EP1017398A1 - Proteasome inhibitoren, inhibitoren des ubiquitinweges oder verbindungen, die mit der durch den ubiquitinweg enstehenden nf-kb aktivierung interferieren zur behandlung von entzündlichen und autoimmunerkrankungen - Google Patents

Proteasome inhibitoren, inhibitoren des ubiquitinweges oder verbindungen, die mit der durch den ubiquitinweg enstehenden nf-kb aktivierung interferieren zur behandlung von entzündlichen und autoimmunerkrankungen

Info

Publication number
EP1017398A1
EP1017398A1 EP98949490A EP98949490A EP1017398A1 EP 1017398 A1 EP1017398 A1 EP 1017398A1 EP 98949490 A EP98949490 A EP 98949490A EP 98949490 A EP98949490 A EP 98949490A EP 1017398 A1 EP1017398 A1 EP 1017398A1
Authority
EP
European Patent Office
Prior art keywords
lactacystin
proteasome
lactone
agent
glucocorticoid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP98949490A
Other languages
English (en)
French (fr)
Inventor
Peter Elliot
Julian Adams
Louis Plamondon
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Millennium Pharmaceuticals Inc
Original Assignee
Proscript Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Proscript Inc filed Critical Proscript Inc
Publication of EP1017398A1 publication Critical patent/EP1017398A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics

Definitions

  • This invention is directed to compositions and methods for treatment of inflammatory and autoimmune diseases.
  • Eukaryotic cells contain multiple proteolytic systems, including lysosomal proteases, calpains, ATP-ubiquitin-proteasome dependent pathway, and an ATP- independent nonlysosomal process.
  • the major neutral proteolytic activity in the cytosol and nucleus is the proteasome, a 20S (700 kDa) particle with multiple peptidase activities.
  • the 20S complex is the proteolytic core of a 26S (1500 kDa) complex that degrades or processes ubiquitin-conjugated proteins. Ubiquitination marks a protein for hydrolysis by the 26S proteasome complex. Many abnormal or short-lived normal polypeptides are degraded by the ubiquitin-proteasome-dependent pathway.
  • Abnormal peptides include oxidant-damaged proteins (e.g., those having oxidized disulfide bonds), products of premature translational termination (e.g., those having exposed hydrophobic groups which are recognized by the proteasome, and stress-induced denatured or damaged proteins (where stress is induced by, e.g., changes in pH or temperature, or exposure to metals).
  • the proteasome also participates in the rapid elimination and post-translational processing of proteins involved in cellular regulation (e.g., cell cycle, gene transcription, and metabolic pathways), intercellular communication, and the immune response (e.g., antigen presentation).
  • the transcription factor NF- ⁇ B is a member of the Rel protein family.
  • the Rel family of transcriptional activator proteins can be divided into two groups. The first group requires proteolytic processing, and includes pi 05 and pi 00, which are processed to p50 and p52, respectively. The second group does not require proteolytic processing and includes p65 (Rel A), Rel (c-Rel), and Rel B.
  • NF- ⁇ B comprises two subunits, p50 and an additional member of the Rel gene family, e.g., p 65. Unprocessed pl05 can also associate with p65 and other members of the Rel family.
  • the p50-p65 heterodimer is present in an inactive form in the cytoplasm, bound to I ⁇ B- ⁇ .
  • the ternary complex can be activated by the dissociation and destruction of I ⁇ B- ⁇ , while the p65/pl05 heterodimer can be activated by processing of pi 05.
  • the ubiquitin-proteasome pathway plays an essential role in the regulation of
  • NF- ⁇ B activity being responsible both for processing of pl05 to p50 and for the degradation of the inhibitor protein I ⁇ B- .
  • I ⁇ B- ⁇ In order to be targeted for degradation by the proteasome, I ⁇ B- ⁇ must first undergo selective phosphorylation at serine residues 32 and 36, followed by ubiquitination (Chen et al. Genes & Development (1995) 9:1586; Chen et al. Cell (1996) S4.853; Brockman et al. Mol. Cell. Biol. (1995) 75:2809; Brown et al. Science (1995) 267:1485).
  • NF- ⁇ B translocates to the nucleus, where it plays a central role in the regulation of a remarkably diverse set of genes involved in the immune and inflammatory responses (Grilli et al, International Review of Cytology (1993) 143: -62).
  • NF- ⁇ B is required for the expression of a number of genes involved in the inflammatory response, such as TNF- ⁇ gene and genes encoding the cell adhesion molecules E-selectin, P-selectin, ICAM, and VCAM (Collins, T., Lab. Invest. (1993) 65:499.
  • NF- ⁇ B is also required for the expression of a large number of cytokine genes such as IL-2, IL-6, granulocyte colony stimulating factor, and IFN- ⁇ .
  • Inducible nitric oxide synthetase is also under regulatory control of NF- ⁇ B.
  • Proteasome inhibitors block I ⁇ B- ⁇ degradation and activation of NF- ⁇ B (Palombella et al. WO 95/25533 published 9/28/95; Traenckner, et al , EMBO J. (1994) 13:5433).
  • Proteasome inhibitors also block
  • TNF- ⁇ induced expression of the leukocyte adhesion molecules E-selectin, VCAM-1, and ICAM-1 (Read, et al, Immunity (1995) 2:493).
  • Cyclins are proteins involved in cell cycle control.
  • the proteasome participates in the degradation of cyclins. Cyclin degradation enables a cell to exit one cell cycle stage (e.g., mitosis) and enter another (e.g., division). There is evidence that cyclin is converted to a form vulnerable to a ubiquitin ligase or that a cyclin-specific ligase is activated during mitosis (Ciechanover Cell (1994) 79:13). Inhibition of the proteasome inhibits cyclin degradation, and therefore inhibits cell proliferation (Kumatori et al. Proc. Natl. Acad. Sci.
  • MS Multiple sclerosis
  • Airway hyperreactivity to a variety of spasmogens and pulmonary inflammation characterized by eosinophilia are pathologies that are characteristic of asthma (Beasely, et al. Am. Rev. Resp. Dis. (1989) 739:806). Asthma is a chronic condition of the airways that involves many types of inflammatory cell and the release of many mediators and neurotransmitters that have multiple effects on the various target cells in the airway. The degree and extent of inflammation in the airway wall are broadly related to the clinical severity of the asthma.
  • the inflammatory response of asthma consists of activation of mast cells resident in the airways, increased numbers of lymphocytes (which are mainly CD4 + T lymphocytes) and an infiltration with eosinophils, which appear to degranulate.
  • lymphocytes which are mainly CD4 + T lymphocytes
  • eosinophils which appear to degranulate.
  • the present invention is directed to methods for treating a patient afflicted with multiple sclerosis or asthma comprising administering to said patient an effective amount of an agent selected from the group consisting of proteasome inhibitors, ubiquitin pathway inhibitors, agents that interfere with the activation of NF- ⁇ B via the ubiquitin proteasome pathway, and mixtures thereof.
  • an agent selected from the group consisting of proteasome inhibitors, ubiquitin pathway inhibitors, agents that interfere with the activation of NF- ⁇ B via the ubiquitin proteasome pathway, and mixtures thereof.
  • the agent is a proteasome inhibitor.
  • the proteasome inhibitor is selected from the group consisting of peptidyl aldehydes, boronic acids, boronic esters, lactacystin, and lactacystin analogs.
  • the proteasome inhibitor is lactacystin or a lactacystin analog, more preferably lactacystin, c/ ⁇ sto-lactacystin ⁇ -lactone, 7-ethyl-c/ ⁇ to-lactacystin ⁇ -lactone, 7- «-propyl-c/-35to-lactacystin ⁇ -lactone, or 7- ⁇ -butyl-c/ ⁇ .sto-lactacystin ⁇ -lactone.
  • the proteasome inhibitor is 7-rc-propyl-c/ ⁇ .sto-lactacystin ⁇ -lactone.
  • the agent is a ubiquitin pathway inhibitor.
  • the agent is one that interferes with the activation of NF- ⁇ B by the ubiquitin-proteasome pathway.
  • the agent that interferes with the activation of NF- ⁇ B is an agent that inhibits phosphorylation of
  • the invention is further directed to methods for treating a patient afflicted with asthma comprising administering to said patient an effective combination of a glucocorticoid and an agent selected from the group consisting of proteasome inhibitors, ubiquitin pathway inhibitors, agents that interfere with the activation of NF- ⁇ B via the ubiquitin proteasome pathway, and mixtures thereof.
  • the combination comprises an amount of the agent sufficient to reduce the dose or treatment frequency required for the glucocorticoid. In certain preferred embodiments, the combination comprises an amount of the glucocorticoid that is less than its standard recommended dosage. In another preferred aspect, the combination comprises an amount of the glucocorticoid sufficient to reduce the dose or treatment frequency required for the agent.
  • the glucocorticoid is selected from the group consisting of flunisolide, triamcinolone acetonide, beclomethasone dipropionate, dexamethasone sodium phosphate, fluticasone propionate, budesonide, hydrocortisone, prednisone, prednisolone, mometasone, tipredane, and butixicort.
  • the combination used to treat a patient afflicted with asthma comprises a glucocorticoid and a proteasome inhibitor. More preferably, the proteasome inhibitor is lactacystin or a lactacystin analog. Most preferably the combination comprises 7-n-propyl-c/ ⁇ zsto-lactacystin ⁇ -lactone and budesonide.
  • the invention is further directed to pharmaceutical compositions comprising a combination of a glucocorticoid and an agent selected from the group consisting of proteasome inhibitors, ubiquitin pathway inhibitors, agents that interfere with the activation of NF- ⁇ B via the ubiquitin proteasome pathway, or mixtures thereof.
  • the pharmaceutical composition is provided in a unit dosage form.
  • the unit dosage form comprises the glucocorticoid in an amount that is less than its standard recommended dosage.
  • Figure 1 is a graphical representation of mean clinical score as a function of time in an experimental autoimmune encephalomyelitis model. These data demonstrate that 3b (7-/z-propyl-c/-7-fto-lactacystin ⁇ -lactone) treatment causes a reduction in relapse rate and in mean clinical score as compared to vehicle- treated animals.
  • Figure 2 is a graphical representation of relapse rate as a function of time in an experimental autoimmune encephalomyelitis model. These data demonstrate that 3b treatment causes a reduction in the rate and severity of relapse.
  • Figure 3 is a graphical representation of leukocyte count in bronchoalveolar lavage fluid from naive (N) or actively sensitized (AS) Brown Norway rats 72 hours following exposure to aerosolized ovalbumin (10 mg/mL). Treatment with 3b causes a dose-dependent reduction in leukocyte influx.
  • Figure 4 is a graphical representation of eosinophil count in bronchoalveolar lavage fluid from naive (N) or actively sensitized (AS) Brown Norway rats 72 hours following exposure to aerosolized ovalbumin (10 mg/mL). Treatment with 3b causes a dose dependent inhibition of eosinophilia in this model.
  • Figure 5 is a graphical representation of leukocyte count in bronchoalveolar lavage fluid from naive, untreated (N); actively sensitized, vehicle-treated (V); or actively sensitized, drug-treated (A-H) Brown Norway rats 72 hours following exposure to aerosolized ovalbumin (10 mg/mL).
  • the combination of budesonide (0J mg/kg) with 3b (0.03 or 0J mg/kg) causes a reduction in leukocyte influx in this model.
  • High-dose budesonide (0.5 mg/kg) is efficacious with or without added 3b.
  • Figure 6 is a graphical representation of eosinophil count in bronchoalveolar lavage fluid from naive, untreated (N); actively sensitized, vehicle-treated (V); or actively sensitized, drug-treated (A-H) Brown Norway rats 72 hours following exposure to aerosolized ovalbumin (10 mg/mL).
  • Treatment with budesonide alone (0J mg/kg) or 3b alone (0.03 or 0J mg/kg) was ineffective.
  • the combination of budesonide (0J mg/kg) with 3b (0.03 or 0J mg/kg) causes a reduction in eosinophilia in this model.
  • High-dose budesonide (0.5 mg/kg) is efficacious with or without added 3b.
  • compositions and methods for treatment of inflammatory and autoimmune diseases are directed to compositions and methods for treatment of inflammatory and autoimmune diseases.
  • All patent applications, patents and literature references cited herein are hereby incorporated by reference in their entirety. In the case of inconsistencies the present disclosure will prevail.
  • the invention provides methods for treating a patient afflicted with multiple sclerosis or asthma comprising administering to said patient an effective amount of an agent selected from the group consisting of proteasome inhibitors, ubiquitin pathway inhibitors, agents that interfere with the activation of NF- ⁇ B via the ubiquitin proteasome pathway, and mixtures thereof. It has now been unexpectedly discovered that the ubiquitin-proteasome pathway is a target for treating multiple sclerosis, asthma, and rheumatoid arthritis.
  • Treating shall mean any amelioration of any symptom pursuant to administration of any proteasome inhibitor, ubiquitin pathway inhibitor, or agent that interferes with activation of NF- ⁇ B via the ubiquitin proteasome pathway.
  • Ultraquitin pathway inhibitor shall mean any substance which specifically inhibits ubiquitination or the transfer of ubiquitin to proteins.
  • Proteinasome inhibitor shall mean any substance which specifically inhibits inhibits the proteasome or the activity thereof.
  • Agents that interfere with activation of NF- ⁇ B by the ubiquitin-proteasome pathway shall mean any substance that 1) specifically inhibits the proteasome or the activity thereof; 2) specifically inhibits ubiquitination of I ⁇ B- ⁇ or pi 05; or 3) specifically inhibits phosphorylation of I ⁇ B- ⁇ or pi 05.
  • “Specifically inhibits” shall mean interferes with the ability of a protein to mediate its biological function at an inhibitor concentration that is lower than the concentration of the inhibitor required to produce another, unrelated biological effect.
  • the concentration of the inhibitor required for such interference is at least 2-fold lower, more preferably at least 5-fold lower, even more preferably at least 10-fold lower, and most preferably at least 20- fold lower than the concentration required to produce an unrelated biological effect.
  • Such inhibitors can act by any of a variety of mechanisms, including without limitation, interfering with the active site or conformation of the protein, interfering with the ability of the protein to interact with another protein, substrate, or co-factor, either by an effect on the protein itself or on the other protein, substrate, or cofactor, and altering the microenvironment in which the biological function of the protein normally occurs.
  • the invention provides methods for treating multiple sclerosis.
  • Multiple sclerosis is an incurable neurological illness that frequently causes chronic disability. It is widely believed that MS is the result of an autoimmune disorder in a genetically susceptible individual, mediated by autoreactive T cells that migrate into the CNS and initiate the inflammatory demyelinating lesion.
  • MS is an autoimmune disease is derived in part from systemic abnormalities of immune function seen in patients with the disease, and in part through similarities with experimental autoimmune encephalomyelitis (EAE), which in turn serves as a model for the human disease (Kennedy, et al J. Neuroimmunol (1987) 76:345; Arnason, et al. Neurol. Clin.
  • EAE is a T-cell-mediated inflammatory, autoimmune demyelinating disease of the CNS.
  • the disease can be induced in a number of experimental laboratory animals, including primates, by the injection of whole brain homogenate, a purified preparation of myelin basic protein (MBP), or proteolipoprotein (PLP) in adjuvant.
  • MBP myelin basic protein
  • PLP proteolipoprotein
  • R-EAE experimental autoimmune encephalomyelitis
  • the clinical disease is characterized by an acute paralytic phase followed by recovery and subsequent relapses. This pattern of relapses and spontaneous recovery in the experimental animal model, which occurs over a period of weeks to months, is very similar to the clinical signs of disease observed in multiple sclerosis (MS) patients over many years.
  • MS multiple sclerosis
  • the method according to this aspect of the invention comprises administering to a patient afflicted with MS an effective amount of an agent selected from the group consisting of proteasome inhibitors, ubiquitin pathway inhibitors, agents that interfere with the activation of NF- ⁇ B via the ubiquitin proteasome pathway, and mixtures thereof.
  • the agent is administered in an amount sufficient to reduce the frequency or severity of relapse of the disease.
  • the proteasome inhibitor 3b When administered during the remission phase at doses of 0.3 or 1.0 mg/kg i.p., the proteasome inhibitor 3b reduced the rate and severity of relapse in the R-EAE model (Figs. 1-2).
  • the invention provides a method for treating asthma.
  • Asthma is an obstructive lung disorder characterized by airway hyperresponsiveness, which is an exaggerated airway narrowing in response to many different stimuli, such as histamine, exercise, cold air, and allergen. Because of the episodic constriction of the bronchial tubes, treatment has been based partly on bronchodilation by ⁇ -adrenergic agonist drugs. More recently, however, it has become appreciated that asthma is a chronic condition of the airways that involves many types of inflammatory cell and the release of many mediators and neurotransmitters that have multiple effects on the various target cells in the airway. The degree and extent of inflammation in the airway wall are broadly related to the clinical severity of the asthma.
  • the inflammatory response of asthma consists of activation of mast cells resident in the airways, increased numbers of lymphocytes (which are mainly
  • the method according to this aspect of the invention comprises administering to a patient afflicted with asthma an effective amount of an agent selected from the group consisting of proteasome inhibitors, ubiquitin pathway inhibitors, agents that interfere with the activation of NF- ⁇ B via the ubiquitin proteasome pathway, and mixtures thereof.
  • the agent is administered in an amount sufficient to reduce the frequency or severity of asthmatic attacks.
  • 3b When administered intratracheally at 1 hour prior to and 24 hours and 48 hours after allergen challenge, 3b (OJ or 0.3 mg/kg) inhibited eosinophilia in actively sensitized Brown Norway rats (Figs. 3-4).
  • asthma is a registered trademark of many drugs used to treat asthma.
  • accepted therapies for asthma include cromoglycate, nedocromil, theophylline, short- and long-acting ⁇ 2 -adrenergic receptor agonists, and inhaled or oral glucocorticoids.
  • More recently developed therapeutics include inhibitors of leukotriene biosynthesis, leukotriene receptor antagonists, and thromboxane antagonists.
  • aAnti-IL-5 and anti-IgE antibodies are being developed (Science (1997) 276: 1643), and antisense approaches are also being investigated (Nyce and Metzger, Nature (1997) 353:721).
  • the agent of the invention is used in an amount sufficient to reduce the dose or treatment frequency required for the other drug or drugs.
  • the other drug or drugs are used in an amount sufficient to reduce the dose or treatment frequency required for the agent of the invention.
  • the agent may be administered at the same time as the other drug or drugs or may be administered at a different time.
  • Steroid therapy is particularly effective for the treatment of asthma, and is an essential line of therapy for severe asthmatics.
  • a number of untoward side-effects result from long-term steroid use, including bone growth suppression, adrenal insufficiency, Cushing's syndrome, cataracts, immunosuppression, and excessive bruising. Many of these effects can be minimized by topical administration of the drug to the lung by inhalation.
  • high doses, such as those required in severe cases result in significant systemic exposure and an increase in the associated side-effects.
  • Drugs that permit the reduction in steroid dose (“steroid-sparing") thus offer very real clinical benefit.
  • the invention provides methods for treating a patient afflicted with asthma comprising administering to the patient a combination of a glucocorticoid and an agent selected from the group consisting of proteasome inhibitors, ubiquitin pathway inhibitors, agents that interfere with the activation of NF- ⁇ B via the ubiquitin proteasome pathway, and mixtures thereof.
  • the glucocorticoid and the agent may be administered at the same or different times, on the same or different days, and with the same or different frequency.
  • the doses of each drug are spaced so as to achieve a combined physiological effect.
  • the glucocorticoid is administered between 0 minutes and about one month before or after the agent of the invention, more preferably between 0 minutes and about one week before or after the agent of the invention, most preferably between 0 minutes and 24 hours before or after the agent of the invention.
  • Glucocorticoids for use in the invention include, but are not limited to, flunisolide, triamcinolone acetonide, beclomethasone dipropionate, dexamethasone sodium phosphate, fluticasone propionate, budesonide, hydrocortisone, prednisone, prednisolone, mometasone, tipredane, and butixicort.
  • the glucocorticoid is budesonide. Suitable formulations, dosages, and routes of administration for glucocorticoids are known in the art (Physician's Desk Reference, 51st Edition, 1997,
  • the agent of the invention is administered in an amount sufficient to reduce the dose or treatment frequency required for the glucocorticoid.
  • the amount of glucocorticoid administered does not exceed the standard recommended dosage, and more preferably the amount of glucocorticoid administered is less than the standard recommended dosage for the drug when administered alone.
  • the amount of glucocorticoid administered is sufficient to reduce the dose or treatment frequency required for the agent selected from the group consisting of proteasome inhibitors, ubiquitin pathway inhibitors, agents that interfere with the activation of NF- ⁇ B via the ubiquitin proteasome pathway, and mixtures thereof.
  • the agent selected from the group consisting of proteasome inhibitors, ubiquitin pathway inhibitors, agents that interfere with the activation of NF- ⁇ B via the ubiquitin proteasome pathway, and mixtures thereof produces efficacy with fewer or less severe side effects or toxicity than treatment with either drug alone.
  • the invention provides pharmaceutical compositions comprising a combination of a glucocorticoid and an agent selected from the group consisting of proteasome inhibitors, ubiquitin pathway inhibitors, agents that interfere with the activation of NF- ⁇ B via the ubiquitin proteasome pathway, and mixtures thereof, are further contemplated within the scope of the invention.
  • the pharmaceutical compositions of the invention can be provided in unit dosage form.
  • the unit dosage form contains an amount of glucocorticoid that is less than its standard recommended dosage when administered by itself.
  • proteasome inhibitors ubiquitin pathway inhibitors
  • agents that interfere with the activation of NF- ⁇ B via the ubiquitin proteasome pathway applies to the pharmaceutical formulations as well as to the methods according to the invention.
  • Non-limiting examples of proteasome inhibitors for use in the present invention include peptidyl aldehydes (Orlowski et al. U.S. Patent 5,580,854; Stein et al. WO 95/24914; Siman et al. WO 91/13904; Iqbal et al. J. Med. Chem. 35:2276-2277 (1995)), peptidyl boronic acids (Adams et al. WO 96/13266; Siman et al. WO 91/13904), other peptidyl derivatives with proteasome inhibitory activity (Iqbal et al. U.S. Patent 5,614,649; Iqbal et al. U.S. Patent 5,550,262; Spaltenstein et al. Tetrahedron Letters
  • the agents disclosed herein may be administered by any route, including intradermally, intraperitoneally, intranasally, intratracheally, subcutaneously, orally or intravenously.
  • administration is preferably by the inhalation route.
  • Peptide aldehyde proteasome inhibitors for use in the present invention preferably are those disclosed in Stein et al. WO 95/24914 published September 21, 1995 or Siman et al WO 91/13904 published September 19, 1991, both hereby incorporated by reference in their entirety.
  • Boronic acid or ester compounds for use in the present invention preferably include those disclosed in Adams et al WO 96/13266, Siman et al. WO 91/13904, or Iqbal et al. U.S. Pat. 5,614,649, each of which is hereby incorporated by reference in its entirety.
  • the boronic acid compound for use in the present invention is selected from the group consisting of : N-acetyl-L-leucine- ⁇ -(l- napthyl) L-alanine-L-leucine boronic acid, ⁇ -(l-napthyl)-L-alanine-L-leucine boronic acid, N-(4-morpholine)carbonyl- ⁇ -( 1 -naphthyl)-L-alanine-L-leucine boronic acid, N-(8-quinoline)sulfonyl- ⁇ -( 1 -naphthyl)-L-alanine-L-leucine boronic acid,
  • Lactacystin and lactacystin analog compounds for use in the present invention preferably include those disclosed in Fenteany et al W ⁇ 96/32105, or Soucy etal.
  • U.S. patent application (08/912,111; filed 08/15/97), each of which is hereby incorporated by reference in its entirety.
  • the lactacystin analog compound is selected from the group consisting of lactacystin, c/ sto-lactacystin ⁇ -lactone, 7-ethyl-c7 ⁇ sto-lactacystin ⁇ -lactone, 7-w-propyl-c/ ⁇ to-lactacystin ⁇ -lactone, and 7- ⁇ -butyl-c/ ⁇ 5to-lactacystin ⁇ -lactone. These compounds can be prepared as illustrated in Schemes 1 and 2. Most preferably, the lactacystin analog compound is 7- «-propyl-c/ ⁇ sto-lactacystin ⁇ -lactone (3b (Scheme 2)).
  • the agent used to treat a patient afflicted with multiple sclerosis or asthma is a proteasome inhibitor.
  • the proteasome inhibitor is lactacystin or a lactacystin analog, more preferably 7- ⁇ -propyl-c/ ⁇ st ⁇ -lactacystin ⁇ -lactone.
  • the combination used to treat a patient afflicted with asthma comprises a glucocorticoid and a proteasome inhibitor. More preferably, the proteasome inhibitor is lactacystin or a lactacystin analog. Most preferably the combination comprises 7-M-propyl-c/ ⁇ st ⁇ -lactacystin ⁇ -lactone and budesonide.
  • Non-limiting examples of ubiquitin pathway inhibitors include those disclosed in Berleth et al, Biochem. 35(5) : 1664- 1671, (1996). Inhibitors of I ⁇ B- ⁇ phosphorylation are also known (Chen, Cell 54:853 (1996); Chen U.S. patent application 08/825,559).
  • the concentration of a disclosed compound in a pharmaceutically acceptable mixture will vary depending on several factors, including the dosage of the compound to be administered, the pharmacokinetic characteristics of the compound(s) employed, and the route of administration.
  • Effective amounts of agents for treating multiple sclerosis, asthma, or rheumatoid arthritis would broadly range between about 10 ⁇ g and about 50 mg per Kg of body weight of a recipient mammal.
  • the agent may be administered in a single dose or in repeat doses. Treatments may be administered daily or more frequently depending upon a number of factors, including the age and overall health of a patient, and the formulation and route of administration of the selected compound(s). Other factors to be considered in determining dosage include kind of concurrent treatment, if any; frequency of treatment and the nature of the effect desired; extent of tissue damage; gender; duration of symptoms; counter indications, if any; and other variables to be assessed by the individual physician.
  • the concentration of the proteasome inhibitor is determined by measuring the activity of the proteasome activity ex vivo after administering the proteasome inhibitor to the mammal. Such measurement comprises obtaining one or more test biological samples from the mammal at one or more specified times after administering the proteasome inhibitor; measuring proteasome activity in the test biological sample or samples; determining the amount of proteasome activity in the test biological sample or samples; and comparing the amount of proteasome activity in the test biological sample to that in a reference biological sample obtained from a mammal to which no proteasome inhibitor has been administered.
  • the biological samples that are obtained from the mammal may include, without limitation, blood, urine, organ, and tissue samples.
  • the biological sample is a blood sample, more preferably a blood sample from which the white blood cells are isolated prior to measuring proteasome activity.
  • Methods for fractionating blood cells are known in the art and are further described in the Examples. Hemoglobin strongly interferes with fluorescence measurements and is thus preferably excluded from test samples when fluorometric assays for proteasome activity are used.
  • the white blood cells are lysed by standard procedures. Variability in lysis efficiency can be corrected for by determining total protein content in the sample using standard procedures and normalizing the proteasome activity measured in the sample relative to the protein content in the sample.
  • a substrate having a detectable label is provided to the reaction mixture and proteolytic cleavage of the substrate is monitored by following disappearance of the substrate or appearance of a cleavage product. Detection of the label may be achieved, for example, by fluorometric, colorimetric, or radiometric assay.
  • the substrate is a peptide substrate and the reaction mixture further comprises a 20S proteasome activator.
  • the activator is one taught in Coux et al. (Ann. Rev. Biochem. 65:
  • the peptide substrate contains a cleavable fluorescent label and release of the label is monitored by fluorometric assay. More preferably, the substrate is N- succinylleucylleucylvalyltyrosyl 7- amino-4-methylcoumarin (Suc-Leu-Leu-Val-Tyr- AMC).
  • test biological sample and the reference biological sample each separately comprise a plurality of samples pooled from a group of mammals, preferably mice, undergoing treatment.
  • test biological sample and the reference biological sample each comprise a single sample obtained from an individual mammal. Assaying of individual samples is presently preferred except when impractical due to the small size of the mammal.
  • a statistical sample is obtained by pooling data from individual test biological samples or from individual reference biological samples.
  • Day-to-day variability in the assay may result from factors such as differences in buffer solutions, operator variability, variability in instrument performance, and temperature variability. Such variability can be minimized by standardizing proteasome activity in both the biological sample and the reference sample relative to a standard proteasome sample.
  • the standard sample comprises purified 20S proteasome, more preferably purified 20S proteasome from rabbit reticulocytes.
  • the reference sample is obtained from the treated mammal prior to initiation of treatment. This embodiment is presently preferred for higher mammals in order to minimize the impact of inter-mammal variability.
  • Clinical monitoring of drug action presently preferably entails this embodiment of the invention, with each patient serving as his or her own baseline control.
  • a decrease in inhibitor activity in the biological sample as compared to the reference sample is indicative of an in vivo effect of the inhibitor at the time the biological sample was obtained.
  • biological samples are obtained at multiple timepoints following administration of the inhibitor.
  • measurement of proteasome activity in the biological samples provides an indication of the extent and duration of in vivo effect of the inhibitor.
  • multiple biological samples are obtained from a single mammal at one or more time points.
  • measurement of proteasome activity in the biological samples provides an indication of the distribution of the inhibitor in the mammal.
  • the dose amount and dose frequency of the proteasome inhibitor are selected so as to avoid excessive proteasome inhibition.
  • excessive proteasome inhibition results in a toxic effect, the toxic effect including, but not being limited to, vomiting, diarrhea, hypovolemia, hypotension, and lethality.
  • the dose amount and dose frequency of the proteasome inhibitor are selected so that proteasome inhibition in any future biological sample does not exceed about 95%.
  • the dose amount and dose frequency of the proteasome inhibitor are selected so that therapeutically useful proteasome inhibition is achieved.
  • therapeutically useful proteasome inhibition results in a therapeutically beneficial antitumor, antiinflammatory, antiviral, or antiparasitic effect.
  • the dose amount and dose frequency of the proteasome inhibitor are selected so that proteasome inhibition of at least about 15%, preferably about 20%, more preferably about 30%, even more preferably about 40%, and still more preferably about 50%, and most preferably from about 50% to about 80% is achieved in a future biological sample, although in some instances proteasome inhibition as high as 95% may be preferred.
  • Agents for use in this invention may be prepared for administration by any of the methods well known in the pharmaceutical art, for example, as described in Remington's Pharmaceutical Sciences (Mack Pub. Co., Easton, PA, 1980). Agents may be prepared for use in parenteral administration in the form of solutions or liquid suspensions; for oral administration in the form of tablets or capsules; for intranasal or intratracheal administration in the form of powders, gels, oily solutions, nasal drops, aerosols, or mists. Formulations for parenteral administration may contain as common excipients sterile water or sterile saline, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, hydrogenated naphthalenes, and the like.
  • Controlled release of an agent may be obtained, in part, by use of biocompatible, biodegradable polymers of lactide, and copolymers of lactide/glycolide or polyoxyethylene/polyoxypropylene.
  • Additional parenteral delivery systems include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes.
  • Formulations for inhalation administration may contain lactose, polyoxyethylene-9-lauryl ether, glycocholate, or deoxycholate.
  • the inhalation route of administration is preferred in order to minimize potential side effects or toxicity resulting from systemic exposure to the agent.
  • an "effective amount" an agent is an amount sufficient to produce any amelioration of any symptom or sign of the disease (Stites et al. Basic & Clinical Immunology Lange Medical Publications, Los Altos, California, 1982).
  • mice Female SJL/J mice, 6 weeks old, were purchased from Harlan Laboratories (Indianapolis, IN), housed in the Northwestern animal care facility, and maintained on standard laboratory food and water ad libitum. Paralyzed mice were afforded easier access to food and water.
  • PLP 139-151 HLGKWLGHPDKF was purchased from Peptides International (Louisville, KY). Amino acid composition was verified by mass spectrometry and purity (>98%) was assessed by HPLC.
  • mice were observed daily for clinical signs of disease. Mice were scored according to their clinical severity as follows: grade 0, no abnormality; grade 1, limp tail; grade 2, limp tail and hind limb weakness (waddling gait); grade 3, partial hind limb paralysis; grade 4, complete hind limb paralysis; and grade 5, moribund.
  • a relapse was defined as an increase of at least one full grade in clinical score after the animal had previously improved at least a full clinical score and had stabilized.
  • Animals treated with 3b showed reduced clinical scores as compared to vehicle-treated animals.
  • the incidence of relapse was 5/10 for the 0.3 mg/kg group and 2/10 for the 1.0 mg/kg group, as compared to 6/10 for the vehicle-treated group.
  • Data are also plotted as mean daily relapse incidence for all animals in a particular treatment group (Fig. 2).
  • the mean maximal clinical score per group is also provided as an indication of disease severity. Animals treated with 3b (both dosage groups) showed reduced rate of relapse and reduced severity of disease as compared with vehicle-treated animals.
  • Ovalbumin (OA; 10 ⁇ g) mixed with aluminum hydroxide gel (10 mg) will be injected (0.5 mL, i.p.) into Brown Norway rats and repeated 7 and 14 days later.
  • sensitized rats were anaesthetized (halothane 5% in O 2 ) and 3b, dexamethasone, or vehicle (lactose) was instilled via a cannula placed directly into the trachea at 1 hour prior to OA exposure. This procedure was repeated at 24 hours and 48 hours after OA exposure.
  • the lungs were lavaged using 3 aliquots (4 mL) of Hank's solution (HBSS x 10, 100 mL; EDTA 100 mM, 100 mL; HEPES 1M, 10 mL made to 11 mL with water); recovered cells were pooled and the total volume of recovered fluid was adjusted to 12 mL by addition of Hank's solution. Total cells were counted (Sysmex Microcell Counter F-500, TOPA Medical Electronics Ltd., Japan). Smears were made by diluting recovered fluid (to approximately 10 6 cells/mL) and spinning an aliquot (100 ⁇ L) in a centrifuge (Cytospin, Shandon, UK).
  • Smears were air dried, fixed using a solution of fast green in methanol (2 mg/L) for 5 seconds and stained with eosin G (5 seconds) and thiazine (5 seconds) (Diff-Quik, Baxter Dade Ltd, Switzerland) in order to differentiate eosinophils, neutrophils, macrophages and lymphocytes.
  • eosin G 5 seconds
  • thiazine 5 seconds
  • a total of 500 cells per smear were counted by light microscopy under oil immersion (x 1000).
  • Ovalbumin challenge resulted in a significant increase in eosinophils, neutrophils, and total leukocytes in BAS fluid from actively sensitized (AS) Brown Norway rats as compared to naive (N) rats.
  • Treatment with dexamethasone (0J mg/kg i.t.) prevented this increase.
  • At doses of 0J and 0.3 mg/kg, 3b reduced the influx of eosinophils and total leukocytes.
  • a significant decrease in lymphocyte count was also observed at all doses (Figs. 3-4).
  • Compound 3b is effective in preventing leukocyte influx following allergen challenge in an animal model of asthma.
  • Example 3 Effect of Treatment with a Combination of 3b and Budesonide on Allergen-Induced Pulmonary Leukocyte Accumulation in Actively Sensitized Brown Norway Rats
  • Ovalbumin (OA; 10 ⁇ g) mixed with aluminum hydroxide gel (10 mg) will be injected (0.5 mL, i.p.) into Brown Norway rats and repeated 7 and 14 days later.
  • sensitized rats were anaesthetized (halothane 5% in O 2 ) and treated intratracheally (i.t.) 1 hour prior to OA exposure with vehicle (group V; lactose, 1 mg), budesonide (group C, OJ mg/kg; group F, 0.5 mg/kg), 3b (group A, 0.03 mg/kg; group B, 0J mg/kg), or mixtures of budesonide and 3b (group D, 0J/0.03 mg/kg; group E, 0J/0J mg/kg; group G, 0.5/0.03 mg/kg; group H, 0.5/0J mg/kg).
  • Drug was instilled via a cannula placed directly into the trachea. This procedure was repeated at 24 hours and 48 hours after OA exposure.
  • the lungs were lavaged using 3 aliquots (4 mL) of Hank's solution (HBSS x 10, 100 mL; EDTA 100 mM, 100 mL; HEPES 1M, 10 mL made to 11 mL with water); recovered cells were pooled and the total volume of recovered fluid was adjusted to 12 mL by addition of Hank's solution. Total cells were counted (Sysmex Microcell Counter F-500, TOPA Medical Electronics Ltd., Japan). Smears were made by diluting recovered fluid (to approximately 10 6 cells/mL) and spinning an aliquot (100 ⁇ L) in a centrifuge (Cytospin, Shandon, UK).
  • Smears were air dried, fixed using a solution of fast green in methanol (2 mg/L) for 5 seconds and stained with eosin G (5 seconds) and" thiazine (5 seconds) (Diff-Quik, Baxter Dade Ltd, Switzerland) in order to differentiate eosinophils, neutrophils, macrophages and lymphocytes.
  • eosin G 5 seconds
  • thiazine 5 seconds
  • a total of 500 cells per smear were counted by light microscopy under oil immersion (x 1000).
  • Ovalbumin challenge resulted in a significant increase in eosinophils, neutrophils, and total leukocytes in BAS fluid from actively sensitized, vehicle treated (V) Brown Norway rats as compared to naive, untreated (N) rats.
  • V vehicle treated
  • OJ mg/kg B
  • 3b failed to prevent this increase.
  • OJ mg/kg C
  • budesonide also had not effect when administered alone.
  • the combination of OJ mg/kg budesonide with 3b at 0.03 mg/kg (D) or 0J mg/kg (E) produced a significant reduction in eosinophil count.
  • the combination of compound 3b with the glucocorticoid budesonide is effective in preventing leukocyte influx following allergen challenge in an animal model of asthma at doses where neither drug alone has any effect.
  • 4- Methylvaleryl chloride was prepared from commercially available 4-methylvaleric acid in the following way: a cold (0 °C) solution of 4-methylvaleric acid (1.85 mL, 15.0 mmol) in 50 mL anhydrous CH 2 C1 2 containing 10 mL of DMF was treated with 1.9-5 ⁇ L oxalyl chloride (22.5 mmol). The mixture was then stirred for 3 h at room temperature, concentrated in vacuo and filtered to affords 1.65 g (100%) of the desired acid chloride as a colorless liquid.
  • acyl oxazolidinone lid was obtained in 80% yield.
  • diethylamide 12a was obtained in 73% yield.
  • diethylamide 12c was obtained in 94% yield. ⁇ NMR (300 MHz, CDC1 3 ) ⁇ 7.35-7.25 (m, 5H), 4.51
  • diethylamide 12d was obtained in 95% yield.
  • aldehyde 14a was obtained in 80% yield.
  • aldehyde 14e was obtained in 97% yield.
  • Aldol product 5b was also obtained in 100% yield by a procedure analogous to that described above but using cis-oxazoline 21 (see below) instead of tr ⁇ ns-oxazoline 4.
  • aldol 5a was hydrogenated at 55 p.s.i. for 48 h to provide ⁇ -lactam 7a in 72% yield.
  • a single dose intravenous pharmacokinetics study with N-(pyrazine)carbonyl-L- phenylalanine-L-leucine boronic acid (1) was conducted in Sprague-Dawley rats (140 to 280 g). Animals were assigned to 3 groups (6/sex in Groups 1 and 2; 9/sex in Group 3). Animals in groups 1, 2, and 3 received 0.03, 0J or 0.3 mg/kg of 1, respectively, in the same dose volume.
  • Blood samples (approximately 1.0 mL) were collected from the jugular vein of animals pre-dose and at approximately 10 and 30 min and 1, 3 and 24 h post-dose on Day 1. The samples were assayed for 1 using a chromatography/mass spectroscopy (LC/MS/MS) method. The lower limit of quantitation for analysis was established at 2.5 ng/mL for 1 in rat plasma and whole blood.
  • LC/MS/MS chromatography/mass spectroscopy
  • t values in plasma and whole blood in males were 14.0 and 18.6 ng » h/mL, respectively and in females were 12.9 and 17.7 ng»h mL, respectively.
  • Estimation of the elimination half-life (tj 2 ) was not possible due to the fluctuation of 1 levels during the terminal phase. The observations suggest that 1 is rapidly cleared from the blood.
  • Example 7 Preparation of blood samples for in vitro measurement of 20S proteasome activity This preparation procedure applies to blood samples collected from mammals, particularly human subjects, Cynomologus monkeys, rats and mice. Peripheral white blood cells are separated from blood samples upon collection for storage at about -70 °C until tested. To prevent interference with the fluroescence assay, it is important that the sample preparation remove all hemoglobin.
  • the required amount of blood is collected into a tube containing anticoagulant.
  • approximately 5 mL of blood is required; for rats, approximately 4 mL of blood is needed; for mice, approximately 1 mL of blood is needed from each of five mice, and the five blood samples are pooled to provide approximately 5 mL.
  • the blood sample is diluted 1:1 (v/v) with sterile saline, and the blood-saline mixture is layered over ⁇ 1 mL of NycoprepTM separation medium (GIBCO BRL
  • Example 8 Assay to measure 20S proteasome activity in peripheral white blood cells
  • the assay is based upon the SDS-inducible chymotrypsin-like activity of free 20S particles. It uses fluorometry to measure the rate at which the 20S proteasome hydrolyzes an amide bond in a small peptide substrate. Measurement of this rate in the absence and in the presence of an inhibitor allows a determination of how enzyme is bound by inhibitor.
  • This assay is used to measure 20S proteasome activity in peripheral white blood cells in mammals, particularly humans, Cynomolgus monkeys, rats and mice.
  • PROCEDURE White blood cells prepared as described in Example 1, are lysed by adding 200 ⁇ L of 5 mM EDTA to each sample. The samples are allowed to stand on ice for at least 1 h.
  • the Ys substrate is dissolved in DMSO.
  • Purified 20S proteasome standard from rabbit reticulocytes is diluted 1:9 (v/v) in 20 mM HEPES/0.5 mM EDTA (pH 7.8).
  • the Ys substrate buffer contains 20 mM HEPES, 0.5 mM EDTA, 0.35% SDS, and 60 ⁇ M Ys substrate.
  • Coomassie protein assay (measuring total protein content) and hemoglobin assay are performed on the test sample following standard procedures using commercially available kits.
  • the amount of white blood cell protein present is calculated accordmg to the following formula:
  • WBC protein ( ⁇ g/mL) total protein ( ⁇ g/mL) - [hemoglobin ( ⁇ g/mL) * 0.56]
  • the 20S proteasome standard is diluted 1:10 in 20 mM HEPES/0.5 mM EDTA (pH 7.8) to form a 12 ⁇ g/mL stock solution and placed on ice. 10 ⁇ L of the standard 20S proteasome solution is added to a cuvette and the reaction is run for 10 minutes. The maximum linear slope is measured on a fluorometer and provides a measurement of standard proteasome activity.
  • the amount of 20S proteasome activity in the test sample is calculated according to the following formula:
  • Blood samples (approximately 2 mL each) were obtained on five occasions from five human volunteers over a period of ten weeks. After collection, white blood cells were isolated from the individual blood samples using a
  • Ny cop repTM The resulting pellet was stored in a freezer set to maintain -60 °C to -80 °C until the day of testing. Samples collected on each occasion were tested together and each sample was tested in duplicate.
  • 20S proteasome activity was determined by measuring the rate of proteolytic hydrolysis of a fluorescent (aAMC)-tagged peptide substrate by the sample and normalizing the activity to the amount of cell-specific protein present in the lysate, as described in Example 2 above. 20S proteasome activity in the sample was determined from the equation:
  • C conversion factor equating the amount of fluorescence to the concentration of free AMC (FU/pmol AMC).
  • Dose formulations of 1 were prepared daily during the course of the study. Dilutions were prepared from a stock solution. The stock solution of 1 was made up in 98% saline (0.9%), 2% ethanol with 0.1% ascorbic acid. Dilutions of the stock were made in the same vehicle.
  • mice Female CD2-F1 mice (18 to 20 g), female BALB/c mice (18 to 20 g), female Wistar rats (150 to 200 g) and male Sprague-Dawley rats (250 to 450 g) were obtained from Taconic Farms (Germantown, NY). Animals were observed for at least one week and examined for general health before study initiation. Animals used in these studies were asymptomatic. Mice were housed 5 per cage and rats 3 per cage in polycarbonate cages. Corn Cob bedding (aAND-1005; farmers Exchange, Framingham, MA) was used during the observation and study periods. Fluorescent lighting was controlled to automatically provide alternate light and dark cycles of approximately 12 hours each.
  • Temperature and humidity were centrally controlled and recorded daily and readings ranged between 21 +/- 2 °C and 45 +/- 5 °C, respectively.
  • Pellets of standard rodent chow (#5001, Purina, St. Louis, MO) were available ad libitum throughout the observation and study periods. Cambridge city tap water was provided by water bottles ad libitum. No contaminants of food and water are known which would be expected to interfere with the study.
  • Drugs were administered in vehicle intravenously (IV) using a dose volume of 100 ⁇ L per mouse or 1.0 mL/kg in rats. Control groups were administered with the vehicle (98% saline [ 0.9%], 2% ethanol, 0.1% ascorbic acid).
  • mice Female CD2-F1 mice (18 to 20 g) and female BaALB/c mice (18 to 20 g) were administered a single intravenous dose of 1 (0.1 to 3.0 mg/kg in a dose volume of 100 ⁇ L).
  • the vehicle was 98% saline [0.9%], 2% ethanol, 0.1% ascorbic acid.
  • Blood samples were collected at 1.0 and 24 h following administration. Due to the blood volume required in the 20S proteasome activity assay, groups of five mice were sacrificed at the same time and their blood pooled to generate single data points.
  • a significant dose-related decrease in 20S proteasome activity was observed 24 h after administration of the last dose for alternate daily intravenous administration of 1 for 14 days.
  • the dose-related decreases in 20S proteasome activity were significant (p ⁇ 0.05) in the dose groups 0.2 mg kg.
  • a significant (p ⁇ 0.05) dose-related decrease in 20S proteasome activity was also observed 24 h after administration of the last dose for once weekly intravenous administration of 1 for 8 weeks.
  • the dose-related decreases in 20S proteasome activity were significant (p ⁇ 0.05) in the dose groups > 0J mg/kg.
  • mice Male and female Cynomolgus monkeys (2.2 to 3.5 kg) were assigned to four groups (5/sex/group). Each group received 0 (vehicle control), 0.045, 0.067 or 0.100 mg/kg/dose of 1 as a single intravenous injection in a dose volume of 0.3 mL/kg twice weekly for 4 weeks (days 1, 5, 8, 12, 15, 19, 22 and 26). The vehicle was 0.1% ascorbic acid/2% ethanol/98% saline (0.9%). Three males from the control, low- and mid-dose groups, two high-dose males, and three females/group were sacrificed at the end of treatment on Day 27. Two animals/sex/group were designated as recovery animals and received treatment for 4 weeks followed by 2 weeks of recovery; they were sacrificed on Day 41.
  • Blood was collected for 20S proteasome activity determination prior to treatment, at 1.0 h after dosing on Days 1, 8, 15 and 22, and at 1.0 h prior to dosing on Days 5, 12, 19 and 26; and on Days 31, 34, 38, and 41 (recovery sacrifice animals). Blood was also collected for 20S proteasome activity determination from the high-dose male before it was sacrificed in moribund condition on Day 26 after receiving 8 doses.
EP98949490A 1997-09-25 1998-09-25 Proteasome inhibitoren, inhibitoren des ubiquitinweges oder verbindungen, die mit der durch den ubiquitinweg enstehenden nf-kb aktivierung interferieren zur behandlung von entzündlichen und autoimmunerkrankungen Withdrawn EP1017398A1 (de)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US6103897P 1997-09-25 1997-09-25
US61038P 1997-09-25
US6956297P 1997-12-12 1997-12-12
US69562P 1997-12-12
US7488798P 1998-02-17 1998-02-17
US74887P 1998-02-17
PCT/US1998/020065 WO1999015183A1 (en) 1997-09-25 1998-09-25 PROTEASOME INHIBITORS, UBIQUITIN PATHWAY INHIBITORS OR AGENTS THAT INTERFERE WITH THE ACTIVATION OF NF-λB VIA THE UBIQUITIN PROTEASOME PATHWAY TO TREAT INFLAMMATORY AND AUTOIMMUNE DISEASES

Publications (1)

Publication Number Publication Date
EP1017398A1 true EP1017398A1 (de) 2000-07-12

Family

ID=27369960

Family Applications (1)

Application Number Title Priority Date Filing Date
EP98949490A Withdrawn EP1017398A1 (de) 1997-09-25 1998-09-25 Proteasome inhibitoren, inhibitoren des ubiquitinweges oder verbindungen, die mit der durch den ubiquitinweg enstehenden nf-kb aktivierung interferieren zur behandlung von entzündlichen und autoimmunerkrankungen

Country Status (5)

Country Link
EP (1) EP1017398A1 (de)
JP (1) JP2001517631A (de)
AU (1) AU9580098A (de)
CA (1) CA2304622A1 (de)
WO (1) WO1999015183A1 (de)

Families Citing this family (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1430903A1 (de) 2000-10-12 2004-06-23 Viromics Gmbh Verwendung von Proteasome Hemmern zur Behandlung von Virus-Infektionen
DE10051716A1 (de) * 2000-10-12 2002-04-25 Ulrich Schubert Mittel zur Hemmung der Freisetzung, Reifung und Replikation von Retroviren
EP1341414B1 (de) 2000-11-16 2013-01-02 The Regents of The University of California Marines actinomyceten-taxon für die wirkstoff-forschung
DK1355910T3 (da) 2001-01-25 2011-06-27 Us Of America Represented By The Secretary Dept Of Health And Human Services Formulering af borsyreforbindelser
US7112588B2 (en) 2001-05-21 2006-09-26 Alcon, Inc. Use of proteasome inhibitors to treat dry eye disorders
CN1509183A (zh) 2001-05-21 2004-06-30 蛋白酶体抑制剂在治疗干眼疾病中的用途
CA2466870A1 (en) * 2001-11-26 2003-06-05 Trustees Of Tufts College Methods for treating autoimmune disorders, and reagents related thereto
US20050203029A1 (en) 2002-04-05 2005-09-15 Ulrich Schubert Agents for treating <I>flaviviridae</I>infections
US7176232B2 (en) 2002-06-24 2007-02-13 The Regents Of The University Of California Salinosporamides and methods for use thereof
KR20060026052A (ko) 2003-06-20 2006-03-22 니리어스 파마슈티컬즈, 인코퍼레이션 암, 염증 및 감염성 질환의 치료를 위한 [3.2.0]헤테로사이클릭 화합물 및 그 유사체의 사용방법
CN1823070A (zh) 2003-06-20 2006-08-23 加利福尼亚大学董事会 盐孢菌酰胺及其使用方法
EP2025679A3 (de) * 2004-04-30 2009-07-08 Nereus Pharmaceuticals, Inc. [3,2,0]-Heterocyclische Verbindungen und Verfahren zu Ihrer Verwendung
US7579371B2 (en) 2004-04-30 2009-08-25 Nereus Pharmaceuticals, Inc. Methods of using [3.2.0] heterocyclic compounds and analogs thereof
EP1637529A1 (de) 2004-09-20 2006-03-22 4Sc Ag Neue Piperidin-4-yl-Thiazol-Carboxamid-Analog als Inhibitoren für die Vermehrung von T-Zellen und ihre Verwendung
EP1830838B1 (de) 2004-12-03 2012-10-03 Dana-Farber Cancer Institute, Inc. Zubereitungen und verfahren zur behandlung von neoplastischen erkrankungen
CN101460457B (zh) 2006-04-06 2012-07-18 尼瑞斯药品公司 Salinosporamide a及其类似物的全合成
US7824698B2 (en) 2007-02-02 2010-11-02 Nereus Pharmaceuticals, Inc. Lyophilized formulations of Salinosporamide A
US7442830B1 (en) 2007-08-06 2008-10-28 Millenium Pharmaceuticals, Inc. Proteasome inhibitors
US8394816B2 (en) 2007-12-07 2013-03-12 Irene Ghobrial Methods of using [3.2.0] heterocyclic compounds and analogs thereof in treating Waldenstrom's Macroglobulinemia
EP2262812A2 (de) 2008-03-07 2010-12-22 Nereus Pharmaceuticals, Inc. Totalsynthese von salinosporamid a und analoga davon
CA2723465A1 (en) 2008-05-12 2009-11-19 Nereus Pharmaceuticals, Inc. Proteasome inhibitors
RS54020B1 (en) 2008-06-17 2015-10-30 Millennium Pharmaceuticals Inc. ORGANOBOR ESTAR UNITS AND THEIR PHARMACEUTICAL COMPOSITIONS
AR075090A1 (es) 2008-09-29 2011-03-09 Millennium Pharm Inc Derivados de acido 1-amino-2-ciclobutiletilboronico inhibidores de proteosoma,utiles como agentes anticancerigenos, y composiciones farmaceuticas que los comprenden.
KR20130036228A (ko) 2010-03-31 2013-04-11 밀레니엄 파머슈티컬스 인코퍼레이티드 1-아미노-2-사이클로프로필에틸보론산의 유도체
DE102010039631A1 (de) * 2010-08-22 2012-02-23 Ulrich Schubert Pharmazeutische Zusammensetzungen mit immunmodulatorischen Eigenschaften
EP2986619A1 (de) 2013-04-16 2016-02-24 Cipla Limited Verfahren zur herstellung von bortezomib-mannitol-ester
CN106132970B (zh) 2014-02-03 2020-09-04 俄亥俄州创新基金会 硼酸酯和其药物制剂
UA121389C2 (uk) 2014-05-20 2020-05-25 Мілленніум Фармасьютікалз, Інк. Борвмісні інгібітори протеасом для застосування після первинної протиракової терапії
EP3270891A1 (de) 2015-03-17 2018-01-24 Leon-Nanodrugs GmbH Nanopartikel mit einer stabilisierten boronsäureverbindung

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6083903A (en) * 1994-10-28 2000-07-04 Leukosite, Inc. Boronic ester and acid compounds, synthesis and uses
US6335358B1 (en) * 1995-04-12 2002-01-01 President And Fellows Of Harvard College Lactacystin analogs

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9915183A1 *

Also Published As

Publication number Publication date
CA2304622A1 (en) 1999-04-01
JP2001517631A (ja) 2001-10-09
AU9580098A (en) 1999-04-12
WO1999015183A9 (en) 1999-07-29
WO1999015183A1 (en) 1999-04-01

Similar Documents

Publication Publication Date Title
EP1017398A1 (de) Proteasome inhibitoren, inhibitoren des ubiquitinweges oder verbindungen, die mit der durch den ubiquitinweg enstehenden nf-kb aktivierung interferieren zur behandlung von entzündlichen und autoimmunerkrankungen
US20010051654A1 (en) Treatment of inflammatory and autoimmune diseases
US5525623A (en) Compositions and methods for the treatment of immunomediated inflammatory disorders
US20060241130A1 (en) Anti-inflammatory compositions and uses thereof
Vargaftig et al. PAF-acether today—relevance for acute experimental anaphylaxis
US20130131052A1 (en) Amelioration Of The Development Of Cataracts And Other Ophthalmic Diseases
JPS61183253A (ja) 新規なペプチダ−ゼ阻害剤
FR2711989A1 (fr) Nouvelles isoquinoléines, leur préparation et leur utilisation comme médicaments.
JP5645918B2 (ja) リソソーム蓄積障害の予防および/または治療方法
EP3458448B1 (de) Fasn-hemmer zur behandlung der nicht-alkoholischen steatohepatitis
US9539221B2 (en) Method of treating airway diseases with β-adrenergic inverse agonists
KR100309616B1 (ko) 면역중재된염증질환의치료를위한조성물및방법
US7528175B2 (en) Method of treating airway diseases with beta-adrenergic inverse agonists
JP4951350B2 (ja) 神経変性障害の予防及び処置のための中性エンドペプチダーゼ(nep)及びヒト可溶性エンドペプチダーゼ(hsep)阻害剤
EP1931656B1 (de) Neue arzneimittel gegen demenz
NZ581748A (en) Treatment of allergic disease with immunomodulator compounds
MXPA00002938A (en) PROTEASOME INHIBITORS, UBIQUITIN PATHWAY INHIBITORS OR AGENTS THAT INTERFERE WITH THE ACTIVATION OF NF-&amp;kgr;B VIA THE UBIQUITIN PROTEASOME PATHWAY TO TREAT INFLAMMATORY AND AUTOIMMUNE DISEASES
EP1101492A1 (de) Arznei gegen allergische krankheiten
AU4653299A (en) Therapeutic agents for allergic diseases
JPH08505143A (ja) ジフルオロペンタペプチド誘導体抗炎症剤
US20020058706A1 (en) Method for treating asthma using pregabalin
US11608320B2 (en) Oxazolidinone hydroxamic acid derivatives
US20230242501A1 (en) Nerve damage preventing and nerve protecting compounds, preparation method thereof, pharmaceutical composition thereof, and their use
EP3858357A1 (de) Verwendung von azolverbindungen zur stimulierung des immunsystems und als inhbitoren von s-pla2gib
CA2288332A1 (en) (3r)-3-amino-4-carboxybutyraldehyde derivatives inhibiting the release of interleukin-1/beta

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20000418

AK Designated contracting states

Kind code of ref document: A1

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

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: MILLENNIUM PHARMACEUTICALS, INC.

17Q First examination report despatched

Effective date: 20020919

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

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

18D Application deemed to be withdrawn

Effective date: 20030401