WO2001056994A1 - Antagonistes de l'integrine - Google Patents

Antagonistes de l'integrine Download PDF

Info

Publication number
WO2001056994A1
WO2001056994A1 PCT/US2001/003347 US0103347W WO0156994A1 WO 2001056994 A1 WO2001056994 A1 WO 2001056994A1 US 0103347 W US0103347 W US 0103347W WO 0156994 A1 WO0156994 A1 WO 0156994A1
Authority
WO
WIPO (PCT)
Prior art keywords
aryl
heteroaryl
alkyl
antagonist
alkynyl
Prior art date
Application number
PCT/US2001/003347
Other languages
English (en)
Inventor
Zhongli Zheng
Mary Beth Carter
Julio H. Cuervo
Original Assignee
Biogen, 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
Application filed by Biogen, Inc. filed Critical Biogen, Inc.
Priority to AU2001234741A priority Critical patent/AU2001234741A1/en
Publication of WO2001056994A1 publication Critical patent/WO2001056994A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D271/00Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms
    • C07D271/02Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms not condensed with other rings
    • C07D271/061,2,4-Oxadiazoles; Hydrogenated 1,2,4-oxadiazoles
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/10Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/16Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/04Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member
    • C07D233/28Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • the present invention relates to organic compounds, which are useful for blocking the activity of integ ⁇ n molecules.
  • This invention also relates to compositions containing such compounds and methods of treatment using such compounds.
  • VLA very late antigen
  • Adhesion molecules of the VLA family presently include VLA-1, -2, -3, -4, -5, -6, -9, -10, -11, -v in which each of the molecules comp ⁇ se a ⁇ l chain non- covalently bound to a ⁇ chain, ( ⁇ l, oc2, oc3, oc4, ⁇ 5, oc6, ⁇ 9, al l, av), respectively.
  • the lnteg ⁇ n a4 ⁇ 7 is also intended to be included within the VLA family
  • Such molecules play an essential role in both normal and pathophysiological processes in a wide variety of tissues. For instance, cellular adhesion and trafficking across the vascular interface plays an essential role in both physiological and pathophysiological processes of acute brain injury (Garcia et al 1994, Am. J. Pathol 144: 188; Becker et al, 1997 PNAS 94: 10873). Further, leukocyte migration into glomeru is a typical feature of human glomeruloneph ⁇ tis (GN) and leukocytes are key mediators of kidney damage Therefore, the targeting of specific and relevant molecules 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-mat ⁇ x interactions.
  • GN human glomeruloneph ⁇ tis
  • VLA-1, -2, -4, -6 and ⁇ 4 ⁇ 7 neutralizing antibodies or blocking peptides that inhibit the interaction between these respective VLA moieties and their ligands are known.
  • VLA-4 ( ⁇ 4 ⁇ l) and its ligand VCAM-1 some antibody antagonists have proven efficacious both prophylactic ally 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 alpha4betal integrin.' Nature, 356, 63 (1993) and E.
  • Keszthelyi et al. Evidence for a prolonged role of alpha4 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., 'alpha4-Integrins mediate antigen-induced late bronchial responses and prolonged airway hyperresponsiveness in sheep.” J. Clin. Invest. 98. 776 (1993) and A. A. Milne and P. P. Piper, Hole of VLA-4 integrin in leukocyte recruitment and bronchial hyperresponsiveness in the guinea-pig.” Eur.
  • One aspect of the invention is a compound having the following structural Formula I:
  • n is an integer from 1 to 2;
  • W is selected from
  • X is selected from S(O) 2 , S(O) 2 NR e , C(O), C(O)O, C(O)NR e , CR f R g
  • Y is selected from N, CR j
  • is selected from R 4 and
  • Q is selected from -CR j -, C(O), O, S(O)lock, NH, NXR 1 B is selected from the group consisting of 1 a bond,
  • heteroatom selected from the group consisting of nitrogen, oxygen, and sulfur;
  • alkyl- heteroaryl wherein alkyl, alkenyl. and alkynyl are optionally substituted with one to four substituents independently selected from R a , and aryl and heteroaryl are optionally substituted with one to four substituents independently selected from
  • alkyl, alkenyl and alkynyl are optionally substituted with one to four substituents selected from phenyl and R x
  • phenyl and Cy are optionally substituted with one to four substituents independently selected from R y R 4 is
  • alkyl, alkenyl and alkynyl are optionally substituted with one to four substituents selected from phenyl and R ⁇ and phenyl and Cy are optionally substituted with one to four substituents independently selected from R y Or R 4 with either R f or R s forms a mono- or bi-cychc ring containing 0-2 heteroatoms selected from nitrogen, oxygen or sulfur, wherein nitrogen is optionally substituted with R J , C(O)R e , SO 2 R e or SO 2 NR d R e R 6 , R 7 , and R 8 are each independently selected from the group consisting of R d and R x or two of R 6 , R 7 , and R 8 and the atom to which both are attached, or two of R 6 , R 7 , and R 8 and the two adjacent atoms to which they are attached, together form a 5-7 membered saturated or unsaturated monocyclic ⁇ ng containing zero to three heteroatoms selected from N, 0 or S
  • alkyl-heteroaryl wherein alkyl, alkenyl and alkynyl are optionally substituted with one to four substituents selected from R x and aryl and heteroaryl are optionally substituted with one to four substituents independently selected from R y ; or R 12 , R n and the carbon to which they are attached form a 3-7 membered mono- or bicyclic ⁇ ng containing 0-2 heteroatoms selected from N, 0 and S, wherein nitrogen is optionally substitiuted with R J , C(O)R c , SO 2 R c or SO 2 NR d R e R 16 is selected from a bond, NR ⁇ O and S(O)terrorism R a ⁇ s 1 Cy, or
  • alkyl- aryl wherein alkyl, alkenyl, .ilkynyl, aryl, heteroaryl are optionally substituted with a group independently sel scted from R c ; R c is
  • R d and R e are independently selected from hydrogen, Cl-10 alkyl, C2-10 alkenyl, C2- 10 alkynyl, Cy and Cy Cl-10 alkyl, aryl, heteroaryl, aryl-substituted aryl, aryl substituted heteroaryl, heteroaryl-substituted heteroaryl wherein alkyl, alkenyl, alkynyl, heteroaryl and Cy is optionally substituted with one to four substituents independently selected from R c ; or R d 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; wherein nitrogen is optionally substitiuted with R J , C(O)R e , SO 2 R e or SO 2 NR d R e
  • R and R g are independently selected from R or R and R g together with the carbon to which they are attached form a ring of 5 to 7 members containing 0-2 heteroatoms mdependently selected from oxygen, sulfur and nitrogen wherein nitrogen is optionally substituted with R J , C(O)R e , SO 2 R e or SO 2 NR d R e R' is
  • alkyl, alkenyl, alkynyl and aryl are each optionally substituted with one to four substituents independently selected from R
  • R J is selected from hydiogen, Cl-10 alkyl, C2-10 alkenyl, C2-10 alkynyl, Cy and Cy Cl-10 alkyl, aryl, heteroaryl, aryl-substituted aryl, aryl substituted heteroaryl, heteroaryl-substituted heteroaryl wherein said alkyl, alkenyl, alkynyl, heteroaryl, and Cy is optionally substituted with one to four substituents independently selected from R c R k and R m are independently selected from hydrogen, Cl-10 alkyl, C2-10 alkenyl, C2-
  • R x is selected from
  • R y is l a group selected from R x ,
  • heteroaryl wherein alkyl, alkenyl, alkynyl, heteroaryl and aryl are each optionally substituted with one to four substituents independently selected from R x ;
  • Cy is cycloalkyl, heterocycyl, aryl, or heteroaryl
  • R 9 is selected from H and R y R z is selected from
  • the present compounds are biologically active small molecules and are generally composed of several domains: a) an acyl (including sulfonyl) moiety and a heterocycle #1 or a substituted aromatic ⁇ ng, b) a heterocycle #2, and c) acid and a sidechain, and are named in a manner similar to that used to name ohgopeptides.
  • an integrin "antagonist” includes any compound that inhibits a "plurality" (defined below) of integ ⁇ ns from binding with an integrin ligand and/or ligand leceptors.
  • an integrin "antagonist” also refers to agents claimed herein which can inhibit or block integ ⁇ n and/or integ ⁇ n hgand-mediated binding or which can otherwise modulate integ ⁇ n and/or integrin ligand function, e.g., by inhibiting or blocking lnteg ⁇ n-hgand mediated integ ⁇ n signal transduction.
  • Such an antagonist of the integ ⁇ n/integ ⁇ n ligand interaction is an agent which has one or more of the following properties: (1) it coats, or binds to, a plurality of integ ⁇ ns (e.g, ⁇ 4 ⁇ 7 , VLA-1, VLA-9 and VLA-1 ) on the surface of such integrin bea ⁇ ng or secreting cell with sufficient specificity to inhibit an integrin ligand/mteg ⁇ n interaction, e.g., the collagen/VLA-1 interaction; (2) it coats, or binds to, a plurality of integ ⁇ ns on the surface of an mteg ⁇ n-bea ⁇ ng or secreting cell with sufficient specificity to modify, and preferably to inhibit, transduction of an mteg ⁇ n-mediated signal e.g., collagen/VLA-1 -mediated signaling; (3) it coats, or binds to, a plurality of integ ⁇ n receptors, (e.g., collagen only or collagen and V
  • the integrin antagonist has one or both of properties 1 and 2. In other preferred embodiments the antagonist has one or both of properties 3 and 4 Moreover, more than one antagonist can be administered to a patient, e.g., an agent which binds to an integrin can be combined with an agent which binds to its ligand.
  • An antagonist of the invention has "biological activity" if it inhibits a plurality integrins from binding with an integ ⁇ n ligand and/or integ ⁇ n receptor as determined by in vitro and in vivo tests known to workers having ordinary skill in the art.
  • a "pan- ⁇ l antagonist” includes any compound that inhibits a plurality (defined below) of integ ⁇ ns containing the ⁇ l subunit from binding with an integ ⁇ n ligand and/or receptor such as any receptor for the ⁇ l subunit.
  • a "pan- ⁇ l antagonist” also refers to agents claimed herein which can inhibit or block integ ⁇ n and/or integ ⁇ n gand-mediated binding or which can otherwise modulate integrin and/or integ ⁇ n ligand function, e.g., by inhibiting or blocking lnleg ⁇ n-hgand mediated integ ⁇ n signal transduction.
  • Such an antagonist of the lnteg ⁇ n/integ ⁇ n ligand interaction is an agent which has one or more of the properties (1) through (4) as described above.
  • a pan- ⁇ l antagonist of the invention has "biological activity" if it inhibits a plurality of ⁇ l subunit containing integrins (including ⁇ 4 ⁇ 7) from binding with an integrin ligand and/or receptor such as any receptoi for the ⁇ l subunit or, as defined above, any receptor for the ⁇ 4 ⁇ 7 integ ⁇ n.
  • biological activity is determined by in vitro and in vivo tests known to workers having oidinary skill in the art
  • Alkyl as well as other groups having the prefix "alk”, such as alkoxy, alkanoyl, means carbon chains which may be lmeai or branched or combinations thereof Examples ot 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.
  • alkenyl examples 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 t ⁇ ple bond, and which may be linear or branched or combinations thereof.
  • alkynyl examples include ethynyl, propargyl, 3-methyl-l-pentenyl, 2-heptynyl and the like.
  • Cycloalkyl means mono- or bicyclic saturated carbocyc c ⁇ ngs, each of which having from 3 to 10 carbon atoms. The term also includes monocyclic ⁇ ngs 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 ⁇ ngs 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-benzod ⁇ oxanyl, and the like
  • Heteroaryl means a mono- or bicyclic aromatic ⁇ ng containing at least one heteroatom selected from N, 0 and S, with each ⁇ ng containing 5 to 6 atoms.
  • heteroaryl include pyrrolyl, isoxazolyl, isothiazolyl, pyrazolyl, py ⁇ dyl, oxazolyl, oxadiazolyl, thiadiazolyl, thiazolyl, lmidazolyl, t ⁇ azolyl, tetrazolyl, furanyl, t ⁇ azinyl, thienyl, py ⁇ midyl, py ⁇ dazinyl, pyrazinyl, benzoxazolyl, benzothiazolyl, benzimidazolyl, benzofuranyl, benzothiophenyl, furo(2,3-b)py ⁇ dyl, quinolyl, indolyl, isoquinolyl, and the like.
  • Heterocyclyl means mono- or bicyclic saturated ⁇ ngs containing at least one heteroatom selected from N, S and 0, each ot 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 pyrro dinyl, pipe ⁇ dinyl, piperazinyl, lmidazohdinyl, 2,3-d ⁇ hydrofuro(2,3-b) pyndyl, benzoxazinyl, tetrahydrohydroquinohnyl, tetrahydroisoquinohnyl, dihydroindolyl, and the like.
  • the term also includes partially unsaturated monocyclic rings that are not aromatic, such as 2- or 4 pyndones attached through the nitrogen or ⁇ -substituted- (1H,3H) py ⁇ m ⁇ d ⁇ ne-2,4-d ⁇ ones ( ⁇ -substituted uracils)
  • Halogen includes fluo ⁇ ne, chlorine, bromine and iodine. "Plurality” is intended to mean. (I) any single integrm; or (II) two or more integ ⁇ ns
  • the present methods utilize (l) molecules capable of inhibiting any combination of two or more different integrins such as a molecule capable of antagonizing both VLA-4 ( ⁇ 4 ⁇ l) and ⁇ 4 ⁇ 7 or VLA-2 ( ⁇ 2 ⁇ l), VLA-6 ( ⁇ 6 ⁇ l) and VLA-4, and so on; or (n) molecules capable of selectively inhibiting any one integ ⁇ n, such as ⁇ 4 ⁇ 7 only or VLA-4 only.
  • a pan-betal antagonist inhibits any single integ ⁇ n containing a betal subunit (including ⁇ 4 ⁇ 7) or two or more integ ⁇ ns that contain a betal subunit (such two or more integ ⁇ ns which may include ⁇ 4 ⁇ 7).
  • Polymer has its art recognized meaning as being a molecule constructed from many smaller structural units called “monomers”, bonded together (preferably covalently) in any pattern.
  • the term includes linear molecules and branched molecules.
  • the term also includes homopolymers where only one species of monomer is used to build the molecule, or copolymers where the molecule is composed of two different types of monomers and so on Copolymers also include polymers where the distribution of monomers is random, alternating copolymers, block copolymers and graft copolymers.
  • the polymer is 'biocompatible'
  • a "biocompatible" substance as that term is used herein, is one that has no unacceptable toxic or injurious effects on biological function
  • Antagonists of the invention are 'small molecules' which are organic molecules.
  • a "small molecule”, as defined herein, has a molecular weight generally less than 2000.
  • the term "effective amount” as used herein, means an amount of a compound of the present invention which inhibits a "plurality" (defined herein) of integrins from binding with an integrin ligand and/or integ ⁇ n receptor, as determined by in vitro and in vivo tests known to workers having ordinary skill in the art.
  • Compounds of Formula I contain one or more asymmetric centers and can thus occur as racemates and racemic mixtures, single enantiomers, diastereome ⁇ c mixtures and individual diastereomers.
  • the present invention is meant to comprehend all such lsome ⁇ c forms of the compounds of Formula I.
  • Some of the compounds desc ⁇ bed herein contain olefinic double bonds, and unless specified otherwise, are meant to include both E and Z geomet ⁇ c isomers.
  • Some ot the compounds desc ⁇ bed 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 diastereoisome ⁇ c 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 resolv ng agent.
  • any enantiomer of a compound of the general Formula I may be obtai ⁇ ed by stereospecific synthesis using optically pure starting materials or reagents of known configuration.
  • Ar is aryl, heteroaryl, aryl-substituted aryl, aryl substituted heteroaryl which are substituted with R 7 and R 0 .
  • R 9 is selected from H and R y R z is selected from 1 OR d
  • Ar is aryl, heteroaryl, aryl-substituted aryl, aryl substituted heteroaryl which are optionally substituted with one to four substituents independently selected from R x and R is selected from H and R y .
  • R z is selected from
  • Ar is selected from aryl, heteroaryl, aryl substitiuted aryl, aryl substituted heteroaryl, nd heteroaryl substituted heteroaryl and further wherein
  • aryl, heteroaryl, aryl substitiuted aryl, aryl substituted heteroaryl, heteroaryl substituted heteroaryl are optionally substituted with 1 to 4 substituents independently selected from R and all other designations and substituents are as recited above.
  • Ar is selected from aryl, heteroaryl, aryl substitiuted aryl, aryl substituted heteroaryl, and heteroaryl substituted heteroaryl and further wherein said aryl, heteroaryl, aryl substitiuted aryl, aryl substituted heteroaryl, heteroaryl substituted heteroaryl are optionally substituted with 1 to 4 substituents independently selected from R x , R 9 is selected from H and R y and all other designations and substituents are as recited above
  • Other antagonists of the invention are shown below as Formula VI:
  • Ar is selected from aryl, heteroaryl, aryl substitiuted aryl, aryl substituted heteroaryl, and heteroaryl substituted heteroaryl and further wherein said aryl, heteroaryl, aryl substitiuted aryl, aryl substituted heteroaryl, heteroaryl substituted heteroaryl are optionally substituted with 1 to 4 substituents independently selected from R x and all other designations and substituents are as recited above.
  • Further se ⁇ es are those of Formulae VIII, IX, XI, XII, and XIII, where all designations and substituents are as recited above VIII
  • Ar is selected from aryl, heteroaryl, aryl substitiuted aryl, aryl substituted heteroaryl, and heteroaryl substituted heteroaryl and further wherein said aryl, heteroaryl, aryl substitiuted aryl, aryl substituted heteroaryl, heteroaryl substituted heteroaryl are optionally substituted with 1 to 4 substituents independently selected from R x , R 9 is selected from H and R y , R 7 is selected from
  • a single polymer molecule may be employed for conjugation with a integrin antagonist, although it is also contemplated that more than one polymer molecule can be attached as well.
  • Conjugated integrin antagonist compositions of the invention may find utility in both in vivo as well as non- in vivo applications.
  • the conjugating polymer may utilize any other groups, moieties, or other conjugated species, as appropriate to the end use application. By way of example, it may be useful in some applications to covalently bond to the polymer a functional moiety imparting UV-degradation resistance, or antioxidation, or other properties or characteristics to the polymer.
  • the polymer may contain any functionality, repeating groups, linkages, or other constitutent structures which do not preclude the efficacy of the conjugated integ ⁇ n antagonist composition for its intended purpose.
  • Illustrative polymers that may usefully be employed to achieve these desirable charactenstics are described herein below in exemplary reaction schemes.
  • the polymer may be coupled to the antagonist to form stable bonds that are not significantly cleavable by human enzymes.
  • bonds that are not 'significantly' cleavable requires that no more than about 20% of the bonds of the compound are cleaved within a 24 hour period, as measured by standard techniques in the art including, but not limited to, high pressure liquid chromatography (HPLC).
  • HPLC high pressure liquid chromatography
  • Integ ⁇ n antagonists of the invention are conjugated most preferably via a terminal reactive group on the polymer although conjugations can also be branched from non-terminal reactive groups.
  • the polymer with the reactive group(s) is designated herein as "activated polymer".
  • the reactive group selectively reacts with reactive groups on the antagonist molecule.
  • the activated polymer(s) is reacted so that attachment may occur at any available integ ⁇ n antagonist functional group.
  • Ammo, carbon, free carboxylic groups suitably activated carbonyl groups, hydroxyl, guanidyl, oxidized carbohydrate moieties, amino, carbon and mercapto groups of the integ ⁇ n antagonist (if available) can be used as attachment sites.
  • the polyme ⁇ c substanc es included herein are preferably water-soluble at room temperature.
  • a non-limiting list of such polymers includes polyalkylene oxide homopolymers such as polyethylene glycol (PEG) or polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof, provided that the water solubility of the block copolymers is maintained.
  • polyalkylene glycol residues of C1-C4 alkyl polyalkylene glycols preferably polyethylene glycol (PEG), or poly(oxy)alkylene glycol residues of such glycols are advantageously incorporated in the polymer systems of interest.
  • the polymer to which the antagonist is attached can be a homopolymer of polyethylene glycol (PEG) or is a polyoxyethylated polyol, provided in all cases that the polymer is soluble in water at room temperature.
  • Non- hmiting examples of such polymers include polyalkylene oxide homopolymers such as PEG or polypropylene glycols, polyoxyethylenated glycols, copolymers thereof and block copolymers thereof, provided that the water solubility of the block copolymer is maintained.
  • polyoxyethylated polyols include, for example, polyoxyethylated glycerol, polyoxyethylated sorbitol, polyoxyethylated glucose, or the like.
  • the glycerol backbone of polyoxyethylated glycerol is the same backbone occurring naturally in, for example, animals and humans in mono-, di-, and t ⁇ glyce ⁇ des. Therefore, this branching would not necessarily be seen as a foreign agent in the body.
  • a general formula for PEG and its derivatives is R"-(CH2CH2O)[x]-(CH2)[y]- R', where (x) represents the degree of polymerization or number of repeating units in the polymer chain and is dependent on the molecular weight of the polymer, (y) represents a positive integer, R' is (CHR 1 ), where R 1 is as defined in claim 1 and R" is a capping group (including, without limitation, OH, C[l-4 jalkyl moieties, or va ⁇ ous biologically active and inactive moieties) or is R'.
  • polyethylene glycols PEG's
  • mono-activated, C[l-4 ]alkyl-termmated PAO's such as mono-methyl- terminated polyethylene glycols (mPEG's) are preferred when mono- substituted polymers are desired; bis-activated polyethylene oxides are preferred when disubstituted antagonists are desired.
  • polyalkylene oxides dextran, polyvinyl pyrrolidones, polyacrylamides, polyvinyl alcohols, carbohydrate-based polymers and the like may be used.
  • the polymer need not have any particular molecular weight, but it is preferred that the molecular weight be between about 300 and 100,000, more preferably between 10,000 and 40,000. In particular, sizes of 20,000 or more are best at preventing loss of the product due to filtration in the kidneys.
  • Polyalkylene glycol de ⁇ vatization has a number of advantageous properties in the formulation of polymer- integ ⁇ n antagonist conjugates in the practice of the present invention, as associated with the following properties of polyalkylene glycol derivatives- improvement of aqueous solubility, while at the same time eliciting no antigenic or lmmunogenic response; high degrees of biocompatibihty; absence of in vivo bi ode gradation of the polyalkylene glycol derivatives; and ease of excretion by living organisms.
  • PEG Polyethylene glycol
  • PAO's polyalkylene oxides
  • PCT WO 93/24476 PEG has also been conjugated to proteins, peptides and enzymes to increase aqueous solubility and circulating life in vivo as well as reduce antigenicity.
  • U.S. Pat Nos. 5,298,643 and 5,321,095, both to Greenwald, et al PCT WO 93/24476 discloses using an ester linkage to covalently bind an organic molecule to water-soluble polyethylene glycols.
  • an integ ⁇ n antagonist covalently bonded to the polymer component in which the nature of the conjugation involves one or more noncleavable covalent chemical bonds which, preferably, are resistant to degradation by human enzymes.
  • Greenwald et al., supra disclose biologically-active conjugates having substantially hydrolysis-resistant bonds (linkages) between a polyalkylene oxide and the target moiety.
  • a noncleavable linker suitable for the antagonists of the present invention is-
  • R )0 and Rn are independently selected from the group consisting of H, C ⁇ _ 6 alkyls, aryls, substituted aryls, aralkyls, heteroalkyls, substituted heteroalkyls and substituted Ci 6 alkyls, q is a positive integer and F is selected from O, NR 1 , S, SO, SO 2 .
  • the linkages between a polymer and the antagonist of the invention is cleavable, allowing for control in terms of the time course over which the polymer may be cleaved from the integrin antagonist. This covalent bond between the integ ⁇ n antagonist and the polymer may be cleaved by chemical or enzymatic reaction.
  • PAO acids can be synthesized by converting mPEG-OH to an ethyl ester. See also Gehrhardt, H., et al. Polymer Bulletin 18: 487 (1987) and Veronese, F. M., et al., J. Controlled Release 10; 145 (1989).
  • the PAO-acid can be synthesized by converting mPEG-OH into a t-butyl ester. Ohya, et al., J. Bioactive and Compatible Polymers Vol.
  • doxorubicin-PEG conjugates which are prepared by linking the two substituents via va ⁇ ous linkages including esters.
  • other polyalkylene oxide de ⁇ vatives of the foregoing such as the polypropylene glycol acids, POG acids, etc., as well as other bifunctional linking groups are also contemplated.
  • the polymer- integrin antagonist product retains an acceptable amount of activity. Concurrently, portions of polyethylene glycol are present in the conjugating polymer to endow the polymer- integ ⁇ n antagonist conjugate with high aqueous solubility and prolonged blood circulation capability.
  • reaction schemes desc ⁇ bed herein are provided for the purposes of illustration only and are not to be limiting with respect to the reactions and structures which may be utilized in the modification of the integ ⁇ n antagonist, e.g , to achieve solubility, stabilization, and cell membrane affinity for parenteral and oral administration.
  • the activity and stability of the integ ⁇ n antagonist conjugates can be va ⁇ ed in several ways by using a polymer of different molecular size. Solubilities of the conjugates can be va ⁇ ed by changing the proportion and size of the polyethylene glycol fragment incorporated in the polymer composition. Salts
  • salts refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids. Salts de ⁇ ved from inorganic bases include aluminum, ammonium, calcium, copper, feme, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particularly preferred are the ammonium, calcium, magnesium, potassium, and sodium salts.
  • Salts de ⁇ ved from pharmaceutically acceptable organic nontoxic bases include salts of pnmary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resms, such as arginine, betaine, caffeine, choline, N, N'-dibenzylethylenediamine, diethylamine, 2- diethylaminoethanol, 2-d ⁇ methylam ⁇ noethanol, ethanolamine, ethylenediamine, N-ethyl- morpho ne, N-ethylpipe ⁇ dine, glucamine, glucosamine, histidine, isopropylamine, lysine, methylglucamine, morphohne, piperazine, pipe ⁇ dine, polyamine resins, procaine, pu ⁇ nes, theobromine, t ⁇ ethylamine, t ⁇ methylamine, tnpropylamine, tromethamine,
  • Such acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfomc, fuma ⁇ c, gluconic, glutamic, hydrobromic, hydrochloric, lsethionic, lactic, maleic, malic, mandehc, methanesulfonic, mucic, nitric, pamoic, pantothenic, phospho ⁇ c, succinic, sulfu ⁇ c, tartaric, p-toluenesulfonic acid, and the like.
  • Particularly preferred are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfu ⁇ c, and tartaric acids.
  • references to the compounds of Formula I aie meant to also include the pharmaceutically acceptable salts.
  • Utilities The ability of the compounds of Formula I to antagonize the actions of any VLA integ ⁇ n containing a ⁇ l subunit (including alpha4 beta 7) makes them useful for preventing or reversing the symptoms, disorders or diseases induced by the binding of VLA to its va ⁇ ous hgands.
  • these antagonists will inhibit cell adhesion processes including cell activation, migration, proliferation and differentiation and be useful in conditions such as acute or chronic renal failure or acute brain injury.
  • another aspect of the present invention piovides a method for the treatment (including prevention, alleviation, amelioration or suppression) of diseases or disorders or symptoms, including fibrotic conditions and an inflammatory disorder mediated by integrin binding and cell adhes on activation, which comprises administering to a mammal an effective amount cf a compound of Formula I.
  • an infla nmatory disorder includes, but is not limited to, skin related conditions such as psoriasis, eczema, burns and dermatitis.
  • Other inflammatory disorders contemplated for treatment by the methods of the present invention include, but are not limited to the treatment of asthma, bronchitis, menstrual cramps, tendinitis, bursitis, and the treatment of pain and headaches, or as an antipyretic for the treatment of fever.
  • the methods of the invention also would be useful to treat gastrointestinal conditions such as inflammatory bowel disease, Crohn's disease, gastritis, irritable bowel syndrome and ulcerative colitis and for the prevention of colorectal cancer.
  • the methods of the invention would be useful in treating inflammation in such diseases as vascular diseases, migraine headaches, periarteritis nodosa, thyroiditis, aplastic anemia, Hodgkin's disease, rheumatic fever, type I diabetes, myasthenia gravis, multiple sclerosis, sarcoidosis, nephrotic syndrome, Behcet's syndrome, polymyositis, gingivitis, hypersensitivity, conjunctivitis, swelling occurring after injury, myocardial ischemia, and the like.
  • diseases as vascular diseases, migraine headaches, periarteritis nodosa, thyroiditis, aplastic anemia, Hodgkin's disease, rheumatic fever, type I diabetes, myasthenia gravis, multiple sclerosis, sarcoidosis, nephrotic syndrome, Behcet's syndrome, polymyositis, gingivitis, hypersensitivity, conjunctivitis, swelling occurring after injury, myocardi
  • the methods of the invention would also be useful in the treatment of allergic rhinitis, respiratory distress syndrome, endotoxin shock syndrome, and atherosclerosis as well as asthma, allergic rhinitis, allergic conjunctivitis, inflammatory lung diseases, rheumatoid arthritis, septic arthritis, organ transplantation rejection, restenosis, autologous bone marrow transplantation, inflammatory sequelae of viral infections, myocarditis, tumor metastasis and atherosclerosis.
  • Antagonists of the present invention may also be useful in treating a subject with a fibrotic condition.
  • fibrotic condition refers to, but is not limited to, subjects afflicted with fibrosis of an internal organ, subjects afflicted with a dermal fibrosing disorder, and subjects afflicted with fibrotic conditions of the eye.
  • Fibrosis of internal organs occurs in disorders such as pulmonary fibrosis, myelofibrosis, liver cirrhosis, mesangial proliferative glomerulonephritis, crescentic glomerulonephritis, diabetic nephropathy, renal interstitial fibrosis, renal fibrosis in patients receiving cyclosporin, and HIV associated nephropathy.
  • Dermal fibrosing disorders include, but are not limited to, scleroderma, morphea, keloids, hypertrophic scars, familial cutaneous collagenoma, and connective tissue nevi of the collagen type.
  • Fibrotic conditions of the eye include conditions such as diabetic retinopathy, postsurgical scarring (for example, after glaucoma filtering surgery and after cross-eye surgery), and prohferative vitreoretinopathy
  • Additional fibrotic conditions which may be treated by the methods of the present invention include: rheumatoid arth ⁇ tis, diseases associated with prolonged joint pain and deteriorated joints; progressive systemic sclerosis, polymyositis, dermatomyositis, eosinophihc fascitis, morphea, Raynaud's syndrome, and nasal polyposis.
  • fibrotic conditions which may be treated the methods of present invention also include inhibiting overproduction of scarring in patients who are known to form keloids or hypertrophic scars, inhibiting or preventing scar ⁇ ng or overproduction of scarring du ⁇ ng healing of various types of wounds including surgical incisions, surgical abdominal wounds and traumatic lacerations, preventing or inhibiting scar ⁇ ng and reclosing of arteries following coronary angioplasty, preventing or inhibiting excess scar or fibious tissue formation associated with cardiac fibrosis after infarction and in hypersensitive vasculopathy
  • the cell adhesion inhibitory activity of these compounds may be measured by determining the concentration of inhibitor required to block the binding of cells expressing integrins to extracellular matrix components such as collagen or fibronectin coated plates In this assay microtiter wells are coated with, for example, collagen.
  • test compound may be added first and allowed to incubate with the coated wells prior to the addition of the cells. The cells are allowed to incubate in the wells for at least 30 minutes Following incubation, the wells are emptied and washed Inhibition of binding is measured by quantitating the fluorescence or radioactivity bound to the plate for each of the various concentrations of test compound, as well as for controls containing no test compound
  • Integrin expressing cells that may be utilized in this assay include Ramos cells, Jurkat cells, A375 melanoma cells, as well as human peripheral blood lymphocytes (PBLs). These cells are commercially available and may be fluorescently or radioactively labeled if desired.
  • a direct binding assay may also be employed to quantitate the inhibitory activity of the compounds of this invention (“DBA”)
  • in vitro assays such as the adhesion inhibition and direct binding assays desc ⁇ bed above, substitute the appropnate lnteg ⁇ n-expressing cell and corresponding ligand.
  • PMNs polymorphonuclear cells express integ ⁇ ns on their surface and bind to ICAM. Integ ⁇ ns are involved in platelet aggregation and inhibition may be measured in a standard platelet aggregation assay.
  • VLA-5 binds specifically to Arg-Gly-Asp sequences, while VLA-6 binds to laminin. IN VIVO TESTING
  • A. Contact Hypersensitivity An exemplary animal model is desc ⁇ bed by P.L. Chisholm et al., "Monoclonal Antibodies to the Integ ⁇ n ⁇ -4 Subunit Inhibit the Mu ⁇ ne Contact Hypersensitivity Response", Eur J. Immunol., 23, pp. 682-688 (1993) and in "Current Protocols in Immunology", J E. Cohgan, et al., Eds., John Wiley & Sons, New York, 1, pp. 4.2.1- 4.2 5 (1991), the disclosures of which are herein incorporated by reference.
  • the skin of the animal is sensitized by exposure to an irritant, such as dinitrofluorobenzene, followed by light physical imtation, such as scratching the skin lightly with a sharp edge.
  • an irritant such as dinitrofluorobenzene
  • the animals are re-sensitized following the same procedure.
  • one ear of the animal is exposed to the chemical irritant, while the other ear is treated with a non-irntant control solution.
  • the animals are given va ⁇ ous doses of the antagonists by subcutaneous injection.
  • In vivo inhibition of cell adhesion-associated inflammation is assessed by measunng the ear swelling response of the animal in the treated versus untreated ear.
  • mice Delayed hypersensitivity SRBC-induced delayed type hypersensitivity (DTH) responses are adapted from the protocol of Hurtrel et al. 1992 Cell Immunol. 142:252-263.
  • DTH delayed type hypersensitivity
  • antagonists of the invention which are prepared according to the methods described in the Examples is given prior to antigen challenge on d 5.
  • SRBC- induced DTH is a well characterized in vivo model of inflammation, and in particular psoriasis, that has been used to demonstrate the importance of a variety of cytokines and adhesion molecules in inflammation (Tedder et al. 1995 J. Exp. Med. 181:2259- 2264, Terashita et al.1996 J. Immunol. 156:4638-4643).
  • C. Asthma Another in vivo assay that may be employed to test the antagonists of this invention is the sheep asthma assay. This assay is performed essentially as described in W. M. Abraham et al., " ⁇ -Integrins Mediate Antigen-induced Late Bronchial Responses and Prolonged Airway Hyperresponsiveness in Sheep", J. Clin. Invest., 93, pp. 776-87 (1994), the disclosure of which is herein incorporated by reference. This assay measures inhibition of Ascaris antigen-induced late phase airway responses and airway hyperresponsiveness in allergic sheep D. Renal Failure
  • the agents of the present invention also may be tested in animal models of chronic renal failure.
  • Mammalian models of chronic renal failure in, for example, mice, rats, guinea pigs, cats, dogs, sheep, goats, pigs, cows, horses, and non-human primates, may be created by causing an appropriate direct or indirect injury or insult to the renal tissues of the animal.
  • Animal models of chronic renal failure may, for example, be created by performing a partial (e.g., 5/6) nephrectomy which reduces the number of functioning nephron units to a level which initiates compensatory renal hypertrophy, further nephron loss, and the progressive decline in renal function which characterizes chronic renal failure.
  • the agents of the present invention may be evaluated for their therapeutic efficacy in causing a clinically significant improvement in a standard marker of renal function when administered to a mammalian subject (e.g., a human patient) in, or at risk of, chronic renal failure.
  • a mammalian subject e.g., a human patient
  • markers of renal function are well known in the medical literature and include, without being limited to, rates of increase in BUN levels, rates of increase in serum creatinine, static measurements of BUN, static measurements of serum creatinine, glomerular filtration rates (GFR), ratios of BUN/creatinine, serum concentrations of sodium (Na+), urine/plasma ratios for creatinine, urine/plasma ratios for urea, urine osmolality, daily urine output, and the like (see, for example, Brenner and Lazarus (1994), in Harrison's Principles of Internal Medicme, 13th edition, Isselbac ler et al., ed..., McGraw Hill Text, New York; Luke and Strom (1994), in Internal M ;d ⁇ c ⁇ ne, 4th Edition, J H. Stein, ed., Mosby-Year Book, Inc. St. Louis.).
  • E. Acute Brain Injury Male Sprague Dawley (SD) or spontaneously hypertensive rats (SHRS) are anesthetized using isoflurane and the ⁇ ght middle cerebral artery (MCAO) occluded by insertion of a 4-0 nylon monofilament up the internal carotid artery to the ongm of the middle cerebral artery (MCA) (Zea Longa et al, 1989 Stroke 20:84). After lh the filament is retracted, the lschemic temtory reperfused and the animal allowed to recover After 24h the rats are sacrificed, at which time brains were removed and analyzed histologically to quantify infarct volume.
  • SD Sprague Dawley
  • SHRS spontaneously hypertensive rats
  • PBS vehicle
  • antagonist of the invention by continuous subcutaneous infusion via osmotic mini-pumps.
  • Pnmed mini osmotic pumps for example from Alza Corp.
  • the pumps are loaded to release antagonist.
  • Expe ⁇ mental treatments include a se ⁇ es of injections of antagonist given every other day (post- operation). After 14 days post balloon catheter denudation, all rats are anesthetized and the carotid arte ⁇ es fixed by perfusion at 120 mm Hg pressure with 1% paraformaldehyde and 1.25% glutaraldehyde in 0 1M phosphate buffer, pH 7 4 via a large cannula placed retrograde in the abdominal aorta. Ten minutes before fixation, these animals are given an intravenous injection of Evans blue (0 3 ml in 5% saline solution).
  • Intimal areas are measured by determining the area between lumen and internal elastic lamina. Medial areas are derermined by measu ⁇ ng the area beween internal and external elastic lamina. Intimal/medial area ratios are calculated form the measurements
  • chronic respiratory disease- free Golden Syrain hamster weighing 120-130g are purchased (e.g., from Charles River, Boston, MA) and housed in plastic cages in groups of 4 in facilities approved by the American Association for Accreditation of Laboratory Animal Care. The animals are allowed to acclimate for one week to laboratory conditions pnor to starting the expe ⁇ ments.
  • hamsters in appropnate group receive either bleomycin (5.5 un ⁇ ts/kg/4ml) or an equivalent volume (4ml/kg) of pyrogen free isotonic saline through transoral route
  • the antagonists of the invention are administered by lntrape ⁇ toneal (IP) or intratrachial route at a therapeutic dose to hamster in appropriate groups twice a week for 21-28 days post installation Thereafter, the animals in each group are killed by an overdose ot sodium pentobarbital (100-125 mg/kg lp) and their lungs processed tor biochemical and histopathological studies
  • the experimental lesion is acute mesangial pro ferative glomerulonephntis and is characterized by expansion of the mesangial matrix and hypercellulanty.
  • the nephritis reproducibly progresses through glomerular and tubulointerstitial scarring, to end stage renal disease
  • glomerulonephntis is induced in rats with a single injection of anti- glomerular basement membrane nephrotoxm serum (NTS), derived in rabbits.
  • NTS anti- glomerular basement membrane nephrotoxm serum
  • two groups of rats are treated with daily intravenous injections of saline (the negative control group) or antagonists of the invention
  • the animals are sacnficed and slides are made of the kidneys, which are stained with pe ⁇ odic acid- Schiff solution to emphasize the pathological changes.
  • the extent of glomerular injury can be quantitated by performing glomerular cell counts from 30 randomly selected glomeruh from normal animals and nephntic animals in each group.
  • Another measure of the effect of antagonists of the invention on the disease process is to quantitate the amount of extracellular mat ⁇ x accumulation in the glomeruh.
  • the degree of glomerular matnx expansion is determined as the percentage of each glomerulus occupied by the mesangial matnx according to the method of Raij et al. (1984) Kidney Int. 26: 137-43.
  • Joints are clinically monitored by determining the amount of joint erythema, swelling and distortion on a scale of 0 (normal) to 4 (severe inflammation). Radiographs are taken and are evaluated for soft tissue swelling, joint space narrowing, bone erosions and deformity. Tissue specimens are obtained and prepared for histopathologic analysis as desc ⁇ bed in Brandes et al., ibid. Total RNA is isolated from excised synovial tissues according to the method of Allen et al. (1990) J. Exp. Med. 171:231.
  • prophylactic or therapeutic dose of a compound of Formula I will, of course, vary with the nature of the seventy 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 he 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.
  • compositions which compnses a compound of Formula I and a pharmaceutically acceptable carrier.
  • composition as in pharmaceutical composition, is intended to encompass a product comp ⁇ sing the active ingredients, and the inert ingredients (pharmaceutically acceptable excipients) that make up the carrier, as well as any product which results, directly oi 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 ingredients, 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, p. renteral, ocular, pulmonary, nasal, and the like may be employed.
  • Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, ae osok, and the like.
  • compositions of the present invention compnse a compound of Formula I as an active ingredient or a pharmaceutically acceptable salt thereof, and may also contain a pharmaceutically acceptable earner 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 compositions include compositions suitable for oral, rectal, topical, parenteral
  • the compounds of the present invention are conveniently delivered in the form of an aerosol spray presentation from pressu ⁇ zed packs or nebu sers.
  • 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.
  • MDI metered dose inhalation
  • suitable propellants such as fluorocarbons or hydrocarbons
  • 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 earner according to conventional pharmaceutical compounding techniques.
  • the earner 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,, flavo ⁇ ng agents, preservatives, colo ⁇ ng 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, microcrystalhne 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 earners 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 earner which constitutes one or moie necessary ingredients.
  • the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid earners 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) VLA-4 antagonists such as those described in US 5,510,332, W097/03094, W097/02289P W096t4O781P W096/22966, W096/20216, W096101644, W096/06108, W095/15973 and W096131206; (b) steroids such as declomethasone, methylprednisolone, betamethasone, prednisone, dexamethasone, and hydrocortisone; (c) immunosuppressants such as cyclosponn, tacrolimus, rapamycin and other FK-506 type immunosuppressants; (d) antihistammes (Hl-histamine antagonists) such as bromopheniramine, chlorpheniramine, dexchlorpheniramine, triprolidine, clemastme,
  • Example 1 Cell Adhesion Assay Protocol
  • Protocol determines the ability of such organic compounds to inhibit and prevent collagen-based cell adhesion. Protocol :
  • BCECF.AM [2',7'-b ⁇ s(2-carboxyethyl)-5-(-6)-carboxyfluoresce ⁇ n, acetoxymethyl ester] at 37°C for 15 min.) each at 25 wl/well.
  • Example 2 Illustrative Synthetic Approaches
  • the artisan identifies the chemical structure of a compound having betal subur it containing integrin activity, such as, for example, the following (A):
  • the amide group adjacent to the acidic moiety in A_is identified and phantom bonds can be formed between the carbonyl of the amide and the ⁇ carbon of the amino acid to form (B) (B)
  • the amide group of (E) adjacent to the acidic moiety is identified and phantom bonds can be formed between the carbonyl of the amide and the ⁇ carbon of the amino acid (F).

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention porte sur de nouveaux antagonistes de l'intégrine de la formule (I) dans laquelle une liaison amide peptidique a été remplacée. Toutes les définitions sont telles que décrites dans les revendications.
PCT/US2001/003347 2000-02-04 2001-02-01 Antagonistes de l'integrine WO2001056994A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2001234741A AU2001234741A1 (en) 2000-02-04 2001-02-01 Integrin antagonists

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US18022500P 2000-02-04 2000-02-04
US60/180,225 2000-02-04

Publications (1)

Publication Number Publication Date
WO2001056994A1 true WO2001056994A1 (fr) 2001-08-09

Family

ID=22659685

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/003347 WO2001056994A1 (fr) 2000-02-04 2001-02-01 Antagonistes de l'integrine

Country Status (2)

Country Link
AU (1) AU2001234741A1 (fr)
WO (1) WO2001056994A1 (fr)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003051854A1 (fr) * 2001-12-11 2003-06-26 Kyowa Hakko Kogyo Co., Ltd. Dérivé de thiadiazolines
WO2004092147A1 (fr) 2003-04-18 2004-10-28 Kyowa Hakko Kogyo Co., Ltd. Inihibiteur de kinesine de stade m
EP1635822A2 (fr) * 2003-06-25 2006-03-22 Elan Pharmaceuticals, Inc. Methodes et compositions de traitement de polyarthrite rhumatoide
US7122580B2 (en) 2002-08-09 2006-10-17 Transtech Pharma, Inc. Aryl and heteroaryl compounds and methods to modulate coagulation
US7208601B2 (en) 2003-08-08 2007-04-24 Mjalli Adnan M M Aryl and heteroaryl compounds, compositions, and methods of use
US7449486B2 (en) 2004-10-19 2008-11-11 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US7459472B2 (en) 2003-08-08 2008-12-02 Transtech Pharma, Inc. Aryl and heteroaryl compounds, compositions, and methods of use
US7795282B2 (en) 2005-05-02 2010-09-14 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US7910611B2 (en) 2005-06-24 2011-03-22 Kyowa Hakko Kirin Co., Ltd. Therapeutic agent for restenosis
US8324257B2 (en) 2006-10-03 2012-12-04 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US8765735B2 (en) 2009-05-18 2014-07-01 Infinity Pharmaceuticals, Inc. Isoxazolines as inhibitors of fatty acid amide hydrolase
US8927551B2 (en) 2009-05-18 2015-01-06 Infinity Pharmaceuticals, Inc. Isoxazolines as inhibitors of fatty acid amide hydrolase
US9149465B2 (en) 2009-05-18 2015-10-06 Infinity Pharmaceuticals, Inc. Isoxazolines as inhibitors of fatty acid amide hydrolase
WO2017135472A1 (fr) 2016-02-05 2017-08-10 Eaファーマ株式会社 Dérivé de sulfonamide et composition pharmaceutique le contenant
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
WO2000000477A1 (fr) * 1998-06-30 2000-01-06 Pfizer Products Inc. Inhibiteurs non peptidyle de la liaison cellulaire dependant de vla-4 utiles dans le traitement des maladies inflammatoires, auto-immunes et respiratoires

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000000477A1 (fr) * 1998-06-30 2000-01-06 Pfizer Products Inc. Inhibiteurs non peptidyle de la liaison cellulaire dependant de vla-4 utiles dans le traitement des maladies inflammatoires, auto-immunes et respiratoires

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003051854A1 (fr) * 2001-12-11 2003-06-26 Kyowa Hakko Kogyo Co., Ltd. Dérivé de thiadiazolines
US7902234B2 (en) 2001-12-11 2011-03-08 Kyowa Hakko Kirin Co., Ltd. Thiadiazoline derivative
US7425636B2 (en) 2001-12-11 2008-09-16 Kyowa Hakko Kogyo Co., Ltd. Thiadiazoline derivative
US7759371B2 (en) 2001-12-11 2010-07-20 Kyowa Hakko Kinn Co., Ltd. Thiadiazoline derivative
US7122580B2 (en) 2002-08-09 2006-10-17 Transtech Pharma, Inc. Aryl and heteroaryl compounds and methods to modulate coagulation
US7851635B2 (en) 2003-04-18 2010-12-14 Kyowa Hakko Kirin Co., Ltd. Mitotic kinesin inhibitor
US8318782B2 (en) 2003-04-18 2012-11-27 Kyowa Hakko Kirin Co., Ltd. Mitotic kinesin inhibitor
WO2004092147A1 (fr) 2003-04-18 2004-10-28 Kyowa Hakko Kogyo Co., Ltd. Inihibiteur de kinesine de stade m
EP1635822A2 (fr) * 2003-06-25 2006-03-22 Elan Pharmaceuticals, Inc. Methodes et compositions de traitement de polyarthrite rhumatoide
EP1635822A4 (fr) * 2003-06-25 2009-06-24 Elan Pharm Inc Methodes et compositions de traitement de polyarthrite rhumatoide
US7208601B2 (en) 2003-08-08 2007-04-24 Mjalli Adnan M M Aryl and heteroaryl compounds, compositions, and methods of use
US7459472B2 (en) 2003-08-08 2008-12-02 Transtech Pharma, Inc. Aryl and heteroaryl compounds, compositions, and methods of use
US10017482B2 (en) 2004-10-19 2018-07-10 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US7956073B2 (en) 2004-10-19 2011-06-07 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US8236825B2 (en) 2004-10-19 2012-08-07 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US9102639B2 (en) 2004-10-19 2015-08-11 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US8623895B2 (en) 2004-10-19 2014-01-07 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US7449486B2 (en) 2004-10-19 2008-11-11 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US9499503B2 (en) 2004-10-19 2016-11-22 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US8268871B2 (en) 2005-05-02 2012-09-18 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US9221841B2 (en) 2005-05-02 2015-12-29 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US8580828B2 (en) 2005-05-02 2013-11-12 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US7795282B2 (en) 2005-05-02 2010-09-14 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US7910611B2 (en) 2005-06-24 2011-03-22 Kyowa Hakko Kirin Co., Ltd. Therapeutic agent for restenosis
US8324257B2 (en) 2006-10-03 2012-12-04 Array Biopharma Inc. Mitotic kinesin inhibitors and methods of use thereof
US9149465B2 (en) 2009-05-18 2015-10-06 Infinity Pharmaceuticals, Inc. Isoxazolines as inhibitors of fatty acid amide hydrolase
US8927551B2 (en) 2009-05-18 2015-01-06 Infinity Pharmaceuticals, Inc. Isoxazolines as inhibitors of fatty acid amide hydrolase
US8765735B2 (en) 2009-05-18 2014-07-01 Infinity Pharmaceuticals, Inc. Isoxazolines as inhibitors of fatty acid amide hydrolase
WO2017135472A1 (fr) 2016-02-05 2017-08-10 Eaファーマ株式会社 Dérivé de sulfonamide et composition pharmaceutique le contenant
JPWO2017135472A1 (ja) * 2016-02-05 2018-11-29 Eaファーマ株式会社 スルホンアミド誘導体及びそれを含有する医薬組成物
US10562898B2 (en) 2016-02-05 2020-02-18 Ea Pharma Co., Ltd. Substituted benzenesulfonamides as inhibitors of alpha-4 beta-7 integrin activity
EA036432B1 (ru) * 2016-02-05 2020-11-10 Эа Фарма Ко., Лтд. Производные сульфонамида и содержащие их фармацевтические композиции
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
AU2001234741A1 (en) 2001-08-14

Similar Documents

Publication Publication Date Title
US20030114514A1 (en) Pharmaceutical compositions containing anti-beta 1 integrin compounds and uses
WO2001056994A1 (fr) Antagonistes de l'integrine
JP5048201B2 (ja) ジケトピペラジン類の使用方法とジケトピペラジン類を含む組成物
AU772024B2 (en) Inhibitors of urokinase and blood vessel formation
EP1808440B1 (fr) Inhibiteurs non covalents d'urokinase et formation de vaisseaux sanguins
AU2006235835B2 (en) Non-covalent inhibitors of urokinase and blood vessel formation
JP4461217B2 (ja) オリゴまたはポリアルキレングリコール結合トロンビン阻害剤
EP0822931B1 (fr) Piperazinones utiles comme inhibiteurs d'agregation plaquettaire
CN111788184A (zh) 用作C5aR抑制剂的二芳基取代的5,5-稠合环化合物
EP3405477B1 (fr) Inhibiteurs de transglutaminases
US20090004264A1 (en) Methods for treating and ameliorating the symptons of inflammatory bowel diseases
JP2011241214A (ja) 線維症の処置におけるC5a受容体アンタゴニストの使用
JP2014210790A (ja) 液性腫瘍を治療するための方法および組成物
KR20170141757A (ko) 테트라하이드로나프티리디닐 프로피온산 유도체 및 이의 용도
JP4189216B2 (ja) Lfa−1アンタゴニストとして有用なジアザシクロアルカンジオン誘導体
JP5016178B2 (ja) 抗血栓活性を有するマロンアミドおよびマロンアミド酸エステル誘導体、それらの製造および使用
US20220041653A1 (en) Mitochondria-targeting peptides
JP2003529528A (ja) プロテアーゼインヒビターとしてのアミノ酸アミジノヒドラゾン、アルコキシグアニジン、およびアミノグアニジン
JPH0625285A (ja) 2−ピペラジノン誘導体およびその用途
AU2003269602B2 (en) Treatment of inflammatory bowel disease
CN117320718A (zh) 用于预防或治疗纤维化的药物组合物
JP2004511489A (ja) ポリヒドロキシアルキル基およびポリヒドロキシシクロアルキル基を有するセリンプロでアーゼの低分子インヒビター
AU2003269602A1 (en) Treatment of inflammatory bowel disease

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ 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 TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

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

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP