WO1999054317A1 - Inhibiteurs de cysteine proteases - Google Patents

Inhibiteurs de cysteine proteases Download PDF

Info

Publication number
WO1999054317A1
WO1999054317A1 PCT/US1999/008501 US9908501W WO9954317A1 WO 1999054317 A1 WO1999054317 A1 WO 1999054317A1 US 9908501 W US9908501 W US 9908501W WO 9954317 A1 WO9954317 A1 WO 9954317A1
Authority
WO
WIPO (PCT)
Prior art keywords
inhibitor
alkyl
protease
aryl
optionally substituted
Prior art date
Application number
PCT/US1999/008501
Other languages
English (en)
Inventor
Lyle W. Spruce
Albert C. Gyorkos
John C. Cheronis
Val S. Goodfellow
Axel H. Leimer
John M. Young
James I. Gerrity
Original Assignee
Cortech Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/065,258 external-priority patent/US6004933A/en
Application filed by Cortech Inc. filed Critical Cortech Inc.
Priority to NZ507696A priority Critical patent/NZ507696A/en
Priority to CA002329712A priority patent/CA2329712A1/fr
Priority to AU39651/99A priority patent/AU750369B2/en
Priority to IL13918799A priority patent/IL139187A0/xx
Publication of WO1999054317A1 publication Critical patent/WO1999054317A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06078Dipeptides with the first amino acid being neutral and aromatic or cycloaliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06034Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms
    • C07K5/06043Leu-amino acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06034Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms
    • C07K5/06052Val-amino acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0812Tripeptides with the first amino acid being neutral and aromatic or cycloaliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0819Tripeptides with the first amino acid being acidic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • cysteine proteases have been identified in biological systems.
  • a "protease” is an enzyme which degrades proteins or peptides into smaller components.
  • the term “cysteine protease” refers to proteases which are distinguished by the presence of a cysteine residue which plays a critical role in the catalytic process. Mammalian systems, including humans, normally degrade and process proteins via a variety of mechanisms including the actions of cysteine proteases. However, when present at elevated levels or when abnormally activated, or where introduced into a biological system in the context of a viral, bacterial or parasitic infection, cysteine proteases are thought to be involved in numerous pathophysiological processes and disease states.
  • calpains calcium-activated neutral proteases
  • calpain I and II Three major calpains have been identified: calpain I and II, and p94.
  • the calpain family of cysteine proteases has been implicated in many diseases and disorders, including stroke, neurodegeneration, such as Alzheimer's disease, amyotrophy and motor neuron damage; acute central nervous system injury, muscular dystrophy, bone resorption, platelet aggregation, cataracts and inflammation.
  • Calpain I has been implicated in excitatory arnino-acid induced neurotoxicity disorders including ischemia, hypoglycemia and epilepsy.
  • cysteine protease p94 a muscle-specific member of the calpain family, has been identified as a gene product responsible for limb girdle muscular dystrophy (Barrett A.J., et al. ICOP Newsletter, 1-2 (1996)).
  • Lysosomal cysteine proteases or cathepsins belong to the papain superfamily of cysteine proteases. They are widely distributed and differentially expressed among tissues. Intracellularly, they serve a variety of digestive and processing functions. Extracellularly, they may be involved in tissue remodeling and in pathologies such as arthritis, inflammation, myocardial infarction, Alzheimer's disease, cancer and muscular dystrophy (Elliott E., et al., Per. in Drug Disc. andDes., 6: 12-32 (1996)).
  • Interleukin-1 ⁇ converting enzyme is a member of the caspase family of cysteine proteases which catalyzes the formation of interleukin-l ⁇ (IL-l ⁇ ), as well as the formation of interferon- ⁇ inducing factor (I GIF) from their inactive precursors, proIL-l ⁇ and pro-IGIF, respectively.
  • Interleukin-l ⁇ is an immunoregulatory protein implicated in inflammation, diabetes, septic shock, rheumatoid arthritis and Alzheimer's disease.
  • ICE and/or other caspases have also been linked to the apoptotic cell death of neurons which is implicated in a variety of neurodegenerative disorders including Parkinson's disease, ischemia and amyotrophic lateral sclerosis (ALS)(Dinarello C, et al., New Eng. J. Med., 328: 106-113 (1993)).
  • ALS amyotrophic lateral sclerosis
  • Cysteine proteases are also produced by various viral pathogens and appear to be involved in every stage of reproduction including DNA and RNA translation and synthesis, and capsid formation (Gorbalenya A., et al., Per. In Drug Disc, 6:64-86 (1996); Krausslich et al., Ann. Rev. Biochem., 57:701-54 (1988)).
  • viral pathogens include Picornaviridae, which includes the genera Enterovirus, Rhinovirus, Cardiovirus, and Aphthovirus, which cause numerous human disease syndromes, ranging from fatal paralysis, encephalitis, meningitis, hepatitis and myocarditis to the common cold (Krausslich et al., Ann.
  • the picomaviral 3C proteinases which are produced by all picornaviruses, are responsible for processing viral polyproteins, an essential stage in viral growth (Malcolm B., et al. Biochemistry, 34:8172-8179 (1995)).
  • cysteine proteinases play significant roles in host-parasite interactions and pathogenesis (Robertson C, et al., Pers. in Drug Disc. andDes., 6:99-118 (1996)). For example, most of the proteinase activity detected in trypanosomes and various Leishmania species has been characterized as belonging to the cysteine protease class.
  • Other proteases are produced by Clostridium histolyticum and malaria parasites, such as Plasmodium falciparum and Plasmodium vinckei strains, and Schistosoma.
  • Cancer procoagulant, CP a cysteine proteinase from malignant cells, has emerged as a probable activator of the coagulation system in cancer (Alessio M.G., et al., Eur. J. Haematol, 45: 78-81 (1990); Gordon S., Methods in Enz., 244:568-581 (1994); Gordon S., Sem. in Thromb. andHemo., 18, 4:424-433 (1992)).
  • cysteine protease inhibitors are primarily irreversible in nature; only weakly inhibit the enzymatic activity of the targeted protease and/or are toxic. Thus, there is a need for effective inhibitors of cysteine proteases as therapeutic and as prophylactic agents for the treatment and/or prevention of cysteine protease mediated pathologies.
  • the present invention relates to cysteine protease inhibitors of the general formula (I):
  • Z is a cysteine protease binding moiety, Z being a carbonyl containing group, preferably an amino carbonyl containing group, wherein the carbon of the heterocycle is attached directly to the carbonyl group of Z.
  • R is alkyl or alkenyl optionally substituted with 1-3 halo or hydroxy; alkylamino, dialkylamino, alkyldialkylamino; or cycloalkyl, alkylcycloalkyl, alkenylcycloalkyl, (C 3 -C 12 )aryl, (C 5 -C 12 )arylalkyl or (C 5 -C, 2 )arylalkenyl optionally comprising 1- 4 heteroatoms selected from N, O and S, and optionally substituted with halo, cyano, nitro, haloalkyl, amino, aminoalkyl, dialkylamino, alkyl, alkenyl, alkynyl, alkoxy, haloalkoxy, carboxyl, carboalkoxy, alkylcarboxamide, (C 5 -C 6 )aryl, -0-(C 5 -C 6 )aryl, arylcarboxamide, alkylcarboxamide, (
  • R is methyl, dimethylamino, phenyl or benzyl optionally substituted with methyl, halo, methylenedioxy, methoxy, dimethoxy, trimethoxy, trifluoromethyl and dimethylamino.
  • X is O and Y is N; X is N and Y is O; or both X and Y are N.
  • Z comprises 1 to 5 amino acid residues or mimetics thereof.
  • Z may, for example, comprise a pentapeptidyl, tetrapeptidyl, tripeptidyl or dipeptidyl binding moiety.
  • Z is of the formula (II):
  • AA,, AA 2 , AA 3 , AA 4 and AA 5 are independently an amino acid residue or amino acid residue mimetic; a direct bond or absent;
  • R, and R/ are independently -C(0)R 5 , -C(0)NHR 5 , -S(0) 2 R 5 , -C(0)OR 5 , -CR 5 or R 5 , where R 5 is H, alkyl, alkenyl or alkynyl optionally substituted with halo, cyano, nitro, haloalkyl, amino, aminoalkyl, dialkylamino, haloalkoxy, carboxyl, carboalkoxy or alkylcarboxamide; cycloalkyl, alkylcycloalkyl, (C 5 -C 12 ) aryl or (C 5 -C 12 )arylalkyl optionally comprising 1-4 heteroatoms selected from N, O and S, and optionally substituted with halo, cyano, nitro, haloalkyl, amino, aminoalkyl, dialkylamino, haloalkoxy, carboxyl, carboalkoxy, alkylcarboxamide, alkyl
  • Typical terminal R, groups include Cbz, succinic acid derivatives of the formulas -C(0)CH(-CH 2 CH(CH 3 ) 2 )CH 2 COOH, - C(0)CH 2 CH 2 COOH, and -C(0)CH 2 CH 2 C(0)OC(CH 3 ) 3 ; toluenesulfonyl, methane sulfonyl, FMOC, (t)-menthyloxy-CO- and acetyl.
  • the amino acids are selected from arginine or an arginine mimetic, proline; aspartic and glutamic acid and the aryl and alkyl esters thereof; alanine and glycine optionally substituted at the ⁇ -carbon or ⁇ -nitrogen with alkyl, cycloalkyl or aryl; leucine, isoleucine; cysteine optionally substituted at the sulfur atom with alkyl, alkenyl or phenyl optionally substituted with halo, cyano, nitro, haloalkyl, amino, aminoalkyl, dialkylamino, alkyl, alkoxy, haloalkoxy, carboxyl, carboalkoxy, alkylcarboxamide, arylcarboxamide, alkylthio or haloalkylthio; phenylalanine, homo-phenylalanine, dehydro-phenylalanine, indoline-2-carboxylic acid; te
  • X' is CR 2 ' or N
  • R 2 , R 2 ' and R 2 " are independently H; alkyl or alkenyl optionally substituted with 1-3 halo, hydroxy, thio, alkylthio, amino, alkylarnino, dialkylamino, alkylguanidinyl, dialkylguanidinyl, guanidinyl; -RCOR', -RCOOR', -RNR'R"R° or -RC(0)NR'R” where R is alkyl or alkenyl, and R', R" and R° are independently H, alkyl, alkenyl, cycloalkyl or (C 5 - C 6 )aryl; or cycloalkyl, alkylcycloalkyl, alkenylcycloalkyl, alkyl-oxyaryl, alkyl-thioaryl, (C 5 -C 12 ) aryl, (C j -C 12 )arylalkyl or (Cj-C 12 )ary
  • R 2 and R 2 ' together with X' form a ring comprising 4-7 atoms selected from C, N, S and O, said ring optionally subsitituted with hydroxy, halo, cyano, nitro, haloalkyl, amino, aminoalkyl, dialkylamino, amidine, alkylamidine, dialkyl amidine, alkyl, alkenyl, alkynyl, alkoxy, haloalkoxy, carboxyl, carboalkoxy, alkylcarboxamide, (C 5 -C 6 )aryl, -0-(C 5 -C 6 )aryl, arylcarboxamide, alkylthio or haloalkylthio.
  • AA 2 may be a residue of the formula (111b):
  • R 3 , R' 3 and R" 3 are independently H; alkyl or alkenyl optionally substituted with 1-3 halo, hydroxy, thio, alkylthio, amino, alkylamino, dialkylamino, alkylguanidinyl, dialkylguanidinyl, guanidinyl; -RCOR', -RCOOR' or -RC(0)NR'R" where R is alkyl or alkenyl, and R' and R" are independently H, alkyl, alkenyl, cycloalkyl or (C 5 -C 6 )aryl; or cycloalkyl, alkylcycloalkyl, alkenylcycloalkyl, alkyl-oxyaryl, alkyl-thioaryl, (C 5 -C 12 ) aryl, (C 5 -C 12 )arylalkyl or (C 5 -C 12 )arylalkenyl optionally comprising 1-4 heteroatoms selected
  • G is -C(O)-, -NHC(O)-, -S(0) 2 -, -OC(O)-, -C- or a direct bond;
  • Rj, R 7 , R' 6 , R' 7 are independently H, alkyl, alkenyl, halo, alkoxy, carboxyl, carboalkoxy, amino, aminoalkyl, dialkylamino; cycloalkyl, (C 3 -C 6 ) aryl or (C 5 -C 6 ) arylalkyl optionally comprising 1-3 heteroatoms selected from N, O and S, and optionally substituted with alkyl, alkenyl, alkynyl, halo, cyano, nitro, haloalkyl, haloalkoxy, amino, alkylamino, dialkylamino, alkoxy, haloalkoxy, carboxyl, carboalkoxy, alkylcarboxamide, alkylthio, gu
  • U, V, W and Y' are independently or together N, C, C(O), N(R,) where R, is H, alkyl, halo, alkoxy, carboalkoxy, cycloalkoxy, carboxyl, alkylthio, amino, alkylamino, dialkylamino, or aryl, fused aryl or cycloalkyl optionally comprising 1 or more heteroatoms selected from O, S and N, and optionally subsituted with halo or alkyl; N(R 10 ) where R 10 is H, alkyl, alkenyl or cycloalkyl, aryl, arylalkyl or fused aryl-cycloalkyl optionally comprising 1-4 heteroatoms selected from N, O and S, and optionally substituted with alkyl, alkenyl, alkynyl, halo, cyano, nitro, haloalkyl, haloalkoxy, amino, alkylamino,
  • X' and X" are C, and R' 2 and R' 3 are H. In another embodiment, X' and/or X" are N.
  • R 5 is benzyl, isoquinolinyl, quinolinyl, naphthyl or HOOCCH 2 C(CH 2 CH(CH 3 ) 2 )-; or R, is Cbz wherein the phenyl is optionally substituted with nitro.
  • R may be toluenesulfonyl, methanesulfonyl, FMOC or (+)-menthyl- oxy-CO-.
  • AA 3 is leucine
  • AA 4 and AA 5 are direct bonds or absent
  • R 5 is alkyl
  • R 4 Leu-Leu-; R ⁇ Leu-Leu-Phe-; R,-Leu-Abu-; R,-Val-Phe-; R,-Leu-Leu-Nle-;
  • Z is Cbz-Leu-Nle-; or Cbz-Leu-Vak
  • -AA 2 -AA,- are selected from: -Phe-hPhe-; -Arg-hPhe-; -Arg mim
  • Z may be a cathepsin L binding moiety, where R 3 is preferably benzyl or (CH 3 ) 2 CHCH 2 -; and R 2 is -CH 2 -benzyl. Where Z is a cathepsin S binding moiety; preferably R 2 and R 3 are alkyl; more preferably
  • R 3 is benzyl, (CH 3 ) 2 CHCH 2 - or (CH 3 ) 2 CH-; and R 2 is -CH 2 - benzyl.
  • AA 3 , AA 4 and AA 3 are direct bonds or absent;
  • R 5 is benzyl, isoquinolinyl, quinolinyl, naphthyl or HOOCCH 2 C(CH 2 CH(CH 3 ) 2 )-; or R, is Cbz.
  • Z is a cathepsin H binding moiety; Z is preferably R,-hPhe-; or HCMiPhe-. Z may also be a cathepsin K binding moiety; where preferably R 3 is benzyl, (CH 3 ) 2 CHCH 2 - or (CH 3 ) 2 CH-; and preferably AA 3 is Gly; and AA 4 is Val or D-Val.
  • R 3 is benzyl, (CH 3 ) 2 CHCH 2 - or (CH 3 ) 2 CH-; and preferably AA 3 is Gly; and AA 4 is Val or D-Val.
  • AA is Arg, Arg mimetic or hPhe; AA 2 is Pro; AA 3 is Gly; and AA 4 is Val or D-Val; or preferably Z is
  • Z is a caspase binding moiety; preferably R 2 is -RCOOR'; where preferably R is -CH 2 - and preferably R' is H; where preferably AA 3 and AA 4 are amino acid residues and AA 5 is a direct bond.
  • Z is an interleukin-l ⁇ converting enzyme binding moiety
  • AA may be optionally substituted tyrosine or leucine
  • AA 3 may be valine, glutamate or an ester of glutamate
  • R 3 may be -CH 3 or (CH 3 ) 2 CH-.
  • R 3 is -CH 3 or imidazolyl-CH 2 -; AA 3 is valine or glutamate; and
  • R 3 is -CH 3 .
  • Z may also be R 4 -AA 5 -AA 4 -AA 3 -Pro-AA 1 ; where AA, is Asp or Asp ester; where -AA 5 - AA 4 -AA 3 - may be -Ala-; -Glu-; -Val-; -Tyr-Ala-; -Tyr-GIu-; -Tyr-Val-; -Leu-Ala-; -Leu-Glu-; or -Leu-Val-.
  • AA 2 is of the formula (VI); wherein X" is CR' 3 where preferably R' 3 is H; and
  • R is -RCOOR' where R is alkyl or alkenyl, and R' is H, alkyl, alkenyl, cycloalkyl or (C 3 - C 6 ) aryl.
  • AA 4 and AA 5 are direct bonds or absent, AA 3 is Tyr or Tyr(O-R') or a direct bond or absent;
  • R 2 is -RCOOR' where R is alkyl or alkenyl, and R' is H, alkyl, alkenyl, cycloalkyl or (C 3 -C 6 ) aryl;
  • Rj is phenyl or benzyl substituted with halo; and
  • R 5 may be benzyl, isoquinolinyl, quinolinyl, naphthyl or HOOCCH 2 C(CH 2 CH(CH 3 ) 2 )-.
  • R 2 is -RCOOR' where preferably R is -CH 2 - and A ( is Asp or an ester thereof.
  • AA 3 is optionally substituted glutamine or glutamic acid or an ester thereof.
  • R 2 is (CH 3 ) 2 CH- or CH 3 SCH 2 CH 2 -.
  • Z is a FLICE binding moiety
  • R 2 is -RCOOR', where preferably R is -CH 2 -; AA 4 is optionally substituted lysine; and preferably AA 3 is glutamic acid; and R 3 is (CH 3 ) 2 CH-.
  • Z may also be a viral or microbial cysteine protease binding moiety.
  • Z is a gingipain binding moiety.
  • R 2 is preferably -RNR'R"R° where preferably R is (C,-C 4 )alkyl; R' is H; and preferably R" and R° are H or (C,- C 3 )alkyl.
  • R 2 is + H 3 N(CH 2 ) 3 CH 2 -.
  • Z is a gingipain R binding moiety
  • AA 2 is proline, where Z is R,-Leu-Pro-AA,-, where AA, is arginine or an arginine mimetic.
  • Z may also be a human coronavirus protease binding moiety, where preferably R 2 is
  • R' is cycloalkyl, aryl or arylalkyl optionally substituted with one or more heteroatoms selected from N, S or O; and R" and R° are alkyl or cycloalkyl; and preferably R 3 is (CH 3 ) 2 CH-, (CH 3 ) 2 CHCH 2 - or CH 3 (CH 2 ) 2 CH 2 -; AA 3 is Asp or ester thereof, Leu, Arg or Arg mimetic, or direct bond; AA 4 and AA 3 are direct bonds or absent; and R 3 is alkyl.
  • R 2 is preferably -RC(0)NR'R" where R' and R" are H or -CH 3 ; or RCOOR' where R' is CH 3 ; and AA 3 and AA 4 are amino acid residues.
  • AA 4 is Leu; R 3 is -CH 3 and AA 3 is Ala.
  • Z may also be a hepatitis A virus 3C proteinase binding moiety, where Z is R,-Leu-AA 3 -Thr-Gln-;
  • R 2 and R 3 are preferably H; AA 3 is alanine; AA 4 is leucine; AA 3 is a direct bond; and R, is absent.
  • R 2 is RCOOR' where R is -CH 2 -; R 3 is benzyl; and AA 3 is leucine, isoleucine or a direct bond.
  • R 2 is -RC(0)NR'R" where R' and R" are H, -CH 3 or -
  • RCOOR' where R' is -CH 3 or -CH 2 CH 3 .
  • Z may also be a human picornain 2A protease; where R 3 is -CH(OR')CH 3 where R' is H, alkyl or aryl; and preferably R 2 is a hydrophobic side chain.
  • R 3 is -CH(OR')CH 3 where R' is H, alkyl or aryl; and preferably R 2 is a hydrophobic side chain.
  • AA is Val or dehydro-Phe
  • AA 2 is Pro
  • AA 3 is Val.
  • Examples include compounds CE-2072, CE-2060 and CE-2061 , the structures of which are shown below.
  • Z is
  • Z may be a protozoan protease binding moiety, such as a Trypanosoma
  • Z may be also selected from -Pro-Phe-Arg-; -Phe-Arg-;-Val-Leu-Lys-; -Leu-Val-Tyr-; Suc-Leu-Tyr- or -Phe-Ala-.
  • Z is a Plasmodium protease binding moiety
  • Z is R,-Phe-Arg-;
  • Z is R ⁇ -Phe-AA,-; or R ⁇ -Leu-AA,-;
  • AA is optionally substituted lysine; and where R, may be morpholino.
  • R may be morpholino.
  • AA 3 , AA 4 and AA 3 are direct bonds or absent, and R ⁇ is Cbz.
  • the present invention also provides methods of inhibiting the enzymatic activity of one or more cysteine proteases comprising contacting a protease with an inhibitory amount of a compound described herein.
  • the compound is selected from:
  • the present invention also provides a method of inhibiting cancer cell growth or tumor progression or tumor metastasis or invasion, by inhibiting the enzymatic activity of cysteine proteases associated with such growth or progression, such as cathepsin B or cathepsin L.
  • Suitable pathogenic targets include, by example only, hepatitis A virus 3C
  • the present invention also provides a method of treating the symptoms associated with allergic responses by inhibiting the enzymatic activity of cysteine proteases associated with certain allergens, such as, for example Derp I.
  • the invention provides a method of treating the symptoms associated with neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, multiple sclerosis.
  • the invention further provides a method of treating the symptoms associated with stroke.
  • a method of treating the symptoms associated with inflammatory and degenerative diseases such as arthridities, including rheumatoid arthritis or osteoarthritis, or periodontal disease.
  • cyste protease binding moiety means a chemical group capable of binding to the substrate binding site of a cysteine protease, typically defined in the literature as the S,-S sold site.
  • the term includes both peptides and peptide mimetics.
  • the binding moiety is selected such that when linked to the keto-heterocycle, the moiety provides the resulting compound with inhibitory activity against the target cysteine protease of less than 100 ⁇ M (K, value); and more preferably of less than 10 ⁇ M.
  • alkyl means C,-C 15 , however, preferably C,-C 7 .
  • alkenyl means C,-C 13 , however, preferably -C 7 .
  • alkynyl means C,-C 13 , however, preferably C,-C 7 .
  • alkyl, alkenyl and alkynyl groups may be linear or branched.
  • aryl means aryl groups preferably comprising 5 to 12 carbons, and more preferably 5 to 6 carbons. Unless otherwise indicated, the term "aryl,” unless otherwise stated, means aryl groups preferably comprising 5 to 12 carbons, and more preferably 5 to 6 carbons. Unless otherwise indicated, the
  • aryl includes mono-and bi-cyclic, as well as fused ring systems.
  • arylalkyl includes mono-substituted alkyl groups (e.g., benzyl), as well as di-substituted alkyl groups such as -alkyl(phenyl) 2 (e.g., -CH(phenyl) 2 ).
  • arylalkyl or arylalkenyl is defined by the general formula (C x -C y )arylalkyl or (C x .C y )arylalkenyl, x and y refer to the number of carbons making up the aryl group.
  • alkyl group is as defined above.
  • arylalkenyl includes aryl compounds having an alkenyl chain comprising 1-3 or more double bonds.
  • Figure 1 shows the inhibition of the production of mature IL-l ⁇ in THP-1 cell line by certain compounds of the present invention.
  • Figures 2 A and 2B show the inhibition of the production of mature IL-l ⁇ in whole blood by certain compounds of the present invention.
  • FIG. 3 is a schematic representation of the synthesis of a compound according to the invention (CM-0019).
  • the present invention provides compounds which are useful as cysteine protease inhibitors. These compounds are characterized by their relatively low molecular weight, reversible inhibition, high potency and selectivity with respect to various types of cysteine
  • the compounds can be implemented to prevent, alleviate and/or otherwise treat diseases which are mediated by the effects associated with the presence of cysteine proteases. Their usage is of particluar importance as they relate to various human treatments in vivo and as well as diagnostic tools in vitro.
  • Peptidyl inhibitors of serine proteases comprising serine protease binding moieties and certain keto-heterocycles have been previously described (see WO 96/16080). It has been surprisingly found that compounds comprising cysteine protease binding moieties and these keto-heterocycles are highly potent and specific inhibitors of a wide variety of cysteine proteases as well.
  • the inhibiting activity can be directed against any cysteine protease by identifying the binding moiety specific for that protease.
  • the characteristics for the P, . . . P n residues using substrate nomenclature by Schechter and Berger (Biochem. Biophys. Res. Commun. 27: 157 (1967); Biochem. Biophys. Res. Commun. 32: 898 (1968)), which define the minimum recognition sequence of enzymes for small synthetic peptide substrates or inhibitors are known for many enzymes or can be determined by measuring rates of hydrolysis of various substrates. Some examples are listed
  • Hepatitis A Virus Gin or its Ala, Val, Leu, Nle, P3 - Arg or its 14 3C proteinase derivatives e.g. Phe mimetics, Ala dimethylGln, P4 - hydrophobic Azogln), ' Glu and residues its derivatives
  • the substituent on the heterocycle i.e., R,
  • R the substituent on the heterocycle
  • the compound CM-0019 comprises the binding moiety specific for papain and a substituted 1,3,4-oxadiazole:
  • the compounds CQ-0010 and CQ-0011 inhibit caspases.
  • Compounds CQ-0002 and CQ-0008 are analogs of leupeptin, the structure of which is provided below for comparison.
  • inhibitors include compounds CE-2072, CE-2060 and CE-2061, which have shown inhibitory activity against picornain 2 A protease (100% inhibition at 100 ⁇ M):
  • these compounds can be synthesized in a single step by refluxing hydrazides of common carboxylic acids with excess ethyl orthoformate at high temperature.
  • the excess orthoformate is hydrolyzed in the workup and the 5-substituted 1,3,4-oxadiazoles are often obtained in essentially pure form without further purification necessary.
  • This entire method of synthesis is illustrated in general form in scheme 1 below. Instances where R 2 correlates to the amino acid side chains of aspartic acid, arginine, and alanine are provided in the Examples.
  • AA n means AA 2 . . . AA 3 .
  • the compounds described herein are useful in inhibiting the activity of cysteine proteases, by contacting the compound with the targeted protease, either in an in vivo or an in vitro environment.
  • contacting means directly or indirectly causing the inhibitor and the protease to come into physical association with each other. Contacting thus includes physical acts such as placing the inhibitor and protease together in a container, or administering the inhibitors to a patient.
  • administering a compound of the invention to a human patient evidencing a disease or disorder associated with abnormal and/or aberrant activity of such proteases in a method for inhibiting the enzymatic activity of such proteases which are associated with disease or disorder falls within the scope of the definition of the term "contacting.”
  • compositions include organic and inorganic acid addition salts such as chloride, acetate, maleate, fumarate, tartrate and citrate.
  • pharmaceutically acceptable metal salts are alkali metal salts such as sodium salt or potassium salt, alkaline earth metal salts such as magnesium salt and calcium salt, aluminum salt and zinc salt.
  • pharmaceutically acceptable ammonium salts are ammonium salt, trishydroxymethylaminomethane and tetramethylammonium salt.
  • pharmaceutically acceptable amino acid addition salts are salts with lysine, glycine and phenylalanine.
  • Cysteine proteases which may be inhibited by the compounds described herein include mammalian, bacterial, parasite, viral, fungal, insect and plant cysteine proteases. Cysteine proteases include papain, actinidain, aleurain (barley), allergen (Dermatophagoides), allergen (Euroglyphus), ananain (Ananas comosus), asclepain (Asepias syriaca), bleomycin hydrolase, calotropin (Calotropis), caricain, clostripain, cathepsin B, cathepsin H, cathepsin L, cathepsin S, cathepsin O, cathepsin K, cathepsin T, chymopain, cysteine aminopeptidase (Lactococcus), cysteine endopeptidases 2 and 3 (barley), cysteine endopeptidases (Brassica napus), cyste
  • endopeptidase Hemerocallis
  • cysteine endopeptidases 1 , 2 and 3 Homarus
  • cysteine endopeptidase Leishmania
  • cysteine endopeptidase mung bean
  • cysteine endopeptidase Ostertagia
  • cysteine endopeptidase pea
  • cysteine endopeptidase Plasmodium
  • cysteine protease tpr Porphyromonas
  • cysteine endopeptidase Tetrahymena
  • cysteine endopeptidase Theileria
  • cysteine endopeptidase tobacco
  • cysteine endopeptidase Trypanosoma
  • dipeptidyl peptidase I endopeptidase (baculovirus ofAutographa), endopeptidase EP-C1 (Phaseolus vulgaris), glycyl endopeptidase, oryzain (include
  • cysteine protease inhibitors described herein, used in prophylaxis, cure or therapy include:
  • Cardiovascular disorders ischemia reperfusion injury from transplantation and/or vascular surgery, angiogenesis, neovascularization, acute cardiac allograft dysfunction, ischemic cardiac damage, chemotherapy-induced myocardial suppression;
  • Inflammatory disorders - rheumatoid arthritis and other inflammatory arthritidies, inflammatory bowel disease, inflammatory peritonitis, sepsis, systemic inflammatory response syndrome, multiple organ failure; Musculo-skeletal disorders—osteoarthritis, osteoporosis, muscular dystrophy, myositis;
  • Neurological disorders multiple sclerosis, stroke, Alzheimer's disease, prion-associated disorders, ataxia telangiectasia, central nervous system injury;
  • Pulmonary disorders asthma, COPD, adult respiratory distress syndrome, Wegener's granulomatosis, emphysema; Allergic, immunologic and autoimmune disorders—house dust mite allergy, transplant rejection, graft verses host disease, Type 1 diabetes mellitus, autoimmune thyroiditis, psoriasis, antibody-mediated autoimmune diseases, lupus erythematosus, Sjogren's syndrome, autoimmune encephalomyelitis;
  • cancer associated coagulopathies including deep venous thrombosis, coronary artery disorder, pulmonary embolism, diss
  • Infectious diseases and associated syndromes septic shock (including Gram-negative sepsis), HJVinfection and AIDS, genital herpes, zoster, chickenpox, EBV infections and encephalitis, CMV-choreoretinitis or encephalitis, cytomegalovirus infections in neonates
  • Kidney disorders polycystic kidney disease, glomerulonephritis
  • miscellaneous disorders periodontal disease, alcohol hepatitis, prostate hypertrophy, trauma, cutaneous mastocytosis, radiation- and HIV-induced diarrhea, cachexia (including acompanying cancer and malnutrition).
  • Caspase 8 caspases Solid tumors, B cell lymphoma, chronic lymphocytic leukemia, prostate hypertrophy, preneoplastic liver foci, resistance to chemotherapy, stroke, Alzheimer's disease, Parkinson's disease, multiple sclerosis, prion-associated disorders, ataxia telangiectasia, ischemic cardiac damage, chemotherapy-induced myocardial suppression, AIDS, type I diabetes, lupus erythematosus, Sjogren's syndrome, glomerulonephritis, dysentery, inflammatory bowel disease, radiation- and HTV induced diarrhea, polycystic kidney disease, anemia or erythropoiesis.
  • Cathepsin B Cathepsin L
  • Osteoarthritis Osteoarthritis
  • osteoporosis Velasco
  • Parkinson's disease Elliott et al., Persp. in Drug leukemia, lymphoma, Disc, and Des., 6:12-32 hodgkin's disease, tumors, (1996) including those of the bladder, brain, lung, pancreas, prostate, stomach and thyroid
  • Cancer Procoagulant Cancer including carcinomas Alessio et al., Eur. J. of the liver, lung, breast, Haematology, 45:78-81 kidney, colon, kidney; osteo-, (1990); Gordon, Seminars in chondro-, and liposarcoma; Thrombosis and Hemostasis, neuroblastoma; melanoma; 18:424-433 (1992) nonlympocytic leukemia; lymphocytic leukemia)
  • Calpain p94 Muscular dystrophy Calpain p94 and limb-girdle muscular dystrophy, ICOP Letters 1996.
  • compositions comprising one or
  • compositions include those suitable for oral, topical or parenteral
  • compositions may, where appropriate, be conveniently presented in discrete unit dosage forms and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the active compound with liquid carriers, solid matrices, semi-solid carriers, finely divided solid carriers or combination thereof, and then, if necessary, shaping the product into the desired delivery system.
  • compositions suitable for oral administration may be presented as discrete unit dosage forms such as hard or soft gelatin capsules, capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or as granules; as a solution, a suspension or as an emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • Tablets and capsules for oral administration may contain conventional excipients such as binding agents, fillers, lubricants, disintegrants, or wetting agents.
  • the tablets may be coated according to methods well known in the art., e.g., with enteric coatings.
  • Oral liquid preparations may be in the form of, for example, aqueous or oily suspension, solutions, emulsions, syrups or elixirs, or may be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparations may contain conventional additives such as suspending agents, emulsifying agents, non-aqueous vehicles (which may include edible oils), or preservative.
  • the compounds may also be formulated for parenteral administration (e.g., by injection, for example, bolus injection or continuous infusion) and may be presented in unit dose form in ampules, pre-filled syringes, small bolus infusion containers or in multi-dose containers with an added preservative.
  • parenteral administration e.g., by injection, for example, bolus injection or continuous infusion
  • the compositions may take such forms as suspensions, solutions, emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form, obtained
  • the compounds may be formulated as ointments, creams or lotions, or as the active ingredient of a transdermal patch.
  • Suitable transdermal delivery systems are disclosed, for example, in Fisher et al. (U.S. Patent No.
  • Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents.
  • the active ingredient can also be delivered via iontophoresis, e.g., as disclosed in U.S. Patent Nos. 4,140,122, 4,383,529, or 4,051,842.
  • compositions suitable for topical administration in the mouth include unit dosage forms such as lozenges comprising active ingredient in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; mucoadherent gels, and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • unit dosage forms such as lozenges comprising active ingredient in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; mucoadherent gels, and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • compositions can be adapted to provide sustained release of the active ingredient employed, e.g., by combination thereof with certain hydrophilic polymer matrices, e.g., comprising natural gels, synthetic polymer gels or mixtures thereof.
  • compositions according to the invention may also contain other adjuvants such as flavorings, coloring, antimicrobial agents, or preservatives.
  • compositions may also be administered via inhalation, using a suitable delivery vehicle.
  • amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician.
  • a suitable dose will be in the range of from about 0.5 to about 100 mg/kg/day, e.g., from about 10 to about 75 mg/kg of body weight per day, such as 3 to about 50 mg per kilogram body weight of the recipient per day, preferably in the range of 6 to 90 mg/kg/day, most preferably in the range of 15 to 60 mg kg/day.
  • the compound is conveniently administered in unit dosage form; for example, containing
  • the active ingredient should be administered to achieve peak plasma concentrations of the active compound of from about 0.5 to about 75 ⁇ M, most preferably, about 1 to 50 ⁇ M, most preferably, about 2 to about 30 ⁇ M. This may be achieved, for example, by the intravenous injection of a 0.05 to 5% solution of the active ingredient, optionally in saline, or orally administered as a bolus containing about 1-100 mg of the active ingredient. Desirable blood levels may be maintained by continuous infusion to provide about 0.01-5.0 mg/kg/hr or by intermittent infusions containing about 0.4-15 mg/kg of the active ingredient(s).
  • the desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
  • the sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
  • the inhibitors described herein may be also used for the detection and quantification of the activity of a cysteine protease in a pure sample, mixture or biological fluid or tissue. The activity can be measured with a protease substrate in the absence and presence of a known concentration of the inhibitor. Specific inhibitors can also be used to confirm that the observed activity is due to a particular protease.
  • the inhibitors described herein may also be used to identify and purify cysteine proteases.
  • the inhibitors can be covalently linked to a solid support, such as an affinity column or beads used in batch methods, and used to purify a protease or enrich a mixture containing the protease.
  • the inhibitor may be linked to the solid support or bead either directly or via a linker of variable length, such that linkage does not interfere with the binding properties (see, e.g.,
  • 35 methylpentanoic acid (2.3 g, 7.11 mmol) in 40 mL of DMF under a nitrogen atmosphere at 0°C was added 1.31 g (9.69 mmol) of HOBT and 1.36 g (7.09 mmol) of EDCI.
  • 1.20 g (7.31 mmol) of 3-methylphenyl acetic hydrazide [prepared analogously to the monoacid hydrazides cited by Rabins et. al. (J. Org. Chem., 30:2486 (1965))] and 1.0 mL (9.10 mmol) of NMM were added. The reaction was allowed to warm to room temperature and stir overnight.
  • Diisopropylamine (5.05 mL, 36 mmol) was diluted with dry THF (20 mL) and chilled to -20°C under N 2 .
  • n-Butyl lithium (1.6 M in hexane, 23 mL, 36 mmol) was added dropwise to maintain the temperature ⁇ -10°C.
  • the temperature was increased to 4°C and stirred 30 minutes to generate LDA.
  • the flask was chilled to -78 °C and (4S)-4-Benzyl-3-[4'-(methyl)pentanoyl]- 2-oxazolidinone in dry THF (15 mL) was added dropwise to maintain the temperature ⁇ -70 °C.
  • Benzoyl hydrazide (200 mg) freshly crystallized from chloroform was suspended in 5 mL of triethyl orthoformate and heated at reflux under nitrogen in a 160 °C oil bath for 3 hours. The mixture was cooled to room temperature, chilled in ice, and treated with 50 mL water and 10 mL 10% KHS0 4 solution. The mixture was stirred approximately 2 minutes then 50 mL of EtOAc was added and stirring continued for 10 minutes. The organic layer was separated and the aqueous layer was extracted three times with ethyl acetate. All ethyl acetate layers were combined and were washed with 10% sodium bicarbonate solution and saturated sodium chloride
  • Acetyl-L-leucyl-L-leucyl-arginine(Mtr) (N-methyl)-(N-Methoxy)-amide Acetyl-Leu- Leu-OH (133 mg) and arginine (Mtr) -N-methyl-N-methoxy amide (200 mg) were dissolved in 10 mL of DMF and were treated with 243 uL of DIEA and 212 mg of HBTU. The reaction stirred at room temperature for 15 hours and was worked up according to method A.
  • Acetyl-L-leucyl-N-[l-[2-[(5-phe ⁇ yl)-l,3,4-oxadiazolyl]carbonyl]-4-(guanidino)- butylJ-L-leucyl amide One half of the crude product from step B was dissolved in a pre-formed solution of 2 mL of TFA and 100 uL thioanisole. The reaction stirred under nitrogen for 4 hours. The solvent was removed in vacuo, and the product was precipitated with dry ether. The precipitate was taken up in methanol and concentrated in vacuo, the residue was triturated with
  • N a -benzyloxycarbonyl ⁇ L-alanine (N-methyl-N-methoxy) amide Cbz-L-Alanine (1.0 g) was dissolved in 10 mL dry DMF with 1.55 mL of DIEA.
  • HBTU (1.78 g) was added and the reaction Stirred 30 minutes.
  • N'-benzyloxycarbonyl-L-alaninal A solution of 6 mL of 1 M lithium aluminum hydride in THF was chilled under nitrogen to 0°C and a solution of compound A (0.68 g ) in 4 mL DMF was added dropwise. After stirring 15 minutes at 0°C the reaction was carefully quenched with 20 mL of EtOAc and 10 mL of 10% KHS0 4 solution. The organic layer was washed with 1 N HC1 and 10% NaHC0 3 solution. Drying over sodium sulfate, removal of the solvent by rotary evaporation, and drying under high vacuum provided 0.38 g of a colorless oil.
  • N-chlorosuccinimide (75.4 mg was suspended in dry toluene and chilled to 0°C. Dimethyl sulfide (60 uL) was added and the suspension stirred 30 minutes at 0°C and was then chilled to - 25 °C. Compound I ( 60 mg) was added in 2 mL dichloromethane and the reaction stirred 2.5 hours at -25 °C. Triethyl amine (84 uL) was added and the reaction warmed to room temperature.
  • N'-t-butoxycarbonyl-L-(Mtr)-argininal Compound A (2 .00 g) was dissolved in 20 mL of dry THF and chilled to 0°C. To this solution was added 4.72 mL of a 1 M solution of LiAlH 4 in THF, dropwise over 30 minutes at 0°C. The reaction was quenched at 0°C by the slow addition of 50 mL EtOAc, followed by 15 mL of 10% KHS0 4 solution. The mixture was
  • N'-Benzyloxycarbonyl-L-Aspartyl(O-t-butyl) N-methyl-N-methoxy amide Cbz-L- Aspartic Acid (O-t-butyl) (1.0 g, 2.93 mmole), N,0-dimethyl hydroxyl amine hydrochloride (0.357 g, 3.66 mmole, were suspended in 15 mL of DMF and treated with 1.53 mL (8.79 mmoles) of DIEA under N 2 atmosphere.
  • HBTU (1.22 g, 3.22 mmoles) was added and the reaction stirred approximately 15 hours at room temperature.
  • the reaction mixture was worked up according to general extractive work up method A.
  • Acetyl-L-tyrosinyl-L- Valyl-L-Alanyl-L-Aspartyl-(O-t-butyl) N-methyl-N-methoxy amide Compound B (150 mg) and Acetyl-Tyr-Val-Ala-OH (230 mg , prepared using conventional peptide synthesis) were combined and suspended in 10 mL of DMF. DIEA (305 uL) was added followed by HBTU.
  • Acetyl-L-Aspartyl(Ot-Bu)-L-Valyl-L-Glutamyl (O-t-Bu)-L-Aspartyl-(O-t-butyl) N- methyl-N-methoxy amide Acetyl-Asp(0-t-Bu)-Val-Glu-(0-t-Bu)-OH (0.302 g, 0.586 mmoles, prepared by conventional peptide synthesis) and H-Asp-(0-t-Bu)-N-(CH 3 )-OCH 3 (0.150 g, 0.645 mmole, prepared as in example VIII) were combined in 5 mL of DMF and DIEA (305 uL) was added.
  • HBTU (277 mg) was added. After 2 hours an additional 200 uL of DIEA was added and the reaction was allowed to stir approximately 15 hours at room temperature. The reaction was worked up according to method A, dried over Na 2 S0 4 and concentrated to an oil. Preparative reverse phase chromatography (C18, 5 - 60% CH 3 CN, 0.1% TFA, 30 minute gradient), and lyophilization of appropriate fractions provided 0.231 g of a colorless lyophilate.
  • reaction mixture was diluted with 5-10 volumes of EtOAc and washed three times each with equivalent volumes of 1 N HCl solution, then saturated NaHC0 3 solution, and finally with saturated NaCl solution.
  • the enzyme cathepsin B (E.C. 3.4.22.01) was obtained from Calbiochem (San Diego,
  • Cathepsm L was assayed in 0.34 M sodium acetate buffer, pH 5.5, containing 0.1 % (v/v) Brij 35, 2.5 mM dithiothreitol (DTT) and 5 mM Na 2 -EDTA.
  • Cathepsin B was assayed under the same conditions, except that the buffer was adjusted to pH 6.
  • Papain was assayed in 50 mM sodium phosphate buffer, pH 6.8, containing 0.2 M sodium chloride, 2 mM DTT, 1 mM Na 2 -EDTA and 0.025 % Brij 35 (v/v).
  • Gingipain assay All assays were carried out in a 96 well microtiter plate reader and cleavage of BAPNA (N ⁇ -benzoyl-DL-arginine-p-nitroanilide hydrochloride) was detected at 405 nm.
  • BAPNA N ⁇ -benzoyl-DL-arginine-p-nitroanilide hydrochloride
  • CM-0019B is an inhibitor of papain and cathepsin L and more selective against cathepsin B than is leupeptin.
  • Compound CQ-0002 which shares the same recognition sequence (Leu-Leu-Arg) with the broad spectrum inhibitor leupeptin, is nearly as potent as leupeptin versus cathepsin B, but surpisingly has a much higher
  • compound CQ-0002 inhibits gingipain R with a potency equivalent to that of leupeptin.
  • Compounds CQ-0004 and CQ-0008 are also potent and selective cathepsin L inhibitors.
  • THP-1 assay Two x 10 6 THP-1 cells were added to 24 well plates in 1 ml RPMI supplemented with 1% FCS, glutamine and 5 x 10' 5 M mercaptoethanol. Two-fold serial dilutions ofthe inhibitors, CQ-0010, CQ-0011 and the commmercially available Ac-YVAD- CHO (Biomol Research Laboratories Inc., Plymouth Meeting, PA), were preincubated with the cells for 15 min at 37°C. LPS was then added to a final concentration of lug/ml and the plates
  • Supematants were harvested after 4 hr and assayed by ELISA for the presence of TNF ⁇ and IL-l ⁇ using commercially available kits (PerSeptive Biosystems, Framingham, MA and R&D Systems, Minneapolis, MN, respectively).
  • Human whole blood assay Heparinized whole blood (19.7 U heparin per ml) from healthy volunteers was collected and dispensed into 12 x 75 mm polystyrene tubes (0.25 ml per tube). The inhibitors, CQ-0010, CQ-0011 and Ac-YVAD-CHO were dissolved in DMSO, then diluted and added to the tubes in 0.25 ml and preincubated with the blood for 15 min at 37°C. LPS was then added to a final concentration of 10 or 100 ug/ml.
  • the tubes were loosely-capped and incubated in a water bath for 4 hr at 37°C after which they were immersed briefly in an ice-water bath. Supematants were harvested by centrifugation and stored at -70°C. The presence of TNF ⁇ and IL-l ⁇ was detected by commercially-available ELISA kits. Assay for other caspase and granzyme B inhibition
  • Inhibition constants were measured photometrically for YAMA (caspase 3), Lap3 (caspase 7), FLICE (caspase 8), Mch2 (caspase 6) and granzyme B.
  • the buffer used for all enzymes consisted of 50 mM Hepes, 100 mM sodium chloride, 10% (v/v) sucrose, 0.1 % (v/v) CHAPS and 10 mM dithiothreitol (DTT). In the case of granzyme B, only 1 mM DTT was used. Enzymes were incubated at 37°C for 10 minutes in 100 ⁇ L well plates and synthetic substrate and inhibitor were added simultaneously. Final substrate concentration was 20 ⁇ M in all cases.
  • the synthetic substrate Ac-DEVD-pNA was used for all caspases and Succ-AAPD- pNA was used for granzyme B.
  • the appearance of product was monitored over 10 minutes at 410 nm using a Spectromax 340 and IC 30 curves were calculated from the initial slopes at varying inhibitor concentrations and inhibition constants were calculated. The results are shown in Table 4.
  • CQ-0010 is an extremely potent and specific inhibitor of IL-l ⁇ production, capable of almost completely inhibiting the production of this cytokine at 5 ⁇ M (Figure 1) while having no dose-dependent effect on levels of TNF ⁇ produced (results not shown).
  • the IC 30 of CQ-0010 was estimated from these dose curves to be 0.3 ⁇ M.
  • CQ-0011 also inhibited ILl ⁇ production but with approximately 10-fold less potency (Figure 1; Table 4).
  • CQ-0010 was equipotent to the aldehyde equivalent (Ac-YVAD- CHO) in inhibiting ICE, but showed improved inhibition against FLICE with a Kj of ⁇ 20 nM.
  • Compound CQ-0011 is a potent inhibitor of Lap3 and FLICE.
  • the compounds are selective and potent caspase inhibitors as shown by their inactivity with respect to granzyme B.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des inhibiteurs de cystéine protéases de formule générale (I), dans laquelle Z est une fraction de liaison des cystéine protéases; X et Y représentent S, O ou N éventuellement substitué; et R1 représente un alkyle ou un aryle éventuellement substitué.
PCT/US1999/008501 1998-04-23 1999-04-23 Inhibiteurs de cysteine proteases WO1999054317A1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
NZ507696A NZ507696A (en) 1998-04-23 1999-04-23 Triazole, oxadiazole and thiadiazole derivatives useful as cysteine protease inhibitors
CA002329712A CA2329712A1 (fr) 1998-04-23 1999-04-23 Inhibiteurs de cysteine proteases
AU39651/99A AU750369B2 (en) 1998-04-23 1999-04-23 Cysteine protease inhibitors
IL13918799A IL139187A0 (en) 1998-04-23 1999-04-23 Cysteine protease inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US09/065,258 1998-04-23
US09/065,258 US6004933A (en) 1997-04-25 1998-04-23 Cysteine protease inhibitors

Publications (1)

Publication Number Publication Date
WO1999054317A1 true WO1999054317A1 (fr) 1999-10-28

Family

ID=22061449

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/008501 WO1999054317A1 (fr) 1998-04-23 1999-04-23 Inhibiteurs de cysteine proteases

Country Status (5)

Country Link
AU (1) AU750369B2 (fr)
CA (1) CA2329712A1 (fr)
IL (1) IL139187A0 (fr)
NZ (1) NZ507696A (fr)
WO (1) WO1999054317A1 (fr)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002030455A2 (fr) * 2000-10-12 2002-04-18 Viromics Gmbh Moyens pour le traitement d'infections a des virus
US6797720B2 (en) 1999-12-03 2004-09-28 Ono Pharmaceutical Co., Ltd. 1,3,4-oxadiazoline derivative and an agent comprising its derivative as active ingredient
WO2005011649A2 (fr) * 2003-05-19 2005-02-10 Vanderbilt University Procedes pour traiter les infections par le virus du syndrome respiratoire aigu severe
US7144901B2 (en) 1999-12-03 2006-12-05 Ono Pharmaceutical Co.,Ltd. Oxadiazole derivatives and drugs containing these derivatives as the active ingredient
US7202262B2 (en) 2003-04-11 2007-04-10 Ptc Therapeutics, Inc. Benzoic acid or benzoate substituted 1,2,4-oxadiazole compounds and their use for the treatment of disease
US7282512B2 (en) 2002-01-17 2007-10-16 Smithkline Beecham Corporation Cycloalkyl ketoamides derivatives useful as cathepsin K inhibitors
WO2010092114A1 (fr) 2009-02-13 2010-08-19 Guerbet Utilisation de tampons pour la complexation de radionucléides
WO2012084981A1 (fr) 2010-12-20 2012-06-28 Guerbet Nanoemulsion de chelate pour irm
WO2013045333A1 (fr) 2011-09-26 2013-04-04 Guerbet Nanoemulsions et leur utilisation comme agents de contraste
WO2014114724A1 (fr) 2013-01-23 2014-07-31 Guerbet Magneto-emulsion vectorisee
US8895497B2 (en) 2009-12-04 2014-11-25 Dcb-Usa, Llc Cathepsin S inhibitors
US8926945B2 (en) 2005-10-07 2015-01-06 Guerbet Compounds comprising a biological target recognizing part, coupled to a signal part capable of complexing gallium
US8986650B2 (en) 2005-10-07 2015-03-24 Guerbet Complex folate-NOTA-Ga68
US9289398B2 (en) 2006-03-30 2016-03-22 Ptc Therapeutics, Inc. Methods for the production of functional protein from DNA having a nonsense mutation and the treatment of disorders associated therewith
US9873677B2 (en) 2014-03-06 2018-01-23 Ptc Therapeutics, Inc. Pharmaceutical compositions and salts of a 1,2,4-oxadiazole benzoic acid
US10517853B2 (en) 2015-10-30 2019-12-31 Ptc Therapeutics, Inc. Methods for treating epilepsy
WO2020007822A1 (fr) 2018-07-02 2020-01-09 Conservatoire National Des Arts Et Metiers (Cnam) Nanoparticules de bismuth métallique (0), procédé de fabrication et utilisations de celles-ci

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5618792A (en) * 1994-11-21 1997-04-08 Cortech, Inc. Substituted heterocyclic compounds useful as inhibitors of (serine proteases) human neutrophil elastase

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5618792A (en) * 1994-11-21 1997-04-08 Cortech, Inc. Substituted heterocyclic compounds useful as inhibitors of (serine proteases) human neutrophil elastase

Cited By (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6797720B2 (en) 1999-12-03 2004-09-28 Ono Pharmaceutical Co., Ltd. 1,3,4-oxadiazoline derivative and an agent comprising its derivative as active ingredient
US7144901B2 (en) 1999-12-03 2006-12-05 Ono Pharmaceutical Co.,Ltd. Oxadiazole derivatives and drugs containing these derivatives as the active ingredient
WO2002030455A3 (fr) * 2000-10-12 2002-08-08 Ulrich Schubert Moyens pour le traitement d'infections a des virus
EP1430903A1 (fr) * 2000-10-12 2004-06-23 Viromics Gmbh Inhibiteurs du proteasome pour le traitement des infections virales
WO2002030455A2 (fr) * 2000-10-12 2002-04-18 Viromics Gmbh Moyens pour le traitement d'infections a des virus
EP2305291A1 (fr) * 2000-10-12 2011-04-06 ViroLogik GmbH Inhibiteurs du proteasome pour le traitement des infections HIV
US7282512B2 (en) 2002-01-17 2007-10-16 Smithkline Beecham Corporation Cycloalkyl ketoamides derivatives useful as cathepsin K inhibitors
US8227494B2 (en) 2003-04-11 2012-07-24 Ptc Therapeutics, Inc. Pharmaceutical compositions of 1,2,4-oxadiazole benzoic acid and their use for the treatment of disease
US8163782B2 (en) 2003-04-11 2012-04-24 Ptc Therapeutics, Inc. 1,2,4-oxadiazole benzoic acid compositions
US7419991B2 (en) 2003-04-11 2008-09-02 Ptc Therapeutics, Inc. 3-[5-(2-fluoro-phenyl)-[1,2,4]oxadiazol-3-yl]-benzoic acid, compositions, and methods for the use thereof
US7683082B2 (en) 2003-04-11 2010-03-23 Ptc Therapeutics, Inc. 1,2,4-oxadiazole benzoic acid compositions and their use in bioassays
US7772259B2 (en) 2003-04-11 2010-08-10 Ptc Therapeutics, Inc. 1,2,4-Oxadiazole benzoic acid compounds and their use for nonsense suppression and the treatment of disease
US10071081B2 (en) 2003-04-11 2018-09-11 Ptc Therapeutics, Inc. Compositions of 1,2,4-oxadiazole benzoic acid compounds and methods for their use
US8796322B2 (en) 2003-04-11 2014-08-05 Ptc Therapeutics, Inc. Methods for using 1,2,4-oxadiazole benzoic acid compounds
US8017636B2 (en) 2003-04-11 2011-09-13 Ptc Therapeutics, Inc. 1,2,4-Oxadiazole benzoic acid compositions and their use in bioassays
US8129540B2 (en) 2003-04-11 2012-03-06 Ptc Therapeutics, Inc. Methods for the synthesis of 1,2,4-oxadiazole benzoic acid compounds
US8975287B2 (en) 2003-04-11 2015-03-10 Ptc Therapeutics, Inc. Methods for using 1,2,4-Oxadiazole benzoic acid compounds
US9861617B2 (en) 2003-04-11 2018-01-09 Ptc Therapeutics, Inc. Compositions of 1,2,4-oxadiazole benzoic acid compounds and methods for their use
US9205088B2 (en) 2003-04-11 2015-12-08 Ptc Therapeutics, Inc. Compositions of 1,2,4-oxadiazol benzoic acid compounds and methods for their use
US8299105B2 (en) 2003-04-11 2012-10-30 Ptc Therapeutics, Inc. 1,2,4-oxadiazole benzoic acid compositions and their use in bioassays
US7202262B2 (en) 2003-04-11 2007-04-10 Ptc Therapeutics, Inc. Benzoic acid or benzoate substituted 1,2,4-oxadiazole compounds and their use for the treatment of disease
US8486982B2 (en) 2003-04-11 2013-07-16 Ptc Therapeutics, Inc. 1,2,4-oxadiazole benzoic acids
WO2005011649A2 (fr) * 2003-05-19 2005-02-10 Vanderbilt University Procedes pour traiter les infections par le virus du syndrome respiratoire aigu severe
WO2005011649A3 (fr) * 2003-05-19 2005-03-17 Univ Vanderbilt Procedes pour traiter les infections par le virus du syndrome respiratoire aigu severe
US8986650B2 (en) 2005-10-07 2015-03-24 Guerbet Complex folate-NOTA-Ga68
US8926945B2 (en) 2005-10-07 2015-01-06 Guerbet Compounds comprising a biological target recognizing part, coupled to a signal part capable of complexing gallium
US9289398B2 (en) 2006-03-30 2016-03-22 Ptc Therapeutics, Inc. Methods for the production of functional protein from DNA having a nonsense mutation and the treatment of disorders associated therewith
WO2010092114A1 (fr) 2009-02-13 2010-08-19 Guerbet Utilisation de tampons pour la complexation de radionucléides
US8895497B2 (en) 2009-12-04 2014-11-25 Dcb-Usa, Llc Cathepsin S inhibitors
US9770520B2 (en) 2010-12-20 2017-09-26 Guerbet Chelate nanoemulsion for MRI
WO2012084981A1 (fr) 2010-12-20 2012-06-28 Guerbet Nanoemulsion de chelate pour irm
WO2013045333A1 (fr) 2011-09-26 2013-04-04 Guerbet Nanoemulsions et leur utilisation comme agents de contraste
WO2014114724A1 (fr) 2013-01-23 2014-07-31 Guerbet Magneto-emulsion vectorisee
US9873677B2 (en) 2014-03-06 2018-01-23 Ptc Therapeutics, Inc. Pharmaceutical compositions and salts of a 1,2,4-oxadiazole benzoic acid
US10233161B2 (en) 2014-03-06 2019-03-19 Ptc Therapeutics, Inc. Pharmaceutical compositions and salts of a 1,2,4-oxadiazole benzoic acid
US10618877B2 (en) 2014-03-06 2020-04-14 Ptc Therapeutics, Inc. Pharmaceutical compositions and salts of a 1,2,4-oxadiazole benzoic acid
US10517853B2 (en) 2015-10-30 2019-12-31 Ptc Therapeutics, Inc. Methods for treating epilepsy
WO2020007822A1 (fr) 2018-07-02 2020-01-09 Conservatoire National Des Arts Et Metiers (Cnam) Nanoparticules de bismuth métallique (0), procédé de fabrication et utilisations de celles-ci

Also Published As

Publication number Publication date
CA2329712A1 (fr) 1999-10-28
NZ507696A (en) 2003-10-31
AU3965199A (en) 1999-11-08
AU750369B2 (en) 2002-07-18
IL139187A0 (en) 2001-11-25

Similar Documents

Publication Publication Date Title
US6004933A (en) Cysteine protease inhibitors
AU750369B2 (en) Cysteine protease inhibitors
US6548638B2 (en) Peptoid and nonpeptoid containing α-keto oxadiazoles as serine protease inhibitors
US6159938A (en) Serine protease inhibitors comprising α-keto heterocycles
US5869455A (en) Serine protease inhibitors-N-substituted derivatives
JP2003535022A6 (ja) セリンプロテアーゼ阻害剤としてのアルファー‐ケトオキサジアゾール含有ペプトイド及び非ペプトイド
KR20010083140A (ko) 유로키나제 및 혈관 형성의 억제제
JPH06504547A (ja) ペプチドケトアミド、ケト酸およびケトエステル
JP2004531466A (ja) 血管新生を調節するための治療薬およびその使用方法
TW523513B (en) Serine protease inhibitors
JPH02212434A (ja) レトロウイルスプロテアーゼインヒビターとしてのペプチドアイソスターの使用
US6001814A (en) Serine protease inhibitors
JPH02256657A (ja) N―置換アミド
KR19990087415A (ko) 세린 프로테아제 억제제
US20040048327A1 (en) Aza-peptide epoxides
US6001813A (en) Val-pro containing α-keto oxadiazoles as serine protease inhibitors
JP2001525315A (ja) 新規メタロプロテアーゼ阻害剤、その治療的利用およびその合成における開始化合物の製造方法
KR20020062312A (ko) 1,3,4-옥사디아졸린 유도체 및 이들을 유효 성분으로하는 약제
US11149067B2 (en) Tailored cyclodepsipeptides as potent non-covalent serine protease inhibitors
NZ517603A (en) Azacycloalkanone serine protease inhibitors
KR20040081182A (ko) 엘라스타제-저해 활성을 가진 이종고리 화합물 및 그 중간체
WO2000009542A1 (fr) INHIBITEURS DE SERINE PROTEASE CONTENANT DES α-CETO HETEROCYCLES
US6100238A (en) Indole and tetrahydroisoquinoline containing Alpha-keto oxadiazoles as serine protease inhibitors
EA000966B1 (ru) Производные пеницилламинамида
JP3125212B2 (ja) 2−ピペラジノン誘導体およびその用途

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref document number: 2329712

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 507696

Country of ref document: NZ

Ref document number: 39651/99

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 139187

Country of ref document: IL

NENP Non-entry into the national phase

Ref country code: KR

WWE Wipo information: entry into national phase

Ref document number: PA/a/2000/010379

Country of ref document: MX

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
WWG Wipo information: grant in national office

Ref document number: 39651/99

Country of ref document: AU