and IA above, R 3 includes all of the isomers arising by substitution with methyl, phenyl, benzyl, diphenylmethyl, -CH 2
CH
2 -COOH, -CH 2 -COOH, 2-amidoethyl, iso-butyl, t 20 butyl, -CH 2 0-benzyl and hydroxymethyl. Additionally, in another preferred embodiment, R 3 and R 2 together with the nitrogen atom bound to R 2 can form a heterocyclic group or substituted heterocyclic group. Q is preferably -C(O)NH- or -C(S)NH-. 25
R
5 is preferably selected from all possible isomers arising by substitution with the following groups: t-butyl-OC(O)CH 2 -, -CH 2
C(O)NH
2 , CH 2
CH
2
C(O)NH
2 , t-butyl-OC(O)CH 2
CH
2 -, BocNH-(CH 2
)
4 -, 30 (4-CH 2 -OC(O)NH-(CH2) 4 -, benzyloxy-CH 2 -, cyclohexyl-CH 2 -, N-benzylpiperid-4-yl-CH 2 -, N-Boc-piperidin-4-yl-CH 2
-,
WO 99/06432 PCT/US98/15325 -- 14- N-(phenylcarbonyl)piperidin-4-yl-CH 2 -, allyloxy-C(O)NH-(CH 2
)
4 -, allyloxy-C(O)NH(CH 2
)
3 -, allyloxy-C(O)NH(CH 2
)
2 -, -CH=, 4-methylphenyl-SO 2
-N(CH
3
)CH
2
C(O)NH(CH
2
)
4 -, -CH 2
C(O)NH(CH
2
)
4 4,
-(CH
2
)
4
NHC(O)CH
2 -3-indolyl, -(CH 2
)
4
NHC(O)CH
2
CH
2 -3-indolyl, 5 -(CH 2
)
4 NHC(O)CH20-4-fluorophenyl, -CH 2
C(O)NHCH(CH
3 )4, -CH2C(O)NHCH 2 -(4-dimethylamino)-4, -CH 2
C(O)NHCH
2 -4-nitrophenyl,
-CH
2
CH
2
C(O)N(CH
3
)CH
2 -4, -CH 2
C(O)N(CH
3 )CH-1 2 4,
-CH
2
CH
2
C(O)NHCH
2
CH
2 -(N-methyl)-2-pyrrolyl,
-CH
2
CH
2
C(O)NHCH
2
CH
2
CH
2
CH
3 , -CH2CH 2 C(O)NHCH2CH 2 -3-indolyl, 10 -CH 2
C(O)N(CH
3 )CHzphenyl, -CH 2
C(O)NH(CH
2
)
2 -(N-methyl)-2-pyrrolyl, -CH2C(O)NHCH 2
CH
2
CH
2
CH
3 , -CH 2
C(O)NHCH
2 CH2-3-indolyl, -(CHz) 2
C(O)NHCH(CH
3 )4, -(CH 2 )2C(O)NHCH 2 -4-dimethylaminophenyl,
-(CH
2
)
2
C(O)NHCH
2 -4-nitrophenyl, -CHC(O)NH-4-[-NHC(O)CH 3 -phenyl],
-CH
2 C(O)NH-4-pyridyl, -CH 2 C(O)NH-4-[dimethylaminophenyl], 15 -CH 2 C(O)NH-3-methoxyphenyl, -CH 2
CH
2 C(O)NH-4-chlorophenyl, -CH2CH 2 C(O)NH-2-pyridyl, -CH 2
CH
2 C(O)NH-4-methoxyphenyl,
-CH
2
CH
2 C(O)NH-3-pyridyl, -(CH 2
)
3 NHC(NH)NH-SO2-4-methylphenyl,
-(CH
2
)
4 NHC(O)NHCHzCH 3 , -(CH 2
)
4 NHC(O)NH-phenyl, -(CHz) 4 NHC(O)NH-4-methoxyphenyl, -CH 2 C(O)NHCH2CH 2
N(CH
3
)
2 , 20 [BocNHCH 2 C(O)NH-]butyl, 2-[4-hydroxy-4-(3-methoxythien-2-yl)piperidin 1-yl]ethyl, 4-[(1-Cbz-piperidin-4-yl)C(O)NH-]butyl, 4-[(N-toluenesulfonylpyrrolidin-2'-yl)C(O)NH-]butyl, 4-[-NHC(O)-4'-piperidinyl]butyl, N-Cbz-NHCH 2 -, (CH 3
)
2
NC(O)CH
2 -, and N-Boc-2-aminoethyl. 25 In the compounds of formula IA, R 6 is preferably 2,4-dioxo tetrahydrofuran-3-yl (3,4-enol), methoxy, ethoxy, iso-propoxy, n-butoxy, t-butoxy, cyclopentoxy, neo-pentoxy, 2-a-iso-propyl-4-p methylcyclohexoxy, 2-p-isopropyl-4-[-methylcyclohexoxy, 30 -NH 2 , benzyloxy, -NHCH2COOH, -NHCH 2
CH
2 COOH, -NH-adamantyl, WO 99/06432 PCT/US98/15325 -- 15 -
-NHCH
2
CH
2
COOCH
2
CH
3 , -NHSO 2 -p-CH 3 -, -NHOR 8 where R 8 is hydrogen, methyl, iso-propyl or benzyl, O-(N-succinimidyl), -O-cholest-5-en-3-p-yl, -OCH 2
-OC(O)C(CH
3
)
3 , -O(CH 2 )zNHC(O)W where z is 1 or 2 and W is selected from the group consisting of pyrid-3-yl, N 5 methylpyridyl, and N-methyl-1,4-dihydro-pyrid-3-yl, -NR"C(O)-R' where R' is aryl, heteroaryl or heterocyclic and R" is hydrogen or
-CH
2 C(O)OCH2CH 3 . Preferred compounds within the scope of formula I and IA above 10 include by way of example: N-(toluene-4-sulfonyl)-L-prolyl-L-aspartic acid 4-tert-butyl ester N-(toluene-4-sulfonyl)-L-prolyl-L-asparagine 15 N-(toluene-4-sulfonyl)-L-prolyl-Ne-(tert-butoxycarbonyl)-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-L-glutamic acid 5-tert-butyl ester N-(toluene-4-sulfonyl)-L-prolyl-L-glutamine 20 N-(toluene-4-sulfonyl)-L-prolyl-Ne-(carbobenzyloxy)-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-O-benzyl-L-serine 25 N-(toluene-4-sulfonyl)-L-prolyl-p-cyclohexyl-L-alanine N-(toluene-4-sulfonyl)sarcosyl-0-(N-tert-butoxycarbonylpiperidin-4 yl)-D,L-alanine 30 N-(toluene-4-sulfonyl)sarcosyl-p-(N-benzoylpiperidin-4-yl)-D,L alanine N-(toluene-4-sulfonyl)sarcosyl-Ne-tert-butoxycarbonyl-L-lysine 35 N-(toluene-4-sulfonyl)-L-prolyl-NE-tert-butoxycarbonyl-D-lysine N-(toluene-4-sulfonyl)-L-prolyl-NE-(allyloxycarbonyl)-L-lysine N-(3,5-ditrifluoromethylbenzenesulfonyl)-L-prolyl-Ne 40 (allyloxycarbonyl)-L-lysine WO 99/06432 PCT/US98/15325 -- 16 - N-(toluene-4-sulfonyl)sarcosyl-NE-(allyloxycarbonyl)-L-lysine N-(toluene-4-sulfonyl)sarcosyl-5-(allyloxycarbonylamino)pentanoic acid 5 N-(toluene-4-sulfonyl)sarcosyl-4-(allyloxycarbonylamino)butanoic acid N-(toluene-4-sulfonyl)-L-prolyl-Ne-(allyloxycarbonyl)-L-lysine 10 N-(toluene-4-sulfonyl)-L-prolyl-4-(allyloxycarbonylamino)butanoic acid N-(toluene-4-sulfonyl)-L-glutaminyl-L-asparagine 15 N-(toluene-4-sulfonyl)-L-prolyl-Ne-[N-(toluene-4-sulfonyl)sarcosyl] L-lysine N-(toluene-4-sulfonyl)-L-prolyl-(2,3-dehydro)phenylalanine 20 N-(toluene-4-sulfonyl)-L-prolyl-Ny-(4-phenyl)butyl-L-asparagine N-(toluene-4-sulfonyl)-L-prolyl-N-(indol-3-ylacetyl)-L-ly sine 25 N-(toluene-4-sulfonyl)-L-prolyl-NE-[3-(indol-3-yl)propionyl)-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-N-(5-methoxyindol-3-carbonyl)-L lysine 30 N-(toluene-4-sulfonyl)-L-prolyl-N-(4-fluorophenoxyacetyl)-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-Ny-R-(1-phenyl)ethyl-L-asparagine N-(toluene-4-sulfonyl)-L-prolyl-Ny-S-(1-phenyl)ethyl-L-asparagine 35 N-(toluene-4-sulfonyl)-L-prolyl-Ny-(4-nitrophenyl)methyl-L asparagine N-(toluene-4-sulfonyl)-L-prolyl-N8-benzyl-N6-methyl-L-glutamine 40 N-(toluene-4-sulfonyl)-L-prolyl-N8-2-(1-methylpyrrol-2-yl)ethyl-L glutamine N-(toluene-4-sulfonyl)-L-prolyl-N8-n-butyl-L-glutamine 45 WO 99/06432 PCT/US98/1 5325 -- 17 - N-(toluene-4-sulfony1)-L-proly1-N8-2-(indo--3-y1)ethyl-L-glutamifle N-(toluene-4-sulfonyl)sarcosyl-Ny -benzyl-Ny -methyl-L-asparagine 5 N-(toluene-4-sulfonyl)sarcosyl-Ny-2-(l1-methylpyrrol-2-yI)ethyl-L asparagine N-(toluene-4-.sulfonyl)sarcosyl-Ny-n-butyl-L-asparagine 10 N-(toluene-4-sulfony1)sarcosyI-Ny-2-(indo1-3-y1)ethy1-L-asparagile N-(toluene-4-sulfonyl)sarcosyl-Na-R-( 1 -phenyl)ethyl-L-glutamine N-(toluene-4-sulfonyl)sarcosyl-N6-S-( 1 -phenyl)ethyl-L-glutamine 15 N-(toluene-4-sulfonyl)sarcosyl-N-(4-N,N-dimethylamilo phenyl)methyl-L-glutamine N-(toluene-4.-sufony)sarcosy1-N6-(4-nitrophenfl)methyl-L-g1utamifle 20 N-(toluene-4-sulfonyl)-N-2-(thien-2-yl)glycinyl-N 6 -S-(l1-phenyl)ethyl L-glutamine N-(toluene-4-sulfonyl)-N-2-(thien-2-yl)glycinyl-NY -(4-N,N 25 dimethylaminophenyl)methyl-L-asparagifle N-(toluene-4-sulfonyl)-N-2-(thien-2-yl)glyciflyl-Ny-( 4 nitrophenyl)methyl-L-asparagine 30 N-(toluene-4-sulfonyl)-N-2-(thien-2-y)glycilyl-Ny-beflNy methyl-L-asparagine N-(toluene-4-sulfonyl)-N-2-(thien-2-yl)glyciflyl-N 6 -2-(l1-methylpyrrol 2-yl)ethyl-L-glutamine 35 N-(toluene-4-sulfonyl)-N-2-(thien-2-yl)glyciflyl-N 6 -fl-butyl-L glutamine N-(toluene-4-sulfonyl)-N-2-(thien-2-y)glycil-N6-2-(ildo1-3 40 yl)ethyl-L-glutamine N-(toluene-4-sulfonyl)-N-2-(thien-2-yl)glycinyl-N 6 -R-(l1-phenyl)ethyl L-glutamine WO 99/06432 PCT/US98/1 5325 -- 18 - N-(toluene-4-sulfonyl)-L-prolyl-Ny-(4-acetamidophenyl)-L-asparagine N-(toluene-4-sulfonyl)-L-prolyl-Ny-(pyrid-4-yl)-L-asparagine 5 N-(toluene-4-sulfonyl)-L-prolyl-Ny-(4-N,N-dimethylaminophenyl)-L asparagine N-(toluene-4-sulfonyl)-L-prolyl-Ny-(3-methoxyphenyl)-L-asparagine 10 N-(toluene-4-sulfonyl)-L-prolyl-N5-(4-chlorophenyl)-L-glutamine N-(toluene-4-sulfonyl)-L-prolyl-N5-(pyrid-2-yl)-L-glutamine N-(toluene-4-sulfonyl)-L-prolyl-N6-(4-methoxyphenyl)-L-glutamine 15 N-(toluene-4-sulfonyl)-L-prolyl-N6-(pyrid-3-yl)-L-glutamine N-(toluene-4-sulfony1)-L-prolyl-N )-(toluene-4-sulfonyl)-L-arginine 20 N-(toluene-4-sulfonyl)-L-prolyl-NE-(ethylaminocarbonyl)-L-lysile N-(toluene-4-sulfonyl)-L-prolyl-NE-(phenylaminocarbonyl)-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-NE-(4-methoxyphenylamino 25 carbonyl)-L-lysine N-(toluene-4-sulfonyl)-L-(5 ,5-dimethyl)thiaprolyl-NE-(tert butoxycarbonyl)-L-lysine methyl ester 30 N-(toluene-4-sulfonyl)-L-(5 ,5 -dimethyl)thiaprolyl-NE-(tert butoxycarbonyl)-L-lysine N-(toluene-4-sulfonyl)-L-(5 ,5-dimethyl)thiaprolyl-L-asparagine 35 N-(toluene-4-sulfonyl)-L-prolyl-Ny -(4-N,N-dimethylamino phenyl)methyl-L-asparagine N-(toluene-4-sulfonyl)-L-prolyl-Ny -(4-N,N-dimethylamino phenyl)methyl-L-asparagine methyl ester 40 N-(toluene-4-sulfonyl)-L-prolyl-Ny -2-(N,N-dimethylamino)ethyl-L asparagine N-(toluene-4-sulfonyl)-L-prolyl-NE-(ethylaminocarbonyl)-L-lysine 45 468 WO 99/06432 PCT/US98/15325 -- 19- N-(toluene-4-sulfonyl)-L-prolyl-NE-(N-tert-butoxycarbonylglycinyl) L-lysine N-(toluene-4-sulfonyl)-L-prolyl-Ne-[N-(carbobenzyloxy)iso 5 nipecotoyl]-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-Ne-(N-toluene-4-sulfonyl-L-prolyl) L-lysine 10 N-(toluene-4-sulfonyl)-L-prolyl-NE-(isonipecotoyl)-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-3-[N-(carbobenzyl oxy)amino]propionic acid 15 N-(toluene-4-sulfonyl)-L-prolyl-Ny,Ny-dimethyl-L-asparagine N-(toluene-4-sulfonyl)-L-prolyl-3-[N-(tert-butoxycarbonyl)amino]-2S propionic acid methyl ester 20 N-(toluene-4-sulfonyl)-L-(5-oxo) prolyl-L-asparagine and pharmaceutically acceptable salts thereof as well as any of the ester compounds recited above wherein one ester is replaced with another ester selected from the group consisting of methyl ester, ethyl ester, n-propyl 25 ester, isopropyl ester, n-butyl ester, isobutyl ester, sec-butyl ester and tert butyl ester. This invention also provides methods for binding VLA-4 in a biological sample which method comprises contacting the biological sample 30 with a compound of formula I or IA above under conditions wherein said compound binds to VLA-4. Certain of the compounds of formula I and IA above are also useful in reducing VLA-4 mediated inflammation in vivo. 35 This invention also provides pharmaceutical compositions comprising a pharmaceutically acceptable carrier and a therapeutically effective amount WO 99/06432 PCT/US98/15325 -- 20 of one or more the compounds of formula I or IA above with the exception that R 3 and R 5 are derived from L-amino acids or other similarly configured starting materials. Alternatively, racemic mixtures can be used. 5 The pharmaceutical compositions may be used to treat VLA-4 mediated disease conditions. Such disease conditions include, by way of example, asthma, Alzheimer's disease, atherosclerosis, AIDS dementia, diabetes (including acute juvenile onset diabetis), inflammatory bowel disease (including ulcerative colitis and Crohn's disease), multiple sclerosis, 10 rheumatoid arthritis, tissue transplantation, tumor metastasis, meningitis, encephalitis, stroke, and other cerebral traumas, nephritis, retinitis, atopic dermatitis, psoriasis, myocardial ischemia and acute leukocyte-mediated lung injury such as that which occurs in adult respiratory distress syndrome. 15 Accordingly, this invention also provides methods for the treatment of an inflammatory disease in a patient mediated by VLA-4 which methods comprise administering to the patient the pharmaceutical compositions described above. 20 Preferred compounds of formula I and IA above include those set forth in Table I below: WO 99/06432 PCTIUS98/1 5325 -- 21 z 9 9 08 o U u WO 99/06432 PCTIUS98/1 5325 z 0r
II
Qd4 9 0 0 6 I o CF M WO 99/06432 PCT/US98/15325 -- 23 z uI I 9I II III O & o o o a co a o o o 0N - . 0 0 U 0 0 U U 0 0 z Z z z o0 a 00 0< o. oN,2 -, 0,o._ IU U 0 - - u o zz cz oF e Z9 0 1= u u Q WO 99/06432 PCT/US98/1 5325 z 0 00 9 I u u UU I U u WO 99/06432 PCTIUS98/1 5325 -- 25 zz 0 n 0o 0 J 0 0 0 4 U m' 0) u WO 99/06432 PCT/US98/15325 -- 26 - 0 z uI oC) & o o a o o o o00 eqq u eq 1'4 UU UU ,.a 'a 3 a. . a '£ 9 9--a c 0 ,,o_ o • .- . 'U U o I I C)0 I aI I) I 0000 4 4 4 e 4444 I£I I I£ I £I £ 0 0U 0 0 0 oUoUU WO 99/06432 PCTIUS98/1 5325 z Or cz w ~ _ _ -& Q 0 ,.- = ~ 7_ 717 7 r4 . . .
WO 99/06432 PCT/US98/15325 -- 28 - II z II 9 9 9 9 I I 9 OE co u: M u M""" m ct >t , >., Q :£~ u Q "- .- , o- a . 00 -e OR 0~ z me= z 0 U o 4 - -" x--- 4 U UU U U U
II
WO 99/06432 PCT/US98/1 5325 In m WO 99/06432 PCT/US98115325 z Ii U 01 U U1 u u I ZU41U WO 99/06432 PCT/US98/1 5325 -- 31 zz z 0) zN u E E 0 Q Z z wc ll~ ~ ~ I- lr-0 l -3 1- = z 1= - m C:4Csmc
UCZ)
WO 99/06432 PCT/US98/1 5325 -- 32 z CCl Z40 cU WO 99/06432 PCT/US98/15325 -- 33 - DETAILED DESCRIPTION OF THE INVENTION As above, this invention relates to compounds which inhibit leukocyte adhesion and, in particular, leukocyte adhesion mediated by VLA 4. However, prior to describing this invention in further detail, the 5 following terms will first be defined. Definitions As used herein, "alkyl" refers to alkyl groups preferably having from 1 to 10 carbon atoms and more preferably 1 to 6 carbon atoms. This term is 10 exemplified by groups such as methyl, t-butyl, n-heptyl, octyl and the like. "Substituted alkyl" refers to an alkyl group, preferably of from 1 to 10 carbon atoms, having from 1 to 5 substituents selected from the group consisting of alkoxy, substituted alkoxy, acyl, acylamino, 15 thiocarbonylamino, acyloxy, amino, amidino, alkyl amidino,thioamidino, aminoacyl, aminocarbonylamino, aminothiocarbonylamino, aminocarbonyloxy, aryl, substituted aryl, aryloxy, substituted aryloxy, aryloxylaryl, substituted aryloxyaryl, cyano, halogen, hydroxyl, nitro, carboxyl, carboxylalkyl, carboxyl-substituted alkyl, carboxyl-cycloalkyl, 20 carboxyl-substituted cycloalkyl, carboxylaryl, carboxyl-substituted aryl, carboxylheteroaryl, carboxyl-substituted heteroaryl, carboxylheterocyclic, carboxyl-substituted heterocyclic, cycloalkyl, substituted cycloalkyl, guanidino, guanidinosulfone, thiol, thioalkyl, substituted thioalkyl, thioaryl, substituted thioaryl, thiocycloalkyl, substituted thiocycloalkyl, 25 thioheteroaryl, substituted thioheteroaryl, thioheterocyclic, substituted thioheterocyclic, heteroaryl, substituted aryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, oxycarbonylamino, oxythiocarbonylamino, -OS(O) 2 -alkyl, 30 OS(O) 2 -substituted alkyl, -OS(O) 2 -aryl, -OS(O) 2 -substituted aryl, -OS(O) 2
-
WO 99/06432 PCT/US98/15325 -- 34 heteroaryl, -OS(O) 2 -substituted heteroaryl, -OS(O) 2 -heterocyclic, -OS(0)2 substituted heterocyclic, -OSO 2 -NRR where R is hydrogen or alkyl, -NRS(0) 2 -alkyl, -NRS(0) 2 -substituted alkyl, -NRS(0) 2 -aryl, -NRS(0) 2 substituted aryl, -NRS(0) 2 -heteroaryl, -NRS(0) 2 -substituted heteroaryl, 5 -NRS(0) 2 -heterocyclic, -NRS(0) 2 -substituted heterocyclic, -NRS(0) 2
-NR
alkyl, -NRS(0) 2 -NR-substituted alkyl, -NRS(O) 2 -NR-aryl, -NRS(0) 2
-NR
substituted aryl, -NRS(0) 2 -NR-heteroaryl, -NRS(0) 2 -NR-substituted heteroaryl, -NRS(0) 2 -NR-heterocyclic, -NRS(0) 2 -NR-substituted heterocyclic where R is hydrogen or alkyl, mono- and di-alkylamino, mono 10 and di-(substituted alkyl)amino, mono- and di-arylamino, mono- and di substituted arylamino, mono- and di-heteroarylamino, mono- and di substituted heteroarylamino, mono- and di-heterocyclic amino, mono- and di-substituted heterocyclic amino, unsymmetric di-substituted amines having different substituents selected from alkyl, substituted alkyl, aryl, substituted 15 aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic and substituted alkyl groups having amino groups blocked by conventional blocking groups such as Boc, Cbz, formyl, and the like or alkyl/substituted alkyl groups substituted with -SO 2 -alkyl, -SO 2 -substituted alkyl, -SO 2 -alkenyl, -SO 2 -substituted alkenyl, -SO2-cycloalkyl, -S02 20 substituted cycloalkyl, -SO 2 -aryl, -SO 2 -substituted aryl, -SO 2 -heteroaryl, SO 2 -substituted heteroaryl, -SO 2 -heterocyclic, -SO 2 -substituted heterocyclic and -SO 2 NRR where R is hydrogen or alkyl. "Alkoxy" refers to the group "alkyl-O-" which includes, by way of 25 example, methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1,2-dimethylbutoxy, and the like. "Substituted alkoxy" refers to the group "substituted alkyl-O-".
WO 99/06432 PCT/US98/15325 -- 35 - "Acyl" refers to the groups H-C(O)-, alkyl-C(O)-, substituted alkyl C(O)-, alkenyl-C(O)-, substituted alkenyl-C(O)-, alkynyl-C(O)-, substituted alkynyl-C(O)- cycloalkyl-C(O)-, substituted cycloalkyl-C(O)-, aryl-C(O)-, substituted aryl-C(O)-, heteroaryl-C(O)-, substituted heteroaryl-C(O), 5 heterocyclic-C(O)-, and substituted heterocyclic-C(O)- provided that a nitrogen atom of the heterocyclic or substituted heterocyclic is not bound to the -C(O)- group wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and 10 substituted heterocyclic are as defined herein. The "acylamino" refers to the group -C(O)NRR where each R is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, 15 aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic and where each R is joined to form together with the nitrogen atom a heterocyclic or substituted heterocyclic ring wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted 20 cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein. The term "substituted alkylcarbonylamino" refers only to the acylamino group -C(O)NRR where each R is independently selected from 25 hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic provided that at least one R is substituted alkyl. The term "substituted alkenylcarbonylamino" refers only to the 30 acylamino group -C(O)NRR where each R is independently selected from WO 99/06432 PCT/US98/15325 -- 36 hydrogen, alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic provided that at least one R is substituted alkenyl. 5 The term "substituted alkynylcarbonylamino" refers only to the acylamino group -C(O)NRR where each R is independently selected from hydrogen, alkyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 10 heterocyclic and substituted heterocyclic provided that at least one R is substituted alkynyl. The term "heterocyclylcarbonylamino" refers only to the acylamino group -C(O)NRR where each R is independently selected from hydrogen, 15 alkyl, substituted alkyl, alkenyl, substituted alkenyl alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic provided that at least one R is heterocyclic. 20 "Thiocarbonylamino" refers to the group -C(S)NRR where each R is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic and where each 25 R is joined to form, together with the nitrogen atom a heterocyclic or substituted heterocyclic ring wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein. 30 WO 99/06432 PCT/US98/15325 -- 37 - "Acyloxy" refers to the groups alkyl-C(O)O-, substituted alkyl C(O)O-, alkenyl-C(O)O-, substituted alkenyl-C(O)O-, alkynyl-C(O)O-, substituted alkynyl-C(O)O-, aryl-C(O)O-, substituted aryl-C(O)O-, cycloalkyl-C(O)O-, substituted cycloalkyl-C(O)O-, heteroaryl-C(O)O-, 5 substituted heteroaryl-C(O)O-, heterocyclic-C(O)O-, and substituted heterocyclic-C(O)O- wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein. 10 "Alkenyl" refers to alkenyl group preferably having from 2 to 10 carbon atoms and more preferably 2 to 6 carbon atoms and having at least 1 and preferably from 1-2 sites of alkenyl unsaturation. 15 "Substituted alkenyl" refers to alkenyl groups having from 1 to 5 substituents selected from the group consisting of alkoxy, substituted alkoxy, acyl, acylamino, thiocarbonylamino, acyloxy, amino, amidino, alkylamidino, thioamidino, aminoacyl, aminocarbonylamino, aminothiocarbonylamino, aminocarbonyloxy, aryl, substituted aryl, aryloxy, 20 substituted aryloxy, aryloxyaryl, substituted aryloxyaryl, halogen, hydroxyl, cyano, nitro, carboxyl, carboxylalkyl, carboxyl-substituted alkyl, carboxyl cycloalkyl, carboxyl-substituted cycloalkyl, carboxylaryl, carboxyl substituted aryl, carboxylheteroaryl, carboxyl-substituted heteroaryl, carboxylheterocyclic, carboxyl-substituted heterocyclic, cycloalkyl, 25 substituted cycloalkyl, guanidino, guanidinosulfone, thiol, thioalkyl, substituted thioalkyl, thioaryl, substituted thioaryl, thiocycloalkyl, substituted thiocycloalkyl, thioheteroaryl, substituted thioheteroaryl, thioheterocyclic, substituted thioheterocyclic, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkoxy, substituted WO 99/06432 PCT/US98/15325 -- 38cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, oxycarbonylamino, oxythiocarbonylamino,
-OS(O)
2 -alkyl, -OS(O) 2 -substituted alkyl, -OS(O) 2 -aryl, -OS(O) 2 -substituted aryl, -OS(O) 2 -heteroaryl, -OS(O) 2 -substituted heteroaryl, -OS(O) 2 5 heterocyclic, -OS(O) 2 -substituted heterocyclic, -OSO2-NRR where R is hydrogen or alkyl, -NRS(O) 2 -alkyl, -NRS(O) 2 -substituted alkyl, -NRS(O) 2 aryl, -NRS(O) 2 -substituted aryl, -NRS(O) 2 -heteroaryl, -NRS(O) 2 -substituted heteroaryl, -NRS(O) 2 -heterocyclic, -NRS(O) 2 -substituted heterocyclic,
-NRS(O)
2 -NR-alkyl, -NRS(O) 2 -NR-substituted alkyl, -NRS(O) 2 -NR-aryl, 10 -NRS(O) 2 -NR-substituted aryl, -NRS(O) 2 -NR-heteroaryl, -NRS(O) 2
-NR
substituted heteroaryl, -NRS(O) 2 -NR-heterocyclic, -NRS(O) 2 -NR-substituted heterocyclic where R is hydrogen or alkyl, mono- and di-alkylamino, mono and di-(substituted alkyl)amino, mono- and di-arylamino, mono- and di substituted arylamino, mono- and di-heteroarylamino, mono- and di 15 substituted heteroarylamino, mono- and di-heterocyclic amino, mono- and di-substituted heterocyclic amino, unsymmetric di-substituted amines having different substituents selected from alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic and substituted alkenyl groups having amino groups blocked by 20 conventional blocking groups such as Boc, Cbz, formyl, and the like or alkenyl/substituted alkenyl groups substituted with -SO 2 -alkyl, -SO 2 substituted alkyl, -S0 2 -alkenyl, -SO 2 -substituted alkenyl, -SO 2 -cycloalkyl,
-SO
2 -substituted cycloalkyl, -SO 2 -aryl, -SO 2 -substituted aryl, -SO2 heteroaryl, -SO,-substituted heteroaryl, -SO 2 -heterocyclic, -SO 2 -substituted 25 heterocyclic and -SO 2 NRR where R is hydrogen or alkyl. "Alkynyl" refers to alkynyl group preferably having from 2 to 10 carbon atoms and more preferably 3 to 6 carbon atoms and having at least 1 and preferably from 1-2 sites of alkynyl unsaturation. 30 WO 99/06432 PCT/US98/15325 -- 39 - "Substituted alkynyl" refers to alkynyl groups having from 1 to 5 substituents selected from the group consisting of alkoxy, substituted alkoxy, acyl, acylamino, thiocarbonylamino, acyloxy, amino, amidino, alkylamidino, thioamidino, aminoacyl, aminocarbonylamino, 5 aminothiocarbonylamino, aminocarbonyloxy, aryl, substituted aryl, aryloxy, substituted aryloxy, aryloxyaryl, substituted aryloxyaryl, halogen, hydroxyl, cyano, nitro, carboxyl, carboxylalkyl, carboxyl-substituted alkyl, carboxyl cycloalkyl, carboxyl-substituted cycloalkyl, carboxylaryl, carboxyl substituted aryl, carboxylheteroaryl, carboxyl-substituted heteroaryl, 10 carboxylheterocyclic, carboxyl-substituted heterocyclic, cycloalkyl, substituted cycloalkyl, guanidino, guanidinosulfone, thiol, thioalkyl, substituted thioalkyl, thioaryl, substituted thioaryl, thiocycloalkyl, substituted thiocycloalkyl, thioheteroaryl, substituted thioheteroaryl, thioheterocyclic, substituted thioheterocyclic, heteroaryl, substituted 15 heteroaryl, heterocyclic, substituted heterocyclic, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, oxycarbonylamino, oxythiocarbonylamino,
-OS(O)
2 -alkyl, -OS(O) 2 -substituted alkyl, -OS(O) 2 -aryl, -OS(O) 2 -substituted aryl, -OS(O) 2 -heteroaryl, -OS(O) 2 -substituted heteroaryl, -OS(0)2 20 heterocyclic, -OS(0) 2 -substituted heterocyclic, -OSO 2 -NRR where R is hydrogen or alkyl, -NRS(0) 2 -alkyl, -NRS(0) 2 -substituted alkyl, -NRS(0) 2 aryl, -NRS(0) 2 -substituted aryl, -NRS(0) 2 -heteroaryl, -NRS(0) 2 -substituted heteroaryl, -NRS(O) 2 -heterocyclic, -NRS(0) 2 -substituted heterocyclic, -NRS(0) 2 -NR-alkyl, -NRS(O) 2 -NR-substituted alkyl, -NRS(O) 2 -NR-aryl, 25 -NRS(0) 2 -NR-substituted aryl, -NRS(O) 2 -NR-heteroaryl, -NRS(0) 2
-NR
substituted heteroaryl, -NRS(O) 2 -NR-heterocyclic, -NRS(O) 2 -NR-substituted heterocyclic where R is hydrogen or alkyl, mono- and di-alkylamino, mono and di-(substituted alkyl)amino, mono- and di-arylamino, mono- and di substituted arylamino, mono- and di-heteroarylamino, mono- and di 30 substituted heteroarylamino, mono- and di-heterocyclic amino, mono- and WO 99/06432 PCT/US98/15325 -- 40 di-substituted heterocyclic amino, unsymmetric di-substituted amines having different substituents selected from alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic and substituted alkynyl groups having amino groups blocked by 5 conventional blocking groups such as Boc, Cbz, formyl, and the like or alkynyl/substituted alkynyl groups substituted with -SO2-alkyl, -SO 2 substituted alkyl, -SO 2 -alkenyl, -SO 2 -substituted alkenyl, -SO 2 -cycloalkyl,
-SO
2 -substituted cycloalkyl, -S0 2 -aryl, -S0 2 -substituted aryl, -S02 heteroaryl, -SO 2 -substituted heteroaryl, -SO 2 -heterocyclic, -SO2-substituted 10 heterocyclic and -SO 2 NRR where R is hydrogen or alkyl. "Amidino" refers to the group H 2 NC(=NH)- and the term "alkylamidino" refers to compounds having 1 to 3 alkyl groups (e.g., alkylHNC(= NH)-). 15 "Thioamidino" refers to the group RSC(=NH)- where R is hydrogen or alkyl. "Aminoacyl" refers to the groups -NRC(O)alkyl, 20 -NRC(O)substituted alkyl, -NRC(O)cycloalkyl, -NRC(O)substituted cycloalkyl, -NRC(O)alkenyl, -NRC(O)substituted alkenyl, -NRC(O)alkynyl, -NRC(O)substituted alkynyl, -NRC(O)aryl, -NRC(O)substituted aryl, -NRC(O)heteroaryl, -NRC(O)substituted heteroaryl, -NRC(O)heterocyclic, and -NRC(O)substituted heterocyclic where R is hydrogen or alkyl and 25 wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein. 30 "Aminocarbonyloxy" refers to the groups -NRC(O)O-alkyl, WO 99/06432 PCT/US98/15325 -- 41 - -NRC(O)O-substituted alkyl, -NRC(O)O-alkenyl, -NRC(O)O-substituted alkenyl, -NRC(O)O-alkynyl, -NRC(O)O-substituted alkynyl, -NRC(O)O cycloalkyl, -NRC(O)O-substituted cycloalkyl, -NRC(O)O-aryl, -NRC(O)O substituted aryl, -NRC(O)O-heteroaryl, -NRC(O)O-substituted heteroaryl, 5 -NRC(O)O-heterocyclic, and -NRC(O)O-substituted heterocyclic where R is hydrogen or alkyl and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein. 10 "Oxycarbonylamino" refers to the groups -OC(O)NH 2 , -OC(O)NRR, -OC(O)NR-alkyl, -OC(O)NR-substituted alkyl, -OC(O)NR-alkenyl, -OC(O)NR-substituted alkenyl, -OC(O)NR-alkynyl, -OC(O)NR-substituted alkynyl, -OC(O)NR-cycloalkyl, -OC(O)NR-substituted cycloalkyl, 15 -OC(O)NR-aryl, -OC(O)NR-substituted aryl, -OC(O)NR-heteroaryl, -OC(O)NR-substituted heteroaryl,- OC(O)NR-heterocyclic, and -OC(O)NR-substituted heterocyclic where R is hydrogen, alkyl or where each R is joined to form, together with the nitrogen atom a heterocyclic or substituted heterocyclic ring and wherein alkyl, substituted alkyl, alkenyl, 20 substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein. "Oxythiocarbonylamino" refers to the groups -OC(S)NH 2 , 25 -OC(S)NRR, -OC(S)NR-alkyl, -OC(S)NR-substituted alkyl, -OC(S)NR alkenyl, -OC(S)NR-substituted alkenyl, -OC(S)NR-alkynyl, -OC(S)NR substituted alkynyl, -OC(S)NR-cycloalkyl, -OC(S)NR-substituted cycloalkyl, -OC(S)NR-aryl, -OC(S)NR-substituted aryl, -OC(S)NR-heteroaryl, OC(S)NR-substituted heteroaryl, -OC(S)NR-heterocyclic, and WO 99/06432 PCT/US98/15325 -- 42 - -OC(S)NR-substituted heterocyclic where R is hydrogen, alkyl or where each R is joined to form together with the nitrogen atom a heterocyclic or substituted heterocyclic ring and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted 5 cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein. "Aminocarbonylamino" refers to the groups -NRC(O)NRR, -NRC(O)NR-alkyl, -NRC(O)NR-substituted alkyl, -NRC(O)NR-alkenyl, 10 -NRC(O)NR-substituted alkenyl, -NRC(O)NR-alkynyl, -NRC(O)NR-substituted alkynyl, -NRC(O)NR-aryl, -NRC(O)NR-substituted aryl, -NRC(O)NR-cycloalkyl, -NRC(O)NR-substituted cycloalkyl, NRC(O)NR-heteroaryl, and -NRC(O)NR-substituted heteroaryl, NRC(O)NR-heterocyclic, and -NRC(O)NR-substituted heterocyclic where 15 each R is independently hydrogen, alkyl or where each R is joined to form together with the nitrogen atom a heterocyclic or substituted heterocyclic ring as well as where one of the amino groups is blocked by conventional blocking groups such as Boc, Cbz, formyl, and the like and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, 20 cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein. "Aminothiocarbonylamino" refers to the groups -NRC(S)NRR, 25 -NRC(S)NR-alkyl, -NRC(S)NR-substituted alkyl, -NRC(S)NR-alkenyl, -NRC(S)NR-substituted alkenyl, -NRC(S)NR-alkynyl, -NRC(S)NR substituted alkynyl, -NRC(S)NR-aryl, -NRC(S)NR-substituted aryl, -NRC(S)NR-cycloalkyl, -NRC(S)NR-substituted cycloalkyl, -NRC(S)NR heteroaryl, and -NRC(S)NR-substituted heteroaryl, -NRC(S)NR 30 heterocyclic, and -NRC(S)NR-substituted heterocyclic where each R is WO 99/06432 PCT/US98/15325 -- 43 independently hydrogen, alkyl or where each R is joined to form together with the nitrogen atom a heterocyclic or substituted heterocyclic ring as well as where one of the amino groups is blocked by conventional blocking groups such as Boc, Cbz, formyl, and the like and wherein alkyl, substituted 5 alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein. "Aryl" or "Ar" refers to an unsaturated aromatic carbocyclic group 10 of from 6 to 14 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl) which condensed rings may or may not be aromatic (e.g., 2-benzoxazolinone, 2H-1,4-benzoxazin-3(4H) one-7yl, and the like). Preferred aryls include phenyl and naphthyl. 15 Substituted aryl refers to aryl groups which are substituted with from 1 to 3 substituents selected from the group consisting of hydroxy, acyl, acylamino, thiocarbonylamino, acyloxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amidino, alkylamidino, thioamidino, amino, aminoacyl, aminocarbonyloxy, 20 aminocarbonylamino, aminothiocarbonylamino, aryl, substituted aryl, aryloxy, substituted aryloxy, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, carboxyl, carboxylalkyl, carboxyl-substituted alkyl, carboxyl-cycloalkyl, carboxyl-substituted cycloalkyl, carboxylaryl, carboxyl 25 substituted aryl, carboxylheteroaryl, carboxyl-substituted heteroaryl, carboxylheterocyclic, carboxyl-substituted heterocyclic, carboxylamido, cyano, thiol, thioalkyl, substituted thioalkyl, thioaryl, substituted thioaryl, thioheteroaryl, substituted thioheteroaryl, thiocycloalkyl, substituted thiocycloalkyl, thioheterocyclic, substituted thioheterocyclic, cycloalkyl, 30 substituted cycloalkyl, guanidino, guanidinosulfone, halo, nitro, heteroaryl, WO 99/06432 PCT/US98/15325 -- 44 substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, oxycarbonylamino, oxythiocarbonylamino, -S(O)2-alkyl, 5 -S(O) 2 -substituted alkyl, -S(O) 2 -cycloalkyl, -S(O) 2 -substituted cycloalkyl, -S(O)2-alkenyl, -S(O) 2 -substituted alkenyl, -S(O) 2 -aryl, -S(O) 2 -substituted aryl, -S(O) 2 -heteroaryl, -S(O)2-substituted heteroaryl, -S(O) 2 -heterocyclic,
-S(O)
2 -substituted heterocyclic, -OS(O) 2 -alkyl, -OS(O) 2 -substituted alkyl,
-OS(O)
2 -aryl, -OS(O) 2 -substituted aryl, -OS(O) 2 -heteroaryl, -OS(O) 2 10 substituted heteroaryl, -OS(O) 2 -heterocyclic, -OS(O) 2 -substituted heterocyclic, -OSO 2 -NRR where R is hydrogen or alkyl, -NRS(O)-alkyl,
-NRS(O)
2 -substituted alkyl, -NRS(O) 2 -aryl, -NRS(O) 2 -substituted aryl,
-NRS(O)
2 -heteroaryl, -NRS(O) 2 -substituted heteroaryl, -NRS(O) 2 heterocyclic, -NRS(O) 2 -substituted heterocyclic, -NRS(O) 2 -NR-alkyl, 15 -NRS(O) 2 -NR-substituted alkyl, -NRS(O) 2 -NR-aryl, -NRS(O) 2
-NR
substituted aryl, -NRS(O) 2 -NR-heteroaryl, -NRS(O) 2 -NR-substituted heteroaryl, -NRS(O) 2 -NR-heterocyclic, -NRS(O) 2 -NR-substituted heterocyclic where R is hydrogen or alkyl, mono- and di-alkylamino, mono and di-(substituted alkyl)amino, mono- and di-arylamino, mono- and di 20 substituted arylamino, mono- and di-heteroarylamino, mono- and di substituted heteroarylamino, mono- and di-heterocyclic amino, mono- and di-substituted heterocyclic amino, unsymmetric di-substituted amines having different substituents selected from alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted 25 heterocyclic and amino groups on the substituted aryl blocked by conventional blocking groups such as Boc, Cbz, formyl, and the like or substituted with -SO 2 NRR where R is hydrogen or alkyl. "Aryloxy" refers to the group aryl-O- which includes, by way of 30 example, phenoxy, naphthoxy, and the like.
WO 99/06432 PCT/US98/15325 -- 45 - "Substituted aryloxy" refers to substituted aryl-O- groups. "Aryloxyaryl" refers to the group -aryl-O-aryl. 5 "Substituted aryloxyaryl" refers to aryloxyaryl groups substituted with from 1 to 3 substituents on either or both aryl rings selected from the group consisting of hydroxy, acyl, acylamino, thiocarbonylamino, acyloxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amidino, alkylamidino, thioamidino, 10 amino, aminoacyl, aminocarbonyloxy, aminocarbonylamino, aminothiocarbonylamino, aryl, substituted aryl, aryloxy, substituted aryloxy, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, carboxyl, carboxylalkyl, carboxyl-substituted alkyl, carboxyl-cycloalkyl, carboxyl 15 substituted cycloalkyl, carboxylaryl, carboxyl-substituted aryl, carboxylheteroaryl, carboxyl-substituted heteroaryl, carboxylheterocyclic, carboxyl-substituted heterocyclic, carboxylamido, cyano, thiol, thioalkyl, substituted thioalkyl, thioaryl, substituted thioaryl, thioheteroaryl, substituted thioheteroaryl, thiocycloalkyl, substituted thiocycloalkyl, 20 thioheterocyclic, substituted thioheterocyclic, cycloalkyl, substituted cycloalkyl, guanidino, guanidinosulfone, halo, nitro, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, oxycarbonylamino, oxythiocarbonylamino, 25 -S(O) 2 -alkyl, -S(O) 2 -substituted alkyl, -S(O) 2 -cycloalkyl, -S(O) 2 -substituted cycloalkyl, -S(O) 2 -alkenyl, -S(O) 2 -substituted alkenyl, -S(O) 2 -aryl, -S(O) 2 substituted aryl, -S(O) 2 -heteroaryl, -S(O) 2 -substituted heteroaryl, -S(O) 2 heterocyclic, -S(O) 2 -substituted heterocyclic, -OS(O) 2 -alkyl, -OS(O) 2 substituted alkyl, -OS(O) 2 -aryl, -OS(O) 2 -substituted aryl, -OS(O) 2 -heteroaryl, WO 99/06432 PCT/US98/15325 -- 46 -
-OS(O)
2 -substituted heteroaryl, -OS(O) 2 -heterocyclic, -OS(O) 2 -substituted heterocyclic, -OSO 2 -NRR where R is hydrogen or alkyl,
-NRS(O)
2 -alkyl, -NRS(O) 2 -substituted alkyl, -NRS(O) 2 -aryl, -NRS(O) 2 substituted aryl, -NRS(O) 2 -heteroaryl, -NRS(O) 2 -substituted heteroaryl, 5 -NRS(O) 2 -heterocyclic, -NRS(O) 2 -substituted heterocyclic, -NRS(O) 2
-NR
alkyl, -NRS(O) 2 -NR-substituted alkyl, -NRS(O) 2 -NR-aryl, -NRS(O) 2
-NR
substituted aryl, -NRS(O) 2 -NR-heteroaryl, -NRS(O) 2 -NR-substituted heteroaryl, -NRS(O) 2 -NR-heterocyclic, -NRS(O) 2 -NR-substituted heterocyclic where R is hydrogen or alkyl, mono- and di-alkylamino, mono 10 and di-(substituted alkyl)amino, mono- and di-arylamino, mono- and di substituted arylamino, mono- and di-heteroarylamino, mono- and di substituted heteroarylamino, mono- and di-heterocyclic amino, mono- and di-substituted heterocyclic amino, unsymmetric di-substituted amines having different substituents selected from alkyl, substituted alkyl, aryl, substituted 15 aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic and amino groups on the substituted aryl blocked by conventional blocking groups such as Boc, Cbz, formyl, and the like or substituted with -SO 2 NRR where R is hydrogen or alkyl. 20 "Cycloalkyl" refers to cyclic alkyl groups of from 3 to 8 carbon atoms having a single cyclic ring including, by way of example, cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl and the like. Excluded from this definition are multi-ring alkyl groups such as adamantanyl, etc. 25 "Cycloalkenyl" refers to cyclic alkenyl groups of from 3 to 8 carbon atoms having single or multiple unsaturation but which are not aromatic. "Substituted cycloalkyl" and "substituted cycloalkenyl" refer to a cycloalkyl and cycloalkenyl groups, preferably of from 3 to 8 carbon atoms, 30 having from 1 to 5 substituents selected from the group consisting of oxo WO 99/06432 PCT/US98/15325 -- 47 - (=0O), thioxo (=S), alkoxy, substituted alkoxy, acyl, acylamino, thiocarbonylamino, acyloxy, amino, amidino, alkylamidino, thioamidino, aminoacyl, aminocarbonylamino, aminothiocarbonylamino, aminocarbonyloxy, aryl, substituted aryl, aryloxy, substituted aryloxy, 5 aryloxyaryl, substituted aryloxyaryl, halogen, hydroxyl, cyano, nitro, carboxyl, carboxylalkyl, carboxyl-substituted alkyl, carboxyl-cycloalkyl, carboxyl-substituted cycloalkyl, carboxylaryl, carboxyl-substituted aryl, carboxylheteroaryl, carboxyl-substituted heteroaryl, carboxylheterocyclic, carboxyl-substituted heterocyclic, cycloalkyl, substituted cycloalkyl, 10 guanidino, guanidinosulfone, thiol, thioalkyl, substituted thioalkyl, thioaryl, substituted thioaryl, thiocycloalkyl, substituted thiocycloalkyl, thioheteroaryl, substituted thioheteroaryl, thioheterocyclic, substituted thioheterocyclic, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, 15 substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, oxycarbonylamino, oxythiocarbonylamino, -OS(0) 2 -alkyl, -OS(0) 2 substituted alkyl, -OS(0) 2 -aryl, -OS(0) 2 -substituted aryl, -OS(0) 2 -heteroaryl, -OS(0) 2 -substituted heteroaryl, -OS(0) 2 -heterocyclic, -OS(0) 2 -substituted heterocyclic, -OSO 2 -NRR where R is hydrogen or alkyl, 20 -NRS(0) 2 -alkyl, -NRS(0) 2 -substituted alkyl, -NRS(0) 2 -aryl, -NRS(0) 2 substituted aryl, -NRS(0) 2 -heteroaryl, -NRS(0) 2 -substituted heteroaryl, -NRS(0) 2 -heterocyclic, -NRS(0) 2 -substituted heterocyclic, -NRS(0) 2 -NR-alkyl, -NRS(0) 2 -NR-substituted alkyl, -NRS(0) 2 -NR-aryl, -NRS(0) 2 -NR-substituted aryl, -NRS(0) 2 -NR-heteroaryl, -NRS(0) 2
-NR
25 substituted heteroaryl, -NRS(0) 2 -NR-heterocyclic, -NRS(0) 2 -NR-substituted heterocyclic where R is hydrogen or alkyl, mono- and di-alkylamino, mono and di-(substituted alkyl)amino, mono- and di-arylamino, mono- and di substituted arylamino, mono- and di-heteroarylamino, mono- and di substituted heteroarylamino, mono- and di-heterocyclic amino, mono- and 30 di-substituted heterocyclic amino, unsymmetric di-substituted amines having WO 99/06432 PCT/US98/15325 -- 48 different substituents selected from alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic and substituted alkynyl groups having amino groups blocked by conventional blocking groups such as Boc, Cbz, formyl, and the like or 5 alkynyl/substituted alkynyl groups substituted with -SO 2 -alkyl, -S02 substituted alkyl, -SO 2 -alkenyl, -SO 2 -substituted alkenyl, -SO 2 -cycloalkyl,
-SO
2 -substituted cycloalkyl, -SO 2 -aryl, -SO 2 -substituted aryl, -SO2 heteroaryl, -SO 2 -substituted heteroaryl, -SO 2 -heterocyclic, -SO 2 -substituted heterocyclic and -SO 2 NRR where R is hydrogen or alkyl. 10 "Cycloalkoxy" refers to -O-cycloalkyl groups. "Substituted cycloalkoxy" refers to -O-substituted cycloalkyl groups. "Guanidino" refers to the groups -NRC(=NR)NRR, 15 -NRC(=NR)NR-alkyl, -NRC(=NR)NR-substituted alkyl, -NRC(=NR)NR alkenyl, -NRC(=NR)NR-substituted alkenyl, -NRC(=NR)NR-alkynyl, -NRC(= NR)NR-substituted alkynyl, -NRC(= NR)NR-aryl, -NRC(= NR)NR-substituted aryl, -NRC(= NR)NR-cycloalkyl, -NRC(=NR)NR-heteroaryl, -NRC(=NR)NR-substituted heteroaryl, 20 -NRC(= NR)NR-heterocyclic, and -NRC(= NR)NR-substituted heterocyclic where each R is independently hydrogen and alkyl as well as where one of the amino groups is blocked by conventional blocking groups such as Boc, Cbz, formyl, and the like and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted 25 cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein. "Guanidinosulfone" refers to the groups -NRC(=NR)NRSO 2 -alkyl,
-NRC(=NR)NRSO
2 -substituted alkyl, -NRC(=NR)NRSO 2 -alkenyl, 30 -NRC(= NR)NRSO 2 -substituted alkenyl, -NRC(= NR)NRSO 2 -alkynyl, WO 99/06432 PCT/US98/15325 -- 49 -
-NRC(=NR)NRSO
2 -substituted alkynyl, -NRC(=NR)NRSO 2 -aryl,
-NRC(=NR)NRSO
2 -substituted aryl, -NRC(=NR)NRSO2-cycloalkyl,
-NRC(=NR)NRSO
2 -substituted cycloalkyl, -NRC(=NR)NRSO 2 -heteroaryl, and -NRC(= NR)NRSO 2 -substituted heteroaryl, -NRC(= NR)NRSO 2 5 heterocyclic, and -NRC(=NR)NRSO 2 -substituted heterocyclic where each R is independently hydrogen and alkyl and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic are as defined herein. 10 "Halo" or "halogen" refers to fluoro, chloro, bromo and iodo and preferably is either chloro or bromo. "Heteroaryl" refers to an aromatic carbocyclic group of from 2 to 10 15 carbon atoms and 1 to 4 heteroatoms selected from oxygen, nitrogen and sulfur within the ring. Such heteroaryl groups can have a single ring (e.g., pyridyl or furyl) or multiple condensed rings (e.g., indolizinyl or benzothienyl). Preferred heteroaryls include pyridyl, pyrrolyl, indolyl and furyl. 20 "Substituted heteroaryl" refers to heteroaryl groups which are substituted with from 1 to 3 substituents selected from the group consisting of hydroxy, acyl, acylamino, thiocarbonylamino, acyloxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, alkenyl, substituted alkenyl, alkynyl, 25 substituted alkynyl, amidino, alkylamidino, thioamidino, amino, aminoacyl, aminocarbonyloxy, aminocarbonylamino, aminothiocarbonylamino, aryl, substituted aryl, aryloxy, substituted aryloxy, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, carboxyl, carboxylalkyl, carboxyl-substituted 30 alkyl, carboxyl-cycloalkyl, carboxyl-substituted cycloalkyl, carboxylaryl, WO 99/06432 PCT/US98/15325 -- 50 carboxyl-substituted aryl, carboxylheteroaryl, carboxyl-substituted heteroaryl, carboxylheterocyclic, carboxyl-substituted heterocyclic, carboxylamido, cyano, thiol, thioalkyl, substituted thioalkyl, thioaryl, substituted thioaryl, thioheteroaryl, substituted thioheteroaryl, 5 thiocycloalkyl, substituted thiocycloalkyl, thioheterocyclic, substituted thioheterocyclic, cycloalkyl, substituted cycloalkyl, guanidino, guanidinosulfone, halo, nitro, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted 10 heterocyclyloxy, oxycarbonylamino, oxythiocarbonylamino, -S(O)2-alkyl,
-S(O)
2 -substituted alkyl, -S(O) 2 -cycloalkyl, -S(O) 2 -substituted cycloalkyl,
-S(O)
2 -alkenyl, -S(O) 2 -substituted alkenyl, -S(O) 2 -aryl, -S(O) 2 -substituted aryl, -S(O) 2 -heteroaryl, -S(O) 2 -substituted heteroaryl, -S(O) 2 -heterocyclic,
-S(O)
2 -substituted heterocyclic, -OS(O) 2 -alkyl, -OS(O) 2 -substituted alkyl, 15 -OS(O) 2 -aryl, -OS(O) 2 -substituted aryl, -OS(O) 2 -heteroaryl, -OS(O) 2 substituted heteroaryl, -OS(O) 2 -heterocyclic, -OS(O) 2 -substituted heterocyclic, -OS0 2 -NRR where R is hydrogen or alkyl, -NRS(O)-alkyl,
-NRS(O)
2 -substituted alkyl, -NRS(O) 2 -aryl, -NRS(O) 2 -substituted aryl,
-NRS(O)
2 -heteroaryl, -NRS(O) 2 -substituted heteroaryl, -NRS(O) 2 20 heterocyclic, -NRS(O) 2 -substituted heterocyclic, -NRS(O) 2 -NR-alkyl,
-NRS(O)
2 -NR-substituted alkyl, -NRS(O) 2 -NR-aryl, -NRS(O) 2
-NR
substituted aryl, -NRS(O) 2 -NR-heteroaryl, -NRS(O) 2 -NR-substituted heteroaryl, -NRS(O) 2 -NR-heterocyclic, -NRS(O) 2 -NR-substituted heterocyclic where R is hydrogen or alkyl, mono- and di-alkylamino, mono 25 and di-(substituted alkyl)amino, mono- and di-arylamino, mono- and di substituted arylamino, mono- and di-heteroarylamino, mono- and di substituted heteroarylamino, mono- and di-heterocyclic amino, mono- and di-substituted heterocyclic amino, unsymmetric di-substituted amines having different substituents selected from alkyl, substituted alkyl, aryl, substituted 30 aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted WO 99/06432 PCT/US98/15325 -- 51 heterocyclic and amino groups on the substituted aryl blocked by conventional blocking groups such as Boc, Cbz, formyl, and the like or substituted with -SO 2 NRR where R is hydrogen or alkyl. 5 "Heteroaryloxy" refers to the group -O-heteroaryl and "substituted heteroaryloxy" refers to the group -O-substituted heteroaryl. "Heterocycle" or "heterocyclic" refers to a saturated or unsaturated group having a single ring or multiple condensed rings, from 1 to 10 carbon 10 atoms and from 1 to 4 hetero atoms selected from nitrogen, sulfur or oxygen within the ring wherein, in fused ring systems, one or more the rings can be aryl or heteroaryl. "Saturated heterocyclic" refers to heterocycles of single or multiple 15 condensed rings lacking unsaturation in any ring (e.g., carbon to carbon unsaturation, carbon to nitrogen unsaturation, nitrogen to nitrogen unsaturation, and the like). "Unsaturated heterocyclic" refers to non-aromatic heterocycles of 20 single or multiple condensed rings having unsaturation in any ring (e.g., carbon to carbon unsaturation, carbon to nitrogen unsaturation, nitrogen to nitrogen unsaturation, and the like). "Substituted heterocyclic" refers to heterocycle groups which are 25 substituted with from 1 to 3 substituents selected from the group consisting of oxo (= O), thioxo (=S), alkoxy, substituted alkoxy, acyl, acylamino, thiocarbonylamino, acyloxy, amino, amidino, alkylamidino, thioamidino, aminoacyl, aminocarbonylamino, aminothiocarbonylamino, aminocarbonyloxy, aryl, substituted aryl, aryloxy, substituted aryloxy, 30 aryloxyaryl, substituted aryloxyaryl, halogen, hydroxyl, cyano, nitro, WO 99/06432 PCT/US98/15325 -- 52 carboxyl, carboxylalkyl, carboxyl-substituted alkyl, carboxyl-cycloalkyl, carboxyl-substituted cycloalkyl, carboxylaryl, carboxyl-substituted aryl, carboxylheteroaryl, carboxyl-substituted heteroaryl, carboxylheterocyclic, carboxyl-substituted heterocyclic, cycloalkyl, substituted cycloalkyl, 5 guanidino, guanidinosulfone, thiol, thioalkyl, substituted thioalkyl, thioaryl, substituted thioaryl, thiocycloalkyl, substituted thiocycloalkyl, thioheteroaryl, substituted thioheteroaryl, thioheterocyclic, substituted thioheterocyclic, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, 10 substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, oxycarbonylamino, oxythiocarbonylamino, -OS(O) 2 -alkyl, -OS(O) 2 substituted alkyl, -OS(O) 2 -aryl, -OS(O) 2 -substituted aryl, -OS(O) 2 -heteroaryl,
-OS(O)
2 -substituted heteroaryl, -OS(O) 2 -heterocyclic, -OS(O) 2 -substituted heterocyclic, -OS0 2 -NRR where R is hydrogen or alkyl, 15 -NRS(O) 2 -alkyl, -NRS(O) 2 -substituted alkyl, -NRS(O) 2 -aryl, -NRS(O) 2 substituted aryl, -NRS(O) 2 -heteroaryl, -NRS(O) 2 -substituted heteroaryl,
-NRS(O)
2 -heterocyclic, -NRS(O) 2 -substituted heterocyclic,
-NRS(O)
2 -NR-alkyl, -NRS(O) 2 -NR-substituted alkyl, -NRS(O) 2 -NR-aryl,
-NRS(O)
2 -NR-substituted aryl, -NRS(O) 2 -NR-heteroaryl, -NRS(O) 2
-NR
20 substituted heteroaryl, -NRS(O) 2 -NR-heterocyclic, -NRS(O) 2 -NR-substituted heterocyclic where R is hydrogen or alkyl, mono- and di-alkylamino, mono and di-(substituted alkyl)amino, mono- and di-arylamino, mono- and di substituted arylamino, mono- and di-heteroarylamino, mono- and di substituted heteroarylamino, mono- and di-heterocyclic amino, mono- and 25 di-substituted heterocyclic amino, unsymmetric di-substituted amines having different substituents selected from alkyl, substituted alkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic and substituted alkynyl groups having amino groups blocked by conventional blocking groups such as Boc, Cbz, formyl, and the like or WO 99/06432 PCT/US98/15325 -- 53 alkynyl/substituted alkynyl groups substituted with -SO 2 -alkyl, -SO 2 substituted alkyl, -SO 2 -alkenyl, -SO 2 -substituted alkenyl, -SO 2 -cycloalkyl,
-SO
2 -substituted cycloalkyl, -SO 2 -aryl, -SO2-substituted aryl, -SO2 heteroaryl, -S0 2 -substituted heteroaryl, -SO 2 -heterocyclic, -SO 2 -substituted 5 heterocyclic and -SO 2 NRR where R is hydrogen or alkyl. Examples of heterocycles and heteroaryls include, but are not limited to, azetidine, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, dihydroindole, indazole, purine, 10 quinolizine, isoquinoline, quinoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, phenanthroline, isothiazole, phenazine, isoxazole, phenoxazine, phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline, phthalimide, 1,2,3,4-tetrahydroisoquinoline, 4,5,6,7 15 tetrahydrobenzo[b]thiophene, thiazole, thiazolidine, thiophene, benzo[b]thiophene, morpholino, thiomorpholino, piperidinyl, pyrrolidine, tetrahydrofuranyl, and the like. "Saturated substituted heterocyclic" refers to substituted heterocycles 20 of single or multiple condensed rings lacking unsaturation in any ring (e.g., carbon to carbon unsaturation, carbon to nitrogen unsaturation, nitrogen to nitrogen unsaturation, and the like). "Unsaturated substituted heterocyclic" refers to non-aromatic 25 substituted heterocycles of single or multiple condensed rings having unsaturation in any ring (e.g., carbon to carbon unsaturation, carbon to nitrogen unsaturation, nitrogen to nitrogen unsaturation, and the like). "Heterocyclyloxy" refers to the group -O-heterocyclic and 30 "substituted heterocyclyloxy" refers to the group -O-substituted heterocyclic.
WO 99/06432 PCT/US98/15325 - 54 - "Thiol" refers to the group -SH. "Thioalkyl" refers to the groups -S-alkyl 5 "Substituted thioalkyl" refers to the group -S-substituted alkyl. "Thiocycloalkyl" refers to the groups -S-cycloalkyl. "Substituted thiocycloalkyl" refers to the group -S-substituted 10 cycloalkyl. "Thioaryl" refers to the group -S-aryl and "substituted thioaryl" refers t the group -S-substituted aryl. 15 "Thioheteroaryl" refers to the group -S-heteroaryl and "substituted thioheteroaryl" refers to the group -S-substituted heteroaryl. "Thioheterocyclic" refers to the group -S-heterocyclic and "substituted thioheterocyclic" refers to the group -S-substituted heterocyclic. 20 "Pharmaceutically acceptable salt" refers to pharmaceutically acceptable salts of a compound of Formula I which salts are derived from a variety of organic and inorganic counter ions well known in the art and include, by way of example only, sodium, potassium, calcium, magnesium, 25 ammonium, tetraalkylammonium, and the like; and when the molecule contains a basic functionality, salts of organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like. 30 WO 99/06432 PCT/US98/15325 -- 55 - Compound Preparation The compounds of this invention can be prepared from readily available starting materials using the following general methods and procedures. It will be appreciated that where typical or preferred process 5 conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures. 10 Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. Suitable protecting groups for various functional groups as well as suitable conditions for 15 protecting and deprotecting particular functional groups are well known in the art. For example, numerous protecting groups are described in T. W. Greene and G. M. Wuts, Protecting Groups in Organic Synthesis, Second Edition, Wiley, New York, 1991, and references cited therein. 20 Furthermore, the compounds of this invention will typically contain one or more chiral centers. Accordingly, if desired, such compounds can be prepared or isolated as pure stereoisomers, i.e., as individual enantiomers or diastereomers, or as stereoisomer-enriched mixtures. All such stereoisomers (and enriched mixtures) are included within the scope of this invention, 25 unless otherwise indicated. Pure stereoisomers (or enriched mixtures) may be prepared using, for example, optically active starting materials or stereoselective reagents well-known in the art. Alternatively, racemic mixtures of such compounds can be separated using, for example, chiral column chromatography, chiral resolving agents and the like. 30 WO 99/06432 PCT/US98/15325 -- 56 - In a preferred method of synthesis, the compounds of formula I and IA wherein Q is -C(O)NR 7 - are prepared by first coupling an amino acid of formula II: 5 R 3 I
R
2 -NH-C-COOH II H 10 wherein R 2 and R 3 are as defined above, with a sulfonyl chloride of formula III:
R
1
-S
O
2 -Cl III 15 wherein R' is as defined above, to provide an N-sulfonyl amino acid of formula IV:
R
3 20
R'-SO
2
-N(R
2 )-C-COOH IV I H 25 wherein R'-R 3 are as defined above. This reaction is typically conducted by reacting the amino acid of formula II with at least one equivalent, preferably about 1.1 to about 2 equivalents, of sulfonyl chloride III in an inert diluent such as 30 dichloromethane and the like. Generally, the reaction is conducted at a temperature ranging from about -70'C to about 40'C for about 1 to about 24 hours. Preferably, this reaction is conducted in the presence of a suitable base to scavenge the acid generated during the reaction. Suitable bases WO 99/06432 PCT/US98/15325 -- 57 include, by way of example, tertiary amines, such as triethylamine, diisopropylethylamine, N-methylmorpholine and the like. Alternatively, the reaction can be conducted under Schotten-Baumann-type conditions using aqueous alkali, such as sodium hydroxide and the like, as the base. Upon 5 completion of the reaction, the resulting N-sulfonyl amino acid IV is recovered by conventional methods including neutralization, extraction, precipitation, chromatography, filtration, and the like. The amino acids of formula II employed in the above reaction are 10 either known compounds or compounds that can be prepared from known compounds by conventional synthetic procedures. Examples of suitable amino acids for use in this reaction include, but are not limited to, L-proline, trans-4-hydroxyl-L-proline, cis-4-hydroxyl-L-proline, trans-3-phenyl-L proline, cis-3-phenyl-L-proline, L-(2-methyl)proline, L-pipecolinic acid, L 15 azetidine-2-carboxylic acid, L-indoline-2-carboxylic acid, L-1,2,3,4 tetrahydroisoquinoline-3-carboxylic acid, L-thiazolidine-4-carboxylic acid, L-(5,5-dimethyl)thiazolidine-4-carboxylic acid, L-thiamorpholine-3 carboxylic acid, glycine, 2-tert-butylglycine, D,L-phenylglycine, L-alanine, a-methylalanine, N-methyl-L-phenylalanine, L-diphenylalanine, sarcosine, 20 D,L-phenylsarcosine, L-aspartic acid P-tert-butyl ester, L-glutamic acid y tert-butyl ester, L-(O-benzyl)serine, 1-aminocyclopropanecarboxylic acid, 1 aminocyclobutanecarboxylic acid, 1-aminocyclopentanecarboxylic acid (cycloleucine) 1-aminocyclohexanecarboxylic acid, L-serine and the like. If desired, the corresponding carboxylic acid esters of the amino acids of 25 formula II, such as the methyl esters, ethyl esters and the like, can be employed in the above reaction with the sulfonyl chloride III. Subsequent hydrolysis of the ester group to the carboxylic acid using conventional reagents and conditions, i.e., treatment with an alkali metal hydroxide in an inert diluent such as methanol/water, then provides the N-sulfonyl amino 30 acid IV.
WO 99/06432 PCT/US98/15325 -- 58 - Similarly, the sulfonyl chlorides of formula III employed in the above reaction are either known compounds or compounds that can be prepared from known compounds by conventional synthetic procedures. Such compounds are typically prepared from the corresponding sulfonic acid, i.e., 5 from compounds of the formula R 1
-SO
3 H where R' is as defined above, using phosphorous trichloride and phosphorous pentachloride. This reaction is generally conducted by contacting the sulfonic acid with about 2 to 5 molar equivalents of phosphorous trichloride and phosphorous pentachloride, either neat or in an inert solvent, such as dichloromethane, at temperature in 10 the range of about 0OC to about 80 0 C for about 1 to about 48 hours to afford the sulfonyl chloride. Alternatively, the sulfonyl chlorides of formula III can be prepared from the corresponding thiol compound, i.e., from compounds of the formula R'-SH where R 1 is as defined above, by treating the thiol with chlorine (Cl 2 ) and water under conventional reaction conditions. 15 Examples of sulfonyl chlorides suitable for use in this invention include, but are not limited to, methanesulfonyl chloride, 2-propanesulfonyl chloride, 1-butanesulfonyl chloride, benzenesulfonyl chloride, 1 naphthalenesulfonyl chloride, 2-naphthalenesulfonyl chloride, p 20 toluenesulfonyl chloride, a-toluenesulfonyl chloride, 4 acetamidobenzenesulfonyl chloride, 4-amidinobenzenesulfonyl chloride, 4 tert-butylbenzenesulfonyl chloride, 4-bromobenzenesulfonyl chloride, 2 carboxybenzenesulfonyl chloride, 4-cyanobenzenesulfonyl chloride, 3,4 dichlorobenzenesulfonyl chloride, 3,5-dichlorobenzenesulfonyl chloride, 3,4 25 dimethoxybenzenesulfonyl chloride, 3,5-ditrifluoromethylbenzenesulfonyl chloride, 4-fluorobenzenesulfonyl chloride, 4-methoxybenzenesulfonyl chloride, 2-methoxycarbonylbenzenesulfonyl chloride, 4 methylamidobenzenesulfonyl chloride, 4-nitrobenzenesulfonyl chloride, 4 thioamidobenzenesulfonyl chloride, 4-trifluoromethylbenzenesulfonyl 30 chloride, 4-trifluoromethoxybenzenesulfonyl chloride, 2,4,6- WO 99/06432 PCT/US98/15325 -- 59 trimethylbenzenesulfonyl chloride, 2-phenylethanesulfonyl chloride, 2 thiophenesulfonyl chloride, 5-chloro-2-thiophenesulfonyl chloride, 2,5 dichloro-4-thiophenesulfonyl chloride, 2-thiazolesulfonyl chloride, 2-methyl 4-thiazolesulfonyl chloride, 1-methyl-4-imidazolesulfonyl chloride, 1 5 methyl-4-pyrazolesulfonyl chloride, 5-chloro-1,3-dimethyl-4 pyrazolesulfonyl chloride, 3-pyridinesulfonyl chloride, 2-pyrimidinesulfonyl chloride, and the like. If desired, a sulfonyl fluoride, sulfonyl bromide or sulfonic acid anhydride may be used in place of the sulfonyl chloride in the above reaction to form the N-sulfonyl amino acids of formula IV. 10 The intermediate N-sulfonyl amino acids of formula IV can also be prepared by reacting a sulfonamide of formula V:
R
1
'-S
O
2
-NH-R
2 V 15 wherein R 1 and R 2 are as defined above, with a carboxylic acid derivative of the formula L(R 3 )CHCOOR or where L is a leaving group, such as chloro, bromo, iodo, mesylate, tosylate and the like, R 3 is as defined above and R is 20 hydrogen or an alkyl group. This reaction is typically conducted by contacting the sulfonamide V with at least one equivalent, preferably 1.1 to 2 equivalents, of the carboxylic acid derivative in the presence of a suitable base, such as triethylamine, in an inert diluent, such as DMF, at a temperature ranging from about 24oC to about 37oC for about 0.5 to about 4 25 hours. This reaction is further described in Zuckermann et al., J. Am. Chem. Soc., 1992, 114, 10646-10647. Preferred carboxylic acid derivatives for use in this reaction are a-chloro and a-bromocarboxylic acid esters such as tert-butyl bromoacetate and the like. When an carboxylic acid ester is employed in this reaction, the ester group is subsequently hydrolyzed using 30 conventional procedures to afford an N-sulfonyl amino acid of formula IV.
WO 99/06432 PCT/US98/15325 - 60 - The compounds of formula I are then prepared by coupling the intermediate N-sulfonyl amino acid of formula IV with an amino acid derivative of formula VI: 5 0O
R'-NH-CH-C-R
6 VI I
R
5 10 wherein R-R' are as defined above and, in addition, R 6 can be hydroxyl. This coupling reaction is typically conducted using well-known coupling reagents such as carbodiimides, BOP reagent (benzotriazol-1-yloxy tris(dimethylamino)phosphonium hexafluorophosphonate) and the like. 15 Suitable carbodiimides include, by way of example, dicyclohexylcarbodiimide (DCC), 1-(3-dimethylaminopropyl)-3 ethylcarbodiimide (EDC) and the like. If desired, polymer supported forms of carbodiimide coupling reagents may also be used including, for example, those described in Tetrahedron Letters, 34(48), 7685 (1993). Additionally, 20 well-known coupling promoters, such as N-hydroxysuccinimide, 1 hydroxybenzotriazole and the like, may be used to facilitate the coupling reaction. This coupling reaction is typically conducted by contacting the N 25 sulfonylamino acid IV with about 1 to about 2 equivalents of the coupling reagent and at least one equivalent, preferably about 1 to about 1.2 equivalents, of amino acid derivative VI in an inert diluent, such as dichloromethane, chloroform, acetonitrile, tetrahydrofuran, N,N dimethylformamide and the like. Generally, this reaction is conducted at a 30 temperature ranging from about O'C to about 37'C for about 12 to about 24 hours. Upon completion of the reaction, the compound of formula I is WO 99/06432 PCT/US98/15325 -- 61 recovered by conventional methods including neutralization, extraction, precipitation, chromatography, filtration, and the like. Alternatively, the N-sulfonyl amino acid IV can be converted into an 5 acid halide and the acid halide coupled with amino acid derivative VI to provide compounds of formula I. The acid halide of VI can be prepared by contacting VI with an inorganic acid halide, such as thionyl chloride, phosphorous trichloride, phosphorous tribromide or phosphorous pentachloride, or preferably, with oxalyl chloride under conventional 10 conditions. Generally, this reaction is conducted using about 1 to 5 molar equivalents of the inorganic acid halide or oxalyl chloride, either neat or in an inert solvent, such as dichloromethane or carbon tetrachloride, at temperature in the range of about 0OC to about 80 0 C for about 1 to about 48 hours. A catalyst, such as N,N-dimethylformamide, may also be used in this 15 reaction. The acid halide of N-sulfonyl amino acid IV is then contacted with at least one equivalent, preferably about 1.1 to about 1.5 equivalents, of amino acid derivative VI in an inert diluent, such as dichloromethane, at a 20 temperature ranging from about -70oC to about 40oC for about 1 to about 24 hours. Preferably, this reaction is conducted in the presence of a suitable base to scavenge the acid generated during the reaction. Suitable bases include, by way of example, tertiary amines, such as triethylamine, diisopropylethylamine, N-methylmorpholine and the like. Alternatively, the 25 reaction can be conducted under Schotten-Baumann-type conditions using aqueous alkali, such as sodium hydroxide and the like. Upon completion of the reaction, the compound of formula I is recovered by conventional methods including neutralization, extraction, precipitation, chromatography, filtration, and the like. 30 WO 99/06432 PCT/US98/15325 -- 62 - Alternatively, the compounds of formula I can be prepared by first forming a diamino acid derivative of formula VII:
R
3
R
7 O 5 | |
R
2 -NH-C-C(0)N-CH-C-R 6 VII I I H R 5 10 wherein R 2
-R
7 are as defined above. The diamino acid derivatives of formula VII can be readily prepared by coupling an amino acid of formula II with an amino acid derivative of formula VI using conventional amino acid coupling techniques and reagents, such carbodiimides, BOP reagent and the like, as described above. Diamino acid VII can then be sulfonated using a 15 sulfonyl chloride of formula III and using the synthetic procedures described above to provide a compound of formula I. The amino acid derivatives of formula VI employed in the above reactions are either known compounds or compounds that can be prepared 20 from known compounds by conventional synthetic procedures. For example, amino acid derivatives of formula VI can be prepared by C alkylating commercially available diethyl 2-acetamidomalonate (Aldrich, Milwaukee, Wisconsin, USA) with an alkyl or substituted alkyl halide. This reaction is typically conducted by treating the diethyl 2-acetamidomalonate 25 with at least one equivalent of sodium ethoxide and at least one equivalent of an alkyl or substituted alkyl halide in refluxing ethanol about 6 to about 12 hours. The resulting C-alkylated malonate is then deacetylated, hydrolyzed and decarboxylated by heating in aqueous hydrochloric acid at reflux for about 6 to about 12 hours to provide the amino acid, typically as the 30 hydrochloride salt.
WO 99/06432 PCT/US98/15325 -- 63 - Examples of amino acid derivatives of formula VI suitable for use in the above reactions include, but are not limited to, P-tert-butyl-L-aspartic acid methyl ester, L-asparagine tert-butyl ester, E-Boc-L-lysine methyl ester, E-Cbz-L-lysine methyl ester, y-tert-butyl-L-glutamic acid methyl ester, L 5 glutamine tert-butyl ester, and the like. If desired, of course, other esters or amides of the above-described compounds may also be employed. For ease of synthesis, the compounds of formula I are typically prepared as an ester, i.e., where R 6 is an alkoxy or substituted alkoxy group 10 and the like. If desired, the ester group can be hydrolysed using conventional conditions and reagents to provide the corresponding carboxylic acid. Typically, this reaction is conducted by treating the ester with at least one equivalent of an alkali metal hydroxide, such as lithium, sodium or potassium hydroxide, in an inert diluent, such as methanol or mixtures of 15 methanol and water, at a temperature ranging about 0OC to about 24'C for about 1 to about 12 hours. Alternatively, benzyl esters may be removed by hydrogenolysis using a palladium catalyst, such as palladium on carbon. The resulting carboxylic acids may be coupled, if desired, to amines such as P-alanine ethyl ester, hydroxyamines such as hydroxylamine and N 20 hydroxysuccinimide, alkoxyamines and substituted alkoxyamines such as O methylhydroxylamine and O-benzylhydroxylamine, and the like, using conventional coupling reagents and conditions as described above. As will be apparent to those skilled in the art, other functional groups 25 present on any of the substituents of the compounds of formula I can be readily modified or derivatized either before or after the above-described coupling reactions using well-known synthetic procedures. For example, a nitro group present on a substituent of a compound of formula I or an intermediate thereof may be readily reduced by hydrogenation in the 30 presence of a palladium catalyst, such as palladium on carbon, to provide the WO 99/06432 PCT/US98/15325 -- 64 corresponding amino group. This reaction is typically conducted at a temperature of from about 20'C to about 50'C for about 6 to about 24 hours in an inert diluent, such as methanol. Compounds having a nitro group on the R 3 substituent can be prepared, for example, by using a 4 5 nitrophenylalanine derivative and the like in the above-described coupling reactions. Similarly, a pyridyl group can be hydrogenated in the presence of a platinum catalyst, such as platinum oxide, in an acidic diluent to provide the 10 corresponding piperidinyl analogue. Generally, this reaction is conducted by treating the pyridine compound with hydrogen at a pressure ranging from about 20 psi to about 60 psi, preferably about 40 psi, in the presence of the catalyst at a temperature of about 20 0 C to about 50'C for about 2 to about 24 hours in an acidic diluent, such as a mixture of methanol and aqueous 15 hydrochloric acid. Compounds having a pyridyl group can be readily prepared by using, for example, P-(2-pyridyl)-, P-(3-pyridyl)- or P-(4 pyridyl)-L-alanine derivatives in the above-described coupling reactions. Additionally, when the R 5 substituent of a compound of formula I or 20 an intermediate thereof contains a primary or secondary amino group, such amino groups can be further derivatized either before or after the above coupling reactions to provide, by way of example, amides, sulfonamides, ureas, thioureas, carbamates, secondary or tertiary amines and the like. Compounds having a primary amino group on the R 5 substituent may be 25 prepared, for example, by reduction of the corresponding nitro compound as described above. Alternatively, such compounds can be prepared by using an amino acid derivative of formula VI derived from lysine, and the like in the above-described coupling reactions.
WO 99/06432 PCT/US98/15325 -- 65 - By way of illustration, a compound of formula I or an intermediate thereof having a substituent containing a primary or secondary amino group, such as where R 5 is a 4-aminobutyl group, can be readily N-acylated using conventional acylating reagents and conditions to provide the corresponding 5 amide. This acylation reaction is typically conducted by treating the amino compound with at least one equivalent, preferably about 1.1 to about 1.2 equivalents, of a carboxylic acid in the presence of a coupling reagent such as a carbodiimide, BOP reagent (benzotriazol-1-yloxy tris(dimethylamino)phosphonium hexafluorophosphonate) and the like, in an 10 inert diluent, such as dichloromethane, chloroform, acetonitrile, tetrahydrofuran, N,N-dimethylformamide and the like, at a temperature ranging from about 0 0 C to about 37 0 C for about 4 to about 24 hours. Preferably, a promoter, such as N-hydroxysuccinimide, 1 hydroxybenzotriazole and the like, is used to facilitate the acylation reaction. 15 Examples of carboxylic acids suitable for use in this reaction include, but are not limited to, N-tert-butyloxycarbonylglycine, N-tert-butyloxycarbonyl-L phenylalanine, N-tert-butyloxycarbonyl-L-aspartic acid benzyl ester, benzoic acid, N-tert-butyloxycarbonylisonipecotic acid, N-methylisonipecotic acid, N-tert-butyloxycarbonylnipecotic acid, N-tert-butyloxycarbonyl-L 20 tetrahydroisoquinoline-3-carboxylic acid, N-(toluene-4-sulfonyl)-L-proline and the like. Alternatively, a compound of formula I or an intermediate thereof containing a primary or secondary amino group can be N-acylated using an 25 acyl halide or a carboxylic acid anhydride to form the corresponding amide. This reaction is typically conducted by contacting the amino compound with at least one equivalent, preferably about 1.1 to about 1.2 equivalents, of the acyl halide or carboxylic acid anhydride in an inert diluent, such as dichloromethane, at a temperature ranging from about of about -70 0 C to 30 about 40 0 C for about 1 to about 24 hours. If desired, an acylation catalyst WO 99/06432 PCT/US98/15325 -- 66 such as 4-(N,N-dimethylamino)pyridine may be used to promote the acylation reaction. The acylation reaction is preferably conducted in the presence of a suitable base to scavenge the acid generated during the reaction. Suitable bases include, by way of example, tertiary amines, such 5 as triethylamine, diisopropylethylamine, N-methylmorpholine and the like. Alternatively, the reaction can be conducted under Schotten-Baumann-type conditions using aqueous alkali, such as sodium hydroxide and the like. Examples of acyl halides and carboxylic acid anhydrides suitable for 10 use in this reaction include, but are not limited to, 2-methylpropionyl chloride, trimethylacetyl chloride, phenylacetyl chloride, benzoyl chloride, 2-bromobenzoyl chloride, 2-methylbenzoyl chloride, 2 trifluoromethylbenzoyl chloride, isonicotinoyl chloride, nicotinoyl chloride, picolinoyl chloride, acetic anhydride, succinic anhydride, and the like. 15 Carbamyl chlorides, such as N,N-dimethylcarbamyl chloride, N,N diethylcarbamyl chloride and the like, can also be used in this reaction to provide ureas. Similarly, dicarbonates, such as di-tert-butyl dicarbonate, may be employed to provide carbamates. 20 In a similar manner, a compound of formula I or an intermediate thereof containing a primary or secondary amino group may be N-sulfonated to form a sulfonamide using a sulfonyl halide or a sulfonic acid anhydride. Sulfonyl halides and sulfonic acid anhydrides suitable for use in this reaction include, but are not limited to, methanesulfonyl chloride, 25 chloromethanesulfonyl chloride, p-toluenesulfonyl chloride, trifluoromethanesulfonic anhydride, and the like. Similarly, sulfamoyl chlorides, such as dimethylsulfamoyl chloride, can be used to provide sulfamides (e.g., > N-SO 2 -N <).
WO 99/06432 PCT/US98/15325 -- 67 - Additionally, a primary and secondary amino group present on a substituent of a compound of formula I/IA or an intermediate thereof can be reacted with an isocyanate or a thioisocyanate to give a urea or thiourea, respectively. This reaction is typically conducted by contacting the amino 5 compound with at least one equivalent, preferably about 1.1 to about 1.2 equivalents, of the isocyanate or thioisocyanate in an inert diluent, such as toluene and the like, at a temperature ranging from about 240C to about 37oC for about 12 to about 24 hours. The isocyanates and thioisocyanates used in this reaction are commercially available or can be prepared from 10 commercially available compounds using well-known synthetic procedures. For example, isocyanates and thioisocyanates are readily prepared by reacting the appropriate amine with phosgene or thiophosgene. Examples of isocyanates and thioisocyanates suitable for use in this reaction include, but are not limited to, ethyl isocyanate, n-propyl isocyanate, 4-cyanophenyl 15 isocyanate, 3-methoxyphenyl isocyanate, 2-phenylethyl isocyanate, methyl thioisocyanate, ethyl thioisocyanate, 2-phenylethyl thioisocyanate, 3 phenylpropyl thioisocyanate, 3-(N,N-diethylamino)propyl thioisocyanate, phenyl thioisocyanate, benzyl thioisocyanate, 3-pyridyl thioisocyanate, fluorescein isothiocyanate (isomer I) and the like. 20 Furthermore, when a compound of formula I/IA or an intermediate thereof contains a primary or secondary amino group, the amino group can be reductively alkylated using aldehydes or ketones to form a secondary or tertiary amino group. This reaction is typically conducted by contacting the 25 amino compound with at least one equivalent, preferably about 1.1 to about 1.5 equivalents, of an aldehyde or ketone and at least one equivalent based on the amino compound of a metal hydride reducing agent, such as sodium cyanoborohydride, in an inert diluent, such as methanol, tetrahydrofuran, mixtures thereof and the like, at a temperature ranging from about O'C to 30 about 50 0 C for about 1 to about 72 hours. Aldehydes and ketones suitable WO 99/06432 PCT/US98/15325 - 68 for use in this reaction include, by way of example, benzaldehyde, 4 chlorobenzaldehyde, valeraldehyde, and the like. In a similar manner, when a compound of formula I/IA or an 5 intermediate thereof has a substituent containing a hydroxyl group, the hydroxyl group can be further modified or derivatized either before or after the above coupling reactions to provide, by way of example, ethers, carbamates and the like. Compounds having a hydroxyl group on, e.g., the
R
3 substituent, for example, can be prepared using an amino acid derivative 10 of formula VI derived from tyrosine and the like in the above-described reactions. By way of example, a compound of formula I/IA or an intermediate thereof having a substituent containing a hydroxyl group, such as where R 3 15 is a (4-hydroxyphenyl)methyl group, can be readily O-alkylated to form ethers. This O-alkylation reaction is typically conducted by contacting the hydroxy compound with a suitable alkali or alkaline earth metal base, such as potassium carbonate, in an inert diluent, such as acetone, 2-butanone and the like, to form the alkali or alkaline earth metal salt of the hydroxyl group. 20 This salt is generally not isolated, but is reacted in situ with at least one equivalent of an alkyl or substituted alkyl halide or sulfonate, such as an alkyl chloride, bromide, iodide, mesylate or tosylate, to afford the ether. Generally, this reaction is conducted at a temperature ranging from about 60 0 C to about 150oC for about 24 to about 72 hours. Preferably, a catalytic 25 amount of sodium or potassium iodide is added to the reaction mixture when an alkyl chloride or bromide is employed in the reaction. Examples of alkyl or substituted alkyl halides and sulfonates suitable for use in this reaction include, but are not limited to, tert-butyl 30 bromoacetate, N-tert-butyl chloroacetamide, 1-bromoethylbenzene, ethyl c- WO 99/06432 PCT/US98/15325 -- 69 bromophenylacetate, 2-(N-ethyl-N-phenylamino)ethyl chloride, 2-(N,N ethylamino)ethyl chloride, 2-(N,N-diisopropylamino)ethyl chloride, 2-(N,N dibenzylamino)ethyl chloride, 3-(N,N-ethylamino)propyl chloride, 3-(N benzyl-N-methylamino)propyl chloride, N-(2-chloroethyl)morpholine, 2 5 (hexamethyleneimino)ethyl chloride, 3-(N-methylpiperazine)propyl chloride, 1-(3-chlorophenyl)-4-(3-chloropropyl)piperazine, 2-(4-hydroxy-4 phenylpiperidine)ethyl chloride, N-tert-butyloxycarbonyl-3-piperidinemethyl tosylate, and the like. 10 Alternatively, a hydroxyl group present on a substituent of a compound of formula I or an intermediate thereof can be O-alkylating using the Mitsunobu reaction. In this reaction, an alcohol, such as 3-(N,N dimethylamino)-1-propanol and the like, is reacted with about 1.0 to about 1.3 equivalents of triphenylphosphine and about 1.0 to about 1.3 equivalents 15 of diethyl azodicarboxylate in an inert diluent, such as tetrahydrofuran, at a temperature ranging from about -10 0 C to about 5'C for about 0.25 to about 1 hour. About 1.0 to about 1.3 equivalents of a hydroxy compound, such as N-tert-butyltyrosine methyl ester, is then added and the reaction mixture is stirred at a temperature of about 0°C to about 30'C for about 2 to about 48 20 hours to provide the O-alkylated product. In a similar manner, a compound of formula I or an intermediate thereof containing a aryl hydroxy group can be reacted with an aryl iodide to provide a diaryl ether. Generally, this reaction is conducted by forming the 25 alkali metal salt of the hydroxyl group using a suitable base, such as sodium hydride, in an inert diluent such as xylenes at a temperature of about -25 'C to about 10oC. The salt is then treated with about 1.1 to about 1.5 equivalents of cuprous bromide dimethyl sulfide complex at a temperature ranging from about 10 0 C to about 30 0 C for about 0.5 to about 2.0 hours, 30 followed by about 1.1 to about 1.5 equivalents of an aryl iodide, such as WO 99/06432 PCT/US98/15325 -- 70 sodium 2-iodobenzoate and the like. The reaction is then heated to about 70'C to about 150oC for about 2 to about 24 hours to provide the diaryl ether. 5 Additionally, a hydroxy-containing compound can also be readily derivatized to form a carbamate. In one method for preparing such carbamates, a hydroxy compound of formula I or an intermediate thereof is contacted with about 1.0 to about 1.2 equivalents of 4-nitrophenyl chloroformate in an inert diluent, such as dichloromethane, at a temperature 10 ranging from about -25 0 C to about 0 0 C for about 0.5 to about 2.0 hours. Treatment of the resulting carbonate with an excess, preferably about 2 to about 5 equivalents, of a trialkylamine, such as triethylamine, for about 0.5 to 2 hours, followed by about 1.0 to about 1.5 equivalents of a primary or secondary amine provides the carbamate. Examples of amines suitable for 15 using in this reaction include, but are not limited to, piperazine, 1 methylpiperazine, 1-acetylpiperazine, morpholine, thiomorpholine, pyrrolidine, piperidine and the like. Alternatively, in another method for preparing carbamates, a 20 hydroxy-containing compound is contacted with about 1.0 to about 1.5 equivalents of a carbamyl chloride in an inert diluent, such as dichloromethane, at a temperature ranging from about 25 0 C to about 70 0 C for about 2 to about 72 hours. Typically, this reaction is conducted in the presence of a suitable base to scavenge the acid generated during the 25 reaction. Suitable bases include, by way of example, tertiary amines, such as triethylamine, diisopropylethylamine, N-methylmorpholine and the like. Additionally, at least one equivalent (based on the hydroxy compound) of 4 (N,N-dimethylamino)pyridine is preferably added to the reaction mixture to facilitate the reaction. Examples of carbamyl chlorides suitable for use in WO 99/06432 PCT/US98/15325 -- 71 this reaction include, by way of example, dimethylcarbamyl chloride, diethylcarbamyl chloride and the like. Likewise, when a compound of formula I/IA or an intermediate 5 thereof contains a primary or secondary hydroxyl group, such hydroxyl groups can be readily converted into a leaving group and displaced to form, for example, amines, sulfides and fluorides. For example, derivatives of 4 hydroxy-L-proline can be converted into the corresponding 4-amino, 4-thio or 4-fluoro-L-proline derivatives via nucleophilic displacement of the 10 derivatized hydroxyl group. Generally, when a chiral compound is employed in these reactions, the stereochemistry at the carbon atom attached to the derivatized hydroxyl group is typically inverted. These reactions are typically conducted by first converting the 15 hydroxyl group into a leaving group, such as a tosylate, by treatment of the hydroxy compound with at least one equivalent of a sulfonyl halide, such as p-toluenesulfonyl chloride and the like, in pyridine. This reaction is generally conducted at a temperature of from about 0OC to about 70 0 C for about 1 to about 48 hours. The resulting tosylate can then be readily 20 displaced with sodium azide, for example, by contacting the tosylate with at least one equivalent of sodium azide in an inert diluent, such as a mixture of N,N-dimethylformamide and water, at a temperature ranging from about 0OC to about 37oC for about 1 to about 12 hours to provide the corresponding azido compound. The azido group can then be reduced by, for example, 25 hydrogenation using a palladium on carbon catalyst to provide the amino (
NH
2 ) compound. Similarly, a tosylate group can be readily displaced by a thiol to form a sulfide. This reaction is typically conducted by contacting the tosylate with 30 at least one equivalent of a thiol, such as thiophenol, in the presence of a WO 99/06432 PCT/US98/15325 -- 72 suitable base, such as 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU), in an inert diluent, such as N,N-dimethylformamide, at a temperature of from about 0 0 C to about 37 0 C for about 1 to about 12 hours to provide the sulfide. Additionally, treatment of a tosylate with morpholinosulfur trifluoride in an 5 inert diluent, such as dichloromethane, at a temperature ranging from about 0OC to about 37oC for about 12 to about 24 hours affords the corresponding fluoro compound. Furthermore, a compound of formula I/IA or an intermediate thereof 10 having a substituent containing an iodoaryl group, for example, when R 3 is a (4-iodophenyl)methyl group, can be readily converted either before or after the above coupling reactions into a biaryl compound. Typically, this reaction is conducted by treating the iodoaryl compound with about 1.1 to about 2 equivalents of an arylzinc iodide, such as 2 15 (methoxycarbonyl)phenylzinc iodide, in the presence of a palladium catalyst, such as palladium tetra(triphenylphosphine), in an inert diluent, such as tetrahydrofuran, at a temperature ranging from about 24oC to about 30 0 C until reaction completion. This reaction is further described, for example, in Rieke, J. Org. Chem. 1991, 56, 1445. 20 In some cases, the compounds of formula I/IA or intermediates thereof may contain substituents having one or more sulfur atoms. Such sulfur atoms will be present, for example, when the amino acid of formula II employed in the above reactions is derived from L-thiazolidine-4-carboxylic 25 acid, L-(5,5-dimethyl)thiazolidine-4-carboxylic acid, L-thiamorpholine-3 carboxylic acid and the like. When present, such sulfur atoms can be oxidized either before or after the above coupling reactions to provide a sulfoxide or sulfone compound using conventional reagents and reaction conditions. Suitable reagents for oxidizing a sulfide compound to a sulfoxide 30 include, by way of example, hydrogen peroxide, 3-chloroperoxybenzoic acid WO 99/06432 PCT/US98/15325 -- 73 - (MCPBA), sodium periodate and the like. The oxidation reaction is typically conducted by contacting the sulfide compound with about 0.95 to about 1.1 equivalents of the oxidizing reagent in an inert diluent, such as dichloromethane, at a temperature ranging from about -50oC to about 75 C 5 for about 1 to about 24 hours. The resulting sulfoxide can then be further oxidized to the corresponding sulfone by contacting the sulfoxide with at least one additional equivalent of an oxidizing reagent, such as hydrogen peroxide, MCPBA, potassium permanganate and the like. Alternatively, the sulfone can be prepared directly by contacting the sulfide with at least two 10 equivalents, and preferably an excess, of the oxidizing reagent. Such reactions are described further in March, "Advanced Organic Chemistry", 4th Ed., pp. 1201-1202, Wiley publisher, 1992. As described above, the compounds of formula I having an R 15 substituent other an hydrogen can be prepared using an N-substituted amino acid of formula II, such as sarcosine, N-methyl-L-phenylalanine and the like, in the above-described coupling reactions. Alternatively, such compounds can be prepared by N-alkylation of a sulfonamide of formula I or IV (where
R
2 is hydrogen) using conventional synthetic procedures. Typically, this N 20 alkylation reaction is conducted by contacting the sulfonamide with at least one equivalent, preferably 1.1 to 2 equivalents, of an alkyl or substituted alkyl halide in the presence of a suitable base, such as potassium carbonate, in an inert diluent, such as acetone, 2-butanone and the like, at a temperature ranging from about 25 0 C to about 70oC for about 2 to about 48 hours. 25 Examples of alkyl or substituted alkyl halides suitable for use in this reaction include, but are not limited to, methyl iodide, and the like. Additionally, the sulfonamides of formula I or IV wherein R 2 is hydrogen and R 1 is a 2-alkoxycarbonylaryl group can be intramolecularly 30 cyclized to form 1,2-benzisothiazol-3-one derivatives or analogues thereof.
WO 99/06432 PCT/US98/15325 -- 74 - This reaction is typically conducted by treating a sulfonamide, such as N-(2 methoxycarbonylphenylsulfonyl)glycine-L-phenylalanine benzyl ester, with about 1.0 to 1.5 equivalents of a suitable base, such as an alkali metal hydride, in a inert diluent, such as tetrahydrofuran, at a temperature ranging 5 from about 0 0 C to about 30oC for about 2 to about 48 hours to afford the cyclized 1,2-benzisothiazol-3-one derivative. Lastly, the compounds of formula I where Q is -C(S)NR 7 - are can prepared by using an amino thionoacid derivative in place of amino acid II in 10 the above described synthetic procedures. Such amino thionoacid derivatives can be prepared by the procedures described in Shalaky, et al., J. Org. Chem., 61:9045-9048 (1996) and Brain, et al., J. Org. Chem., 62:3808 3809 (1997) and references cited therein. 15 Pharmaceutical Formulations When employed as pharmaceuticals, the compounds of formula I and IA are usually administered in the form of pharmaceutical compositions. These compounds can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and 20 intranasal. These compounds are effective as both injectable and oral compositions. Such compositions are prepared in a manner well known in the pharmaceutical art and comprise at least one active compound. This invention also includes pharmaceutical compositions which 25 contain, as the active ingredient, one or more of the compounds of formula I and IA above associated with pharmaceutically acceptable carriers. In making the compositions of this invention, the active ingredient is usually mixed with an excipient, diluted by an excipient or enclosed within such a carrier which can be in the form of a capsule, sachet, paper or other 30 container. When the excipient serves as a diluent, it can be a solid, semi- WO 99/06432 PCT/US98/15325 -- 75 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 5 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. In preparing a formulation, it may be necessary to mill the active 10 compound to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it ordinarily is milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size is normally adjusted by milling to provide a substantially uniform distribution in the 15 formulation, e.g. about 40 mesh. Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, 20 polyvinylpyrrolidone, cellulose, 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 25 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. The compositions are preferably formulated in a unit dosage form, 30 each dosage containing from about 5 to about 100 mg, more usually about 10 WO 99/06432 PCT/US98/15325 -- 76 to about 30 mg, of the active ingredient. 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 5 association with a suitable pharmaceutical excipient. The active compound is effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It, will be understood, however, that the amount of the compound actually administered 10 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, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like. 15 For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation 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 20 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. This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, 0.1 to about 500 mg of the active ingredient of the present invention. 25 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 30 envelope over the former. The two components can separated by enteric WO 99/06432 PCT/US98/15325 -- 77 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 5 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 10 suspensions, and flavored emulsions with edible oils such as 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 15 suspensions in pharmaceutically 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 20 pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device may be attached to a face masks tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from 25 devices which deliver the formulation in an appropriate manner. The following formulation examples illustrate the pharmaceutical compositions of the present invention. 30 WO 99/06432 PCT/US98/15325 -- 78 - Formulation Example 1 Hard gelatin capsules containing the following ingredients are prepared: Quantity 5 Ingredient (mg/capsule) Active Ingredient 30.0 Starch 305.0 Magnesium stearate 5.0 10 The above ingredients are mixed and filled into hard gelatin capsules in 340 mg quantities. Formulation Example 2 15 A tablet formula is prepared using the ingredients below: Quantity Ingredient (mg/tablet) Active Ingredient 25.0 20 Cellulose, microcrystalline 200.0 Colloidal silicon dioxide 10.0 Stearic acid 5.0 The components are blended and compressed to form tablets, each 25 weighing 240 mg. Formulation Example 3 A dry powder inhaler formulation is prepared containing the following components: 30 Ingredient Weight % Active Ingredient 5 Lactose 95 35 WO 99/06432 PCT/US98/15325 -- 79 - The active mixture is mixed with the lactose and the mixture is added to a dry powder inhaling appliance. Formulation Example 4 5 Tablets, each containing 30 mg of active ingredient, are prepared as follows: Quantity Ingredient (mg/tablet) 10 Active Ingredient 30.0 mg Starch 45.0 mg Microcrystalline cellulose 35.0 mg Polyvinylpyrrolidone (as 10% solution in water) 4.0 mg 15 Sodium carboxymethyl starch 4.5 mg Magnesium stearate 0.5 mg Talc 1.0 mg Total 120 mg 20 The active ingredient, starch and cellulose are passed through a No. 20 mesh U.S. sieve and mixed thoroughly. The solution of polyvinyl pyrrolidone is mixed with the resultant powders, which are then passed through a 16 mesh U.S. sieve. The granules so produced are dried at 500 to 60'C and passed through a 16 mesh U.S. sieve. The sodium carboxymethyl 25 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 150 mg. Formulation Example 5 30 Capsules, each containing 40 mg of medicament are made as follows: Quantity Ingredient (mg/capsule) Active Ingredient 40.0 mg 35 Starch 109.0 mg Magnesium stearate 1.0 mg Total 150.0 mg WO 99/06432 PCT/US98/15325 -- 80 - The active ingredient, cellulose, starch, an magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 150 mg quantities. 5 Formulation Example 6 Suppositories, each containing 25 mg of active ingredient are made as follows: 10 Ingredient Amount Active Ingredient 25 mg Saturated fatty acid glycerides to 2,000 mg 15 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 20 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 follows: 25 Ingredient Amount Active Ingredient 50.0 mg Xanthan gum 4.0 mg 30 Sodium carboxymethyl cellulose (11%) Microcrystalline cellulose (89%) 50.0 mg Sucrose 1.75 g Sodium benzoate 10.0 mg Flavor and Color q.v. 35 Purified water to 5.0 ml WO 99/06432 PCT/US98/15325 -- 81 - The medicament, 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 5 water and added with stirring. Sufficient water is then added to produce the required volume. Formulation Example 8 Quantity 10 Ingredient (mg/capsule) Active Ingredient 15.0 mg Starch 407.0 mg Magnesium stearate 3.0 mg 15 Total 425.0 mg The active ingredient, cellulose, starch, and magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into hard 20 gelatin capsules in 560 mg quantities. Formulation Example 9 An intravenous formulation may be prepared as follows: 25 Ingredient Ouantity Active Ingredient 250.0 mg Isotonic saline 1000 ml 30 Formulation Example 10 A topical formulation may be prepared as follows: Ingredient Quantity 35 Active Ingredient 1-10 g Emulsifying Wax 30 g WO 99/06432 PCT/US98/15325 -- 82 - Liquid Paraffin 20 g White Soft Paraffin to 100 g 5 The white soft paraffin is heated until molten. The liquid paraffin and emulsifying wax are incorporated and stirred until dissolved. The active ingredient is added and stirring is continued until dispersed. The mixture is then cooled until solid. 10 Another preferred formulation employed 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 15 pharmaceutical agents is well known in the art. See. e.g., U.S. Patent 5,023,252, issued June 11, 1991, herein incorporated by reference. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents. 20 When it is 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 25 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 30 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 WO 99/06432 PCT/US98/15325 -- 83 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. 5 Utility The compounds of this invention can be employed to bind VLA-4 (A3 1 integrin) in biological samples and, accordingly have utility in, for example, assaying such samples for VLA-4. In such assays, the compounds can be 10 bound to a solid support and the VLA-4 sample added thereto. The amount of VLA-4 in the sample can be determined by conventional methods such as use of a sandwich ELISA assay. Alternatively, labeled VLA-4 can be used in a competitive assay to measure for the presence of VLA-4 in the sample. Other suitable assays are well known in the art. 15 In addition, certain of the compounds of this invention inhibit, in vivo, adhesion of leukocytes to endothelial cells mediated by VLA-4 and, accordingly, can be used in the treatment of diseases mediated by VLA-4. Such diseases include inflammatory diseases in mammalian patients 20 such as asthma, Alzheimer's disease, atherosclerosis, AIDS dementia, diabetes (including acute juvenile onset diabetes), inflammatory bowel disease (including ulcerative colitis and Crohn's disease), multiple sclerosis, rheumatoid arthritis, tissue transplantation, tumor metastasis, meningitis, encephalitis, stroke, and other cerebral traumas, nephritis, retinitis, atopic 25 dermatitis, psoriasis, myocardial ischemia and acute leukocyte-mediated lung injury such as that which occurs in adult respiratory distress syndrome. The biological activity of the compounds identified above may be assayed in a variety of systems. For example, a compound can be 30 immobilized on a solid surface and adhesion of cells expressing VLA-4 can WO 99/06432 PCT/US98/15325 -- 84 be measured. Using such formats, large numbers of compounds can be screened. Cells suitable for this assay include any leukocytes known to express VLA-4 such as T cells, B cells, monocytes, eosinophils, and basophils. A number of leukocyte cell lines can also be used, examples 5 include Jurkat and U937. The test compounds can also be tested for the ability to competitively inhibit binding between VLA-4 and VCAM-1, or between VLA-4 and a labeled compound known to bind VLA-4 such as a compound of this 10 invention or antibodies to VLA-4. In these assays, the VCAM-1 can be immobilized on a solid surface. VCAM-1 may also be expressed as a recombinant fusion protein having an Ig tail (e.g., IgG) so that binding to VLA-4 may be detected in an immunoassay. Alternatively, VCAM-1 expressing cells, such as activated endothelial cells or VCAM-1 transfected 15 fibroblasts can be used. For assays to measure the ability to block adhesion to brain endothelial cells, the assays described in International Patent Application Publication No. WO 91/05038 are particularly preferred. This application is incorporated herein by reference in its entirety. 20 Many assay formats employ labelled assay components. The labelling systems can be in a variety of forms. The label may be coupled directly or indirectly to the desired component of the assay according to methods well known in the art. A wide variety of labels may be used. The component may be labelled by any one of several methods. The most common method 25 of detection is the use of autoradiography with 3 H, 1251, 35 S, 14 C, or 32p labelled compounds or the like. Non-radioactive labels include ligands which bind to labelled antibodies, fluorophores, chemiluminescent agents, enzymes and antibodies which can serve as specific binding pair members for a labelled ligand. The choice of label depends on sensitivity required, WO 99/06432 PCT/US98/15325 -- 85 ease of conjugation with the compound, stability requirements, and available instrumentation. Appropriate in vivo models for demonstrating efficacy in treating 5 inflammatory responses include EAE (experimental autoimmune encephalomyelitis) in mice, rats, guinea pigs or primates, as well as other inflammatory models dependent upon a4 integrins. Compounds having the desired biological activity may be modified as 10 necessary to provide desired properties such as improved pharmacological properties (e.g., in vivo stability, bio-availability), or the ability to be detected in diagnostic applications. For instance, inclusion of one or more D-amino acids in the sulfonamides of this invention typically increases in vivo stability. Stability can be assayed in a variety of ways such as by 15 measuring the half-life of the proteins during incubation with peptidases or human plasma or serum. A number of such protein stability assays have been described (see, e.g., Verhoef, et al., Eur. J. Drug Metab. Pharmacokinet., 1990, 15.1(2):83-93). 20 For diagnostic purposes, a wide variety of labels may be linked to the compounds, which may provide, directly or indirectly, a detectable signal. Thus, the compounds of the subject invention may be modified in a variety of ways for a variety of end purposes while still retaining biological activity. In addition, various reactive sites may be introduced at the terminus for 25 linking to particles, solid substrates, macromolecules, or the like. Labeled compounds can be used in a variety of in vivo or in vitro applications. A wide variety of labels may be employed, such as radionuclides (e.g., gamma-emitting radioisotopes such as technetium-99 or 30 indium-111), fluorescers (e.g., fluorescein), enzymes, enzyme substrates, WO 99/06432 PCT/US98/15325 - 86 enzyme cofactors, enzyme inhibitors, chemiluminescent compounds, bioluminescent compounds, and the like. Those of ordinary skill in the art will know of other suitable labels for binding to the complexes, or will be able to ascertain such using routine experimentation. The binding of these 5 labels is achieved using standard techniques common to those of ordinary skill in the art. In vitro uses include diagnostic applications such as monitoring inflammatory responses by detecting the presence of leukocytes expressing 10 VLA-4. The compounds of this invention can also be used for isolating or labeling such cells. In addition, as mentioned above, the compounds of the invention can be used to assay for potential inhibitors of VLA-4/VCAM-1 interactions. 15 For in vivo diagnostic imaging to identify, e.g., sites of inflammation, radioisotopes are typically used in accordance with well known techniques. The radioisotopes may be bound to the peptide either directly or indirectly using intermediate functional groups. For instance, chelating agents such as diethylenetriaminepentacetic acid (DTPA) and ethylenediaminetetraacetic 20 acid (EDTA) and similar molecules have been used to bind proteins to metallic ion radioisotopes. The complexes can also be labeled with a paramagnetic isotope for purposes of in vivo diagnosis, as in magnetic resonance imaging (MRI) or 25 electron spin resonance (ESR), both of which were well known. In general, any conventional method for visualizing diagnostic imaging can be used. Usually gamma- and positron-emitting radioisotopes are used for camera imaging and paramagnetic isotopes are used for MRI. Thus, the compounds can be used to monitor the course of amelioration of an inflammatory 30 response in an individual. By measuring the increase or decrease in WO 99/06432 PCT/US98/15325 -- 87 lymphocytes expressing VLA-4 it is possible to determine whether a particular therapeutic regimen aimed at ameliorating the disease is effective. The pharmaceutical compositions of the present invention can be used to 5 block or inhibit cellular adhesion associated with a number of diseases and disorders. For instance, a number of inflammatory disorders are associated with integrins or leukocytes. Treatable disorders include, e.g., transplantation rejection (e.g., allograft rejection), Alzheimer's disease, atherosclerosis, AIDS dementia, diabetes (including acute juvenile onset 10 diabetes), retinitis, cancer metastases, rheumatoid arthritis, acute leukocyte-mediated lung injury (e.g., adult respiratory distress syndrome), asthma, nephritis, and acute and chronic inflammation, including atopic dermatitis, psoriasis, myocardial ischemia, and inflammatory bowel disease (including Crohn's disease and ulcerative colitis). In preferred embodiments 15 the pharmaceutical compositions are used to treat inflammatory brain disorders, such as multiple sclerosis (MS), viral meningitis and encephalitis. Inflammatory bowel disease is a collective term for two similar diseases referred to as Crohn's disease and ulcerative colitis. Crohn's disease is an 20 idiopathic, chronic ulceroconstrictive inflammatory disease characterized by sharply delimited and typically transmural involvement of all layers of the bowel wall by a granulomatous inflammatory reaction. Any segment of the gastrointestinal tract, from the mouth to the anus, may be involved, although the disease most commonly affects the terminal ileum and/or colon. 25 Ulcerative colitis is an inflammatory response limited largely to the colonic mucosa and submucosa. Lymphocytes and macrophages are numerous in lesions of inflammatory bowel disease and may contribute to inflammatory injury.
WO 99/06432 PCT/US98/15325 - 88 - Asthma is a disease characterized by increased responsiveness of the tracheobronchial tree to various stimuli potentiating paroxysmal constriction of the bronchial airways. The stimuli cause release of various mediators of inflammation from IgE-coated mast cells including histamine, eosinophilic 5 and neutrophilic chemotactic factors, leukotrines, prostaglandin and platelet activating factor. Release of these factors recruits basophils, eosinophils and neutrophils, which cause inflammatory injury. Atherosclerosis is a disease of arteries (e.g., coronary, carotid, aorta 10 and iliac). The basic lesion, the atheroma, consists of a raised focal plaque within the intima, having a core of lipid and a covering fibrous cap. Atheromas compromise arterial blood flow and weaken affected arteries. Myocardial and cerebral infarcts are a major consequence of this disease. Macrophages and leukocytes are recruited to atheromas and contribute to 15 inflammatory injury. Rheumatoid arthritis is a chronic, relapsing inflammatory disease that primarily causes impairment and destruction of joints. Rheumatoid arthritis usually first affects the small joints of the hands and feet but then may 20 involve the wrists, elbows, ankles and knees. The arthritis results from interaction of synovial cells with leukocytes that infiltrate from the circulation into the synovial lining of the joints. See e.g., Paul, Immunology (3d ed., Raven Press, 1993). 25 Another indication for the compounds of this invention is in treatment of organ or graft rejection mediated by VLA-4. Over recent years there has been a considerable improvement in the efficiency of surgical techniques for transplanting tissues and organs such as skin, kidney, liver, heart, lung, pancreas and bone marrow. Perhaps the principal outstanding problem is the 30 lack of satisfactory agents for inducing immunotolerance in the recipient to WO 99/06432 PCT/US98/15325 -- 89the transplanted allograft or organ. When allogeneic cells or organs are transplanted into a host (i.e., the donor and donee are different individuals from the same species), the host immune system is likely to mount an immune response to foreign antigens in the transplant (host-versus-graft 5 disease) leading to destruction of the transplanted tissue. CD8 + cells, CD4 cells and monocytes are all involved in the rejection of transplant tissues. Compounds of this invention which bind to alpha-4 integrin are useful, inter alia, to block alloantigen-induced immune responses in the donee thereby preventing such cells from participating in the destruction of the transplanted 10 tissue or organ. See, e.g., Paul et al., Transplant International 9, 420-425 (1996); Georczynski et al., Immunology 87, 573-580 (1996); Georcyznski et al., Transplant. Immunol. 3, 55-61 (1995); Yang et al., Transplantation 60, 71-76 (1995); Anderson et al., APMIS 102, 23-27 (1994). 15 A related use for compounds of this invention which bind to VLA-4 is in modulating the immune response involved in "graft versus host" disease (GVHD). See e.g., Schlegel et al., J. Immunol. 155, 3856-3865 (1995). GVHD is a potentially fatal disease that occurs when immunologically competent cells are transferred to an allogeneic recipient. In this situation, 20 the donor's immunocompetent cells may attack tissues in the recipient. Tissues of the skin, gut epithelia and liver are frequent targets and may be destroyed during the course of GVHD. The disease presents an especially severe problem when immune tissue is being transplanted, such as in bone marrow transplantation; but less severe GVHD has also been reported in 25 other cases as well, including heart and liver transplants. The therapeutic agents of the present invention are used, inter alia, to block activation of the donor T-cells thereby interfering with their ability to lyse target cells in the host.
WO 99/06432 PCT/US98/15325 -- 90 - A further use of the compounds of this invention is inhibiting tumor metastasis. Several tumor cells have been reported to express VLA-4 and compounds which bind VLA-4 block adhesion of such cells to endothelial cells. Steinback et al., Urol. Res. 23, 175-83 (1995); Orosz et al., Int. J. 5 Cancer 60, 867-71 (1995); Freedman et al., Leuk. Lymphoma 13, 47-52 (1994); Okahara et al., Cancer Res. 54, 3233-6 (1994). A further use of the compounds of this invention is in treating multiple sclerosis. Multiple sclerosis is a progressive neurological autoimmune 10 disease that affects an estimated 250,000 to 350,000 people in the United States. Multiple sclerosis is thought to be the result of a specific autoimmune reaction in which certain leukocytes attack and initiate the destruction of myelin, the insulating sheath covering nerve fibers. In an animal model for multiple sclerosis, murine monoclonal antibodies directed 15 against VLA-4 have been shown to block the adhesion of leukocytes to the endothelium, and thus prevent inflammation of the central nervous system and subsequent paralysis in the animals 6 . Pharmaceutical compositions of the invention are suitable for use in a 20 variety of drug delivery systems. Suitable formulations for use in the present invention are found in Remington's Pharmaceutical Sciences, Mace Publishing Company, Philadelphia, PA, 17th ed. (1985). In order to enhance serum half-life, the compounds may be 25 encapsulated, introduced into the lumen of liposomes, prepared as a colloid, or other conventional techniques may be employed which provide an extended serum half-life of the compounds. A variety of methods are available for preparing liposomes, as described in, e.g., Szoka, et al., U.S. Patent Nos. 4,235,871, 4,501,728 and 4,837,028 each of which is 30 incorporated herein by reference.
WO 99/06432 PCT/US98/15325 -- 91 - The amount administered to the patient will vary depending upon what is being administered, the purpose of the administration, such as prophylaxis or therapy, the state of the patient, the manner of administration, and the like. In therapeutic applications, compositions are administered to a patient 5 already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. An amount adequate to accomplish this is defined as "therapeutically effective dose." Amounts effective for this use will depend on the disease condition being treated as well as by the judgment of the attending clinician depending 10 upon factors such as the severity of the inflammation, the age, weight and general condition of the patient, and the like. The compositions administered to a patient are in the form of pharmaceutical compositions described above. These compositions may be 15 sterilized by conventional sterilization techniques, or may be sterile filtered. The resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the compound preparations typically will be between 3 and 11, more preferably from 5 to 9 20 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts. The therapeutic dosage of the compounds of the present invention will 25 vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. For example, for intravenous administration, the dose will typically be in the range of about 20 kg to about 500 gg per kilogram body weight, preferably 30 about 100 Mg to about 300 /ag per kilogram body weight. Suitable dosage WO 99/06432 PCT/US98/15325 -- 92 ranges for intranasal administration are generally about 0.1 pg to 1 mg per kilogram body weight. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems. 5 The following synthetic and biological examples are offered to illustrate this invention and are not to be construed in any way as limiting the scope of this invention. Unless otherwise stated, all temperatures are in degrees Celsius. 10 EXAMPLES In the examples below, the following abbreviations have the following meanings. If an abbreviation is not defined, it has its generally accepted meaning. 15 aq or aq. = aqueous AcOH = acetic acid bd = broad doublet bm = broad multiplet bs = broad singlet 20 Bn = benzyl Boc = N-tert-butoxylcarbonyl Boc 2 0 = di-tert-butyl dicarbonate BOP = benzotriazol-1-yloxy tris(dimethylamino)phosphonium 25 hexafluorophosphate Cbz = carbobenzyloxy CHC1 3 = chloroform
CH
2 C1 2 = dichloromethane (COC1) 2 = oxalyl chloride 30 d = doublet dd = doublet of doublets dt = doublet of triplets DBU = 1,8-diazabicyclo[5.4.0]undec-7-ene DCC = 1,3-dicyclohexylcarbodiimide 35 DMAP = 4-N,N-dimethylaminopyridine DME = ethylene glycol dimethyl ether DMF = N,N-dimethylformamide DMSO = dimethylsulfoxide WO 99/06432 PCT/US98/15325 -- 93 - EDC 1-(3-dimethylaminopropyl)-3 ethylcarbodiimide hydrochloride Et 3 N = triethylamine Et 2 0 = diethyl ether 5 EtOAc = ethyl acetate EtOH = ethanol eq or eq. = equivalent Fmoc = N-(9-fluorenylmethoxycarbonyl) FmocONSu = N-(9-fluorenylmethoxycarbonyl) 10 succinimide g = grams h = hour H20 = water HBr = hydrobromic acid 15 HCI = hydrochloric acid HOBT = 1-hydroxybenzotriazole hydrate hr = hour
K
2
CO
3 = potassium carbonate L = liter 20 m = multiplet MeOH = methanol mg = milligram MgSO 4 = magnesium sulfate mL = milliliter 25 mm = millimeter mM = millimolar mmol = millimol mp = melting point N = normal 30 NaCl = sodium chloride Na 2
CO
3 = sodium carbonate NaHCO 3 = sodium bicarbonate NaOEt = sodium ethoxide NaOH = sodium hydroxide 35 NH 4 C1 = ammonium chloride NMM = N-methylmorpholine Phe = L-phenylalanine Pro = L-proline psi = pounds per square inch 40 PtO 2 = platinum oxide q = quartet quint. = quintet rt = room temperature s = singlet 45 sat = saturated WO 99/06432 PCT/US98/15325 -- 94 t = triplet t-BuOH = tert-butanol TFA = trifluoroacetic acid THF = tetrahydrofuran 5 TLC or tic = thin layer chromatography Ts = tosyl TsCl = tosyl chloride TsOH = tosylate L = microliter 10 In the examples below, all temperatures are in degrees Celcius (unless otherwise indicated). The following Methods were used to prepare the compounds set forth below as indicated. 15 Method 1 N-Tosylation Procedure N-Tosylation of the appropriate amino acid was conducted via the 20 method of Cupps, Boutin and Rapoport J. Org. Chem. 1985, 50, 3972. Method 2 Methyl Ester Preparation Procedure Amino acid methyl esters were prepared using the method of Brenner 25 and Huber Helv. Chim. Acta 1953, 36, 1109. Method 3 BOP Coupling Procedure The desired dipeptide ester was prepared by the reaction of a suitable N 30 protected amino acid (1 equivalent) with the appropriate amino acid ester or amino acid ester hydrochloride (1 equivalent), benzotriazol-1-yloxy tris(dimethylamino)phosphonium hexafluorophosphate [BOP] (2.0 equivalent), triethylamine (1.1 equivalent), and DMF. The reaction mixture WO 99/06432 PCT/US98/15325 - 95 was stirred at room temperature overnight. The crude product is purified flash chromatography to afford the dipeptide ester. Method 4 5 Hydrogenation Procedure I Hydrogenation was performed using 10% palladium on carbon (10% by weight) in methanol at 30 psi overnight. The mixture was filtered through a pad of Celite and the filtrate concentrated to yield the desired amino compound. 10 Method 5 Hydrolysis Procedure I To a chilled (0'C) THF/H 2 0 solution (2:1, 5 - 10 mL) of the appropriate ester was added LiOH (or NaOH) (0.95 equivalents). The 15 temperature was maintained at 0oC and the reaction was complete in 1-3 hours. The reaction mixture was extracted with ethyl acetate and the aqueous phase was lyophilized resulting in the desired carboxylate salt. Method 6 20 Ester Hydrolysis Procedure II To a chilled (0OC) THF/H 2 0 solution (2:1, 5 - 10 mL) of the appropriate ester was added LiOH (1.1 equivalents). The temperature was maintained at 00C and the reaction was complete in 1-3 hours. The reaction mixture was concentrated and the residue was taken up into H20 and the pH 25 adjusted to 2-3 with aqueous HC1. The product was extracted with ethyl acetate and the combined organic phase was washed with brine, dried over MgSO 4 , filtered and concentrated to yield the desired acid. Method 7 30 Ester Hydrolysis Procedure III WO 99/06432 PCT/US98/15325 -- 96 - The appropriate ester was dissolved in dioxane/H 2 0 (1:1) and 0.9 equivalents of 0.5 N NaOH was added. The reaction was stirred for 3-16 hours and than concentrated. The resulting residue was dissolved in IO and extracted with ethyl acetate. The aqueous phase was lyophilized to yield 5 the desired carboxylate sodium salt. Method 8 Sulfonylation Procedure I To the appropriately protected aminophenylalanine analog (11.2 mmol), 10 dissolved in methylene chloride (25ml) and cooled to -78 'C was added the desired sulfonyl chloride (12 mmol) followed by dropwise addition of pyridine (2 mL). The solution was allowed to warm to room temperature and was stirred for 48 hr. The reaction solution was transferred to a 250 mL separatory funnel with methylene chloride (100 mL) and extracted with 1N 15 HCI (50 mL x 3), brine (50 mL), and water (100 mL). The organic phase was dried (MgSO4) and the solvent concentrated to yield the desired product. Method 9 20 Reductive Amination Procedure Reductive amination of Tos-Pro-p-NH2-Phe with the appropriate aldehyde was conducted using acetic acid, sodium triacetoxyborohydride, methylene chloride and the combined mixture was stirred at room temperature overnight. The crude product was purified by flash 25 chromatography. Method 10 BOC Removal Procedure Anhydrous hydrochloride (HC1) gas was bubbled through a methanolic 30 solution of the appropriate Boc-amino acid ester at 0 0 C for 15 minutes and WO 99/06432 PCT/US98/15325 - 97 the reaction mixture was stirred for three hours. The solution was concentrated to a syrup and dissolved in Et 2 0 and reconcentrated. This procedure was repeated and the resulting solid was placed under high vacuum overnight. 5 Method 11 Tert-Butyl Ester Hydrolysis Procedure I The tert-butyl ester was dissolved in CH 2 C1 2 and treated with TFA. The reaction was complete in 1-3 hr at which time the reaction mixture was 10 concentrated and the residue dissolved in H20 and lyophilized to yield the desired acid. Method 12 EDC Coupling Procedure I 15 To a CH 2 C1 2 solution (5-20 mL) of N-(toluene-4-sulfonyl)-L-proline (1 equivalent), the appropriate amino acid ester hydrochloride (1 equivalent), N-methylmorpholine (1.1-2.2 equivalents) and 1-hydroxybenzotriazole (2 equivalents) were mixed, placed in an ice bath and 1-(3 dimethylaminopropyl)-3-ethyl carbodiimide (1.1 equivalents) added. The 20 reaction was allowed to rise to room temperature and stirred overnight. The reaction mixture was poured into H20 and the organic phase was washed with sat. NaHCO 3 , brine, dried (MgSO 4 or Na 2
SO
4 ), filtered and concentrated. The crude product was purified by column chromatography. 25 Method 13 EDC Coupling Procedure II To a DMF solution (5-20 mL) of the appropriate N-protected amino acid (1 equivalent), the appropriated amino acid ester hydrochloride (1 equivalent), Et 3 N (1.1 equivalents) and 1-hydroxybenzotriazole (2 30 equivalents) were mixed, placed in an ice batch and 1-(3- WO 99/06432 PCT/US98/15325 -- 98 dimethylaminopropyl)-3-ethyl carbodiimide (1.1 equivalents) added. The reaction was allowed to rise to room temperature and stirred overnight. The reaction mixture was partitioned between EtOAc and H20 and the organic phase washed with 0.2 N citric acid, H20, sat. NaHCO 3 , brine, dried 5 (MgSO 4 or Na 2
SO
4 ), filtered and concentrated. The crude product was purified by column chromatography or preparative TLC. Method 14 Sulfonylation Procedure II 10 The appropriate sulfonyl chloride was dissolved in CH 2 C1 2 and placed in an ice bath. L-Pro-L-Phe-OMe o HCI (1 equivalent) and Et 3 N (1.1 equivalent) was added and the reaction allowed to warm to room temperature and stirred overnight under an atmosphere of nitrogen. The reaction mixture was concentrated and the residue partitioned between EtOAc and H20 and 15 the organic phase washed with sat. NaHCO 3 , brine, dried (MgSO 4 or Na 2
SO
4 ), filtered and concentrated. The crude product was purified by column chromatography or preparative TLC. Method 15 20 Sulfonylation Procedure III To a solution of L-Pro-L-4-(3-dimethylaminopropyloxy)-Phe-OMe [prepared using the procedure described in Method 10] (1 equivalent) in
CH
2 Cl 2 was added Et 3 N (5 equivalents) followed by the appropriate sulfonyl chloride (1.1 equivalent). The reaction was allowed to warm to room 25 temperature and stirred overnite under an atmosphere of nitrogen. The mixture was concentrated, dissolved in EtOAc, washed with sat. NaHCO 3 and 0.2 N citric acid. The aqueous phase was made basic with solid NaHCO 3 and the product extracted with EtOAc. The organic phase was washed with brine, dried (MgSO 4 or Na 2
SO
4 ), filtered and concentrated.
WO 99/06432 PCT/US98/15325 -- 99 - The crude methyl ester was purified by preparative TLC. The corresponding acid was prepared using the procedure described in Method 7. Method 16 5 Hydrogenation Procedure II To a methanol (10 -15 mL) solution of the azlactone, was added NaOAc (1 equivalent) and 10% Pd/C. This mixture was placed on the hydrogenator at 40 psi H2. After 8 - 16 hours, the reaction mixture was filtered through a pad of Celite and the filtrate concentrated to yield the dehydrodipeptide 10 methyl ester. The ester was dissolved in dioxane/H 2 0 (5- 10 mL), to which was added 0.5 N NaOH (1.05 equivalents). After stirring for 1- 3 hours, the reaction mix was concentrated and the residue was redissolved in H20 and washed with EtOAc. The aqueous phase was made acidic with 0.2 N HCI and the product was extracted with EtOAc. The combined organic phase 15 was washed with brine (1 x 5 mL), dried (MgSO 4 or Na 2
SO
4 ), filtered and concentrated to yield the acid as approximately a 1:1 mixture of diastereomers. Method 17 20 Tert-Butyl Ester Hydrolysis Procedure II The tert-butyl ester was dissolved in CH 2 C1 2 (5 mL) and treated with TFA (5 mL). The reaction was complete in 1-3 hours at which time the reaction mixture was concentrated and the residue dissolved in H20 and concentrated. The residue was redissolved in H20 and lyophilized to yield 25 the desired product.
WO 99/06432 PCT/US98/15325 -- 100 - Example 1 Synthesis of N-(Toluene-4-sulfonyl)-L-prolyl-L aspartic Acid 4-tert-Butyl Ester 5 N-(Toluene-4-sulfonyl)-L-proline hydrate was coupled to P-tert-butyl L aspartic acid methyl ester hydrochloride using the procedure described in Method 3. The title compound was prepared, via hydrolysis of the methyl ester using LiOH in THF/water, as a solid, mp = 153-155oC. 10 NMR data was as follows: 1H NMR (CDC13): 6 = 7.76 (d, 2H, J = 8.2 Hz), 7.73 (s, 1H), 7.36 (d, 2H, J = 8.0 Hz), 4.82 (m, 1H), 4.14 (m, 1H), 3.52 (m, 1H), 3.21 (m, 1H), 3.00 (dd, 1H, J = 4.8, 16.9 Hz), 2.84 (dd, 1H, J = 5.0, 16.9 Hz), 2.45 (s, 3H), 2.14 (m, 1H), 1.77-1.50 (3H), 1.47 (s, 9H). 15 13C NMR (CDCl 3 ): 6 = 174.5, 172.3, 170.2, 145.0, 133.5, 130.6, 128.5, 82.8, 62.7, 50.3, 49.7, 37.9, 30.7, 28.6, 25.0, 22.2. Mass Spectroscopy: FAB m/e 441 (M+H). Example 2 20 Synthesis of N-(Toluene-4-sulfonyl)-L-prolyl-L-asparagine N-(Toluene-4-sulfonyl)-L-proline hydrate was coupled to L-asparagine tert-butyl ester hydrochloride using the procedure described in Method 3. 25 The title compound was prepared, via cleavage of the tert-butyl ester using trifluoroacetic acid in CH 2 C1 2 , to provide a solid, mp = 178-180'C. NMR data was as follows: 'H NMR (DMSO-d 6 ): 6 = 12.6 (s, 1H), 8.15 (d, 2H, J = 8.0 Hz), 7.75 (d, 2H, J = 8.2 Hz), 7.43 (d, 2H, J = 8.4 Hz), 7.40 (bs, 1H), 6.95 (bs, 30 1H), 4.50 (m, 1H), 4.15 (dd, 1H, J = 2.8, 8.5 Hz), 3.34 (m, 1H), 3.12 (m, 1H), 2.53 (m, 2H), 2.41 (s, 3H), 1.76 (m, 2H), 1.50 (m, 2H).
WO 99/06432 PCT/US98/15325 -- 101 - 13C NMR (DMSO-d 6 ): 8 = 172.8, 171.7, 171.2, 144.0, 134.3, 130.2, 127.9, 61.7, 49.3, 49.1, 36.9, 30.8, 24.2, 21.4. Mass Spectroscopy: FAB m/e 384 (M + H). 5 Example 3 Synthesis of N-(Toluene-4-sulfonyl)-L-prolyl Ne-(tert-butoxycarbonyl)-L-lysine 10 N-(Toluene-4-sulfonyl)-L-proline hydrate was coupled to Ne-Boc-lysine methyl ester hydrochloride using the procedure described in Method 3. The title compound was prepared via hydrolysis of the methyl ester using LiOH in THF/water. NMR data was as follows: 15 'H NMR (CDCl 3 ): 6 = 7.77 (d, 2H, J = 8.2 Hz), 7.56 (m, 1H), 7.35 (d, 2H, J = 8.0 Hz), 4.77 (bs, 1H), 4.57 (m, 1H), 4.15 (m, 1H), 3.57 (m, 1H), 3.20 (m, 1H), 3.10 (m, 2H), 2.44 (s, 3H), 2.13 (m, 1H), 1.99 (m, 1H), 1.83 (m, 2H), 1.77-1.44 (6H), 1.41 (s, 9H). 13C NMR (CDCl 3 ): 6 = 175.0, 172.3, 156.8, 145.0, 133.5, 128.5, 20 79.8, 62.9, 53.0, 50.3, 40.8, 32.1, 32.0, 30.6, 29.9, 29.0, 25.1, 23.1, 22.2. Mass Spectroscopy: FAB m/e 498 (M + H). Example 4 Synthesis of 25 N-(Toluene-4-sulfonyl)-L-prolyl L-glutamic Acid y-tert-Butyl Ester N-(Toluene-4-sulfonyl)-L-proline hydrate was coupled to y-tert-butyl L glutamic acid methyl ester hydrochloride using the procedure described in 30 Method 3. The title compound was prepared, via hydrolysis of the methyl ester using LiOH in THF/water, as a solid, mp = 164-166oC. NMR data was as follows: WO 99/06432 PCT/US98/15325 -- 102 - 'H NMR (CDCl 3 ): 8 = 7.76 (d, 2H, J = 8.3 Hz), 7.71 (d, 1H11, J = 7.4 Hz), 7.36 (d, 2H, J = 8.3 Hz), 4.56 (dd, 1H, J = 1.8, 7.0 Hz), 4.12 (dd, 1H, J = 3.2, 8.5 Hz), 3.59 (m, 1H), 3.23 (m, 1H), 2.45 (s, 3H), 2.43 (m, 2H), 2.25-2.07 (3H), 1.79 (m, 1H), 1.68 (m, 2H), 1.46 (s, 9H). 5 13C NMR (CDC1 3 ): 8 = 174.6, 173.1, 172.8, 145.0, 133.4, 130.6, 128.5, 81.7, 62.8, 52.7, 50.4, 32.1, 30.8, 28.6, 27.6, 25.1, 22.2. Mass Spectroscopy: FAB m/e 455 (M+H). Example 5 10 Synthesis of N-(Toluene-4-sulfonyl)-L-prolyl-L-glutamine N-(Toluene-4-sulfonyl)-L-proline hydrate was coupled to L-glutamine tert-butyl ester hydrochloride using the procedure described in Method 3. 15 The title compound, prepared via cleavage of the tert-butyl ester using trifluoroacetic acid in CH 2 C1 2 , was recovered as a solid, mp = 45-55°C. NMR data was as follows: 'H NMR (DMSO-d 6 ): 8 = 8.22 (d, 1H, J = 7.7 Hz), 7.75 (d, 2H, J = 8.3 Hz), 7.44 (d, 2H, J = 8.0 Hz), 7.27 (bs, 1H), 6.81 (bs, 1H11), 4.15 (m, 20 2H), 3.38 (m, 1H), 3.12 (m, 1H), 2.41 (s, 3H), 2.15 (m, 2H), 1.96 (m, 1H), 1.78 (m, 1H), 1.64 (m, 4H), 1.47 (m, 1H). 13C NMR (CD 3 OD): 8 = 178, 3, 175,3, 174, 9, 146.3, 135.7, 131.6, 129.5, 63.7, 53.8, 51.2, 33.1, 32.5, 29.0, 26.2, 22.1. Mass Spectroscopy: FAB m/e 398 (M+H). 25 Example 6 Synthesis of N-(Toluene-4-sulfonyl)-L-prolyl-Ne-(carbobenzyloxy)-L-lysine 30 N-(Toluene-4-sulfonyl)-L-proline hydrate was coupled to NE-Cbz-L lysine methyl ester hydrochloride using the procedure described in Method WO 99/06432 PCT/US98/15325 -- 103 - 3. The title compound was prepared via hydrolysis of the methyl ester using LiOH in THF/water. NMR data was as follows: 'H NMR (CDC1 3 ): 8 = 7.75 (d, 2H, J = 8.2 Hz), 7.60 (m ,1H), 7.32 5 (7H), 5.14 (m, 1H), 5.07 (s, 2H), 4.57 (m, 1H), 4.13 (m, 1H), 3.55 (m, 1H), 3.18 (m, 3H), 2.43 (s, 3H), 2.14 (m, 1H), 1.96 (m, 1H), 1.84 (m, 1H), 1.79 (m, 1H), 1.57 (m, 4H), 1.41 (m, 2H). 13C NMR (CDCl 3 ): 8 = 175.0, 172.4, 157.3, 145.0, 137.2, 133.4, 130.6, 129.0, 128.6, 128.5, 128.4, 67.2, 62.8, 53.0, 50.4, 41.3, 32.0, 10 30.6, 29.7, 25.1, 23.0, 22.2. Mass Spectroscopy: FAB m/e 532 (M+H). Example 7 Synthesis of 15 N-(Toluene-4-sulfonyl)-L-prolyl-O-benzyl-L-serine N-(Toluene-4-sulfonyl)-L-proline hydrate was coupled to O-benzylserine methyl ester hydrochloride using the procedure described in Method 3. The title compound was prepared via hydrolysis of the methyl ester using LiOH 20 in THF/water. NMR data was as follows: 1H NMR (CDC1 3 ): 6 = 7.75 (m, 3H11), 7.32 (6H), 4.73 (m, 1H), 4.60 (d, 1H, J = 12.3 Hz), 4.51 (d, 1H, J = 12.3 Hz), 4.19 (dd, 1H, J = 3.0, 8.5 Hz), 3.95 (dd, 1H, J = 3.7, 9.6 Hz), 3.78 (m, 1H), 3.50 (m, 1H), 3.20 25 (m, 1H11), 2.43 (s, 3H), 2.11 (m, 1H), 1.64 (3H). 13C NMR (CDC13): 6 = 173.0, 171.8, 144.4, 137.3, 132.9, 130.0, 128.5, 128.4, 127.9, 127.7, 73.2, 69.0, 62.1, 52.8, 49.7, 30.0, 24.4, 21.5. Mass Spectroscopy: FAB m/e 447 (M + H). 30 WO 99/06432 PCT/US98/15325 -- 104 - Example 8 Synthesis of N-(Toluene-4-sulfonyl)-L prolyl-p-cyclohexyl-L-alanine 5 N-(Toluene-4-sulfonyl)-L-proline hydrate was coupled to P cyclohexylalanine methyl ester hydrochloride using the procedure described in Method 3. The title compound was prepared via hydrolysis of the methyl ester using LiOH in THF/water. 10 NMR data was as follows: 'H NMR (CDC13): 6 = 7.76 (d, 2H, J = 8.2 Hz), 7.36 (d, 2H, J = 8.0 Hz), 6.95 (bs, 1H), 4.60 (m, 1H), 4.17 (dd, 1H, J = 2.5, 8.7 Hz), 3.77 (m, 1H), 3.54 (m, 1H), 3.21 (m, 1H), 2.45 (s, 3H), 2.21 (m, 1H), 1.89-0.90 (15H). 15 13C NMR (CDCl 3 ): 8 = 175.8, 171.5, 144.4, 132.8, 130.0, 127.8, 62.2, 50.6, 49.7, 39.1, 34.3, 33.4, 32.3, 29.6, 26.3, 26.1, 26.0, 24.4, 21.6. Mass Spectroscopy: FAB m/e 423 (M + H). Preparative Example A 20 Synthesis of N-(Toluene-4-sulfonyl)-L-prolyl D-glutamic acid y-tert-Butyl Ester N-Benzyloxycarbonyl-D-glutamic acid 5-tert-butyl ester was converted 25 to the methyl ester using the procedure described in Method 2. y-tert-Butyl D-glutamic methyl ester was prepared from the product of the previous step utilizing the procedure described in Method 4. N-(Toluene-4-sulfonyl)-L proline hydrate was coupled to the resulting y-tert-butyl-D-glutamic methyl ester utilizing the procedure described in Method 3. The title compound was 30 prepared via hydrolysis of the methyl ester using the procedure described in Method 6. The product was isolated as a white solid, mp = 50 0 C. NMR data was as follows: WO 99/06432 PCT/US98/15325 -- 105 - 'H NMR (CDC1 3 , 300 MHz): 6 = 7.73 (d, 2H, J = 10.1 Hz); 7.58 (d, 1H, J = 8.3 Hz); 7.35 (d, 2H, J = 8.2 Hz); 6.05 (bs, 1H); 4.63 (m, 1H); 4.10 (m, 1H); 3.62 (m, 1H); 2.44 (m, 5H), 2.30 (m, 1H); 2.05 (m, 2H); 1.80 (m, 1 H); 1.61 (m, 2H); 1.45 (s, 9H). 5 13C NMR (CDC1 3 , 75 MHz): 6 = 175.1, 173.1, 172.8, 145.1, 133.1, 130.6, 128.5, 81.7, 62.9, 52.3, 50.6, 31.9, 31.2, 28.6, 27.6, 24.8, 22.2. Mass Spectroscopy: (PI-FAB) 455, (M+H)*. Preparative Example B 10 Synthesis of N-(Toluene-4-sulfonyl)-L-prolyl D-asparatic Acid P-tert-Butyl Ester N-Benzyloxycarbonyl-D-aspartic acid 4-tert-butyl ester was converted to 15 the methyl ester using the procedure described in Method 2. P-tert-Butyl-D aspartic methyl ester was prepared from the product of the previous step utilizing the procedure described in Method 4. N-(Toluene-4-sulfonyl)-L proline hydrate was coupled to the resulting P-tert-Butyl-D-aspartic methyl ester utilizing the procedure described in Method 3. The title compound was 20 prepared via hydrolysis of the methyl ester using the procedure described in Method 6. The product was isolated as a white solid, mp = 55'C. NMR data was as follows: 1H NMR(CDC13, 300 MHz): 6 = 7.82 (d, 1H, J = 9.1 Hz); 7.72 (d, 2H, J = 8.2 Hz); 7.35 (d, 2H., J = 7.0 Hz); 4.90 (m, 1H); 4.15 (m, 1H); 25 3.61 (m, 1H); 3.15 (m, 1H); 2.75 (m, 1H); 2.44 (s. 3H); 2.15 (m, 1 H); 1.80 (m, 1H); 1.65 (m, 2H); 1.48 (s, 9H). 13C NMR(CDC1 3 , 75 MHz): 6 = 144.9, 13175.3, 172.4, 170.4, 0.6, 128.5, 83.14, 63.0, 50.5, 48.9, 37.8, 31.2, 28.6, 24.8, 22.2. Mass Spectroscopy: (PI-FAB) 441, (M +H) . 30 WO 99/06432 PCT/US98/15325 -- 106 - Preparative Example C Synthesis of N-(Toluene-4-sulfonyl)sarcosyl 0-(N-benzylpiperidin-4-yl)-D,L-alanine 5 N-(Toluene-4-sulfonyl)sarcosyl-p-(pyrid-4-yl)-D,L-alanine methyl ester was employed in this reaction and was prepared as follows. Sodium metal (2 eq.) was dissolved in EtOH containing diethyl acetamidomalonate (1 eq.) and 4-picolylchloride hydrochloride (1 eq.). The mixture was heated to reflux 10 for 6 hr, and then cooled and filtered to remove NaCl (washed with EtOH). The solvent was removed in vacuo and the mixture was taken up into saturated aq NaHCO 3 and extracted with EtOAc. The solvent was removed and the residue purified by silica gel flash chromatography (95:5
CH
2 C1 2 /MeOH) to give diethyl 2-(4-pyridylmethyl)-2-acetamidomalonate. 15 Diethyl 2-(4-pyridylmethyl)-2-acetamidomalonate was dissolved in 6N HCI and heated to reflux for about 19 hr whereupon it was cooled to room temperature and the HCI solution was removed by evaporation in vacuo. The intermediate amino acid dihydrochloride salt was taken up into MeOH 20 saturated with HCI gas and stirred for 3.5 hr. The MeOH/HCl was removed by evaporation in vacuo to give P-(3-pyridyl)alanine methyl ester dihydrochloride (2.235 g, 100%). N-(Toluene-4-sulfonyl)sarcosine was coupled to 3-(4-pyridyl)alanine 25 methyl ester dihydrochloride using the procedure described in Method 3 to give N-(toluene-4-sulfonyl)sarcosyl-p-(4-pyridyl)alanine. N-(Toluene-4-sulfonyl)sarcosyl-D,L-P-(4-pyridyl)alanine methyl ester (266 mg, 0.656 mmol) was dissolved in methanol (6 mL) and 12 N HC1 (273 30 itL) and PtO 2 (25 mg) were added. The mixture was hydrogenated at 40 psi
H
2 for 4 hr. The mixture was filtered and the solvent was removed in vacuo WO 99/06432 PCT/US98/15325 -- 107 to give N-(toluene-4-sulfonyl)sarcosyl-D,L-P-(4-piperidinyl)alanine methyl ester hydrochloride (260 mg, 88%). N-(Toluene-4-sulfonyl)sarcosyl-p-(piperidin-4-yl)-D,L-alanine methyl 5 ester hydrochloride (180 mg, 0.402 mmol) was dissolved in MeOH (3 mL). Triethylamine (56 ItL), benzaldehyde (53 mg, 0.502 mmol) and 1.0 M NaBH 3 CN in THF (400 yL) were added and the mixture was stirred for 5 hr. 1N HCI (3 mL) was added and the mixture was stirred for 5 minutes before diluting with saturated aqueous NaHCO 3 (30 mL). The mixture was 10 extracted with EtOAc (3 x 25 mL) and the combined extracts were dried (Na 2
SO
4 ), filtered and evaporated in vacuo to give a residue which was purified by silica gel flash chromatography (95:5:0.5
CH
2 Cl 2 /MeOH/NH 4 OH) to give N-(toluene-4-sulfonyl)sarcosyl-3-(N benzylpiperidin-4-yl)-D,L-alanine methyl ester (105 mg, 52%). The title 15 compound was prepared via hydrolysis of the methyl ester using 0.5 N aqueous NaOH in THF/water as a very hygroscopic solid. NMR data was as follows: 'H NMR (CD 3 OD): 6 = 7.73 (d, 2H, J = 8.2 Hz), 7.43 (d, 2H, J = 8.2 Hz), 7.33 (min, 5H), 4.34 (min, 1H), 3.74 (d, 1H, J = 16.5 Hz), 3.62 (s, 20 2H), 3.58 (d, 1H, J = 16.5 Hz), 2.94 (min, 2H), 2.79 (s, 3H), 2.44 (s, 3H), 2.17 (min, 1H), 1.90-1.26 (8H). 13C NMR (CD 3 OD): 6 = 179.6, 170.0, 146.1, 137.6, 135.7, 131.8, 131.6, 130.0, 129.4, 64.4, 55.0, 54.6, 41.3, 37.8, 34.0, 33.5, 32.4, 22.1. Mass Spectroscopy: FAB m/e 488 (M+H). 25 30 WO 99/06432 PCT/US98/15325 -- 108 - Example 9 Synthesis of N-(Toluene-4-sulfonyl)sarcosyl P-(N-tert-butoxycarbonylpiperidin-4-yl)-D,L-alanine 5 N-(Toluene-4-sulfonyl)sarcosyl-p-(4-piperidinyl)-D,L-alanine methyl ester hydrochloride (see Preparative Example C (109) above) (0.257 mmol) was dissolved in CHC1 3 (3 mL). Triethylamine (43 AL) and di-tert-butyl dicarbonate (67 mg, 0.309 mmol) were added and the mixture was stirred 10 for 1 hr. The mixture was diluted with saturated aqueous NaHCO 3 (20 mL) and extracted with CHC1 3 (2 x 20 mL). The extracts were dried (Na 2
SO
4 ), filtered and evaporated in vacuo to give a residue which was purified by silica gel flash chromatography (95:5 CH 2
C
2 /MeOH) to yield N-(toluene-4 sulfonyl)sarcosyl-p-(N-tert-butoxycarbonylpiperidin-4-yl)-D,L-alanine 15 methyl ester (121 mg, 92%). The title compound was prepared via hydrolysis of the methyl ester using LiOH in THF/water (70 mg, 99%). NMR data was as follows: 'H NMR (CDC13): 8 = 7.69 (d, 2H, J = 8.2 Hz), 7.37 (d, 2H, J = 8.0 Hz), 7.14 (d, 1H, J = 8.7 Hz), 6.60 (bs, 1H), 4.69 (min, 1H), 4.09 (min, 2H), 20 3.91 (d, 1H, J = 16.7 Hz), 3.42 (d, 1H, J = 16.5 Hz), 2.81 (s, 3H), 2.68 (min, 2H), 2.45 (s, 3H), 1.89-1.08 (7H), 1.45 (s, 9H). 13C NMR (CDCl 3 ): 8 = 175.1, 168.3, 155.0, 144.5, 132.7, 130.0, 127.6, 79.7, 77.2, 54.0, 49.6, 38.5, 37.0, 32.5, 32.1, 31.1, 28.4, 21.6. Mass Spectroscopy: FAB m/e 504 (M+Li). 25 Example 10 Synthesis of N-(Toluene-4-sulfonyl)sarcosyl P-(N-benzoylpiperidin-4-yl)-D,L-alanine 30 N-(Toluene-4-sulfonyl)sarcosyl-p-(piperidin-4-yl)-D,L-alanine methyl ester hydrochloride (see Preparative Example C (109)) (65 mg, 0.15 mmol) was dissolved in CHCl1 3 (3 mL) and cooled in an ice bath. Triethylamine (61 WO 99/06432 PCT/US98/15325 -- 109 - /uL) and benzoyl chloride (21 mg, 0.15 mmol) were added and the mixture was stirred for 1 hr. The mixture was diluted with 1N HCl (20 mL) and extracted with EtOAc (2 x 25 mL). The extracts were dried (Na 2
SO
4 ), filtered and evaporated in vacuo to give a residue which was purified by 5 silica gel flash chromatography (95:5 CH 2 Cl 2 /MeOH) to yield N-(toluene-4 sulfonyl)sarcosyl-p-(N-benzoylpiperidin-4-yl)-D,L-alanine methyl ester (67 mg, 89%). The title compound was prepared via hydrolysis of the methyl ester using LiOH in THF/water (71 mg, 100%). NMR data was as follows: 10 1 H NMR (CDC1 3 ): 6 7.68 (d, 2H, J = 8.3 Hz), 7.39 (m, 7H), 7.17 (d, 1H, J = 8.3 Hz), 6.55 (bs, 1H), 4.68 (m, 2H), 3.92 (d, 1H, J = 16.5 Hz), 3.72 (m, 1H), 3.37 (d, 1H, J = 16.7 Hz), 3.00 (m, 1H), 2.78 (m, 1H), 2.80 (s, 3H), 2.45 (s, 3H), 2.90-1.60 (5H), 1.20 (m, 2H). 13C NMR (CDC1 3 ): 8 = 174.4, 170.7, 168.2, 144.5, 135.6, 132.7, 15 130.0, 129.7, 128.4, 127.6, 126.8, 77.3, 77.2, 54.0, 49.6, 42.5, 37.0, 32.5, 21.6, Mass Spectroscopy: FAB m/e 502 (M+H). Example 11 20 Synthesis of N-(Toluene-4-sulfonyl)sarcosyl Nc-tert-butoxycarbonyl-L-lysine N-(Toluene-4-sulfonyl)sarcosine was coupled to NE-Boc-L-lysine methyl 25 ester hydrochloride using the procedure described in Method 3 to give N (toluene-4-sulfonyl)sarcosyl-NE-Boc-L-lysine methyl ester. The title compound was prepared via hydrolysis of the methyl ester using LiOH in THF/water. NMR data was as follows: 30 1 H NMR (DMSO-d6): 8 = 8.21 (d, 1H, J = 7.7 Hz), 7.67 (d, 2H, J = 7.7 Hz), 7.43 (d, 2H, J = 8.2 Hz), 6.76 (m, 1H), 4.13 (m, 1H), 3.69 (s, WO 99/06432 PCT/US98/15325 -- 110- 2H), 3.34 (bs, 1H), 2.88 (min, 2H), 2.69 (s, 3H), 2.41 (s, 3H), 1.68 (min, 1H), 1.58 (min, 1H), 1.37 (s, 9H), 1.27 (min, 2H).
'
3 C NMR (DMSO-d 6 ): 6 = 173.7, 167.4, 155.9, 143.7, 134.4, 130.1, 127.7, 77.7, 52.3, 52.2, 39.8, 36.1, 31.0, 29.4, 28.6, 23.1, 21.4. 5 Mass Spectroscopy: FAB m/e 478 (M+Li). Example 12 Synthesis of N-(Toluene-4-sulfonyl)-L-prolyl 10 Ne-tert-butoxycarbonyl-D-lysine A CH 2 Cl 2 solution of Nac-Cbz-NE-Boc-D-lysine was chilled to -15 0 C (dry ice/CH 3 CN bath) to which was added diethylisopropylamine (1.5 eq.), methanol (3.0 eq.) and BOP (1.1 eq) to form the methyl ester. The 15 reaction was allowed to warm temperature and stirred overnight. The reaction mixture was then poured into 0.1 M HCI and the organic phase washed with H20, saturated NaHCO 3 , and brine, and then dried over MgSO 4 , filtered and concentrated. The crude methyl ester was purified by column chromatography. Deprotection with 10% Pd(C) in MeOH at 40 psi 20 H2 gave the free amine which was coupled to Tos-Pro-OH using the procedure described in Method 3. The ester was hydrolyzed using the procedure described in Method 6. The product was isolated as a white solid. NMR data was as follows: 1H NMR(CDC13, 300 MHz): 6 = 7.74 (d, 2H, J = 8.0 Hz); 7.40 25 7.34 (min, 3H); 4.91 (bs, 1H); 4.60 (min, 1H); 4.06 (min, 1H); 3.63 (min, 1 H); 3.15 (min, 3H), 2.44 (s, 3H); 2.22-1.90 (min, 2H); 1.80 (min, 2H); 1.70 - 1.30 (bm, 15 H). 1 3 C NMR(CDC1 3 , 75 MHz): 5 = 175.3, 172.3, 156.9, 145.1, 133.1, 130.6, 128.5, 79.7, 63.1, 52.6, 50.6, 40.7, 32.1, 31.2, 29.7, 28.9, 24.8, 30 22.9, 22.2. Mass Spectroscopy: (PI-FAB) 498, (M+H)
+
.
WO 99/06432 PCT/US98/15325 -- 111 - Example 13 Synthesis of N-(Toluene-4-sulfonyl)-L-prolyl-(2,3-dehydro)phenylalanine 5 N-(Toluene-4-sulfonyl)-L-prolyl-p-hydroxy-D,L-phenylalanine methyl ester (501 mg, 1.12 mmol) was dissolved in CH 2 Cl 2 (10 mL) and cooled in an ice bath. Triethylamine (195 yL) was added, followed by methanesulfonyl chloride (135 mg, 1.13 mmol). The mixture was stirred 10 was stirred for 45 minutes before additional triethylamine (313 pL) was added. The mixture was warmed to room temperature and stirred for 3 hr. The mixture was diluted with CH 2 C2 and washed with saturated aqueous NaHCO 3 (2 X 30 mL), dried (NaSO 4 ), filtered and evaporated in vacuo to give a residue which was purified by silica gel flash chromatography (95:5 15 CH 2 Cl 2 /MeOH) to give the dehydro ester (453 mg, 94%). The title compound was prepared via hydrolysis of the methyl ester using NaOH in methanol and purified by silica gel flash chromatography (9/1 CH 2 Cl 2 / MeOH) to give the compound as a white solid (120 mg, 62%). NMR data was as follows: 20 'H NMR (CD 3 OD): 8 = 7.92-7.68 (min, 4H), 7.52-7.27 (min, 6H), 4.28 (min, 1H), 3.68 (m, 1H), 3.31 (min, 3H), 2.44 (s, 3H), 2.09-1.46 (4H). 13C NMR (CD 3 OD): 8 = 174.6, 146.4, 135.6, 135.3, 132.0, 131.7, 131.1, 130.3, 130.0, 129.9, 129.6, 64.4, 51.5, 32.4, 26.1, 22.2. Mass Spectroscopy: FAB m/e 439 (M+Li). 25 Example 14 Synthesis of N-(Toluene-4-sulfonyl)-L-(5,5-dimethyl)thiaprolyl Ne-(tert-butoxycarbonyl)-L-lysine Methyl Ester 30 The title compound was prepared using the procedure described in Method 13 employing suitable starting materials to provide for a solid, mp = 51-53 0
C.
WO 99/06432 PCT/US98/15325 -- 112- NMR data was as follows: 1 H NMR (CDCl 3 , 300 MHz): 8 = 1.14 (s, 3H), 1.36 (s, 3H), 1.42 (s, 9H), 1.43 (m, 4H), 1.80 (m, 2H), 2.45 (s, 3H), 3.08 (m, 2H), 3.78 (s, 3H), 3.87 (s, 1H), 4.42 (d, 1H, J= 9.7 Hz), 4.57 (m, 1H), 4.65 (d, 1H, J = 9.7 5 Hz), 7.09 (d, 1H, J = 7.7 Hz), 7.35 (d, 2H, 8.1 Hz), 7.77 (d, 2H, J = 8.2 Hz). 1 3 C NMR (CDCl 3 , 75 MHz): 8 = 22.2, 23.0, 24.9, 29.0, 30.0, 32.5, 40.8, 51.7, 52.8, 53.1, 55.2, 74.1, 128.7, 130.6, 133.3, 145.4, 156.5, 169.1, 172.5. 10 Mass Spectroscopy: (FAB+) 558 (M+H). Example 15 Synthesis of N-(Toluene-4-sulfonyl)-L-(5,5-dimethyl)thiaprolyl 15 NE-(tert-butoxycarbonyl)-L-lysine The title compound was prepared from the product of Example 14 (348) using the procedure described in Method 7 as a solid, mp = > 150'C (dec.). NMR data was as follows: 20 'H NMR (CDC13, 300 MHz): 8 = 1.15 (s, 3H), 1.37 (s, 3H), 1.42 (s, 9H), 1.49 (m, 4H), 2.05 (m, 2H), 2.45 (s, 3H), 3.10 (br s, 2H), 3.92 (s, 1H), 4.47-4.73 (m, 3H), 7.37 (d, 2H, J = 8.2 Hz), 7.80 (d, 2H, J = 8.2 Hz). 13C NMR (CDCl 3 , 75 MHz): 8 = 22.3, 25.1, 28.9, 29.5, 30.0, 30.9, 25 40.9, 51.1, 54.6, 54.7, 55.3, 74.2, 79.2, 128.5, 128.6, 130.6, 133.3, 145.4, 156.77, 156.81. Mass Spectroscopy: (FAB +) 544 (M +H). 30 WO 99/06432 PCT/US98/15325 -- 113 - Example 16 Synthesis of N-(Toluene-4-sulfonyl)-L-(5,5-dimethyl) thiaprolyl-L-asparagine 5 The product was prepared from suitable starting materials via Method 3 and Method 11 and was isolated as a solid. NMR data was as follows: 'H NMR (DMSO-d6, 300 MHz): 6 = 8.39 (d, 1H, J = 7.6 Hz); 7.73 10 (d, 2H, J = 8.2 Hz); 7.42 (d, 2H, J = 8.0 Hz); 6.96 (br s, 1H); 4.60 - 4.51 (br m, 1H); 4.6 (m, 1H); 4.13 (s, 1H); 2.56 (m, 1H); 2.43 (m, lh); 2.40 (s, 3H); 1.28 (s, 3H); 1.18 (s, 3H). 13C NMR (DMSO-d 6 , 75 MHz): 6 = 172.8, 171.6, 167.8, 144.2, 134.3, 130.2, 127.9, 72.03, 54.9, 50.5, 48.9, 30.1, 24.9, 21.4. 15 Mass Spectroscopy: (PI-FAB) 430, (M +H) . Other compounds prepared by the methods described above include those set forth in Table II below: 20 WO 99/06432 PCTIUS98/1 5325 C; 00 9 9 z R_ _ 72_____"
U>
WO 99/06432 PCT/US98/1 5325 -115 - 0 -z 0 u 0 cz c z WO 99/06432 PCT/US98/15325 N 0 a aa -- 11 - C's cz cz m m o 0 0~ IR "x "x " a c u -" , o o O 0o U) U U r,. 0 U U 0 9- ") - Z -Zb C .) . . . : o . o C z WO 99/06432 PCT/US98/1 5325 9x 0 9- 99 71l Z -Z Ln c oM C)N Uz ct z -Ez WO 99/06432 PCT/US98/15325 -- 118 - 00 -- u 9 9 * .@ (8 x = o - : ' z o, z a o _ z z 0 . - om a. z 0 "^ OU U zU U &It SU U U 0 .0 U U U U U U U U U U U U U U U U WO 99/06432 PCT/US98/15325 -- 119- . 0 a - a r -- I I I 71 u I zuOu u, z z 0 a 0 a 0 a 0 -0 oO cz C.ISC. Z Z U C. u u u uf f u u u u uf a x = . " IU I - - 4h - - - z II I I III 4u 4 44, 4 e e e en e e U C. U U U U C. C.
WO 99/06432 PCT/US98/1 5325 -120- 00 0 Q C0 0 EE Z Z WO 99/06432 PCT/US98/15325 -- 121 - 9 0 a 0 a 0 I I r ar a 0o cO c o Zo - z - Zz 0 a Z11,- , ".-.
u. z - z Q ~~~ x E E E uS3 uc uc zc 'r . _ z o '00(UN 0 IN 0 I 4 I I In In InI I U U U U U UU 73 a WO 99/06432 PCT/US98/1 5325 -- 122- 0- 0 "o "a"a7 o 9Z, WO 99/06432 PCT/US98/1 5325 -123- 00 w 0 E. n e in U WO 99/06432 PCT/US98/15325 -- 124 - Example 79 In vitro Assay For Determining Binding of Candidate Compounds to VLA-4 5 An in vitro assay was used to assess binding of candidate compounds to c4p 1 integrin. Compounds which bind in this assay can be used to assess VCAM-1 levels in biological samples by conventional assays (e.g., competitive assays). This assay is sensitive to ICso values as low as about lnM. 10 The activity of a 4 1 integrin was measured by the interaction of soluble VCAM-1 with Jurkat cells (e.g., American Type Culture Collection Nos. TIB 152, TIB 153, and CRL 8163), a human T-cell line which expresses high levels of a 4 1 integrin. VCAM-1 interacts with the cell surface in an 15 4
P
1 integrin-dependent fashion (Yednock, et al. J. Biol. Chem., 1995, 270:28740). Recombinant soluble VCAM-1 was expressed as a chimeric fusion protein containing the seven extracellular domains of VCAM-1 on the N 20 terminus and the human IgG, heavy chain constant region on the C-terminus. The VCAM-1 fusion protein was made and purified by the manner described by Yednock, supra. Jurkat cells were grown in RPMI 1640 supplemented with 10% fetal 25 bovine serum, penicillin, streptomycin and glutamine as described by Yednock, supra. Jurkat cells were incubated with 1.5 mM MnCl 2 and 5 pg/mL 15/7 antibody for 30 minutes on ice. Mn +2 activates the receptor to enhance 30 ligand binding, and 15/7 is a monoclonal antibody that recognizes an activated/ligand occupied conformation of a43 1 integrin and locks the WO 99/06432 PCT/US98/15325 -- 125 molecule into this conformation thereby stabilizing the VCAM-1/a4pl integrin interaction. Yednock, et al., supra. Antibodies similar to the 15/7 antibody have been prepared by other investigators (Luque, et al, 1996, J. Biol. Chem. 271:11067) and may be used in this assay. 5 Cells were then incubated for 30 minutes at room temperature with candidate compounds, in various concentrations ranging from 66 AM to 0.01 AM using a standard 5-point serial dilution. 15 AL soluble recombinant VCAM-1 fusion protein was then added to Jurkat cells and incubated for 30 10 minutes on ice. (Yednock et al., supra.). Cells were then washed two times and resuspended in PE-conjugated goat F(ab') 2 anti-mouse IgG Fc (Immunotech, Westbrook, ME) at 1:200 and incubated on ice, in the dark, for 30 minutes. Cells were washed twice and 15 analyzed with a standard fluorescence activated cell sorter ("FACS") analysis as described in Yednock, et al., supra. Compounds having an IC 50 so of less than about 15AM possess binding affinity to ( 4 1 . 20 When tested in this assay, each of the compounds in Examples 1-78 has an IC 5 0 so of 15 4M or less. 25 Example 80 In vitro Saturation Assay For Determining Binding of Candidate Compounds to a 4 1 30 The following describes an in vitro assay to determine the plasma levels needed for a compound to be active in the Experimental Autoimmune WO 99/06432 PCT/US98/15325 -- 126- Encephalomyelitis ("EAE") model, described in the next example, or in other in vivo models. Log-growth Jurkat cells are washed and resuspended in normal animal 5 plasma containing 20 yg/ml of the 15/7 antibody (described in the above example). The Jurkat cells are diluted two-fold into either normal plasma samples containing known candidate compound amounts in various concentrations 10 ranging from 66 yM to 0.01 tM, using a standard 12 point serial dilution for a standard curve, or into plasma samples obtained from the peripheral blood of candidate compound-treated animals. Cells are then incubated for 30 minutes at room temperature, washed 15 twice with phosphate-buffered saline ("PBS") containing 2% fetal bovine serum and 1mM each of calcium chloride and magnesium chloride (assay medium) to remove unbound 15/7 antibody. The cells are then exposed to phycoerythrin-conjugated goat F(ab') 2 anti 20 mouse IgG Fc (Immunotech, Westbrook, ME), which has been adsorbed for any non-specific cross-reactivity by co-incubation with 5 % serum from the animal species being studied, at 1:200 and incubated in the dark at 40C for 30 minutes. 25 Cells are washed twice with assay medium and resuspended in the same. They are then analyzed with a standard fluorescence activated cell sorter ("FACS") analysis as described in Yednock et al. J. Biol. Chem., 1995, 270:28740.
WO 99/06432 PCT/US98/15325 -- 127 - The data is then graphed as fluorescence versus dose, e.g., in a normal dose-response fashion. The dose levels that result in the upper plateau of the curve represent the levels needed to obtain efficacy in an in vivo model. 5 This assay may also be used to determine the plasma levels needed to saturate the binding sites of other integrins, such as the c3 1 , integrin, which is the integrin most closely related a 4 p 1 (Palmer et al, 1993, J. Cell Bio., 123:1289). Such binding is predictive of in vivo utility for inflammatory conditions mediated by a 9 p 1 integrin, including by way of example, airway 10 hyper-responsiveness and occlusion that occurs with chronic asthma, smooth muscle cell proliferation in atherosclerosis, vascular occlusion following angioplasty, fibrosis and glomerular scarring as a result of renal disease, aortic stenosis, hypertrophy of synovial membranes in rheumatoid arthritis, and inflammation and scarring that occur with the progression of ulcerative 15 colitis and Crohn's disease. Accordingly, the above-described assay may be performed with a human colon carcinoma cell line, SW 480 (ATTC #CCL228) transfected with cDNA encoding c 9 integrin (Yokosaki et al., 1994, J. Biol. Chem., 20 269:26691), in place of the Jurkat cells, to measure the binding of the a3p integrin. As a control, SW 480 cells which express other a and k subunits may be used. Accordingly, another aspect of this invention is directed to a method for 25 treating a disease in a mammalian patient, which disease is mediated by Cf3 1 , and which method comprises administering to said patient a therapeutically effective amount of a compound of this invention. Such compounds are preferably administered in a pharmaceutical composition described herein above. Effective daily dosing will depend upon the age, weight, condition of 30 the patient which factors can be readily ascertained by the attending WO 99/06432 PCT/US98/15325 -- 128clinician. However, in a preferred embodiment, the compounds are administered from about 20 to 500 ig/kg per day. Example 81 5 In vivo Evaluation The standard multiple sclerosis model, Experimental Autoimmune (or Allergic) Encephalomyelitis ("EAE"), is used to determine the effect of candidate compounds to reduce motor impairment in rats or guinea pigs. 10 Reduction in motor impairment is based on blocking adhesion between leukocytes and the endothelium and correlates with. anti-inflammatory activity in the candidate compound. This model has been previously described by Keszthelyi et al., Neurology, 1996, 47:1053-1059, and measures the delay of onset of disease. 15 Brains and spinal cords of adult Hartley guinea pigs are homogenized in an equal volume of phosphate-buffered saline. An equal volume of Freund's complete adjuvant (100 mg mycobacterium tuberculosis plus 10 ml Freund's incomplete adjuvant) is added to the homogenate. The mixture is emulsified 20 by circulating it repeatedly through a 20 ml syringe with a peristaltic pump for about 20 minutes. Female Lewis rats (2-3 months old, 170-220 g) or Hartley guinea pigs (20 day old, 180-200 g) are anesthetized with isoflurane and three injections 25 of the emulsion, 0.1 ml each, are made in each flank. Motor impairment onset is seen in approximately 9 days. Candidate compound treatment begins on Day 8, just before onset of symptoms. Compounds are administered subcutaneously ("SC"), orally 30 ("PO") or intraperitoneally ("IP"). Doses are given in a range of 10mg/kg WO 99/06432 PCT/US98/15325 -- 129 to 200 mg/kg, bid, for five days, with typical dosing of 10 to 100 mg/kg SC, 10 to 50 mg/kg PO, and 10 to 100 mg/kg IP. Antibody GG5/3 against 4 f3 1 integrin (Keszthelyi et al., Neurology, 5 1996, 47:1053-1059), which delays the onset of symptoms, is used as a positive control and is injected subcutaneously at 3 mg/kg on Day 8 and 11. Body weight and motor impairment are measured daily. Motor impairment is rated with the following clinical score: 10 0 no change 1 tail weakness or paralysis 2 hindlimb weakness 3 hindlimb paralysis 15 4 moribund or dead A candidate compound is considered active if it delays the onset of symptoms, e.g., produces clinical scores no greater than 2 or slows body weight loss as compared to the control. 20 Example 82 Asthma Model Inflammatory conditions mediated by a 4 p 1 integrin include, for example, airway hyper-responsiveness and occlusion that occurs with chronic asthma. 25 The following describes an asthma model which can be used to study the in vivo effects of the compounds of this invention for use in treating asthma. Following the procedures described by Abraham et al, J. Clin. Invest, 93:776-787 (1994) and Abraham et al, Am J. Respir Crit Care Med, 30 156:696-703 (1997), both of which are incorporated by reference in their entirety, compounds of this invention are formulated into an aerosol and administered to sheep which are hypersensitive to Ascaris suum antigen. Compounds which decrease the early antigen-induced bronchial response WO 99/06432 PCT/US98/15325 -- 130and/or block the late-phase airway response, e.g., have a protective effect against antigen-induced late responses and airway hyper-responsiveness ("AHR"), are considered to be active in this model. 5 Allergic sheep which are shown to develop both early and late bronchial responses to inhaled Ascaris suum antigen are used to study the airway effects of the candidate compounds. Following topical anesthesia of the nasal passages with 2% lidocaine, a balloon catheter is advanced through one nostril into the lower esophagus. The animals are then intubated with a 10 cuffed endotracheal tube through the other nostril with a flexible fiberoptic bronchoscope as a guide. Pleural pressure is estimated according to Abraham (1994). Aerosols (see formulation below) are generated using a disposable medical nebulizer 15 that provides an aerosol with a mass median aerodynamic diameter of 3.2 ym as determined with an Andersen cascade impactor. The nebulizer is connected to a dosimeter system consisting of a solenoid valve and a source of compressed air (20 psi). The output of the nebulizer is directed into a plastic T-piece, one end of which is connected to the inspiratory port of a 20 piston respirator. The solenoid valve is activated for 1 second at the beginning of the inspiratory cycle of the respirator. Aerosols are delivered at VT of 500 ml and a rate of 20 breaths/minute. A 0.5% sodium bicarbonate solution only is used as a control. 25 To assess bronchial responsiveness, cumulative concentration-response curves to carbachol can be generated according to Abraham (1994). Bronchial biopsies can be taken prior to and following the initiation of treatment and 24 hours after antigen challenge. Bronchial biopsies can be preformed according to Abraham (1994). 30 WO 99/06432 PCT/US98/15325 -- 131 - An in vitro adhesion study of alveolar macrophages can be performed according to Abraham (1994), and a percentage of adherent cells is calculated. 5 Aerosol Formulation A solution of the candidate compound in 0.5% sodium bicarbonate/saline (w/v) at a concentration of 30.0 mg/mL is prepared using the following procedure: 10 A. Preparation of 0.5% Sodium Bicarbonate/Saline Stock Solution: 100.0 mL Ingredient Gram / 100.0 mL Final Concentration Sodium Bicarbonate 0.5 g 0.5% 15 Saline q.s. ad 100.0 mL q.s. ad 100% Procedure: 1. Add 0.5g sodium bicarbonate into a 100 mL volumetric flask. 2. Add approximately 90.0 mL saline and sonicate until dissolved. 20 3. Q.S. to 100.0 mL with saline and mix thoroughly. B. Preparation of 30.0 mg/mL Candidate Compound: 10.0 mL Ingredient Gram / 10.0 mL Final Concentration Candidate Compound 0.300 g 30.0 mg/mL 25 0.5% Sodium Bicarbonate / q.s. ad 10.0 mL q.s ad 100% Saline Stock Solution Procedure: 1. Add 0.300 g of the candidate compound into a 10.0 mL 30 volumetric flask.
WO 99/06432 PCT/US98/15325 -- 132- 2. Add approximately 9.7 mL of 0.5% sodium bicarbonate / saline stock solution. 3. Sonicate until the candidate compound is completely dissolved. 4. Q.S. to 10.0 mL with 0.5% sodium bicarbonate / saline stock 5 solution and mix thoroughly. Using a conventional oral formulation, compounds of this invention would be active in this model. 10

Claims (16)

1. A compound of formula I: 5 R 3 O R'-S O 2 -N(R 2 )-C-Q-CH-C-OH I I H R 5 10 where R' is selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted 15 heterocylic, heteroaryl and substituted heteroaryl; R 2 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, and R 1 and R 2 together with the nitrogen 20 atom bound to R 2 and the SO 2 group bound to R 1 can form a heterocyclic or a substituted heterocyclic group; R 3 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic and, when R 2 25 does not form a heterocyclic group with R 1 , R 2 and R 3 together with the nitrogen atom bound to R 2 and the carbon atom bound to R 3 can form a heterocyclic or a substituted heterocyclic group; R 5 is -ALK-X or =CH-Y where ALK is an alkyl group of from 1 to 10 carbon atoms attached via a methylene group (-CH 2 -) to the carbon atom to 30 which it is attached; X is selected from the group consisting of substituted alkylcarbonylamino, substituted alkenylcarbonylamino, substituted alkynylcarbonylamino, heterocyclylcarbonylamino, substituted WO 99/06432 PCT/US98/15325 -- 134 - heterocyclylcarbonylamino, acyl, acyloxy, aminocarbonyloxy, acylamino, oxycarbonylamino, alkoxycarbonyl, substituted alkoxycarbonyl, aryloxycarbonyl, substituted aryloxycarbonyl, cycloalkoxycarbonyl, substituted cycloalkoxycarbonyl, heteroaryloxycarbonyl, substituted 5 heteroaryloxycarbonyl, heterocyclyloxycarbonyl, substituted heterocyclyloxycarbonyl, cycloalkyl, substituted cycloalkyl, saturated heterocyclic, substituted saturated heterocyclic, substituted alkoxy, substituted alkenoxy, substituted alkynoxy, heterocyclyloxy, substituted heterocycloxy, substituted thioalkyl, substituted thioalkenyl, substituted 10 thioalkynyl, aminocarbonylamino, aminothiocarbonylamino, guanidino, amidino, alkylamidino, thioamidino, halogen, cyano, nitro, -OS(O) 2 -alkyl, -OS(O) 2 -substituted alkyl, -OS(O) 2 -cycloalkyl, -OS(O) 2 -substituted cycloalkyl, -OS(O) 2 -aryl, -OS(O) 2 -substituted aryl, -OS(O) 2 -heteroaryl, -OS(O) 2 -substituted heteroaryl, -OS(O) 2 -heterocyclic, -OS(O) 2 -substituted 15 heterocyclic, -OS0 2 -NRR where R is hydrogen or alkyl, -NRS(O)-alkyl, -NRS(O) 2 -substituted alkyl, -NRS(O) 2 -cycloalkyl, -NRS(O) 2 -substituted cycloalkyl, -NRS(O) 2 -aryl, -NRS(O) 2 -substituted aryl, -NRS(O) 2 -heteroaryl, -NRS(O) 2 -substituted heteroaryl, -NRS(O) 2 -heterocyclic, -NRS(O) 2 substituted heterocyclic, -NRS(O) 2 -NR-alkyl, -NRS(O) 2 -NR-substituted 20 alkyl, -NRS(O) 2 -NR-cycloalkyl, -NRS(O) 2 -NR-substituted cycloalkyl, -NRS(O) 2 -NR-aryl, -NRS(O) 2 -NR-substituted aryl, -NRS(O) 2 -NR-heteroaryl, -NRS(O) 2 -NR-substituted heteroaryl, -NRS(O) 2 -NR-heterocyclic, -NRS(O) 2 NR-substituted heterocyclic where R is hydrogen or alkyl, -S(O) 2 -alkyl, S(O) 2 -substituted alkyl, -S(O) 2 -aryl, -S(O) 2 -substituted aryl, -S(O) 2 25 substituted heteroaryl, -S(O) 2 -substituted heteroaryl, -S(O) 2 -heterocyclic, S(O) 2 -substituted heterocyclic, mono- and di-(substituted alkyl)amino, N,N (alkyl, substituted alkyl)amino, N,N-(aryl, substituted alkyl)amino, N,N (substituted aryl, substituted alkyl)amino, N,N-(heteroaryl, substituted alkyl)amino, N,N-(substituted heteroaryl, substituted alkyl)amino, N,N 30 (heterocyclic, substituted alkyl)amino, N,N-N,N-(substituted heterocyclic, WO 99/06432 PCT/US98/15325 -- 135 - substituted alkyl)amino, mono- and di-(heterocyclic)amino, mono- and di (substituted heterocyclic)amino, N,N-(alkyl, heterocyclic)amino, N,N-(alkyl, substituted heterocyclic)amino, N,N-(aryl, heterocyclic)amino, N,N (substituted aryl, heterocyclic)amino, N,N-(aryl, substituted 5 heterocyclic)amino, N,N-(substituted aryl, substituted heterocyclic)amino, N,N-(heteroaryl, heterocyclic)amino, N,N-(heteroaryl, substituted heterocyclic)amino, N,N-(substituted heteroaryl, heterocyclic)amino, and N,N-(substituted heteroaryl, substituted heterocyclic)amino; and Y is selected from the group consisting of alkyl, substituted alkyl, aryl, 10 substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic; Q is -C(X)NR 7 - wherein R 7 is selected from the group consisting of hydrogen and alkyl; and X is selected from the group consisting of oxygen and sulfur; 15 and pharmaceutically acceptable salts thereof with the provisos that A. when R' is p-methylphenyl, R 2 and R 3 are joined together with the nitrogen atom bound to R 2 and the carbon atom bound to R 3 to form a pyrrolidinyl ring and Q is -C(O)NH-, then R 5 is not -CH 2 C(O)-O-t-butyl or 20 CH 2 CH 2 C(O)-O-t-butyl; and B. when R' is p-methylphenyl, R 2 is methyl, R 3 is hydrogen and Q is -C(O)NH-, then R s is not -CH 2 (N-benzylpiperin-4-yl).
2. A compound of formula IA: 25 R 3 O I 0 R'-S O 2 -N(R 2 )-C-Q-CH-C-R 6 IA 30 H R 5 where WO 99/06432 PCT/US98/15325 -- 136 - R 1 is selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocylic, heteroaryl and substituted heteroaryl; R 2 is selected from the group consisting of hydrogen, alkyl, substituted 5 alkyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, and RI and R 2 together with the nitrogen atom bound to R 2 and the SO 2 group bound to R 1 can form a heterocyclic or a substituted heterocyclic group; 10 R 3 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic and, when R 2 does not form a heterocyclic group with R 1 , R 2 and R 3 together with the nitrogen atom bound to R 2 and the carbon atom bound to R 3 can form a 15 heterocyclic or a substituted heterocyclic group; R 5 is -ALK-X or =CH-Y where ALK is an alkyl group of from 1 to 10 carbon atoms attached via a methylene group (-CH 2 -) to the carbon atom to which it is attached; X is selected from the group consisting of substituted alkylcarbonylamino, substituted alkenylcarbonylamino, substituted 20 alkynylcarbonylamino, heterocyclylcarbonylamino, substituted heterocyclylcarbonylamino, acyl, acyloxy, aminocarbonyloxy, acylamino, oxycarbonylamino, alkoxycarbonyl, substituted alkoxycarbonyl, aryloxycarbonyl, substituted aryloxycarbonyl, cycloalkoxycarbonyl, substituted cycloalkoxycarbonyl, heteroaryloxycarbonyl, substituted 25 heteroaryloxycarbonyl, heterocyclyloxycarbonyl, substituted heterocyclyloxycarbonyl, cycloalkyl, substituted cycloalkyl, saturated heterocyclic, substituted saturated heterocyclic, substituted alkoxy, substituted alkenoxy, substituted alkynoxy, heterocyclyloxy, substituted heterocycloxy, substituted thioalkyl, substituted thioalkenyl, substituted WO 99/06432 PCT/US98/15325 -- 137- thioalkynyl, aminocarbonylamino, aminothiocarbonylamino, guanidino, amidino, alkylamidino, thioamidino, halogen, cyano, nitro, -OS(O)-alkyl, -OS(O) 2 -substituted alkyl, -OS(O) 2 -cycloalkyl, -OS(O) 2 -substituted cycloalkyl, -OS(O) 2 -aryl, -OS(O) 2 -substituted aryl, -OS(O) 2 -heteroaryl, 5 -OS(O) 2 -substituted heteroaryl, -OS(O) 2 -heterocyclic, -OS(O) 2 -substituted heterocyclic, -OSO 2 -NRR where R is hydrogen or alkyl, -NRS(O) 2 -alkyl, -NRS(O) 2 -substituted alkyl, -NRS(O) 2 -cycloalkyl, -NRS(O) 2 -substituted cycloalkyl, -NRS(O) 2 -aryl, -NRS(O) 2 -substituted aryl, -NRS(O) 2 -heteroaryl, -NRS(O) 2 -substituted heteroaryl, -NRS(O) 2 -heterocyclic, -NRS(O) 2 10 substituted heterocyclic, -NRS(O) 2 -NR-alkyl, -NRS(O) 2 -NR-substituted alkyl, -NRS(O) 2 -NR-cycloalkyl, -NRS(O) 2 -NR-substituted cycloalkyl, -NRS(O) 2 -NR-aryl, -NRS(O) 2 -NR-substituted aryl, -NRS(O)z-NR-heteroaryl, -NRS(O) 2 -NR-substituted heteroaryl, -NRS(O) 2 -NR-heterocyclic, -NRS(O) 2 NR-substituted heterocyclic where R is hydrogen or alkyl, -S(O) 2 -alkyl, 15 S(O) 2 -substituted alkyl, -S(O) 2 -aryl, -S(O) 2 -substituted aryl, -S(O) 2 substituted heteroaryl, -S(O) 2 -substituted heteroaryl, -S(O) 2 -heterocyclic, S(O) 2 -substituted heterocyclic, mono- and di-(substituted alkyl)amino, N,N (alkyl, substituted alkyl)amino, N,N-(aryl, substituted alkyl)amino, N,N (substituted aryl, substituted alkyl)amino, N,N-(heteroaryl, substituted 20 alkyl)amino, N,N-(substituted heteroaryl, substituted alkyl)amino, N,N (heterocyclic, substituted alkyl)amino, N,N-N,N-(substituted heterocyclic, substituted alkyl)amino, mono- and di-(heterocyclic)amino, mono- and di (substituted heterocyclic)amino, N,N-(alkyl, heterocyclic)amino, N,N-(alkyl, substituted heterocyclic)amino, N,N-(aryl, heterocyclic)amino, N,N 25 (substituted aryl, heterocyclic)amino, N,N-(aryl, substituted heterocyclic)amino, N,N-(substituted aryl, substituted heterocyclic)amino, N,N-(heteroaryl, heterocyclic)amino, N,N-(heteroaryl, substituted heterocyclic)amino, N,N-(substituted heteroaryl, heterocyclic)amino, and N,N-(substituted heteroaryl, substituted heterocyclic)amino; and Y is 30 selected from the group consisting of alkyl, substituted alkyl, aryl, WO 99/06432 PCT/US98/15325 -- 138- substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic; R 6 is selected from the group consisting of 2,4-dioxo-tetrahydrofuran-3 yl (3,4-enol), amino, alkoxy, substituted alkoxy, cycloalkoxy, substituted 5 cycloalkoxy, -O-(N-succinimidyl), -NH-adamantyl, -O-cholest-5-en-3-P-yl, NHOY where Y is hydrogen, alkyl, substituted alkyl, aryl, and substituted aryl, -NH(CH 2 )pCOOY where p is an integer of from 1 to 8 and Y is as defined above, -OCH 2 NR 9 R 1 o where R 9 is selected from the group consisting of -C(O)-aryl and -C(O)-substituted aryl and R' is selected from the group 10 consisting of hydrogen and -CH 2 COOR" where R" is alkyl, and -NHSO 2 Z where Z is alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic; Q is -C(X)NR 7 - wherein R 7 is selected from the group consisting of 15 hydrogen and alkyl; and X is selected from the group consisting of oxygen and sulfur; and pharmaceutically acceptable salts thereof with the proviso that A. when R' is p-methylphenyl, R 2 and R 3 are joined together with 20 the nitrogen atom bound to R 2 and the carbon atom bound to R 3 to form a pyrrolidinyl ring, R 6 is methoxy, and Q is -C(O)NH-, then R is not -CH 2 CH 2 COO-t-butyl or -CH 2 COO-t-butyl.
3. A compound according to Claims 1 or 2 wherein R 1 is selected 25 from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, heterocyclic, substituted heterocylic, heteroaryl and substituted heteroaryl.
4. A compound according to Claim 3 wherein R is selected from the group consisting of 4-methylphenyl, methyl, benzyl, n-butyl, 4 30 chlorophenyl, 1-naphthyl, 2-naphthyl, 4-methoxyphenyl, phenyl, 2,4,6- WO 99/06432 PCT/US98/15325 -- 139 - trimethylphenyl, 2-(methoxycarbonyl)phenyl, 2-carboxyphenyl, 3,5 dichlorophenyl, 4-trifluoromethylphenyl, 3,4-dichlorophenyl, 3,4 dimethoxyphenyl, 4-(CH 3 C(O)NH-)phenyl, 4-trifluoromethoxyphenyl, 4 cyanophenyl, isopropyl, 3,5-di-(trifluoromethyl)phenyl, 4-t-butylphenyl, 4-t 5 butoxyphenyl, 4-nitrophenyl, 2-thienyl, 1-N-methyl-3-methyl-5 chloropyrazol-4-yl, phenethyl, 1-N-methylimidazol-4-yl, 4-bromophenyl, 4 amidinophenyl, 4-methylamidinophenyl, 4-[CH 3 SC(=NH)]phenyl, 5-chloro 2-thienyl, 2,5-dichloro-4-thienyl, 1-N-methyl-4-pyrazolyl, 2-thiazolyl, 5 methyl-1,3,4-thiadiazol-2-yl, 4-[H 2 NC(S)]phenyl, 4-aminophenyl, 4 10 fluorophenyl, 2-fluorophenyl, 3-fluorophenyl, 3,5-difluorophenyl, pyridin-3 yl, pyrimidin-2-yl, 4-(3'-dimethylamino-n-propoxy)-phenyl and 1 methylpyrazol-4-yl.
5. A compound according to Claims 1 or 2 wherein R 2 is selected 15 from the group consisting of hydrogen, methyl, phenyl, benzyl, -(CH 2 ) 2 -2 thienyl and -(CH 2 ) 2 -4
6. A compound according to Claims 1 or 2 wherein R 1 and R 2 together with the nitrogen atom bound to R 2 and the SO 2 group bound to RI 20 are joined to form a heterocyclic group or substituted heterocyclic group.
7. A compound according to Claims 1 or 2 wherein R 2 and R 3 together with the nitrogen atom bound to R 2 substituent and the carbon bound to the R 3 substituent form a heterocyclic group or a substituted 25 heterocyclic group.
8. A compound according to Claims 1 or 2 wherein R 3 is selected from the group consisting of methyl, phenyl, benzyl, diphenylmethyl, -CH 2 CHz-COOH, -CH 2 -COOH, 2-amidoethyl, iso-butyl, t-butyl, -CH 2 0 30 benzyl and hydroxymethyl. WO 99/06432 PCT/US98/15325 -- 140 -
9. A compound according to Claims 1 or 2 wherein R is selected from the group consisting of t-butyl-OC(O)CH 2 -, -CH 2 C(O)NH 2 , -CH 2 CH 2 C(O)NH 2 , t-butyl-OC(O)CH 2 CH 2 -, BocNH-(CH 2 ) 4 -, (4-CH 2 -OC(O)NH-(CH 2 ) 4 -, benzyloxy-CH 2 -, cyclohexyl-CH 2 -, 5 N-benzylpiperid-4-yl-CH 2 -, N-Boc-piperidin-4-yl-CH 2 -, N-(phenylcarbonyl)piperidin-4-yl-CH 2 -, allyloxy-C(O)NH-(CH 2 ) 4 -, allyloxy-C(O)NH(CH2) 3 -, allyloxy-C(O)NH(CH 2 ) 2 -, #-CH =, 4-methylphenyl-SO 2 -N(CH3)CH 2 C(O)NH(CH 2 ) 4 -, -CH 2 C(O)NH(CH 2 )4, -(CH 2 ) 4 NHC(O)CH 2 -3-indolyl, -(CH 2 ) 4 NHC(O)CHzCH 2 -3-indolyl, 10 -(CH 2 ) 4 NHC(O)CH 2 0-4-fluorophenyl, -CH 2 C(O)NHCH(CH 3 ), -CH 2 C(O)NHCH 2 -(4-dimethylamino)-4, -CH 2 C(O)NHCH2-4-nitrophenyl, -CH 2 CH 2 C(O)N(CH 3 )CH 2 -, -CH 2 C(O)N(CH 3 )CH 2 -, -CH 2 CH 2 C(O)NHCH 2 CH 2 -(N-methyl)-2-pyrrolyl, -CH 2 CHzC(O)NHCH 2 CH 2 CH 2 CH 3 , -CH 2 CH 2 C(O)NHCH 2 CHz-3-indolyl, 15 -CH 2 C(O)N(CH 3 )CH 2 phenyl, -CHzC(O)NH(CH 2 )-(N-methyl)-2-pyrrolyl, -CHzC(O)NHCH 2 CH 2 CH 2 CH 3 , -CH 2 C(O)NHCH 2 CH 2 -3-indolyl, -(CH 2 ) 2 C(O)NHCH(CH 3 )4, -(CHz) 2 C(O)NHCH 2 -4-dimethylaminophenyl, -(CH 2 ) 2 C(O)NHCH2-4-nitrophenyl, -CH 2 C(O)NH-4-[-NHC(O)CH 3 -phenyl], -CH 2 C(O)NH-4-pyridyl, -CH 2 C(O)NH-4-[dimethylaminophenyl], 20 -CH 2 C(O)NH-3-methoxyphenyl, -CH 2 CH 2 C(O)NH-4-chlorophenyl, -CH 2 CH 2 C(O)NH-2-pyridyl, -CH 2 CH 2 C(O)NH-4-methoxyphenyl, -CH 2 CH 2 C(O)NH-3-pyridyl, -(CH 2 ) 3 NHC(NH)NH-SO2-4-methylphenyl, -(CH2) 4 NHC(O)NHCH 2 CH 3 , -(CH 2 ) 4 NHC(O)NH-phenyl, -(CH2) 4 NHC(O)NH-4-methoxyphenyl, -CH 2 C(O)NHCH 2 CH 2 N(CH 3 ) 2 , 25 [BocNHCH 2 C(O)NH-]butyl, 2-[4-hydroxy-4-(3-methoxythien-2-yl)piperidin 1-yl]ethyl, 4-[(1-Cbz-piperidin-4-yl)C(O)NH-]butyl, 4-[(N-toluenesulfonylpyrrolidin-2'-yl)C(O)NH-]butyl, 4-[-NHC(O)-4'-piperidinyl]butyl, N-Cbz-NHCH 2 -, (CH 3 ) 2 NC(O)CH 2 -, and N-Boc-2-aminoethyl. 30 WO 99/06432 PCT/US98/15325 -- 141 -
10. A compound according to Claim 2 wherein R 6 is selected from the group consisting of 2,4-dioxo-tetrahydrofuran-3-yl (3,4-enol), methoxy, ethoxy, iso-propoxy, n-butoxy, t-butoxy, cyclopentoxy, neo-pentoxy, 2-a iso-propyl-4-3-methylcyclohexoxy, 2-p3-isopropyl-4-p-methylcyclohexoxy, 5 -NH 2 , benzyloxy, -NHCH 2 COOH, -NHCH 2 CH 2 COOH, -NH-adamantyl, -NHCH 2 CH 2 COOCH 2 CH 3 , -NHSO 2 -p-CH 3 -4, -NHOR 8 where R 8 is hydrogen, methyl, iso-propyl or benzyl, O-(N-succinimidyl), -O-cholest-5-en-3-p-yl, -OCH 2 -OC(O)C(CH 3 ) 3 , -O(CH 2 )zNHC(O)W where z is 1 or 2 and W is selected from the group consisting of pyrid-3-yl, N 10 methylpyridyl, and N-methyl-1,4-dihydro-pyrid-3-yl, -NR"C(O)-R' where R' is aryl, heteroaryl or heterocyclic and R" is hydrogen or -CH 2 C(O)OCH 2 CH 3 .
11. A compound according to Claims 1 or 2 wherein Q is preferably 15 -C(O)NH- or -C(S)NH-.
12. A compound selected from the group consisting of: N-(toluene-4-sulfonyl)-L-prolyl-L-aspartic acid 4-tert-butyl ester 20 N-(toluene-4-sulfonyl)-L-prolyl-L-asparagine N-(toluene-4-sulfonyl)-L-prolyl-N-(tert-butoxycarbonyl)-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-L-glutamic acid 5-tert-butyl ester 25 N-(toluene-4-sulfonyl)-L-prolyl-L-glutamine N-(toluene-4-sulfonyl)-L-prolyl-N-(carbobenzyloxy)-L-lysine 30 N-(toluene-4-sulfonyl)-L-prolyl-O-benzyl-L-serine N-(toluene-4-sulfonyl)-L-prolyl-p-cyclohexyl-L-alanine N-(toluene-4-sulfonyl)sarcosyl-p-(N-tert-butoxycarbonylpiperidin-4-yl) 35 D,L-alanine WO 99/06432 PCT/US98/15325 -- 142 - N-(toluene-4-sulfonyl)sarcosyl-3-(N-benzoylpiperidin-4-yl)-D,L-alanine N-(toluene-4-sulfonyl)sarcosyl-Ne-tert-butoxycarbonyl-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-N-tert-butoxycarbonyl-D-lysine 5 N-(toluene-4-sulfonyl)-L-prolyl-NE-(allyloxycarbonyl)-L-lysine N-(3,5-ditrifluoromethylbenzenesulfonyl)-L-prolyl-NE (allyloxycarbonyl)-L-lysine 10 N-(toluene-4-sulfonyl)sarcosyl-Ne-(allyloxycarbonyl)-L-lysine N-(toluene-4-sulfonyl)sarcosyl-5-(allyloxycarbonylamino)pentanoic acid 15 N-(toluene-4-sulfonyl)sarcosyl-4-(allyloxycarbonylamino)butanoic acid N-(toluene-4-sulfonyl)-L-prolyl-Ne-(allyloxycarbonyl)-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-4-(allyloxycarbonylamino)butanoic acid 20 N-(toluene-4-sulfonyl)-L-glutaminyl-L-asparagine N-(toluene-4-sulfonyl)-L-prolyl-NE-[N-(toluene-4-sulfonyl)sarcosyl]-L lysine 25 N-(toluene-4-sulfonyl)-L-prolyl-(2,3-dehydro)phenylalanine N-(toluene-4-sulfonyl)-L-prolyl-Ny-(4-phenyl)butyl-L-asparagine 30 N-(toluene-4-sulfonyl)-L-prolyl-N-(indol-3-ylacetyl)-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-NE-[3-(indol-3-yl)propionyl)-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-N-(5-methoxyindol-3-carbonyl)-L 35 lysine N-(toluene-4-sulfonyl)-L-prolyl-Ne-(4-fluorophenoxyacetyl)-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-Ny-R-(1-phenyl)ethyl-L-asparagine 40 N-(toluene-4-sulfonyl)-L-prolyl-Ny-S-(1-phenyl)ethyl-L-asparagine N-(toluene-4-sulfonyl)-L-prolyl-Ny-(4-nitrophenyl)methyl-L-asparagine 45 N-(toluene-4-sulfonyl)-L-prolyl-N8-benzyl-N8-methyl-L-glutamine WO 99/06432 PCTIUS98/1 5325 -- 143 - N-(toluene-4-sulfonyl)-L-prolyl-N5-2-(l1-methylpyrrol-2-yl)ethyl-L glutamine N-(toluene-4-sulfonyl)-L-prolyl-N6-n-butyl-L-glutamine 5 N-(toluene-4-sulfonyl)-L-prolyl-N5-2-(indol-3-yl)ethyl-L-glutamine N-(toluene-4-sulfonyl)sarcosyl-Ny -benzyl-Ny-methyl-L-asparagine 10 N-(toluene-4-sulfonyl)sarcosyl-Ny -2-( 1 -methylpyrrol-2-yl)ethyl-L asparagine N-(toluene-4-sulfonyl)sarcosyl-Ny-n-butyl-L-asparagine 15 N-(toluene-4-sulfonyl)sarcosyl-Ny -2-(indol-3 -yl)ethyl-L-asparagine N-(toluene-4-sulfonyl)sarcosyl-N6-R-( 1 -phenyl)ethyl-L-glutamine N-(toluene-4-sulfonyl)sarcosyl-N&-S-(l1-phenyl)ethyl-L-glutamine 20 N-(toluene-4-sulfonyl)sarcosyl-N6-(4-N,N-dimethylamino phenyl)methyl-L-glutamine N-(toluene-4-.sulfonyl)sarcosyl-N&-(4-nitrophenyl)methyl-L-glutamile 25 N-(toluene-4-sulfonyl)-N-2-(thien-2-yl)glycinyl-N&S-( 1 -phenyl)ethyl-L glutamine N-(toluene-4-sulfonyl)-N-2-(thien-2-yl)glycinyl-Ny -(4-N,N 30 dimethylaminophenyl)methyl-L-asparagifle N-(toluene-4-sulfonyl)-N-2-(thien-2-yl)glycinyl-Ny -(4 nitrophenyl)methyl-L-asparagine 35 N-(toluene-4-sulfonyl)-N-2-(thien-2-yl)glycinyl-Ny-benzyl-Ny-methyl-L asparagine N-(toluene-4-sulfonyl)-N-2-(thien-2-yl)glycinyl-N&-2-(l1-methylpyrrol-2 yl)ethyl-L-glutamine 40 N-.(toluene-4-sulfonyl)-N-2-(thien-2-yl)glycinyl-N-n-butyl-L-glutamfile N-(toluene-4-sulfonyl)-N-2-(thien-2-yl)glycinyl-N-2-(ildol-3-y)ethyl-L glutamine 45 WO 99/06432 PCT/US98/15325 -- 144 - N-(toluene-4-sulfonyl)-N-2-(thien-2-yl)glycinyl-N6-R-(1-phenyl)ethyl-L glutamine N-(toluene-4-sulfonyl)-L-prolyl-Ny-(4-acetamidophenyl)-L-asparagine 5 N-(toluene-4-sulfonyl)-L-prolyl-Ny-(pyrid-4-yl)-L-asparagine N-(toluene-4-sulfonyl)-L-prolyl-Ny-(4-N,N-dimethylaminophenyl)-L asparagine 10 N-(toluene-4-sulfonyl)-L-prolyl-Ny-(3-methoxyphenyl)-L-asparagine N-(toluene-4-sulfonyl)-L-prolyl-N8-(4-chlorophenyl)-L-glutamine 15 N-(toluene-4-sulfonyl)-L-prolyl-N6-(pyrid-2-yl)-L-glutamine N-(toluene-4-sulfonyl)-L-prolyl-N8-(4-methoxyphenyl)-L-glutamine N-(toluene-4-sulfonyl)-L-prolyl-N6-(pyrid-3-yl)-L-glutamine 20 N-(toluene-4-sulfonyl)-L-prolyl-N?-(toluene-4-sulfonyl)-L-arginine N-(toluene-4-sulfonyl)-L-prolyl-N-(ethylaminocarbonyl)-L-lysine 25 N-(toluene-4-sulfonyl)-L-prolyl-N-(phenylaminocarbonyl)-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-N-(4-methoxyphenylamino-carbonyl) L-lysine 30 N-(toluene-4-sulfonyl)-L-(5,5-dimethyl)thiaprolyl-Ne-(tert butoxycarbonyl)-L-lysine methyl ester N-(toluene-4-sulfonyl)-L-(5,5-dimethyl)thiaprolyl-Ne-(tert butoxycarbonyl)-L-lysine 35 N-(toluene-4-sulfonyl)-L-(5,5-dimethyl)thiaprolyl-L-asparagine N-(toluene-4-sulfonyl)-L-prolyl-Ny-(4-N,N-dimethylamino phenyl)methyl-L-asparagine 40 N-(toluene-4-sulfonyl)-L-prolyl-Ny-(4-N,N-dimethylamino phenyl)methyl-L-asparagine methyl ester N-(toluene-4-sulfonyl)-L-prolyl-Ny-2-(N,N-dimethylamino)ethyl-L 45 asparagine WO 99/06432 PCT/US98/15325 -- 145 - N-(toluene-4-sulfonyl)-L-prolyl-NE-(ethylaminocarbonyl)-L-lysine 468 N-(toluene-4-sulfonyl)-L-prolyl-Ne-(N-tert-butoxycarbonylglycinyl)-L lysine 5 N-(toluene-4-sulfonyl)-L-prolyl-N-[N-(carbobenzyloxy)iso-nipecotoyl] L-lysine N-(toluene-4-sulfonyl)-L-prolyl-Ne-(N-toluene-4-sulfonyl-L-prolyl)-L 10 lysine N-(toluene-4-sulfonyl)-L-prolyl-Ne-(isonipecotoyl)-L-lysine N-(toluene-4-sulfonyl)-L-prolyl-3-[N-(carbobenzyl-oxy)amino]propionic 15 acid N-(toluene-4-sulfonyl)-L-prolyl-Ny,Ny-dimethyl-L-asparagine N-(toluene-4-sulfonyl)-L-prolyl-3-[N-(tert-butoxycarbonyl)amino]-2S 20 propionic acid methyl ester N-(toluene-4-sulfonyl)-L-(5-oxo)prolyl-L-asparagine and pharmaceutically acceptable salts thereof as well as any of the ester 25 compounds recited above wherein one ester is replaced with another ester selected from the group consisting of methyl ester, ethyl ester, n-propyl ester, isopropyl ester, n-butyl ester, isobutyl ester, sec-butyl ester and tert butyl ester. 30
13. A method for binding VLA-4 in a biological sample which method comprises contacting the biological sample with a compound of formula A under conditions wherein said compound binds to VLA-4: R 3 O 35 I 11 R'-SO 2 -N(R 2 )-C-Q-CH-C-R 6' A HR 40 where WO 99/06432 PCT/US98/15325 -- 146 - R' is selected from the group consisting of alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocylic, heteroaryl and substituted heteroaryl; R 2 is selected from the group consisting of hydrogen, alkyl, substituted 5 alkyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, and R' and R 2 together with the nitrogen atom bound to R 2 and the SO 2 group bound to R' can form a heterocyclic or a substituted heterocyclic group; 10 R 3 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic and, when R 2 does not form a heterocyclic group with R', R 2 and R 3 together with the nitrogen atom bound to R 2 and the carbon atom bound to R 3 can form a 15 heterocyclic or a substituted heterocyclic group; R 5 is -ALK-X or =CH-Y where ALK is an alkyl group of from 1 to 10 carbon atoms attached via a methylene group (-CH 2 -) to the carbon atom to which it is attached; X is selected from the group consisting of substituted alkylcarbonylamino, substituted alkenylcarbonylamino, substituted 20 alkynylcarbonylamino, heterocyclylcarbonylamino, substituted heterocyclylcarbonylamino, acyl, acyloxy, aminocarbonyloxy, acylamino, oxycarbonylamino, alkoxycarbonyl, substituted alkoxycarbonyl, aryloxycarbonyl, substituted aryloxycarbonyl, cycloalkoxycarbonyl, substituted cycloalkoxycarbonyl, heteroaryloxycarbonyl, substituted 25 heteroaryloxycarbonyl, heterocyclyloxycarbonyl, substituted heterocyclyloxycarbonyl, cycloalkyl, substituted cycloalkyl, saturated heterocyclic, substituted saturated heterocyclic, substituted alkoxy, substituted alkenoxy, substituted alkynoxy, heterocyclyloxy, substituted heterocycloxy, substituted thioalkyl, substituted thioalkenyl, substituted WO 99/06432 PCT/US98/15325 -- 147 - thioalkynyl, aminocarbonylamino, aminothiocarbonylamino, guanidino, amidino, alkylamidino, thioamidino, halogen, cyano, nitro, -OS(O)-alkyl, -OS(O) 2 -substituted alkyl, -OS(O) 2 -cycloalkyl, -OS(O) 2 -substituted cycloalkyl, -OS(O) 2 -aryl, -OS(O) 2 -substituted aryl, -OS(O) 2 -heteroaryl, 5 -OS(O) 2 -substituted heteroaryl, -OS(O) 2 -heterocyclic, -OS(O) 2 -substituted heterocyclic, -OSO 2 -NRR where R is hydrogen or alkyl, -NRS(O) 2 -alkyl, -NRS(O) 2 -substituted alkyl, -NRS(O) 2 -cycloalkyl, -NRS(O) 2 -substituted cycloalkyl, -NRS(O) 2 -aryl, -NRS(O) 2 -substituted aryl, -NRS(O) 2 -heteroaryl, -NRS(O) 2 -substituted heteroaryl, -NRS(O) 2 -heterocyclic, -NRS(O) 2 10 substituted heterocyclic, -NRS(O) 2 -NR-alkyl, -NRS(O) 2 -NR-substituted alkyl, -NRS(O) 2 -NR-cycloalkyl, -NRS(O) 2 -NR-substituted cycloalkyl, -NRS(O) 2 -NR-aryl, -NRS(O) 2 -NR-substituted aryl, -NRS(O) 2 -NR-heteroaryl, -NRS(O) 2 -NR-substituted heteroaryl, -NRS(O) 2 -NR-heterocyclic, -NRS(O) 2 NR-substituted heterocyclic where R is hydrogen or alkyl, -S(O) 2 -alkyl, 15 S(O) 2 -substituted alkyl, -S(O) 2 -aryl, -S(O) 2 -substituted aryl, -S(O) 2 -substituted heteroaryl, -S(O) 2 -substituted heteroaryl, -S(O) 2 -heterocyclic, -S(O) 2 -substituted heterocyclic, mono- and di-(substituted alkyl)amino, N,N-(alkyl, substituted alkyl)amino, N,N-(aryl, substituted alkyl)amino, N,N-(substituted aryl, substituted alkyl)amino, N,N 20 (heteroaryl, substituted alkyl)amino, N,N-(substituted heteroaryl, substituted alkyl)amino, N,N-(heterocyclic, substituted alkyl)amino, N,N-N,N (substituted heterocyclic, substituted alkyl)amino, mono- and di (heterocyclic)amino, mono- and di-(substituted heterocyclic)amino, N,N (alkyl, heterocyclic)amino, N,N-(alkyl, substituted heterocyclic)amino, N,N 25 (aryl, heterocyclic)amino, N,N-(substituted aryl, heterocyclic)amino, N,N (aryl, substituted heterocyclic)amino, N,N-(substituted aryl, substituted heterocyclic)amino, N,N-(heteroaryl, heterocyclic)amino, N,N-(heteroaryl, substituted heterocyclic)amino, N,N-(substituted heteroaryl, heterocyclic)amino, and N,N-(substituted heteroaryl, substituted 30 heterocyclic)amino; and Y is selected from the group consisting of alkyl, WO 99/06432 PCT/US98/15325 -- 148- substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic and substituted heterocyclic with the proviso that when R 5 is = CH-Y then (H) is removed from the formula; 5 R 6 ' is selected from the group consisting of 2,4-dioxo-tetrahydrofuran-3 yl (3,4-enol), hydroxyl, amino, alkoxy, substituted alkoxy, cycloalkoxy, substituted cycloalkoxy, -O-(N-succinimidyl), -NH-adamantyl, -O-cholest-5 en-3-p-yl, -NHOY where Y is hydrogen, alkyl, substituted alkyl, aryl, and substituted aryl, -NH(CH 2 )pCOOY where p is an integer of from 1 to 8 and 10 Y is as defined above, -OCH 2 NR 9 Rlo where R 9 is selected from the group consisting of -C(O)-aryl and -C(O)-substituted aryl and R 1 o is selected from the group consisting of hydrogen and -CH 2 COOR" where R 1 is alkyl, and NHSO 2 Z where Z is alkyl, substituted alkyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 15 heterocyclic and substituted heterocyclic; Q is -C(X)NR 7 - wherein R 7 is selected from the group consisting of hydrogen and alkyl; and X is selected from the group consisting of oxygen and sulfur; and pharmaceutically acceptable salts thereof 20 with the provisos that A. when R 1 is p-methylphenyl, R 2 and R 3 are joined together with the nitrogen atom bound to R 2 and the carbon atom bound to R 3 to form a pyrrolidinyl ring, R 6 I - is hydroxyl, and Q is -C(O)NH-, then R 5 is not -CH 2 C(O)-O-t-butyl or -CH 2 CH 2 C(O)-O-t-butyl; 25 B. when R' is p-methylphenyl, R 2 is methyl, R 3 is hydrogen, R 6 ' is hydroxyl and Q is -C(O)NH-, then R is not -CH 2 (N-benzylpiperin-4-yl); and C. when R' is p-methylphenyl, R 2 and R 3 are joined together with the nitrogen atom bound to R 2 and the carbon atom bound to R 3 to form a 30 pyrrolidinyl ring, R 6 ' is methoxy, and Q is -C(O)NH-, then R 5 is not WO 99/06432 PCT/US98/15325 -- 149 - -CH 2 CH 2 COO-t-butyl or -CH 2 COO-t-butyl.
14. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of one or more 5 compounds of Claim 13.
15. A method for treating an inflammatory disease in a mammalian patient which disease is mediated by VLA-4 which method comprises administering to said patient a therapeutically effective amount of the 10 pharmaceutical composition of Claim 14.
16. The method according to Claim 15 wherein said inflammatory condition is selected from the group consisting of asthma, Alzheimer's disease, atherosclerosis, AIDS dementia, diabetes (including acute juvenile 15 onset diabetis), inflammatory bowel disease (including ulcerative colitis and Crohn's disease), multiple sclerosis, rheumatoid arthritis, tissue transplantation, tumor metastasis, meningitis, encephalitis, stroke, and other cerebral traumas, nephritis, retinitis, atopic dermatitis, psoriasis, myocardial ischemia and acute leukocyte-mediated lung injury such as that which occurs 20 in adult respiratory distress syndrome.
AU85850/98A 1997-07-31 1998-07-31 Dipeptide and related compounds which inhibit leukocyte adhesion mediated by vla-4 Abandoned AU8585098A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US90441797A 1997-07-31 1997-07-31
US08904417 1997-07-31
PCT/US1998/015325 WO1999006432A1 (en) 1997-07-31 1998-07-31 Dipeptide and related compounds which inhibit leukocyte adhesion mediated by vla-4

Publications (1)

Publication Number Publication Date
AU8585098A true AU8585098A (en) 1999-02-22

Family

ID=25419120

Family Applications (1)

Application Number Title Priority Date Filing Date
AU85850/98A Abandoned AU8585098A (en) 1997-07-31 1998-07-31 Dipeptide and related compounds which inhibit leukocyte adhesion mediated by vla-4

Country Status (12)

Country Link
EP (1) EP1001971A1 (en)
JP (1) JP2001512135A (en)
KR (1) KR20010022406A (en)
CN (1) CN1265670A (en)
AU (1) AU8585098A (en)
BR (1) BR9812111A (en)
CA (1) CA2290749A1 (en)
HU (1) HUP0002495A3 (en)
IL (1) IL133639A0 (en)
NO (1) NO20000410L (en)
PL (1) PL338373A1 (en)
WO (1) WO1999006432A1 (en)

Families Citing this family (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6903075B1 (en) 1997-05-29 2005-06-07 Merck & Co., Inc. Heterocyclic amide compounds as cell adhesion inhibitors
EP0984981B1 (en) * 1997-05-30 2003-12-17 Celltech Therapeutics Limited Anti-inflammatory tyrosine derivatives
US6482849B1 (en) 1997-06-23 2002-11-19 Tanabe Seiyaku Co., Ltd. Inhibitors of α4β1 mediated cell adhesion
US6939855B2 (en) 1997-07-31 2005-09-06 Elan Pharmaceuticals, Inc. Anti-inflammatory compositions and method
US6559127B1 (en) 1997-07-31 2003-05-06 Athena Neurosciences, Inc. Compounds which inhibit leukocyte adhesion mediated by VLA-4
ATE267168T1 (en) * 1997-11-24 2004-06-15 Merck & Co Inc BETA-ALANINE DERIVATIVES AS CELL ADHESION INHIBITORS
US6645939B1 (en) 1997-11-24 2003-11-11 Merck & Co., Inc. Substituted β-alanine derivatives as cell adhesion inhibitors
MY153569A (en) 1998-01-20 2015-02-27 Mitsubishi Tanabe Pharma Corp Inhibitors of ?4 mediated cell adhesion
US6329372B1 (en) 1998-01-27 2001-12-11 Celltech Therapeutics Limited Phenylalanine derivatives
US6555562B1 (en) 1998-02-26 2003-04-29 Celltech R&D Limited Phenylalanine derivatives
US6521626B1 (en) 1998-03-24 2003-02-18 Celltech R&D Limited Thiocarboxamide derivatives
GB9811159D0 (en) 1998-05-22 1998-07-22 Celltech Therapeutics Ltd Chemical compounds
GB9811969D0 (en) 1998-06-03 1998-07-29 Celltech Therapeutics Ltd Chemical compounds
US6685617B1 (en) 1998-06-23 2004-02-03 Pharmacia & Upjohn Company Inhibitors of α4β1 mediated cell adhesion
GB9814414D0 (en) 1998-07-03 1998-09-02 Celltech Therapeutics Ltd Chemical compounds
US6339101B1 (en) * 1998-08-14 2002-01-15 Gpi Nil Holdings, Inc. N-linked sulfonamides of N-heterocyclic carboxylic acids or isosteres for vision and memory disorders
US6333340B1 (en) * 1998-08-14 2001-12-25 Gpi Nil Holdings, Inc. Small molecule sulfonamides for vision and memory disorders
GB9821061D0 (en) 1998-09-28 1998-11-18 Celltech Therapeutics Ltd Chemical compounds
GB9821222D0 (en) 1998-09-30 1998-11-25 Celltech Therapeutics Ltd Chemical compounds
GB9825652D0 (en) 1998-11-23 1999-01-13 Celltech Therapeutics Ltd Chemical compounds
GB9826174D0 (en) 1998-11-30 1999-01-20 Celltech Therapeutics Ltd Chemical compounds
CA2359112A1 (en) 1999-01-22 2000-07-27 Elan Pharmaceuticals, Inc. Fused ring heteroaryl and heterocyclic compounds which inhibit leukocyte adhesion mediated by vla-4
US6479492B1 (en) 1999-01-22 2002-11-12 Elan Pharmaceuticals, Inc. Compounds which inhibit leukocyte adhesion mediated by VLA-4
US6436904B1 (en) 1999-01-25 2002-08-20 Elan Pharmaceuticals, Inc. Compounds which inhibit leukocyte adhesion mediated by VLA-4
US6407066B1 (en) 1999-01-26 2002-06-18 Elan Pharmaceuticals, Inc. Pyroglutamic acid derivatives and related compounds which inhibit leukocyte adhesion mediated by VLA-4
AU2623800A (en) * 1999-01-26 2000-08-07 American Home Products Corporation Pyroglutamic acid derivatives and related compounds which inhibit leukocyte adhesion mediated by vla-4
US6518283B1 (en) 1999-05-28 2003-02-11 Celltech R&D Limited Squaric acid derivatives
BR0013248A (en) * 1999-08-13 2002-07-23 Biogen Inc Cell adhesion inhibitors
US6534513B1 (en) 1999-09-29 2003-03-18 Celltech R&D Limited Phenylalkanoic acid derivatives
US6455539B2 (en) 1999-12-23 2002-09-24 Celltech R&D Limited Squaric acid derivates
AU2018301A (en) 1999-12-28 2001-07-24 Pfizer Products Inc. Non-peptidyl inhibitors of vla-4 dependent cell binding useful in treating inflammatory, autoimmune, and respiratory diseases
EP1332132B1 (en) 2000-04-17 2007-10-10 UCB Pharma, S.A. Enamine derivatives as cell adhesion molecules
US6545013B2 (en) 2000-05-30 2003-04-08 Celltech R&D Limited 2,7-naphthyridine derivatives
US6403608B1 (en) 2000-05-30 2002-06-11 Celltech R&D, Ltd. 3-Substituted isoquinolin-1-yl derivatives
AU2001267753A1 (en) 2000-07-07 2002-01-21 Celltech R And D Limited Squaric acid derivatives containing a bicyclic heteroaromatic ring as integrin antagonists
JP2004505110A (en) 2000-08-02 2004-02-19 セルテック アール アンド ディ リミテッド 3-position substituted isoquinolin-1-yl derivative
MY129000A (en) 2000-08-31 2007-03-30 Tanabe Seiyaku Co INHIBITORS OF a4 MEDIATED CELL ADHESION
JP2005022976A (en) * 2001-07-18 2005-01-27 Ajinomoto Co Inc Carboxylic acid derivative
MY140707A (en) 2002-02-28 2010-01-15 Mitsubishi Tanabe Pharma Corp Process for preparing a phenylalanine derivative and intermediates thereof
ES2197003B1 (en) * 2002-04-08 2005-03-16 J. URIACH &amp; CIA S.A. NEW ANTAGONIST COMPOUNDS OF INTEGRINAS ALFA.
TW200307671A (en) 2002-05-24 2003-12-16 Elan Pharm Inc Heteroaryl compounds which inhibit leukocyte adhesion mediated by α 4 integrins
TWI281470B (en) 2002-05-24 2007-05-21 Elan Pharm Inc Heterocyclic compounds which inhibit leukocyte adhesion mediated by alpha4 integrins
US7807847B2 (en) * 2004-07-09 2010-10-05 Vascular Biogenics Ltd. Process for the preparation of oxidized phospholipids
US7196112B2 (en) 2004-07-16 2007-03-27 Biogen Idec Ma Inc. Cell adhesion inhibitors
AU2008219007A1 (en) 2007-02-20 2008-08-28 Merrimack Pharmaceuticals, Inc. Methods of treating multiple sclerosis by administration of alpha-fetoprotein in combination with an integrin antagonist
AU2009234253C1 (en) 2008-04-11 2015-05-07 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
WO2015136468A1 (en) 2014-03-13 2015-09-17 Prothena Biosciences Limited Combination treatment for multiple sclerosis
WO2017173302A2 (en) 2016-04-01 2017-10-05 The Regents Of The University Of California Inhibitors of integrin alpha 5 beta 1 and methods of use
CN109516925B (en) * 2018-10-31 2021-07-16 陕西慧康生物科技有限责任公司 Synthesis method of glutamic acid-1-methyl ester-5-tert-butyl ester
CN114605348B (en) * 2022-05-12 2022-08-16 北京鑫开元医药科技有限公司 Compounds having HDAC inhibitory activity, preparation method, compositions and uses thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2679903B1 (en) * 1991-08-02 1993-12-03 Elf Sanofi DERIVATIVES OF N-SULFONYL INDOLINE CARRYING AN AMIDIC FUNCTION, THEIR PREPARATION, THE PHARMACEUTICAL COMPOSITIONS CONTAINING SAME.
WO1994007815A2 (en) * 1992-09-25 1994-04-14 Abbott Laboratories Small peptide anaphylatoxin receptor ligands
EP0673247A4 (en) * 1992-12-01 1996-05-01 Merck & Co Inc Fibrinogen receptor antagonists.
JPH0873422A (en) * 1994-09-07 1996-03-19 Kdk Corp New amino acid ester and method for detecting leukocyte, esterase or protease
US6306840B1 (en) * 1995-01-23 2001-10-23 Biogen, Inc. Cell adhesion inhibitors

Also Published As

Publication number Publication date
CN1265670A (en) 2000-09-06
CA2290749A1 (en) 1999-02-11
HUP0002495A1 (en) 2000-12-28
KR20010022406A (en) 2001-03-15
HUP0002495A3 (en) 2001-01-29
PL338373A1 (en) 2000-10-23
IL133639A0 (en) 2001-04-30
BR9812111A (en) 2000-07-18
NO20000410L (en) 2000-03-28
JP2001512135A (en) 2001-08-21
NO20000410D0 (en) 2000-01-27
EP1001971A1 (en) 2000-05-24
WO1999006432A1 (en) 1999-02-11

Similar Documents

Publication Publication Date Title
EP1001974B1 (en) 4-amino-phenylalanine type compounds which inhibit leukocyte adhesion mediated by vla-4
AU756696B2 (en) Substituted phenylalanine type compounds which inhibit leukocyte adhesion mediated by VLA-4
EP1000051B1 (en) Carbamyloxy compounds which inhibit leukocyte adhesion mediated by vla-4
AU8585098A (en) Dipeptide and related compounds which inhibit leukocyte adhesion mediated by vla-4
US7229970B2 (en) Carbamyloxy compounds which inhibit leukocyte adhesion mediated by VLA-4
EP0994895A1 (en) Dipeptide compounds which inhibit leukocyte adhesion mediated by vla-4
AU8678698A (en) Compounds which inhibit leukocyte adhesion mediated by vla-4
AU8585198A (en) Benzyl compounds which inhibit leukocyte adhesion mediated by vla-4
AU8823498A (en) Sulfonylated dipeptide compounds which inhibit leukocyte adhesion mediated by vla-4
US6949570B2 (en) Compounds which inhibit leukocyte adhesion mediated by VLA-4
US6492421B1 (en) Substituted phenylalanine type compounds which inhibit leukocyte adhesion mediated by VLA-4
US6423688B1 (en) Dipeptide and related compounds which inhibit leukocyte adhesion mediated by VLA-4
US6362341B1 (en) Benzyl compounds which inhibit leukocyte adhesion mediated by VLA-4
US6291453B1 (en) 4-amino-phenylalanine type compounds which inhibit leukocyte adhesion mediated by VLA-4
WO2000043413A2 (en) Pyroglutamic acid derivatives and related compounds which inhibit leukocyte adhesion mediated by vla-4
US7030114B1 (en) Compounds which inhibit leukocyte adhesion mediated by VLA-4
US7166580B2 (en) Compounds which inhibit leukocyte adhesion mediated by VLA-4
EP1150997A1 (en) Compounds which inhibit leukocyte adhesion mediated by vla-4

Legal Events

Date Code Title Description
MK5 Application lapsed section 142(2)(e) - patent request and compl. specification not accepted