WO1998053818A1 - Sulfonamides as cell adhesion inhibitors - Google Patents

Sulfonamides as cell adhesion inhibitors Download PDF

Info

Publication number
WO1998053818A1
WO1998053818A1 PCT/US1998/010952 US9810952W WO9853818A1 WO 1998053818 A1 WO1998053818 A1 WO 1998053818A1 US 9810952 W US9810952 W US 9810952W WO 9853818 A1 WO9853818 A1 WO 9853818A1
Authority
WO
WIPO (PCT)
Prior art keywords
aryl
optionally substituted
independently selected
ioalkyl
alkyl
Prior art date
Application number
PCT/US1998/010952
Other languages
French (fr)
Inventor
Philippe L. Durette
William K. Hagmann
Malcolm Maccoss
Sander G. Mills
Richard A. Mumford
Original Assignee
Merck & Co., Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB9714335.8A external-priority patent/GB9714335D0/en
Priority claimed from GBGB9800684.4A external-priority patent/GB9800684D0/en
Application filed by Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to EP98924989A priority Critical patent/EP0998282A4/en
Priority to JP50093999A priority patent/JP2002501537A/en
Priority to AU77032/98A priority patent/AU728435B2/en
Priority to US09/424,823 priority patent/US6221888B1/en
Priority to CA002291708A priority patent/CA2291708A1/en
Publication of WO1998053818A1 publication Critical patent/WO1998053818A1/en

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
    • C07K5/06191Dipeptides containing heteroatoms different from O, S, or N
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C07K5/06008Dipeptides 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
    • 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
    • C07K5/06034Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms
    • 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
    • C07K5/06034Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms
    • C07K5/06052Val-amino acid
    • 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
    • C07K5/06078Dipeptides with the first amino acid being neutral and aromatic or cycloaliphatic
    • 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
    • C07K5/06113Asp- or Asn-amino acid
    • 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

Definitions

  • the compounds of the present invention are antagonists of the VLA-4 integrin ("very late antigen- 4"; CD49d/CD29; or c-4 ⁇ l) and/or the ⁇ 4 ⁇ 7 integrin (LPAM-1 and ⁇ 4 ⁇ p), thereby blocking the binding of VLA-4 integrin ("very late antigen- 4"; CD49d/CD29; or c-4 ⁇ l) and/or the ⁇ 4 ⁇ 7 integrin (LPAM-1 and ⁇ 4 ⁇ p), thereby blocking the binding of VLA-4 integrin ("very late antigen- 4"; CD49d/CD29; or c-4 ⁇ l) and/or the ⁇ 4 ⁇ 7 integrin (LPAM-1 and ⁇ 4 ⁇ p), thereby blocking the binding of VLA-4 integrin ("very late antigen- 4"; CD49d/CD29; or c-4 ⁇ l) and/or the ⁇ 4 ⁇ 7 integrin (LPAM-1 and ⁇ 4 ⁇ p), thereby blocking the binding of VLA-4 integrin ("very late antigen
  • VLA-4 to its various ligands, such as VCAM-1 and regions of fibronectin and/or ⁇ 4 ⁇ 7 to its various ligands, such as MadCAM-1, VCAM-1 and fibronectin.
  • these antagonists are useful in inhibiting cell adhesion processes including cell activation, migration, proliferation and differentiation.
  • VLA-4 and/or ⁇ 4 ⁇ 7 binding and cell adhesion and activation are useful in the treatment, prevention and suppression of diseases mediated by VLA-4 and/or ⁇ 4 ⁇ 7 binding and cell adhesion and activation, such as multiple sclerosis, asthma, allergic rhinitis, allergic conjunctivitis, inflammatory lung diseases, rheumatoid arthritis, septic arthritis, type I diabetes, organ transplantation, restenosis, autologous bone marrow transplantation, inflammatory sequelae of viral infections, myocarditis, inflammatory bowel disease including ulcerative colitis and Crohn 's disease, certain types of toxic and immune-based nephritis, contact dermal hypersensitivity, psoriasis, tumor metastasis, and atherosclerosis.
  • diseases mediated by VLA-4 and/or ⁇ 4 ⁇ 7 binding and cell adhesion and activation such as multiple sclerosis, asthma, allergic rhinitis, allergic conjunctivitis, inflammatory lung diseases, rhe
  • the present invention relates to sulfonamide derivatives which are useful for the inhibition and prevention of leukocyte adhesion and leukocyte adhesion-mediated pathologies.
  • This invention also relates to compositions containing such compounds and methods of treatment using such compounds.
  • CAMs cell adhesion molecules
  • selectins include the selectins, integrins, cadherins and immunoglobulins.
  • CAMs play an essential role in both normal and pathophysiological processes. Therefore, the targetting of specific and relevant CAMs in certain disease conditions without interfering with normal cellular functions is essential for an effective and safe therapeutic agent that inhibits cell-cell and cell-matrix interactions.
  • the integrin superfamily is made up of structurally and functionally related glycoproteins consisting of ⁇ and ⁇ heterodimeric, transmembrane receptor molecules found in various combinations on nearly every mammalian cell type, (for reviews see: E. C. Butcher, Cell, 61, 1033 (1991); T. A. Springer, Cell, 76, 301 (1994); D. Cox et al., "The Pharmacology of the Integrins.” Medicinal Research Rev, 14. 195 (1994) and V, W. Engleman et al., "Cell Adhesion Integrins as Pharmaceutical Targets.” in Ann. Repts. in Medicinal Chemistry. Vol. 31, J. A. Bristol, Ed.; Acad. Press, NY, 1996, p. 191).
  • VLA-4 (“very late antigen-4"; CD49d/CD29; or ⁇ -i ⁇ l) is an integrin expressed on all leukocytes, except platelets and mature neutrophils, including dendritic cells and macrophage-like cells and is a key mediator of the cell-cell and cell-matrix interactions of of these cell types (see M. E. Hemler, "VLA Proteins in the Integrin Family: Structures, Functions, and Their Role on Leukocytes.” Ann. Rev. Immunol. 8, 365 (1990)).
  • the ligands for VLA-4 include vascular cell adhesion molecule-1 (VCAM-1) and the CS-1 domain of fibronectin (FN).
  • VCAM-1 is a member of the Ig superfamily and is expressed in vivo on endothelial cells at sites of inflammation. (See R. Lobb et al. "Vascular Cell Adhesion Molecule 1.” in Cellular and Molecular Mechanisms of Inflammation, C. G. Cochrane and M. A. Gimbrone, Eds.; Acad. Press, San Diego, 1993, p. 151.) VCAM-1 is produced by vascular endothelial cells in response to pro-inflammatory cytokines (See A. J. H. Gearing and W. Newman, "Circulating adhesion molecules in disease.”,
  • the CS-1 domain is a 25 amino acid sequence that arises by alternative splicing within a region of fibronectin.
  • a role for VLA-4/CS-1 interactions in inflammatory conditions has been proposed (see M. J. Elices, "The integrin c ⁇ 4 ⁇ (VLA-
  • ⁇ 4 ⁇ 7 (also referred to as LPAM-1 and ⁇ 4 ⁇ p) is an integrin expressed on leukocytes and is a key mediator of leukocyte trafficking and homing in the gastrointestinal tract (see C. M. Parker et al., Proc. Natl. Acad. Sci. USA, r ⁇ , 1924 (1992)).
  • the ligands for ⁇ 4 ⁇ 7 include mucosal addressing cell adhesion molecule-1 (MadCAM-1) and, upon activation of ⁇ 4 ⁇ 7, VCAM-1 and fibronectin (Fn).
  • MadCAM-1 is a member of the Ig superfamily and is expressed in vivo on endothelial cells of gut-associated mucosal tissues of the small and large intestine ("Peyer 's Patches") and lactating mammary glands. (See M. J. Briskin et al., Nature. 363. 461 (1993); A. Hamann et al., J. Immunol.. 152.3282 (1994)). MadCAM-1 can be induced in vitro by proinflammatory stimuli (See E. E. Sikorski et al. J. Immunol.. 151. 5239 (1993)). MadCAM-1 is selectively expressed at sites of lymphocyte extravasation and specifically binds to the integrin, o-4 ⁇ 7.
  • Neutralizing anti-0.4 antibodies or blocking peptides that inhibit the interaction between VLA-4 and/or ⁇ 4 ⁇ 7 and their ligands have proven efficacious both prophylactically and therapeutically in several animal models of disease, including i) experimental allergic encephalomyelitis, a model of neuronal demyelination resembling multiple sclerosis (for example, see T. Yednock et al., "Prevention of experimental autoimmune encephalomyelitis by antibodies against 0.4 ⁇ i integrin.” Nature. 356, (_3 (1993) and E. Keszthelyi et al., "Evidence for a prolonged role of 0.4 integrin throughout active experimental allergic encephalomyelitis.” Neurology.
  • VLA-4 interactions in other diseases, including rheumatoid arthritis; various melanomas, carcinomas, and sarcomas; inflammatory lung disorders; acute respiratory distress syndrome (ARDS); atherosclerotic plaque formation; restenosis; uveitis and circulatory shock (for examples, see A. A. Postigo et al., "The ⁇ 4 ⁇ /VCAM-l adhesion pathway in physiology and disease.”, Res. Immunol.. 144. 723 (1994) and J.-X. Gao and A. C. Issekutz, "Expression of VCAM-1 and VLA-4 dependent T-lymphocyte adhesion to dermal fibroblasts stimulated with proinflammatory cytokines.” Immunol. 89, 375 (1996)).
  • VLA-4- and ⁇ 4 ⁇ 7-dependent cell adhesion that have improved pharmacokinetic and pharmacodynamic properties such as oral bioavailability and significant duration of action.
  • Such compounds would prove to be useful for the treatment, prevention or suppression of various pathologies mediated by VLA-4 and ⁇ 4 ⁇ 7 binding and cell adhesion and activation.
  • One aspect of the present invention provides a method for the treatment of diseases, disorders, conditions or symptoms mediated by cell adhesion in a mammal which comprises administering to said mammal an effective amount of a compound Formula I:
  • Rl is 1) Ci-ioalkyl
  • R2 and R3 are independently selected from
  • R is 1) hydrogen
  • heteroaryl-Ci_io a lkyl wherein alkyl, alkenyl, and alkynyl are optionally substituted with one to four substituents independently selected from R a ; and aryl and heteroaryl optionally substituted with one to four substituents independently selected from R ⁇ ; or
  • R3, R4 and the carbon to which they are attached form a 3-7 membered mono- or bicyclic ring containing 0-2 heteroatoms selected from N, O and S;
  • B is 1) hydrogen, 2) Ci-io alkyl,
  • R is 1) hydrogen
  • R7 is 1) hydrogen
  • heteroaryl-C ⁇ _ oalkyl wherein alkyl, alkenyl and alkynyl are optionally substituted with one to four substituents selected from Rx, and aryl and heteroaryl are optionally substituted with one to four substituents independently selected from R ; or
  • R a is 1) Cy, or 2) a group selected from Rx; wherein Cy is optionally substituted with one to four substituents independently selected from R c ;
  • Rk is 1) a group selected from R a ,
  • aryl Ci-ioalkyl 6) heteroaryl Ci-io alkyl, wherein alkyl, alkenyl, alkynyl, aryl, heteroaryl are optionally substituted with a group independently selected from R c ;
  • R c is 1) halogen
  • Rd and R e are independently selected from hydrogen, Ci-ioalkyl, C2-10alkenyl, C2-10alkynyl, Cy and Cy Ci-ioalkyl, wherein alkyl, alkenyl, alkynyl and Cy is optionally substituted with one to four substituents independently selected from R c ; or R" and R e together with the atoms to which they are attached form a heterocyclic ring of 5 to 7 members containing 0-2 additional heteroatoms independently selected from oxygen, sulfur and nitrogen;
  • W and Rg are independently selected from hydrogen, Ci-ioalkyl, Cy and Cy Ci-ioalkyl; or R* and RS together with the carbon to which they are attached form a ring of 5 to 7 members containing 0-2 heteroatoms independently selected from oxygen, sulfur and nitrogen;
  • R n is 1) hydrogen
  • alkyl, alkenyl, and alkynyl are optionally substituted with one to four substituents independently selected from R a ; and aryl and heteroaryl are each optionally substituted with one to four substituents independently selected from R D ;
  • Ri is 1) Ci-ioalkyl
  • alkyl, alkenyl, alkynyl and aryl are each optionally substituted with one to four substituents independently selected from R c ;
  • R X is 1) -ORd
  • heterocyclyl wherein alkyl, alkenyl, alkynyl and aryl are each optionally substituted with one to four substituents independently selected from Rx;
  • Cy is cycloalkyl, heterocyclyl, aryl, or heteroaryl
  • n is an integer from 1 to 2;
  • n is an integer from 1 to 10;
  • X is 1) -C(O)ORd
  • compounds of formula I are those wherein R ⁇ is Cy.
  • Rl Cy is preferably aryl optionally substituted with one to four substituents selected from R D . More preferred R* is phenyl with a substituent on the
  • compounds of Formula I are those wherein R ⁇ is H or Ci-6alkyl.
  • R ⁇ is H or Ci-3alkyl, more preferably H or methyl.
  • compounds of Formula I are those wherein one of R3 and R4 is other than hydrogen.
  • R3 and R4 are hydrogen and the other is Cy, Cy-C]-. ⁇ oalkyl, or C-j_.iQa_ls.yl, each of which is optionally substituted as provided under Formula I.
  • R3/R4 Cy is preferably aryl or heteroaryl each optionally substituted with one to four substituents selected from R D ; and alkyl is optionally substituted with one to four substituents independently selected from R a .
  • R /R i s selected from phenyl; Cy-C ⁇ _3alkyl wherein Cy is thienyl, pyridyl or phenyl optionally substituted with one or two groups selected from phenyl, hydroxy, halogen, cyano, and nitro; and C ⁇ .galkyl optionally substituted with CO2H or CO H2.
  • compounds of Formula I are those wherein one of R6 and R? is other than hydrogen.
  • one of R ⁇ and R? is hydrogen and the other is aryl, aryl-Ci-l ⁇ alkyl, or Ci-ioalkyl, wherein aryl is optionally substituted with one to four substituents independently selected from Ry, and alkyl is optionally substituted with one to four substituents selected from Rx.
  • compounds of Formula I are those wherein X is -C(O)ORd.
  • the cell adhesion is mediated by VLA-4, and the compounds are of Formula la as described hereinbelow.
  • R2 is 1) hydrogen, or
  • Ci- ⁇ alk l one of R and R4 is hydrogen and the other is 1) aryl, 2) heteroaryl,
  • Ci.ioalkyl wherein aryl and heteroaryl are each optionally substituted with one to four substituents selected from R"; and alkyl is optionally substituted with one to four substituents independently selected from R a ; or R3, R4 and the carbon to which they are attached form a 3-7 membered mono- or bicyclic ring containing 0-2 heteroatoms selected from N, O and S; one of R6 and R ⁇ is hydrogen and the other is
  • Ci.ioalkyl wherein aryl is optionally substituted with one to four substituents independently selected from Ry, and alkyl is optionally substituted with one to four substituents selected from R x ;
  • R a , R D , R x and Ry are as defined above under Formula I.
  • Rb is selected from Ci-ioalkoxy, halogen, cyano, and trifluoromethyl;
  • R2 is H or methyl; one of R3 and R4 is hydrogen and the other is phenyl, Cy-C ⁇ _3alkyl (wherein Cy is thienyl, pyridyl or phenyl optionally substiuted with one or two groups selected from phenyl, hydroxy, halogen, cyano, and nitro); and Ci. alkyl optionally substituted with CO2H or CONH2; one of R6 and R? is hydrogen and the other is aryl-C ⁇ _
  • the present compounds are generally composed of three domains: 1) a sulfonyl moiety, 2) amino acid 1, and 3) amino acid 2, and are named in a manner similar to that used to name oligopeptides.
  • Alkyl as well as other groups having the prefix “alk”, such as alkoxy, alkanoyl, means carbon chains which may be linear or branched or combinations thereof.
  • alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec- and tert-butyl, pentyl, hexyl, heptyl, octyl, nonyl, and the like.
  • alkenyl means carbon chains which contain at least one carbon-carbon double bond, and which may be linear or branched or combinations thereof. Examples of alkenyl include vinyl, allyl, isopropenyl, pentenyl, hexenyl, heptenyl, 1-propenyl, 2-butenyl, 2- methyl-2-butenyl, and the like.
  • Alkynyl means carbon chains which contain at least one carbon-carbon triple bond, and which may be linear or branched or combinations thereof. Examples of alkynyl include ethynyl, propargyl, 3-m ethyl- 1-pentynyl, 2-heptynyl and the like.
  • Cycloalkyl means mono- or bicyclic saturated carbocyclic rings, each of which having from 3 to 10 carbon atoms. The term also includes monocyclic rings fused to an aryl group in which the point of attachment is on the non-aromatic portion.
  • cycloalkyl examples include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, tetrahydronaphthyl, decahydronaphthyl, indanyl, and the like.
  • Aryl means mono- or bicyclic aromatic rings containing only carbon atoms.
  • the term also includes aryl group fused to a monocyclic cycloalkyl or monocyclic heterocyclyl group in which the point of attachment is on the aromatic portion.
  • aryl include phenyl, naphthyl, indanyl, indenyl, tetrahydronaphthyl, 2,3- dihydrobenzofuranyl, benzopyranyl, 1,4-benzodioxanyl, and the like.
  • Heteroaryl means a mono- or bicyclic aromatic ring containing at least one heteroatom selected from N, O and S, with each ring containing 5 to 6 atoms.
  • heteroaryl include pyrrolyl, isoxazolyl, isothiazolyl, pyrazolyl, pyridyl, oxazolyl, oxadiazolyl, thiadiazolyl, thiazolyl, imidazolyl, triazolyl, tetrazolyl, furanyl, triazinyl, thienyl, pyrimidyl, pyridazinyl, pyrazinyl, benzoxazolyl, benzothiazolyl, benzimidazolyl, benzofuranyl, benzothiophenyl, furo(2,3-b)pyridyl, quinolyl, indolyl, isoquinolyl, and the like.
  • Heterocyclyl means mono- or bicyclic saturated rings containing at least one heteroatom selected from N, S and O, each of said ring having from 3 to 10 atoms in which the point of attachment may be carbon or nitrogen.
  • the term also includes monocyclic heterocycle fused to an aryl or heteroaryl group in which the point of attachment is on the non-aromatic portion.
  • heterocyclyl examples include pyrrolidinyl, piperidinyl, piperazinyl, imidazolidinyl, 2,3-dihydrofuro(2,3-b)pyridyl, benzoxazinyl, tetrahydrohydroquinolinyl, tetrahydroisoquinolinyl, dihydroindolyl, and the like.
  • the term also includes partially unsaturated monocyclic rings that are not aromatic, such as 2- or 4- pyridones attached through the nitrogen or N-substituted-(lH,3H)- pyrimidine-2,4-diones (N-substituted uracils).
  • Halogen includes fluorine, chlorine, bromine and iodine.
  • Optical Isomers Diastereomers - Geometric Isomers - Tautomers
  • Compounds of Formula I contain one or more asymmetric centers and can thus occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. The present invention is meant to comprehend all such isomeric forms of the compounds of Formula I.
  • tautomers Some of the compounds described herein may exist with different points of attachment of hydrogen, referred to as tautomers. Such an example may be a ketone and its enol form known as keto-enol tautomers. The individual tautomers as well as mixture thereof are encompassed with compounds of Formula I.
  • Compounds of the Formula I may be separated into diastereoisomeric pairs of enantiomers by, for example, fractional crystallization from a suitable solvent, for example methanol or ethyl acetate or a mixture thereof.
  • a suitable solvent for example methanol or ethyl acetate or a mixture thereof.
  • the pair of enantiomers thus obtained may be separated into individual stereoisomers by conventional means, for example by the use of an optically active acid as a resolving agent.
  • any enantiomer of a compound of the general Formula I or la may be obtained by stereospecific synthesis using optically pure starting materials or reagents of known configuration.
  • salts refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids.
  • Salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particularly preferred are the ammonium, calcium, magnesium, potassium, and sodium salts.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N -dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl-morpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
  • basic ion exchange resins such as
  • salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids.
  • acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p- toluenesulfonic acid, and the like.
  • Particularly preferred are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, and tartaric acids.
  • Another aspect of the present invention provides a method for the treatment (including prevention, alleviation, amelioration or suppression) of diseases or disorders or symptoms mediated by VLA-4 and or ⁇ 4 ⁇ 7 binding and cell adhesion and activation, which comprises administering to a mammal an effective amount of a compound of Formula I.
  • Such diseases, disorders, conditions or symptoms are for example (1) multiple sclerosis, (2) asthma, (3) allergic rhinitis, (4) allergic conjunctivitis, (5) inflammatory lung diseases, (6) rheumatoid arthritis, (7) septic arthritis, (8) type I diabetes, (9) organ transplantation rejection, (10) restenosis, (11) autologous bone marrow transplantation, (12) inflammatory sequelae of viral infections, (13) myocarditis, (14) inflammatory bowel disease including ulcerative colitis and Crohn 's disease, (15) certain types of toxic and immune-based nephritis, (16) contact dermal hypersensitivity, (17) psoriasis, (18) tumor metastasis, and (19) atherosclerosis.
  • prophylactic or therapeutic dose of a compound of Formula I will, of course, vary with the nature of the severity of the condition to be treated and with the particular compound of Formula I and its route of administration. It will also vary according to the age, weight and response of the individual patient. In general, the daily dose range lie within the range of from about 0.001 mg to about 100 mg per kg body weight of a mammal, preferably 0.01 mg to about 50 mg per kg, and most preferably 0.1 to 10 mg per kg, in single or divided doses. On the other hand, it may be necessary to use dosages outside these limits in some cases.
  • a suitable dosage range is from about 0.001 mg to about 25 mg (preferably from 0.01 mg to about 1 mg) of a compound of Formula I per kg of body weight per day and for cytoprotective use from about 0.1 mg to about 100 mg (preferably from about 1 mg to about 100 mg and more preferably from about 1 mg to about 10 mg) of a compound of Formula I per kg of body weight per day.
  • a suitable dosage range is, e.g.
  • ophthalmic preparations for ocular administration comprising 0.001-1% by weight solutions or suspensions of the compounds of Formula I in an acceptable ophthalmic formulation may be used.
  • compositions which comprises a compound of Formula I and a pharmaceutically acceptable carrier.
  • composition is intended to encompass a product comprising the active ingredient(s), and the inert ingredient(s) (pharmaceutically acceptable excipients) that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients.
  • the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of Formula I, additional active ingredient(s), and pharmaceutically acceptable excipients.
  • any suitable route of administration may be employed for providing a mammal, especially a human with an effective dosage of a compound of the present invention.
  • oral, rectal, topical, parenteral, ocular, pulmonary, nasal, and the like may be employed.
  • Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, aerosols, and the like.
  • the pharmaceutical compositions of the present invention comprise a compound of Formula I as an active ingredient or a pharmaceutically acceptable salt thereof, and may also contain a pharmaceutically acceptable carrier and optionally other therapeutic ingredients.
  • pharmaceutically acceptable salts refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic bases or acids and organic bases or acids.
  • the compounds of the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or nebulisers.
  • the compounds may also be delivered as powders which may be formulated and the powder composition may be inhaled with the aid of an insufflation powder inhaler device.
  • the preferred delivery systems for inhalation are metered dose inhalation (MDI) aerosol, which may be formulated as a suspension or solution of a compound of Formula I in suitable propellants, such as fluorocarbons or hydrocarbons and dry powder inhalation (DPI) aerosol, which may be formulated as a dry powder of a compound of Formula I with or without additional excipients.
  • MDI metered dose inhalation
  • DPI dry powder inhalation
  • Suitable topical formulations of a compound of formula I include transdermal devices, aerosols, creams, ointments, lotions, dusting powders, and the like.
  • the compounds of Formula I can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous).
  • any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, capsules and tablets, with the solid oral preparations being preferred over the liquid preparations. Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques.
  • compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient, as a powder or granules or as a solution or a suspension in an aqueous liquid, a non-aqueous liquid, an oil-in-water emulsion or a water-in-oil liquid emulsion.
  • compositions may be prepared by any of the methods of pharmacy but all methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more necessary ingredients.
  • the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation.
  • a tablet may be prepared by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine, the active ingredient in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent. Desirably, each tablet contains from about 1 mg to about 500 mg of the active ingredient and each cachet or capsule contains from about 1 to about 500 mg of the active ingredient.
  • Compounds of Formula I may be used in combination with other drugs that are used in the treatment/prevention/suppression or amelioration of the diseases or conditions for which compounds of Formula I are useful. Such other drugs may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with a compound of Formula I.
  • a pharmaceutical composition containing such other drugs in addition to the compound of Formula I is preferred.
  • the pharmaceutical compositions of the present invention include those that also contain one or more other active ingredients, in addition to a compound of Formula I.
  • Examples of other active ingredients that may be combined with a compound of Formula I, either administered separately or in the same pharmaceutical compositions, include, but are not limited to: (a) other VLA-4 antagonists such as those described in US 5,510,332, WO97/03094, WO97/02289, WO9640781, W096/22966, WO96/20216,
  • steroids such as beclomethasone, methylprednisolone, betamethasone, prednisone, dexamethasone, and hydrocortisone;
  • immunosuppressants such as cyclosporin, tacrolimus, rapamycin and other FK-506 type immunosuppressants;
  • antihistamines Hl-histamine antagonists
  • the weight ratio of the compound of the Formula I to the second active ingredient may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used. Thus, for example, when a compound of the Formula I is combined with an NSAID the weight ratio of the compound of the Formula I to the NSAID will generally range from about 1000:1 to about 1:1000, preferably about 200:1 to about 1:200. Combinations of a compound of the Formula I and other active ingredients will generally also be within the aforementioned range, but in each case, an effective dose of each active ingredient should be used.
  • Compounds of the present invention may be prepared by procedures illustrated in the accompanying schemes.
  • the next Fmoc- protected amino acid derivative D is coupled to C employing standard peptide (in this instance, 2-(lH-benzotriazol-l-yl)-l, 1,3,3- tetramethyluronium hexafluorophosphate (HBTU), HOBt, and N,N- diisopropylethylamine (DIEA) in DMF) to yield dipeptide E.
  • standard peptide in this instance, 2-(lH-benzotriazol-l-yl)-l, 1,3,3- tetramethyluronium hexafluorophosphate (HBTU), HOBt, and N,N- diisopropylethylamine (DIEA) in DMF
  • the Fmoc group is removed with piperidine in DMF to yield the free amine F.
  • a sulfonyl chloride derivative is reacted with F in the presence of DIEA to yield G.
  • the final product is removed from the resin with strong
  • Step A Loading of N-Fmoc-amino acid derivatives onto resins. N-Fmoc-amino acids were loaded on either Wang®
  • Chloro (2-chorotrityl) resin typically 0.2 mmol
  • a solution of N-Fmoc- amino acid (0.2 mmol) in dimethylformamide (3 ml) was added to the resin, followed by the addition of N,N-diisopropylethylamine(0.4 mmol).
  • the resin was gently stirred for 2 hours, filtered and washed sequentially with dimethylformamide (3 times) and dichloromethane (3 times).
  • the resin was finally washed with 10% methanol in dichloromethane and vacuum dried.
  • the amino acid substitution value obtained after vacuum drying typically ranged between 0.05 to 0.1 mmol.
  • Step B Deprotection of the N-Fmoc group.
  • N-Fmoc protecting group was removed from the resin from Step A by treatment with 20% piperidine in dimethylformamide for 30 minutes. Following filtration, the resin was washed sequentially with dimethylformamide (3 times), dichloromethane (1 time) and dimethylformamide (2 times) and used in the subsequent reaction. Step C. Coupling of the next N-Fmoc-amino acid derivative
  • Step D Deprotection of the N-Fmoc group.
  • the N-Fmoc protecting group was removed from the resin from Step C by the procedure described in Step B and used in the subsequent reaction.
  • Step E Acylation (or sulfonylation) of the terminal amino group.
  • the desired N-terminal capping reagent (sulfonylchloride or acylchloride) (0.4 mol) was dissolved in dimethylformamide (2 ml), mixed with N,N-diisopropylethylamine(0.8 mmol) and added to the resin from Step D. After approximately two hours, the resin was sequentially washed with dimethylformamide (3 times) and dichloromethane (3 times).
  • Step F Cleavage of the desired products from the resins.
  • the final desired products were cleaved from the resins from Step E by gently stirring with a solution of trifluoroacetic acid:thioanisole:ethanedithiol (95:2.5:2.5); 3 hours for Wang® resin and 30 minutes for the Chloro (2-chorotrityl) resin. Following filtration, the solvents were removed by evaporation and the residue dissolved in acetonitrile (3 mL). Insoluble material was removed by filtration. The final products were purified by reverse phase chromatography with a linear gradient of buffer A (0.1% trifluoroacetic acid in water) and buffer B (0.1% trifluoroacetic acid in acetonitrile) and isolated by lyophilization. Molecular ions were obtained by electrospray ionization mass spectrometry or matrix-assisted laser desorption ionization time-of- flight mass spectrometry to confirm the structure of each peptide.
  • Untreated 96 well polystyrene flat bottom plates were coated with bovine serum albumin (BSA; 20 ⁇ g/ml) for 2 hours at room temperature and washed twice with phosphate buffered saline (PBS).
  • BSA bovine serum albumin
  • PBS phosphate buffered saline
  • the albumin coating was next derivatized with 10 ⁇ g/ml 3-(2- pyridyldithio) propionic acid N-hydroxysuccinimide ester (SPDP), a heterobifunctional crosslinker, for 30 minutes at room temperature and washed twice with PBS.
  • SPDP 3-(2- pyridyldithio) propionic acid N-hydroxysuccinimide ester
  • the CS-1 peptide (Cys-Leu-His-Gly-Pro-Glu-Ile- Leu-Asp-Val-Pro-Ser-Thr), which was synthesized by conventional solid phase chemistry and purified by reverse phase HPLC, was next added to the derivatized BSA at a concentration of 2.5 ⁇ g/ml and allowed to react for 2 hours at room temperature. The plates were washed twice with PBS and stored at 4°C.
  • Jurkat cells obtained from the American Type Culture Collection (Rockville, MD; cat # ATCC TIB-152) were grown and maintained in RPMI-1640 culture medium containing 10% fetal calf serum (FCS), 50 units/ml penicillin, 50 ⁇ g/ml streptomycin and 2 mM glutamine. Fluorescence activated cell sorter analysis with specific monoclonal antibodies confirmed that the cells expressed both the ⁇ 4 and ⁇ l chains of VLA-4. The cells were centrifuged at 400xg for five minutes and washed twice with PBS.
  • FCS fetal calf serum
  • the cells were incubated at a concentration of 2 x 10 cells/ml in PBS containing a 1 ⁇ M concentration of a fluorogenic esterase substrate (2', 7'-bis-(2-carboxyethyl)-5-(and -6)- carboxyfluorescein, acetoxymethyl ester; BCECF-AM; Molecular Probes Inc., Eugene, Oregon; catalog #B-1150) for 30-60 minutes at 37°C in a 5% C ⁇ 2/air incubator.
  • the fluorescently labeled Jurkat cells were washed two times in PBS and resuspended in RPMI containing 0.25% BSA at a final concentration of 2.0 x 10 cells/ml.
  • Compounds of this invention were prepared in DMSO at lOOx the desired final assay concentration. Final concentrations were selected from a range between 0.001 nM-100 ⁇ M. Three ⁇ L of diluted compound, or vehicle alone, were premixed with 300 ⁇ L of cell suspension in 96-well polystyrene plates with round bottom wells. 100 ⁇ L aliquots of the cell /compound mixture were then transferred in duplicate to CS-1 coated wells. The cells were next incubated for 30 minutes at room temperature. The non-adherent cells were removed by two gentle washings with PBS.
  • VCAM-Ig The signal peptide as well as domains 1 and 2 of human
  • VCAM (GenBank Accession no. M30257) were amplified by PCR using the human VCAM cDNA (R & D Systems) as template and the following primer sequences: 3'-PCR primer:5'-AATTATAATTTGATCAACTTAC
  • the 5'-PCR primer contained EcoRI and PvuII restriction sites followed by a Kozak consensus sequence (CCACC) proximal to the initiator methionine ATG.
  • the 3'-PCR primer contained a Bell site and a splice donor sequence. PCR was performed for 30 cycles using the following parameters: 1 min. at 94 C, 2 min. at 55 C, and 2 min. at 72 C.
  • the pig-Tail vector contains the genomic fragment which encodes the hinge region, CH2 and CH3 of human IgGl (GenBank Accession no. Z17370).
  • the DNA sequence of the resulting VCAM fragment was verified using Sequenase (US Biochemical, Cleveland, OH).
  • the fragment encoding the entire VCAM-Ig fusion was subsequently excised from pig-Tail with EcoRI and NotI and ligated to pCI-neo (Promega, Madison, WI) digested with EcoRI and NotI.
  • the resulting vector, designated pCI-neo/VCAM-Ig was transfected into CHO-K1 (ATCC CCL 61) cells using calcium-phosphate DNA precipitation (Specialty Media, Lavalette, NJ).
  • VCAM-Ig producing clones were selected according to standard protocols using 0.2-0.8 mg/ml active G418 (Gibco, Grand Island, NY), expanded, and cell supernatants were screened for their ability to mediate Jurkat adhesion to wells previously coated with 1.5 ⁇ g/ml (total protein) goat anti-human IgG (Sigma, St. Louis, MO).
  • a positive CHO-Kl/VCAM-Ig clone was subsequently adapted to CHO-SFM serum-free media (Gibco) and maintained under selection for stable expression of VCAM-Ig.
  • VCAM- Ig was purified from crude culture supernatants by affinity chromatography on Protein A/G Sepharose (Pierce, Rockford, IL) according to the manufacturer's instructions and desalted into 50 mM sodium phosphate buffer, pH 7.6, by ultrafiltration on a YM-30 membrane (Amicon, Beverly, MA).
  • Step 2 Preparation of 1 0 I-VCAM-Ig VCAM-Ig was labeled to a specific radioactivity greater that
  • the labeled protein was separated from unincorporated isotope by means of a calibrated HPLC gel filtration column (G2000SW; 7.5 x 600 mm; Tosoh, Japan) using uv and radiometric detection.
  • Compounds of this invention were prepared in DMSO at lOOx the desired final assay concentration. Final concentrations were selected from a range between 0.001 nM-100 ⁇ M.
  • Jurkat cells were centrifuged at 400xg for five minutes and resuspended in binding buffer (25 M HEPES, 150 M NaCl, 3 mM KC1, 2 mM glucose, 0.1% bovine serum albumin, pH 7.4). The cells were centrifuged again and resuspended in binding buffer supplemented with MnCl 2 at a final concentration of 1 mM.
  • Compounds were assayed in Millipore MHVB multiscreen plates (cat# MHVBN4550, Millipore Corp., MA) by making the following additions to duplicate wells: (i) 200 ⁇ L of binding buffer
  • I- VCAM-Ig in the absence of cells was usually less than 5% of that observed using cells in the presence of vehicle. Percent inhibition was then calculated for each test well and the IC 50 was determined from a ten point titration using a validated four parameter fit algorithm.
  • Step 1 ⁇ 4 ⁇ 7 Cell line.
  • RPMI-8866 cells (a human B cell line ⁇ ⁇ j / ⁇ /; a gift from Prof. John Wilkins, University of Manitoba, Canada) were grown in RPMI/10% fetal calf serum/ 100 U penicillin 100 ⁇ g streptomycin/2 mM L-glutamine at 37°C, 5 % carbon dioxide. The cells were pelleted at 1000 rpm for 5 minutes and then washed twice and resuspended in binding buffer (25 M Hepes, 150 mM NaCl , 0.1 % BSA, 3 mM KC1, 2 mM Glucose, pH 7.4).
  • binding buffer 25 M Hepes, 150 mM NaCl , 0.1 % BSA, 3 mM KC1, 2 mM Glucose, pH 7.4
  • Compounds of this invention were prepared in DMSO at lOOx the desired final assay concentration. Final concentrations were selected from a range between 0.001 nM-100 ⁇ M. Compounds were assayed in Millipore MHVB multiscreen plates (Cat# MHVBN4550) by making the following sequential additions to duplicate wells: (i) 100 ⁇ l well of binding buffer containing 1.5 mM MnCl 2 ; (ii) 10 ⁇ l/well 125 I- VCAM-Ig in binding buffer (final assay concentration ⁇ 500 pM); (iii) 1.5 ⁇ l/well test compound or DMSO alone; (iv) 38 ⁇ l/well RPMI-8866 cell suspension (1.25 x 10 6 cells/well).
  • the plates were incubated at room temperature for 45 minutes on a plate shaker at 200 rpm, filtered on a vacuum box, and washed on the same apparatus by the addition of 100 ⁇ L of binding buffer containing 1 mM MnCl 2 .
  • 100 ⁇ L of Microscint-20 (Packard cat# 6013621) was added to each well.
  • the plates were then sealed, placed on a shaker for 30 seconds, and counted on a Topcount microplate scintillation counter (Packard).
  • Control wells containing DMSO alone were used to determine the level of VCAM-Ig binding corresponding to 0% inhibition.
  • Wells in which cells were omitted were used to determine the level of binding corresponding to 100% inhibition. Percent inhibition was then calculated for each test well and the IC 50 was determined from a ten point titration using a validated four parameter fit algorithm.

Abstract

Compounds of formula (I) are antagonists of VLA-4 and/or α4-β7, and as such are useful in the inhibition or prevention of cell adhesion and cell-adhesion mediated pathologies. These compounds may be formulated into pharmaceutical compositions and are suitable for use in the treatment of asthma, allergies, inflammation, multiple sclerosis, and other inflammatory and autoimmune disorders.

Description

TITLE OF THE INVENTION
SULFONAMIDES AS CELL ADHESION INHIBITORS
SUMMARY OF THE INVENTION The compounds of the present invention are antagonists of the VLA-4 integrin ("very late antigen- 4"; CD49d/CD29; or c-4βl) and/or the α4β7 integrin (LPAM-1 and α4βp), thereby blocking the binding of
VLA-4 to its various ligands, such as VCAM-1 and regions of fibronectin and/or α4β7 to its various ligands, such as MadCAM-1, VCAM-1 and fibronectin. Thus, these antagonists are useful in inhibiting cell adhesion processes including cell activation, migration, proliferation and differentiation. These antagonists are useful in the treatment, prevention and suppression of diseases mediated by VLA-4 and/or α4β7 binding and cell adhesion and activation, such as multiple sclerosis, asthma, allergic rhinitis, allergic conjunctivitis, inflammatory lung diseases, rheumatoid arthritis, septic arthritis, type I diabetes, organ transplantation, restenosis, autologous bone marrow transplantation, inflammatory sequelae of viral infections, myocarditis, inflammatory bowel disease including ulcerative colitis and Crohn 's disease, certain types of toxic and immune-based nephritis, contact dermal hypersensitivity, psoriasis, tumor metastasis, and atherosclerosis.
BACKGROUND OF THE INVENTION
The present invention relates to sulfonamide derivatives which are useful for the inhibition and prevention of leukocyte adhesion and leukocyte adhesion-mediated pathologies. This invention also relates to compositions containing such compounds and methods of treatment using such compounds.
Many physiological processes require that cells come into close contact with other cells and/or extracellular matrix. Such adhesion events may be required for cell activation, migration, proliferation and differentiation. Cell-cell and cell-matrix interactions are mediated through several families of cell adhesion molecules (CAMs) including the selectins, integrins, cadherins and immunoglobulins. CAMs play an essential role in both normal and pathophysiological processes. Therefore, the targetting of specific and relevant CAMs in certain disease conditions without interfering with normal cellular functions is essential for an effective and safe therapeutic agent that inhibits cell-cell and cell-matrix interactions. The integrin superfamily is made up of structurally and functionally related glycoproteins consisting of α and β heterodimeric, transmembrane receptor molecules found in various combinations on nearly every mammalian cell type, (for reviews see: E. C. Butcher, Cell, 61, 1033 (1991); T. A. Springer, Cell, 76, 301 (1994); D. Cox et al., "The Pharmacology of the Integrins." Medicinal Research Rev, 14. 195 (1994) and V, W. Engleman et al., "Cell Adhesion Integrins as Pharmaceutical Targets." in Ann. Repts. in Medicinal Chemistry. Vol. 31, J. A. Bristol, Ed.; Acad. Press, NY, 1996, p. 191).
VLA-4 ("very late antigen-4"; CD49d/CD29; or α-iβl) is an integrin expressed on all leukocytes, except platelets and mature neutrophils, including dendritic cells and macrophage-like cells and is a key mediator of the cell-cell and cell-matrix interactions of of these cell types (see M. E. Hemler, "VLA Proteins in the Integrin Family: Structures, Functions, and Their Role on Leukocytes." Ann. Rev. Immunol. 8, 365 (1990)). The ligands for VLA-4 include vascular cell adhesion molecule-1 (VCAM-1) and the CS-1 domain of fibronectin (FN). VCAM-1 is a member of the Ig superfamily and is expressed in vivo on endothelial cells at sites of inflammation. (See R. Lobb et al. "Vascular Cell Adhesion Molecule 1." in Cellular and Molecular Mechanisms of Inflammation, C. G. Cochrane and M. A. Gimbrone, Eds.; Acad. Press, San Diego, 1993, p. 151.) VCAM-1 is produced by vascular endothelial cells in response to pro-inflammatory cytokines (See A. J. H. Gearing and W. Newman, "Circulating adhesion molecules in disease.",
Immunol. Today. 14, 506 (1993). The CS-1 domain is a 25 amino acid sequence that arises by alternative splicing within a region of fibronectin. (For a review, see R. O. Hynes "Fibronectins.", Springer- Velag, NY, 1990.) A role for VLA-4/CS-1 interactions in inflammatory conditions has been proposed (see M. J. Elices, "The integrin c<4βι (VLA-
4) as a therapeutic target" in Cell Adhesion and Human Disease. Ciba Found. Symp., John Wiley & Sons, NY, 1995, p. 79). α4β7 (also referred to as LPAM-1 and α4βp) is an integrin expressed on leukocytes and is a key mediator of leukocyte trafficking and homing in the gastrointestinal tract (see C. M. Parker et al., Proc. Natl. Acad. Sci. USA, rø, 1924 (1992)). The ligands for α4β7 include mucosal addressing cell adhesion molecule-1 (MadCAM-1) and, upon activation of α4β7, VCAM-1 and fibronectin (Fn). MadCAM-1 is a member of the Ig superfamily and is expressed in vivo on endothelial cells of gut-associated mucosal tissues of the small and large intestine ("Peyer 's Patches") and lactating mammary glands. (See M. J. Briskin et al., Nature. 363. 461 (1993); A. Hamann et al., J. Immunol.. 152.3282 (1994)). MadCAM-1 can be induced in vitro by proinflammatory stimuli (See E. E. Sikorski et al. J. Immunol.. 151. 5239 (1993)). MadCAM-1 is selectively expressed at sites of lymphocyte extravasation and specifically binds to the integrin, o-4β7.
Neutralizing anti-0.4 antibodies or blocking peptides that inhibit the interaction between VLA-4 and/or α4β7 and their ligands have proven efficacious both prophylactically and therapeutically in several animal models of disease, including i) experimental allergic encephalomyelitis, a model of neuronal demyelination resembling multiple sclerosis (for example, see T. Yednock et al., "Prevention of experimental autoimmune encephalomyelitis by antibodies against 0.4 βi integrin." Nature. 356, (_3 (1993) and E. Keszthelyi et al., "Evidence for a prolonged role of 0.4 integrin throughout active experimental allergic encephalomyelitis." Neurology. 47, 1053 (1996)); ii) bronchial hyperresponsiveness in sheep and guinea pigs as models for the various phases of asthma (for example, see W. M. Abraham et al., "0.4-Integrins mediate antigen-induced late bronchial responses and prolonged airway hyperresponsiveness in sheep." J. Clin. Invest. 93. 776 (1993) and A. A. Y. Milne and P. P. Piper, "Role of VLA-4 integrin in leucocyte recruitment and bronchial hyperresponsiveness in the gunea-pig." Eur. J. Pharmacol.. 282. 243 (1995)); iii) adjuvant-induced arthritis in rats as a model of inflammatory arthritis (see C. Barbadillo et al., "Anti-VLA-4 mAb prevents adjuvant arthritis in Lewis rats." Arthr. Rheuma. (Suppl.), 36 95 (1993) and D. Seiffge, "Protective effects of monoclonal antibody to VLA-4 on leukocyte adhesion and course of disease in adjuvant arthritis in rats." J. Rheumatol.. 23, 12 (1996)); iv) adoptive autoimmune diabetes in the NOD mouse (see J. L. Baron et al., "The pathogenesis of adoptive murine autoimmune diabetes requires an interaction between 0.4 -integrins and vascular cell adhesion molecule-
1.", J. Clin. Invest.. 93, 1700 (1994), A. Jakubowski et al., "Vascular cell adhesion molecule-Ig fusion protein selectively targets activated α4- integrin receptors in vivo: Inhibition of autoimmune diabetes in an adoptive transfer model in nonobese diabetic mice." J. Immunol.. 155. 938 (1995), and X. D. Yang et al., "Involvement of beta 7 integrin and mucosal addressin cell adhesion molecule-1 (MadCAM-1) in the development of diabetes in nonobese diabetic mice", Diabetes, 46, 1542 (1997)); v) cardiac allograft survival in mice as a model of organ transplantation (see M. Isobe et al., "Effect of anti-VCAM-1 and anti- VLA-4 monoclonal antibodies on cardiac allograft survival and response to soluble antigens in mice.", Tranplant. Proc. 26, 867 (1994) and S. Molossi et al., "Blockade of very late antigen-4 integrin binding to fibronectin with connecting segment- 1 peptide reduces accelerated coronary arteripathy in rabbit cardiac allografts." J. Clin Invest.. 95, 2601 (1995)); vi) spontaneous chronic colitis in cotton-top tamarins which resembles human ulcerative colitis, a form of inflammatory bowel disease (see D. K. Podolsky et al., "Attenuation of colitis in the Cotton-top tamarin by anti-c.4 integrin monoclonal antibody.", J. Clin. Invest.. 92,
372 (1993)); vii) contact hypersensitivity models as a model for skin allergic reactions (see T. A. Ferguson and T. S. Kupper, "Antigen- independent processes in antigen-specific immunity.", J. Immunol., 150. 1172 (1993) and P. L. Chisholm et al., "Monoclonal antibodies to the integrin α-4 subunit inhibit the murine contact hypersensitivity response." Eur. J. Immunol.. 23, 682 (1993)); viii) acute neurotoxic nephritis (see M. S. Mulligan et al., "Requirements for leukocyte adhesion molecules in nephrotoxic nephritis.", J. Clin. Invest.. 91, 577 (1993)); ix) tumor metastasis (for examples, see M. Edward, "Integrins and other adhesion molecules involved in melanocytic tumor progression.", Curr. Opin. Oncol.. __, 185 (1995)); x) experimental autoimmune thyroiditis (see R. W. McMurray et al., "The role of α4 integrin and intercellular adhesion molecule-1 (ICAM-1) in murine experimental autoimmune thyroiditis." Autoimmunity. 23. 9 (1996); and xi) ischemic tissue damage following arterial occlusion in rats (see F. Squadrito et al., "Leukocyte integrin very late antigen-4/vascular cell adhesion molecule- 1 adhesion pathway in splanchnic artery occlusion shock." Eur. J. Pharmacol.. 318. 153 (1996; xii) inhibition of TH2 T-cell cytokine production including IL-4 and IL-5 by VLA-4 antibodies which would attenuate allergic responses (J. Clinical Investigation 1O0, 3083 (1997). The primary mechanism of action of such antibodies appears to be the inhibition of lymphocyte and monocyte interactions with CAMs associated with components of the extracellular matrix, thereby limiting leukocyte migration to extravascular sites of injury or inflammation and/or limiting the priming and/or activation of leukocytes.
There is additional evidence supporting a possible role for VLA-4 interactions in other diseases, including rheumatoid arthritis; various melanomas, carcinomas, and sarcomas; inflammatory lung disorders; acute respiratory distress syndrome (ARDS); atherosclerotic plaque formation; restenosis; uveitis and circulatory shock (for examples, see A. A. Postigo et al., "The α4βι/VCAM-l adhesion pathway in physiology and disease.", Res. Immunol.. 144. 723 (1994) and J.-X. Gao and A. C. Issekutz, "Expression of VCAM-1 and VLA-4 dependent T-lymphocyte adhesion to dermal fibroblasts stimulated with proinflammatory cytokines." Immunol. 89, 375 (1996)).
At present, there is a humanized monoclonal antibody (Antegren® Athena Neurosciences/Elan ) against VLA-4 in clinical development for the treatment of "flares" associated with multiple sclerosis and a humanized monoclonal antibody (ACT-l®/LDP-02 LeukoSite) against 0C4β7 in clinical development for the treatment of inflammatory bowel disease. Several peptidyl antagonists of VLA-4 have been described (D. Y. Jackson et al., "Potent o-4βl peptide antagonists as potential anti-inflammatory agents", J. Med. Chem.. 40, 3359 (1997); H. N. Shroff et al., "Small peptide inhibitors of α4β7 mediated MadCAM-1 adhesion to lymphocytes", Bioorpr. Med. Chem. Lett.. 6, 2495 (1996); US 5,510,332, WO97/03094, WO97/02289, WO96/40781, WO96/22966, WO96/20216, WO96/01644, WO96/06108, W095/15973). There is one report of nonpeptidyl inhibitors of the ligands for 0.4-integrins (WO96/31206).
There still remains a need for low molecular weight, specific inhibitors of VLA-4- and α4β7-dependent cell adhesion that have improved pharmacokinetic and pharmacodynamic properties such as oral bioavailability and significant duration of action. Such compounds would prove to be useful for the treatment, prevention or suppression of various pathologies mediated by VLA-4 and α4β7 binding and cell adhesion and activation.
DETAILED DESCRIPTION OF THE INVENTION One aspect of the present invention provides a method for the treatment of diseases, disorders, conditions or symptoms mediated by cell adhesion in a mammal which comprises administering to said mammal an effective amount of a compound Formula I:
Figure imgf000008_0001
I
or a pharmaceutically acceptable salt thereof wherein: Rl is 1) Ci-ioalkyl,
2) C2-10alkenyl, 3) C2-10alkynyl,
4) Cy, 5) Cy-Ci-ioalkyl,
6) Cy-C2-10alkenyl,
7) Cy-C2-10alkynyl, wherein alkyl, alkenyl, and alkynyl are optionally substituted with one to four substituents independently selected from Ra; and Cy is optionally substituted with one to four substituents independently selected from RD;
R2 and R3 are independently selected from
1) hydrogen, or 2) a group selected from Rl;
R is 1) hydrogen,
2) Ci.ioalkyl,
3) C2_loalkenyl, 4) C2-loalkynyl,
5) aryl,
6) aryl-Cι_ιoalkyl,
7) heteroaryl,
8) heteroaryl-Ci_ioalkyl, wherein alkyl, alkenyl, and alkynyl are optionally substituted with one to four substituents independently selected from Ra; and aryl and heteroaryl optionally substituted with one to four substituents independently selected from R^; or
R3, R4 and the carbon to which they are attached form a 3-7 membered mono- or bicyclic ring containing 0-2 heteroatoms selected from N, O and S;
B is 1) hydrogen, 2) Ci-io alkyl,
3) Cy, or
4) Cy-Ci-io alkyl, wherein alkyl is optionally substituted with one to four substituents independently selected from Ra; and Cy is optionally substituted with one to four substituents independently selected from RD;
R is 1) hydrogen,
2) Cl-ioalkyl,
3) C2-10alkenyl,
4) C2-10alkynyl,
5) Cy, 6) Cy-Ci-ioalkyl,
7) Cy-C2-10alkenyl,
8) Cy-C2-lθalkynyl, wherein alkyl, alkenyl and alkynyl are optionally substituted with one to four substituents selected from Rχ, and Cy is optionally substituted with one to four substituents independently selected from RY;
R7 is 1) hydrogen,
2) Ci.ioalkyl,
3) C2-loalkenyl, 4) C2-loalkynyl,
5) aryl,
6) aryl-Cι_ιoalkyl,
7) heteroaryl,
8) heteroaryl-Cι_ oalkyl, wherein alkyl, alkenyl and alkynyl are optionally substituted with one to four substituents selected from Rx, and aryl and heteroaryl are optionally substituted with one to four substituents independently selected from R ; or
R6, R7 together with the atoms to which they are attached form a 3-7 membered ring containing 0-2 heteroatoms selected from N, O and S;
Ra is 1) Cy, or 2) a group selected from Rx; wherein Cy is optionally substituted with one to four substituents independently selected from Rc;
Rk is 1) a group selected from Ra,
2) Ci-io alkyl,
3) C2-10 alkenyl,
4) C2-10 alkynyl,
5) aryl Ci-ioalkyl, 6) heteroaryl Ci-io alkyl, wherein alkyl, alkenyl, alkynyl, aryl, heteroaryl are optionally substituted with a group independently selected from Rc;
Rc is 1) halogen,
2) amino,
3) carboxy,
4) Ci-4alkyl,
5) Ci-4alkoxy,
6) aryl,
7) aryl Ci-4alkyl, or
8) aryloxy;
Rd and Re are independently selected from hydrogen, Ci-ioalkyl, C2-10alkenyl, C2-10alkynyl, Cy and Cy Ci-ioalkyl, wherein alkyl, alkenyl, alkynyl and Cy is optionally substituted with one to four substituents independently selected from Rc; or R" and Re together with the atoms to which they are attached form a heterocyclic ring of 5 to 7 members containing 0-2 additional heteroatoms independently selected from oxygen, sulfur and nitrogen;
W and Rg are independently selected from hydrogen, Ci-ioalkyl, Cy and Cy Ci-ioalkyl; or R* and RS together with the carbon to which they are attached form a ring of 5 to 7 members containing 0-2 heteroatoms independently selected from oxygen, sulfur and nitrogen;
Rn is 1) hydrogen,
2) Ci-ioalkyl,
3) C2-10alkenyl,
4) C2-10alkynyl,
5) cyano,
6) aryl,
7) aryl Ci-ioalkyl,
8) heteroaryl,
9) heteroaryl Ci-ioalkyl, or
10) -SO2R1; wherein alkyl, alkenyl, and alkynyl are optionally substituted with one to four substituents independently selected from Ra; and aryl and heteroaryl are each optionally substituted with one to four substituents independently selected from RD;
Ri is 1) Ci-ioalkyl,
2) C2-10alkenyl,
3) C2-10alkynyl, or
4) aryl; wherein alkyl, alkenyl, alkynyl and aryl are each optionally substituted with one to four substituents independently selected from Rc;
RX is 1) -ORd,
2) -NO2,
3) halogen
4) -S(O)mRd
5) -SRd,
6) -S(0)2ORd,
7) -S(0)mNRdRe) 8) -NRdRe,
9) -O(CRfRg)nNRdRe,
10) -C(0)Rd?
11) -CO2Rd,
12) -CO2(CRfRg)nCONRdRe,
13) -OC(O)Rd,
14) -CN,
15) -C(O)NRdRe,
16) -NRdC(O)Re,
17) -OC(O)NRdRe,
18) -NRdC(O)ORe,
19) -NR<*C(O)NRdRe.
20) -CRd(N-ORe), or
21) -CF3;
Ry -s 1) a group selected from Rx,
2) Ci-io alkyl,
3) C2-10 alkenyl,
4) C2-IO alkynyl,
5) aryl Ci-ioalkyl,
6) heteroaryl Ci-io alkyl,
7) cycloalkyl,
8) heterocyclyl; wherein alkyl, alkenyl, alkynyl and aryl are each optionally substituted with one to four substituents independently selected from Rx;
Cy is cycloalkyl, heterocyclyl, aryl, or heteroaryl;
m is an integer from 1 to 2;
n is an integer from 1 to 10;
X is 1) -C(O)ORd,
2) -P(0)2ORd 3) -S(0)mORd,
4) -C(O)NRdRh, or
5) -5-tetrazolyl.
In one embodiment of the present method compounds of formula I are those wherein R^ is Cy. For the purpose of Rl Cy is preferably aryl optionally substituted with one to four substituents selected from RD. More preferred R* is phenyl with a substituent on the
3-position and optionally a second substituent;the more preferred substituents are selected from Cι_ιoalkoxy, halogen, cyano, and trifluoromethyl.
In another embodiment of the present method compounds of Formula I are those wherein R^ is H or Ci-6alkyl. Preferred R^ is H or Ci-3alkyl, more preferably H or methyl.
In another embodiment of the present method compounds of Formula I are those wherein one of R3 and R4 is other than hydrogen.
In a preferred embodiment one of R3 and R4 is hydrogen and the other is Cy, Cy-C]-.ιoalkyl, or C-j_.iQa_ls.yl, each of which is optionally substituted as provided under Formula I. For the purpose of R3/R4 Cy is preferably aryl or heteroaryl each optionally substituted with one to four substituents selected from RD; and alkyl is optionally substituted with one to four substituents independently selected from Ra. More preferred R /R is selected from phenyl; Cy-Cι_3alkyl wherein Cy is thienyl, pyridyl or phenyl optionally substituted with one or two groups selected from phenyl, hydroxy, halogen, cyano, and nitro; and C^.galkyl optionally substituted with CO2H or CO H2.
Yet in another embodiment of the present method compounds of Formula I are those wherein one of R6 and R? is other than hydrogen. In a preferred embodiment one of Rβand R? is hydrogen and the other is aryl, aryl-Ci-lθalkyl, or Ci-ioalkyl, wherein aryl is optionally substituted with one to four substituents independently selected from Ry, and alkyl is optionally substituted with one to four substituents selected from Rx. More preferred R6 / R7 is selected from aryl-C ι_3alkyl wherein aryl is phenyl or naphthyl each optionally substituted with one or two groups selected from Ey.
Yet in another embodiment of the present method compounds of Formula I are those wherein X is -C(O)ORd.
In a more preferred embodiment, the cell adhesion is mediated by VLA-4, and the compounds are of Formula la as described hereinbelow.
In another aspect the present invention provides compounds of formula la:
Figure imgf000015_0001
la
or a pharmaceutically acceptable salt thereof, wherein
R2 is 1) hydrogen, or
2) Ci-βalk l; one of R and R4 is hydrogen and the other is 1) aryl, 2) heteroaryl,
3) aryl-Cι_ιoalkyl,
4) heteroaryl-Cι-ιoalkyl, or
5) Ci.ioalkyl, wherein aryl and heteroaryl are each optionally substituted with one to four substituents selected from R"; and alkyl is optionally substituted with one to four substituents independently selected from Ra; or R3, R4 and the carbon to which they are attached form a 3-7 membered mono- or bicyclic ring containing 0-2 heteroatoms selected from N, O and S; one of R6 and R^ is hydrogen and the other is
1) aryl,
2) aryl-Cι_ιoalkyl, or
3) Ci.ioalkyl, wherein aryl is optionally substituted with one to four substituents independently selected from Ry, and alkyl is optionally substituted with one to four substituents selected from Rx; and
Ra, RD, Rx and Ry are as defined above under Formula I.
In a more preferred embodiment of compounds of Formula la, Rb is selected from Ci-ioalkoxy, halogen, cyano, and trifluoromethyl;
R2 is H or methyl; one of R3 and R4 is hydrogen and the other is phenyl, Cy-Cι_3alkyl (wherein Cy is thienyl, pyridyl or phenyl optionally substiuted with one or two groups selected from phenyl, hydroxy, halogen, cyano, and nitro); and Ci. alkyl optionally substituted with CO2H or CONH2; one of R6 and R? is hydrogen and the other is aryl-Cι_
2alkyl, wherein aryl is optionally substituted with one to two substituents independently selected from halogen; and Ry is as defined above under
Formula I.
The present compounds are generally composed of three domains: 1) a sulfonyl moiety, 2) amino acid 1, and 3) amino acid 2, and are named in a manner similar to that used to name oligopeptides.
Representative names used herein and their corresponding structures are shown below (without the stereochemistry) to illustrate the nomenclature used in the application. N-(3,4-dimethoxybenzenesulfonyl)-(L)-3-(2-thienyl)alanyl-(L)-norleucine
Figure imgf000016_0001
N-(3,5-dichlorobenzenesulfonyl)-(L)-N-methylvalyl-(L)-2- naphthylalanine
Figure imgf000017_0001
N-(3-chlorobenzenesulfonyl)-2-amino-indan-2-carbonyl-(L)-4- fluorophenylalanine
Figure imgf000017_0002
"Alkyl", as well as other groups having the prefix "alk", such as alkoxy, alkanoyl, means carbon chains which may be linear or branched or combinations thereof. Examples of alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec- and tert-butyl, pentyl, hexyl, heptyl, octyl, nonyl, and the like.
"Alkenyl" means carbon chains which contain at least one carbon-carbon double bond, and which may be linear or branched or combinations thereof. Examples of alkenyl include vinyl, allyl, isopropenyl, pentenyl, hexenyl, heptenyl, 1-propenyl, 2-butenyl, 2- methyl-2-butenyl, and the like.
"Alkynyl" means carbon chains which contain at least one carbon-carbon triple bond, and which may be linear or branched or combinations thereof. Examples of alkynyl include ethynyl, propargyl, 3-m ethyl- 1-pentynyl, 2-heptynyl and the like. "Cycloalkyl" means mono- or bicyclic saturated carbocyclic rings, each of which having from 3 to 10 carbon atoms. The term also includes monocyclic rings fused to an aryl group in which the point of attachment is on the non-aromatic portion. Examples of cycloalkyl include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, tetrahydronaphthyl, decahydronaphthyl, indanyl, and the like.
"Aryl" means mono- or bicyclic aromatic rings containing only carbon atoms. The term also includes aryl group fused to a monocyclic cycloalkyl or monocyclic heterocyclyl group in which the point of attachment is on the aromatic portion. Examples of aryl include phenyl, naphthyl, indanyl, indenyl, tetrahydronaphthyl, 2,3- dihydrobenzofuranyl, benzopyranyl, 1,4-benzodioxanyl, and the like.
"Heteroaryl" means a mono- or bicyclic aromatic ring containing at least one heteroatom selected from N, O and S, with each ring containing 5 to 6 atoms. Examples of heteroaryl include pyrrolyl, isoxazolyl, isothiazolyl, pyrazolyl, pyridyl, oxazolyl, oxadiazolyl, thiadiazolyl, thiazolyl, imidazolyl, triazolyl, tetrazolyl, furanyl, triazinyl, thienyl, pyrimidyl, pyridazinyl, pyrazinyl, benzoxazolyl, benzothiazolyl, benzimidazolyl, benzofuranyl, benzothiophenyl, furo(2,3-b)pyridyl, quinolyl, indolyl, isoquinolyl, and the like.
"Heterocyclyl" means mono- or bicyclic saturated rings containing at least one heteroatom selected from N, S and O, each of said ring having from 3 to 10 atoms in which the point of attachment may be carbon or nitrogen. The term also includes monocyclic heterocycle fused to an aryl or heteroaryl group in which the point of attachment is on the non-aromatic portion. Examples of "heterocyclyl" include pyrrolidinyl, piperidinyl, piperazinyl, imidazolidinyl, 2,3-dihydrofuro(2,3-b)pyridyl, benzoxazinyl, tetrahydrohydroquinolinyl, tetrahydroisoquinolinyl, dihydroindolyl, and the like. The term also includes partially unsaturated monocyclic rings that are not aromatic, such as 2- or 4- pyridones attached through the nitrogen or N-substituted-(lH,3H)- pyrimidine-2,4-diones (N-substituted uracils).
"Halogen" includes fluorine, chlorine, bromine and iodine. Optical Isomers - Diastereomers - Geometric Isomers - Tautomers
Compounds of Formula I contain one or more asymmetric centers and can thus occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. The present invention is meant to comprehend all such isomeric forms of the compounds of Formula I.
Some of the compounds described herein contain olefinic double bonds, and unless specified otherwise, are meant to include both E and Z geometric isomers.
Some of the compounds described herein may exist with different points of attachment of hydrogen, referred to as tautomers. Such an example may be a ketone and its enol form known as keto-enol tautomers. The individual tautomers as well as mixture thereof are encompassed with compounds of Formula I.
Compounds of the Formula I may be separated into diastereoisomeric pairs of enantiomers by, for example, fractional crystallization from a suitable solvent, for example methanol or ethyl acetate or a mixture thereof. The pair of enantiomers thus obtained may be separated into individual stereoisomers by conventional means, for example by the use of an optically active acid as a resolving agent.
Alternatively, any enantiomer of a compound of the general Formula I or la may be obtained by stereospecific synthesis using optically pure starting materials or reagents of known configuration.
Salts
The term "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids. Salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particularly preferred are the ammonium, calcium, magnesium, potassium, and sodium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N -dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl-morpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
When the compound of the present invention is basic, salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids. Such acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p- toluenesulfonic acid, and the like. Particularly preferred are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, and tartaric acids.
It will be understood that, as used herein, references to the compounds of Formula I are meant to also include the pharmaceutically acceptable salts.
Utilities
The ability of the compounds of Formula I to antagonize the actions of VLA-4 and/or α4β7 integrin makes them useful for preventing or reversing the symptoms, disorders or diseases induced by the binding of VLA-4 and or α4β7to their various respective ligands. Thus, these antagonists will inhibit cell adhesion processes including cell activation, migration, proliferation and differentiation. Accordingly, another aspect of the present invention provides a method for the treatment (including prevention, alleviation, amelioration or suppression) of diseases or disorders or symptoms mediated by VLA-4 and or α4β7 binding and cell adhesion and activation, which comprises administering to a mammal an effective amount of a compound of Formula I. Such diseases, disorders, conditions or symptoms are for example (1) multiple sclerosis, (2) asthma, (3) allergic rhinitis, (4) allergic conjunctivitis, (5) inflammatory lung diseases, (6) rheumatoid arthritis, (7) septic arthritis, (8) type I diabetes, (9) organ transplantation rejection, (10) restenosis, (11) autologous bone marrow transplantation, (12) inflammatory sequelae of viral infections, (13) myocarditis, (14) inflammatory bowel disease including ulcerative colitis and Crohn 's disease, (15) certain types of toxic and immune-based nephritis, (16) contact dermal hypersensitivity, (17) psoriasis, (18) tumor metastasis, and (19) atherosclerosis.
Dose Ranges
The magnitude of prophylactic or therapeutic dose of a compound of Formula I will, of course, vary with the nature of the severity of the condition to be treated and with the particular compound of Formula I and its route of administration. It will also vary according to the age, weight and response of the individual patient. In general, the daily dose range lie within the range of from about 0.001 mg to about 100 mg per kg body weight of a mammal, preferably 0.01 mg to about 50 mg per kg, and most preferably 0.1 to 10 mg per kg, in single or divided doses. On the other hand, it may be necessary to use dosages outside these limits in some cases.
For use where a composition for intravenous administration is employed, a suitable dosage range is from about 0.001 mg to about 25 mg (preferably from 0.01 mg to about 1 mg) of a compound of Formula I per kg of body weight per day and for cytoprotective use from about 0.1 mg to about 100 mg (preferably from about 1 mg to about 100 mg and more preferably from about 1 mg to about 10 mg) of a compound of Formula I per kg of body weight per day. In the case where an oral composition is employed, a suitable dosage range is, e.g. from about 0.01 mg to about 100 mg of a compound of Formula I per kg of body weight per day, preferably from about 0.1 mg to about 10 mg per kg and for cytoprotective use from 0.1 mg to about 100 mg (preferably from about 1 mg to about 100 mg and more preferably from about 10 mg to about 100 mg) of a compound of Formula I per kg of body weight per day.
For the treatment of diseases of the eye, ophthalmic preparations for ocular administration comprising 0.001-1% by weight solutions or suspensions of the compounds of Formula I in an acceptable ophthalmic formulation may be used.
Pharmaceutical Compositions
Another aspect of the present invention provides pharmaceutical compositions which comprises a compound of Formula I and a pharmaceutically acceptable carrier. The term "composition", as in pharmaceutical composition, is intended to encompass a product comprising the active ingredient(s), and the inert ingredient(s) (pharmaceutically acceptable excipients) that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of Formula I, additional active ingredient(s), and pharmaceutically acceptable excipients.
Any suitable route of administration may be employed for providing a mammal, especially a human with an effective dosage of a compound of the present invention. For example, oral, rectal, topical, parenteral, ocular, pulmonary, nasal, and the like may be employed. Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, aerosols, and the like. The pharmaceutical compositions of the present invention comprise a compound of Formula I as an active ingredient or a pharmaceutically acceptable salt thereof, and may also contain a pharmaceutically acceptable carrier and optionally other therapeutic ingredients. The term "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic bases or acids and organic bases or acids. The compositions include compositions suitable for oral, rectal, topical, parenteral (including subcutaneous, intramuscular, and intravenous), ocular (ophthalmic), pulmonary (aerosol inhalation), or nasal administration, although the most suitable route in any given case will depend on the nature and severity of the conditions being treated and on the nature of the active ingredient. They may be conveniently presented in unit dosage form and prepared by any of the methods well- known in the art of pharmacy.
For administration by inhalation, the compounds of the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or nebulisers. The compounds may also be delivered as powders which may be formulated and the powder composition may be inhaled with the aid of an insufflation powder inhaler device. The preferred delivery systems for inhalation are metered dose inhalation (MDI) aerosol, which may be formulated as a suspension or solution of a compound of Formula I in suitable propellants, such as fluorocarbons or hydrocarbons and dry powder inhalation (DPI) aerosol, which may be formulated as a dry powder of a compound of Formula I with or without additional excipients.
Suitable topical formulations of a compound of formula I include transdermal devices, aerosols, creams, ointments, lotions, dusting powders, and the like.
In practical use, the compounds of Formula I can be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous). In preparing the compositions for oral dosage form, any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as, for example, powders, capsules and tablets, with the solid oral preparations being preferred over the liquid preparations. Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be coated by standard aqueous or nonaqueous techniques.
In addition to the common dosage forms set out above, the compounds of Formula I may also be administered by controlled release means and/or delivery devices such as those described in U.S. Patent Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123; 3,630,200 and 4,008,719. Pharmaceutical compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient, as a powder or granules or as a solution or a suspension in an aqueous liquid, a non-aqueous liquid, an oil-in-water emulsion or a water-in-oil liquid emulsion. Such compositions may be prepared by any of the methods of pharmacy but all methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more necessary ingredients. In general, the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation. For example, a tablet may be prepared by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine, the active ingredient in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent. Desirably, each tablet contains from about 1 mg to about 500 mg of the active ingredient and each cachet or capsule contains from about 1 to about 500 mg of the active ingredient.
The following are examples of representative pharmaceutical dosage forms for the compounds of Formula I:
Injectable Suspension (I.M.) m /mL
Compound of Formula I 10
Methylcellulose 5.0
Tween 80 0.5
Benzyl alcohol 9.0
Benzalkonium chloride 1.0
Water for injection to a total volume of 1 mL
Tablet mg/tablet
Compound of Formula I 25
Microcrystalline Cellulose 415
Povidone 14.0
Pregelatinized Starch 43.5
Magnesium Stearate 2-5
500
Capsule m /capsule
Compound of Formula I 25
Lactose Powder 573.5
Magnesium Stearate 1-5
600 Aerosol Per canister
Compound of Formula I 24 mg
Lecithin, NF Liquid Concentrate 1.2 mg Trichlorofluoromethane, NF 4.025 g
Dichlorodifluoromethane, NF 12.15 g
Combination Therapy
Compounds of Formula I may be used in combination with other drugs that are used in the treatment/prevention/suppression or amelioration of the diseases or conditions for which compounds of Formula I are useful. Such other drugs may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with a compound of Formula I. When a compound of Formula I is used contemporaneously with one or more other drugs, a pharmaceutical composition containing such other drugs in addition to the compound of Formula I is preferred. Accordingly, the pharmaceutical compositions of the present invention include those that also contain one or more other active ingredients, in addition to a compound of Formula I. Examples of other active ingredients that may be combined with a compound of Formula I, either administered separately or in the same pharmaceutical compositions, include, but are not limited to: (a) other VLA-4 antagonists such as those described in US 5,510,332, WO97/03094, WO97/02289, WO9640781, W096/22966, WO96/20216,
WO96/01644, WO96/06108, WO95/15973 and WO96/31206; (b) steroids such as beclomethasone, methylprednisolone, betamethasone, prednisone, dexamethasone, and hydrocortisone; (c) immunosuppressants such as cyclosporin, tacrolimus, rapamycin and other FK-506 type immunosuppressants; (d) antihistamines (Hl-histamine antagonists) such as bromopheniramine, chlorpheniramine, dexchlorpheniramine, triprolidine, clemastine, diphenhydramine, diphenylpyraline, tripelennamine, hydroxyzine, methdilazine, promethazine, trimeprazine, azatadine, cyproheptadine, antazoline, pheniramine pyrilamine, astemizole, terfenadine, loratadine, cetirizine, fexofenadine, descarboethoxyloratadine, and the like; (e) non-steroidal anti-asthmatics such as β2-agonists (terbutaline, metaproterenol, fenoterol, isoetharine, albuterol, bitolterol, and pirbuterol), theophylline, cromolyn sodium, atropine, ipratropium bromide, leukotriene antagonists (zafirlukast, montelukast, pranlukast, iralukast, pobilukast, SKB-106,203), leukotriene biosynthesis inhibitors (zileuton, BAY-1005); (f) non-steroidal antiinflammatory agents (NSAIDs) such as propionic acid derivatives (alminoprofen, benoxaprofen, bucloxic acid, carprofen, fenbufen, fenoprofen, fluprofen, flurbiprofen, ibuprofen, indoprofen, ketoprofen, miroprofen, naproxen, oxaprozin, pirprofen, pranoprofen, suprofen, tiaprofenic acid, and tioxaprofen), acetic acid derivatives (indomethacin, acemetacin, alclofenac, clidanac, diclofenac, fenclofenac, fenclozic acid, fentiazac, furofenac, ibufenac, isoxepac, oxpinac, sulindac, tiopinac, tolmetin, zidometacin, and zomepirac), fenamic acid derivatives (flufenamic acid, meclofenamic acid, mefenamic acid, niflumic acid and tolfenamic acid), biphenylcarboxylic acid derivatives (diflunisal and flufenisal), oxicams (isoxicam, piroxicam, sudoxicam and tenoxican), salicylates (acetyl salicylic acid, sulfasalazine) and the pyrazolones (apazone, bezpiperylon, feprazone, mofebutazone, oxyphenbutazone, phenylbutazone); (g) cyclooxygenase-2 (COX-2) inhibitors such as celecoxib; (h) inhibitors of phosphodiesterase type IV (PDE-IN); (i) antagonists of the chemokine receptors, especially CCR-1, CCR-2, and CCR-3; (j) cholesterol lowering agents such as HMG- CoA reductase inhibitors (lovastatin, simvastatin and pravastatin, fluvastatin, atorvastatin, and other statins), sequestrants (cholestyramine and colestipol), nicotinic acid, fenofibric acid derivatives (gemfibrozil, clofibrat, fenofibrate and benzafibrate), and probucol; (k) anti-diabetic agents such as insulin, sulfonylureas, biguanides (metformin), α-glucosidase inhibitors (acarbose) and glitazones (troglitazone, pioglitazone, englitazone, MCC-555, BRL49653 and the like); (1) preparations of interferon beta (interferon beta-la, interferon beta- lb); (m) anticholinergic agents such as muscarinic antagonists (ipratropium bromide); (n) other compounds such as 5-aminosalicylic acid and prodrugs thereof, antimetabolites such as azathioprine and 6- mercaptopurine, and cytotoxic cancer chemotherapeutic agents. The weight ratio of the compound of the Formula I to the second active ingredient may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used. Thus, for example, when a compound of the Formula I is combined with an NSAID the weight ratio of the compound of the Formula I to the NSAID will generally range from about 1000:1 to about 1:1000, preferably about 200:1 to about 1:200. Combinations of a compound of the Formula I and other active ingredients will generally also be within the aforementioned range, but in each case, an effective dose of each active ingredient should be used. Compounds of the present invention may be prepared by procedures illustrated in the accompanying schemes. In the first method (Scheme 1), a resin-based synthetic strategy is outlined where the resin employed is represented by the ball ( ^ ). An N-Fmoc-protected amino acid derivative A (Fmoc = fluorenylmethoxycarbonyl) is loaded on to the appropriate hydroxyl-containing resin using dicyclohexylcarbodiimide (DCC) and 1-hydroxybenzotriazole (HOBt) in dimethylformamide (DMF) to give R The Fmoc protecting group is removed with piperidine in DMF to yield free amine C. The next Fmoc- protected amino acid derivative D is coupled to C employing standard peptide (in this instance, 2-(lH-benzotriazol-l-yl)-l, 1,3,3- tetramethyluronium hexafluorophosphate (HBTU), HOBt, and N,N- diisopropylethylamine (DIEA) in DMF) to yield dipeptide E. The Fmoc group is removed with piperidine in DMF to yield the free amine F. A sulfonyl chloride derivative is reacted with F in the presence of DIEA to yield G. The final product is removed from the resin with strong acid (in this instance, trifluoroacetic acid (TFA) in the presence of thioanisole and dithiane) to yield compounds of the present invention H. Scheme 1
Figure imgf000029_0001
Figure imgf000029_0002
Figure imgf000029_0003
H In the second method (Scheme 2), standard solution phase synthetic methodology is outlined. An N-Boc-protected amino acid derivative A (Boc = tert-butyloxycarbonyl) is treated with tert-butyl 2,2,2- trichloroacetimidate in the presence of boron trifluoride etherate to yield tert-butyl ester B which is subsequently coupled to Cbz-protected amino acid derivative C (Cbz = carbobenzyloxy) in the presence of l-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC), HOBt, and N-methylmorpholine (NMM) in methylene chloride (CH2CI2) to yield dipeptide D. Catalytic hydrogenation of D in the presence of a palladium-on-carbon (Pd/C) catalyst yields E. Reaction of E with a sulfonyl chloride in the presence of DIEA and 4-dimethylaminopyridine (DMAP) yields F which is subsequently reacted with strong acid (TFA) to yield the desired product G.
Scheme 2
Figure imgf000031_0001
B
Figure imgf000031_0002
Figure imgf000031_0003
Figure imgf000031_0004
GENERAL PROCEDURE FOR THE SOLID-PHASE SYNTHESIS OF COMPOUNDS OF FORMULA I.
Step A. Loading of N-Fmoc-amino acid derivatives onto resins. N-Fmoc-amino acids were loaded on either Wang®
(Calbiochem-Novabiochem Corp.) or Chloro (2-chlorotrityl) resin. Wang® resin, typically 0.3 mmol, was washed with dimethylformamide three times. A solution of N-Fmoc-amino acid (0.3 mmol) in dimethylformamide (3 mL) was transferred to the pre-swollen Wang® resin. Dicyclohexylcarbodiimide (0.3 mmol) and 1-N- hydroxybenztriazole (0.3 mmol) was added and the mixture gently swirled for 2 hours. Following filtration, the resin was sequentially washed with dimethylformamide (3 times) and dichloromethane (3 times). The amino acid substitution value obtained after vacuum drying typically ranged between 0.07 to 0.1 mmol.
Alternatively, Chloro (2-chorotrityl) resin, typically 0.2 mmol, was pre-swollen in dimethylformamide. A solution of N-Fmoc- amino acid (0.2 mmol) in dimethylformamide (3 ml) was added to the resin, followed by the addition of N,N-diisopropylethylamine(0.4 mmol). The resin was gently stirred for 2 hours, filtered and washed sequentially with dimethylformamide (3 times) and dichloromethane (3 times). The resin was finally washed with 10% methanol in dichloromethane and vacuum dried. The amino acid substitution value obtained after vacuum drying typically ranged between 0.05 to 0.1 mmol.
Step B. Deprotection of the N-Fmoc group.
The N-Fmoc protecting group was removed from the resin from Step A by treatment with 20% piperidine in dimethylformamide for 30 minutes. Following filtration, the resin was washed sequentially with dimethylformamide (3 times), dichloromethane (1 time) and dimethylformamide (2 times) and used in the subsequent reaction. Step C. Coupling of the next N-Fmoc-amino acid derivative
A solution of the next desired N-Fmoc-amino acid derivative (0.4 mmol) in dimethylformamide (2 mL) was mixed with 2-(lH- benzotriazol-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate (0.4 mmol), l-N-hydroxybenztriazole(0.4 mmol) and diisopropylethylamine (0.6 mmol). This solution was transferred to resin from Step B and typically allowed to react for 2 hours. Couplings were monitored by ninhydrin reaction. The coupling mixture was filtered and the resin washed with dimethylformamide (3 times) and used in the subsequent reaction.
Step D. Deprotection of the N-Fmoc group.
The N-Fmoc protecting group was removed from the resin from Step C by the procedure described in Step B and used in the subsequent reaction.
Step E. Acylation (or sulfonylation) of the terminal amino group.
The desired N-terminal capping reagent (sulfonylchloride or acylchloride) (0.4 mol) was dissolved in dimethylformamide (2 ml), mixed with N,N-diisopropylethylamine(0.8 mmol) and added to the resin from Step D. After approximately two hours, the resin was sequentially washed with dimethylformamide (3 times) and dichloromethane (3 times).
Step F. Cleavage of the desired products from the resins.
The final desired products were cleaved from the resins from Step E by gently stirring with a solution of trifluoroacetic acid:thioanisole:ethanedithiol (95:2.5:2.5); 3 hours for Wang® resin and 30 minutes for the Chloro (2-chorotrityl) resin. Following filtration, the solvents were removed by evaporation and the residue dissolved in acetonitrile (3 mL). Insoluble material was removed by filtration. The final products were purified by reverse phase chromatography with a linear gradient of buffer A (0.1% trifluoroacetic acid in water) and buffer B (0.1% trifluoroacetic acid in acetonitrile) and isolated by lyophilization. Molecular ions were obtained by electrospray ionization mass spectrometry or matrix-assisted laser desorption ionization time-of- flight mass spectrometry to confirm the structure of each peptide.
The following compounds were prepared by the general procedure described above using the appropriate amino acid derivatives and sulfonyl chloride. These examples are provided to illustrate the present invention and are not to be construed as limiting its scope in any manner.
Ex. No. Compound Name MS*
(1) N-(3,4-dimethoxybenzenesulfonyl)-(L)-3-(2- 485 thienyDalanyl-norleucine;
(2) N-(3,4-dimethoxybenzenesulfonyl)-(L)-3,3- 555 diphenylalanyl-(L)-norleucine;
(3) N-(3,4-dimethoxybenzenesulfonyl)-(L)-aspartyl-(L)- 447 norleucine;
(4) N-(3,4-dimethoxybenzenesulfonyl)-(L)-3-(3- 481 pyridyl)alanyl-(L)-norleucine;
(5) N-(3,4-dimethoxybenzenesulfonyl)-(L)-alanyl-(L)- 403 norleucine;
(6) N-(3,4-dimethoxybenzenesulfonyl)-(L)- 493 homophenylalanyl-(L)-norleucine;
(7) N-(3,4-dimethoxybenzenesulfonyl)-(L)-valyl-(L)-3-(2- 515 naphthyl)alanine;
(8) N-(3,4-dimethoxybenzenesulfonyl)-(L)-asparagyl-(L)- 446 norleucine;
(9) N-(3,4-dimethoxybenzenesulfonyl)-(L)-isoleucyl-(L)- 529 3-(2-naphthyl)alanine;
(10) N-(3,4-dimethoxybenzenesulfonyl)-(L)-3-(4- 555 biphenyl)alanyl-(L)-norleucine; (11) N-(3,5-dichlorobenzenesulfonyl)-(L)-N-methylvalyl- 537 (L)-3-(2-naphthyl)alanine;
(12) N-(3,5-di(trifluoromethyl)-benzenesulfonyl)-(L)-N- 605 methylvalyl-(L)-3-(2-naphthyl)alanine;
(13) N-(3,5-dichlorobenzenesulfonyl)-2(S)-aminobutyryl- 509 (L)-3-(2-naphthyl)alanine;
(14) N-(3,4-dimethoxybenzenesulfonyl)-(L)-N- 543 methylisoleucyl-(L)-3-(2-naphthyl)alanine;
(15) N-(3,4-dimethoxybenzenesulfonyl)-(L)-N- 529 methylvalyl-(L)-3-(2-naphthyl)alanine;
(16) N-(3,4-dimethoxybenzenesulfonyl)-(L)-sarcosinyl-(L)- 487 3-(2-naphthyl)alanine;
(17) N-(3,4-dimethoxybenzenesulfonyl)-(L)-phenylalanyl- 479 (L)-norleucine;
(18) N-(3,4-dimethoxybenzenesulfonyl)-(L)-N- 492 methylphenylalanyl-(L)-norleucine;
(19) N-(3,4-dimethoxybenzenesulfonyl)-(L)-α- 465 phenylglycyl-(L)-norleucine ;
(20) N-(3,4-dimethoxybenzenesulphonyl)-(L)-N- 417 methylalanyl-(L)-norleucine;
(21) N-(3,5-dichlorobenzenesulphonyl)-(L)-tyrosyl-(L)-4- 555 fluorophenylalanine;
(22) N-(3,5-dichlorobenzenesulphonyl)-(L)-3-(4- 540 pyridyl)alanine-(L)-4-fluorophenylalanine;
(23) N-(3,5-dichlorobenzenesulphonyl)-(L)-phenylalanyl- 539.1 (L)-4-fluorophenylalanine;
(24) N-(3,5-dichlorobenzenesulphonyl)-(L)-4- 557.1 fluorophenylalanyl-(L)-4-fluorophenylalanine;
(25) N-(3,5-dichlorobenzenesulphonyl)-(L)-3,5- 575.1 difluorophenylalanyl-(L)-4-fluorophenylalanine;
(26) N-(3,5-dichlorobenzenesulphonyl)-(L)-3-(2- 540 pyridyl)alanyl-(L)-4-fluorophenylalanine;
(27) N-(3,5-dichlorobenzenesulphonyl)-(L)-N- 543.3 methylaspartyl-(L)-3-(2-naphthyl)alanine;
(28) N-(3-fluorobenzenesulρhonyl)-(L)-N-methylvalyl-(L)- 487.2 3-(2-naphthyl)alanine;
(29) N-(3,5-dichlorobenzenesulphonyl)-(L)-4- 575.1 chlorophenylalanyl-(L)-4-fluorophenylalanine;
(30) N-(3,5-dichlorobenzenesulphonyl)-(L)-4- 564.0 cyanophenylalanyl-(L)-4-fluorophenylalanine;
(31) N-(3,5-dichlorobenzenesulphonyl)-(L)-4- 584.1 nitrophenylalanyl-(L)-4-fluorophenylalanine;
(32) N-(3,5-dichlorobenzenesulphonyl)-(L)-aspartyl-(L)-4- 507.0 fluorophenylalanine;
(33) N-(3,5-dichlorobenzenesulphonyl)-(L)-3-(3- 540.2 pyridyl)alanyl-(L)-4-fluorophenylalanine;
(34) N-(3,5-dichlorobenzenesulphonyl)-(L)-N-methylvalyl- 604.2 (L)-4-fluorophenylalanine;
(35) N-(3,5-dichlorobenzenesulphonyl)-(D)-N-methylvalyl- 537.0 (L)- 3-(2-naphthyl)alanine;
(36) N-(3-chlorobenzenesulphonyl)-2-amino-indan-2- 517.2 carbonyl-(L)-4-fluorophenylalanine;
(37) N-(3-fluorobenzenesulphonyl)-(L)-2-phenylglycyl-(L)- 475.2 4-fluorophenylalanine;
(38) N-(3,5-dichlorobenzenesulphonyl)-(L)-leucyl-(L)- 455.1 aspartic acid.
*m/e, (M)+ or (M + 1) + or (M + NH4)+.
EXAMPLE 39
Inhibition of VLA-4 Dependent Adhesion to BSA-CS-1 Conjugate
Step 1. Preparation of CS-1 Coated Plates
Untreated 96 well polystyrene flat bottom plates were coated with bovine serum albumin (BSA; 20 μg/ml) for 2 hours at room temperature and washed twice with phosphate buffered saline (PBS). The albumin coating was next derivatized with 10 μg/ml 3-(2- pyridyldithio) propionic acid N-hydroxysuccinimide ester (SPDP), a heterobifunctional crosslinker, for 30 minutes at room temperature and washed twice with PBS. The CS-1 peptide (Cys-Leu-His-Gly-Pro-Glu-Ile- Leu-Asp-Val-Pro-Ser-Thr), which was synthesized by conventional solid phase chemistry and purified by reverse phase HPLC, was next added to the derivatized BSA at a concentration of 2.5 μg/ml and allowed to react for 2 hours at room temperature. The plates were washed twice with PBS and stored at 4°C.
Step 2. Preparation of Fluorescently Labeled Jurkat Cells
Jurkat cells, clone E6-1, obtained from the American Type Culture Collection (Rockville, MD; cat # ATCC TIB-152) were grown and maintained in RPMI-1640 culture medium containing 10% fetal calf serum (FCS), 50 units/ml penicillin, 50 μg/ml streptomycin and 2 mM glutamine. Fluorescence activated cell sorter analysis with specific monoclonal antibodies confirmed that the cells expressed both the α4 and βl chains of VLA-4. The cells were centrifuged at 400xg for five minutes and washed twice with PBS. The cells were incubated at a concentration of 2 x 10 cells/ml in PBS containing a 1 μM concentration of a fluorogenic esterase substrate (2', 7'-bis-(2-carboxyethyl)-5-(and -6)- carboxyfluorescein, acetoxymethyl ester; BCECF-AM; Molecular Probes Inc., Eugene, Oregon; catalog #B-1150) for 30-60 minutes at 37°C in a 5% Cθ2/air incubator. The fluorescently labeled Jurkat cells were washed two times in PBS and resuspended in RPMI containing 0.25% BSA at a final concentration of 2.0 x 10 cells/ml.
Step 3. Assay Procedure
Compounds of this invention were prepared in DMSO at lOOx the desired final assay concentration. Final concentrations were selected from a range between 0.001 nM-100 μM. Three μL of diluted compound, or vehicle alone, were premixed with 300 μL of cell suspension in 96-well polystyrene plates with round bottom wells. 100 μL aliquots of the cell /compound mixture were then transferred in duplicate to CS-1 coated wells. The cells were next incubated for 30 minutes at room temperature. The non-adherent cells were removed by two gentle washings with PBS. The remaining adherent cells were quantitated by reading the plates on a Cytofluor II fluorescence plate reader (Perseptive Biosystems Inc., Framingham, MA; excitation and emission filter settings were 485 nm and 530 nm, respectively). Control wells containing vehicle alone were used to determine the level of cell adhesion corresponding to 0% inhibition. Control wells coated with BSA and crosslinker (no CS-1 peptide) were used to determine the level of cell adhesion corresponding to 100% inhibition. Cell adhesion to wells coated with BSA and crosslinker was usually less than 5% of that observed to CS-1 coated wells in the presence of vehicle. Percent inhibition was then calculated for each test well and the IC50 was determined from a ten point titration using a validated four parameter fit algorithm.
EXAMPLE 40
Antagonism of VLA-4 Dependent Binding to VCAM-Ig Fusion Protein.
1. Preparation of VCAM-Ig The signal peptide as well as domains 1 and 2 of human
VCAM (GenBank Accession no. M30257) were amplified by PCR using the human VCAM cDNA (R & D Systems) as template and the following primer sequences: 3'-PCR primer:5'-AATTATAATTTGATCAACTTAC
CTGTCAATTCTTTTACAGCCTGCC-3'; 5'-PCR primer:
5'-ATAGGAATTCCAGCTGCCACCATGCCTGGGAAGATGGTCG-3'. The 5'-PCR primer contained EcoRI and PvuII restriction sites followed by a Kozak consensus sequence (CCACC) proximal to the initiator methionine ATG. The 3'-PCR primer contained a Bell site and a splice donor sequence. PCR was performed for 30 cycles using the following parameters: 1 min. at 94 C, 2 min. at 55 C, and 2 min. at 72 C. The amplified region encoded the following sequence of human VCAM-1:
MPGKMWILGASNILWIMFAASQAFKIETTPESRYLAQIGDSVSLTC STTGCESPFFSWRTQIDSPLNGKVTNEGTTSTLTMNPVSFGNEHSYLC TATCESRl- EKGIQVΕ-ΥSFPKDPEIHLSGPLEAGKPITVKCSVADVY PFDRLEIDLLKGDHLMKSQEFLEDADRKSLETKSLEVTFTPVIEDIGKV LVC-----ΗLHIDEMDSVPTVRQAVKEL. The resulting PCR product of 650 bp was digested with EcoRI and Bell and ligated to expression vector pig-Tail (R & D Systems, Minneapolis, MN) digested with EcoRI and BamHI. The pig-Tail vector contains the genomic fragment which encodes the hinge region, CH2 and CH3 of human IgGl (GenBank Accession no. Z17370). The DNA sequence of the resulting VCAM fragment was verified using Sequenase (US Biochemical, Cleveland, OH). The fragment encoding the entire VCAM-Ig fusion was subsequently excised from pig-Tail with EcoRI and NotI and ligated to pCI-neo (Promega, Madison, WI) digested with EcoRI and NotI. The resulting vector, designated pCI-neo/VCAM-Ig was transfected into CHO-K1 (ATCC CCL 61) cells using calcium-phosphate DNA precipitation (Specialty Media, Lavalette, NJ). Stable VCAM-Ig producing clones were selected according to standard protocols using 0.2-0.8 mg/ml active G418 (Gibco, Grand Island, NY), expanded, and cell supernatants were screened for their ability to mediate Jurkat adhesion to wells previously coated with 1.5 μg/ml (total protein) goat anti-human IgG (Sigma, St. Louis, MO). A positive CHO-Kl/VCAM-Ig clone was subsequently adapted to CHO-SFM serum-free media (Gibco) and maintained under selection for stable expression of VCAM-Ig. VCAM- Ig was purified from crude culture supernatants by affinity chromatography on Protein A/G Sepharose (Pierce, Rockford, IL) according to the manufacturer's instructions and desalted into 50 mM sodium phosphate buffer, pH 7.6, by ultrafiltration on a YM-30 membrane (Amicon, Beverly, MA).
Step 2. Preparation of 1 0I-VCAM-Ig VCAM-Ig was labeled to a specific radioactivity greater that
125 1000 Ci/mmole with I-Bolton Hunter reagent (New England Nuclear,
Boston, MA; cat # NEX120-0142) according to the manufacturer's instructions.The labeled protein was separated from unincorporated isotope by means of a calibrated HPLC gel filtration column (G2000SW; 7.5 x 600 mm; Tosoh, Japan) using uv and radiometric detection.
Step 3. VCAM-Ig Binding Assay
Compounds of this invention were prepared in DMSO at lOOx the desired final assay concentration. Final concentrations were selected from a range between 0.001 nM-100 μM. Jurkat cells were centrifuged at 400xg for five minutes and resuspended in binding buffer (25 M HEPES, 150 M NaCl, 3 mM KC1, 2 mM glucose, 0.1% bovine serum albumin, pH 7.4). The cells were centrifuged again and resuspended in binding buffer supplemented with MnCl2 at a final concentration of 1 mM. Compounds were assayed in Millipore MHVB multiscreen plates (cat# MHVBN4550, Millipore Corp., MA) by making the following additions to duplicate wells: (i) 200 μL of binding buffer
125 containing 1 mM MnCl2; (ii) 20 μL of I-VCAM-Ig in binding buffer containing 1 mM MnCl2 (final assay concentration ~ 100 pM); (iii) 2.5 μL of compound solution or DMSO; (iv) and 0.5 x 10 cells in a volume of 30 μL. The plates were incubated at room temperature for 30 minutes, filtered on a vacuum box, and washed on the same apparatus by the addition of 100 μL of binding buffer containing 1 mM MnCl2. After insertion of the multiscreen plates into adapter plates (Packard, Meriden, CT, cat# 6005178), 100 μL of Microscint-20 (Packard cat#
6013621) was added to each well. The plates were then sealed, placed on a shaker for 30 seconds, and counted on a Topcount microplate scintillation counter (Packard). Control wells containing DMSO alone were used to determine the level of VCAM-Ig binding corresponding to 0% inhibition. Contol wells in which cells were omitted were used to determine the level of binding corresponding to 100% inhibition. Binding
125 of I- VCAM-Ig in the absence of cells was usually less than 5% of that observed using cells in the presence of vehicle. Percent inhibition was then calculated for each test well and the IC50 was determined from a ten point titration using a validated four parameter fit algorithm.
EXAMPLE 41
Antagonism of α4β7 Dependent Binding to VCAM-Ig Fusion Protein.
Step 1. α4β7 Cell line.
RPMI-8866 cells (a human B cell line α βj/β/; a gift from Prof. John Wilkins, University of Manitoba, Canada) were grown in RPMI/10% fetal calf serum/ 100 U penicillin 100 μg streptomycin/2 mM L-glutamine at 37°C, 5 % carbon dioxide. The cells were pelleted at 1000 rpm for 5 minutes and then washed twice and resuspended in binding buffer (25 M Hepes, 150 mM NaCl , 0.1 % BSA, 3 mM KC1, 2 mM Glucose, pH 7.4).
Step 2. VCAM-Ig Binding Assay
Compounds of this invention were prepared in DMSO at lOOx the desired final assay concentration. Final concentrations were selected from a range between 0.001 nM-100 μM. Compounds were assayed in Millipore MHVB multiscreen plates (Cat# MHVBN4550) by making the following sequential additions to duplicate wells: (i) 100 μl well of binding buffer containing 1.5 mM MnCl2; (ii) 10 μl/well 125I- VCAM-Ig in binding buffer (final assay concentration < 500 pM); (iii) 1.5 μl/well test compound or DMSO alone; (iv) 38 μl/well RPMI-8866 cell suspension (1.25 x 106 cells/well). The plates were incubated at room temperature for 45 minutes on a plate shaker at 200 rpm, filtered on a vacuum box, and washed on the same apparatus by the addition of 100 μL of binding buffer containing 1 mM MnCl2. After insertion of the multiscreen plates into adapter plates (Packard, Meriden, CT, cat# 6005178), 100 μL of Microscint-20 (Packard cat# 6013621) was added to each well. The plates were then sealed, placed on a shaker for 30 seconds, and counted on a Topcount microplate scintillation counter (Packard). Control wells containing DMSO alone were used to determine the level of VCAM-Ig binding corresponding to 0% inhibition. Wells in which cells were omitted were used to determine the level of binding corresponding to 100% inhibition. Percent inhibition was then calculated for each test well and the IC50 was determined from a ten point titration using a validated four parameter fit algorithm.

Claims

WHAT IS CLAIMED IS:
1. A method for the treatment of diseases, disorders, conditions or symptoms mediated by cell adhesion in a mammal which comprises administering to said mammal an effective amount of a compound of Formula I:
Figure imgf000043_0001
I
or a pharmaceutically acceptable salt thereof wherein: Rl is 1) Ci-ioalkyl,
2) C2-10alkenyl,
3) C2-10alkynyl,
4) Cy, 5) Cy-Ci-ioalkyl,
6) Cy-C2-10alkenyl,
7) Cy-C2-10alkynyl, wherein alkyl, alkenyl, and alkynyl are optionally substituted with one to four substituents independently selected from Ra; and Cy is optionally substituted with one to four substituents independently selected from RD;
R2 and R3 are independently selected from
1) hydrogen, or
2) a group selected from Rl;
R4 IS 1) hydrogen,
2) C╬╣_╬╣oalkyl,
3) C2-ioalkenyl,
4) C2_╬╣oalkynyl,
5) aryl,
6) aryl-C╬╣_╬╣oalkyl, 7) heteroaryl,
8) heteroaryl-C╬╣_╬╣oalkyl, wherein alkyl, alkenyl, and alkynyl are optionally substituted with one to four substituents independently selected from Ra; and aryl and heteroaryl optionally substituted with one to four substituents independently selected from RD; or
R3, R4 and the carbon to which they are attached form a 3-7 membered mono- or bicyclic ring containing 0-2 heteroatoms selected from N, O and S;
R5 is 1) hydrogen,
2) Ci-io alkyl,
3) Cy, or 4) Cy-Cl-io alkyl, wherein alkyl is optionally substituted with one to four substituents independently selected from Ra; and Cy is optionally substituted with one to four substituents independently selected from RD;
R6 is 1) hydrogen,
2) Ci-ioalkyl,
3) C2-10alkenyl,
4) C2-10alkynyl,
5) Cy, 6) Cy-Ci-ioalkyl,
7) Cy-C2-10alkenyl,
8) Cy-C2-10alkynyl, wherein alkyl, alkenyl and alkynyl are optionally substituted with one to four substituents selected from Rχ, and Cy is optionally substituted with one to four substituents independently selected from Ry;
R7 is 1) hydrogen,
2) Ci.ioalkyl, 3) C2-ioalkenyl,
4) C2-ioalkynyl,
5) aryl,
6) aryl-Ci.ioalkyl, 7) heteroaryl,
8) heteroaryl-C╬╣_╬╣oalkyl, wherein alkyl, alkenyl and alkynyl are optionally substituted with one to four substituents selected from Rx, and aryl and heteroaryl are optionally substituted with one to four substituents independently selected from Ry; or
R6, R7 together with the atoms to which they are attached form a 3-7 membered ring containing 0-2 heteroatoms selected from N, O and S;
Ra is 1) Cy, or
2) a group selected from Rx; wherein Cy is optionally substituted with one to four substituents independently selected from Rc;
Rk is 1) a group selected from Ra,
2) Cl-10 alkyl,
3) C2-10 alkenyl,
4) C2-10 alkynyl,
5) aryl Ci-ioalkyl, 6) heteroaryl Ci-io alkyl, wherein alkyl, alkenyl, alkynyl, aryl, heteroaryl are optionally substituted with a group independently selected from Rc;
RC is 1) halogen,
2) amino,
3) carboxy,
4) Ci_4alkyl,
5) Cl-4alkoxy, 6) aryl,
7) aryl Ci-4alkyl, or
8) aryloxy;
Rd and Re are independently selected from hydrogen, Ci-ioalkyl, C2-10 lkenyl, C2-10 lkynyl, Cy and Cy Cl-ioalkyl, wherein alkyl, alkenyl, alkynyl and Cy is optionally substituted with one to four substituents independently selected from Rc; or
Rd and Re together with the atoms to which they are attached form a heterocyclic ring of 5 to 7 members containing 0-2 additional heteroatoms independently selected from oxygen, sulfur and nitrogen;
Rf and R£ are independently selected from hydrogen, Cl-ioalkyl, Cy and Cy Ci-ioalkyl; or R* and R£ together with the carbon to which they are attached form a ring of 5 to 7 members containing 0-2 heteroatoms independently selected from oxygen, sulfur and nitrogen;
Rn is 1) hydrogen, 2) Cl-ioalkyl,
3) C2-10alkenyl,
4) C2-10alkynyl,
5) cyano,
6) aryl, 7) aryl Ci-ioalkyl,
8) heteroaryl,
9) heteroaryl Cl-ioalkyl, or
10) -S02R1; wherein alkyl, alkenyl, and alkynyl are optionally substituted with one to four substituents independently selected from Ra; and aryl and heteroaryl are each optionally substituted with one to four substituents independently selected from RD; Ri is 1) Cl-ioalkyl,
2) C2-l╬╕alkenyl,
3) C2-10alkynyl, or
4) aryl; wherein alkyl, alkenyl, alkynyl and aryl are each optionally substituted with one to four substituents independently selected from Rc;
Rx is 1) ΓûáORd,
2) ΓûáN02,
Figure imgf000047_0001
7) -S(O)mNRdRe,
8) -NRdRe,
9) -O(CRfRg)nNRdRe,
10) -C(0)Rd?
11) -C╬╕2Rd,
12) -C╬╕2(CRfRg)nCONRdRe>
13) -OC(0)Rd,
14) -CN,
15) -C(0)NRdRe,
16) -NRdC(0)Re,
17) -OC(0)NRdRe,
18) -NRdC(0)ORe,
19) -NRdC(0)NRdRe,
20) -CRd(N-ORe), or
21) -CF3;
Ry -s 1) a group selected from Rx,
2) Ci-io alkyl,
3) C2-10 alkenyl,
4) C2-IO alkynyl, 5) aryl Cl-ioalkyl,
6) heteroaryl Cl_l╬╕ alkyl,
7) cycloalkyl,
8) heterocyclyl; wherein alkyl, alkenyl, alkynyl and aryl are each optionally substituted with one to four substituents independently selected from Rx; Cy is cycloalkyl, heterocyclyl, aryl, or heteroaryl;
m is an integer from 1 to 2;
n is an integer from 1 to 10;
Figure imgf000048_0001
3) -S(0)mORd,
4) -C(0)NRdRh, or
5) -5-tetrazolyl.
2. A method of Claim 1 wherein said disease is selected from asthma, allergic rhinitis, multiple sclerosis, inflammation, inflammatory bowel disease and atherosclerosis.
3. A method of Claim 1 wherein said cell adhesion is mediated by VLA-4.
4. A method of Claim 1 wherein in compounds of Formula I
Rl is aryl optionally substituted with one to four substituents selected from RD; R2 is H or Ci-3alkyl; one of R and R4 is hydrogen and the other is Cy, Cy-Cι_ιoalkyl, or Cι_ IQalkyl, each of which is optionally substituted as provided in Claim 1; one of Rβand Η is hydrogen and the other is aryl, aryl-Cl-ioalkyl, or Ci- lOalkyl, wherein aryl is optionally substituted with one to four substituents independently selected from Ry, and alkyl is optionally substituted with one to four substituents selected from Ifr
X is -C(0)ORd;
Rb, Rd, Rx and Ry are as defined in Claim 1.
5. A compound having the formula la:
Figure imgf000049_0001
la
or a pharmaceutically acceptable salt thereof, wherein
R2 is 1) hydrogen, or
2) Ci-╬▓alkyl; one of R and R is hydrogen and the other is 1) aryl, 2) heteroaryl,
3) aryl-Ci.ioalkyl,
4) heteroaryl-Ci-ioalkyl, or
5) Cuoalkyl, wherein aryl and heteroaryl are each optionally substituted with one to four substituents selected from RD; and alkyl is optionally substituted with one to four substituents independently selected from Ra; or R3, R4 and the carbon to which they are attached form a 3-7 membered mono- or bicyclic ring containing 0-2 heteroatoms selected from N, O and S; one of R6 and R*7 is hydrogen and the other is
1) aryl,
2) aryl-C╬╣_╬╣oalkyl, or
3) Ci-ioalkyl, wherein aryl is optionally substituted with one to four substituents independently selected from Ry, and alkyl is optionally substituted with one to four substituents selected from Rx; and Ra, Rb, Rχ and Ry are as defined in Claim 1.
6. A compound of Claim 5 wherein
Rb is selected from Ci-ioalkoxy, halogen, cyano, and trifluoromethyl;
R2 is H or methyl; one of R3 and R4 is hydrogen and the other is phenyl, Cy-C╬╣_3alkyl
(wherein Cy is thienyl, pyridyl or phenyl optionally substiuted with one or two groups selected from phenyl, hydroxy, halogen, cyano, and nitro); and Cι_6alkyl optionally substituted with CO2H or CONH2; one of R6 and Η is hydrogen and the other is aryl-Cι_2alkyl, wherein aryl is optionally substituted with one to two substituents independently selected from halogen; and Ry is as defined in Claim 1.
7. A compound selected from the group consisting of:
N-(3,4-dimethoxybenzenesulfonyl)-(L)-3-(2-thienyl)alanyl-norleucine;
N-(3,4-dimethoxybenzenesulfonyl)-(L)-3,3-diphenylalanyl-(L)-norleucine; N-(3,4-dimethoxybenzenesulfonyl)-(L)-aspartyl-(L)-norleucine;
N-(3,4-dimethoxybenzenesulfonyl)-(L)-3-(3-pyridyl)alanyl-(L)-norleucine;
N-(3,4-dimethoxybenzenesulfonyl)-(L)-alanyl-(L)-norleucine;
N-(3,4-dimethoxybenzenesulfonyl)-(L)-homophenylalanyl-(L)-norleucine;
N-(3,4-dimethoxybenzenesulfonyl)-(L)-valyl-(L)-3-(2-naphthyl)alanine; N-(3,4-dimethoxybenzenesulfonyl)-(L)-asparagyl-(L)-norleucine;
N-(3,4-dimethoxybenzenesulfonyl)-(L)-isoleucyl-(L)-3-(2- naphthyDalanine; N-(3,4-dimethoxybenzenesulfonyl)-(L)-3-(4-biphenyl)alanyl-(L)- norleucine;
N-(3,5-dichlorobenzenesulfonyl)-(L)-N-methylvalyl-(L)-3-(2- naphthyl)alanine; N-(3,5-di(trifluoromethyl)-benzenesulfonyl)-(L)-N-methylvalyl-(L)-3-(2- naphthyl)alanine;
N-(3,5-dichlorobenzenesulfonyl)-2(S)-aminobutyryl-(L)-3-(2- naphthyl)alanine;
N-(3,4-dimethoxybenzenesulfonyl)-(L)-N-methylisoleucyl-(L)-3-(2- naphthyl)alanine;
N-(3,4-dimethoxybenzenesulfonyl)-(L)-N-methylvalyl-(L)-3-(2- naphthy 1 )alanine ;
N-(3,4-dimethoxybenzenesulfonyl)-(L)-sarcosinyl-(L)-3-(2- naphthyl)alanine; N-(3,4-dimethoxybenzenesulfonyl)-(L)-phenylalanyl-(L)-norleucine;
N-(3,4-dimethoxybenzenesulfonyl)-(L)-N-methylphenylalanyl-(L)- norleucine;
N-(3,4-dimethoxybenzenesulfonyl)-(L)-╬▒-phenylglycyl-(L)-norleucine;
N-(3,4-dimethoxybenzenesulfonyl)-(L)-N-methylalanyl-(L)-norleucine; N-(3,5-dichlorobenzenesulfonyl)-(L)-tyrosyl-(L)-4-fluorophenylalanine;
N-(3,5-dichlorobenzenesulfonyl)-(L)-3-(4-pyridyl)alanine-(L)-4- fluorophenylalanine ;
N-(3,5-dichlorobenzenesulfonyl)-(L)-phenylalanyl-(L)-4- fluorophenylalanine; N-(3,5-dichlorobenzenesulfonyl)-(L)-4-fluorophenylalanyl-(L)-4- fluorophenylalanine;
N-(3,5-dichlorobenzenesulfonyl)-(L)-3,5-difluorophenylalanyl-(L)-4- fluorophenylalanine ;
N-(3,5-dichlorobenzenesulfonyl)-(L)-N-methylaspartyl-(L)-2- naphthylalanine;
N-(3,5-dichlorobenzenesulfonyl)-(L)-3-(2-pyridyl)alanyl-(L)-4- fluorophenylalanine ; N-(3,5-dichlorobenzenesulfonyl)-(L)-N-methylvalyl-(L)-3-(2- naphthyl)alanine;
N-(3-fluorobenzenesulfonyl)-(L)-N-methylvalyl-(L)-3-(2-naphthyl)alanine;
N-(3,5-dichlorobenzenesulfonyl)-(L)-4-chlorophenylalanyl-(L)-4- fluorophenylalanine;
N-(3,5-dichlorobenzenesulfonyl)-(L)-4-cyanophenylalanyl-(L)-4- fluorophenylalanine ;
N-(3,5-dichlorobenzenesulfonyl)-(L)-4-nitrophenylalanyl-(L)-4- fluorophenylalanine ; N-(3,5-dichlorobenzenesulfonyl)-(L)-aspartyl-(L)-4-fluorophenylalanine;
N-(3,5-dichlorobenzenesulfonyl)-(L)-3-(3-pyridyl)alanyl-(L)-4- fluorophenylalanine; and
N-(3,5-dichlorobenzenesulfonyl)-(D)-N-methylvalyl-(L)-4- fluorophenylalanine; N-(3,5-dichlorobenzenesulfonyl)-(L)-N-methylvalyl-(L)- 3-(2- naphthyl)alanine;
N-(3-chlorobenzenesulfonyl)-2-amino-indan-2-carbonyl-(L)-4- fluorophenylalanine ;
N-(3-fluorobenzenesulfonyl)-(L)-2-phenylglycyl-(L)-4- fluorophenylalanine; and
N-(3,5-dichlorobenzenesulfonyl)-(L)-leucyl-(L)-aspartic acid.
8. A method for the treatment of diseases, disorders, conditions or symptoms mediated by cell adhesion in a mammal which comprises administering to said mammal an effective amount of a compound of Claim 5.
9. A method for the treatment of asthma, allergic rhinitis, multiple sclerosis, atherosclerosis inflammatory bowel disorder, or inflammation in a mammal which comprises administering to said mammal an effective amount of a compound of Claim 5.
10. A pharmaceutical composition which comprises a compound of Claim 5 and a pharmaceutically acceptable carrier thereof.
PCT/US1998/010952 1997-05-29 1998-05-29 Sulfonamides as cell adhesion inhibitors WO1998053818A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP98924989A EP0998282A4 (en) 1997-05-29 1998-05-29 Sulfonamides as cell adhesion inhibitors
JP50093999A JP2002501537A (en) 1997-05-29 1998-05-29 Sulfonamides as cell adhesion inhibitors
AU77032/98A AU728435B2 (en) 1997-05-29 1998-05-29 Sulfonamides as cell adhesion inhibitors
US09/424,823 US6221888B1 (en) 1997-05-29 1998-05-29 Sulfonamides as cell adhesion inhibitors
CA002291708A CA2291708A1 (en) 1997-05-29 1998-05-29 Sulfonamides as cell adhesion inhibitors

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US4795497P 1997-05-29 1997-05-29
US60/047,954 1997-05-29
GBGB9714335.8A GB9714335D0 (en) 1997-07-07 1997-07-07 Sulfonamides as cell adhesion inhibitors
GB9714335.8 1997-07-07
US6678797P 1997-11-25 1997-11-25
US60/066,787 1997-11-25
GBGB9800684.4A GB9800684D0 (en) 1998-01-14 1998-01-14 Sulfonamides as cell adhesion inhibitors
GB9800684.4 1998-01-14

Publications (1)

Publication Number Publication Date
WO1998053818A1 true WO1998053818A1 (en) 1998-12-03

Family

ID=27451678

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/010952 WO1998053818A1 (en) 1997-05-29 1998-05-29 Sulfonamides as cell adhesion inhibitors

Country Status (5)

Country Link
EP (1) EP0998282A4 (en)
JP (1) JP2002501537A (en)
AU (1) AU728435B2 (en)
CA (1) CA2291708A1 (en)
WO (1) WO1998053818A1 (en)

Cited By (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000037444A1 (en) * 1998-12-18 2000-06-29 Glaxo Group Limited Compounds useful in the treatment of inflammatory diseases
EP1017382A1 (en) * 1997-05-29 2000-07-12 Merck &amp; Co., Inc. (a New Jersey corp.) Biarylalkanoic acids as cell adhesion inhibitors
US6093696A (en) * 1997-05-30 2000-07-25 Celltech Therapeutics, Limited Tyrosine derivatives
US6110945A (en) * 1998-06-03 2000-08-29 Celltech Therapeutics Limited Aromatic amine derivatives
US6274577B1 (en) 1998-09-30 2001-08-14 Celltech Therapeutics Limited Benzodiazepines
US6291453B1 (en) 1997-07-31 2001-09-18 Athena Neurosciences, Inc. 4-amino-phenylalanine type compounds which inhibit leukocyte adhesion mediated by VLA-4
US6291511B1 (en) 1997-05-29 2001-09-18 Merck & Co., Inc. Biarylalkanoic acids as cell adhesion inhibitors
US6319922B1 (en) 1998-11-23 2001-11-20 Celltech Therapeutics Limited Propanoic acid derivatives
US6329372B1 (en) 1998-01-27 2001-12-11 Celltech Therapeutics Limited Phenylalanine derivatives
US6348463B1 (en) 1998-09-28 2002-02-19 Celltech Therapeutics Limited Phenylalanine derivatives
US6362204B1 (en) 1998-05-22 2002-03-26 Celltech Therapeutics, Ltd Phenylalanine derivatives
US6362341B1 (en) 1997-07-31 2002-03-26 Athena Neurosciences, Inc. Benzyl compounds which inhibit leukocyte adhesion mediated by VLA-4
US6403608B1 (en) 2000-05-30 2002-06-11 Celltech R&D, Ltd. 3-Substituted isoquinolin-1-yl derivatives
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
US6423688B1 (en) 1997-07-31 2002-07-23 Athena Neurosciences, Inc. Dipeptide and related compounds which inhibit leukocyte adhesion mediated by VLA-4
US6455539B2 (en) 1999-12-23 2002-09-24 Celltech R&D Limited Squaric acid derivates
US6465471B1 (en) 1998-07-03 2002-10-15 Celltech Therapeutics Limited Cinnamic acid derivatives
US6469025B1 (en) 2000-08-02 2002-10-22 Celltech R&D Ltd. 3-substituted isoquinolin-1-yl derivatives
US6482849B1 (en) 1997-06-23 2002-11-19 Tanabe Seiyaku Co., Ltd. Inhibitors of α4β1 mediated cell adhesion
US6489300B1 (en) 1997-07-31 2002-12-03 Eugene D. Thorsett Carbamyloxy compounds which inhibit leukocyte adhesion mediated by VLA-4
US6492421B1 (en) 1997-07-31 2002-12-10 Athena Neurosciences, Inc. Substituted phenylalanine type compounds which inhibit leukocyte adhesion mediated by VLA-4
US6518283B1 (en) 1999-05-28 2003-02-11 Celltech R&D Limited Squaric acid derivatives
US6521666B1 (en) 1998-01-20 2003-02-18 Tanabe Seiyaku Co., Ltd. Inhibitors of α4 mediated cell adhesion
US6521626B1 (en) 1998-03-24 2003-02-18 Celltech R&D Limited Thiocarboxamide derivatives
US6534513B1 (en) 1999-09-29 2003-03-18 Celltech R&D Limited Phenylalkanoic acid derivatives
US6545013B2 (en) 2000-05-30 2003-04-08 Celltech R&D Limited 2,7-naphthyridine derivatives
US6555562B1 (en) 1998-02-26 2003-04-29 Celltech R&D Limited Phenylalanine derivatives
WO2003026575A3 (en) * 2001-09-24 2003-05-01 Synaptic Pharma Corp Molecules specific for npff receptors and uses thereof
US6559127B1 (en) 1997-07-31 2003-05-06 Athena Neurosciences, Inc. Compounds which inhibit leukocyte adhesion mediated by VLA-4
US6583139B1 (en) 1997-07-31 2003-06-24 Eugene D. Thorsett Compounds which inhibit leukocyte adhesion mediated by VLA-4
US6610700B2 (en) 2000-04-17 2003-08-26 Celltech R & D Limited Enamine derivatives
US6630503B1 (en) 1999-08-13 2003-10-07 Biogen, Inc. Cell adhesion inhibitors
US6645939B1 (en) 1997-11-24 2003-11-11 Merck & Co., Inc. Substituted β-alanine derivatives as cell adhesion inhibitors
US6667331B2 (en) 1999-12-28 2003-12-23 Pfizer Inc Non-peptidyl inhibitors of VLA-4 dependent cell binding useful in treating inflammatory, autoimmune, and respiratory diseases
US6685617B1 (en) 1998-06-23 2004-02-03 Pharmacia & Upjohn Company Inhibitors of α4β1 mediated cell adhesion
WO2004041279A1 (en) 2002-10-30 2004-05-21 Merck & Co., Inc. Gamma-aminoamide modulators of chemokine receptor activity
US6740654B2 (en) 2000-07-07 2004-05-25 Celltech R & D Limited Squaric acid derivatives
US6756378B2 (en) 1999-06-30 2004-06-29 Pharmacopeia Drug Discovery, Inc. VLA-4 inhibitor compounds
US6939855B2 (en) 1997-07-31 2005-09-06 Elan Pharmaceuticals, Inc. Anti-inflammatory compositions and method
US6953798B1 (en) 1998-11-30 2005-10-11 Celltech R&D Limited β-alanine derivates
US7026501B2 (en) 2000-08-31 2006-04-11 Tanabe Seiyaku Co., Ltd. Inhibitors of α4 mediated cell adhesion
US7030114B1 (en) 1997-07-31 2006-04-18 Elan Pharmaceuticals, Inc. Compounds which inhibit leukocyte adhesion mediated by VLA-4
US7196112B2 (en) 2004-07-16 2007-03-27 Biogen Idec Ma Inc. Cell adhesion inhibitors
US7253171B2 (en) 2001-01-19 2007-08-07 Laboratorios Almirall, S.A. Urea derivatives as integrin α4 antagonists
WO2008151211A1 (en) * 2007-06-05 2008-12-11 Sanofi-Aventis Substituted benzoylamino-indan-2-carboxylic acids and related compounds
WO2009126920A2 (en) 2008-04-11 2009-10-15 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
WO2010061329A1 (en) 2008-11-26 2010-06-03 Pfizer Inc. 3-aminocyclopentanecarboxamides as chemokine receptor modulators
US7741362B2 (en) 2003-10-06 2010-06-22 Siegfried Wurster Somatostatin receptor 1 and/or 4 selective agonists and antagonists
WO2010090764A1 (en) 2009-02-09 2010-08-12 Supergen, Inc. Pyrrolopyrimidinyl axl kinase inhibitors
EP2510941A2 (en) 2007-02-20 2012-10-17 Merrimack Pharmaceuticals, Inc. Methods of treating multiple sclerosis by administration of alpha-fetoprotein in combination with an integrin antagonist
WO2018136935A1 (en) * 2017-01-23 2018-07-26 University Of Hawaii 2-arylsulfonamido-n-arylacetamide derivatized stat3 inhibitors
US11116760B2 (en) 2018-10-30 2021-09-14 Gilead Sciences, Inc. Quinoline derivatives
US11174256B2 (en) 2018-10-30 2021-11-16 Gilead Sciences, Inc. Imidazopyridine derivatives
US11179383B2 (en) 2018-10-30 2021-11-23 Gilead Sciences, Inc. Compounds for inhibition of α4β7 integrin
US11224600B2 (en) 2018-10-30 2022-01-18 Gilead Sciences, Inc. Compounds for inhibition of alpha 4 beta 7 integrin
US11578069B2 (en) 2019-08-14 2023-02-14 Gilead Sciences, Inc. Compounds for inhibition of α4 β7 integrin

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5663297A (en) * 1989-02-23 1997-09-02 Hoffmann-La Roche Inc. Antithrombic peptides

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6306840B1 (en) * 1995-01-23 2001-10-23 Biogen, Inc. Cell adhesion inhibitors
AU706064B2 (en) * 1995-05-10 1999-06-10 Darwin Discovery Limited Peptide compounds which inhibit metalloproteinase and TNF liberation, and their therapeutic use
PL338506A1 (en) * 1997-07-31 2000-11-06 Elan Pharm Inc Compounds inhibiting adhesion of leucocytes occuring through the mediation of vla-4
CA2291475A1 (en) * 1997-07-31 1999-02-11 Elan Pharmaceuticals, Inc. Dipeptide compounds which inhibit leukocyte adhesion mediated by vla-4
AR016133A1 (en) * 1997-07-31 2001-06-20 Wyeth Corp CARBAMILOXI COMPOUND INHIBITING THE ADHESION OF LEUKOCYTES THROUGH VLA-4, COMPOUNDS THAT ARE DRUGS OF THESE COMPOUNDS, PHARMACEUTICAL COMPOSITION, METHOD FOR SETTING VLA-4 TO A BIOLOGICAL SAMPLE, METHOD FOR THE TREATMENT OF A TREATMENT

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5663297A (en) * 1989-02-23 1997-09-02 Hoffmann-La Roche Inc. Antithrombic peptides

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP0998282A4 *

Cited By (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6291511B1 (en) 1997-05-29 2001-09-18 Merck & Co., Inc. Biarylalkanoic acids as cell adhesion inhibitors
EP1017382A1 (en) * 1997-05-29 2000-07-12 Merck &amp; Co., Inc. (a New Jersey corp.) Biarylalkanoic acids as cell adhesion inhibitors
EP1017382A4 (en) * 1997-05-29 2000-08-30 Merck & Co Inc Biarylalkanoic acids as cell adhesion inhibitors
US6093696A (en) * 1997-05-30 2000-07-25 Celltech Therapeutics, Limited Tyrosine derivatives
US6596752B1 (en) 1997-06-23 2003-07-22 Tanabe Seiyaku Co., Ltd. Inhibitors of α4β1 mediated cell adhesion
US6482849B1 (en) 1997-06-23 2002-11-19 Tanabe Seiyaku Co., Ltd. Inhibitors of α4β1 mediated cell adhesion
US7288526B2 (en) 1997-07-31 2007-10-30 Elan Pharmaceuticals, Inc. Dipeptide and related compounds which inhibit leukocyte adhesion mediated by VLA-4
US6939855B2 (en) 1997-07-31 2005-09-06 Elan Pharmaceuticals, Inc. Anti-inflammatory compositions and method
US7229970B2 (en) 1997-07-31 2007-06-12 Elan Pharmaceuticals, Inc. Carbamyloxy compounds which inhibit leukocyte adhesion mediated by VLA-4
US6586602B2 (en) 1997-07-31 2003-07-01 Eugene D. Thorsett Benzyl compounds which inhibit leukocyte adhesion mediated by VLA-4
US6583139B1 (en) 1997-07-31 2003-06-24 Eugene D. Thorsett Compounds which inhibit leukocyte adhesion mediated by VLA-4
US6559127B1 (en) 1997-07-31 2003-05-06 Athena Neurosciences, Inc. Compounds which inhibit leukocyte adhesion mediated by VLA-4
US6362341B1 (en) 1997-07-31 2002-03-26 Athena Neurosciences, Inc. Benzyl compounds which inhibit leukocyte adhesion mediated by VLA-4
US6900179B2 (en) 1997-07-31 2005-05-31 Eugene D. Thorsett Carbamyloxy compounds which inhibit leukocyte adhesion mediated by VLA-4
US7030114B1 (en) 1997-07-31 2006-04-18 Elan Pharmaceuticals, Inc. Compounds which inhibit leukocyte adhesion mediated by VLA-4
US7166580B2 (en) 1997-07-31 2007-01-23 Elan Pharmaceuticals, Inc. Compounds which inhibit leukocyte adhesion mediated by VLA-4
US6423688B1 (en) 1997-07-31 2002-07-23 Athena Neurosciences, Inc. Dipeptide and related compounds which inhibit leukocyte adhesion mediated by VLA-4
US6525026B2 (en) 1997-07-31 2003-02-25 Elan Pharmaceuticals, Inc. Carbamyloxy compounds which inhibit leukocyte adhesion mediated by VLA-4
US6291453B1 (en) 1997-07-31 2001-09-18 Athena Neurosciences, Inc. 4-amino-phenylalanine type compounds which inhibit leukocyte adhesion mediated by VLA-4
US7320960B2 (en) 1997-07-31 2008-01-22 Elan Pharmaceuticals, Inc. Carbamyloxy compounds which inhibit leukocyte adhesion mediated by VLA-4
US6492421B1 (en) 1997-07-31 2002-12-10 Athena Neurosciences, Inc. Substituted phenylalanine type compounds which inhibit leukocyte adhesion mediated by VLA-4
US6489300B1 (en) 1997-07-31 2002-12-03 Eugene D. Thorsett Carbamyloxy compounds which inhibit leukocyte adhesion mediated by VLA-4
US6645939B1 (en) 1997-11-24 2003-11-11 Merck & Co., Inc. Substituted β-alanine derivatives as cell adhesion inhibitors
US6521666B1 (en) 1998-01-20 2003-02-18 Tanabe Seiyaku Co., Ltd. Inhibitors of α4 mediated cell adhesion
US6855843B2 (en) 1998-01-20 2005-02-15 Tanabe Seiyaku Co., Ltd. Inhibitors of α4 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
US6362204B1 (en) 1998-05-22 2002-03-26 Celltech Therapeutics, Ltd Phenylalanine derivatives
US6369229B1 (en) 1998-06-03 2002-04-09 Celltech Therapeutics, Limited Pyridylalanine derivatives
US6110945A (en) * 1998-06-03 2000-08-29 Celltech Therapeutics Limited Aromatic amine derivatives
US6685617B1 (en) 1998-06-23 2004-02-03 Pharmacia & Upjohn Company Inhibitors of α4β1 mediated cell adhesion
US6465471B1 (en) 1998-07-03 2002-10-15 Celltech Therapeutics Limited Cinnamic acid derivatives
US6677339B2 (en) 1998-09-28 2004-01-13 Celltech R & D Limited Phenylalanine derivatives
US6348463B1 (en) 1998-09-28 2002-02-19 Celltech Therapeutics Limited Phenylalanine derivatives
US6274577B1 (en) 1998-09-30 2001-08-14 Celltech Therapeutics Limited Benzodiazepines
US6319922B1 (en) 1998-11-23 2001-11-20 Celltech Therapeutics Limited Propanoic acid derivatives
US6953798B1 (en) 1998-11-30 2005-10-11 Celltech R&D Limited β-alanine derivates
WO2000037444A1 (en) * 1998-12-18 2000-06-29 Glaxo Group Limited Compounds useful in the treatment of inflammatory diseases
US6867192B1 (en) 1998-12-18 2005-03-15 Duncan Robert Armour Compounds useful in the treatment of inflammatory diseases
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
US7101855B2 (en) 1999-01-26 2006-09-05 Elan Pharmaceuticals, Inc. 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
US6756378B2 (en) 1999-06-30 2004-06-29 Pharmacopeia Drug Discovery, Inc. VLA-4 inhibitor compounds
US7179819B2 (en) 1999-06-30 2007-02-20 Daiichi Pharmaceutical Co., Ltd. VLA-4 inhibitor compounds
US6630503B1 (en) 1999-08-13 2003-10-07 Biogen, Inc. Cell adhesion inhibitors
US7034043B2 (en) 1999-08-13 2006-04-25 Biogen Idec Ma 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
US6668527B2 (en) 1999-12-28 2003-12-30 Pfizer Inc. Non-peptidyl inhibitors of VLA-4 dependent cell binding useful in treating inflammatory, autoimmune, and respiratory diseases
US6903128B2 (en) 1999-12-28 2005-06-07 Pfizer Inc Non-peptidyl inhibitors of VLA-4 dependent cell binding useful in treating inflammatory, autoimmune, and respiratory diseases
US6667331B2 (en) 1999-12-28 2003-12-23 Pfizer Inc Non-peptidyl inhibitors of VLA-4 dependent cell binding useful in treating inflammatory, autoimmune, and respiratory diseases
US6780874B2 (en) 2000-04-17 2004-08-24 Celltech R & D Limited Enamine derivatives
US6610700B2 (en) 2000-04-17 2003-08-26 Celltech R & D Limited Enamine derivatives
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
US6740654B2 (en) 2000-07-07 2004-05-25 Celltech R & D Limited Squaric acid derivatives
US6469025B1 (en) 2000-08-02 2002-10-22 Celltech R&D Ltd. 3-substituted isoquinolin-1-yl derivatives
US7872151B2 (en) 2000-08-31 2011-01-18 Mitsubishi Tanabe Pharma Corporation Inhibitors of α4 mediated cell adhesion
US7456217B2 (en) 2000-08-31 2008-11-25 Mitsubishi Tanabe Pharma Corporation Inhibitors of alpha4 mediated cell adhesion
US7026501B2 (en) 2000-08-31 2006-04-11 Tanabe Seiyaku Co., Ltd. Inhibitors of α4 mediated cell adhesion
US8501809B2 (en) 2000-08-31 2013-08-06 Mitsubishi Tanabe Pharma Corporation Inhibitors of α4 mediated cell adhesion
US7671090B2 (en) 2000-08-31 2010-03-02 Mitsubishi Tanabe Pharma Corporation Inhibitors of α4 mediated cell adhesion
US7253171B2 (en) 2001-01-19 2007-08-07 Laboratorios Almirall, S.A. Urea derivatives as integrin α4 antagonists
WO2003026575A3 (en) * 2001-09-24 2003-05-01 Synaptic Pharma Corp Molecules specific for npff receptors and uses thereof
WO2004041279A1 (en) 2002-10-30 2004-05-21 Merck & Co., Inc. Gamma-aminoamide modulators of chemokine receptor activity
US7741362B2 (en) 2003-10-06 2010-06-22 Siegfried Wurster Somatostatin receptor 1 and/or 4 selective agonists and antagonists
US7196112B2 (en) 2004-07-16 2007-03-27 Biogen Idec Ma Inc. Cell adhesion inhibitors
EP2510941A2 (en) 2007-02-20 2012-10-17 Merrimack Pharmaceuticals, Inc. Methods of treating multiple sclerosis by administration of alpha-fetoprotein in combination with an integrin antagonist
WO2008151211A1 (en) * 2007-06-05 2008-12-11 Sanofi-Aventis Substituted benzoylamino-indan-2-carboxylic acids and related compounds
US8569535B2 (en) 2007-06-05 2013-10-29 Sanofi Substituted benzoylamino-indan-2-carboxylic acids and related compounds
CN101679346B (en) * 2007-06-05 2014-10-22 塞诺菲-安万特股份有限公司 Substituted benzoylamino-indan-2-carboxylic acids and related compounds
RU2477279C2 (en) * 2007-06-05 2013-03-10 Санофи-Авентис Substituted benzoylaminoindan-2-carboxylic acids and related compounds
AU2008259804B2 (en) * 2007-06-05 2013-04-18 Sanofi-Aventis Substituted benzoylamino-indan-2-carboxylic acids and related compounds
TWI446905B (en) * 2007-06-05 2014-08-01 Sanofi Aventis Substituted benzoylamino-indan-2-carboxylic acids and related compounds
EP2860260A1 (en) 2008-04-11 2015-04-15 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
WO2009126920A2 (en) 2008-04-11 2009-10-15 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
WO2010061329A1 (en) 2008-11-26 2010-06-03 Pfizer Inc. 3-aminocyclopentanecarboxamides as chemokine receptor modulators
WO2010090764A1 (en) 2009-02-09 2010-08-12 Supergen, Inc. Pyrrolopyrimidinyl axl kinase inhibitors
WO2018136935A1 (en) * 2017-01-23 2018-07-26 University Of Hawaii 2-arylsulfonamido-n-arylacetamide derivatized stat3 inhibitors
US11299480B2 (en) 2017-01-23 2022-04-12 University Of Hawaii 2-arylsulfonamido-N-arylacetamide derivatized STAT3 inhibitors
US11116760B2 (en) 2018-10-30 2021-09-14 Gilead Sciences, Inc. Quinoline derivatives
US11174256B2 (en) 2018-10-30 2021-11-16 Gilead Sciences, Inc. Imidazopyridine derivatives
US11179383B2 (en) 2018-10-30 2021-11-23 Gilead Sciences, Inc. Compounds for inhibition of α4β7 integrin
US11224600B2 (en) 2018-10-30 2022-01-18 Gilead Sciences, Inc. Compounds for inhibition of alpha 4 beta 7 integrin
US11578069B2 (en) 2019-08-14 2023-02-14 Gilead Sciences, Inc. Compounds for inhibition of α4 β7 integrin

Also Published As

Publication number Publication date
JP2002501537A (en) 2002-01-15
AU7703298A (en) 1998-12-30
CA2291708A1 (en) 1998-12-03
EP0998282A4 (en) 2000-08-30
AU728435B2 (en) 2001-01-11
EP0998282A1 (en) 2000-05-10

Similar Documents

Publication Publication Date Title
AU728435B2 (en) Sulfonamides as cell adhesion inhibitors
EP1017382B1 (en) Biarylalkanoic acids as cell adhesion inhibitors
EP1034164B1 (en) Substituted beta-alanine derivatives as cell adhesion inhibitors
US6221888B1 (en) Sulfonamides as cell adhesion inhibitors
EP1001764A1 (en) Heterocyclic amide compounds as cell adhesion inhibitors
US6645939B1 (en) Substituted β-alanine derivatives as cell adhesion inhibitors
US6271252B1 (en) Cyclic amino acid derivatives as cell adhesion inhibitors
WO1999064395A1 (en) Heterocyclic amide compounds as cell adhesion inhibitors
US6291511B1 (en) Biarylalkanoic acids as cell adhesion inhibitors
US6353099B1 (en) Substituted ureas as cell adhesion inhibitors
WO1999020272A1 (en) Azapeptide acids as cell adhesion inhibitors
WO1999026923A1 (en) Para-aminomethylaryl carboxamide derivatives
US6191171B1 (en) Para-aminomethylaryl carboxamide derivatives
WO1999026922A1 (en) Substituted pyrrole derivatives as cell adhesion inhibitors
US6020347A (en) 4-substituted-4-piperidine carboxamide derivatives
US6559174B2 (en) N-arylsulfonyl aryl aza-bicyclic derivatives as potent cell adhesion inhibitors
WO1999025685A1 (en) 4-substituted-4-piperidine carboxamide derivatives
GB2354440A (en) Aryl amides as cell adhesion inhibitors
US6855708B2 (en) N-arylsulfonyl aza-bicyclic derivatives as potent cell adhesion inhibitors
US6482840B2 (en) Substituted cyclic amidine derivatives as inhibitors of cell adhesion
US6734311B2 (en) Substituted amidine derivatives as inhibitors of cell adhesion
US6069163A (en) Azapeptide acids as cell adhesion inhibitors
US6090841A (en) Substituted pyrrole derivatives as cell adhesion inhibitors
US6903075B1 (en) Heterocyclic amide compounds as cell adhesion inhibitors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AU AZ BA BB BG BR BY CA CN CU CZ EE GE GW HU ID IL IS JP KG KR KZ LC LK LR LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK SL TJ TM TR TT UA US UZ VN YU

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 1998924989

Country of ref document: EP

ENP Entry into the national phase

Ref country code: JP

Ref document number: 1999 500939

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase

Ref document number: 2291708

Country of ref document: CA

Ref country code: CA

Ref document number: 2291708

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 09424823

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 77032/98

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 1998924989

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 77032/98

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 1998924989

Country of ref document: EP