EP0845987A1 - Inhibiteurs de la c-proteinase destines au traitement des affections liees a la surproduction de collagene - Google Patents

Inhibiteurs de la c-proteinase destines au traitement des affections liees a la surproduction de collagene

Info

Publication number
EP0845987A1
EP0845987A1 EP96930499A EP96930499A EP0845987A1 EP 0845987 A1 EP0845987 A1 EP 0845987A1 EP 96930499 A EP96930499 A EP 96930499A EP 96930499 A EP96930499 A EP 96930499A EP 0845987 A1 EP0845987 A1 EP 0845987A1
Authority
EP
European Patent Office
Prior art keywords
alkyl
mono
group
sulfonyl
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP96930499A
Other languages
German (de)
English (en)
Other versions
EP0845987A4 (fr
Inventor
Mitch Brenner
Wen-Bin Ho
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Fibrogen Inc
Original Assignee
Fibrogen 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 Fibrogen Inc filed Critical Fibrogen Inc
Publication of EP0845987A1 publication Critical patent/EP0845987A1/fr
Publication of EP0845987A4 publication Critical patent/EP0845987A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/02Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link
    • C07K5/022Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link containing the structure -X-C(=O)-(C)n-N-C-C(=O)-Y-; X and Y being heteroatoms; n being 1 or 2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • 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
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • 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
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C259/00Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups
    • C07C259/04Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups without replacement of the other oxygen atom of the carboxyl group, e.g. hydroxamic acids
    • C07C259/06Compounds containing carboxyl groups, an oxygen atom of a carboxyl group being replaced by a nitrogen atom, this nitrogen atom being further bound to an oxygen atom and not being part of nitro or nitroso groups without replacement of the other oxygen atom of the carboxyl group, e.g. hydroxamic acids having carbon atoms of hydroxamic groups bound to hydrogen atoms or to acyclic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/22Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound oxygen atoms
    • C07C311/29Sulfonamides, the carbon skeleton of the acid part being further substituted by singly-bound oxygen atoms having the sulfur atom of at least one of the sulfonamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/51Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/60Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton with the carbon atom of at least one of the carboxyl groups bound to nitrogen atoms
    • 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
    • C07D207/46Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with hetero atoms directly attached to the ring nitrogen atom
    • C07D207/48Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • C07F9/38Phosphonic acids RP(=O)(OH)2; Thiophosphonic acids, i.e. RP(=X)(XH)2 (X = S, Se)
    • C07F9/40Esters thereof
    • C07F9/4003Esters thereof the acid moiety containing a substituent or a structure which is considered as characteristic
    • C07F9/4006Esters of acyclic acids which can have further substituents on alkyl

Definitions

  • Collagen is integral to, among other things, the proper formation of connective tissue.
  • the over- or under-production of collagen or the production of abnormal collagen has been linked with numerous connective tissue diseases and disorders.
  • C-proteinase is an essential key enzyme for the proper maturation of collagen, and therefore appears to be an ideal target for the inhibition, control and/or modulation of collagen formation.
  • the present invention relates to organic molecules capable of inhibiting C-proteinase activity in order to regulate, modulate and/or inhibit abnormal collagen formation. More specifically, the invention relates to the use of compounds and pharmaceutical compositions thereof for the treatment of various diseases relating to the inappropriate or unregulated production of collagen.
  • Collagen Structure At present nineteen types of collagens have been identified. These collagens, including fibrillar collagen types I, II, III are synthesized as procollagen precursor molecules which contain amino- and carboxy-terminal peptide extensions. These peptide extensions, referred to as “pro-regions, " are designated as N- and C-propeptides, respectively.
  • the pro-regions are typically cleaved upon secretion of the procollagen triple helical precursor molecule from the cell to yield a mature triple helical collagen molecule.
  • the "mature" collagen molecule is capable of association, for example, into highly structured collagen fibers. See e.g. , Fessler and Fessler, 1978, Annu. Rev. Biochem. 47: 129-162; Bornstein and Traub, 1979, in: The Proteins (eds. Neurath, H. and Hill, R.H.), Academic Press, New York, pp. 412-632; Kivirikko et al. , 1984, in: Extracellular Matrix Biochemistry (eds. Piez, K.A.
  • An array of critical diseases has been associated with the inappropriate or unregulated production of collagen, including pathological fibrosis or scarring, including endocardial sclerosis, idiopathic interstitial fibrosis, interstitial pulmonary fibrosis, perimuscular fibrosis, Symmers' fibrosis, pericentral fibrosis, hepatitis, dermatof ⁇ broma, biliary cirrhosis, alcoholic cirrhosis, acute pulmonary fibrosis, idiopathic pulmonary fibrosis, acute respiratory distress syndrome, kidney fibrosis/glomerulonephritis, kidney fibrosis/diabetic nephropathy, scleroderma/systemic, scleroderma/local, keloids, hypertrophic scars, severe joint adhesions/arthritis, myelofibrosis, corneal scarring, cystic fibrosis, muscular dystrophy (duch
  • fibrotic disorders may be induced or initiated by surgery, including scar revision plastic surgeries, glaucoma, cataract fibrosis, corneal scarring, joint adhesions, graft vs. host disease, tendon surgery, nerve entrapment, dupuytren's contracture, OB/GYN adhesions/fibrosis, pelvic adhesions, peridural fibrosis, restenosis.
  • One strategy for the treatment of these diseases is to inhibit the pathological overproduction of collagen.
  • identification and isolation of molecules which control, inhibit and/or modulate the production of collagen are of major medical interest.
  • C-proteinase is the essential key enzyme that catalyzes the cleavage of the C-propeptide of, for example, fibrillar collagens, including type I, type II, and type III collagen. See, U.S. Application Ser. No. 60/002,038, filed August 8, 1995 as a provisional application, and references disclosed therein. " C-proteinase was first observed in the culture media of human and mouse fibroblasts (Goldberg et al , 1975, Cell 4:45-50; Kessler and Goldberg, 1978, Anal. Biochem. 86:463-469), and chick tendon fibroblasts (Duskin et al , 1978, Arch.
  • a partially purified protein having C-proteinase activity was obtained from chick calvaria in 1982. Njieha et al. , 1982, Biochemistry 23:757-764.
  • chicken C-proteinase was isolated, purified and characterized from 0 conditioned media of chick embryo tendons. Hojima et al. , 1985, J. Biol. Chem. 260: 15996-16003.
  • Murine C-proteinase has been subsequently purified from media of cultured mouse fibroblasts. Kessler et al. , 1986, Collagen Relat. Res. 6:249-266; Kessler and Adar, 1989, Eur. J. Biochem. 186:115- 121.
  • the cDNA encoding human C-proteinase has been identified, as 5 set forth in the above-referenced related applications and references disclosed therein.
  • Dithiothreitol, SDS, concanavalin A, Zn 2+ , Cu 2 ' , and Cd 2+ are similarily reported to be inhibitory at low concentrations.
  • some reducing agents, several amino acids, phosphate, and ammonium sulfate were inhibitory at concentrations of 1-lOmM.
  • the enzyme was shown to be inhibited by the basic amino acids lysine and arginine. Leung et al. , supra; Ryhanen et al. , 1982, Arch. Biochem. Biophys. 215:230-236.
  • high concentrations of NaCI or Tris-HCI buffer were found to inhibit C- proteinase's activity.
  • C-proteinase activity and its inhibition have been determined using a wide array of assays. See e.g. , Kessler and Goldberg, 1978, Anal. Biochem. 86:463; Njieha et al. , 1982, Biochemistry 21:757-764. Despite the availability of such assays, large scale review and testing of potential C- proteinase inhibitors has not been performed to date due to the limited availability of human C-proteinase. As articulated in numerous publications, the enzyme is difficult to isolate by conventional biochemical means and the identity of the cDNA sequence encoding such enzyme was not known until reported in the above-referenced and related patent applications.
  • C- proteinase appears to be an ideal target for the treatment of disorders associated with the inappropriate or unregulated production and maturation of collagen.
  • none of the inhibitors so far identified has proven an effective therapeutic for the treatment of collagen-related diseases or even an inhibitor to C-proteinase activity.
  • the present invention relates to organic molecules capable of modulating, regulating and/or inhibiting production and/or maturation of collagen by affecting C-proteinase activity.
  • the compounds of the present invention have the formulae:
  • R is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, halo substitituted aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, 5 acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfmyl or sulfonyl)-alkyl;
  • R 2 is selected from the group consisting of H, lower alkyl
  • R 3 is selected from the group consisting of H, lower alkyl, mono- or * • • " poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, halo substitituted aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfmyl or sulfonyl)-alkyl;
  • * ⁇ Rj is selected from the group consisting of aryl, heteroaryl, alkyl, aralkyl, heteroaralkyl, alkylamino, arylalky lamin
  • Y is selected from the group consisting of OH, HOHN(hydroxylamine), H 2 N, alkylamino; 2 " Z is a direct bond; methylene, oxygen, sulf or, amino; n is 0 or 1 ;
  • Rj is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl- (thio, sulfmyl or sulfonyl)-alkyl;
  • R 2 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl- (thio, sulfmyl or sulfonyl)-alkyl;
  • R 3 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfmyl or sulfonyl)-alkyl;
  • R 4 is selected from the group consisting of H, lower alkyl
  • R 5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl;
  • R is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfmyl or sulfonyl)-alkyl;
  • R 2 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfmyl or sulfonyl)-alkyl;
  • R 3 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfmyl or sulfonyl)-alkyl; j is selected from the group consisting of H, lower alkyl; R 5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufmyl or sul
  • H is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfmyl or sulfonyl)-alkyl;
  • R 2 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfmyl or sulfonyl)-alkyl;
  • R 3 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfinyl or sulfonyl)-alkyl;
  • R 4 is selected from the group consisting of H, lower alkyl
  • Rj is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl;
  • Rj is selected from the group consisting of OH, alkoxyl, lower alkyl, alkylamino, peptide
  • X is selected from the group consisting of N, C;
  • R 2 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfmyl or sulfonyl)-alkyl;
  • R 3 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfmyl or sulfonyl)-alkyl;
  • R 4 is selected from the group consisting of H, lower alkyl
  • R 5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl.
  • the present invention is further directed to pharmaceutical compositions comprising a pharmaceutically effective amount of the above- described compounds and a pharmaceutically acceptable carrier or excipient. Such a composition should modulate the production and/or maturation of 3 collagen by inhibiting C-proteinase activity.
  • the present invention is also directed to the use of the disclosed compounds and compositions for the treatment of disorders associated with the inappropriate or unregulated production of collagen by modulating, inhibiting and/or regulating C-proteinase activity.
  • the compositions of the present invention may be included in methods for treating diseases associated with inappropriate or unregulated production of collagen, including, but not limited to, rheumatoid arthritis, scleroderma, pathological fibrosis or scarring.
  • C-proteinase shall be construed to mean an enzyme capable of processing collagen molecules, derivatives or fragments, or their precursors by cleaving through -Ala_l_Asp-Asp- and/or -GlyJ_Asp-Glu-.
  • the term shall include human C-proteinase and derivatives, analogs, fragments and variants
  • “Pharmaceutically acceptable salts” refers to those salts which retain the biological effectiveness and properties of the free acids and which are obtained by reaction with inorganic or organic bases such as sodium hydroxide, magnesium hydroxide, ammonia, trialkylamine, dialkylamine,
  • Alkyl refers to a saturated aliphatic hydrocarbon, including straight- chain, branched-chain, and cyclic alkyl groups.
  • the alkyl group has 1 to 12 carbons. More preferably, it is a lower alkyl of from 1 to 7
  • Typical alkyl groups includes methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl, hexyl
  • the alkyl group may be substituted or unsubstituted.
  • the substituted group(s) is preferably, carboxyl, hydroxyl, mercapto, cycloalkyl, heterocycloalkyl, halo, alkoxyl, alkylamino.
  • Aryl refers to an aromatic group which has at least one ring having a conjugated pi electron system and includes carbocylic aryl, heterocyclic aryl and biaryl groups, all of which may be optionally substimted.
  • the aryl is a substimted or unsubstituted phenyl or pyridyl.
  • Preferred aryl substituent(s) preferably phenyl or pydridyl are halogen, trihalomethyl, hydroxyl, SH, NO 2 , amine, thioether, cyano, alkoxy, and groups.
  • the present invention relates to compounds capable of regulating and/or modulating collagen formation by inhibiting C-proteinase activity.
  • the present invention is directed to compounds which inhibit C-proteinase activity as a therapeutic approach to cure or manage various connective tissue disorders, including fibrotic disorders, arthritic disorders, or disorders induced or initiated by surgical or dramatic insults.
  • the invention is generally directed to compounds and/or compositions comprising compounds having the formulae:
  • R[ is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, halo substitimted aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfinyl or sulfonyl)-alkyl;
  • R 2 is selected from the group consisting of H, lower alkyl
  • R 3 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, halo substitimted aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thk), sulfinyl or sulfonyl)-alkyl;
  • R is selected from the group consisting of aryl, heteroaryl, alkyl, aralkyl, heteroaralkyl, alkylamino, arylalky lamino;
  • Y is selected from the group consistmg of OH, HOHN(hydroxylamine), H 2 N, alkylamino;
  • Z is a direct bond; methylene, oxygen, sulfor, amino; n is 0 or 1 ; or:
  • R is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl- (thio, sulfinyl or sulfonyl )-alkyl;
  • R 2 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl- (thio, sulfinyl or sulfonyl)-alkyl;
  • R 3 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfinyl or sulfonyl)-alkyl;
  • R is selected from the group consistmg of H, lower alkyl
  • R 5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulf ony l)alky 1 , alkoyalkylacylalkyl ; or:
  • R is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfmyl or sulfonyl)-alkyl;
  • R 2 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfmyl or sulfonyl)-alkyl;
  • R 3 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfmyl or sulfonyl)-alkyl;
  • R is selected from the group consisting of H, lower alkyl
  • R 5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl; or:
  • Ri is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfinyl or sulfonyl)-alkyl;
  • R 2 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfinyl or sulfonyl)-alkyl;
  • R 3 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfinyl or sulfonyl)-alkyl;
  • R 4 is selected from the group consisting of H, lower alkyl
  • R 5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl- (thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl; or:
  • R- is selected from the group consisting of OH, alkoxyl, lower alkyl, alkylamino, peptide
  • X is selected from the group consisting of N, C;
  • R 2 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfinyl or sulfonyl)-alkyl;
  • R 3 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfinyl or sulfonyl)-alkyl;
  • R 4 is selected from the group consisting of H, lower alkyl
  • R 5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl.
  • the compounds of the present invention may have the following formulae:
  • the invention is further directed, where applicable, to solvated as well as unsolvated forms of the compounds (e.g. hydrated forms) having the ability to inhibit, regulate and/or modulate the production and/or mamration of collagen by inhibiting C-proteinase activity.
  • the compounds described above may be prepared by any process TM known to be applicable to the preparation of chemically-related compounds. Suitable processes are illustrated by the fallowing representative examples. Necessary starting materials may be obtained by standard procedures of organic chemistry.
  • a compound is subjected to a series of screens to determine the compound's ability to modulate, regulate and/or inhibit the production and mamration of collagen.
  • screens include biochemical assays, cell culmre assays and animal models.
  • Disorders associated with the inappropriate or unregulated production and/or mamration of collagen can be treated with the * J compounds and compositions of the present invention.
  • diseases or disorders include pathological fibrosis or scarring, such as endocardial sclerosis, idiopathic interstitial fibrosis, interstitial pulmonary fibrosis, perimuscular fibrosis, Symmers' fibrosis, pericentral fibrosis, hepatitis, dermatofibroma, biliary cirrhosis, alcoholic cirrhosis, acute pulmonary fibrosis, idiopathic pulmonary fibrosis, acute respiratory distress syndrome, kidney fibrosis/glomerulonephritis, kidney fibrosis/diabetic
  • pathological fibrosis or scarring such as endocardial sclerosis, idiopathic interstitial fibrosis, interstitial pulmonary fibrosis, perimuscular fibrosis, Symmers' fibrosis, pericentral fibrosis, hepatitis, dermatofibroma, biliary cirrhosis, alcoholic cirrhosis
  • nephropathy 35 nephropathy, scleroderma/ systemic, scleroderma/local, keloids, hypertrophic scars, severe joint adhesions/arthritis, myelofibrosis, corneal scarring, cystic fibrosis, muscular dystrophy (duchenne's), cardiac fibrosis, muscular fibrosis/retinal separation, esophageal stricture, payronles disease.
  • fibrotic disorders may be induced or initiated by surgery such as scar revision/plastic surgeries, glaucoma, cataract fibrosis, corneal scarring, joint adhesions, graft vs. host disease, tendon surgery, nerve entrapment, dupuytren's contracture, OB/GYN adhesions/fibrosis, pelvic adhesions, peridural fibrosis, restenosis. Still further fibrotic disorders may be induced l" by chemotherapy, including, for example lung fibrosis and the like.
  • the identified compounds can be administered to a patient in need, by itself, or in pharmaceutical compositions where it is mixed with *•*••*• > suitable carriers or excipient(s) at doses to treat or ameliorate a variety of disorders.
  • a therapeutically effective dose further refers to that amount of the compound sufficient to result in amelioration of symptoms.
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral 25 delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intra ventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.
  • a local rather than systemic manner for example, via injection of the compound directly into a ** '" arthritic joints or in fibrotic tissue, often in a depot or sustained release formulation.
  • the compounds may be administered topically, for example, as eye drops.
  • one may administer the drug in a targeted drug delivery system for example, in a liposome coated with a specific antibody, targeting, 3 for example, arthritic or fibrotic tissue.
  • a targeted drug delivery system for example, in a liposome coated with a specific antibody, targeting, 3 for example, arthritic or fibrotic tissue.
  • the liposomes will be targeted to and taken up selectively by the afflicted tissue.
  • compositions of the present 1" invention may be manufacmred in a manner that is itself known, e.g. , by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • Pharmaceutical compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more l *3 physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the agents of the invention may be formulated in aqueous 2 " solutions, preferably in physiologically compatible buffers such as Hanks 's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks 's solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the compounds can be formulated readily by 3 combining the active compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be obtained solid excipient, J j optionally grinding a resulting mixmre, and processing the mixmre of granules, after adding suitable auxiliaries, if desired, to obtain tablets or
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium
  • polyviny lpyrrolidone PVP
  • disintegrating agents such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally,
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • suitable liquids such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
  • the compositions may take the form of
  • the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g. , dichlorodifluoromethane, trichlorofluoromethane,
  • the dosage unit may be determined by providing a valve
  • Capsules and cartridges of, e.g. , gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the compounds may be formulated for parenteral administration by 3 injection, e.g. , by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g. , in ampoules or in multi- dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or *•** " dispersing agents.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents which increase the
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g. , sterile pyrogen-free water, before use.
  • a suitable vehicle e.g. , sterile pyrogen-free water
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g. , containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds may also be formulated as a depot preparation. Such long acting formulations
  • the implantation for example subcutaneously or intramuscularly
  • intramuscular injection for example, the
  • 35 compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • a pharmaceutical carrier for the hydrophobic compounds of the invention is a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • the cosolvent system may be the VPD co-solvent system.
  • VPD is a solution of 3 % w/v benzyl alcohol, 8% w/v of the nonpolar surfactant polysorbate 80,
  • VPD VPD co-solvent system
  • VPD:5W VPD diluted 1 : 1 with a 5 % dextrose in water solution.
  • This co-solvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration.
  • the proportions of a co-solvent system may be
  • co-solvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g.
  • polyvinyl pyrrolidone and other sugars or polysaccharides may substitute for dextrose.
  • Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs.
  • organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity.
  • the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials have been established and are well known by those
  • Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days.
  • compositions also may comprise suitable solid or
  • gel phase carriers or excipients examples include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • Such pharmaceutically acceptable base addition salts are those salts which retain the biological effectiveness and properties of the free acids and which are obtained by reaction with inorganic or organic bases such as sodium hydroxide, magnesium hydroxide, ammonia, trialkylamine, dialkylamine,
  • compositions suitable for use in the 2 " present invention include compositions wherein the active ingredients are contained in an effective amount to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount effective to prevent development of or to alleviate the existing symptoms of the subject being treated. Determination of the effective amounts is well within the 25 capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • the therapeutically effective dose can be estimated imtially from cell culmre assays.
  • a dose can be formulated in animal models to achieve a • - * -'" circulating concentration range that includes the IC 50 as determined in cell culmre (i. e. , the concentration of the test compound which achieves a half-
  • a therapeutically effective dose refers to that amount of the compound that results in amelioration of symptoms or a prolongation of survival in a patient.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell culmres or experimental animals, e.g. , for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD 50 and ED 50 .
  • Compounds which exhibit high therapeutic indices are preferred. The data obtained from these cell culmre assays and animal studies can be used in formulating a range of dosage for use in human.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. See, e.g. , Fingl et al. , 1975, in "The Pharmacological Basis of Therapeutics", Ch. 1 p. l .
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the C- proteinase inhibiting effects, or minimal effective concentration (MEC).
  • MEC minimal effective concentration
  • the MEC will vary for each compound but can be estimated from in vitro data; for example, the concentration necessary to achieve 50-90% inhibition of the C-proteinase using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using MEC value.
  • Compounds should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90% .
  • the effective local concentration of the drug may not be related to plasma concentration.
  • composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
  • compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may, for example, comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be * accompanied by instructions for administration.
  • Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labelled for treatment of an indicated condition. Suitable conditions indicated on the label may include treatment of arthritis or any other fibrotic disorder.
  • the compounds of the present invention may be synthesized according to known techniques. The following represent preferred methods for synthesizing and testing the compounds of the claimed invention:
  • Boc-Glu (OBn)OH and Boc-Asp(OBn)OH have been synthesized using methods readily known in the art.
  • reaction mixmre was allowed to warm to -30°C over 2h, then a 40% aquoeus solution of methyl amine (22 mmol) was add, and the reaction mixmre was allowed to warm to room temperamre. After the reaction had stirred for a further hour, diethyl ether (50 ml) and water (70 ml) were added. The organic layer was separated, washed with 1 M NaHCO 3 , 10% citric acid, and saturated NaCI solution, and dried over MgSO 4 . The solvent was evaporated in vacuo to give a white solid (5.0 g, 72% yield).
  • ester hydroxamate (28) also designated as FG-058, as a white solid.
  • Methoxybenzenesulfonyl)-L-proline hydroxamate also designated as FG054, (Example for Inhibitor A) is as follows:
  • the prefened method for synthesizing N- hydroxy-2-[[N'-(4-methoxybenzenesulfonyl)-N'-(4-methoxybenzyl)]amino]- acetamide (35), also designated as FG-067 (Example for Inhibitor A) is essentially as described for the synthesis of synthesizing N-hydroxy-2-[[N'-(4- methoxybenzenesulfonyl)-N-(4-chlorobenzyl)]amino]-acetamide, using as starting material 4-methoxybenzaldehyde instead of 4-chlorobenzaldehyde.
  • N-(4-Methoxybenzenesulfonyl)- ⁇ -benzyl-(L)-aspartic Acid (40).
  • b-benzyl-(L)-aspartic acid HCl salt (38) (2.00 g, 8.96 mmol)
  • p-methoxybenzenesulfonyl chloride (39) (1.76 g, 8.53 mmol)
  • triethylamine (1.81 g, 17.91 mmol) at room temperamre.
  • the reaction mixmre was quenched with 1 N HCl (60 ml), and extracted with CH 2 C1 2 (3 x 50 ml).
  • N-Benzyloxy-N'-(4-Methoxybenzenesulfonyl)- ⁇ -benzyl-(L)-aspartic amide (41).
  • N-(4-methoxybenzenesulfonyl)-b-benzyl- (L)-aspartic acid 300 mg, 0.76 mmol
  • O- benzylhydroxylamine/HCl in an 0 anhydride solution of (7/3) THF/DMF (10 ml) was added
  • N-hydroxybenzotriazole (HOBT) (103 mg, 0.76 mmol), N-ethy lmorpholine (204 mg, 1.68 mmol) and then diisopropylcarbodiimide (106 mg, 0.84 mmol) at room temperamre. After stirring over the weekend (2.5 days), the reaction mixmre was diluted with (1/1) hexanes/EtOAc (40 ml), washed successively 5 with 1 N HCl (2 x 20 ml), saturated NaHCO3 aqueous solution (2 x 20 mL) and brine. The organic layer was dried over MgSO 4 and concentrated.
  • HOBT N-hydroxybenzotriazole
  • the following assay may be used to determine the level of activity and effect of the different compounds of the present invention on C-proteinase activity.
  • radiolabeled ( 14 C) procollagen is added to 10 units/ml of chicken C-proteinase in a solution of 0.1 M Tris-HCI, 0.1 M NaCI, 0.02%
  • the protein bands are detected by autoradiography.
  • the amount of enzyme activity is based on the disappearance of the band conesponding to uncleaved procollagen.
  • the IC 50 of inhibitors can be determined by plotting the % activity versus inhibitor concentration and estimating the inhibitor concentration which results in 50% activity.
  • TABLE I IC S0 Of Various Identified C-Proteinase Inhibitors.
  • the IC 50 value of the inhibitors can also be determined by a filtration ELISA assay.
  • a filtration ELISA assay In this assay about 25 ng of unlabeled human procollagen I were incubated with the C-Proteinase as, see, section 6.2.1 but for one hour. The reaction was stopped with the addition of 40 ⁇ l precipitation buffer (0.5 X Reaction buffer, 0.1 mg/ml chicken collagen II, 10 ⁇ g/ml BSA, 7.5 mM EDTA). 25 ⁇ l of 75% ethanol was added and the reactions were mixed and incubated on ice for one hour to precipitate the procollagen.
  • 40 ⁇ l precipitation buffer 0.5 X Reaction buffer, 0.1 mg/ml chicken collagen II, 10 ⁇ g/ml BSA, 7.5 mM EDTA
  • the soluble c-propeptide was separated from the precipitated collagen by filtering through a Millipore multiscreen-HV 0.45 ⁇ m hydrophilic plate using a Millipore multiscreen vacuum manifold. 20 ⁇ l of the filtrate was removed and the amount of cleaved c-propeptide was determined by using the procollagen type I C-peptide (PIP) EIA kit from Takara Biomedicals.
  • PIP procollagen type I C-peptide
  • IC 50 of the inhibitors was determined by plotting the % activity * * ⁇ " versus inhibitor concentration and estimating the inhibitor concentration which gives 50 % activity. IC 50 values are shown in TABLE II
  • tissue culture assays by measurement of the production of procollagen and mature collagen in conditioned medium before and after treatment with a compound.
  • the ratio of collagen and procollagen will directly conelate to the cellular conversion of the precursor to the mature collagen product, and as such indicate the C-proteinase activity.
  • the media content of C-propeptide/cell may be determined, and compared for untreated cells and inhibitor-treated cells.
  • animal models which mimic clinical disorders related to unregulated or inappropriate collagen production are known in the art and may be employed to determine the in vivo efficacy of the compounds of the invention. These animal models include a wound chamber model in rats (Schilling et al., 1959, Surgery 46:702-710), an estradiol stimulated uterus expansion model (Mandell et al., 1982, The Journal of Biological Chemistry 257:5268-5273), and an induced angiogenesis model (Matrigel) (Passaniti et al, 1992, Laboratory Investigation 67:519-528).
  • liver fibrosis models include clinical disorder models like liver fibrosis models (Tsukamoto et al., 1990, Seminar in Liver Disease 10:56-65; Kock-Weser, 1952, Laboratory Investigation 1:324-331 ; Manione, 1949, American Journal of Pathology 25:273-285; Tarns, 1957, American Journal of Pathology 33:13-27; Wahl et ai, 1986, Journal of Experimental Medicine 163:884-902), a pulmonary fibrosis model (Kelly et ai, 1980, Journal of Laboratory Clinical Medicine 96:954-964), arterial restenosis models (Jackson, 1994, Trends of Cardiovascular Medicine 4:122-130; Clowes et al., 1983, Laboratory Investigation 49:327-333), a kidney fibrosis model (Yamamoto et al, 1987, Kidney International 3_2:514-525), a tendon repairing model (Franklin et al,
  • cytotoxicity assays are studied in cytotoxicity assays in order to determine whether there is an effect on cell survival or proliferation. These assays may involve the use of rapidly proliferating or quiescent cells. A known number of cells is seeded and exposed for increasing periods of time to a concentration range of potential inhibitors. Cell numbers are determined by cell counting or staining (e.g. Crystal Violet).
  • Cytotoxicity is evaluated as a function of cellular survival and cell proliferation.
  • Cellular survival involves the use of quiescent cells and is determined by cell number (counting or staining). A decrease in cell number indicates cell loss, and thus an effect on cell survival.
  • Cell proliferation involves the use of rapidly proliferating cells and is, as well, determined by cell number. Here a decrease in cell number relative to the untreated controls indicates an effect on cell proliferation.

Abstract

Utilisation nouvelle de molécules organiques capables d'inhiber l'activité de la C-protéinase afin de réguler, moduler et/ou prévenir une formation anormale de collagène.
EP96930499A 1995-08-08 1996-08-08 Inhibiteurs de la c-proteinase destines au traitement des affections liees a la surproduction de collagene Withdrawn EP0845987A4 (fr)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US203895P 1995-08-08 1995-08-08
US602038 1995-08-08
US60120396A 1996-02-14 1996-02-14
US601203 1996-02-14
US60918796A 1996-03-01 1996-03-01
US609187 1996-03-01
PCT/US1996/012876 WO1997005865A1 (fr) 1995-08-08 1996-08-08 Inhibiteurs de la c-proteinase destines au traitement des affections liees a la surproduction de collagene

Publications (2)

Publication Number Publication Date
EP0845987A1 true EP0845987A1 (fr) 1998-06-10
EP0845987A4 EP0845987A4 (fr) 2000-05-24

Family

ID=27357060

Family Applications (1)

Application Number Title Priority Date Filing Date
EP96930499A Withdrawn EP0845987A4 (fr) 1995-08-08 1996-08-08 Inhibiteurs de la c-proteinase destines au traitement des affections liees a la surproduction de collagene

Country Status (9)

Country Link
EP (1) EP0845987A4 (fr)
JP (1) JPH11511137A (fr)
KR (1) KR19990036271A (fr)
CN (1) CN1198096A (fr)
AU (1) AU6951296A (fr)
BR (1) BR9609883A (fr)
CA (1) CA2229098A1 (fr)
MX (1) MX9801093A (fr)
WO (1) WO1997005865A1 (fr)

Families Citing this family (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5863949A (en) * 1995-03-08 1999-01-26 Pfizer Inc Arylsulfonylamino hydroxamic acid derivatives
BR9610070A (pt) * 1995-08-08 1999-07-27 Univ Jefferson Sequência de ácido nucleico polipetídeos compostos antifibrótico e processo para tratar distúrbios ou doenças relacionadas com fibrose para usar a sequência de dna e para produzir c-proteinase recombinante
US5994351A (en) * 1998-07-27 1999-11-30 Pfizer Inc. Arylsulfonylamino hydroxamic acid derivatives
CN1230884A (zh) * 1996-07-22 1999-10-06 孟山都公司 硫羟砜金属蛋白酶抑制剂
US6747027B1 (en) 1996-07-22 2004-06-08 Pharmacia Corporation Thiol sulfonamide metalloprotease inhibitors
BR9711223A (pt) * 1996-08-23 1999-08-17 Pfizer Derivados de cido arilsulfonilamino-hidrox mico
ZA98376B (en) * 1997-01-23 1998-07-23 Hoffmann La Roche Sulfamide-metalloprotease inhibitors
US6376506B1 (en) 1997-01-23 2002-04-23 Syntex (U.S.A.) Llc Sulfamide-metalloprotease inhibitors
GB9706255D0 (en) 1997-03-26 1997-05-14 Smithkline Beecham Plc Novel compounds
US6037139A (en) * 1997-06-03 2000-03-14 Wisconsin Alumni Research Foundation System for assaying modulators of procollagen maturation
CN1198607C (zh) 1997-07-22 2005-04-27 盐野义制药株式会社 治疗或预防肾小球病的组合物
NZ503945A (en) * 1997-07-31 2002-11-26 Procter & Gamble Metalloprotease-inhibiting amide derivatives
US6130220A (en) * 1997-10-16 2000-10-10 Syntex (Usa) Inc. Sulfamide-metalloprotease inhibitors
US6107291A (en) * 1997-12-19 2000-08-22 Amgen Inc. Azepine or larger medium ring derivatives and methods of use
US6107337A (en) * 1998-08-06 2000-08-22 Pfizer Inc. Arylsulfonylamino hydroxamic acid derivatives
EP1137661A1 (fr) 1998-12-10 2001-10-04 F. Hoffmann-La Roche Ag Inhibiteurs de la c-proteinase de procollagene
CA2355902A1 (fr) * 1998-12-22 2000-06-29 F. Hoffmann-La Roche Ag Hydroxamates de sulfonamide
US6492394B1 (en) 1998-12-22 2002-12-10 Syntex (U.S.A.) Llc Sulfonamide hydroxamates
US6800646B1 (en) 1999-02-08 2004-10-05 Pharmacia Corporation Sulfamato hydroxamic acid metalloprotease inhibitor
US6506936B1 (en) * 1999-02-25 2003-01-14 Fibrogen, Inc. N-substituted arylsulfonylamino hydroxamic acids useful as inhibitors of c-proteinase and for treating or preventing disorders related to unregulated collagen production
CA2366954A1 (fr) 1999-03-03 2000-09-08 The Procter & Gamble Company Inhibiteurs de metalloproteases dihetero-substitues
TR200102523T2 (tr) 1999-03-03 2002-02-21 The Procter & Gamble Company Alkenil ve alkinil içeren metaloproteaz inhibitörleri
US6448278B2 (en) 1999-12-23 2002-09-10 Pfizer Inc. Procollagen C-proteinase inhibitors
US6462063B1 (en) 2000-02-04 2002-10-08 Fibrogen, Inc. C-proteinase inhibitors
US6645993B2 (en) 2001-03-30 2003-11-11 Warner-Lambert Company 3-heterocyclylpropanohydroxamic acid PCP inhibitors
MXPA04010555A (es) 2002-04-25 2005-02-17 Pharmacia Corp Acidos piperidinil- y piperazinil-sulfonilmetil hidroxamicos y su uso como inhibidores de proteasa.
DK1511472T3 (da) * 2002-05-29 2009-08-24 Merck & Co Inc Anvendelige forbindelser til behandling af Anthrax og hæmning af letalfaktor
US7105539B2 (en) 2002-12-03 2006-09-12 Enobia Pharma Derivatives of succinic and glutaric acids and analogs thereof useful as inhibitors of phex
EP1697309B1 (fr) 2003-12-19 2014-04-23 Basf Se Amides de phenylalanine a substitution benzoyle
AU2005250351A1 (en) 2004-05-11 2005-12-15 Merck & Co., Inc. Process for making n-sulfonated-amino acid derivatives
US8093236B2 (en) * 2007-03-13 2012-01-10 Takeda Pharmaceuticals Company Limited Weekly administration of dipeptidyl peptidase inhibitors
CA2686735A1 (fr) 2007-05-04 2008-11-13 Mdrna, Inc. Lipides d'acides amines et leurs utilisations
WO2009097893A1 (fr) * 2008-02-04 2009-08-13 Proyecto De Biomedicina Cima, S.L. Procédés de traitement d'une maladie cardiaque associée à une fibrose myocardique à l'aide d'un inhibiteur de pcp
CA3114240C (fr) 2018-10-30 2023-09-05 Gilead Sciences, Inc. Derives d'imidazopyridine utilises en tant qu'inhibiteurs de l'integrine alpha4beta7
CN112969504B (zh) 2018-10-30 2024-04-09 吉利德科学公司 用于抑制α4β7整合素的化合物
BR112021007213A2 (pt) 2018-10-30 2021-08-10 Gilead Sciences, Inc. derivados de quinolina como inibidores de integrina alfa4beta7
CA3115820A1 (fr) 2018-10-30 2020-05-07 Gilead Sciences, Inc. Composes pour l'inhibition de l'integrine .alpha.4.beta.7
AU2020329207B2 (en) 2019-08-14 2024-02-29 Gilead Sciences, Inc. Compounds for inhibition of alpha 4 beta 7 integrin

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4687841A (en) * 1985-10-18 1987-08-18 Monsanto Company Peptide hydroxamic acid derivatives
EP0262399A2 (fr) * 1986-08-29 1988-04-06 Takeda Chemical Industries, Ltd. Dérivés d'énols cycliques, préparation et utilisation
WO1991015507A1 (fr) * 1990-04-10 1991-10-17 Beecham Group Plc Phosphonopeptides inhibiteurs de collagenase
EP0606046A1 (fr) * 1993-01-06 1994-07-13 Ciba-Geigy Ag Arylsulfonamido-substitués dérivés d'acides hydroxamic

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4599361A (en) * 1985-09-10 1986-07-08 G. D. Searle & Co. Hydroxamic acid based collagenase inhibitors
US5292926A (en) * 1988-01-25 1994-03-08 Santen Pharmaceutical Co., Ltd. Cysteine derivatives
FR2626882B1 (fr) * 1988-02-08 1991-11-08 Ire Celltarg Sa Conjugues de derives de vinca comportant une chaine detergente en position c-3
NZ229004A (en) * 1988-05-19 1993-09-27 Immunobiology Res Inst Inc Tetrapeptides having t cell helper acitivity
FR2655339B2 (fr) * 1989-04-19 1992-04-10 Medgenix Group Sa Composes et complexes utiles notamment en imagerie medicale.
US5149794A (en) * 1990-11-01 1992-09-22 State Of Oregon Covalent lipid-drug conjugates for drug targeting
US5114953A (en) * 1990-11-21 1992-05-19 University Of Florida Treatment for tissue ulceration
US5443815A (en) * 1991-11-27 1995-08-22 Diatech, Inc. Technetium-99m labeled peptides for imaging
US5256657A (en) * 1991-08-19 1993-10-26 Sterling Winthrop, Inc. Succinamide derivative matrix-metalloprotease inhibitors
US5237057A (en) * 1992-04-06 1993-08-17 Biosite Diagnostics, Inc. Tetrahydrocannabinol derivatives and protein and polypeptide tetrahydrocannabinol derivative conjugates and labels
US5393902A (en) * 1994-04-26 1995-02-28 Lever Brothers Company, Division Of Conopco, Inc. Process for the preparation of bis(amidocarboxylic acids)

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4687841A (en) * 1985-10-18 1987-08-18 Monsanto Company Peptide hydroxamic acid derivatives
EP0262399A2 (fr) * 1986-08-29 1988-04-06 Takeda Chemical Industries, Ltd. Dérivés d'énols cycliques, préparation et utilisation
WO1991015507A1 (fr) * 1990-04-10 1991-10-17 Beecham Group Plc Phosphonopeptides inhibiteurs de collagenase
EP0606046A1 (fr) * 1993-01-06 1994-07-13 Ciba-Geigy Ag Arylsulfonamido-substitués dérivés d'acides hydroxamic

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BRINGMANN, GERHARD ET AL: "Acetogenic isoquinoline alkaloids. 66. First total synthesis of korupensamines A and B" HETEROCYCLES ( 1994 ), 39(2), 503-8 ,1994, XP000561812 *
CHEMICAL ABSTRACTS, vol. 110, no. 23, 5 June 1989 (1989-06-05) Columbus, Ohio, US; abstract no. 205068, LELIEVRE, Y. ET AL: "Synovial collagenase inhibition by actinonin. Structure-activity relations" XP002134086 & PATHOL. BIOL. ( 1989 ), 37(1), 43-6 , 1989, *
CHEMICAL ABSTRACTS, vol. 113, no. 17, 22 October 1990 (1990-10-22) Columbus, Ohio, US; abstract no. 147795, ODAKE, SHINJIRO ET AL: "Vertebrate collagenase inhibitor. I. Tripeptidyl hydroxamic acids" XP002134085 & CHEM. PHARM. BULL. ( 1990 ), 38(4), 1007-11 ,1990, *
CHEMICAL ABSTRACTS, vol. 116, no. 17, 27 April 1992 (1992-04-27) Columbus, Ohio, US; abstract no. 174753, PERICH, JOHN W. ET AL: "Synthesis of casein-related peptides and phosphopeptides. XVI. The efficient synthesis of the casein-related O-phosphoseryl-containing peptide Ac-Glu-Ser(P)-Leu-Ser(P)-Ser(P)-Ser(P)-Glu -Glu-NHMe" XP002134088 & AUST. J. CHEM. ( 1992 ), 45(2), 385-94 , 1992, *
CHEMICAL ABSTRACTS, vol. 123, no. 23, 4 December 1995 (1995-12-04) Columbus, Ohio, US; abstract no. 314466, OUCHI, T. ET AL: "Synthesis of polydepsipeptide with pendant carboxylic acid groups and its hydrolysis behavior" XP002134087 & STUD. POLYM. SCI. ( 1994 ), 12(BIODEGRADABLE PLASTICS AND POLYMERS), 528-33 ,1994, *
See also references of WO9705865A1 *

Also Published As

Publication number Publication date
EP0845987A4 (fr) 2000-05-24
AU6951296A (en) 1997-03-05
CN1198096A (zh) 1998-11-04
BR9609883A (pt) 1999-03-23
JPH11511137A (ja) 1999-09-28
CA2229098A1 (fr) 1997-02-20
WO1997005865A1 (fr) 1997-02-20
MX9801093A (es) 1998-04-30
KR19990036271A (ko) 1999-05-25

Similar Documents

Publication Publication Date Title
EP0845987A1 (fr) Inhibiteurs de la c-proteinase destines au traitement des affections liees a la surproduction de collagene
MXPA98001093A (en) Inhibitors of proteinase c for the treatment of diseases related to the overproduction of colag
US5639781A (en) Alcohol or aldehyde derivatives and their use
EP0358398B1 (fr) Glutaramides cycloalkyl substitués comme agents antihypertensifs
US4918105A (en) Novel compounds with collagenase-inhibiting activity, a process for their preparation and pharmaceutical compositions in which these compounds are present
US5442110A (en) Hydroxamic acid derivative
KR100432602B1 (ko) 매트릭스메탈로프로테아제억제제
JPH1160551A (ja) スルホニルアミノカルボン酸
KR20000015785A (ko) 페닐설폰아미드 유도체
JP2000515493A (ja) パラ―置換フェニレン誘導体
KR19980086893A (ko) 치환된 디아미노카복실산
WO1999017790A1 (fr) Inhibiteurs de cysteine et de serine proteases, a base d'alpha-cetoamides contenant des peptides
JP2844329B2 (ja) 選択的トロンビン抑制剤
US6506936B1 (en) N-substituted arylsulfonylamino hydroxamic acids useful as inhibitors of c-proteinase and for treating or preventing disorders related to unregulated collagen production
WO1997012903A1 (fr) Composes, compositions et procedes permettant d'inhiber la liaison de proteines contenant un domaine sh2 avec des proteines phosphorylees de meme origine
FI112660B (fi) Menetelmä matriksin metalloproteaasien inhibiittoreina käyttökelpoisten trisyklisten indolijohdannaisten valmistamiseksi
EP0343911A2 (fr) Glutaramide cycloalkyl substituée comme agent diurétique
WO1993014112A1 (fr) Derives substitues de peptidyle contenant de l'acide phosphinique, utilises comme agents antidegeneratifs
US6462063B1 (en) C-proteinase inhibitors
KR19980086989A (ko) 치환된 6- 및 7- 아미노-테트라하이드로이소퀴놀린 카복실산
RU2106356C1 (ru) Псевдопептиды или их соли, способы их получения, фармацевтическая композиция
PL198827B1 (pl) ω-Amidy N-arylosulfonyloaminokwasów, sposób ich wytwarzania, środek farmaceutyczny i zastosowanie ω-amidów N-arylosulfonyloaminokwasów
HU206194B (en) Process for producing cyclomethylene-1,2-dicarboxylic acid derivatives and pharmaceutical compositions comprising same
EP0712838B1 (fr) Derive d'acylphenylglycine et agent preventif et curatif, concernant des maladies causees par une activite collagenase accrue, contenant un tel compose comme ingredient actif
AU4973500A (en) Alpha-amino-beta-sulfonyl hydroxamic acid compounds

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19980306

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

RIC1 Information provided on ipc code assigned before grant

Free format text: 7A 61K 31/215 A, 7A 61K 31/38 B, 7A 61K 31/40 B, 7A 61K 31/66 B, 7A 61K 31/165 B, 7A 61K 31/70 B, 7A 61K 31/34 B, 7C 07C 259/06 B, 7C 07C 311/29 B, 7C 07D 207/48 B, 7C 07C 323/60 B, 7C 07K 5/02 B, 7C 07F 9/40 B

A4 Supplementary search report drawn up and despatched

Effective date: 20000411

AK Designated contracting states

Kind code of ref document: A4

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20000301