WO2011109355A1 - Compounds for immunoproteasome inhibition - Google Patents

Compounds for immunoproteasome inhibition Download PDF

Info

Publication number
WO2011109355A1
WO2011109355A1 PCT/US2011/026629 US2011026629W WO2011109355A1 WO 2011109355 A1 WO2011109355 A1 WO 2011109355A1 US 2011026629 W US2011026629 W US 2011026629W WO 2011109355 A1 WO2011109355 A1 WO 2011109355A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
hydrogen
aralkyl
aryl
Prior art date
Application number
PCT/US2011/026629
Other languages
English (en)
French (fr)
Inventor
Kevin D. Shenk
Francesco Parlati
Mark K. Bennett
Original Assignee
Onyx Therapeutics, 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=43983939&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2011109355(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to JP2012556160A priority Critical patent/JP6042724B2/ja
Priority to BR112012022060A priority patent/BR112012022060A2/pt
Priority to KR1020127025901A priority patent/KR20130075723A/ko
Priority to AU2011223795A priority patent/AU2011223795B2/en
Priority to MA35248A priority patent/MA34133B1/fr
Priority to CN201180022010XA priority patent/CN102892417A/zh
Priority to SG2012064531A priority patent/SG183843A1/en
Priority to US13/580,112 priority patent/US9359398B2/en
Priority to EA201290844A priority patent/EA201290844A1/ru
Priority to MX2012010017A priority patent/MX2012010017A/es
Application filed by Onyx Therapeutics, Inc. filed Critical Onyx Therapeutics, Inc.
Priority to EP11715320.5A priority patent/EP2542238B1/en
Priority to CA2791651A priority patent/CA2791651C/en
Publication of WO2011109355A1 publication Critical patent/WO2011109355A1/en
Priority to ZA2012/06479A priority patent/ZA201206479B/en
Priority to CU2012000125A priority patent/CU20120125A7/es
Priority to ECSP12012140 priority patent/ECSP12012140A/es

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/4045Indole-alkylamines; Amides thereof, e.g. serotonin, melatonin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • 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
    • C07K5/0827Tripeptides containing heteroatoms different from O, S, or N
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/18Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D209/20Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals substituted additionally by nitrogen atoms, e.g. tryptophane
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/10Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D241/14Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D241/24Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/10Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by doubly bound oxygen or sulphur atoms
    • C07D295/104Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by doubly bound oxygen or sulphur atoms with the ring nitrogen atoms and the doubly bound oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D303/00Compounds containing three-membered rings having one oxygen atom as the only ring hetero atom
    • C07D303/02Compounds containing oxirane rings
    • C07D303/36Compounds containing oxirane rings with hydrocarbon radicals, substituted by nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic Table
    • C07F5/02Boron compounds
    • C07F5/04Esters of boric acids

Definitions

  • proteasome-mediated degradation In eukaryotes, protein degradation is predominately mediated through the ubiquitin pathway in which proteins targeted for destruction are ligated to the 76 amino acid polypeptide ubiquitin. Once targeted, ubiquitinated proteins then serve as substrates for the 26S proteasome, a multicatalytic protease, which cleaves proteins into short peptides through the action of its three major proteolytic activities. While having a general function in intracellular protein turnover, proteasome-mediated degradation also plays a key role in many processes such as major histocompatibility complex (MHC) class I presentation, apoptosis and cell viability, antigen processing, NF- ⁇ activation, and transduction of pro- inflammatory signals.
  • MHC major histocompatibility complex
  • the 20S proteasome is a 700 kDa cylindrical-shaped multicatalytic protease complex comprised of 28 subunits, classified as a- and ⁇ -type, that are arranged in 4 stacked heptameric rings. In yeast and other eukaryotes, 7 different a subunits form the outer rings and 7 different ⁇ subunits comprise the inner rings. The a subunits serve as binding sites for the 19S (PA700) and 1 IS (PA28) regulatory complexes, as well as a physical barrier for the inner proteolytic chamber formed by the two ⁇ subunit rings. Thus, in vivo, the proteasome is believed to exist as a 26S particle ("the 26S proteasome"). In vivo experiments have shown that inhibition of the 20S form of the proteasome can be readily correlated to inhibition of 26S proteasome.
  • N-terminal nucleophile (Ntn) ATTY REF: 26500-0023WO1 hydrolases where the nucleophilic N-terminal residue is, for example, Cys, Ser, Thr, and other nucleophilic moieties.
  • This family includes, for example, penicillin G acylase (PGA), penicillin V acylase (PVA), glutamine PRPP amidotransferase (GAT), and bacterial glycosylasparaginase.
  • PGA penicillin G acylase
  • PVA penicillin V acylase
  • GAT glutamine PRPP amidotransferase
  • bacterial glycosylasparaginase bacterial glycosylasparaginase.
  • higher vertebrates also possess three interferon- ⁇ - inducible ⁇ subunits (LMP7, LMP2 and MECLl), which replace their normal counterparts, ⁇ 5, ⁇ and ⁇ 2, respectively.
  • LMP7, LMP2 and MECLl interferon- ⁇ - inducible ⁇ subunits
  • the proteasome is referred to as an "immunoproteasome".
  • eukaryotic cells can possess two forms of proteasomes in varying ratios.
  • proteolytic activities have been defined for the eukaryote 20S proteasomes: chymotrypsin-like activity (CT-L), which cleaves after large hydrophobic residues; trypsin-like activity (T-L), which cleaves after basic residues; and peptidylglutamyl peptide hydrolyzing activity (PGPH), which cleaves after acidic residues.
  • C-L chymotrypsin-like activity
  • T-L trypsin-like activity
  • PGPH peptidylglutamyl peptide hydrolyzing activity
  • Two additional less characterized activities have also been ascribed to the proteasome: BrAAP activity, which cleaves after branched-chain amino acids; and SNAAP activity, which cleaves after small neutral amino acids.
  • small molecules which have been used to inhibit proteasome activity; however, these compounds generally lack the specificity to delineate between the two forms of the proteasome. Thus, the ability to explore and exploit the roles of each specific proteasome form at the cellular and molecular level has not been possible. Therefore, the creation of small molecule inhibitor(s) that preferentially inhibit a single form of the proteasome is needed to allow the exploration of the roles of each proteasome form at the cellular and molecular level.
  • One aspect of the invention relates to inhibitors that preferentially inhibit
  • the invention relates to the treatment of immune related diseases, comprising administering a ATTY REF: 26500-0023WO1 compound of the invention. In certain embodiments, the invention relates to the treatment of cancer, comprising administering a compound of the invention.
  • One aspect of the invention relates to compounds having a structure of formula (I) or a pharmaceutically acceptable salt thereof,
  • A is optionally a covalent bond when adjacent to an occurrence of Z; Y is absent or is N(R 7 )(R 8 ); M is absent or is Ci_i 2 alkyl; each Z is independently selected from O, S, NH, and N-Ci_ 6 alkyl; or Z is optionally a covalent bond when adjacent to an occurrence of A
  • R 1 is selected from hydrogen, -Ci_ 6 alkyl-Y, Ci_ 6 hydroxyalkyl, Ci_ 6 alkoxyalkyl, aryl, and Ci_ 6aralkyl;
  • R 2 is selected from aryl, heteroaryl, Ci_ 6 aralkyl and Ci_ 6 heteroaralkyl;
  • R 3 is selected from aryl, heteroaryl, Ci_ 6 heteroaralkyl, Ci_ 6 aralkyl and Ci_ 6 alkyl;
  • R 4 and R 5 are each independently selected from hydrogen, Ci_ 6 aralkyl, and Ci_ 6 alkyl; or
  • R 4 and R 5 together are Ci_i 2 alkyl, thereby forming a ring;
  • R 7 is selected from hydrogen, OH, and Ci_ 6 alkyl
  • R 8 is an N-terminal protecting group
  • R 7 and R 8 are independently selected from hydrogen, Ci_ 6 alkyl, and Ci_ 6 aralkyl, preferably hydrogen;
  • R 9 and R 10 are independently selected from hydrogen, metal cation, Ci_ 6 alkyl, Ci_ 6 alkenyl, Ci_ 6 alkynyl, aryl, heteroaryl, Ci_ 6 aralkyl, and Ci_ 6 heteroaralkyl, preferably from hydrogen, metal cation, and Ci_ 6 alkyl, or R 11 and R 12 together are Ci_ 6 alkyl, thereby forming a ring;
  • each R 11 is independently selected from hydrogen and Ci_ 6 alkyl, preferably Ci_ 6 alkyl;
  • R 12 is independently selected from hydrogen, Ci_ 6 alkyl, Ci_ 6 alkenyl, Ci_ 6 alkynyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, Ci_ 6 aralkyl, and Ci_ 6 heteroaralkyl.
  • the invention relates to compounds that are useful as enzyme inhibitors. These compounds are generally useful to inhibit enzymes having a nucleophilic group at the N- terminus. For example, activities of enzymes or enzyme subunits having N-terminal amino acids with nucleophiles in their side chains, such as threonine, serine, or cysteine can be successfully inhibited by the enzyme inhibitors described herein. Activities of enzymes or enzyme subunits having non-amino acid nucleophilic groups at their N-termini, such as, for example, protecting groups or carbohydrates, can also be successfully inhibited by the enzyme inhibitors described herein.
  • N- terminal nucleophiles of Ntn form covalent adducts with the boronic acid or boronic ester functional group of the enzyme inhibitors as described herein.
  • Peptides can have a repeating backbone structure with side chains extending from the backbone units.
  • each backbone unit has a side chain associated with it, although in some cases, the side chain is a hydrogen atom. In other embodiments, not every backbone unit has an associated side chain.
  • the side chains extending from the backbone units can include natural aliphatic or aromatic amino acid side chains, such as hydrogen (glycine), methyl (alanine), isopropyl (valine), sec-butyl (isoleucine), isobutyl (leucine), phenylmethyl (phenylalanine), and the side chain constituting the amino acid proline.
  • the side chains can also be other branched or unbranched aliphatic or aromatic groups such as ethyl, n-propyl, n-butyl, t-butyl, and aryl substituted derivatives such as 1-phenylethyl, 2-phenylethyl, (l-naphthyl)methyl, (2- naphthyl)methyl, l-(l-naphthyl)ethyl, l-(2-naphthyl)ethyl, 2-(l-naphthyl)ethyl, 2-(2- naphthyl)ethyl, and similar compounds.
  • aryl substituted derivatives such as 1-phenylethyl, 2-phenylethyl, (l-naphthyl)methyl, (2- naphthyl)methyl, l-(l-naphthyl)ethyl, l-(2-naphthyl)e
  • the aryl groups can be further substituted with branched or unbranched Ci_ 6 alkyl groups, or substituted alkyl groups, acetyl and the like, or further aryl groups, or substituted aryl groups, such as benzoyl and the like. Heteroaryl groups can also be used as side chain substituents.
  • Heteroaryl groups include nitrogen-, oxygen-, and sulfur-containing aryl groups such as thienyl, benzothienyl, naphthothienyl, thianthrenyl, furyl, pyranyl, isobenzofuranyl, chromenyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, indolyl, purinyl, quinolyl, and the like.
  • aryl groups such as thienyl, benzothienyl, naphthothienyl, thianthrenyl, furyl, pyranyl, isobenzofuranyl, chromenyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, indolyl, purinyl, quinolyl, and the like.
  • polar or charged residues can be introduced into the peptide boronates.
  • naturally occurring amino acids such as hydroxy-containing (Thr, Tyr, Ser) or sulfur-containing (Met, Cys) can be introduced, as well as non-essential amino acids, for example, taurine, carnitine, citrulline, cystine, ornithine, norleucine and others.
  • Non-naturally occurring side chain substituents with charged or polar moieties can also be included, such as, for example, Cl-6alkyl chains or C6-12aryl groups with one or more hydroxy, short chain alkoxy, sulfide, thio, carboxyl, ester, phospho, amido or amino groups, or such substituents substituted with one or more halogen atoms.
  • R is the side chain.
  • Such a designation does not exclude the naturally occurring amino ATTY REF: 26500-0023WO1 acid proline, or other non-naturally occurring cyclic secondary amino acids, which will be recognized by those of skill in the art.
  • the backbone units are N-alkylated amide units (for example, N-methyl and the like), olefmic analogs (in which one or more amide bonds are replaced by olefmic bonds), tetrazole analogs (in which a tetrazole ring imposes a cis-configuration on the backbone), or combinations of such backbone linkages.
  • the amino acid a-carbon is modified by a-alkyl substitution, for example, aminoisobutyric acid.
  • side chains are locally modified, for example, by ⁇ or ⁇ dehydro modification, in which a double bond is present between the a and ⁇ atoms of the side chain, or for example by ⁇ or ⁇ cyclopropyl modification, in which a cyclopropyl group is present between the a and ⁇ atoms of the side chain.
  • D-amino acids can be used.
  • Further embodiments can include side chain-to-backbone cyclization, disulfide bond formation, lactam formation, azo linkage, and other modifications discussed in “Peptides and Mimics, Design of Conformationally Constrained" by Hruby and Boteju, in "Molecular Biology and Biotechnology: A
  • the invention relates to compounds having a structure of formula (I) or a pharmaceutically acceptable salt thereof,
  • A is optionally a covalent bond when adjacent to an occurrence
  • Y is absent or is N(R 7 )(R 8 );
  • M is absent or is Ci_i 2 alkyl; ATTY REF: 26500-0023WO1 each Z is independently selected from O, S, NH, and N-Ci_ 6 alkyl; or
  • Z is optionally a covalent bond when adjacent to an occurrence of A
  • R 1 is selected from hydrogen, -Ci_ 6 alkyl-Y, Ci_ 6 hydroxyalkyl, Ci_ 6 alkoxyalkyl, aryl, and Ci_ 6aralkyl;
  • R 2 is selected from aryl, heteroaryl, Ci_ 6 aralkyl and Ci_ 6 heteroaralkyl;
  • R 3 is selected from aryl, heteroaryl, Ci_ 6 heteroaralkyl, Ci_ 6 aralkyl and Ci_ 6 alkyl;
  • R 4 and R 5 are each independently selected from hydrogen, Ci_ 6 aralkyl, and Ci_ 6 alkyl; or
  • R 4 and R 5 together are Ci_i 2 alkyl, thereby forming a ring;
  • R 6 is selected from hydrogen, Ci_ 6 alkyl, Ci_ 6 alkenyl, Ci_ 6 alkynyl, carbocyclyl, heterocyclyl, an N-terminal protecting group, aryl, Ci_ 6 aralkyl, heteroaryl, Ci_ 6 heteroaralkyl,
  • R 7 is selected from hydrogen, OH, and Ci_ 6 alkyl
  • R 8 is an N-terminal protecting group
  • R 7 and R 8 are independently selected from hydrogen, Ci_ 6 alkyl, and Ci_ 6 aralkyl, preferably hydrogen;
  • R 9 and R 10 are independently selected from hydrogen, metal cation, Ci_ 6 alkyl, Ci_ 6 alkenyl, Ci_ 6 alkynyl, aryl, heteroaryl, Ci_ 6 aralkyl, and Ci_ 6 heteroaralkyl, preferably from hydrogen, metal cation, and Ci_ 6 alkyl, or R 11 and R 12 together are Ci_ 6 alkyl, thereby forming a ring;
  • each R 11 is independently selected from hydrogen and Ci_ 6 alkyl, preferably Ci_ 6 alkyl;
  • R 12 is independently selected from hydrogen, Ci_ 6 alkyl, Ci_ 6 alkenyl, Ci_ 6 alkynyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, Ci_ 6 aralkyl, and Ci_ 6 heteroaralkyl.
  • R 1 is -Ci_ 6 alkyl-Y.
  • R 1 is substituted with one or more substituents selected from amide, amine, carboxylic acid (or a salt thereof), ester (including Ci_ 6 alkyl ester, Ci_ 5 alkyl ester, and aryl ester), thiol, or ATTY REF: 26500-0023WO1 thioether.
  • R 1 is substituted with one or more substituents selected from carboxylic acid and ester.
  • Y is absent and R 1 is selected from methyl, ethyl, isopropyl, carboxymethyl, and benzyl, preferably methyl.
  • R 2 is selected from Ci_ 6 aralkyl and Ci_ 6 heteroaralkyl.
  • R 2 is selected from Ci_ 6 alkyl-phenyl, Ci_6alkyl-indolyl, Ci_ 6 alkyl- thienyl, Ci_ 6 alkyl-thiazolyl, and Ci_ 6 alkyl-isothiazolyl, wherein the alkyl moiety may contain six, five, four, three, two, or one carbon atoms, preferably one or two.
  • R 2 is substituted with one or more substituents selected from hydroxy, halogen, amide, amine, carboxylic acid (or a salt thereof), ester (including Ci_ 6 alkyl ester, Ci_ 5 alkyl ester, and aryl ester), thiol, or thioether.
  • R 2 is substituted with a substituent selected from alkyl, trihaloalkyl, alkoxy, hydroxy, or cyano, preferably hydroxy or alkoxy, such as methoxy.
  • R 2 is selected from Ci_6alkyl- phenyl and Ci_6alkyl-indolyl.
  • R 2 is selected from
  • R H or any suitable protecting group
  • D is selected from H, OMe, OBu , OH, CN, CF 3 and CH 3 .
  • D is selected from H, OMe, OH, CN, CF 3 and CH 3 .
  • D is attached at the 4-position relative to the point of attachment, preferably excluding embodiments where the 4-position of the ring is occupied by the nitrogen of a pyridine ring.
  • R 3 is selected from Ci_ 6 aralkyl and Ci_ 6 alkyl.
  • the alkyl moiety may contain six, five, four, three, two, or one carbon atoms, ATTY REF: 26500-0023WO1 preferably one or two.
  • R 3 is substituted with one or more substituents selected from hydroxy, halogen, amide, amine, carboxylic acid (or a salt thereof), ester (including Ci_ 6 alkyl ester, Ci_ 5 alkyl ester, and aryl ester), thiol, or thioether.
  • R 3 is substituted with a substituent selected from alkyl, trihaloalkyl, alkoxy, hydroxy, or cyano.
  • R 3 is s Ci_ 6 alkyl-phenyl.
  • R 3 is selected from
  • D is selected from H, OMe, OBu , OH, CN, CF 3 or CH 3 .
  • D is selected from H, OMe, OH, CN, CF 3 or CH 3 .
  • R 4 and R 5 are independently selected from hydrogen and
  • Ci_ 6 alkyl preferably R 4 and R 5 are both hydrogen. In certain alternative embodiments, R 4 and R 5 together are Ci_i 2 alkyl, thereby forming a ring. In certain preferred such
  • R 4 and R 5 together with the two adjacent oxygen atoms and the boron form a pinacol boronate ester.
  • R 6 is selected from carbocyclyl, aryl, and heterocyclylM-. In certain such embodiments, R 6 is carbocyclyl or aryl. In certain such embodiments, R 6 is an indene, such as methylindene, particularly 3-methylindene. In certain alternative,
  • R 6 is heterocyclylM-, wherein the heterocyclyl is selected from morpholino, piperidino, piperazino, and pyrrolidino, preferably morpholino.
  • M is Ci_i 2 alkyl.
  • the stereochemical configuration of the carbons bearing R 1 , R 2 , or R 3 are independently D or L. In certain preferred embodiments, the stereochemical configuration of at least one of the carbons bearing R 1 , R 2 , and R 3 respectively is D. In certain preferred such embodiments, the stereochemical configuration of the carbon bearing R 1 is D. In certain such embodiments, the stereochemical configuration of the carbon bearing R 2 is D. In certain such embodiments, the stereochemical configuration of the carbon bearing R 3 is D. In certain embodiments the stereochemical configuration of at least two of the carbons bearing R 1 , R 2 , and R 3 respectively is D. In yet another preferred embodiment, the ATTY REF: 26500-0023WO1 stereochemical configuration of all three of the carbons bearing R 1 , R 2 , and R 3 respectively is D.
  • One aspect of the invention relates to inhibitors that preferentially inhibit
  • the EC50 ratio of a compound of formula I in an assay of constitutive proteasome activity as compared to the EC50 of the same compound in an assay of immunoproteasome activity is greater than 1. In certain such embodiments, the EC50 is greater than 2, 3, 4 or even 5.
  • C x _ y alkyl refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups that contain from x to y carbons in the chain, including haloalkyl groups such as trifluoromethyl and 2,2,2- tirfluoroethyl, etc.
  • Coalkyl indicates a hydrogen where the group is in a terminal position, a bond if internal.
  • C2_ y alkenyl and “C2_ y alkynyl” refer to substituted or unsubstituted unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
  • alkoxy refers to an alkyl group having an oxygen attached thereto.
  • alkoxy groups include methoxy, ethoxy, propoxy, tert-butoxy and the like.
  • An "ether" is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxy.
  • Ci_ 6 alkoxyalkyl refers to a Ci_ 6 alkyl group substituted with an alkoxy group, thereby forming an ether.
  • Ci_ 6 aralkyl refers to a Ci_ 6 alkyl group substituted with an aryl group.
  • amine and “amino” are art-recognized and refer to both unsubstituted and substituted amines and salts thereof, e.g., a moiety that can be represented by the general formulae: ATTY REF: 26500-0023WO1
  • R 9 , R 10 and R 10 each independently represent a hydrogen, an alkyl, an alkenyl, -(Cl3 ⁇ 4)m-R 8 , or R 9 and R 10 taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure;
  • R 8 represents an aryl, a cycloalkyl, a cycloalkenyl, a heterocyclyl or a polycyclyl; and
  • m is zero or an integer from 1 to 8.
  • only one of R 9 or R 10 can be a carbonyl, e.g., R 9 , R 10 , and the nitrogen together do not form an imide.
  • R 9 and R 10 each independently represent a hydrogen, an alkyl, an alkenyl, or - (CI3 ⁇ 4)m-R 8 .
  • an amino group is basic, meaning it has a pKa > 7.00. The protonated forms of these functional groups have pK a s above 7.00.
  • amide and “amido” are art-recognized as an amino-substituted carbonyl and includes a moiety that can be represented by the general formula:
  • R 9 , R 10 are as defined above.
  • Preferred embodiments of the amide will not include imides which may be unstable.
  • aryl as used herein includes 5-, 6-, and 7-membered substituted or unsubstituted single-ring aromatic groups in which each atom of the ring is carbon.
  • aryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Aryl groups include benzene, naphthalene, phenanthrene, phenol, aniline, and the like.
  • carrier refers to a non-aromatic substituted or unsubstituted ring in which each atom of the ring is carbon.
  • carrier also include polycyclic ring systems having two or more ATTY REF: 26500-0023WO1 cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is carbocyclic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • carbonyl is art-recognized and includes such moieties as can be represented by the general formula:
  • X is a bond or represents an oxygen or a sulfur
  • R 11 represents a hydrogen, an alkyl, an alkenyl, -(CI3 ⁇ 4) m -R 8 or a pharmaceutically acceptable salt
  • R 11 represents a hydrogen, an alkyl, an alkenyl or -(CI3 ⁇ 4) m -R 8 , where m and R 8 are as defined above.
  • X is an oxygen and R 11 or R 11 is not hydrogen, the formula represents an
  • enzyme can be any partially or wholly proteinaceous molecule which carries out a chemical reaction in a catalytic manner.
  • Such enzymes can be native enzymes, fusion enzymes, proenzymes, apoenzymes, denatured enzymes, farnesylated enzymes, ubiquitinated enzymes, fatty acylated enzymes, gerangeranylated enzymes, GPI- linked enzymes, lipid-linked enzymes, prenylated enzymes, naturally-occurring or artificially-generated mutant enzymes, enzymes with side chain or backbone modifications, enzymes having leader sequences, and enzymes complexed with non-proteinaceous material, such as proteoglycans, proteo liposomes.
  • Enzymes can be made by any means, including natural expression, promoted expression, cloning, various solution-based and solid-based peptide syntheses, and similar methods known to those of skill in the art.
  • Ci_ 6 heteroaralkyl refers to a Ci_ 6 alkyl group substituted with a heteroaryl group.
  • heteroaryl includes substituted or unsubstituted aromatic 5- to 7- membered ring structures, more preferably 5- to 6-membered rings, whose ring structures include one to four heteroatoms.
  • heteroaryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining ATTY REF: 26500-0023WO1 rings wherein at least one of the rings is heteroaromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, isoxazole, oxazole, oxadiazole, thiazole, thiadiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like.
  • heteroatom as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, phosphorus, and sulfur.
  • heterocyclyl or “heterocyclic group” refer to substituted or unsubstituted non-aromatic 3- to 10-membered ring structures, more preferably 3- to 7-membered rings, whose ring structures include one to four heteroatoms.
  • heterocyclyl or “heterocyclic group” refer to substituted or unsubstituted non-aromatic 3- to 10-membered ring structures, more preferably 3- to 7-membered rings, whose ring structures include one to four heteroatoms.
  • heterocyclyl or
  • heterocyclic group also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heterocyclic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Heterocyclyl groups include, for example, tetrahydropyran, piperidine, piperazine, pyrrolidine, morpholine, lactones, lactams, and the like.
  • Ci_ 6 hydroxyalkyl refers to a Ci_ 6 alkyl group substituted with a hydroxy group.
  • inhibitor is meant to describe a compound that blocks or reduces an activity of an enzyme.
  • An inhibitor can act with competitive, uncompetitive, or noncompetitive inhibition.
  • An inhibitor can bind reversibly or irreversibly, and therefore the term includes compounds that are suicide substrates of an enzyme.
  • An inhibitor can modify one or more sites on or near the active site of the enzyme, or it can cause a conformational change elsewhere on the enzyme.
  • peptide includes not only standard amide linkage with standard a-substituents, but commonly utilized peptidomimetics, other modified linkages, non-naturally occurring side chains, and side chain modifications, as detailed below.
  • polycyclyl or “polycyclic” refer to two or more rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls) in which two or more ATTY REF: 26500-0023WO1 carbons are common to two adjoining rings, e.g., the rings are "fused rings".
  • rings of the polycycle can be substituted or unsubstituted.
  • preventing is art-recognized, and when used in relation to a condition, such as a local recurrence (e.g., pain), a disease such as cancer, a syndrome complex such as heart failure or any other medical condition, is well understood in the art, and includes administration of a composition which reduces the frequency of, or delays the onset of, symptoms of a medical condition in a subject relative to a subject which does not receive the composition.
  • a condition such as a local recurrence (e.g., pain)
  • a disease such as cancer
  • a syndrome complex such as heart failure or any other medical condition
  • prevention of cancer includes, for example, reducing the number of detectable cancerous growths in a population of patients receiving a prophylactic treatment relative to an untreated control population, and/or delaying the appearance of detectable cancerous growths in a treated population versus an untreated control population, e.g., by a statistically and/or clinically significant amount.
  • Prevention of an infection includes, for example, reducing the number of diagnoses of the infection in a treated population versus an untreated control population, and/or delaying the onset of symptoms of the infection in a treated population versus an untreated control population.
  • Prevention of pain includes, for example, reducing the magnitude of, or alternatively delaying, pain sensations experienced by subjects in a treated population versus an untreated control population.
  • prodrug encompasses compounds that, under physiological conditions, are converted into therapeutically active agents.
  • a common method for making a prodrug is to include selected moieties that are hydrolyzed under physiological conditions to reveal the desired molecule.
  • the prodrug is converted by an enzymatic activity of the host animal.
  • prophylactic or therapeutic treatment is art-recognized and includes administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic, (i.e., it protects the host against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).
  • the unwanted condition e.g., disease or other unwanted state of the host animal
  • substituted refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that “substitution” or
  • substituted with includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc.
  • substituted is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
  • Substituents can include, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thio formate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a s
  • a “therapeutically effective amount” of a compound with respect to the subject method of treatment refers to an amount of the compound(s) in a preparation which, when administered as part of a desired dosage regimen (to a mammal, preferably a human) alleviates a symptom, ameliorates a condition, or slows the onset of disease conditions according to clinically acceptable standards for the disorder or condition to be treated or the cosmetic purpose, e.g., at a reasonable benefit/risk ratio applicable to any medical treatment.
  • thioether refers to an alkyl group, as defined above, having a sulfur moiety attached thereto.
  • the "thioether” is represented by -S- alkyl.
  • Representative thioether groups include methylthio, ethylthio, and the like.
  • treating includes reversing, reducing, or arresting the symptoms, clinical signs, and underlying pathology of a condition in manner to improve or stabilize a subject's condition.
  • proteasome inhibition has been suggested as a prevention and/or treatment of a multitude of diseases including, but not limited to, proliferative diseases, neurotoxic/degenerative diseases, ischemic conditions, inflammation, immune-related diseases, HIV, cancers, organ graft rejection, septic shock, viral and parasitic infections, conditions associated with acidosis, macular degeneration, pulmonary conditions, muscle wasting diseases, fibrotic diseases, bone and hair growth diseases.
  • Proteasome inhibitors can be used to treat conditions mediated directly by the proteolytic function of the proteasome such as muscle wasting, or mediated indirectly via proteins which are processed by the proteasome such as NF- ⁇ .
  • the proteasome participates in the rapid elimination and post-translational processing of proteins (e.g., enzymes) involved in cellular regulation (e.g., cell cycle, gene transcription, and metabolic pathways), intercellular communication, and the immune response (e.g., antigen presentation).
  • proteasome inhibitors inhibit both the constitutive and immuno forms of the proteasome. Even bortezomib, the only FDA-approved proteasome inhibitor for the treatment of relapsed multiple myeloma patients, does not distinguish between the two forms (Altun et al., Cancer Res 65:7896, 2005). Thus, what is known about therapeutic proteasome inhibition is based on work with molecules that inhibit both forms of the proteasome. Accordingly, compounds of the invention may be beneficial for reducing the severity of side effects associated with molecules that inhibit both forms of the proteasome.
  • Immunoproteasome expression occurs predominantly in cells and organs that make up the lymphatic system, such as white blood cells(leukocytes), bone marrow, and the thymus, spleen and lymph nodes. Although some organs preferentially express constitutive ATTY REF: 26500-0023WO1 proteasomes (e.g., heart), others such as adrenal, liver, lung and gut, appear to express both forms.
  • the immune system of which leukocytes and lymphoid tissues play a major role, is responsible for protecting an organism from outside biological influences. When functioning properly, it protects the body against bacterial and viral infections.
  • the immune system also screens for autologous cells that have undergone oncogenic transformation. Intracellular proteolysis generates small peptides for presentation to T-lymphocytes to induce MHC class I-mediated immune responses.
  • the proteasome is the main provider of these precursor peptides, however, differences between antigenic peptides have been observed between cells with varying amounts of each proteasome form (Cascio et al., EMBO J 20:2357-2366, 2001).
  • the invention relates to a method for inhibiting antigen presentation in a cell, including exposing the cell to a compound as described herein.
  • the invention relates to a method for altering the repertoire of antigenic peptides produced by the proteasome or other Ntn with multicatalytic activity. For example, if the activity of the immunoproteasome proteasome is selectively inhibited, a different set of antigenic peptides may be produced by the remaining constitutive proteasome and presented in MHC molecules on the surfaces of cells than would be produced and presented without any enzyme inhibition.
  • a further embodiment is a method for suppressing the immune system of a subject, comprising administering to the subject an effective amount of a proteasome inhibitor compound in a manner described herein.
  • Immunodeficiency disorders occur when a part of the immune system is not working properly or is not present. They can affect B lymophyctes, T lymphocytes, or phagocytes and be either inherited (e.g., IgA deficiency, severe combined immunodeficiency (SCID), thymic dysplasia and chronic granulomatous) or acquired (e.g., acquired immunodeficiency syndrome (AIDS), human immunodeficiency virus (HIV) and drug-induced ATTY REF: 26500-0023WO1 immunodeficiencies).
  • a dosing strategy utilizing selective proteasome inhibitors of the invention may be used to treat immune -related conditions such as immunodeficiency disorders.
  • autoimmune disorders In autoimmune disorders, the immune system inappropriately attacks the body's healthy organs and tissues as if they were foreign invaders.
  • An example of an autoimmune disease is Sjogren's Syndrome, which is characterized by infiltration and focal accumulation of lymphocytes in the exocrine glands.
  • Sjogren's Syndrome Sjogren's Syndrome, which is characterized by infiltration and focal accumulation of lymphocytes in the exocrine glands.
  • a study examining the proteasome expression level revealed a significant up-regulation of beta5i (LMP7) exclusively in the salivary glands of SS patients (Egerer et al., Arthritis Rheum 54: 1501-8, 2006).
  • immune-related conditions include lupus (such as lupus nephritis and systemic lupus erythamotosus), rheumatoid arthritis (such as juvenile rheumatoid arthritis and psoriatic rheumatoid arthritis), scleroderma, ankylosing spondylitis, dermatomyositis, psoriasis, multiple sclerosis (both relapsing remitting and chronic progressive forms) and inflammatory bowel disease (such as ulcerative colitis and Crohn's disease).
  • Tissue/organ transplant rejection occurs when the immune system mistakenly attacks the cells being introduced to the host's body.
  • GVHD graft versus host disease
  • Inflammation is the first response of the immune system to infection or irritation.
  • a cellular component of inflammation involves the movement of leukocytes, which express immunoproteasome, from blood vessels into the inflamed tissue. These cells take on the important role of removing the irritant, bacteria, parasite or cell debris.
  • Proteasome inhibitors are already known to have anti-inflammatory activity (Meng et al, PNAS
  • the invention relates to a method of treating inflammation and inflammatory diseases comprising administering a compound as described herein.
  • Inflammatory diseases include acute (e.g., bronchitis, ATTY REF: 26500-0023WO1 conjunctivitis, pancreatitis) and chronic conditions (e.g., chronic cholecstitis, bronchiectasis, aortic valve stenosis, restenosis, psoriasis and arthritis), along with conditions associated with inflammation such as fibrosis, infection and ischemia.
  • the invention relates to methods of treating conditions whereby an organ is inflamed, such as autoimmune myocarditis and autoimmune thyroiditis.
  • tissue damage including damage due to the inflammation process, progression of regeneration and repair begins.
  • lost tissue is replaced by proliferation of cells of the same type, which reconstruct the normal architecture.
  • improper regeneration of the tissue architecture may have severe consequences.
  • the regenerated tissue forms an abnormal nodular architecture leading to cirrhosis and portal hypertension.
  • the repair process results in lost tissue being replaced by a fibrous scar which is produced from granulation tissue. Fibrosis is the excessive and persistent formation of scar tissue resulting from the
  • fibroblasts hyperproliferative growth of fibroblasts and is associated with activation of the TGF- ⁇ signaling pathway. Fibrosis involves extensive deposition of extracellular matrix and can occur within virtually any tissue or across several different tissues. Normally, the level of intracellular signaling protein (Smad) that activate transcription of target genes upon TGF- ⁇ stimulation is regulated by proteasome activity (Xu et al, 2000). However, accelerated degradation of the TGF- ⁇ signaling components has been observed in cancers and other hyperproliferative conditions.
  • Smad intracellular signaling protein
  • the invention relates to a method for treating a hyperproliferative condition selected from diabetes (such as type 1 , type 2 and metabolic syndrome), diabetic retinopathy, macular degeneration, diabetic nephropathy, glomerulosclerosis, IgA nephropathy, cirrhosis, biliary atresia, congestive heart failure, scleroderma, radiation-induced fibrosis, and lung fibrosis (idiopathic pulmonary fibrosis, collagen vascular disease, sarcoidosis, interstitial lung diseases and extrinsic lung disorders), comprising administering a compound as described herein.
  • diabetes such as type 1 , type 2 and metabolic syndrome
  • diabetic retinopathy macular degeneration
  • diabetic nephropathy glomerulosclerosis
  • IgA nephropathy cirrhosis
  • biliary atresia congestive heart failure
  • scleroderma radiation-induced fibrosis
  • the invention relates to the topical or systemic administration of a compound as described herein to treat burns. Wound closure following surgery is often associated with disfiguring scars, which may be prevented by inhibition of fibrosis. Thus, in certain embodiments, the invention relates to a ATTY REF: 26500-0023WO1 method for the prevention or reduction of scarring, comprising administering a compound as described herein.
  • LPS lipopolysaccharide
  • TNFa systemic inflammatory response syndrome
  • the invention relates to preventing or treating septic shock, comprising administering a compound as disclosed herein.
  • compositions are useful for the treatment of a parasitic infection, such as infections caused by protozoan parasites.
  • a parasitic infection such as infections caused by protozoan parasites.
  • the proteasome of these parasites is considered to be involved primarily in cell differentiation and replication activities (Paugam et al., Trends Parasitol. 2003, 19(2): 55-59).
  • entamoeba species have been shown to lose encystation capacity when exposed to proteasome inhibitors (Gonzales, et al, Arch. Med. Res. 1997, 28, Spec No: 139-140). In certain such
  • the compounds as disclosed herein are useful for the treatment of parasitic infections in humans caused by a protozoan parasite selected from Plasmodium sps.
  • Trypanosoma sps. including T. cruzi, which causes Chagas' disease, and T. brucei which causes African sleeping sickness
  • Leishmania sps. including L. amazonesis, L. donovani, L. infantum, L. mexicana, etc.
  • Pneumocystis carinii a protozoan known to cause pneumonia in AIDS and other immunosuppressed patients
  • Toxoplasma gondii Entamoeba histolytica
  • Entamoeba invadens and Giardia lamblia.
  • the disclosed ATTY REF: 26500-0023WO1 compositions are useful for the treatment of parasitic infections in animals and livestock caused by a protozoan parasite selected from Plasmodium hermani, Cryptosporidium sps., Echinococcus granulosus, Eimeria tenella, Sarcocystis neurona, and Neurospora crassa.
  • a protozoan parasite selected from Plasmodium hermani, Cryptosporidium sps., Echinococcus granulosus, Eimeria tenella, Sarcocystis neurona, and Neurospora crassa.
  • Other compounds useful as proteasome inhibitors in the treatment of parasitic diseases are described in WO 98/10779, which is incorporated herein by reference in its entirety.
  • compounds disclosed herein may inhibit proteasome activity in a parasite without recovery in white blood cells.
  • the long half-life of blood cells may provide prolonged protection with regard to therapy against recurring exposures to parasites.
  • the compounds as described herein may provide prolonged protection with regard to chemoprophylaxis against future infection.
  • Viral infections contribute to the pathology of many diseases.
  • Heart conditions such as ongoing myocarditis and dilated cardiomyopathy have been linked to the coxsackievirus B3.
  • all three immunoproteasome subunits were uniformly up-regulated in hearts of mice which developed chronic myocarditis (Szalay et al., Am J Pathol 168: 1542-52, 2006).
  • Some viruses utilize the ubiquitin-proteasome system in the viral entry step where the virus is released from the endosome into the cytosol.
  • the mouse hepatitis virus (MHV) belongs to the Coronaviridae family, which also includes the severe acute respiratory syndrome (SARS) coronvirus.
  • HBV human hepatitis B virus
  • A, C, D and E may also utilize the ubiquitin-proteasome degradation pathway for secretion, morphogenesis and
  • the invention relates to treating a viral infection, comprising administering a compound as disclosed herein.
  • Listeria monocytogenes causes a condition known as listeriosis, the manifestations of which range from mild (nausea, vomiting and diarrhea) to severe ATTY REF: 26500-0023WO1
  • proteasome subunit composition revealed that infection of mice with lymphocytic choriomeningitis virus or Listeria monocytogenes lead to an almost complete replacement of constitutive
  • Prokaryotic infections may include diseases caused by either mycobacteria (such as tuberculosis, leprosy or Buruli Ulcer) or archaebacteria.
  • the invention relates to a method for treating infection (e.g., bacterial, parasitic or viral), comprising contacting a cell with a compound as disclosed herein.
  • infection e.g., bacterial, parasitic or viral
  • the invention relates to a method for treating infection, comprising administering a compound as disclosed herein.
  • the invention relates to a method of treating an ischemic condition or reperfusion injury, comprising administering a compound as disclosed herein.
  • Such conditions or injuries include, but are not limited to, acute coronary syndrome (vulnerable plaques), arterial occlusive disease (cardiac, cerebral, peripheral arterial and vascular occlusions), atherosclerosis (coronary sclerosis, coronary artery disease), infarctions, heart failure, pancreatitis, myocardial hypertrophy, stenosis, and restenosis.
  • Cachexia is a syndrome characterized by wasting of skeletal muscle associated with enhanced proteolysis due to the ubiquitin-proteasome pathway. Inhibiting the proteasome reduces proteolysis, thereby reducing both muscle protein loss and the nitrogenous load on kidneys or liver (Tawa et al, JCI 100: 197-203, 1997). In cachexia, elevated expression of proinflammatory cytokines, TNF-a and IFN- ⁇ , both of which stimulate expression of immunoproteasome subunits, have been reported (Acharyya et al, JCI 114:370-378, 2004). In fact, most types of muscle atrophy exhibit elevated rates of protein degradation (Lecker et al., FASEB J 18:39-51, 2004).
  • the invention relates to the treatment of cachexia or muscle -wasting disease, comprising administering a compound as disclosed herein.
  • Compounds of the invention may be useful for treating conditions such as cancer, chronic infectious diseases, fever, muscle disuse (atrophy) and denervation, nerve injury, fasting, renal failure associated with acidosis, and hepatic failure. See, e.g., Goldberg, U.S. Patent No. 5,340,736.
  • proteasome inhibitors block both degradation and processing of ubiquitinated NF- ⁇ in vitro and in vivo.
  • proteasome inhibitors also block ⁇ - ⁇ degradation and NF- ⁇ activation (Palombella et al, Cell (1994) 78:773-785; and Traenckner et al, EMBO J. (1994) 13:5433- 5441).
  • the invention relates to a method for inhibiting ⁇ - ⁇ degradation, comprising contacting the cell with a compound as described herein.
  • the invention relates to methods for affecting cyclin- dependent eukaryotic cell cycles, including exposing a cell (in vitro or in vivo) to a compound as disclosed herein.
  • Cyclins are proteins involved in cell cycle control. The proteasome participates in the degradation of cyclins. Examples of cyclins include mitotic cyclins, Gl cyclins, and cyclin B. Degradation of cyclins enables a cell to exit one cell cycle stage (e.g., mitosis) and enter another (e.g., division). It is believed all cyclins are associated with p34 cdc2 protein kinase or related kinases.
  • proteolysis targeting signal is localized to amino acids 42- 50 (destruction box).
  • cyclin is converted to a form ATTY REF: 26500-0023WO1 vulnerable to a ubiquitin ligase or that a cyclin-specific ligase is activated during mitosis (Ciechanover, A., Cell, (1994) 79: 13-21).
  • Inhibition of the proteasome inhibits cyclin degradation, and therefore inhibits cell proliferation, for example, in cyclin-related cancers (Kumatori et al, Proc. Natl. Acad. Sci. USA (1990) 87:7071-7075).
  • the invention relates to a method for treating a proliferative disease in a subject (e.g., cancer, psoriasis, or restenosis), comprising administering a compound as disclosed herein.
  • a proliferative disease e.g., cancer, psoriasis, or restenosis
  • the invention also relates to a method for treating cyclin-related inflammation, comprising adminstering a compound as described herein.
  • the invention relates to methods for reducing the rate of intracellular protein degradation in a cell. Each of these methods comprises contacting a cell (in vivo or in vitro) with a compound as disclosed herein.
  • AD Alzheimer's disease
  • Pathological hallmarks of the disease include senile amyloid plaques, neurofibrillary tangles, dystrophic neuritis and significant neuronal loss in selected regions of the brain.
  • Microglia the resident macrophages in the brain, release numerous proinflammatory cytokines, including TNF-a, when activated by ⁇ 42, a peptide associated with neuritic and vascular amyloid plaques. This microglial- mediated inflammatory response contributes to significant neuronal loss.
  • HD Huntington's disease
  • another neurodegenerative disorder display motor dysfunction and cognitive decline over a period of years until death.
  • Upon autopsy the presence of inclusions or intraneuronal aggregates, caused by a polyQ expansion mutation (also referred to as a CAG triplet repeat expansion), can be detected, accompanied by significant atrophy in the striatum and cortex portions of the brain.
  • Neurodegenerative diseases and conditions include, but are not limited to, stroke, ischemic damage to the nervous system, neural trauma (e.g., percussive brain damage, spinal cord injury, and traumatic damage to the nervous system), multiple sclerosis and other immune-mediated neuropathies (e.g., Guillain-Barre syndrome and its variants, acute motor axonal neuropathy, acute inflammatory demyelinating polyneuropathy, and Fisher
  • HIV/ AIDS dementia complex HIV/AIDS dementia complex, axonomy, diabetic neuropathy, Parkinson's disease, Huntington's disease, multiple sclerosis, bacterial, parasitic, fungal, and viral meningitis, encephalitis, vascular dementia, multi-infarct dementia, Lewy body dementia, frontal lobe dementia such as Pick's disease, subcortical dementias (such as Huntington or progressive supranuclear palsy), focal cortical atrophy syndromes (such as primary aphasia), metabolic-toxic dementias (such as chronic hypothyroidism or B12 deficiency), and dementias caused by infections (such as syphilis or chronic meningitis).
  • axonomy diabetic neuropathy
  • Parkinson's disease Huntington's disease
  • multiple sclerosis bacterial, parasitic, fungal, and viral meningitis
  • encephalitis vascular dementia
  • multi-infarct dementia Lewy body dementia
  • frontal lobe dementia such as Pick
  • proteasome inhibitors that bind to the 20S proteasome stimulate bone formation in bone organ cultures. Furthermore, when such inhibitors have been administered systemically to mice, certain proteasome inhibitors increased bone volume and bone formation rates over 70% (Garrett, I. R. et al., J. Clin. Invest. (2003) 111 : 1771- 1782), therefore suggesting that the ubiquitin-proteasome machinery regulates osteoblast differentiation and bone formation. Therefore, the disclosed proteasome inhibitor compositions may be useful in the treatment and/or prevention of diseases associated with bone loss, such as osteroporosis. ATTY REF: 26500-0023WO1
  • Cancer is a general term for disease characterized by uncontrolled, abnormal growth of cells. Many cancers arise via multistep pathways involving inactivation of tumor suppressor proteins and activation of oncogenic peptides. Cancer cells can spread to other parts of the body through the lymphatic system or blood stream. Usually, cancer is classified according to the type of tissue or cell most prominently involved. As noted previously, proteasome inhibition has already been validated as a therapeutic strategy for the treatment of cancer, particularly multiple myeloma. Multiple myeloma cells possess both forms of the proteasome, although the ratio can vary somewhat. Multiple myeloma is a hematologic disease characterized by an excessive number of abnormal plasma cells in the bone marrow.
  • lymphomas generally appear to express immunoproteasome. Cancer cells originating from lymphoid cells express 30% or more immunoproteasome.
  • the invention relates to a method for the treatment of cancer, comprising administering a compound as described herein.
  • the cancer is a heme- related disorder.
  • the cancer is selected from a solid tumor, head and neck squamous cell carcinoma, cervical carcinoma and small cell lung carcinoma.
  • the invention relates to a method of treating cancer comprising administering a compound as disclosed herein.
  • Compounds prepared as described herein can be administered in various forms, depending on the disorder to be treated and the age, condition, and body weight of the patient, as is well known in the art. For example, where the compounds are to be administered.
  • they may be formulated as tablets, capsules, granules, powders, or syrups; or for parenteral administration, they may be formulated as injections (intravenous, intramuscular, or subcutaneous), drop infusion preparations, or suppositories.
  • parenteral administration they may be formulated as injections (intravenous, intramuscular, or subcutaneous), drop infusion preparations, or suppositories.
  • injections intravenous, intramuscular, or subcutaneous
  • drop infusion preparations or suppositories.
  • suppositories for application by the ophthalmic mucous membrane route, they may be formulated as eye drops or eye ointments.
  • formulations can be prepared by conventional means, and if desired, ATTY REF: 26500-0023WO1 the active ingredient may be mixed with any conventional additive or excipient, such as a binder, a disintegrating agent, a lubricant, a corrigent, a solubilizing agent, a suspension aid, an emulsifying agent, a coating agent, a cyclodextrin, and/or a buffer.
  • a daily dosage of from 0.01 to 2000 mg of the compound is
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect.
  • the precise time of administration and/or amount of the composition that will yield the most effective results in terms of efficacy of treatment in a given patient will depend upon the activity, pharmacokinetics, and bioavailability of a particular compound, physiological condition of the patient (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication), route of administration, etc.
  • physiological condition of the patient including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication
  • route of administration etc.
  • the above guidelines can be used as the basis for fine-tuning the treatment, e.g., determining the optimum time and/or amount of administration, which will require no more than routine experimentation consisting of monitoring the subject and adjusting the dosage and/or timing.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those ligands, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • phrases "pharmaceutically acceptable carrier” as used herein means a
  • composition such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material.
  • a liquid or solid filler such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose, and sucrose; (2) starches, such as corn ATTY REF: 26500-0023WO1 starch, potato starch, and substituted or unsubstituted ⁇ -cyclodextrin; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol, and polyethylene glycol; (12) esters, such as ethyl o
  • pharmaceutically acceptable salt refers to the relatively non-toxic, inorganic and organic acid addition salts of the inhibitor(s). These salts can be prepared in situ during the final isolation and purification of the inhibitor(s), or by separately reacting a purified inhibitor(s) in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate, mesylate, glucoheptonate, lactobionate, laurylsulphonate salts, and amino acid salts, and the like.
  • sulfate bisulfate
  • phosphate nitrate
  • acetate valerate
  • oleate palmitate
  • stearate laurate
  • benzoate lactate
  • phosphate tosylate
  • citrate maleate
  • fumarate succinate
  • tartrate naphthylate
  • mesylate glucoheptonate
  • lactobionate lactobionate
  • laurylsulphonate salts
  • the inhibitors useful in the methods of the present invention may contain one or more acidic functional groups and, thus, are capable of forming
  • “pharmaceutically acceptable salts” in these instances refers to the relatively non-toxic inorganic and organic base addition salts of an inhibitor(s). These salts can likewise be prepared in situ during the final isolation and purification of the inhibitor(s), or by separately reacting the purified inhibitor(s) in its free acid form with a suitable base, such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation, with ATTY REF: 26500-0023WO1 ammonia, or with a pharmaceutically acceptable organic primary, secondary, or tertiary amine.
  • a suitable base such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation
  • ATTY REF: 26500-0023WO1 ammonia or with a pharmaceutically acceptable organic primary, secondary, or tertiary amine.
  • Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts, and the like.
  • Organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, and the like (see, for example, Berge et al., supra).
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring, and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • antioxidants examples include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite, and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite, and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT
  • Formulations suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert matrix, such as gelatin and glycerin, or sucrose and acacia) and/or as mouthwashes, and the like, each containing a predetermined amount of an inhibitor(s) as an active ingredient.
  • a composition may also be administered as a bolus, electuary, or paste.
  • solid dosage forms for oral administration capsules, tablets, pills, dragees, powders, granules, and the like
  • the active ingredient is mixed with one or more
  • pharmaceutically acceptable carriers such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, cyclodextrins, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example,
  • compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols, and the like.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered inhibitor(s) moistened with an inert liquid diluent.
  • Tablets, and other solid dosage forms may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes, and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
  • compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • opacifying agents include polymeric substances and waxes.
  • the active ingredient can also be in micro- encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents, and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols, and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents, and e
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming, and preservative agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming, and preservative agents.
  • Suspensions in addition to the active inhibitor(s) may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • Formulations for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more inhibitor(s) with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
  • suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent.
  • Formulations which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams, or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration of an inhibitor(s) include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches, and inhalants.
  • the active component may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
  • the ointments, pastes, creams, and gels may contain, in addition to inhibitor(s), excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc, and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc, and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to an inhibitor(s), excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates, and polyamide powder, or mixtures of these substances.
  • Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • the inhibitor(s) can be alternatively administered by aerosol. This is accomplished by preparing an aqueous aerosol, liposomal preparation, or solid particles containing the composition.
  • a nonaqueous (e.g., fluorocarbon propellant) suspension could be used.
  • Sonic nebulizers are preferred because they minimize exposing the agent to shear, which can result in degradation of the compound.
  • an aqueous aerosol is made by formulating an aqueous solution or suspension of the agent together with conventional pharmaceutically acceptable carriers and stabilizers.
  • the carriers and stabilizers vary with the requirements of the particular composition, but typically include nonionic surfactants (T weens, Pluronics, sorbitan esters, lecithin, Cremophors), pharmaceutically acceptable co-solvents such as polyethylene glycol, innocuous proteins like serum albumin, oleic acid, amino acids such as glycine, buffers, salts, sugars, or sugar alcohols.
  • Aerosols generally are prepared from isotonic solutions.
  • Transdermal patches have the added advantage of providing controlled delivery of an inhibitor(s) to the body.
  • dosage forms can be made by dissolving or dispersing the agent in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the inhibitor(s) across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the inhibitor(s) in a polymer matrix or gel.
  • compositions of this invention suitable for parenteral administration comprise one or more inhibitors(s) in combination with one or more pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just ATTY REF: 26500-0023WO1 prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents, and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include tonicity-adjusting agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
  • adjuvants such as preservatives, wetting agents, emulsifying agents, and dispersing agents.
  • Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include tonicity-adjusting agents, such as sugars
  • delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
  • Injectable depot forms are made by forming microencapsule matrices of inhibitor(s) in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.
  • agents may be given orally, parenterally, topically, or rectally. They are, of course, given by forms suitable for each administration route. For example, they ATTY REF: 26500-0023WO1 are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, infusion; topically by lotion or ointment; and rectally by suppositories. Oral administration is preferred.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection, and infusion.
  • systemic administration means the administration of a ligand, drug, or other material other than directly into the central nervous system, such that it enters the patient's system and thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
  • inhibitors(s) may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracisternally, and topically, as by powders, ointments or drops, including buccally and sublingually.
  • the inhibitor(s), which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • compositions of this invention may be provided in an aqueous solution containing about 0.1-10% w/v of a compound disclosed herein, among other substances, for parenteral administration. Typical dose ranges are from about 0.01 to about 50 mg/kg of body weight per day, given in 1-4 divided doses. Each divided dose may contain the same or different compounds of the invention.
  • the dosage will be an effective amount depending on several factors including the overall health of a patient, and the formulation and route of administration of the selected compound(s).
  • Another aspect of the invention provides a conjoint therapy wherein one or more other therapeutic agents are administered with the proteasome inhibitor.
  • Such conjoint treatment may be achieved by way of the simultaneous, sequential, or separate dosing of the individual components of the treatment.
  • a compound of the invention is conjointly administered with one or more other proteasome inhibitor(s).
  • a compound of the invention is conjointly administered with a chemotherapeutic.
  • chemotherapeutics may include, natural products such as vinca alkaloids (e.g., vinblastine, vincristine, and vinorelbine); paclitaxel; epidipodophyllotoxins (e.g., etoposide, teniposide); antibiotics (e.g., dactinomycin (actinomycin D) daunorubicin, doxorubicin and idarubicin); anthracyclines; mitoxantrone; bleomycins; plicamycin
  • vinca alkaloids e.g., vinblastine, vincristine, and vinorelbine
  • paclitaxel e.g., etoposide, teniposide
  • antibiotics e.g., dactinomycin (actinomycin D) daunorubicin, doxorubicin and idarubic
  • antiproliferative/antimitotic alkylating agents such as nitrogen mustards (mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil), ethylenimines and methylmelamines (hexamethylmelamine and thiotepa); alkyl sulfonates (busulfan); nitrosoureas (carmustine (BCNU) and analogs, streptozocin); trazenes (e.g.,dacarbazine (DTIC)); antiproliferative/antimitotic antimetabolites such as folic acid analogs (methotrexate), pyrimidine analogs (fluorouracil, floxuridine, and cytara), antiproliferative/antimitotic alkylating agents such as nitrogen mustards (mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil), ethylenimines and methylmelamine
  • mitotane e.g., mitotane;aminoglutethimide; hormones (e.g., estrogen); and hormone agonists such as ATTY REF: 26500-0023WO1 leutinizing hormone releasing hormone (LHRH) agonists (e.g., goserelin, leuprolide and triptorelin).
  • hormones e.g., estrogen
  • hormone agonists such as ATTY REF: 26500-0023WO1 leutinizing hormone releasing hormone (LHRH) agonists (e.g., goserelin, leuprolide and triptorelin).
  • LHRH leutinizing hormone releasing hormone
  • the other therapeutic agent is an HDAC inhibitor (e.g., Trichostatin A, depsipeptide, apicidin, A-161906, scriptaid, PXD-101, CHAP, butyric acid, depudecin, oxamflatin, phenylbutyrate, valproic acid, SAHA (Vorinostat), MS275 (N-(2- Aminophenyl)-4-[N-(pyridine-3-ylmethoxy-carbonyl)aminomethyl]benzamide),
  • HDAC inhibitor e.g., Trichostatin A, depsipeptide, apicidin, A-161906, scriptaid, PXD-101, CHAP, butyric acid, depudecin, oxamflatin, phenylbutyrate, valproic acid, SAHA (Vorinostat), MS275 (N-(2- Aminophenyl)-4-[N-(pyridine-3-ylmethoxy-carbonyl)aminomethyl
  • the other agent is SAHA (suberoylanilide hydroxamic acid).
  • the other therapeutic agent is a protein kinase inhibitor (e.g., sorafenib, imatinib, dasatinib, sunitinib, pazopanib, and nilotinib).
  • a protein kinase inhibitor e.g., sorafenib, imatinib, dasatinib, sunitinib, pazopanib, and nilotinib.
  • the protein kinase inhibitor is sorafenib.
  • the other chemotherapeutic agent is selected from mechlorethamine, camptothecin, ifosfamide, tamoxifen, raloxifene, gemcitabine, navelbine, or any analog or derivative variant of the foregoing.
  • a compound of the invention is conjointly administered with a steroid.
  • Suitable steroids may include, but are not limited to, 21-acetoxypregnenolone, alclometasone, algestone, amcinonide, beclomethasone, betamethasone, budesonide, chloroprednisone, clobetasol, clocortolone, cloprednol, corticosterone, cortisone, cortivazol, deflazacort, desonide, desoximetasone, dexamethasone, diflorasone, diflucortolone, difuprednate, enoxolone, fluazacort, flucloronide, flumethasone, flunisolide, fluocinolone acetonide, fluocinonide, fluocortin butyl, fluocortolone, fluorometholone, fluperolone acetate, fluprednidene acetate, fluprednisolone, flurandrenolide, flu
  • a compound of the invention is conjointly administered with an immunotherapeutic agent.
  • Suitable immunotherapeutic agents may include, but are not ATTY REF: 26500-0023WO1 limited to, cyclosporine, thalidomide (or thalidomide analogs such as lenalidomide), or monoclonal antibodies.
  • the monoclonal antibodies can be either naked or conjugated such as rituximab, tositumomab, alemtuzumab, epratuzumab, ibritumomab tiuxetan, gemtuzumab ozogamicin, bevacizumab, cetuximab, erlotinib or trastuzumab.
  • Compound B was synthesized according to the procedure in U.S. Patent Application Serial No. 11/820,490.
  • Example 1 Assay to Determine Inhibitory Preference
  • a biochemical assay that can be utilized to determine whether or not a molecule preferentially inhibits the CT-L activity of the constitutive (B5) or immunoproteasome (L7) relies on first determining the EC50 for each subunit. This can be done using enzyme kinetic assays such as those disclosed in U.S. Application Serial No. 09/569748, Example 2 and Stein et al, Biochem. (1996), 35, 3899-3908 using isolated 20S proteasome preps with greater than 90% constitutive proteasome subunits or immunoproteasome subunits to determine. The inhibitory preference of the molecule is then based on the EC50 ratio of the chymotryptic-like activity of the constitutive proteasome (to that of the immunoproteasome (20S ratio).
  • a ratio of less than one denotes the molecule inhibits the CT-L activity of the constitutive proteasome better than that of the immunoproteasome.
  • Ratios greater than one signifies the molecule inhibits chymotrypsin-like activity of the immunoproteasome better ATTY REF: 26500-0023WO1 than that of the constitutive proteasome. The greater the ratio number, the more specific the compound is for the immunoproteasome inhibitor.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Epidemiology (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Hospice & Palliative Care (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Psychiatry (AREA)
  • Child & Adolescent Psychology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Peptides Or Proteins (AREA)
PCT/US2011/026629 2010-03-01 2011-03-01 Compounds for immunoproteasome inhibition WO2011109355A1 (en)

Priority Applications (15)

Application Number Priority Date Filing Date Title
CA2791651A CA2791651C (en) 2010-03-01 2011-03-01 Compounds for immunoproteasome inhibition
MX2012010017A MX2012010017A (es) 2010-03-01 2011-03-01 Compuestos de para la inhibicion de inmunoproteasomas.
EA201290844A EA201290844A1 (ru) 2010-03-01 2011-03-01 Соединения для ингибирования иммунопротеасом
AU2011223795A AU2011223795B2 (en) 2010-03-01 2011-03-01 Compounds for immunoproteasome inhibition
BR112012022060A BR112012022060A2 (pt) 2010-03-01 2011-03-01 composto para a inibição de imunoproteassoma
CN201180022010XA CN102892417A (zh) 2010-03-01 2011-03-01 用于免疫蛋白酶体抑制的化合物
SG2012064531A SG183843A1 (en) 2010-03-01 2011-03-01 Compounds for immunoproteasome inhibition
JP2012556160A JP6042724B2 (ja) 2010-03-01 2011-03-01 イムノプロテアソーム阻害のための化合物
KR1020127025901A KR20130075723A (ko) 2010-03-01 2011-03-01 면역프로테아좀 저해를 위한 화합물
MA35248A MA34133B1 (fr) 2010-03-01 2011-03-01 Composes pour inhibiteurs de l'immunoproteasome
US13/580,112 US9359398B2 (en) 2010-03-01 2011-03-01 Compounds for immunoproteasome inhibition
EP11715320.5A EP2542238B1 (en) 2010-03-01 2011-03-01 Compounds for immunoproteasome inhibition
ZA2012/06479A ZA201206479B (en) 2010-03-01 2012-08-28 Compounds for immunoproteasome inhibition
CU2012000125A CU20120125A7 (es) 2010-03-01 2012-08-30 Compuestos para la inhibición de inmunoproteasomas
ECSP12012140 ECSP12012140A (es) 2010-03-01 2012-09-04 Compuestos de para la inhibición de inmunoproteasomas

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US30936610P 2010-03-01 2010-03-01
US61/309,366 2010-03-01

Publications (1)

Publication Number Publication Date
WO2011109355A1 true WO2011109355A1 (en) 2011-09-09

Family

ID=43983939

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/026629 WO2011109355A1 (en) 2010-03-01 2011-03-01 Compounds for immunoproteasome inhibition

Country Status (20)

Country Link
US (1) US9359398B2 (US07094801-20060822-C00173.png)
EP (1) EP2542238B1 (US07094801-20060822-C00173.png)
JP (1) JP6042724B2 (US07094801-20060822-C00173.png)
KR (1) KR20130075723A (US07094801-20060822-C00173.png)
CN (1) CN102892417A (US07094801-20060822-C00173.png)
AU (1) AU2011223795B2 (US07094801-20060822-C00173.png)
BR (1) BR112012022060A2 (US07094801-20060822-C00173.png)
CA (1) CA2791651C (US07094801-20060822-C00173.png)
CL (1) CL2012002404A1 (US07094801-20060822-C00173.png)
CO (1) CO6612265A2 (US07094801-20060822-C00173.png)
CR (1) CR20120451A (US07094801-20060822-C00173.png)
CU (1) CU20120125A7 (US07094801-20060822-C00173.png)
DO (1) DOP2012000238A (US07094801-20060822-C00173.png)
EA (1) EA201290844A1 (US07094801-20060822-C00173.png)
EC (1) ECSP12012140A (US07094801-20060822-C00173.png)
MA (1) MA34133B1 (US07094801-20060822-C00173.png)
MX (1) MX2012010017A (US07094801-20060822-C00173.png)
SG (1) SG183843A1 (US07094801-20060822-C00173.png)
WO (1) WO2011109355A1 (US07094801-20060822-C00173.png)
ZA (1) ZA201206479B (US07094801-20060822-C00173.png)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014139008A1 (en) * 2013-03-15 2014-09-18 Adaerata, Limited Partnership Small molecule modulators of pcsk9 and methods of use thereof
US8853147B2 (en) 2009-11-13 2014-10-07 Onyx Therapeutics, Inc. Use of peptide epoxyketones for metastasis suppression
US8921324B2 (en) 2007-10-04 2014-12-30 Onyx Therapeutics, Inc. Crystalline peptide epoxy ketone protease inhibitors and the synthesis of amino acid keto-epoxides
US9051353B2 (en) 2009-03-20 2015-06-09 Onyx Therapeutics, Inc. Crystalline tripeptide epoxy ketone protease inhibitors
US9205124B2 (en) 2005-11-09 2015-12-08 Onyx Therapeutics, Inc. Compounds for enzyme inhibition
US9309283B2 (en) 2012-07-09 2016-04-12 Onyx Therapeutics, Inc. Prodrugs of peptide epoxy ketone protease inhibitors
US9359398B2 (en) 2010-03-01 2016-06-07 Onyx Therapeutics, Inc. Compounds for immunoproteasome inhibition
US9511109B2 (en) 2008-10-21 2016-12-06 Onyx Therapeutics, Inc. Combination therapy with peptide epoxyketones
US9657058B2 (en) 2006-06-19 2017-05-23 Onyx Therapeutics, Inc. Compounds for enzyme inhibition
RU2625801C2 (ru) * 2012-12-03 2017-07-19 Ф. Хоффманн-Ля Рош Аг Соединения замещенной триазолбороновой кислоты
AU2014240122B2 (en) * 2013-03-14 2019-02-14 Onyx Therapeutics, Inc. Dipeptide and tripeptide epoxy ketone protease inhibitors
WO2021001697A1 (en) * 2019-07-01 2021-01-07 Latvian Institute Of Organic Synthesis Novel boronic acid containing peptidomimetics as malarial serine protease inhibitors
US11267803B2 (en) 2016-06-21 2022-03-08 Orion Ophthalmology LLC Carbocyclic prolinamide derivatives
US11377439B2 (en) 2016-06-21 2022-07-05 Orion Ophthalmology LLC Heterocyclic prolinamide derivatives

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011136905A2 (en) 2010-04-07 2011-11-03 Onyx Therapeutics, Inc. Crystalline peptide epoxyketone immunoproteasome inhibitor
USRE49816E1 (en) 2014-01-10 2024-01-30 Cornell University Dipeptides as inhibitors of human immunoproteasomes
CN103804469B (zh) * 2014-02-17 2016-04-06 苏州明锐医药科技有限公司 卡非佐米中间体的制备方法
CN114796207A (zh) 2014-08-18 2022-07-29 康奈尔大学 作为人免疫蛋白酶体的抑制剂的二肽模拟物
US10253049B2 (en) * 2014-10-01 2019-04-09 Merck Patent Gmbh Boronic acid derivatives
CN106588965A (zh) * 2015-10-15 2017-04-26 北京大学 脲拟肽硼酸化合物及其药物组合物、制备方法和用途
EP3362754B1 (en) 2015-10-15 2021-12-22 Cornell University Proteasome inhibitors and uses thereof
WO2019075252A1 (en) 2017-10-11 2019-04-18 Cornell University PEPTIDOMIMETIC INHIBITORS OF PROTEASOME
US11225493B2 (en) * 2017-11-16 2022-01-18 Principia Biopharma Inc. Immunoproteasome inhibitors
EP3710063A1 (en) * 2017-11-16 2020-09-23 Amgen Inc. Stable compositions of pegylated carfilzomib compounds

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5340736A (en) 1991-05-13 1994-08-23 The President & Fellows Of Harvard College ATP-dependent protease and use of inhibitors for same in the treatment of cachexia and muscle wasting
WO1998010779A1 (en) 1996-09-13 1998-03-19 New York University Method for treating parasitic diseases with proteasome inhibitors
WO2006086600A1 (en) * 2005-02-11 2006-08-17 Cephalon, Inc. Proteasome inhibitors and methods of using the same
WO2006099261A2 (en) * 2005-03-11 2006-09-21 The University Of North Carolina At Chapel Hill Potent and specific immunoproteasome inhibitors
WO2010036357A1 (en) * 2008-09-29 2010-04-01 Millennium Pharmaceuticals, Inc. Derivatives of 1-amino-2-cyclobutylethylboronic acid
WO2010145376A1 (zh) * 2009-06-19 2010-12-23 北京大学 三肽硼酸(酯)类化合物、其制备方法和应用

Family Cites Families (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4733665C2 (en) 1985-11-07 2002-01-29 Expandable Grafts Partnership Expandable intraluminal graft and method and apparatus for implanting an expandable intraluminal graft
US5135919A (en) 1988-01-19 1992-08-04 Children's Medical Center Corporation Method and a pharmaceutical composition for the inhibition of angiogenesis
DE3827340A1 (de) * 1988-08-12 1990-02-15 Hoechst Ag Verwendung von (alpha)-aminoboronsaeure-derivaten zur prophylaxe und behandlung von viruserkrankungen
JPH0291023U (US07094801-20060822-C00173.png) 1989-01-05 1990-07-19
US5441944A (en) 1989-04-23 1995-08-15 The Trustees Of The University Of Pennsylvania Substituted cyclodextrin sulfates and their uses as growth modulating agents
US5071957A (en) 1989-08-04 1991-12-10 Bristol-Myers Company Antibiotic BU-4061T
US4990448A (en) 1989-08-04 1991-02-05 Bristol-Myers Company Bu-4061T
KR0166088B1 (ko) 1990-01-23 1999-01-15 . 수용해도가 증가된 시클로덱스트린 유도체 및 이의 용도
US5376645A (en) 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
CA2077665A1 (en) 1990-03-05 1991-09-06 Robert Siman Chymotrypsin-like proteases and their inhibitors
GB9300048D0 (en) 1993-01-04 1993-03-03 Wellcome Found Endothelin converting enzyme inhibitors
TW380137B (en) 1994-03-04 2000-01-21 Merck & Co Inc Process for making an epoxide
US5693617A (en) 1994-03-15 1997-12-02 Proscript, Inc. Inhibitors of the 26s proteolytic complex and the 20s proteasome contained therein
US6660268B1 (en) 1994-03-18 2003-12-09 The President And Fellows Of Harvard College Proteasome regulation of NF-KB activity
US6506876B1 (en) 1994-10-11 2003-01-14 G.D. Searle & Co. LTA4 hydrolase inhibitor pharmaceutical compositions and methods of use
US6083903A (en) * 1994-10-28 2000-07-04 Leukosite, Inc. Boronic ester and acid compounds, synthesis and uses
DE19505263A1 (de) 1995-02-16 1996-08-22 Consortium Elektrochem Ind Verfahren zur Reinigung von wasserlöslichen Cyclodextrinderivaten
US6335358B1 (en) 1995-04-12 2002-01-01 President And Fellows Of Harvard College Lactacystin analogs
US6150415A (en) 1996-08-13 2000-11-21 The Regents Of The University Of California Epoxide hydrolase complexes and methods therewith
IL129407A0 (en) 1996-10-18 2000-02-17 Vertex Pharma Inhibitors of serine proteases particularly hepatitis C virus NS3 protease pharmaceutical compositions containing the same and the use thereof
SI0932617T1 (en) 1996-10-18 2002-06-30 Vertex Pharmaceuticals Incorporated Inhibitors of serine proteases, particularly hepatitis c virus ns3 protease
US6046177A (en) 1997-05-05 2000-04-04 Cydex, Inc. Sulfoalkyl ether cyclodextrin based controlled release solid pharmaceutical formulations
US5874418A (en) 1997-05-05 1999-02-23 Cydex, Inc. Sulfoalkyl ether cyclodextrin based solid pharmaceutical formulations and their use
AU750207B2 (en) 1997-06-13 2002-07-11 Cydex Pharmaceuticals, Inc. Polar drug or prodrug compositions with extended shelf-life storage and a method of making thereof
US6133308A (en) 1997-08-15 2000-10-17 Millennium Pharmaceuticals, Inc. Synthesis of clasto-lactacystin beta-lactone and analogs thereof
US6075150A (en) 1998-01-26 2000-06-13 Cv Therapeutics, Inc. α-ketoamide inhibitors of 20S proteasome
US6099851A (en) 1998-06-02 2000-08-08 Weisman; Kenneth M. Therapeutic uses of leuprolide acetate
US6902721B1 (en) 1998-07-10 2005-06-07 Osteoscreen, Inc. Inhibitors of proteasomal activity for stimulating bone growth
US6838436B1 (en) 1998-07-10 2005-01-04 Osteoscreen Inc. Inhibitors of proteasomal activity for stimulating bone growth
US6462019B1 (en) 1998-07-10 2002-10-08 Osteoscreen, Inc. Inhibitors of proteasomal activity and production for stimulating bone growth
US6204257B1 (en) 1998-08-07 2001-03-20 Universtiy Of Kansas Water soluble prodrugs of hindered alcohols
CA2347275C (en) 1998-10-20 2010-03-09 Millennium Pharmaceuticals, Inc. Method for monitoring proteasome inhibitor drug action
US6492333B1 (en) 1999-04-09 2002-12-10 Osteoscreen, Inc. Treatment of myeloma bone disease with proteasomal and NF-κB activity inhibitors
US6831099B1 (en) 1999-05-12 2004-12-14 Yale University Enzyme inhibition
CA2385958A1 (en) 1999-10-20 2001-04-26 Osteoscreen, Inc. Inhibitors of proteasomal activity for stimulating bone and hair growth
PT1326632E (pt) 2000-10-12 2007-01-31 Viromics Gmbh Tratamento para o tratamento de infecções por vírus da hepatite
US6713446B2 (en) 2001-01-25 2004-03-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Formulation of boronic acid compounds
EP1392355B1 (en) 2001-05-21 2007-02-14 Alcon, Inc. Use of proteasome inhibitors to treat dry eye disorders
JP4416501B2 (ja) * 2001-05-30 2010-02-17 ノバルティス アクチエンゲゼルシャフト 2−{[n−(2−アミノ−3−(ヘテロアリールまたはアリール)プロピオニル)−アミノアシル]−アミノ}−アルキルボロン酸誘導体
US7524883B2 (en) 2002-01-08 2009-04-28 Eisai R&D Management Co., Ltd. Eponemycin and epoxomicin analogs and uses thereof
US20040116329A1 (en) 2002-01-29 2004-06-17 Epstein Stephen E. Inhibition of proteasomes to prevent restenosis
WO2003086283A2 (en) 2002-04-09 2003-10-23 Greenville Hospital System Metastasis modulating activity of highly sulfated oligosaccharides
US7968569B2 (en) 2002-05-17 2011-06-28 Celgene Corporation Methods for treatment of multiple myeloma using 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione
AU2003249682A1 (en) 2002-06-03 2003-12-19 Als Therapy Development Foundation Treatment of neurodegenerative diseases using proteasome modulators
US20030224469A1 (en) 2002-06-03 2003-12-04 Buchholz Tonia J. Methods and kits for assays utilizing fluorescence polarization
AU2003256847A1 (en) 2002-07-26 2004-02-16 Advanced Research And Technology Institute At Indiana University Method of treating cancer
US7189740B2 (en) 2002-10-15 2007-03-13 Celgene Corporation Methods of using 3-(4-amino-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione for the treatment and management of myelodysplastic syndromes
TW200418791A (en) 2003-01-23 2004-10-01 Bristol Myers Squibb Co Pharmaceutical compositions for inhibiting proteasome
EP2316431B1 (en) 2003-04-08 2015-09-30 Novartis AG Solid oral composition comprising a S1P receptor agonist and a sugar alcohol
ATE506456T1 (de) 2003-06-10 2011-05-15 David Gladstone Inst Verfahren zur behandlung von lentivirusinfektionen
US7012063B2 (en) 2003-06-13 2006-03-14 Children's Medical Center Corporation Reducing axon degeneration with proteasome inhibitors
BRPI0418386A (pt) 2003-12-31 2007-05-22 Cydex Inc formulação inalante contendo éter sulfoalquìlico gama-ciclodextrina e corticosteróide
GB0400804D0 (en) 2004-01-14 2004-02-18 Innoscience Technology Bv Pharmaceutical compositions
US7232818B2 (en) 2004-04-15 2007-06-19 Proteolix, Inc. Compounds for enzyme inhibition
US8198270B2 (en) 2004-04-15 2012-06-12 Onyx Therapeutics, Inc. Compounds for proteasome enzyme inhibition
PT1745064E (pt) 2004-04-15 2011-03-23 Proteolix Inc Compostos para a inibição de uma enzima de proteassoma
JP5108509B2 (ja) 2004-05-10 2012-12-26 プロテオリックス, インコーポレイテッド 酵素阻害のための化合物
US20050256324A1 (en) 2004-05-10 2005-11-17 Proteolix, Inc. Synthesis of amino acid keto-epoxides
EP1805208A2 (en) 2004-10-20 2007-07-11 Proteolix, Inc. Labeled compounds for proteasome inhibition
HUE027850T2 (en) 2004-12-07 2016-11-28 Onyx Therapeutics Inc Preparation for proteasome inhibition
WO2006113470A2 (en) 2005-04-15 2006-10-26 Geron Corporation Cancer treatment by combined inhibition of proteasome and telomerase activities
GT200600350A (es) 2005-08-09 2007-03-28 Formulaciones líquidas
DK2623113T3 (en) 2005-11-09 2017-07-17 Onyx Therapeutics Inc Enzyme inhibition compound
AR057227A1 (es) 2005-12-09 2007-11-21 Centocor Inc Metodo para usar antagonistas de il6 con inhibidores del proteasoma
US20070207950A1 (en) 2005-12-21 2007-09-06 Duke University Methods and compositions for regulating HDAC6 activity
WO2007122686A1 (ja) 2006-04-14 2007-11-01 Eisai R & D Management Co., Ltd. ベンズイミダゾール化合物
DE102006026464A1 (de) 2006-06-01 2007-12-06 Virologik Gmbh Innovationszentrum Medizintechnik Und Pharma Pharmazeutische Zusammensetzung zur Behandlung von Virusinfektionen und / oder Tumorerkrankungen durch Inhibition der Proteinfaltung und des Proteinabbaus
US7691852B2 (en) 2006-06-19 2010-04-06 Onyx Therapeutics, Inc. Compounds for enzyme inhibition
WO2008033807A2 (en) 2006-09-13 2008-03-20 The Arizona Board Of Regents On Behalf Of The University Of Arizona Synergistic combinations of antineoplastic thiol-binding mitochondrial oxidants and antineoplastic proteasome inhibitors for the treatment of cancer
WO2008091620A2 (en) 2007-01-23 2008-07-31 Gloucester Pharmaceuticals, Inc. Combination therapy comprising romidepsin and i.a. bortezomib
WO2008140782A2 (en) 2007-05-10 2008-11-20 Proteolix, Inc. Compounds for enzyme inhibition
SI2178888T1 (sl) 2007-08-06 2012-11-30 Millennium Pharm Inc Proteasomni inhibitorji
US7442830B1 (en) 2007-08-06 2008-10-28 Millenium Pharmaceuticals, Inc. Proteasome inhibitors
DK2207791T4 (da) 2007-10-04 2019-10-07 Onyx Therapeutics Inc Krystallinske peptidepoxyketon-proteasehæmmere og syntese af aminosyreketoepoxider
WO2009051581A1 (en) 2007-10-16 2009-04-23 Millennium Pharmaceuticals, Inc. Proteasome inhibitors
US7838673B2 (en) 2007-10-16 2010-11-23 Millennium Pharmaceuticals, Inc. Proteasome inhibitors
WO2009067453A1 (en) 2007-11-19 2009-05-28 Syndax Pharmaceuticals, Inc. Combinations of hdac inhibitors and proteasome inhibitors
SG194349A1 (en) 2008-06-17 2013-11-29 Millennium Pharm Inc Boronate ester compounds and pharmaceutical compositions thereof
ES2617560T3 (es) 2008-10-21 2017-06-19 Onyx Therapeutics, Inc. Combinación del inhibidor del proteasoma de epoxicetona peptídica carfilzomib con melfalán para su uso en el tratamiento del mieloma múltiple
TWI504598B (zh) 2009-03-20 2015-10-21 Onyx Therapeutics Inc 結晶性三肽環氧酮蛋白酶抑制劑
EP2498793B1 (en) 2009-11-13 2019-07-10 Onyx Therapeutics, Inc. Oprozomib for use in metastasis suppression
MA34133B1 (fr) 2010-03-01 2013-04-03 Onyx Therapeutics Inc Composes pour inhibiteurs de l'immunoproteasome
NZ602622A (en) 2010-03-31 2015-01-30 Millennium Pharm Inc Derivatives of 1-amino-2-cyclopropylethylboronic acid
WO2011136905A2 (en) 2010-04-07 2011-11-03 Onyx Therapeutics, Inc. Crystalline peptide epoxyketone immunoproteasome inhibitor
FR2962092B1 (fr) * 2010-06-30 2013-06-28 Valeo Systemes Dessuyage Adaptateur d'essuie-glace a contacts electriques

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5340736A (en) 1991-05-13 1994-08-23 The President & Fellows Of Harvard College ATP-dependent protease and use of inhibitors for same in the treatment of cachexia and muscle wasting
WO1998010779A1 (en) 1996-09-13 1998-03-19 New York University Method for treating parasitic diseases with proteasome inhibitors
WO2006086600A1 (en) * 2005-02-11 2006-08-17 Cephalon, Inc. Proteasome inhibitors and methods of using the same
WO2006099261A2 (en) * 2005-03-11 2006-09-21 The University Of North Carolina At Chapel Hill Potent and specific immunoproteasome inhibitors
WO2010036357A1 (en) * 2008-09-29 2010-04-01 Millennium Pharmaceuticals, Inc. Derivatives of 1-amino-2-cyclobutylethylboronic acid
WO2010145376A1 (zh) * 2009-06-19 2010-12-23 北京大学 三肽硼酸(酯)类化合物、其制备方法和应用

Non-Patent Citations (32)

* Cited by examiner, † Cited by third party
Title
ACHARYYA ET AL., JCI, vol. 114, 2004, pages 370 - 378
BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
CASCIO ET AL., EMBO J, vol. 20, 2001, pages 2357 - 2366
CIECHANOVER, A., CELL, vol. 79, 1994, pages 13 - 21
DIAZ-HERNANDEZ ET AL., J NEUROSCI, vol. 23, 2003, pages 11653 - 1161
EGERER ET AL., ARTHRITIS RHEUM, vol. 54, 2006, pages 1501 - 8
FOX, WHITESELL: "Organic Chemistry", 1994, JONES AND BARTLETT PUBLISHERS, pages: 177 - 178
GAN ET AL., J. BIOL. CHEM., vol. 279, 2004, pages 5565 - 5572
GARRETT, 1. R. ET AL., J. CLIN. INVEST., vol. 111, 2003, pages 1771 - 1782
GONZALES ET AL., ARCH. MED. RES., vol. 28, 1997, pages 139 - 140
HRUBY, BOTEJU: "Molecular Biology and Biotechnology: A Comprehensive Desk Reference", 1995, VCH PUBLISHERS, article "Peptides and Mimics, Design of Conformationally Constrained", pages: 658 - 664
J. MED. CHEM., vol. 51, 2008, pages 1068
KHAN ET AL., J IMMUNOL, vol. 167, 2001, pages 6859 - 6868
KUMATORI ET AL., PROC. NATL. ACAD. SCI. USA, vol. 87, 1990, pages 7071 - 7075
LECKER ET AL., FASEB J, vol. 18, 2004, pages 39 - 51
MACARY ET AL., PNAS, vol. 98, 2001, pages 3982 - 3987
MENG ET AL., PNAS, vol. 96, 1999, pages 10403 - 10408
MISHTO ET AL., NEUROBIOL AGING, vol. 27, 2006, pages 54 - 66
PALOMBELLA ET AL., CELL, vol. 78, 1994, pages 773 - 785
PAUGAM ET AL., TRENDS PARASITOL., vol. 19, no. 2, 2003, pages 55 - 59
PIVAZYAN, A.D.; MATTESON, D.S.; FRABRY-ASZTALOS, L.; SINGH, R.P.; LIN, P-F.; BLAIR, W.; GUO, K.; ROBINSON, B.; PRUSOFF, W.H.: "Inhibition of HIC-1 Protease by a Boron-Modified Polypeptide", BIOCHEMICAL PHARMACOLOGY, vol. 60, 2000, pages 927 - 936, XP002638798 *
QURESHI, N. ET AL., J. IMMUN., vol. 171, 2003, pages 1515 - 1525
SIMSEK ET AL., J VIROL, vol. 79, 2005, pages 12914 - 12920
STEIN ET AL., BIOCHEM., vol. 35, 1996, pages 3899 - 3908
SZALAY ET AL., AM J PATHOL, vol. 168, 2006, pages 1542 - 52
TAWA ET AL., JCI, vol. 100, 1997, pages 197 - 203
TRAENCKNER ET AL., EMBO J., vol. 13, 1994, pages 5433 - 5441
WATANABE T ET AL: "Synthesis of boronic acid derivatives of tyropeptin: Proteasome inhibitors", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, PERGAMON, ELSEVIER SCIENCE, GB, vol. 19, no. 8, 15 April 2009 (2009-04-15), pages 2343 - 2345, XP026079469, ISSN: 0960-894X, [retrieved on 20090304], DOI: DOI:10.1016/J.BMCL.2009.02.117 *
YU, LAI, J VIROL, vol. 79, 2005, pages 644 - 648
ZHU Y ET AL: "Design, synthesis and biological evaluation of tripeptide boronic acid proteasome inhibitors", BIOORGANIC & MEDICINAL CHEMISTRY, PERGAMON, GB, vol. 17, no. 19, 1 October 2009 (2009-10-01), pages 6851 - 6861, XP026601532, ISSN: 0968-0896, [retrieved on 20090820], DOI: DOI:10.1016/J.BMC.2009.08.023 *
ZHU Y Q ET AL: "3D-QSAR studies of boron-containing dipeptides as proteasome inhibitors with CoMFA and CoMSIA methods", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, EDITIONS SCIENTIFIQUE ELSEVIER, PARIS, FR, vol. 44, no. 4, 1 April 2009 (2009-04-01), pages 1486 - 1499, XP026003082, ISSN: 0223-5234, [retrieved on 20080724], DOI: DOI:10.1016/J.EJMECH.2008.07.019 *
ZHU, Y.; ZHAO, X.; ZHU, G.; LI, Y.; MA, Y.; YUAN, Y.; YANG, J.; HU, Y.; AI, L.; GAO, Q.: "Design, Synthesis, Biological Evaluation, and Structure - Activity Relationship (SAR) Discussion of Dipeptidyl Boronate Proteasome Inhibitors, Part I: Comprehensive Undertanding of the SAR of a-Amino Acid Boronates", JOURNAL OF MEDICINAL CHEMISTRY, vol. 52, 2009, pages 4192 - 4199, XP002638799 *

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9205124B2 (en) 2005-11-09 2015-12-08 Onyx Therapeutics, Inc. Compounds for enzyme inhibition
US9205125B2 (en) 2005-11-09 2015-12-08 Onyx Therapeutics, Inc. Compounds for enzyme inhibition
US9205126B2 (en) 2005-11-09 2015-12-08 Onyx Therapeutics, Inc. Compounds for enzyme inhibition
US10150794B2 (en) 2005-11-09 2018-12-11 Onyx Therapeutics, Inc. Compounds for enzyme inhibition
US9657058B2 (en) 2006-06-19 2017-05-23 Onyx Therapeutics, Inc. Compounds for enzyme inhibition
US8921324B2 (en) 2007-10-04 2014-12-30 Onyx Therapeutics, Inc. Crystalline peptide epoxy ketone protease inhibitors and the synthesis of amino acid keto-epoxides
US8921583B2 (en) 2007-10-04 2014-12-30 Onyx Therapeutics, Inc. Crystalline peptide epoxy ketone protease inhibitors and the synthesis of amino acid keto-epoxides
US9511109B2 (en) 2008-10-21 2016-12-06 Onyx Therapeutics, Inc. Combination therapy with peptide epoxyketones
USRE47954E1 (en) 2008-10-21 2020-04-21 Onyx Therapeutics, Inc. Combination therapy with peptide epoxyketones
US10596222B2 (en) 2008-10-21 2020-03-24 Onyx Therapeutics, Inc. Combination therapy with peptide epoxyketones
US9403868B2 (en) 2009-03-20 2016-08-02 Onyx Therapeutics, Inc. Crystalline tripeptide epoxy ketone protease inhibitors
US9051353B2 (en) 2009-03-20 2015-06-09 Onyx Therapeutics, Inc. Crystalline tripeptide epoxy ketone protease inhibitors
US8853147B2 (en) 2009-11-13 2014-10-07 Onyx Therapeutics, Inc. Use of peptide epoxyketones for metastasis suppression
US9359398B2 (en) 2010-03-01 2016-06-07 Onyx Therapeutics, Inc. Compounds for immunoproteasome inhibition
US10682419B2 (en) 2012-07-09 2020-06-16 Onyx Therapeutics, Inc. Prodrugs of peptide epoxy ketone protease inhibitors
US9315542B2 (en) 2012-07-09 2016-04-19 Onyx Therapeutics, Inc. Prodrugs of peptide epoxy ketone protease inhibitors
US9309283B2 (en) 2012-07-09 2016-04-12 Onyx Therapeutics, Inc. Prodrugs of peptide epoxy ketone protease inhibitors
US9878047B2 (en) 2012-07-09 2018-01-30 Onyx Therapeutics, Inc. Prodrugs of peptide epoxy ketone protease inhibitors
RU2625801C2 (ru) * 2012-12-03 2017-07-19 Ф. Хоффманн-Ля Рош Аг Соединения замещенной триазолбороновой кислоты
AU2014240122B2 (en) * 2013-03-14 2019-02-14 Onyx Therapeutics, Inc. Dipeptide and tripeptide epoxy ketone protease inhibitors
AU2019203282B2 (en) * 2013-03-14 2021-02-18 Onyx Therapeutics, Inc. Dipeptide and tripeptide epoxy ketone protease inhibitors
US20160031935A1 (en) * 2013-03-15 2016-02-04 Adaerata, Limited Partnership Small molecule modulators of pcsk9 and methods of use thereof
WO2014139008A1 (en) * 2013-03-15 2014-09-18 Adaerata, Limited Partnership Small molecule modulators of pcsk9 and methods of use thereof
US11267803B2 (en) 2016-06-21 2022-03-08 Orion Ophthalmology LLC Carbocyclic prolinamide derivatives
US11377439B2 (en) 2016-06-21 2022-07-05 Orion Ophthalmology LLC Heterocyclic prolinamide derivatives
US11866422B2 (en) 2016-06-21 2024-01-09 Orion Ophthalmology LLC Carbocyclic prolinamide derivatives
WO2021001697A1 (en) * 2019-07-01 2021-01-07 Latvian Institute Of Organic Synthesis Novel boronic acid containing peptidomimetics as malarial serine protease inhibitors

Also Published As

Publication number Publication date
SG183843A1 (en) 2012-10-30
MX2012010017A (es) 2012-10-01
CN102892417A (zh) 2013-01-23
BR112012022060A2 (pt) 2018-05-08
ECSP12012140A (es) 2012-10-30
AU2011223795A1 (en) 2012-09-20
EP2542238A1 (en) 2013-01-09
JP2013521295A (ja) 2013-06-10
CA2791651C (en) 2019-08-20
AU2011223795B2 (en) 2015-11-05
CR20120451A (es) 2012-10-16
CO6612265A2 (es) 2013-02-01
MA34133B1 (fr) 2013-04-03
EP2542238B1 (en) 2015-08-12
EA201290844A1 (ru) 2013-03-29
US20130072422A1 (en) 2013-03-21
KR20130075723A (ko) 2013-07-05
US9359398B2 (en) 2016-06-07
CU20120125A7 (es) 2012-11-15
CA2791651A1 (en) 2011-09-09
DOP2012000238A (es) 2013-01-15
CL2012002404A1 (es) 2012-12-21
ZA201206479B (en) 2015-08-26
JP6042724B2 (ja) 2016-12-14

Similar Documents

Publication Publication Date Title
EP2542238B1 (en) Compounds for immunoproteasome inhibition
US9657058B2 (en) Compounds for enzyme inhibition
AU2005271232B2 (en) Compounds for proteasome enzyme inhibition
US8697646B2 (en) Crystalline peptide epoxyketone immunoproteasome inhibitor
IL179020A (en) Inhibitors of enzymes
AU2013203467A1 (en) Compounds for immunoproteasome inhibition
AU2013203525A1 (en) Crystalline peptide epoxyketone immunoproteasome inhibitor

Legal Events

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

Ref document number: 201180022010.X

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11715320

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 7290/DELNP/2012

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 221574

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2011223795

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 12012501719

Country of ref document: PH

ENP Entry into the national phase

Ref document number: 2791651

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: CR2012-000451

Country of ref document: CR

Ref document number: MX/A/2012/010017

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 0165412

Country of ref document: KE

WWE Wipo information: entry into national phase

Ref document number: 2012002404

Country of ref document: CL

Ref document number: 2012556160

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 1201004390

Country of ref document: TH

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2011223795

Country of ref document: AU

Date of ref document: 20110301

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12170402

Country of ref document: CO

Ref document number: 201290844

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 2011715320

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20127025901

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13580112

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112012022060

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112012022060

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20120831