WO1997005865A1 - C-proteinase inhibitors for the treatment of disorders related to the overproduction of collagen - Google Patents

C-proteinase inhibitors for the treatment of disorders related to the overproduction of collagen Download PDF

Info

Publication number
WO1997005865A1
WO1997005865A1 PCT/US1996/012876 US9612876W WO9705865A1 WO 1997005865 A1 WO1997005865 A1 WO 1997005865A1 US 9612876 W US9612876 W US 9612876W WO 9705865 A1 WO9705865 A1 WO 9705865A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
mono
group
sulfonyl
compound
Prior art date
Application number
PCT/US1996/012876
Other languages
French (fr)
Inventor
Mitch Brenner
Wen-Bin Ho
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.
Priority to JP9508648A priority Critical patent/JPH11511137A/en
Priority to EP96930499A priority patent/EP0845987A4/en
Priority to BR9609883A priority patent/BR9609883A/en
Priority to MX9801093A priority patent/MX9801093A/en
Priority to AU69512/96A priority patent/AU6951296A/en
Publication of WO1997005865A1 publication Critical patent/WO1997005865A1/en

Links

Classifications

    • 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
    • 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
    • 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.
  • 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.
  • 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.
  • 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 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 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
  • 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;
  • 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.
  • 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.
  • 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
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • 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,
  • 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.
  • 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 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,
  • 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.
  • 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 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.
  • 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.
  • 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.
  • 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:
  • 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
  • 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 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

The present invention relates to the novel use of organic molecules capable of inhibiting C-proteinase activity in order to regulate, modulate and/or inhibit abnormal collagen formation.

Description

C-PROTEINASE INHIBITORS
FOR THE TREATMENT OF DISORDERS RELATED TO THE
OVERPRODUCTION OF COLLAGEN
This application is related to and is a continuation-in-part application of the U.S. application Serial Number 08/601,203, entitled "C-Proteinase Inhibitors For The Treatment Of Disorders Related To The Overproduction Of Collagen, " filed February 14, 1996, which is a continuation-in-part application of the provisional U.S. application Serial Number 60/002,038, filed August 8, 1995.
1. FIELD OF THE INVENTION
Collagen is integral to, among other things, the proper formation of connective tissue. Thus, the over- or under-production of collagen or the production of abnormal collagen (including incorrectly processed collagen) has been linked with numerous connective tissue diseases and disorders. Mounting evidence suggests that 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.
2. BACKGROUND OF THE INVENTION
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. Upon cleavage, 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. and Reddi, A.H.), Elsevier Science Publishing Co. , Inc. , New York, pp. 83-118; Prockop and Kivirikko, 1984, N. Engl. J. Med. 311:376-383; Kuhn, 1987, in: Structure and Function of Collagen Types (eds. Mayne, R. and Burgeson, R.E.), Academic Press, Inc. , Orlando, Florida, pp. 1-42.
Diseases Associated With The Abnormal Production of Collagen. 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, dermatofϊbroma, 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 (duchenne's), cardiac fibrosis, muscular fibrosis/retinal separation, esophageal stricture, payronles disease. Further 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. Thus, identification and isolation of molecules which control, inhibit and/or modulate the production of collagen are of major medical interest.
Relationship Between Collagen Formation and C-Proteinase. Recent evidence suggests that 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. Biochem. Biophys. 185:326-332; Leung et al. , 1979, J. Biol. Chem. 254:224-232). An acidic proteinase which removes the 5 C-terminal propeptides from type I procollagen has also been identified. Davidson et al. , 1979, Eur. J. Biochem. 100:551.
A partially purified protein having C-proteinase activity was obtained from chick calvaria in 1982. Njieha et al. , 1982, Biochemistry 23:757-764. In 1985, 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. Finally, the cDNA encoding human C-proteinase has been identified, as 5 set forth in the above-referenced related applications and references disclosed therein.
Experiments conducted with these purified forms of chick and mouse C-proteinase have indicated that the enzyme is instrumental in the formation of functional collagen fibers. Fertala et al. , 1994, J. Biol. Chem. 269: 11584. 0
C-Proteinase Inhibitors. As a consequence of the enzyme's apparent importance to collagen production, scientists have identified a number of C-
5 proteinase inhibitors. See e.g. , Hojima et al , supra. For example, several metal chelators have demonstrated activity as a C-proteinase inhibitor. Likewise, chymostatin and pepstatin A were found to be relatively strong inhibitors of C-proteinase. Additionally, α2-Macroglobulin, ovostatin, and fetal bovine serum appear to at least partially inhibit C-proteinase activity.
Dithiothreitol, SDS, concanavalin A, Zn2+, Cu2' , and Cd2+ are similarily reported to be inhibitory at low concentrations. Likewise, some reducing agents, several amino acids, phosphate, and ammonium sulfate were inhibitory at concentrations of 1-lOmM. Further, 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. Finally, high concentrations of NaCI or Tris-HCI buffer were found to inhibit C- proteinase's activity. For example, it is reported that, with 0.2, 0.3, and 0.5M NaCI, the activity of C-proteinase was reduced 66, 38, and 25%, respectively, of that observed with the standard assay concentration of 0.15M. Tris-HCI buffer in a concentration of 0.2-0.5M markedly inhibited activity. Hojima et al. , supra. In contrast, microbial inhibitors such as leupeptin, phosphoramidon, antipain, bestatin, elastinal, and amastatin, are considered to have weak or no effect on the activity of C-proteinase.
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.
Development Of Compounds To Inhibit C-Proteinase Activity. In view of its essential role in the formation and maturation of collagen, C- proteinase appears to be an ideal target for the treatment of disorders associated with the inappropriate or unregulated production and maturation of collagen. However, 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 identification of effective compounds which specifically inhibit the activity of C-proteinase to regulate and modulate abnormal or inappropriate collagen production is therefore desirable and the object of this invention.
3. SUMMARY OF THE INVENTION
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. Specifically, the compounds of the present invention have the formulae:
Inhibitor A
Figure imgf000007_0001
or:
Figure imgf000007_0002
wherein:
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;
R2 is selected from the group consisting of H, lower alkyl;
R3 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 lamino;
X is selected from the group consisting of SO2, C=O;
Y is selected from the group consisting of OH, HOHN(hydroxylamine), H2N, alkylamino; 2" Z is a direct bond; methylene, oxygen, sulf or, amino; n is 0 or 1 ;
25
30
35 or:
Inhibitor B
Figure imgf000009_0001
wherein:
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;
R2 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;
R3 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;
R4 is selected from the group consisting of H, lower alkyl; R5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl;
or:
c. Inhibitor C
Figure imgf000010_0001
wherein:
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;
R2 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;
R3 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; R5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufmyl or sulfonyl)alkyl, alkoyalkylacylalkyl;
or:
d. Inhibitor D
Figure imgf000011_0001
wherein: 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;
R2 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;
R3 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;
R4 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;
or:
Inhibitor E
Figure imgf000012_0001
wherein:
Rj is selected from the group consisting of OH, alkoxyl, lower alkyl, alkylamino, peptide;
X is selected from the group consisting of N, C;
R2 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;
R3 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;
R4 is selected from the group consisting of H, lower alkyl;
R5 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. *•**" More particularly, 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.
15 4. DEFINITIONS
"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
2U thereof having C-proteinase-like activity.
"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,
23 monoalkylamine, dibasic amino acids, sodium acetate, potassium benzoate, triethanol amine and the like.
"Alkyl" refers to a saturated aliphatic hydrocarbon, including straight- chain, branched-chain, and cyclic alkyl groups. Preferably, the alkyl group has 1 to 12 carbons. More preferably, it is a lower alkyl of from 1 to 7
3" carbons, more preferably 1 to 4 carbons. Typical alkyl groups includes methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl, hexyl
11
35 and the like. The alkyl group may be substituted or unsubstituted. When substituted 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. Preferably, the aryl is a substimted or unsubstituted phenyl or pyridyl. Preferred aryl substituent(s) preferably phenyl or pydridyl are halogen, trihalomethyl, hydroxyl, SH, NO2, amine, thioether, cyano, alkoxy, and groups.
5. DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to compounds capable of regulating and/or modulating collagen formation by inhibiting C-proteinase activity.
More particularly, 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.
5.1. The Compounds
The invention is generally directed to compounds and/or compositions comprising compounds having the formulae:
a. Inhibitor A
Figure imgf000014_0001
or:
Figure imgf000015_0001
wherein:
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;
R2 is selected from the group consisting of H, lower alkyl;
R3 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;
X is selected from the group consisting of SO2, C = O;
Y is selected from the group consistmg of OH, HOHN(hydroxylamine), H2N, alkylamino;
Z is a direct bond; methylene, oxygen, sulfor, amino; n is 0 or 1 ; or:
b. Inhibitor B
Figure imgf000016_0001
wherein:
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;
R2 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;
R3 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;
R5 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:
Inhibitor C
Figure imgf000017_0001
wherein:
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;
R2 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;
R3 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;
R5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl; or:
Inhibitor D
Figure imgf000018_0001
wherein:
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;
R2 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;
R3 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;
R4 is selected from the group consisting of H, lower alkyl;
R5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl- (thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl; or:
e. Inhibitor E
Figure imgf000019_0001
wherein:
R- is selected from the group consisting of OH, alkoxyl, lower alkyl, alkylamino, peptide;
X is selected from the group consisting of N, C;
R2 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;
R3 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;
R4 is selected from the group consisting of H, lower alkyl;
R5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl.
In specific embodiments of the invention, the compounds of the present invention may have the following formulae:
Figure imgf000020_0001
and pharmaceutically acceptable salts thereof;
or:
Figure imgf000020_0002
and pharmaceutically acceptable salts thereof;
or:
Figure imgf000021_0001
and pharmaceutically acceptable salts thereof;
or:
Figure imgf000021_0002
and pharmaceutically acceptable salts thereof;
or:
Figure imgf000021_0003
and pharmaceutically acceptable salts thereof; or:
Figure imgf000022_0001
and pharmaceutically acceptable salts thereof;
or:
Figure imgf000022_0002
and pharmaceutically acceptable salts thereof;
or:
Figure imgf000023_0001
and pharmaceutically acceptable salts thereof;
or:
Figure imgf000023_0002
and pharmaceutically acceptable salts thereof;
or:
Figure imgf000024_0001
and pharmaceutically acceptable salts thereof;
or:
Figure imgf000024_0002
and pharmaceutically acceptable salts thereof; or:
Figure imgf000025_0001
and pharmaceutically acceptable salts thereof;
15 or:
Figure imgf000025_0002
25 and pharmaceutically acceptable salts thereof.
The chemical formulae referred herein may exhibit the phenomenon of tautomerism. As the formulae drawing within this specification can only
J represent one of the possible tautomeric forms, it should be understood that the invention encompasses any tautomeric form which possesses the ability to
23
35 regulate and/or modulate the production and/or mamration of collagen by inhibiting C-proteinase activity. Further, the invention is understood to include all possible stereoisomers of each of the compounds described.
In addition to the above compounds and their pharmaceutically 3 acceptable salts, 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 ™ 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.
An individual compound's relevant activity and efficiency as an agent
* to affect C-proteinase activity may be determined using available techniques.
Preferentially, 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. These screens include biochemical assays, cell culmre assays and animal models. 20
5.2. Indications
Disorders associated with the inappropriate or unregulated production and/or mamration of collagen, including arthritic disorders, fibrotic disorders, and other connective tissue disorders, can be treated with the *J compounds and compositions of the present invention.
These 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
24
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. Further,
•*-* 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.
5.3. Pharmaceutical Formulations And Routes Of Administration
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. Techniques for formulation and administration of the compounds of the instant application may be found in "Remington's Pharmaceutical Sciences, " Mack Publishing
20 Co. , Easton, PA, latest edition.
5.3.1. Routes Of Administration.
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.
Alternately, one may administer the compound in 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. In order to prevent the scarring process frequently occurring as
25
35 complication of glaucoma surgery, the compounds may be administered topically, for example, as eye drops.
Furthermore, 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. The liposomes will be targeted to and taken up selectively by the afflicted tissue.
5.3.2. Composition/Formulation.
The pharmaceutical 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.
For injection, 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. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
For oral administration, 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
26
35 dragee cores. 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
3 carboxymethylcellulose, and/or poly viny lpyrrolidone (PVP). If desired, disintegrating agents may be added, 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
••**" arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
I*3 Pharmaceutical preparations 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,
stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration. For buccal administration,the compositions may take the form of
2*3 tablets or lozenges formulated in conventional manner.
For administration by inhalation, 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,
an
J / dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve
27
35 to deliver a metered amount. 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.
Pharmaceutical formulations 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
15 include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the
20 solubility of the compounds to allow for the preparation of highly concentrated solutions.
Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g. , sterile pyrogen-free water, before use.
25 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.
In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations
30 may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the
28
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.
3 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,
1" and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol. The VPD co-solvent system (VPD:5W) consists of 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. Namrally, the proportions of a co-solvent system may be
*3 varied considerably without destroying its solubility and toxicity characteristics. Furthermore, the identity of the 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.
2" polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.
Alternatively, other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs. Certain
23 organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity. Additionally, the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various of sustained-release materials have been established and are well known by those
3" skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days.
29 35 Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed.
The pharmaceutical compositions also may comprise suitable solid or
3 gel phase carriers or excipients. Examples of such carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
Many of the C-proteinase inhibiting compounds of the invention may
1" be provided as salts with pharmaceutically compatible counterions. 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,
* monoalkylamine, dibasic amino acids, sodium acetate, potassium benzoate, triethanol amine and the like.
5.3.3. Effective Dosage.
Pharmaceutical 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.
For any compound used in the method of the invention, the therapeutically effective dose can be estimated imtially from cell culmre assays. For example, a dose can be formulated in animal models to achieve a -*-'" circulating concentration range that includes the IC50 as determined in cell culmre (i. e. , the concentration of the test compound which achieves a half-
30
35 maximal inhibition of the C-proteinase activity). Such information can be used to more accurately determine useful doses in humans.
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 LD50 (the dose lethal to 50% of the population) and the ED50 (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 LD50 and ED50. 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 ED50 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). 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% .
In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration.
The amount of 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.
10 5.3.4. Packaging
The 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.
20
6. EXAMPLES
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:
25
6.1. Example 1: Compound Synthesis
6.1.1. Synthesis Of N-hydroxy-2-[[N'-(4- methoxybenzenesulfonyI)-N'-(4- chlorobenzyl)]amino]-acetamide
The preferred method for synthesizing N-hydroxy-2-
30
[[N'-(4-methoxybenzenesulfonyl)-N'(4-chlorobenzyl)]amino]-acetamide (5)
32
35 (specific compounds in this specification may be referred to, for nomenclamre purposes as "C#", wherein "#" is an arabic number), also designated as FG047, (Example for Inhibitor A) is as follows:
NaBCNH**-, ZaClZ fcOK
Figure imgf000035_0001
Figure imgf000035_0002
Figure imgf000035_0003
Figure imgf000035_0004
5 (FG (MT)
Synthesis Of Ethyl-2[(4-chlorobenτyl)amino]-acetate (3). To a cold solution of glycine ethyl ester hydrochloride (1) (1.00 g, 7.16mmol) and 4- chlorobenzaldehyde (2) (1.00 g, 7.16 mmol) in anhydrous MeOH (10 ml) was added anhydrous ZnCl2 solid (75 mg, 0.55 mmol) followed by NaBCNH3 solid (0.45 g, 7.16 mmol). After stirring at room temperamre for 18 h, the reaction mixmre was quenched with 1 N HCl (20 ml) aqueous solution, and stirred for another 30 min. The mixmre was concentrated on a rotary evaporator to remove most MeOH solvent and then extracted with ether (20 ml). The aqueous layer was carefully basified in an ice bath with 45% (w/w) of KOH aqueous solution to pH 10, and extracted with EtOAc (2 x 50 ml). The combined EtOAc organic layers was washed with brine, dried over Na2SO4, and concentrated. The crude product was purified by silica gel chromatography (4/1 hexanes/EtOAc) to get ethyl-2[(4-chlorobenzyl)amino]- acetate (3) as an oil.
Synthesis Of Ethyl-2-[[N-(4-methoxybenzene-sulfonyl)-N-(4- chlorobenz l)]amino]acetate (4). Triethylamine (294 mg, 2.90 mmol) was added dropwise to a solution of ethyl-2[(4-chlorobenzyl)amino]-acetate (3) (600 mg, 2.64 mmol) and p-methoxybenzenesulfonyl chloride (545 mg, 2.64 mmol) in anhydrous CH2C12 (7 ml). The mixmre was stirred at room temperamre for 15 h and treated with 1 N HCl (20 ml) solution. The resulting two phases were separated, and the aqueous layer was extracted with CH2C12. The combined orgamc layer was washed with brine, dried over MgSO4, and concentrated. The crude product was purified by silica gel chromatography (3/1 to 2/1 hexanes/EtOAc) to get ethyl-2-[[N-(4- methoxybenzene-sulfonyl)-N-(4-chlorobenzyl)]-amino]acetate (4) as an oil.
Synthesis Of N-hydroxy-2[[N'-(4-methoxy-benzenesulfonyl)-N'(4- chlorobenzyl)]amino]-acetamide (5). Separate solutions of hydroxylamine hydrochloride (171 mg, 2.46 mmol) in MeOH (1.3 ml) and of KOH (207 mg, 3.69 mmol) in MeOH (1.3 ml) were prepared at the boiling point, cooled to 40°C, and then the latter solution was added to the former. After cooling the reaction mixmre in an ice bath for 30 min, potassium chloride solid was filtered off. To the filtrate was added ethyl ester (4) (487 mg, 1.23 mmol). After stirring at room temperamre for 6 h, the reaction mixmre was treated with 1 N HCl (20 ml) solution, and extracted with CH2C12. The organic layer was washed with brine, dried over MgS04, and concentrated to get the crude product. The residue was triturated in ether and the white solid was collected to get the hydroxamate, i.e. , N-hydroxy-2[[N'-(4-methoxy-benzenesulfonyl)- N'(4-chlorobenzyl)] -amino] -acetamide (5), also designated as FG-047. mp: 124 - 125 °C; MS (ES) (M+H)": 385; IH NMR (360 MHz, DMSO-d6)<5 10.47 (s, 1 H, OH), 8.81 (s, 1 H, NH). 7.81 - 7.08 (m, 8 H, Ph), 4.34 (s, 2 H, CH2CO), 3.85 (s, 3 H, OMe), 3.63 (s, 2 H, CH2Ph).
6.1.2. Synthesis Of N-hydroxy-2-[[N'-(4- methoxybenzenesulfonyl)-N'-(4-fluorobenzyl)]amino]- acetamide
The preferred method for synthesizing N-hydroxy-2-
[ [N ' -(4-methoxybenzenesulf onyl)-N ' -(4-fluorobenzy 1)] amino] -acetamide (6) , also designated as FG053, (Example for Inhibitor A) is essentially as described for the synthesis of N-hydroxy-2-[[N'-(4-methoxybenzenesulfonyl)-
N'(4-chlorobenzyl)] amino] -acetamide, using as starting material 4- fluorobenzaldehyde instead of 4-chlorobenzaldehyde. See, Section 6.1.1.
Figure imgf000037_0001
6 (FG 053)
MS (ES) (M-l)":367; 'H NMR (360 MHz, DMSO-d6) δ 10.45 (s, 1 H, OH), 8.83 (s, 1 H, NH).7.81-7.08 (m, 8 H, Ph), 4.33 (s, 2 H, CH2CO), 3.85 (s, 3 H, OMe), 3.62 (s, 2 H, CH2Ph).
6.1.3. Synthesis Of Hydroxamate
The preferred method for synthesizing hydroxamate (11) (Example for Inhibitor B) is as follows: OEt BτCH2CQ2 Bu -' OEt NaH
Figure imgf000038_0001
9
Figure imgf000038_0002
Figure imgf000038_0003
Alkylation of malonic ester (7) with tert-butyl bromoacetate provides tetraester (8). Upon saponification and decarboxylation, the resulting aid (9) is coupled with Glu (OBn) NHMe using peptide coupling reaction to give (10). After subsequent hydrolysis, anhydride formation, and addition of NH2OH, (10) can be converted to the desired hydroxamate (11).
6.1.4. Synthesis Of N-Carboxymethyl Dipeptide
The preferred method for synthesizing N-carboxymethyl dipeptide (16), also designated as FG057, (Example for Inhibitor D) is as follows: COOBn
,COOBn 1. TFA
H.. OH H. NHMe 2. DCC/HOBt
N N
I
Boc O BOC O
12 13
Figure imgf000039_0001
14 15
Hj/Pd
Figure imgf000039_0002
16 (FG 057)
The building blocks (12) and (13) which are readily available by literature procedures, are coupled by the classical DCC/HOBt method leading to the dipeptide (14) in 78% yield. Deprotection of (14) using TFA in CH2C12 and subsequent alkylation with benzyl bromacetate in the presence of NMM resulted in the formation of the N-carboxymethyl dipeptide (15) , which was converted to the free acid (16) by catalytic hydrogenation.
Boc-Glu (OBn)OH and Boc-Asp(OBn)OH have been synthesized using methods readily known in the art. Synthesis Of Boc-Glu(OBn)NHMe (13): Triethylamine (2.0 g, 20 mmol) was added to a solution of Boc-Glu (OBn)OH (6.7 g, 20 mmol) in dry THF (100 ml). The solution was cooled to -78°C under argon atmosphere, then ethyl chloroformate (2.2 g, 20mmol) was added dropwise. The 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 NaHCO3, 10% citric acid, and saturated NaCI solution, and dried over MgSO4. The solvent was evaporated in vacuo to give a white solid (5.0 g, 72% yield).
'H-NMR (200 MHz, CDC13): δ 1.38 (s, 9H, CH3); 1.85 - 2.59 (m, 4H, CH2); 2.75 (d, 3H, NCH3); 4.17 (m, IH, CH); 5.08 (<5, 2H, OCH2); 5.33 (bδ, IH, NH); 6.30 (bs, IH, NH); 7.31 (s, 5H, Ph-H).
Synthesis Of Boc-Asp(OBn)-Glu(OBn)NHMe (14): To a solution of tert-butoxycarbonyl amino ester (13) (700 mg, 2 mmol) in 10 ml CH2C12 was added 1.5 ml TFA and the reaction mixmre was stined for 1 h at room temperamre under argon atmosphere. The excess acid was evaporated under vacuum, the residue was treated several times with diethyl ether and concentrated under reduced pressure, to give a colourless oil, which was without further purification. The TFA-salt, Boc-Asp(OBn)OH (446 mg, 2 mmol), HOBt (170 mg, 2 mmol) and NMM (202 mg, 2 mmol) in CH2C12 (10 ml) was added and the reaction was stirred overnight at room temperamre under argon atmosphere. The precipitate of dicyclohexylurea was removed by filtration and the filtrate was concentrated under reduced pressure. The residue was diluted with EtOAc (30 ml), filtrated again and washed with 1 M NaHC02, 10% citric acid, and saturated NaCI solution. The organic layer was dried over MgSO4, and concentrated in vacuo, to give a white solid in 78% yield (867 mg). 'H-NMR (200 MHz, CDC13): <5 1.41 (s, 9H, CH3); 1.85 - 3.05 (m, 9H, 3CH2, NCH3); 4.40 (m, 2H, CH); 5.01 (m, 4H, OCH2); 5.53 (d, IH, NH); 6.48 (bs, IH, NH); 7.15 - 7.38 (m, 10H, Ph-H).
3 Synthesis Of N-Carboxymethyl-Dipeptide (15): To a solution of Boc -
Asp(OBn)-Glu(OBn)NHMe (14) (555 mg, 1 mmol) in 10 ml CH2C12 was added 1 ml TFA and the reaction mixmre was stirred for 1 h at room temperamre under argon atmosphere. The excess acid was evaporated under vacuum, the residue was treated several times with diethyl ether and
*" concentrated under reduced pressure, to give a colourless oil, which was dissolved in dry THF (20 ml). To this solution was added NMM (101 mg, 1 mmol) and benzyl bromoacetate (230 mg, 1 mmol), and the reaction was stirred overnight at room temperamre under argon atmosphere. The reaction mixmre was concentrated under reduced pressure. The residue was diluted
15 with EtOAc (20 ml), and washed with 1 M NaHCO3, 10% citric acid, and saturated NaCI solution. The organic layer was dried over MgS02, and then concentrated to give a colourless oil, which was purified by flash chromatography on silica gel (ethyl acetate/MeOH 10:1) to afford (15) (410 mg) as a white solid in 88% yield.
20 'H-NMR (200 MHz, CDC13): ό 1.83 - 2.95 (m, 9H, 3CH2, NCH3);
3.30 - 3.60 (m, 4H, CH & N CH2); 4.39 (m, 2H, CH); 5.05 (m, 6H, OCH2); 6.64 (bs, IH, NH); 7.29 - (m, 15H, Ph-H). 8.02 (bs. -IH, NH); 13C-NMR (60 MHz, CDC13); δ 26.17, 27.24, 30.59, 36.57 (3CH2, NCH3); 49.59 (NCH,); 52.54, 59.07 (2CH); 66.46, 66.71, 66.86 (3OCH,); 128.17, 128.26,
25 128.34, 128.55, 128.80, 135.27, 135.44, 135.79 (Ph-C); 171.25, 171.34, 172.01 , 172.61, 173.05 (5C = O).
Synthesis Of N-Carboxymethyl-Asp-Glu-NMe (16): To a solution of benzyl-protected N-carboxymethyl dipeptide (15) (68 mg, 0.113 mmol) in
30 methanol (5 ml) was added portionwise Pd/C powder (50 mg). The mixmre was stirred under H2 atmosphere (balloon pressure) at room temperamre for
39
35 20 h. Catalyst was filtered of through a pad of celite and rinsed with methanol. The filtrate was concentrated and the crude solid was recrystallized from EtOAc/MeOH at -20 °C to afford the product N-Carboxymethyl-Asp- Glu-NMe (16) (23 mg, 0.065 mmol) as a white solid in 58 % yield. 5 mp: 147-149 °C; NMR (360 MHz, DMSO-d6) δ 8.19 (d, J = 8.5 Hz,
1 H), 7.72 (m, 1 H), 4.20 (m, 1 H), 3.45-3.20 (m, 4 H), 2.63-2.44 (m, 5 H), 2.20 (m, 2 H), 1.93 (m, 1 H), 1.72 (m, 1 H).
6.1.5. Synthesis Of Mercapto Compound
*" The prefened method for synthesizing mercapto compound (22), also designated as FG-O74 (Example for Inhibitor C) is as follows:
Figure imgf000042_0001
κ2co3 CH,0
Figure imgf000042_0002
1. Glπ(OBn)NHMc
2. DCC/HOBt
25
Figure imgf000042_0003
21 22
30 (FG-074)
40
35 Alkylation of the diethyl phosphate (17) with tert-butyl bromacetate in the presence of NaH provided the phosphonate (18) in 90 % yield. Horner- Emmons reaction of (18) with formaldehyde using K2CO3, as a base resulted in the formation of the unsaturated ester (19). Saponification of (19) with LIOH and subsequent Michael addition of thiolacetic acid provided the acid (20). Coupling of (20) with Glu(OBn)NHMe using the DCC/HOBt method afforded the dipeptide (21) as a mixmre of diastereomers by flash chromatography. Deprotection of (21) with TFA in CH2C12 provided the mono acid (22) .
Synthesis l-Ethyl-2-diethylphosphono-succinic acid 4-tert-butyl ester (18): To a suspension of NaH (0.48 g, 20 mmol) in dry THF (80 ml) was added dropwise a solution of triethyl phosphonoacetate (4.48 g, 20 mmol) in THF (20 ml) and after 3 h at room temperamre tert. -butyl bromoacetate in THF (20 ml). The resulting suspension was stured for 3 h at room temperamre, water (50 ml) was added and the reaction mixmre was acidified with 1 M HCl to pH 3. After addition of diethyl ether (70 ml), the organic was separated, washed with saturated NaCI solution, dried over MgSO4 and the solvent was evaporated in vacuo, to give a colourless oil in 90 % yield.
Synthesis Of l-Ethyl-2-methylene-succinic acid 4-tert. -butyl ester (19):
A mixmre of compound (18) (5.4 g, 16 mmol), K2CO3 (6.9 g, 50 mmol), and a 30% aqueous solution of formaldehyde (3.2 g, 100 mmol) was refluxed for 3 h. After cooling, the mixmre was extracted with hexane, the organic layer was washed with water and brine and dried over MgS04. After filtration and evaporation in vacuo, an oily residue was obtained in 80% yield (2.7 g).
'H-NMR (200 MHz, CDC13): δ 1.29 (t, j = 7 Hz, 3H, CH2); 1.43 (s, 9H, CH3); 3.24 (s, 2H, CH2; 4.20 (q, J = Hz, 2H, OCH2); 5.63, 6.28 (s, 2H, =CH2). Synthesis Of 2-Acetylsulfanylmethyl-succinic acid 4-tert. -butyl ester (20): The unsaturated ester (19) (2.3 g, 11 mmol) was dissolved in THF (50 ml) and a 0.2 M aqueous solution of LI OH (264 mg, 11 mmol) was added at 0°C. The mixture was allowed to come to room temperamre and was stined additional 3 h. The organic solvent was evaporated and the aqueous layer was extracted with diethyl ether (30 ml) . The aqueous layer was acidified with 1 M HCl to pH 3 and extracted with EtOAc. The organic layer was washed with water and brine and dried over MgS0 . After filtration and evaporation of the solvent, the acid was obtained as a white solid in 60% yield.
'H-NMR (200 MHz, CDC13); δ 1.44 (s, 9H, CH3); 3.26 (s, 2H, CH2); 5.78, 6.42 (s, 2H, =CH2); 10.01 (bs. IH, COOH).
The acid (1.0 g, 5.4 mmol) was dissolved in CHC13 and thioacetic acid (1.45 g, 19 mmol) was added. The mixmre was stirred at 60°C for 49 h. The solvent was evaporated in vacuo, and the compound (20) was obtained with a quantitative yield as a colourless oil.
'H-NMR (200 MHz, CDC13); δ 1.44 (s, 9H, CH3); 2.34 (s, 3H, CH3); 2.45 - 2.75 ( , 2H, CH2; 2.96 - 3.33 (m, 3H, CH2, CH); 9.74 (bs, IH, COOH).
Synthesis Of 3-acetyUhiomethyl-4-oxo-5-aza-6-(R)-methylcarbamoyl-8- benz loxycarbonyl-octanoic Acid, tert-butyl Ester (21): To a solution of tert - butoxycarbonyl amino ester (13) (700 mg, 2 mmol) in 10 ml CH2C12 was added 1 ml TFA and the reaction mixmre was stined for 1 h at room temperamre under argon atmosphere. The excess acid was evaporated under vacuum, the residue was treated several times with diethyl ether (15 ml) and concentrated under reduced pressure, to give a colourless oil, which was used without further purification. The TFA-salt, acid (20) (525 mg, 2 mmol) HOBt (170 mg, 2 mmol) and NMM (202 mg, 2 mmol) were dissolved in CH2C12 (40 ml). A solution of DCC (412 mg, 2 mmol) in CH2CH, (10 ml) was added and the reaction was stined overnight at room temperamre under argon atmosphere. The precipitate of dicyclohexylurea was removed by filtration and the filtrate was concentrated under reduced pressure. The residue was diluted with EtOAc (30 ml), filtrated again and washed with 1 M NaHC03, 10% citric acid, and saturated NaCI solution. The organic layer was dried over MgS04 and concentrated in vacuo, to give (21) as a colourless oil, which was purified by silica gel chromatography using EtOAc as eluant. The afforded yield was 72% (710 mg), as a mixmre of diastereomers. 'H- NMR (200 MHz, CDC13); δ 1.36, 1.39 (2s, 9H, CH3); 1.87 - 3.2 (m, 14H, 4CH3, CH3, NCH3); 4.40 (m, 2H, CH); 5.08, 5.11 (2s, 2H, OCH2); 6.43, 6.76, 7.07 (bs, -2H, NH); 6.76 (2d, IH, NH); 7.31 (m, 15H, Ph-H).
Synthesis of 3-Thiomethyl-4-oxo-5-aza-6-(R)-methylcarbamoyl-8- carboxy-octanoic Acid (22) (FG 074): To a solution of tert-butoxycarbonyl amino ester 21 (495 mg, 1 mmol) in 15 mL CH2CI2 was added TFA (1.5 mL) and the reaction mixmre was stined for lh at room temperamre under argon atmosphere. The solvent and excess acid were evaporated under vacuum, the residue was treated several times with diethyl ether (10 mL) and concentrated under reduced pressure, to give a colourless oil, which was purified by flash chromatography on silica gel (ethyl acetate/MeOH 10:1, containing 1 % acetic acid) to yield the intermediate as a white solid 84% (370mg) yield.
To a solution of the intermediate (30 mg, 0.07 mmol) in 1 mL of (1.5/1) methanol/H2O at room temperamre was added LiOH.H2O (12 mg, 0.27 mmol). After stining for 3 hours at room temperamre, the reaction mixmre was quenched with 0.5mL of 1 N HCI aqueous solution and extracted with EtOAc (2 x 10 mL). The combined organic layers were washed with brine, dried over MgSO , and concentrated to get the thio compound 22 as a gummy solid (19 mg, 0.06 mmol) in 86% yield.
MS (ES) (M + Y: 307
Figure imgf000046_0001
22 (FG-074)
6.1.6. Synthesis Of 2-[[N-(4-methoxybenzenesuIfonyl)-N-(4- chIorobenzyl)]amino]acetic Acid
The prefened method for synthesizing 2-[[N-(4- methoxybenzenesulfonyl)-N-(4-chlorobenzyl)]-amino]acetic acid (23), also designated as FG046, (Example for Inhibitor A) is as follows:
Figure imgf000046_0002
23 (FG 046)
Synthesis Of 2-[[N-(4-methoxybenzenesulfonyl)-N-(4- chlorobenzyl)]amino]acetic Acid (23): To a suspension mixmre of ethyl ester (4) (300 mg, 0.75 mmol) in 1.5: 1 MeOH/H20 (4ml) was added LiOH/H20. After stirring at room temperamre for 5 h, the mixmre was quenched with 1 N HCl solution (20 ml) and extracted with CH2C12 (2x20 ml). The combined organic layer was washed with brine, dried over MgS04, and concentrated. The crude solid was recrystallized from hot ether to afford (23) as a white solid. mp : 139.5-140°C; 'H NMR (360 MHz, DMSO-d6) 57.79 - 7.08 (m,4H,Ph), 4.37 (s,2 H,CH2), 3.85 (s, 3H, OCH3), 3.83 (s,2H, CH2).
6.1.7. Synthesis Of N-hydroxy-2[[N'-(4- methoxybenzenesulfonyl)- N' (carboxymethyI)]amino]acetamide
The prefened method for synthesizing N-hydroxy-2[[N'-
(4-methoxybenzenesulfonyl)-N'(carboxymethyl)]amino]acetamide (30), also designated as FG055, (Example for Inhibitor A) is as follows:
Figure imgf000047_0001
Synthesis Of Ethyl 2-[[N-(4-methoxybenzen-sulfonyl)]amino]acetate (26): To a mixmre of glycine ethyl ester hydrochloride (24) (3.0 g, 21.5 mmol) and 4-methoxybenzenesulfonyl chloride (25) (4.4 g, 21.3 mmol) in anhydrous CH2C12 (60 ml) was added triethylamine (4.79 g, 47.3 mmol). After stirring at room temperamre for 15 h, the reaction mixmre was quenched with 1 N HCl (120 ml) and extracted with CH2C12 (2x100 ml). The 3 combined organic layer was washed with brine, dried over MgSO4, and concentrated. The crude solid was recrystallized from EtOAc/hexanes to give sulfonamide (26) as a white solid.
Synthesis Of Ethyl 2-[[N-(4-methoxybenzene-sulfonyl)-N-tert- *•*■" butyloxycarbonylmethyl)]amino]acetate (27): To a slurry of sodium hydride (60% dispersed in mineral oil) (162 mg, 4.03 mmol) in anhydrous THF (10 ml) in an ice bath was added sulfonamide (26) (1.0 g, 3.66 mmol), followed by bromo tert-butylacetate (785 mg, 4.03 mmol). The mixmre was stined vigorously at 0°C for 30 min and then at rt for 15 h. After cooling the
15 reaction mixmre in an ice bath, it was quenched with water (25 ml) and extracted with ether (2x50 ml). The combined organic layer was washed with brine, dried over MgS04, and concentrated. The residue was purified by silica gel chromatography (2:1 hexanes: EtOAc) to give (27) as a colorless syrup.
20 Synthesis OfN-hydroxy-2[[N'-4-methoxy benzenesulf onyl)-N'-(tert- butyloxycarbonyl-methyl)]amino]acetamide (28): Separate solutions of hydroxylamine hydrochloride (377 mg, 5.43 mmol) in MeOH (2.7 ml) and of KOH (456 mg, 8.13 mmol) in MeOH (2.7 ml) were prepared at the boiling point, cooled to 40 °C, and then the latter solution was added to the former.
25 After cooling in an ice bath for 30 min, potassium chloride solid was filtered off. To the filtrate was added ethyl ester (27) (1.05 g, 2.71 mmol). After stirring at room temperamre for 6 h, the reaction mixmre was neutralized with 1 N HCl solution to pH 4, and partitioned between CH2C12(60 ml) and water (20 ml). The resulting two phases were separated and the aqueous layer was
30 extracted with CH2C12 (60 ml). The combined organic layer was washed with brine, dried over MgS04, and concentrated. The residue was purified by silica
46
35 gel chromatography (11 : 1 CH2Cl2:MeOH) to give ester hydroxamate (28) also designated as FG-058, as a white solid.
Synthesis Of N-hydroxy-2[[N'-(4-methoxybenzenesulfonyl)- N'(carboxymethyl)]-amino] acetamide (29): A solution of tert-butyl ester hydroxamate (28) (520 mg, mmol) in 35 % CF3COOH/CH2Cl2 (9 ml) was stirred at 0°C for 10 min and then at room temperamre for 1.5 h. The mixmre was concentrated and dried in vacuo. The residue was triturated in EtOAc and the solid was collected which was recrystallized from EtOAc/MeOH/hexanes to give acid hydroxamate (29) as a white solid. mp: 160-161 °C; MS (ES) (M + H)+ : 319; 'H NMR (360 MHz, DMSO-d6) δ 12.08 (brs, 1 H, CO2H), 10.69 (s, 1 H, OH), 8.96 (s, 1 H, NH), 7.76 (d, J = 8.7 Hz, 2 H, Ph), 7.09 (d, J = 8.7 Hz, 2 H, Ph), 4.01 (s, 2 H, CH2), 3.84 (s, 3 H, OMe), 3.83 (s, 2 H, CH2).
6.1.8. Synthesis Of N-(4-Methoxybenzene-sulfonyl)-L-proline Hydroxamate
The prefened method for synthesizing N-(4-
Methoxybenzenesulfonyl)-L-proline hydroxamate, also designated as FG054, (Example for Inhibitor A) is as follows:
Figure imgf000050_0001
30 31 32
Figure imgf000050_0002
33 (FG 05 )
Synthesis Of N-(4-Methoxybenzenesulfonyl)-L-Proline Methyl Ester To a solutio*(i3i-#:L-proline methyl ester hydrochloride (30) (1.00 g, 6.03 mmol) and 4-methoxybenzenesulfonyl chloride (31) (1.19 g, 5.75 mmol) in anhydrous CH2C12 (17 ml) was added triethylamine (1.22 g, 12.06 mmol). After stirring at room temperamre for 15 h, the reaction mixmre was quenched with 1 N HCl (30 ml) in aqueous solution and extracted with EtOAc (2 x 100 ml). The combined EtOAc organic layers was washed with brine, dried over Mg2SO , and concentrated to afford (32). This product was directly used for the next reaction without further purification.
Synthesis Of N-(4-Methoxybenzenesulfonyl)-L-Proline Hydroxamate (33): Separate solutions of hydroxylamine hydrochloride (465 mg, 6.68 mmol) in MeOH (3.4 ml) and of KOH (561 mg, 10.0 mmol) in MeOH (3.4 ml) were prepared at the boiling point, cooled to 40° C, and then the latter solution was added to the former. After cooling in an ice bath for 30 min, potassium chloride solid was filtered off. To the filtrate was added ethyl ester (32) (1.0 g, 3.34 mmol). After stining at room temperamre for 15 h, the reaction mixmre was treated with 1 N HCl (40 ml) solution, and extracted with CH2Cl2/MeOH (10: 1) The organic layer was washed with brine, dried over MgSO4, and concentrated to get the crude product. The crude solid was recrystalized from hot MeOH/EtOAc to afford the hydroxamate (33) as a solid.
6.1.9. Synthesis Of N-hydroxy-2-[[N'-(4- methoxybeπzenesulfonyl)-N'-(4- trifluoromethyIbenzyI)3amino]-acetamide
The prefened method for synthesizing N-hydroxy-2-
[[N'-(4-methoxybenzenesulfonyl)-N'-(4-trifluoromethylbenzyl)]amino]- acetamide (34), also designated as FG-066 (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-trifluoromethylbenzaldehyde instead of 4- chlorobenzaldehy de . See , Section 6.1.1., supra . lR NMR (360 MHz, DMSO-d6) δ 10.50 (s, 1 H, OH), 8.83 (s, 1 H,
NH). 7.81 - 7.08 (m, 8 H, Ph), 4.45 (s, 2 H, CH2CO), 3.86 (s, 3 H, OMe),
3.67 (s, 2 H, CH2Ph).
Figure imgf000051_0001
34 (FG-066) 6.1.10. Synthesis Of N-hydroxy-2-[[N'-(4- methoxybenzenesulfonyl)-N'-(4- methoxybenzyl)]amino]-acetamide
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.
See, Section 6.1.1. , supra.
Η NMR (360 MHz, DMSO-d6) δ 10.42 (s, 1 H, OH), 8.79 (s, 1 H,
NH). 7.81 - 6.86 (m, 8 H, Ph), 4.29 (s, 2 H, CH2CO), 3.85 (s, 3 H, OMe),
3. 73 (s, 3 H, OMe), 3.28 (s, 2 H, CH2Ph).
Figure imgf000052_0001
35 (FG-067)
6.1.11. Synthesis Of N-hydroxy-2-[[N'-(4- benzenesulf ony 1)-N ' -(4-chlorobenzyl)] amino] - acetamide
The prefened method for synthesizing N- hydroxy-2- [ [N ' -(4-benzenesulfony 1)-N ' -(4-chlorobenzy l)]amino] -acetamide (36), also designated as FG-080 (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 benzenesulfonyl chloride instead 4-methoxybenzenesulfonyl chloride. See, Section 6.1.1. , supra. MS (ES) (M + RX: 355.
Figure imgf000053_0001
36 (FG-080)
6.1.12. Synthesis Of N-hydroxy-2-[[N'-(4- methoxybenzenesulfonyl)-N'-(benzyI)]amino]- acetamide
The prefened method for synthesizing N- hydroxy-2-[[N'-(4-methoxybenzenesulfonyl)-N'-(benzyl)]amino]-acetamide (37), also designated as FG-061 (Example for Inhibitor A) is essentially as described for the synthesis of synthesizing N-hydroxy-2-[[N'-(4- methoxybenzenesulfonyl)-N'-(carboxymethyl)]amino]-acetamide, using as starting benzyl bromide instead of bromo tert-butylacetate. See, section 6.1.7. , supra.
MS (ES) (M - H) :349
Figure imgf000054_0001
37 (FG-061)
6.1.13. Synthesis Of N-(4-MethoxybenzenesuIfonyl)- /5-benzyl-(L)-Aspartic Acid
The prefened method for synthesizing N-(4-Methoxybenzenesulfonyl)-b-benzyl-(L)- Aspartic Acid (42), also designated as FG084, (Example for Inhibitor A) is as follows:
Synthesis of FG 084 (Inhibitor A)
Figure imgf000054_0002
38 39 40
Figure imgf000054_0003
FG 084
41 42
Inhibitor A
N-(4-Methoxybenzenesulfonyl)-β-benzyl-(L)-aspartic Acid (40). To a suspension mixmre of b-benzyl-(L)-aspartic acid HCl salt (38) (2.00 g, 8.96 mmol) and p-methoxybenzenesulfonyl chloride (39) (1.76 g, 8.53 mmol) in anhydrous CH2C1, was added triethylamine (1.81 g, 17.91 mmol) at room temperamre. After stirring for 15 hrs the reaction mixmre was quenched with 1 N HCl (60 ml), and extracted with CH2C12 (3 x 50 ml). The combined organic layers was washed with brine, dried over MgSO4, and concentrated to give N-(4- methoxybenzenesulfonyl)-b-benzyl-(L)-aspartic acid (3.14 g, 7.99 •*' mmol, 94% yield) as a gummy product.
N-Benzyloxy-N'-(4-Methoxybenzenesulfonyl)-β-benzyl-(L)-aspartic amide (41). To a mixmre of N-(4-methoxybenzenesulfonyl)-b-benzyl- (L)-aspartic acid (300 mg, 0.76 mmol) and 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 MgSO4 and concentrated. The residue was purified by silica gel flash chromatography ((1/1) EtOAc/hexanes) to get N-benzyloxy-NO-(4-methoxybenzenesulfonyl)- b-benzyl-(L)-aspartic amide (114 mg, 0.22 mmol, 30% yield) as a white solid, 0 mp: 128 - 129 .C; MS (ES) (M + H)+ : 499
N-Hydroxy-N'-(4-Methoxybenzenesulfonyl)-(L)-aspartic amide (42).
A mixmre of N-benzyloxy-NO-(4-methoxybenzenesulfonyl)- b-benzyl-(L)-aspartic amide (102 mg, 0.20 mmol) and 10% Pd/C (43 mg) in 5 methanol (7 ml) was stined vigorously in a hydrogen atmosphere (balloon pressure) for 20 hrs. The catalyst was filtered off through a pad of celite and the filtrate was concentrated. The residue was lyophilized from water to get N-hydroxy-NO-(4-methoxybenzenesulfonyl)-(L)-aspartic amide (50 mg, 0.16 mmol, 77% yield) as a hygroscopic fluffy powder. 0
53 5 IH NMR (360 MHz, DMSO-d6) δ 10.60 (s, 1 H), 8.78 (s, 1 H), 7.87 (d, J = 8.2 Hz, 1 H), 3.93 (m, 1 H), 3.82 (s, 3 H), 2.50 (m, 1 H), 2.21 (dd, J = 16.0, 6.5 Hz, 1 H).
6.2. Example 2: C-Proteinase Assays
6.2.1. In Vitro Assay For The Determination Of C-
Proteinase Activity And The IC^ Of Inhibitors
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.
About 125 μg radiolabeled (14C) 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%
Brij-35, and 5 mM CaCl2 in a total volume of 10 μl. The reaction is allowed to proceed for 15 minutes at 35° C and is stopped with one-half volume of 3x stop/loading buffer (30 mM EDTA, 30% glycerol, 6% SDS, 0.006%
Bromophenol-blue). Subsequently, the samples are heated to 100°C for 4 minutes, and resolved by SDS-PAGE (Novex) using 6% polyacryleamide gels.
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 IC50 of inhibitors can be determined by plotting the % activity versus inhibitor concentration and estimating the inhibitor concentration which results in 50% activity.
The IC50 value of the inhibitors which have been tested is shown in TABLE I. TABLE I: ICS0 Of Various Identified C-Proteinase Inhibitors.
Inhibitor Generic Group IC5,
FG-047 A 50μM
FG-061 A lOOμM
FG-053 A lOOμM
FG-0521 B 125μM
FG-066 A 150μM
FG-067 A 150μM
FG-086 (cbz-Pro-Leu-Gly-hydroxamate2) A 200μM
FG-087 (Ac -PYYG-hydroxamate) A 335μM
FG-088 (Actinonin3) B 350μM
FG-054 A 400μM
FG-057 D 1500μM
FG-0584 A 2100μM
FG-055 A 2600μM
FG-0515 C > > lOOμM
FG-046 A > > lOOOμM
1 commercially available from Peptides International (IHN-3850-PI).
2 commercially available from Sigma (C-8537).
3 commercially available from Sigma (A-6671).
4 intermediate compound (28), see, section 6.1.7
5 commercially available from Peptides International (ISN-3835-PI)
6.2.2. In Vitro ELISA Assay For The Determination Of C-
Proteinase Activity And The ICjo Of Inhibitors
The IC50 value of the inhibitors can also be determined by 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 μl precipitation buffer (0.5 X Reaction buffer, 0.1 mg/ml chicken collagen II, 10 μg/ml BSA, 7.5 mM EDTA). 25 μl of 75% ethanol was added and the reactions were mixed and incubated on ice for one hour to precipitate the procollagen. The soluble c-propeptide was separated from the precipitated collagen by filtering through a Millipore multiscreen-HV 0.45 μm hydrophilic plate using a Millipore multiscreen vacuum manifold. 20 μl 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.
For inhibition of hBMP-1 about 20 ng radiolabeled (I25I) human procollagen I was added to 1 to 2 μl of 5 times concentrated recombinant 3 hBMP-1 cell media (Kessler et al. (1996) Science 271 :360) in reaction buffer in a total volume of 10 μl. The reaction was let to proceed for one hour at 35 °C and then stopped with one-half volume of 3x stop/loading buffer and analyzed on SDS-PAGE as above.
The IC50 of the inhibitors was determined by plotting the % activity **" versus inhibitor concentration and estimating the inhibitor concentration which gives 50 % activity. IC50 values are shown in TABLE II
TABLE II:
IC50 Of Various Ide ennttiififieedd CC--PPiroteinase Inhibitors
As Determined by ELISA
15
Inhibitor c Group IC ELISA
FG-061 A 12μM FG-047 A 13μM FG-053 A 22μM FG-067 A 37μM
20 FG-066 A 48μM
6.2.3. Tissue Culture Assay For The Determination Of C- Proteinase Activity And The IC50 Of Inhibitors
C-proteinase activity and the IC50 of inhibitors in vivo
25 may be determined in 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.
30 Alternatively, the media content of C-propeptide/cell may be determined, and compared for untreated cells and inhibitor-treated cells.
56
35 6.3. Animal Models For The Determination Of C-Proteinase Activity And The Efficacy Of Inhibitors
Several 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). Further animal 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,
1986, The Journal of Laboratory and Clinical Medicine 108:103-108), a rumor growth model (Kiohs, et al, 1985, JNCL 75:353-359), a trabeculectomy model (Lahery et al, 1989, Journal of Ocular Pharmacology 5^:155-179), and an abdominal adhesions model (Williams et al, 1992, Journal of Surgical Research 52:65-70).
6.4. Example 4: Measurement Of Cytotoxicity
Potential inhibitors 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.
The present invention is not to be limited in scope by the exemplified embodiments which are intended as illustrations of single aspects of the invention, and any compounds and methods for the use thereof which are functionally equivalent are within the scope of the invention. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope of the appended claims.
All references cited herein are hereby incorporated by reference in their entirety.

Claims

CLAIMS What Is Claimed:
1. A compound having an inhibitory effect on C-proteinase, said compound having a formula selected from the group consisting of:
Figure imgf000061_0001
or:
Figure imgf000061_0002
and pharmaceutically acceptable salts thereof; wherein:
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;
R2 is selected from the group consisting of H, lower alkyl;
R3 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, sulfmyl or sulfonyl)-alkyl; R4 is selected from the group consisting of aryl, heteroaryl, alkyl, aralkyl, heteroaralkyl, alkylamino, arylalky lamino;
X is selected from the group consisting of S02, C=0; Y is selected from the group consisting of OH, HOHN(hydroxylamine). H2N, alkylamino;
Z is a direct bond; methylene, oxygen, sulfor, amino; and n is 0 or 1.
2. A compound having an inhibitory effect on C-proteinase, said " compound having a formula selected from the group consisting of:
Figure imgf000062_0001
and pharmaceutically acceptable salts thereof; wherein:
R, is selected from the group consisting of H, lower alkyl, mono- or 0 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; 5 R2 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 0 sulfonyl)-alkyl;
60 5 R3 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, sulf yl or sulfonyl)-alkyl; R, is selected from the group consisting of H, lower alkyl; and R5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl.
3. A compound having an inhibitory effect on C-proteinase, said compound having a formula selected from the group consisting of:
Figure imgf000063_0001
and pharmaceutically acceptable salts thereof; wherein:
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. sulfinyl or sulfonyl)-alkyl;
R2 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; R3 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; R4 is selected from the group consisting of H, lower alkyl; and R5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl.
4. A compound having an inhibitory effect on C-proteinase, said compound having a formula selected from the group consisting of:
Figure imgf000064_0001
and pharmaceutically acceptable salts thereof; wherein:
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;
R2 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; R3 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; and R5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl.
10
5. A compound having an inhibitory effect on C-proteinase, said compound having a formula selected from the group consisting of:
Figure imgf000065_0001
and pharmaceutically acceptable salts thereof; •*•*" wherein:
R, is selected from the group consisting of OH, alkoxyl, lower alkyl, alkylamino, peptide;
X is selected from the group consisting of N, C; R2 is selected from the group consisting of H, lower alkyl, mono- or 23 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; R3 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,
63
35 mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl. alkyl-(thio, sulf yl or sulfonyl)-alkyl; R4 is selected from the group consisting of H, lower alkyl; and R5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl.
6. The compound of Claim 1 wherein said compound has the formula:
Figure imgf000066_0001
and pharmaceutically acceptable salts thereof.
7. The compound of Claim 1 wherein said compound has the formula:
Figure imgf000066_0002
and pharmaceutically acceptable salts thereof.
The compound of Claim 1 wherein said compound has the formula:
Figure imgf000067_0001
and pharmaceutically acceptable salts thereof.
9. The compound of Claim 1 wherein said compound has the formula:
Figure imgf000067_0002
and pharmaceutically acceptable salts thereof.
10. The compound of Claim 1 wherein said compound has the formula:
Figure imgf000067_0003
and pharmaceutically acceptable salts thereof.
11. The compound of Claim 1 wherein said compound has the formula:
Figure imgf000068_0001
and pharmaceutically acceptable salts thereof.
12. The compound of Claim 1 wherein said compound has the formula:
Figure imgf000068_0002
and pharmaceutically acceptable salts thereof.
13. The compound of Claim 1 wherein said compound has the formula:
Figure imgf000068_0003
and pharmaceutically acceptable salts thereof.
14. The compound of Claim 1 wherein said compound has the formula:
Figure imgf000069_0001
and pharmaceutically acceptable salts thereof.
*"*" 15. The compound of Claim 1 wherein said compound has the formula:
15
Figure imgf000069_0002
and pharmaceutically acceptable salts thereof;
25 16. The compound of Claim 1 wherein said compound has the formula:
30
67
35
Figure imgf000070_0001
and pharmaceutically acceptable salts thereof;
17. The compound of Claim 3 wherein said compound has the formula:
Figure imgf000070_0002
and pharmaceutically acceptable salts thereof.
18. The compound of Claim 3 wherein said compound has the formula:
Figure imgf000070_0003
and pharmaceutically acceptable salts thereof.
19. A pharmaceutical composition comprising a compound selected from the group consisting of:
Figure imgf000071_0001
or:
Figure imgf000071_0002
wherein:
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;
R2 is selected from the group consisting of H, lower alkyl;
R3 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;
R4 is selected from the group consisting of aryl, heteroaryl, alkyl, aralkyl, heteroaralkyl, alkylamino, arylalky lamino;
X is selected from the group consisting of S02, C=0; Y is selected from the group consisting of OH, HOHN(hydroxylamine), H2N, alkylamino;
Z is a direct bond; methylene, oxygen, sulfor, amino; and n is 0 or 1 ; and a pharmaceutically acceptable carrier or excipient;
or:
Figure imgf000072_0001
* wherein:
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, sulfmyl or sulfonyl)-alkyl;
R2 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, 3 (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfinyl or sulfonyl)-alkyl;
R3 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl, 30 biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino,
70
35 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; and R5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufmyl or sulfonyl)alkyl, alkoyalkylacylalkyl; and a pharmaceutically acceptable canier or excipient;
or:
Figure imgf000073_0001
wherein:
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, sulfinyl or sulfonyl)-alkyl;
R2 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;
R3 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; and Rj is selected from the group consistmg of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl;
and a pharmaceutically acceptable earner or excipient;
or:
Figure imgf000074_0001
wherein: *** 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, sulfinyl or sulfonyl)-alkyl; •ώυ R2 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;
R3 is selected from the group consistmg 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; and
72
35 R5 is selected from the group consisting of H, lower alkyl, carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(fhio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl;
and a pharmaceutically acceptable carrier or excipient;
or:
Figure imgf000075_0001
wherein: " R, is selected from the group consisting of OH, alkoxyl, lower alkyl, alkylamino, peptide;
X is selected from the group consisting of N, C;
R2 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl,
^ 20 biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfmyl or sulfonyl)-alkyl; R3 is selected from the group consisting of H, lower alkyl, mono- or poly-haloalkyl, carboxyalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, biaryl,
25 biarylalkyl, hydroxyalkyl, alkyoxyalkyl, acyloxyalkyl, mercaptoalkyl, (amino, mono- or dialkylamino)alkyl, acylaminoalkyl, cycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, alkyl-(thio, sulfinyl or sulfonyl )-alkyl; R4 is selected from the group consisting of H, lower alkyl; and R5 is selected from the group consisting of H, lower alkyl,
30 carboxyalkyl, (mono- or dialkylamino)alkyl, alkyl-(thio, sufinyl or sulfonyl)alkyl, alkoyalkylacylalkyl;
73
35 and a pharmaceutically acceptable carrier or excipient.
20. A pharmaceutical composition comprising a compound selected from the group consisting of:
Figure imgf000076_0001
and a pharmaceutically acceptable carrier or excipient;
or:
Figure imgf000076_0002
and a pharmaceutically acceptable carrier or excipient;
or:
Figure imgf000077_0001
and a pharmaceutically acceptable canier or excipient;
or:
Figure imgf000077_0002
HOHN ^O
and a pharmaceutically acceptable canier or excipient; or:
Figure imgf000077_0003
and a pharmaceutically acceptable canier or excipient; or:
Figure imgf000078_0001
and a pharmaceutically acceptable canier or excipient;
or:
Figure imgf000078_0002
and a pharmaceutically acceptable carrier or excipient;
or:
Figure imgf000078_0003
and a pharmaceutically acceptable carrier or excipient; or:
and a pharmaceutically acceptable canier or excipient;
or:
Figure imgf000079_0002
and a pharmaceutically acceptable canier or excipient;
or:
Figure imgf000080_0001
and a pharmaceutically acceptable canier or excipient;
or:
Figure imgf000080_0002
and a pharmaceutically acceptable carrier or excipient; or:
Figure imgf000081_0001
and a pharmaceutically acceptable canier or excipient;
10
21. A method for treating diseases related to the inappropriate or unregulated production of collagen comprising administration of an effective amount of the composition according to Claim 20.
13 22. A method for treating diseases related to the inappropriate or umegulated production of collagen comprising administration of an effective amount of the composition according to Claim 21.
23. The method of Claim 22 wherein the fibrotic disorder is selected
260υ from the group consisting of hepatic cinhosis and arthritis.
24. The method of Claim 23 wherein the fibrotic disorder is selected from the group consisting of hepatic cinhosis and arthritis.
25
30
79
35
PCT/US1996/012876 1995-08-08 1996-08-08 C-proteinase inhibitors for the treatment of disorders related to the overproduction of collagen WO1997005865A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
JP9508648A JPH11511137A (en) 1995-08-08 1996-08-08 C-proteinase inhibitors for treating diseases associated with collagen overproduction
EP96930499A EP0845987A4 (en) 1995-08-08 1996-08-08 C-proteinase inhibitors for the treatment of disorders related to the overproduction of collagen
BR9609883A BR9609883A (en) 1995-08-08 1996-08-08 Compound having inhibitory effect on proteinase and pharmaceutical composition and process to treat diseases related to inappropriate or unregulated collagen production
MX9801093A MX9801093A (en) 1995-08-08 1996-08-08 C-proteinase inhibitors for the treatment of disorders related to the overproduction of collagen.
AU69512/96A AU6951296A (en) 1995-08-08 1996-08-08 C-proteinase inhibitors for the treatment of disorders related to the overproduction of collagen

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US203895P 1995-08-08 1995-08-08
US60/002,038 1995-08-08
US60120396A 1996-02-14 1996-02-14
US601,203 1996-02-14
US60918796A 1996-03-01 1996-03-01
US609,187 1996-03-01

Publications (1)

Publication Number Publication Date
WO1997005865A1 true WO1997005865A1 (en) 1997-02-20

Family

ID=27357060

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/012876 WO1997005865A1 (en) 1995-08-08 1996-08-08 C-proteinase inhibitors for the treatment of disorders related to the overproduction of collagen

Country Status (9)

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

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998042659A2 (en) * 1997-03-26 1998-10-01 Smithkline Beecham Plc Aryl- or heteroarylsulfonamide substituted hydroxamic acid derivates, process for their preparation and their use as pharmaceuticals
EP0871710A1 (en) * 1995-08-08 1998-10-21 Thomas Jefferson University Recombinant c-proteinase and processes, methods and uses thereof
WO1998055862A1 (en) * 1997-06-03 1998-12-10 Wisconsin Alumni Research Foundation System for assaying modulators of procollagen maturation
US5863949A (en) * 1995-03-08 1999-01-26 Pfizer Inc Arylsulfonylamino hydroxamic acid derivatives
WO1999006340A2 (en) * 1997-07-31 1999-02-11 The Procter & Gamble Company Sulfonylamino substituted hydroxamic acid derivatives as metalloprotease inhibitors
WO1999032451A1 (en) * 1997-12-19 1999-07-01 Amgen Inc. Azepine or larger medium ring derivatives and their use as pharmaceuticals
US5994351A (en) * 1998-07-27 1999-11-30 Pfizer Inc. Arylsulfonylamino hydroxamic acid derivatives
US5998412A (en) * 1997-01-23 1999-12-07 Syntex (U.S.A.) Inc. Sulfamide-metalloprotease inhibitors
US6013649A (en) * 1996-07-22 2000-01-11 Monsanto Company Thiol sulfone metalloprotease inhibitors
WO2000037436A1 (en) * 1998-12-22 2000-06-29 F. Hoffmann-La Roche Ag Sulfonamide hydroxamates
US6107337A (en) * 1998-08-06 2000-08-22 Pfizer Inc. Arylsulfonylamino hydroxamic acid derivatives
US6130220A (en) * 1997-10-16 2000-10-10 Syntex (Usa) Inc. Sulfamide-metalloprotease inhibitors
US6153609A (en) * 1996-08-23 2000-11-28 Pfizer Inc Arylsulfonylamino hydroxamic acid derivatives
US6197770B1 (en) 1999-03-03 2001-03-06 The Procter & Gamble Co. Alkenyl- and alkynl-containing metalloprotease inhibitors
EP1154992A1 (en) * 1999-02-25 2001-11-21 Fibrogen, Inc. N-substituted arylsulfonylamino hydroxamic acids useful as inhibitors of c-proteinase and for treating or preventing disorders related to unregulated collagen production
US6376506B1 (en) 1997-01-23 2002-04-23 Syntex (U.S.A.) Llc Sulfamide-metalloprotease inhibitors
US6423729B1 (en) 1997-07-22 2002-07-23 Shionogi & Co., Ltd. Therapeutic or prophylactic agent for glomerulopathy
US6426402B1 (en) 1998-12-10 2002-07-30 Syntex (U.S.A.) Llc Peptidic procollagen C-proteinase inhibitors
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
US6492394B1 (en) 1998-12-22 2002-12-10 Syntex (U.S.A.) Llc Sulfonamide hydroxamates
US6566381B1 (en) 1999-03-03 2003-05-20 The Procter & Gamble Company Hetero-substituted metalloprotease inhibitors
US6645993B2 (en) 2001-03-30 2003-11-11 Warner-Lambert Company 3-heterocyclylpropanohydroxamic acid PCP inhibitors
US6747027B1 (en) 1996-07-22 2004-06-08 Pharmacia Corporation Thiol sulfonamide metalloprotease inhibitors
WO2004050620A2 (en) * 2002-12-03 2004-06-17 Enobia Pharma Derivatives of succinic and glutaric acids and analogs thereof useful as inhibitors of phex
US6800646B1 (en) 1999-02-08 2004-10-05 Pharmacia Corporation Sulfamato hydroxamic acid metalloprotease inhibitor
EP1511472A2 (en) * 2002-05-29 2005-03-09 Merck & Co., Inc. Compounds useful in the treatment of anthrax and inhibiting lethal factor
WO2005061443A2 (en) 2003-12-19 2005-07-07 Basf Aktiengesellschaft Benzoyl-substituted phenylalanine amides
US7119203B2 (en) 2002-04-25 2006-10-10 Pharmacia Corporation Piperidinyl- and piperazinyl-sulfonylmethyl hydroxamic acids and their use as protease inhibitors
WO2009097893A1 (en) * 2008-02-04 2009-08-13 Proyecto De Biomedicina Cima, S.L. Methods for the treatment of cardiac disease associated to myocardial fibrosis using an inhibitor of pcp
US7579487B2 (en) 2004-05-11 2009-08-25 Merck & Co., Inc. Process for making N-sulfonated-amino acid derivatives
US7939505B2 (en) 2007-05-04 2011-05-10 Marina Biotech, Inc. Amino acid lipids and uses thereof
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

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8093236B2 (en) * 2007-03-13 2012-01-10 Takeda Pharmaceuticals Company Limited Weekly administration of dipeptidyl peptidase inhibitors

Citations (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
US5024835A (en) * 1988-02-08 1991-06-18 Ire-Celltarg S.A. Conjugates of a vinca derivative carrying a detergent chain in the C-3 position
US5114953A (en) * 1990-11-21 1992-05-19 University Of Florida Treatment for tissue ulceration
US5149794A (en) * 1990-11-01 1992-09-22 State Of Oregon Covalent lipid-drug conjugates for drug targeting
US5237057A (en) * 1992-04-06 1993-08-17 Biosite Diagnostics, Inc. Tetrahydrocannabinol derivatives and protein and polypeptide tetrahydrocannabinol derivative conjugates and labels
US5256657A (en) * 1991-08-19 1993-10-26 Sterling Winthrop, Inc. Succinamide derivative matrix-metalloprotease inhibitors
US5276147A (en) * 1989-04-19 1994-01-04 Medgenix Group S.A. Compounds and complexes useful in medical imaging
US5292926A (en) * 1988-01-25 1994-03-08 Santen Pharmaceutical Co., Ltd. Cysteine derivatives
US5298490A (en) * 1988-05-19 1994-03-29 Immunobiology Research Institute, Inc. Tetra and penta-peptides useful in regulating the immune system
US5393902A (en) * 1994-04-26 1995-02-28 Lever Brothers Company, Division Of Conopco, Inc. Process for the preparation of bis(amidocarboxylic acids)
US5443815A (en) * 1991-11-27 1995-08-22 Diatech, Inc. Technetium-99m labeled peptides for imaging

Family Cites Families (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
US4925868A (en) * 1986-08-29 1990-05-15 Takeda Chemical Industries, Ltd. 4-Hydroxy-3-pyrrolin-2-ones and treatment of circulatory disorders therewith
GB9008078D0 (en) * 1990-04-10 1990-06-06 Beecham Group Plc Novel compounds
US5455258A (en) * 1993-01-06 1995-10-03 Ciba-Geigy Corporation Arylsulfonamido-substituted hydroxamic acids

Patent Citations (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
US5024835A (en) * 1988-02-08 1991-06-18 Ire-Celltarg S.A. Conjugates of a vinca derivative carrying a detergent chain in the C-3 position
US5298490A (en) * 1988-05-19 1994-03-29 Immunobiology Research Institute, Inc. Tetra and penta-peptides useful in regulating the immune system
US5276147A (en) * 1989-04-19 1994-01-04 Medgenix Group S.A. Compounds and complexes useful in medical imaging
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
US5256657A (en) * 1991-08-19 1993-10-26 Sterling Winthrop, Inc. Succinamide derivative matrix-metalloprotease inhibitors
US5443815A (en) * 1991-11-27 1995-08-22 Diatech, Inc. Technetium-99m labeled peptides for imaging
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)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP0845987A4 *

Cited By (68)

* 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
EP0871710A1 (en) * 1995-08-08 1998-10-21 Thomas Jefferson University Recombinant c-proteinase and processes, methods and uses thereof
EP0871710A4 (en) * 1995-08-08 2001-03-07 Univ Jefferson Recombinant c-proteinase and processes, methods and uses thereof
US6747027B1 (en) 1996-07-22 2004-06-08 Pharmacia Corporation Thiol sulfonamide metalloprotease inhibitors
US6013649A (en) * 1996-07-22 2000-01-11 Monsanto Company Thiol sulfone metalloprotease inhibitors
US6153609A (en) * 1996-08-23 2000-11-28 Pfizer Inc Arylsulfonylamino hydroxamic acid derivatives
US5998412A (en) * 1997-01-23 1999-12-07 Syntex (U.S.A.) Inc. Sulfamide-metalloprotease inhibitors
US6143744A (en) * 1997-01-23 2000-11-07 Syntex (U.S.A.) Inc. Sulfamide-metalloprotease inhibitors
US6376506B1 (en) 1997-01-23 2002-04-23 Syntex (U.S.A.) Llc Sulfamide-metalloprotease inhibitors
WO1998042659A3 (en) * 1997-03-26 1999-02-25 Smithkline Beecham Plc Aryl- or heteroarylsulfonamide substituted hydroxamic acid derivates, process for their preparation and their use as pharmaceuticals
WO1998042659A2 (en) * 1997-03-26 1998-10-01 Smithkline Beecham Plc Aryl- or heteroarylsulfonamide substituted hydroxamic acid derivates, process for their preparation and their use as pharmaceuticals
US6242467B1 (en) 1997-03-26 2001-06-05 Smithkline Beecham P.L.C. Compounds
US6037139A (en) * 1997-06-03 2000-03-14 Wisconsin Alumni Research Foundation System for assaying modulators of procollagen maturation
WO1998055862A1 (en) * 1997-06-03 1998-12-10 Wisconsin Alumni Research Foundation System for assaying modulators of procollagen maturation
US6423729B1 (en) 1997-07-22 2002-07-23 Shionogi & Co., Ltd. Therapeutic or prophylactic agent for glomerulopathy
US6878739B2 (en) 1997-07-22 2005-04-12 Shionogi & Co., Ltd. Composition for treating or preventing glomerulopathy
WO1999006340A3 (en) * 1997-07-31 1999-09-30 Procter & Gamble Sulfonylamino substituted hydroxamic acid derivatives as metalloprotease inhibitors
WO1999006340A2 (en) * 1997-07-31 1999-02-11 The Procter & Gamble Company Sulfonylamino substituted hydroxamic acid derivatives as metalloprotease inhibitors
US6218389B1 (en) 1997-07-31 2001-04-17 The Procter & Gamble Co. Acyclic metalloprotease inhibitors
US6130220A (en) * 1997-10-16 2000-10-10 Syntex (Usa) Inc. Sulfamide-metalloprotease inhibitors
WO1999032451A1 (en) * 1997-12-19 1999-07-01 Amgen Inc. Azepine or larger medium ring derivatives and their use as pharmaceuticals
US6107291A (en) * 1997-12-19 2000-08-22 Amgen Inc. Azepine or larger medium ring derivatives and methods of use
US6147074A (en) * 1998-07-27 2000-11-14 Pfizer Inc Arylsulfonylamino hydroxamic acid derivatives
US5994351A (en) * 1998-07-27 1999-11-30 Pfizer Inc. Arylsulfonylamino hydroxamic acid derivatives
US6107337A (en) * 1998-08-06 2000-08-22 Pfizer Inc. Arylsulfonylamino hydroxamic acid derivatives
US6951918B2 (en) 1998-12-10 2005-10-04 Syntex (U.S.A.) Llc Peptidic procollagen C-proteinase inhibitors
US6426402B1 (en) 1998-12-10 2002-07-30 Syntex (U.S.A.) Llc Peptidic procollagen C-proteinase inhibitors
AU769319B2 (en) * 1998-12-22 2004-01-22 F. Hoffmann-La Roche Ag Sulfonamide hydroxamates
US6492394B1 (en) 1998-12-22 2002-12-10 Syntex (U.S.A.) Llc Sulfonamide hydroxamates
WO2000037436A1 (en) * 1998-12-22 2000-06-29 F. Hoffmann-La Roche Ag Sulfonamide hydroxamates
US6844366B2 (en) 1998-12-22 2005-01-18 Syntex (U.S.A.) Llc Sulfonamide hydroxamates
US6787559B2 (en) 1998-12-22 2004-09-07 Syntex (U.S.A.) Llc. Sulfonamide compounds
US6800646B1 (en) 1999-02-08 2004-10-05 Pharmacia Corporation Sulfamato hydroxamic acid metalloprotease inhibitor
US7067670B2 (en) 1999-02-08 2006-06-27 Warner Lambert Company 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
EP1154992A4 (en) * 1999-02-25 2003-05-28 Fibrogen Inc N-substituted arylsulfonylamino hydroxamic acids useful as inhibitors of c-proteinase and for treating or preventing disorders related to unregulated collagen production
EP1154992A1 (en) * 1999-02-25 2001-11-21 Fibrogen, Inc. N-substituted arylsulfonylamino hydroxamic acids useful as inhibitors of c-proteinase and for treating or preventing disorders related to unregulated collagen production
US6566381B1 (en) 1999-03-03 2003-05-20 The Procter & Gamble Company Hetero-substituted metalloprotease inhibitors
US6197770B1 (en) 1999-03-03 2001-03-06 The Procter & Gamble Co. Alkenyl- and alkynl-containing metalloprotease inhibitors
US6762198B2 (en) 1999-03-03 2004-07-13 The Procter & Gamble Company Dihetero-substituted metalloprotease inhibitors
US6448278B2 (en) 1999-12-23 2002-09-10 Pfizer Inc. Procollagen C-proteinase inhibitors
US6897306B2 (en) 1999-12-23 2005-05-24 Pfizer Products, Inc. 3-heterocyclylpropanohydroxamic acids
US6462063B1 (en) 2000-02-04 2002-10-08 Fibrogen, Inc. C-proteinase inhibitors
US6831088B2 (en) 2001-03-30 2004-12-14 Pfizer Inc. 3-heterocyclypropanohydroxamic acid PCP inhibitors
US6645993B2 (en) 2001-03-30 2003-11-11 Warner-Lambert Company 3-heterocyclylpropanohydroxamic acid PCP inhibitors
US7119203B2 (en) 2002-04-25 2006-10-10 Pharmacia Corporation Piperidinyl- and piperazinyl-sulfonylmethyl hydroxamic acids and their use as protease inhibitors
EP1511472A2 (en) * 2002-05-29 2005-03-09 Merck & Co., Inc. Compounds useful in the treatment of anthrax and inhibiting lethal factor
US7504425B2 (en) 2002-05-29 2009-03-17 Merck & Co., Inc. Compounds useful in the treatment of anthrax and inhibiting lethal factor
EP1511472A4 (en) * 2002-05-29 2006-09-27 Merck & Co Inc Compounds useful in the treatment of anthrax and inhibiting lethal factor
WO2004050620A3 (en) * 2002-12-03 2004-08-19 Biomep Inc Derivatives of succinic and glutaric acids and analogs thereof useful as inhibitors of phex
WO2004050620A2 (en) * 2002-12-03 2004-06-17 Enobia Pharma Derivatives of succinic and glutaric acids and analogs thereof useful as inhibitors of phex
US7365091B2 (en) 2002-12-03 2008-04-29 Enobia Pharma Derivatives of succinic and glutaric acids and analogs thereof useful as inhibitors of PHEX
US7105539B2 (en) 2002-12-03 2006-09-12 Enobia Pharma Derivatives of succinic and glutaric acids and analogs thereof useful as inhibitors of phex
WO2005061443A3 (en) * 2003-12-19 2005-12-22 Basf Ag Benzoyl-substituted phenylalanine amides
US7687435B2 (en) 2003-12-19 2010-03-30 Basf Aktiengesellschaft Benzoyl-substituted phenylalanineamides
WO2005061443A2 (en) 2003-12-19 2005-07-07 Basf Aktiengesellschaft Benzoyl-substituted phenylalanine amides
US7579487B2 (en) 2004-05-11 2009-08-25 Merck & Co., Inc. Process for making N-sulfonated-amino acid derivatives
US8877729B2 (en) 2007-05-04 2014-11-04 Marina Biotech, Inc. Amino acid lipids and uses thereof
US7939505B2 (en) 2007-05-04 2011-05-10 Marina Biotech, Inc. Amino acid lipids and uses thereof
US8501824B2 (en) 2007-05-04 2013-08-06 Marina Biotech, Inc. Amino acid lipids and uses thereof
US9339461B2 (en) 2007-05-04 2016-05-17 Marina Biotech, Inc. Arginine-based lipids for delivery of therapeutics
US9731016B2 (en) 2007-05-04 2017-08-15 Marina Biotech, Inc. Tyrosine-based lipids for delivery of therapeutics
WO2009097893A1 (en) * 2008-02-04 2009-08-13 Proyecto De Biomedicina Cima, S.L. Methods for the treatment of cardiac disease associated to myocardial fibrosis using an inhibitor of pcp
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
KR19990036271A (en) 1999-05-25
JPH11511137A (en) 1999-09-28
EP0845987A4 (en) 2000-05-24
AU6951296A (en) 1997-03-05
BR9609883A (en) 1999-03-23
CN1198096A (en) 1998-11-04
MX9801093A (en) 1998-04-30
EP0845987A1 (en) 1998-06-10
CA2229098A1 (en) 1997-02-20

Similar Documents

Publication Publication Date Title
EP0845987A1 (en) C-proteinase inhibitors for the treatment of disorders related to the overproduction of collagen
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 (en) Cycloalkyl-substituted glutaramide antihypertensive agents
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 (en) Matrix Metalloprotease Inhibitors
JPH1160551A (en) Sulfonylaminocarboxylic acid
KR20000015785A (en) Phenyl sulfone amid derivative
JP2000515493A (en) Para-substituted phenylene derivatives
KR19980086893A (en) Substituted diaminocarboxylic acids
WO1999017790A1 (en) PEPTIDE-CONTAINING α-KETOAMIDE CYSTEINE AND SERINE PROTEASE INHIBITORS
JP2844329B2 (en) Selective thrombin inhibitors
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 (en) Compounds, compositions and methods for inhibiting the binding of proteins containing an sh2 domain to cognate phosphorylated proteins
FI112660B (en) A process for preparing tricyclic indole derivatives useful as matrix metalloprotease inhibitors
EP0343911A2 (en) Cycloalkyl-substituted glutaramide diuretic agents
WO1993014112A1 (en) Substituted phosphinic acid-containing peptidyl derivatives as antidegenerative agents
US6462063B1 (en) C-proteinase inhibitors
KR19980086989A (en) Substituted 6- and 7-amino-tetrahydroisoquinoline carboxylic acids
RU2106356C1 (en) Pseudopeptides of salts thereof, methods of preparation thereof, and pharmaceutical composition
PL198827B1 (en) N-ARYLSULFONYL AMINO ACID OMEGA AMIDES, method of making them, pharmaceutical agent and their application
HU206194B (en) Process for producing cyclomethylene-1,2-dicarboxylic acid derivatives and pharmaceutical compositions comprising same
EP0712838B1 (en) Acylphenylglycine derivative and preventive and remedy for diseases caused by increased collagenase activity containing said compound as active ingredient
AU4973500A (en) Alpha-amino-beta-sulfonyl hydroxamic acid compounds

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 96197271.8

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

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)
ENP Entry into the national phase

Ref document number: 2229098

Country of ref document: CA

Ref document number: 2229098

Country of ref document: CA

Kind code of ref document: A

Ref document number: 1997 508648

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1019980700939

Country of ref document: KR

Ref document number: PA/a/1998/001093

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1996930499

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1996930499

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1019980700939

Country of ref document: KR

WWW Wipo information: withdrawn in national office

Ref document number: 1996930499

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1019980700939

Country of ref document: KR