US20110319330A1 - Methods For Treating Arthritis, Autoimmune Disease, and Ischemia-Reperfusion Injury - Google Patents

Methods For Treating Arthritis, Autoimmune Disease, and Ischemia-Reperfusion Injury Download PDF

Info

Publication number
US20110319330A1
US20110319330A1 US12/767,858 US76785810A US2011319330A1 US 20110319330 A1 US20110319330 A1 US 20110319330A1 US 76785810 A US76785810 A US 76785810A US 2011319330 A1 US2011319330 A1 US 2011319330A1
Authority
US
United States
Prior art keywords
antitrypsin
methylpropyl
apoptosis
carbonyl
oxadiazolyl
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.)
Abandoned
Application number
US12/767,858
Inventor
Leland Shapiro
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.)
Bio Holding Inc
Original Assignee
Bio Holding 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 Bio Holding Inc filed Critical Bio Holding Inc
Priority to US12/767,858 priority Critical patent/US20110319330A1/en
Publication of US20110319330A1 publication Critical patent/US20110319330A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/55Protease inhibitors
    • A61K38/57Protease inhibitors from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • 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
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to compositions and methods useful in the inhibition of apoptosis. Likewise, the present invention relates to methods of treating diseases associated with excessive or unregulated apoptosis.
  • necrotic cell death is characterized by cytoplasmic swelling, rupturing of cellular membranes, inflammation and disintegration of subcellular and nuclear components.
  • apoptosis is characterized by more organized changes in morphology and molecular structure. Apoptotic cells often condense and shrink, in part, by cytoplasmic membrane blebbing, a process of shedding small packets of membrane-bound cytoplasm.
  • the chromosomes of such cells condense around the nuclear periphery.
  • the chromosomes are degraded by specific nucleases that cleave DNA to produce regular-sized fragments.
  • Macrophages envelop and phagocytose apoptotic cells, thereby digesting and recycling the cellular components.
  • Changes in cell morphology during apoptosis are profound. Detection of the many morphological changes associated with apoptosis is detected using light microscopy or electron microscopy.
  • electron microscopy is useful for evaluating cells with a high nucleus to cytoplasm ratio and light microscopy is useful for immuno- and histochemistry.
  • the changes characteristic of apoptosis include decreased volume, compaction of cytoplasmic organelles, and increased cell density.
  • microvilli disappear, blebs of cytoplasm form at the cell surface, and the blebs dissociate from the cell to form apoptotic bodies.
  • TdT terminal deoxynucleotidyl transferase
  • TUNEL dUTP-biotin nick end labeling
  • ISEL in situ, end labeling
  • proteases can be inhibited by inhibitors including N-tosyl-L-phenylalanylchloromethyl ketone (TPCK) and N-tosyl-L-lysylchloromethyl ketone (TLCK).
  • TPCK N-tosyl-L-phenylalanylchloromethyl ketone
  • TLCK N-tosyl-L-lysylchloromethyl ketone
  • cysteine proteases related to interleukin-1- ⁇ converting enzyme have been identified as components of apoptotic signaling pathways.
  • the interleukin-1- ⁇ converting enzyme-like proteases are referred to as caspases and are identified and have been isolated by molecular cloning.
  • Granzyme B is a serine esterase that can activate several members of the caspase family.
  • Granzyme B may be a mediator of cytotoxic T lymphocyte induced apoptosis.
  • Granzyme B is known to cleave and initiate caspase 3, a likely component of its mode of action.
  • Granzyme B may also initiate nuclear events associated with cytotoxic T lymphocyte-induced apoptosis, consistent with observations that it is passively transported into the nucleus and bind to nuclear proteins.
  • One action of Granzyme B may be in the regulation of conversion of proCPP32 to CPP32.
  • CPP32 is itself a protease thought to cleave poly(ADP-ribose) polymerase (PARP) and may also activate prolamin protease resulting in activation of lamin protease. Cleavage of lamins and inactivation of the DNA repair enzyme PARP promote the development of apoptotic changes in the cell nucleus.
  • PARP poly(ADP-ribose) polymerase
  • TLCK serine protease inhibitor
  • TLCK inhibits apoptosis-associated proteolysis
  • TLCK is known to inhibit cysteine proteases in addition to serine proteases, and has recently been shown to inhibit a member of the interleukin-1 ⁇ converting enzyme family.
  • the effect of TLCK on apoptosis is likely not mediated by an effect as a serine protease inhibitor, given the more established role of cysteine proteases in apoptosis.
  • the lamins are nuclear intermediate filament proteins that form a fibrous layer between the inner nuclear membrane and the chromatin.
  • the resulting lamina is thought to play a role in maintaining nuclear shape and in mediating chromatin-nuclear membrane interactions.
  • the apoptosis-associated changes in nuclear shape might require lamin reorganization.
  • Another polypeptide that is cleaved during apoptosis is poly (ADP-ribose) polymerase (PARP).
  • PARP is an abundant nuclear enzyme that catalyzes the conversion of the dinucleotide NAD + to nicotinamide and protein-linked chains of ADP-ribose. Yet, the detailed role of PARP in the process of apoptosis is unclear. Studies have suggested that inhibitors of PARP delay apoptosis and yet other studies have suggested that inhibition of PARP increases the fragmentation of DNA during apoptosis. It is clear, however, that PARP is proteolytically degraded late in apoptosis.
  • U1 ribonuclear protein U1-70k
  • U1-70k U1 ribonuclear protein
  • PITSLRE ⁇ 1 protein kinase adenomatous polyposis coli (APC) protein
  • APC adenomatous polyposis coli
  • retinoblastoma gene product a gene product of apoptosis
  • nuclear matrix proteins a protein that influences the expression of apoptosis.
  • Cleavage of fodrin an abundant membrane associated cytoskeletal protein, has been detected during apoptosis in a variety of cell lines.
  • PITSLRE ⁇ 1 protein kinase a member of the P34 cdc2 gene family has been shown to induce mitotic delay in CHO cells. Members of this family appear to be cleaved during apoptosis.
  • PITSLRE ⁇ 1 kinase is proteolytically cleaved during FAS— or steroid-induced apoptosis in T-cells.
  • Another major group of protease targets is the caspases, themselves proteases, or precursor forms of caspases.
  • Yet another group of proteins which may well be downstream effectors of caspase-mediated apoptosis include the protein kinases PKC ⁇ , PKC ⁇ , MEKK1, the sterol regulatory element binding proteins 1 and 2, and the DNA fragmentation factor (DFF).
  • Increased levels or apparent induction of apoptosis is associated with a number of diseases including cancer, autoimmune diseases including rheumatoid arthritis, neurodegenerative diseases, myocardial infarction, stroke, sepsis, ischemia-reperfusion injury, toxin induced liver injury, and AIDS (see Kidd, V. J., Annu Rev Physiol, 1998, 60, 533; List, P. J. M., et al., Arterioscler Thromb Vasc Biol 1999, 19, 14; Jabs, T., Biochem Pharmacol 1999 57, 231; Deigner, H. P., et al. Curr Med Chem 1999, 6, 399).
  • the apoptosis appears to be mediated by oxygen free radicals [O.] which have been implicated in various disorders including atherosclerosis, diabetes, sepsis, Alzheimer's disease, arthritis, muscular dystrophy, cancer, Downs syndrome, multiple sclerosis, HIV infection and other inflammatory diseases (Morel, J. B. and Dangle, J. L., Cell Death Differ 1997, 4, 671; Beal, M. F., Curr Opin Neurobiol 1996, 6, 661).
  • the present invention is directed to a method of treating an animal or a patient suffering from a disease characterized by excessive apoptosis.
  • the method of the invention comprises administering a therapeutically effective amount of at least one serine protease inhibitor and subsequently monitoring a decrease in apoptosis.
  • the animal is a human.
  • the agent is ⁇ 1 -antitrypsin (ATT) or an ⁇ 1 -antitrypsin-like agent.
  • peptides of interest are homologous and analogous peptides. All homologues are natural peptides which have sequence homology, analogs will be peptidyl derivatives, e.g., aldehyde or ketone derivatives of such peptides.
  • agents with ⁇ 1 -antitrypsin-like activity are also envisioned.
  • peptide derivatives of ⁇ 1 -antitrypsin compounds like oxydiazole, thiadiazole, CE-2072, UT-77, and triazole peptoids are preferred.
  • the ⁇ 1 -antitrypsin-like agent includes, but is not limited to, small organic molecules including naturally occurring, synthetic, and biosynthetic molecules, small inorganic molecules including naturally-occurring and synthetic molecules, natural products including those produced by plants and fungi, peptides, variants of ⁇ 1 -antitrypsin, chemically modified peptides, and proteins.
  • an individual with risk for a pathological disease or condition that is precipitated at least in part by excessive apoptosis can be treated to prevent the onset of acute disease with a prophylactic treatment of an agent exhibiting ⁇ 1 -antitrypsin or ⁇ 1 -antitrypsin-like activity.
  • a still further embodiment of the invention directed to a method of inhibiting apoptosis comprises allowing a serine protease inhibitor to bind to a protease and measuring the decrease in apoptosis.
  • Another embodiment of the invention is directed to a method of inhibiting apoptosis comprising allowing a serine protease inhibitor to bind to a cell surface receptor and measuring the decrease in apoptosis.
  • a yet still further embodiment of the invention is directed to use of oxidation-resistant and free-radical resistant inhibitors of serine proteases.
  • the oxidation-sensitive Met 358 in ⁇ 1 -antitrypsin can, by genetic engineering, be replaced by Val 358 - ⁇ 1 -antitrypsin, which results in a molecule termed Val 358 - ⁇ 1 -antitrypsin.
  • Val 358 - ⁇ 1 -antitrypsin is a more potent inhibitor of neutrophil elastase than is Met 358 - ⁇ 1 -antitrypsin possibly because of the stability of Val 358 - ⁇ 1 -antitrypsin to the neutrophil oxidative burst.
  • the Met at position 358 is replaced with any hydrophobic or neutral oxidation-resistant amino acid residue, including: alanine, asparagine, ⁇ -amino butyric acid, anthranilic acid, ⁇ -cyanoalanine, ⁇ -(3,4-dihyroxyphenyl) alanine, 3,5-diiodotyrosine, glutamine, glycine, homoserine, 3-hydroxyanthranilic acid, 5-hydroxy-indole-3-acetic acid, 3-hydroxykynurenine, hydroxyproline, 5-hydroxy-tryptophan, indoleacetic acid, 3-iodotyrosine, isoleucine, allo-isoleucine, leucine, leucylglycine, norleucine, norvaline, phenylalanine, proline, prolylglycine, serine, threonine, allo-threonine, throxine, 3,5,3′-tri-
  • FIG. 2 illustrates the effect of ⁇ 1 -antitrypsin and the peptoid CE-2072 on apoptosis in RCG Neuron (rat cerebral granule) cells, also termed RCGC.
  • the current invention teaches methodologies and agents for treating animals and patients that suffer from a disease involving excessive apoptosis.
  • the methods involve administration of therapeutically effective amounts of at least one serine protease inhibitor and testing for changes in apoptosis by any of several means known in the art.
  • the serine proteases that are inhibited by the agent of the invention include trypsin, elastase, cathepsin G, tryptase TL-2, Factor Xa and proteinase-3.
  • the methods further involve inhibition of oxygen free radicals and inhibition of oxygen free radical formation by serine protease inhibitors.
  • the method further includes a pharmaceutically acceptable carrier, any of which are known in the art.
  • Serine protease inhibitors include ⁇ 1 -antitrypsin, or ⁇ 1 -antitrypsin-like agents. In the latter group are included the oxydiazole, thiazole, triazole peptoids, or some combination of these agents.
  • the serine protease inhibitor is optionally derivatized chemically by esterification, acetylation or amidation.
  • a preferred embodiment of the invention is directed toward the treatment of myocardial infarction.
  • Another preferred embodiment of the invention is directed toward treatment of stroke, also known as brain ischemia or cerebrovascular accident.
  • the therapeutically effective amounts of the serine protease inhibitors are sufficient to bring the concentration of the added agent in the biological fluid of the individual to between about 10 pM and 2 mM.
  • the effective concentrations correspond to between about 5 nanograms per milliliter to about 10 milligrams per milliliter of the biological fluid of the individual.
  • the biological fluid of the individual is calculated from the total body weight of the individual or, in diseases that are localized to specific body compartments, from the volume of the compartment.
  • Biological fluid can include, but is not limited to, blood, plasma, serum, lymph, tears, saliva, cerebrospinal fluid, or combinations thereof.
  • the therapeutically effective amount is sufficient to bring the concentration in the biological fluid to between 0.5 ⁇ M and 200 ⁇ M, preferably between 5 ⁇ M and 200 ⁇ M, most preferably about 100 ⁇ M.
  • the agent is advantageously administered according to the weight of the subject.
  • Administration of the therapeutically effective amount of serine protease inhibitor can be in a bolus, for example, of about 0.001 to 7 g of ⁇ 1 -antitrypsin-like agent or about 1 to 70 g of ⁇ 1 -antitrypsin, per kg of body weight of the subject.
  • Preferred amounts are about 0.01 g/kg body weight of oxydiazole, thiazole, or triazole peptoids, and about 1 g/kg body weight of natural or variant ⁇ 1 -antitrypsin.
  • the administration of the agent in the invention can be performed parenterally, orally, vaginally, nasally, buccally, intravenously, intramuscularly, subcutaneously, rectally, intrathecally, epidurally, transdermally, intracerebroventricularly, or combinations thereof.
  • the agent is administered continuously or intermittently by osmotic pump or by implanted osmotic pump, including those of the Alza Corporation. It is a further embodiment of the invention that the therapeutically effective amount of the serine protease inhibitor is administered between about once daily to about once hourly. In a more preferred embodiment of the invention, the serine to protease inhibitor is administered twice per day. It is a further embodiment of the invention that the monitoring of changes in apoptosis be performed on tissue obtained from an animal or patient. Any of several methods for monitoring apoptosis, well known in the art, are suitable.
  • a further method of the invention is directed to encouraging the binding of a serine protease inhibitor to a protease and observing a change in apoptosis.
  • the serine protease inhibitor is ⁇ 1 -antitrypsin or ⁇ 1 -antitrypsin-like agent.
  • the ⁇ 1 -antitrypsin-like agent is also a substituted oxydiazole, substituted thiadiazole, substituted triazole peptoids, or any combination of these agents.
  • the present invention is not limited by the mechanism of action of ⁇ 1 -antitrypsin inhibitors in decreasing apoptosis.
  • the apoptosis may be mediated by tumor necrosis factor, by anti-Fas or by any other mechanism.
  • apoptosis not mediated by tumor necrosis factor is inhibited by the ⁇ 1 -antitrypsin-like agents of the invention.
  • the agents of the invention are effective to inhibit apoptosis in a plurality of organs including, but not limited to brain, heart, spinal cord, peripheral nerves, skin, stomach, liver, pancreas, gut, ovaries, testis, and endocrine glands.
  • 5,874,585 discloses substituted heterocyclic compounds useful as inhibitors of serine proteases including: (benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(3-trifluoromethylbenzyl)-1,2,4-oxadiazolyl)carbonyl)-2-(S)-methylpropyl]-L-prolinamide benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(2-phenylethyl)-1,2,4-oxadiazolyl)carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(2-methoxybenzyl)-1,2,4-oxadiazolyl)carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (benzyloxycarbonyl)-L-valyl
  • U.S. Pat. No. 5,216,022 teaches other small molecules useful for the practice of this invention, including: Benzyloxycarbonyl-L-valyl-N-[1-(2-[5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide (also known as CE-2072), Benzyloxycarbonyl-L-valyl-N-[1-(2-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; Benzyloxycarbonyl-L-valyl-N-[1-(2-(5-(methyl)-1,3,4-oxadiazoly]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; Benzyloxycarbonyl)-L-valyl-N-[1-(2-(
  • Yet another embodiment of the invention is directed toward the inhibition of apoptosis resulting from the interaction between a serine protease inhibitor and a cell surface receptor and resulting in a measurable decrease in apoptosis.
  • the serine protease inhibitor of this embodiment is an ⁇ 1 -antitrypsin or an ⁇ 1 -antitrypsin-like agent.
  • the ⁇ 1 -antitrypsin-like agents includes substituted oxydiazoles, substituted thiadiazole, substituted triazole peptoids, or combinations of these agents.
  • the substituted oxydiazole, thiadiazole, and triazole peptoids are synthesized de novo or derivatized from existing compounds.
  • Specific diseases or disorders for which the therapeutic methods of the invention are beneficial include wasting diseases of various types.
  • the diseases include cancer, neurodegenerative diseases, myocardial infarction, and stroke.
  • the cancers include bladder, breast, kidney, leukemia, lung, myoloma, liposarcoma, lymphoma, tongue, prostate, and uterine cancers.
  • the method of the invention is also applied to Alzheimer's disease, arthritis, muscular dystrophy, Downs Syndrome, sepsis, HIV infection, multiple sclerosis, arteriosclerosis, diabetes, and arthritis. In fact, the invention is applied to any disease associated with elevated levels of apoptosis.
  • the agents are delivered by any of a variety of routes including: by injection (e.g., subcutaneous, intramuscular, intravenous, intra-arterial, and intraperitoneal), by continuous intravenous infusion, transdermally, orally (e.g., tablet, pill, liquid medicine), by implanted osmotic pumps (e.g., ALZA Corp.), by suppository or aerosol spray.
  • routes including: by injection (e.g., subcutaneous, intramuscular, intravenous, intra-arterial, and intraperitoneal), by continuous intravenous infusion, transdermally, orally (e.g., tablet, pill, liquid medicine), by implanted osmotic pumps (e.g., ALZA Corp.), by suppository or aerosol spray.
  • peptides are preferably prepared using recombinant DNA techniques, synthetic techniques, or chemical derivatization of biologically or chemically synthesized peptides.
  • the compounds of the present invention are used as therapeutic agents in the treatment of a physiological, or especially, pathological, condition caused in whole or part by uncontrolled serine protease and apoptosis activity.
  • the peptides or peptoids can be administered as free peptides, free peptoids, or pharmaceutically acceptable salts thereof.
  • the terms used herein conform to those in Budavari, S. (Ed.), “The Merck Index, An Encyclopedia of Chemicals, Drugs, and Biologicals,” Merck Company, Inc., twelfth edition.
  • pharmaceutically acceptable salt refers to those acid addition salts or methyl complexes of the peptides which do not significantly or adversely affect the therapeutic properties including efficacy and toxicity, of the peptides and peptoids.
  • the peptides and peptoids are administered to individuals as a pharmaceutical composition which, in most cases, will comprise the peptide, peptoid, and/or pharmaceutical salts thereof with a phainiaceuticaily acceptable carrier.
  • pharmaceutically acceptable carrier refers to those solid and liquid carriers, which do not significantly or adversely affect the therapeutic properties of the peptides.
  • compositions containing peptides and/or peptoids of the present invention are administered to individuals, particularly humans, either intravenously, subcutaneously, intramuscularly, intranasally, orally, topically, transdermally, parenterally, gastrointestinally, transbronchially, and transalveolarly.
  • Topical administration is accomplished by a topically applied cream, gel, rinse, etc. containing therapeutically effective amounts of inhibitors of serine proteases.
  • Transdermal administration is accomplished by administration of a cream, rinse, gel, etc. capable of allowing the inhibitors of serine proteases to penetrate the skin and enter the blood stream.
  • Parenteral routes of administration include, but are not limited to, direct injection such as intravenous, intramuscular, intraperitoneal, or subcutaneous injection.
  • Gastrointestinal routes of administration include, but are not limited to, ingestion and rectal.
  • Transbroncheal and transalveolar routes of administration include, but are not limited to, inhalation, either via the mouth or intranasally, and direct injection into an area, such as through a tracheotomy, endotracheal tube, or aspirated through a respiratory mist.
  • osmotic pumps are used for administration. The necessary dosage will vary with the particular condition being treated, method of administration, and rate of clearance of the molecule from the body.
  • AAT vehicle is phosphate-buffered saline, or optionally, any pharmaceutically acceptable carrier.
  • the dosage of ⁇ 1 -antitrypsin administered is varied between the amounts necessary to produce a concentration of 10 ⁇ M and 250 ⁇ M in the blood. In general, a concentration of 5 mg/ml of ⁇ 1 -antitrypsin is equivalent to about 100 ⁇ M. In yet other experiments the frequency of administration is varied from once per day to four times per day. Likewise, antielastase and antiproteinase are used.
  • TNF Tumor Necrosis Factor
  • one preferred embodiment of the process is the co-administration of ⁇ 1 -antitrypsin and a free radical scavenger, such as glutathione (1 mg/kg body weight).
  • oxidation-resistant ⁇ 1 -antitrypsin variants are used to avoid inactivation by excess free radicals.
  • synthetic ⁇ 1 -antitrypsin or recombinant ⁇ 1 -antitrypsin produced with alternative and oxidation-resistant amino acid sequences are embodiments of the invention.
  • RCG neuronal cells are seeded into cell culture dishes in 400 ⁇ l cell culture medium (Eagle's basal medium, BME) containing 10% (v/v) FBS. At day two the now conditioned medium is removed and the cells are treated for 10 hours as follows:
  • the medium is replaced with 4% (w/v) paraformaldehyde, incubated for 15 minutes at room temperature, and the cells stained with Hoechst dye 33258 (8 ⁇ g/ml) for 15 minutes at room temperature.
  • the apoptosis in the cells is evaluated using a fluorescence microscope by an evaluator blinded to the method of treatment. The results are shown in FIG. 2 .
  • agent CE-2072 a synthetic inhibitor of serine protease.
  • the latter is formally known as benzyloxcarbonyl-L-valyl-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-Methylpropyl]-L-prolinamide.
  • Human donor organs including kidneys, are subject to ischemia during transport, which can last up to several hours.
  • Biopsies (3 mm) are removed from the top medial surface of donor kidneys undergoing transport prior to implantation, and grouped by time after removal from the donor: 1-2 hours, 2-4 hours, and greater than 4 hours.
  • Donor kidneys transplanted within one hour serve as the first control and the contralateral kidney serves as the second control.
  • Half of the donor kidneys are treated with ⁇ 1 -antitrypsin (10 mg/ml fluid) upon removal from the donor to inhibit apoptosis.
  • Val 358 -antitrypsin and Ile 358 -antitrypsin are produced from the appropriate nucleotide sequences by methods well known in the art, including construction of a plasmid, transfection of the host E. coli , selection of transfected colonies, expansion of the culture, and isolation and purification of the mutant gene product.
  • Amounts of the recombinant agents effective in inhibiting apoptosis, excessive clotting, neutrophil extravasation, ischemia-reperfusion injury, or myocardial damage are applied in an experimental model of myocardial infarction (see Example 6.1, supra). Effective amounts are between 0.03 and 7 g/kg body weight, for example, about 0.5 g/kg.
  • the amount of variant antitrypsin is measured in the blood or other biological fluid. In those tests sufficient variant antitrypsin is administered to provide a concentration of about 1 ⁇ M to about 100 ⁇ M in the blood or biological fluid.
  • ⁇ 1 -antitrypsin Effect of ⁇ 1 -antitrypsin on apoptosis
  • Primary rat brain granule cells are pretreated for one hour in the absence or presence of ⁇ 1 -antitrypsin (3.0 mg/ml), followed by replacement of the cell culture medium with either control medium containing 10% (vol/vol) fetal bovine serum, medium devoid of fetal bovine serum, or medium devoid of fetal bovine serum but containing ⁇ 1 -antitrypsin.
  • control medium 10% (vol/vol) fetal bovine serum, medium devoid of fetal bovine serum, or medium devoid of fetal bovine serum but containing ⁇ 1 -antitrypsin.
  • ⁇ 1 -Antitrypsin completely reverses the apoptosis associated with serum depletion, which results in cell death.

Abstract

The instant invention provides a method of treating an animal suffering a disease characterized by excessive apoptosis by administering a therapeutically effective amount of at least one serine protease inhibitor and thereafter monitoring a decrease in apoptosis. The inhibitor of the invention includes α1-antitrypsin or an α1-antitrypsin-like agent, including, but not limited to oxidation-resistant variants of α1-antitrypsin, and peptoids with antitrypsin activity. The diseases treatable by the invention include cancer, autoimmune disease, sepsis neurodegenerative disease, myocardial infarction, stroke, ischemia-reperfusion injury, toxin induced liver injury and AIDS. The method of the invention is also suitable for the prevention or amelioration of diseases characterized by excessive apoptosis.

Description

  • The present application claims the priority of U.S. application Ser. No. 60/123,167, filed Mar. 5, 1999, the entire disclosure of which is incorporated by reference herein.
  • 1. FIELD OF THE INVENTION
  • The present invention relates to compositions and methods useful in the inhibition of apoptosis. Likewise, the present invention relates to methods of treating diseases associated with excessive or unregulated apoptosis.
  • 2. BACKGROUND OF THE INVENTION
  • Normal development, growth, and function of multi-cellular organisms require control both of processes that produce cells and of those that destroy cells. Mitosis, or cell proliferation, is highly regulated except in specific states termed cell proliferative diseases. There also exist processes for destruction of cells. Cells in multi-cellular organisms die by two distinct mechanisms. One method, termed necrotic cell death, is characterized by cytoplasmic swelling, rupturing of cellular membranes, inflammation and disintegration of subcellular and nuclear components. The other method, apoptosis, by contrast, is characterized by more organized changes in morphology and molecular structure. Apoptotic cells often condense and shrink, in part, by cytoplasmic membrane blebbing, a process of shedding small packets of membrane-bound cytoplasm. The chromosomes of such cells condense around the nuclear periphery. Generally, in apoptotic cells the chromosomes are degraded by specific nucleases that cleave DNA to produce regular-sized fragments. Importantly, there is a requirement for new mRNA and protein expression during the early stages of some forms of apoptosis, indicating that it is an active process. Macrophages envelop and phagocytose apoptotic cells, thereby digesting and recycling the cellular components.
  • Changes in cell morphology during apoptosis are profound. Detection of the many morphological changes associated with apoptosis is detected using light microscopy or electron microscopy. In particular, electron microscopy is useful for evaluating cells with a high nucleus to cytoplasm ratio and light microscopy is useful for immuno- and histochemistry. The changes characteristic of apoptosis include decreased volume, compaction of cytoplasmic organelles, and increased cell density. In addition, microvilli disappear, blebs of cytoplasm form at the cell surface, and the blebs dissociate from the cell to form apoptotic bodies. Other techniques are useful in the analysis of apoptosis including confocal, laser, and scanning microscopy, fluorescent DNA dye binding, and molecular techniques. The molecular techniques permit detection of apoptosis in formalin-fixed and embedded tissue, including terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-biotin nick end labeling (TUNEL) and in situ, end labeling (ISEL).
  • Protease Involvement
  • The progression of apoptosis requires the coordinated action of specific proteases. The proteases can be inhibited by inhibitors including N-tosyl-L-phenylalanylchloromethyl ketone (TPCK) and N-tosyl-L-lysylchloromethyl ketone (TLCK). Furthermore, at least 10 cysteine proteases related to interleukin-1-β converting enzyme have been identified as components of apoptotic signaling pathways. The interleukin-1-β converting enzyme-like proteases are referred to as caspases and are identified and have been isolated by molecular cloning.
  • In addition, there are other proteases involved in apoptosis including the granzymes and cathepsin. Granzyme B is a serine esterase that can activate several members of the caspase family. Granzyme B may be a mediator of cytotoxic T lymphocyte induced apoptosis. Granzyme B is known to cleave and initiate caspase 3, a likely component of its mode of action. Granzyme B may also initiate nuclear events associated with cytotoxic T lymphocyte-induced apoptosis, consistent with observations that it is passively transported into the nucleus and bind to nuclear proteins. One action of Granzyme B may be in the regulation of conversion of proCPP32 to CPP32. CPP32 is itself a protease thought to cleave poly(ADP-ribose) polymerase (PARP) and may also activate prolamin protease resulting in activation of lamin protease. Cleavage of lamins and inactivation of the DNA repair enzyme PARP promote the development of apoptotic changes in the cell nucleus.
  • Serine Proteases
  • In contrast to cysteine proteases, the role of serine proteases in apoptosis is controversial. For a general discussion, see Kaufmann, S. Cancer Res 1993, 53, 3976. For example, it is known that the serine protease inhibitor TLCK inhibits apoptosis-associated proteolysis. However, TLCK is known to inhibit cysteine proteases in addition to serine proteases, and has recently been shown to inhibit a member of the interleukin-1β converting enzyme family. Thus, the effect of TLCK on apoptosis is likely not mediated by an effect as a serine protease inhibitor, given the more established role of cysteine proteases in apoptosis.
  • Cellular Protease Targets
  • Multiple polypeptide species must be modified to produce the wide range of morphological manifestations that characterize apoptosis. For example, the lamins are nuclear intermediate filament proteins that form a fibrous layer between the inner nuclear membrane and the chromatin. The resulting lamina is thought to play a role in maintaining nuclear shape and in mediating chromatin-nuclear membrane interactions. Thus, the apoptosis-associated changes in nuclear shape might require lamin reorganization. Another polypeptide that is cleaved during apoptosis is poly (ADP-ribose) polymerase (PARP). PARP is an abundant nuclear enzyme that catalyzes the conversion of the dinucleotide NAD+ to nicotinamide and protein-linked chains of ADP-ribose. Yet, the detailed role of PARP in the process of apoptosis is unclear. Studies have suggested that inhibitors of PARP delay apoptosis and yet other studies have suggested that inhibition of PARP increases the fragmentation of DNA during apoptosis. It is clear, however, that PARP is proteolytically degraded late in apoptosis.
  • Another proteolytic enzyme target during apoptosis is the U1 ribonuclear protein (U1-70k), a molecule required for splicing of precursor mRNA that is itself cleaved to an inactive 40 kDa fragment during apoptosis. The cleavage of this polypeptide would result in cessation of RNA processing.
  • Other substrates for protease activity during apoptosis include fodrin, the PITSLREβ1 protein kinase, the adenomatous polyposis coli (APC) protein, the retinoblastoma gene product, terminin, and nuclear matrix proteins. Cleavage of fodrin, an abundant membrane associated cytoskeletal protein, has been detected during apoptosis in a variety of cell lines. PITSLREβ1 protein kinase, a member of the P34cdc2 gene family has been shown to induce mitotic delay in CHO cells. Members of this family appear to be cleaved during apoptosis. For example, recent studies indicate that PITSLREβ1 kinase is proteolytically cleaved during FAS— or steroid-induced apoptosis in T-cells. Another major group of protease targets is the caspases, themselves proteases, or precursor forms of caspases. Yet another group of proteins which may well be downstream effectors of caspase-mediated apoptosis, include the protein kinases PKCε, PKCθ, MEKK1, the sterol regulatory element binding proteins 1 and 2, and the DNA fragmentation factor (DFF).
  • Diseases Associated with Apoptosis
  • Increased levels or apparent induction of apoptosis is associated with a number of diseases including cancer, autoimmune diseases including rheumatoid arthritis, neurodegenerative diseases, myocardial infarction, stroke, sepsis, ischemia-reperfusion injury, toxin induced liver injury, and AIDS (see Kidd, V. J., Annu Rev Physiol, 1998, 60, 533; List, P. J. M., et al., Arterioscler Thromb Vasc Biol 1999, 19, 14; Jabs, T., Biochem Pharmacol 1999 57, 231; Deigner, H. P., et al. Curr Med Chem 1999, 6, 399). The apoptosis appears to be mediated by oxygen free radicals [O.] which have been implicated in various disorders including atherosclerosis, diabetes, sepsis, Alzheimer's disease, arthritis, muscular dystrophy, cancer, Downs syndrome, multiple sclerosis, HIV infection and other inflammatory diseases (Morel, J. B. and Dangle, J. L., Cell Death Differ 1997, 4, 671; Beal, M. F., Curr Opin Neurobiol 1996, 6, 661).
  • 3. SUMMARY OF THE INVENTION
  • The present invention is directed to a method of treating an animal or a patient suffering from a disease characterized by excessive apoptosis. The method of the invention comprises administering a therapeutically effective amount of at least one serine protease inhibitor and subsequently monitoring a decrease in apoptosis.
  • In a preferred embodiment, the animal is a human. In another preferred embodiment, the agent is α1-antitrypsin (ATT) or an α1-antitrypsin-like agent. In addition, peptides of interest are homologous and analogous peptides. All homologues are natural peptides which have sequence homology, analogs will be peptidyl derivatives, e.g., aldehyde or ketone derivatives of such peptides. Furthermore, agents with α1-antitrypsin-like activity are also envisioned. In this regard, peptide derivatives of α1-antitrypsin, compounds like oxydiazole, thiadiazole, CE-2072, UT-77, and triazole peptoids are preferred. The α1-antitrypsin-like agent includes, but is not limited to, small organic molecules including naturally occurring, synthetic, and biosynthetic molecules, small inorganic molecules including naturally-occurring and synthetic molecules, natural products including those produced by plants and fungi, peptides, variants of α1-antitrypsin, chemically modified peptides, and proteins. It is a further embodiment of this invention that an individual with risk for a pathological disease or condition that is precipitated at least in part by excessive apoptosis, can be treated to prevent the onset of acute disease with a prophylactic treatment of an agent exhibiting α1-antitrypsin or α1-antitrypsin-like activity.
  • A further embodiment of the invention envisions a method for inhibiting apoptosis in an in vitro mammalian cell culture, an ex vivo mammalian tissue culture, or a mammalian organ, comprising providing to a cell culture, tissue culture, or organ, an amount of a serine protease inhibitor sufficient to inhibit apoptosis in the cell culture, tissue culture, or organ. In the aforementioned embodiment, a measured amount of apoptosis is indicative of expression or activity of apoptosis.
  • A still further embodiment of the invention directed to a method of inhibiting apoptosis comprises allowing a serine protease inhibitor to bind to a protease and measuring the decrease in apoptosis. Another embodiment of the invention is directed to a method of inhibiting apoptosis comprising allowing a serine protease inhibitor to bind to a cell surface receptor and measuring the decrease in apoptosis.
  • A yet still further embodiment of the invention is directed to use of oxidation-resistant and free-radical resistant inhibitors of serine proteases. In this regard, the oxidation-sensitive Met358 in α1-antitrypsin can, by genetic engineering, be replaced by Val3581-antitrypsin, which results in a molecule termed Val3581-antitrypsin. Val3581-antitrypsin is a more potent inhibitor of neutrophil elastase than is Met3581-antitrypsin possibly because of the stability of Val3581-antitrypsin to the neutrophil oxidative burst. The Met at position 358 is replaced with any hydrophobic or neutral oxidation-resistant amino acid residue, including: alanine, asparagine, α-amino butyric acid, anthranilic acid, β-cyanoalanine, β-(3,4-dihyroxyphenyl) alanine, 3,5-diiodotyrosine, glutamine, glycine, homoserine, 3-hydroxyanthranilic acid, 5-hydroxy-indole-3-acetic acid, 3-hydroxykynurenine, hydroxyproline, 5-hydroxy-tryptophan, indoleacetic acid, 3-iodotyrosine, isoleucine, allo-isoleucine, leucine, leucylglycine, norleucine, norvaline, phenylalanine, proline, prolylglycine, serine, threonine, allo-threonine, throxine, 3,5,3′-tri-iodo-thyronine, tryptophan, and tyrosine. The amino acid substitutions are effected by genetic engineering, chemical modification, or a combination thereof.
  • 4. BRIEF DESCRIPTION OF THE DRAWING
  • FIG. 1 illustrates the effect of α1-antitrypsin on apoptosis in primary rat brain cerebral granule cells.
  • FIG. 2 illustrates the effect of α1-antitrypsin and the peptoid CE-2072 on apoptosis in RCG Neuron (rat cerebral granule) cells, also termed RCGC.
  • 5. DETAILED DESCRIPTION OF THE INVENTION
  • 5.1 Standard Methods
  • In accordance with the present invention there can be employed conventional molecular biology, microbiology, and recombinant DNA techniques within the skill of the art. Such techniques are explained fully in the literature. See, e.g., Sambrook, Fritsch & Maniatis, Molecular Cloning: A Laboratory Manual, Second Edition 1989, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.; Animal Cell Culture, R. I. Freshney, ed., 1986).
  • 5.2 Serine Protease Inhibitors
  • The current invention teaches methodologies and agents for treating animals and patients that suffer from a disease involving excessive apoptosis. The methods involve administration of therapeutically effective amounts of at least one serine protease inhibitor and testing for changes in apoptosis by any of several means known in the art. The serine proteases that are inhibited by the agent of the invention include trypsin, elastase, cathepsin G, tryptase TL-2, Factor Xa and proteinase-3. The methods further involve inhibition of oxygen free radicals and inhibition of oxygen free radical formation by serine protease inhibitors. The method further includes a pharmaceutically acceptable carrier, any of which are known in the art. Serine protease inhibitors include α1-antitrypsin, or α1-antitrypsin-like agents. In the latter group are included the oxydiazole, thiazole, triazole peptoids, or some combination of these agents. The serine protease inhibitor is optionally derivatized chemically by esterification, acetylation or amidation.
  • There are numerous diseases that are characterized by excessive apoptosis. Among these diseases are cancer, autoimmune diseases, neurodegenerative diseases, myocardial infarction, stroke, ischemia-reperfusion injury, toxin-induced liver injury, sepsis and AIDS.
  • A preferred embodiment of the invention is directed toward the treatment of myocardial infarction. Another preferred embodiment of the invention is directed toward treatment of stroke, also known as brain ischemia or cerebrovascular accident. The therapeutically effective amounts of the serine protease inhibitors are sufficient to bring the concentration of the added agent in the biological fluid of the individual to between about 10 pM and 2 mM. For α1-antitrypsin the effective concentrations correspond to between about 5 nanograms per milliliter to about 10 milligrams per milliliter of the biological fluid of the individual. The biological fluid of the individual is calculated from the total body weight of the individual or, in diseases that are localized to specific body compartments, from the volume of the compartment. Biological fluid can include, but is not limited to, blood, plasma, serum, lymph, tears, saliva, cerebrospinal fluid, or combinations thereof.
  • In a preferred embodiment of the invention, the therapeutically effective amount is sufficient to bring the concentration in the biological fluid to between 0.5 μM and 200 μM, preferably between 5 μM and 200 μM, most preferably about 100 μM. The agent is advantageously administered according to the weight of the subject. Administration of the therapeutically effective amount of serine protease inhibitor can be in a bolus, for example, of about 0.001 to 7 g of α1-antitrypsin-like agent or about 1 to 70 g of α1-antitrypsin, per kg of body weight of the subject. Preferred amounts are about 0.01 g/kg body weight of oxydiazole, thiazole, or triazole peptoids, and about 1 g/kg body weight of natural or variant α1-antitrypsin.
  • The administration of the agent in the invention can be performed parenterally, orally, vaginally, nasally, buccally, intravenously, intramuscularly, subcutaneously, rectally, intrathecally, epidurally, transdermally, intracerebroventricularly, or combinations thereof.
  • In another embodiment of the invention, the agent is administered continuously or intermittently by osmotic pump or by implanted osmotic pump, including those of the Alza Corporation. It is a further embodiment of the invention that the therapeutically effective amount of the serine protease inhibitor is administered between about once daily to about once hourly. In a more preferred embodiment of the invention, the serine to protease inhibitor is administered twice per day. It is a further embodiment of the invention that the monitoring of changes in apoptosis be performed on tissue obtained from an animal or patient. Any of several methods for monitoring apoptosis, well known in the art, are suitable.
  • A further method of the invention is directed to encouraging the binding of a serine protease inhibitor to a protease and observing a change in apoptosis. In this embodiment, the serine protease inhibitor is α1-antitrypsin or α1-antitrypsin-like agent. The α1-antitrypsin-like agent is also a substituted oxydiazole, substituted thiadiazole, substituted triazole peptoids, or any combination of these agents.
  • Apoptosis is associated with free radical production, including oxygen free radicals. Free radicals are known to inactivate natural α1-antitrypsin. Therefore, it is desirable to supplement α1-antitrypsin in blood with sufficient α1-antitrypsin-like activity which is not inactivated by free radicals. Alternatively, a mutant α1-antitrypsin resistant to inactivation by free radicals, or administration of a synthetic molecule that is not inactivated by free radicals, is contemplated. Also, co-administration of a free radical scavenger or inhibitor is contemplated.
  • The present invention is not limited by the mechanism of action of α1-antitrypsin inhibitors in decreasing apoptosis. Thus the apoptosis may be mediated by tumor necrosis factor, by anti-Fas or by any other mechanism. In a particular embodiment of the invention apoptosis not mediated by tumor necrosis factor is inhibited by the α1-antitrypsin-like agents of the invention. Moreover, the agents of the invention are effective to inhibit apoptosis in a plurality of organs including, but not limited to brain, heart, spinal cord, peripheral nerves, skin, stomach, liver, pancreas, gut, ovaries, testis, and endocrine glands.
  • It is to be understood that the present invention is not limited to the examples described herein, and other serine protease inhibitors known in the art are used within the limitations of the invention. For example, one skilled in the art can easily adopt inhibitors as described in WO 98/24806, which discloses substituted oxadiazole, thiadiazole and triazole as serine protease inhibitors. U.S. Pat. No. 5,874,585 discloses substituted heterocyclic compounds useful as inhibitors of serine proteases including: (benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(3-trifluoromethylbenzyl)-1,2,4-oxadiazolyl)carbonyl)-2-(S)-methylpropyl]-L-prolinamide benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(2-phenylethyl)-1,2,4-oxadiazolyl)carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(2-methoxybenzyl)-1,2,4-oxadiazolyl)carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(trifluoromethyl)-1,2,4-oxadiazolyl)carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(methyl)-1,2,4-oxadiazolyl)carbonyl)-2-(S)-Methylpropyl]-L-Prolinamide; (Benzyloxycarbonyl)-L-Valyl-N-[1-(3-(5-(difluoromethyl)-1,2,4-oxadiazolyl) carbonyl)-2-(S)-Methylpropyl]-L-Prolinamide; (Benzyloxycarbonyl)-L-Valyl-N-[1-(3-(5-(benzyl)-1,2,4-oxadiazolyl)carbonyl)-2-(S)-Methylpropyl]-L-Prolinamide; (Benzyloxycarbonyl)-L-Val yl-N-[1-(3-(5-(3-methoxybenzyl)-1,2,4-oxadiazolypcarbonyl)-2-(S)-Methylpropyl]-L-Prolinamide; (Benzyloxycarbonyl)-L-Valyl-N-[1-(3-(5-(2,6-difluorobenzyl)-1,2,4-oxadiazolyl)carbonyl)-2-(S)-Methylpropyl]-L-Prolinamide; (Benzyloxycarbonyl)-L-Valyl-N-[1-(3-(5-(trans-styryl)-1,2,4-oxadiazolypcarbonyl)-2-(S)-MethylpropyThL-Prolinamide; (Benzyloxycarbonyl)-L-Valyl-N-[1-(3-(5-(trans-4-Trifluoro methylstyryl)-1,2,4-oxadiazolyecarbonyl)-2-(S)-Methylpropyl]-L-Prolinamide; (Benzyloxycarbonyl)-L-Valyl-N-[1-(3-(5-(trans-4-Methoxystyryl)-1,2,4-oxadiazolyl)carbonyl)-2-(S)-Methylpropyl]-L-Prolinamide; (Benzyloxycarbonyl)-L-Valyl-N-[1-(3-(5-(3-Thienylmethyl)-1,2,4-oxadiazolyl)carbonyl)-2-(S)-Methylpropyl]-L-Prolinamide; (Benzyloxycarbonyl)-L-Valyl-N-[1-(3-(5-(Phenyl)-1,2,4-oxadiazolyl)carbonyl)-2-(S)-methylpropyl]-L-prolinamide; and (Benzyloxycarbonyl)-L-Valyl-N-[1-(3-(5-(3-Phenylpropyl)-1,2,4-oxadiazolyecarbonyl)-2-(S)-Methylpropyl]-L-Prolinamide. U.S. Pat. No. 5,216,022 teaches other small molecules useful for the practice of this invention, including: Benzyloxycarbonyl-L-valyl-N-[1-(2-[5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide (also known as CE-2072), Benzyloxycarbonyl-L-valyl-N-[1-(2-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; Benzyloxycarbonyl-L-valyl-N-[1-(2-(5-(methyl)-1,3,4-oxadiazoly]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; Benzyloxycarbonyl)-L-valyl-N-[1-(2-(5-(3-trifluoromethylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(2-(5-(4-Dimethylamino benzyl)-1,3,4-oxadiazolyflcarbonyl)-2-(S)-methylpropyl]-L-prolinamide; Benzyloxycarbonyl)-L-valyl-N-[1-(2-(5-(1-napthylenyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(3,4-methylenedioxybenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(3,5-dimethylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(3,5-dimethoxybenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(3,5-ditrifluoromethylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(3-methylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(biphenylmethine)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(4-phenylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(3-phenylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(3-phenoxybenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(cyclohexylmethylene)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(3-trifluoromethyldimethylmethylene)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(1-napthylmethylene)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(3-pyridylmethyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(3,5-diphenylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; (Benzyloxycarbonyl)-L-valyl-N-[1-(3-(5-(4-dimethylaminobenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-L-prolinamide; 2-(5-[(Benzyloxycarbonyl)amino]-6-oxo-2-(4-fluorophenyl)-1,6-dihydro-1-pyrimidinyl]-N-[1-(3-(5-(3-trifluoromethylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-(S)-2-methylpropyl]acetamide; 2-(5-Amino-6-oxo-2-(4-fluorophenyl)-1,6-dihydro-1-pyrimidinyl]-N-[1-(3-(5-(3-trifluoromethylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 2-(5-[(Benzyloxycarbonyl)amino]-6-oxo-2-(4-fluorophenyl)-1,6-dihydro-1-pyrimidinyl]-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-(S)-2-methylpropyl]acetamide; 2-(5-Amino-6-oxo-2-(4-fluorophenyl)-1,6-dihydro-1-pyrimidinyl]-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-methylpropyl]acetamide; (Pyrrole-2-carbonyl)-N-(benzyl)glycyl-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]amide; (Pyrrole-2-carbonyl)-N-(benzyl)glycyl-N-[1-(3-(5-(3-trifluoromethylbenzyl)]-1,2,4-oxadiazolyl)-(S)-methylpropyl]amide; (2S,5S)-5-Amino-1,2,4,5,6,7-hexahydroazepino-[3,2,1]-indole-4-one-carbon yl-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-(R,S)-2-methylpropyl]amide; BTD-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]amide; (R,S)-3-Amino-2-oxo-5-phenyl-1,4,-benzodiazepine-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; (Benzyloxycarbonyl)-L-valyl-2-L-(2,3-dihydro-1H-indole)-N-[1-(2-[5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]amide; (Benzyloxycarbonyl)-L-valyl-2-L-(2,3-dihydro-1H-indole)-N-[1-(3-(5-(3-trifluoromethylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]amide; Acetyl-2-L-(2,3-dihydro-1H-indole)-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]amide; 3-(S)-(Benzyloxycarbonyl)amino)-ε-lactam-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 3-(S)-(Amino)-ε-lactam-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide trifluoroacetic acid salt; 3-(S)-[(4-morpholino carbonyl-butanoyl)amino]-ε-lactam-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(R,S)-methylpropyl]acetamide; 6-[4-Fluorophenyn-ε-lactam-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 2-(2-(R,S)-Phenyl-4-oxothiazolidin-3-yl]-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 2-(2-(R,S)-phenyl-4-oxothiazolidin-3-yl]-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]hydroxymethyl)-2-(S)-methylpropyl]acetamide; 2-(2-(R,S)-Benzyl-4-oxothiazolidin-3-yl]-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]-acetamide; 2-(2-(R,S)-Benzyl-4-oxothiazolidin-3-yl oxide]-N-[1-(3-(5-(3-trifluoromethylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(R,S,)-methylpropyl]acetamide; (1-Benzoyl-3,8-quinazolinedione)-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; (1-Benzoyl-3,6-piperazinedione)-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; (1-Phenyl-3,6-piperazinedione)-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; [(1-Phenyl-3,6-piperazinedione)-N-[1-(3-(5-(3-trifluoromethylbenzyl)-1,2,4-oxadiazolyl]carbonyl)]-2-(S)-methylpropyl]acetamide; 3-[(Benzyloxycarbonyl)amino]-quinolin-2-one-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 3-[(Benzyloxycarbonyl)amino]-7-piperidinyl-quinolin-2-one-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 3-(Carbomethoxy-quinolin-2-one-N-[1-(2-(5-(3-methybenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 3-(Amino-quinolin-2-one)-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 3-[(4-Morpholino)aceto]amino-quinolin-2-one-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 3,4-Dihydro-quinolin-2-one-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 1-Acetyl-3-(4-fluorobenzylidene) piperazine-2,5-dione-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 1-Acetyl-3-(4-dimethylamino benzylidene)piperazine-2,5-dione-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 1-Acetyl-3-(4-carbomethoxy benzylidene)piperazine-2,5-dione-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 1-Acetyl-3-[(4-pyridyl)methylene]piperazine-2,5-dione-N-[1-(2-(5-(3-methyl benzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 4-[1-Benzyl-3-(R)-benzyl-piperazine-2,5,-dione]-N-[1-(2-[5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 4-[1-Benzyl-3-(S)-benzyl piperazine-2,5,-dione]-N-[1-(2-(5-(3-methyl benzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 4-[1-Benzyl-3(R)-benzylpiperazine-2,5,-dione]-N-[1-(3-(5-(3-trifluoromethylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 441-Benzyl-3-(S)-benzylpiperazine-2,5,-dione]-N-[1-(3-(5-(3-trifluoromethylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 4-[1-Benzyl-3-(S)-benzyl piperazine-2,5,-dione]-N-[1-(3-(5-(2-dimethylaminoethyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 4-[1-Methyl-3-(R,S)-phenylpiperazine-2,5,-dione]-N-[1-(3-(5-(3-trifluoromethylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 4-[1-Methyl-3-(R,S)-phenyl piperazine-2,5,-dione]-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 4-[1-(4-Morpholino ethyl)3-(R)-benzyl piperazine-2,5,-dione]-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 5-(R,S)-Phenyl-2,4-imidazolidinedione-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 5-(R)-Benzyl-2,4-imidazolidinedione-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 5-(S)-Benzyl-2,4-imidazolidinedione-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 5-(S)-Benzyl-2,4-imidazolidinedione-N-[1-(3-(5-(3-trifluoromethylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 5-(R)-Benzyl-2,4-imidazolidinedione-N-[1-(3-(5-(3-trifluoromethylbenzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; 1-Benzyl-4-(R)-benzyl-2,5-imidazolidinedione-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide; and 1-Benzyl-4-(R)-benzyl-2,5-imidazolidinedione-N-[1-(3-(5-(3-trifluoromethyl benzyl)-1,2,4-oxadiazolyl]carbonyl)-2-(S)-methylpropyl]acetamide among others.
  • Yet another embodiment of the invention is directed toward the inhibition of apoptosis resulting from the interaction between a serine protease inhibitor and a cell surface receptor and resulting in a measurable decrease in apoptosis. The serine protease inhibitor of this embodiment is an α1-antitrypsin or an α1-antitrypsin-like agent. The α1-antitrypsin-like agents includes substituted oxydiazoles, substituted thiadiazole, substituted triazole peptoids, or combinations of these agents. The substituted oxydiazole, thiadiazole, and triazole peptoids are synthesized de novo or derivatized from existing compounds.
  • 5.3. Diseases Addressed by the Invention
  • Specific diseases or disorders for which the therapeutic methods of the invention are beneficial include wasting diseases of various types. The diseases include cancer, neurodegenerative diseases, myocardial infarction, and stroke. The cancers include bladder, breast, kidney, leukemia, lung, myoloma, liposarcoma, lymphoma, tongue, prostate, and uterine cancers. The method of the invention is also applied to Alzheimer's disease, arthritis, muscular dystrophy, Downs Syndrome, sepsis, HIV infection, multiple sclerosis, arteriosclerosis, diabetes, and arthritis. In fact, the invention is applied to any disease associated with elevated levels of apoptosis.
  • 5.4. Modes of Administration
  • Modes of administration of the various therapeutic agents used in the invention are exemplified in the examples below. However, the agents are delivered by any of a variety of routes including: by injection (e.g., subcutaneous, intramuscular, intravenous, intra-arterial, and intraperitoneal), by continuous intravenous infusion, transdermally, orally (e.g., tablet, pill, liquid medicine), by implanted osmotic pumps (e.g., ALZA Corp.), by suppository or aerosol spray.
  • Those skilled in the art of biochemical synthesis will recognize that for commercial scale quantities of peptides, such peptides are preferably prepared using recombinant DNA techniques, synthetic techniques, or chemical derivatization of biologically or chemically synthesized peptides.
  • The compounds of the present invention are used as therapeutic agents in the treatment of a physiological, or especially, pathological, condition caused in whole or part by uncontrolled serine protease and apoptosis activity. The peptides or peptoids can be administered as free peptides, free peptoids, or pharmaceutically acceptable salts thereof. The terms used herein conform to those in Budavari, S. (Ed.), “The Merck Index, An Encyclopedia of Chemicals, Drugs, and Biologicals,” Merck Company, Inc., twelfth edition. The term “pharmaceutically acceptable salt” refers to those acid addition salts or methyl complexes of the peptides which do not significantly or adversely affect the therapeutic properties including efficacy and toxicity, of the peptides and peptoids. The peptides and peptoids are administered to individuals as a pharmaceutical composition which, in most cases, will comprise the peptide, peptoid, and/or pharmaceutical salts thereof with a phainiaceuticaily acceptable carrier. The term “pharmaceutically acceptable carrier” refers to those solid and liquid carriers, which do not significantly or adversely affect the therapeutic properties of the peptides.
  • The pharmaceutical compositions containing peptides and/or peptoids of the present invention are administered to individuals, particularly humans, either intravenously, subcutaneously, intramuscularly, intranasally, orally, topically, transdermally, parenterally, gastrointestinally, transbronchially, and transalveolarly. Topical administration is accomplished by a topically applied cream, gel, rinse, etc. containing therapeutically effective amounts of inhibitors of serine proteases. Transdermal administration is accomplished by administration of a cream, rinse, gel, etc. capable of allowing the inhibitors of serine proteases to penetrate the skin and enter the blood stream. Parenteral routes of administration include, but are not limited to, direct injection such as intravenous, intramuscular, intraperitoneal, or subcutaneous injection. Gastrointestinal routes of administration include, but are not limited to, ingestion and rectal. Transbroncheal and transalveolar routes of administration include, but are not limited to, inhalation, either via the mouth or intranasally, and direct injection into an area, such as through a tracheotomy, endotracheal tube, or aspirated through a respiratory mist. In addition, osmotic pumps are used for administration. The necessary dosage will vary with the particular condition being treated, method of administration, and rate of clearance of the molecule from the body.
  • 6. EXAMPLES
  • The following specific examples are provided to better assist the reader in the various aspects of practicing the present invention. As these specific examples are merely illustrative, nothing in the following descriptions should be construed as limiting the invention in any way. Such limitations are, of course, defined solely by the accompanying claims.
  • 6.1. Effect of Therapy with α1-Antitrypsin Following Experimental Myocardial Infarction or Stroke Rats (female, 250-300 g each) are randomly assigned to one of four groups: myocardial infarction control, stroke control, myocardial infarction, and stroke. The rats are subjected to a 30 minute ligation of the coronary arterial supply (for the myocardial infarction group) or the left carotid artery (for the stroke group), followed by release of the ligature. Sham operated controls receive the cut-down and manipulation of the artery but without ligation. Immediately preceding the ligation or sham ligation, half of the animals in each group (by random selection) receive α1-antitrypsin (sufficient to achieve a 50 μM concentration of added agent in the blood, or in the alternative, an amount equal to 10 mg/kg body weight) and the other half receive a body-weight equivalent volume of AAT vehicle, intravenously. The AAT vehicle is phosphate-buffered saline, or optionally, any pharmaceutically acceptable carrier. At twenty-four hours after release of the sham or actual ligation the animals are sacrificed and the hearts and brains removed for analysis of the amount of apoptosis. In other experiments the dosage of α1-antitrypsin administered is varied between the amounts necessary to produce a concentration of 10 μM and 250 μM in the blood. In general, a concentration of 5 mg/ml of α1-antitrypsin is equivalent to about 100 μM. In yet other experiments the frequency of administration is varied from once per day to four times per day. Likewise, antielastase and antiproteinase are used.
  • 6.2. Anti-Apoptosis Therapy for Septic Shock
  • Protection of mouse L929 cells from apoptotic effects of TNF are evaluated using: the agents α1-antitrypsin; (Benzyloxycarbonyl)-L-Valyl-N-[1-2-3(5-(3-methylbenzyl)-1,3,4-oxadiazolypcarbonyl)-2-(S)-Methylpropyl]-L-Prolinamide; (Benzyloxcarbonyl)-L-Valyl-N-[1-(3-(5-(2-Phenylethyl)-1,2,4-oxadiazolyecarbonyl)-2-(S)-Methylpropyl]-L-Prolinamide; and (Benzyloxcarbonyl)-L-Valyl-N-[1-(3-(5-(2-Methoxybenzyl)-1,2,4-oxadiazolyecarbonyl)-2-(S)-Methylpropyl]-L-Prolinamide L929. Cells (105 cells/well) are treated with 300 ng/ml of human Tumor Necrosis Factor (TNF) with or without the agent (added one hour prior to TNF addition) at 0.02, 0.1, 0.2, 1.0, 2.0 and 10 mg agent/ml. One day later the cells are stained for viability using 2′-[4-hydroxyphenyl]-5-[4-methyl-1-piperazinyl]-2,5′-bi-1H-benzimidazole and fluorescence analyzed for apoptosis using a Leitz fluorescence microscope. The results are evaluated in terms of the dose response to the agent.
  • 6.3. Free Radical Scavengers as Co-Inhibitors of Apoptosis
  • Agents that reduce free radical levels do not directly prevent the oxidizing effect of free radicals. Therefore, it is advantageous to administer two or three independently acting agents, as opposed to a single agent. Thus, one preferred embodiment of the process is the co-administration of α1-antitrypsin and a free radical scavenger, such as glutathione (1 mg/kg body weight).
  • 6.4. Free Radical Scavengers as Co-Inhibitors of Apoptosis
  • In yet another embodiment of the invention, oxidation-resistant α1-antitrypsin variants are used to avoid inactivation by excess free radicals. As an example, synthetic α1-antitrypsin or recombinant α1-antitrypsin produced with alternative and oxidation-resistant amino acid sequences are embodiments of the invention.
  • 6.5. Effect of AAT and CE-2072 on Apoptosis in RCG Neuron Cells
  • RCG neuronal cells are seeded into cell culture dishes in 400 μl cell culture medium (Eagle's basal medium, BME) containing 10% (v/v) FBS. At day two the now conditioned medium is removed and the cells are treated for 10 hours as follows:
  • Group Condition Treatment
    1. negative control conditioned medium with serum
    2. positive control BME without serum
    3. experimental BME without serum, with IGF-I (50 ng/ml)
    4. experimental BME without serum, with lyophilized AAT
    (50 μM)
    5. experimental BME without serum, with soluble AAT (50 μM)
    6. experimental BME without serum, with CE-2072 (60 μM) in
    DMSO
    7. diluent control BME without serum, with vehicle
    8. DMSO control BME without serum, with DMSO
  • Then the medium is replaced with 4% (w/v) paraformaldehyde, incubated for 15 minutes at room temperature, and the cells stained with Hoechst dye 33258 (8 μg/ml) for 15 minutes at room temperature. The apoptosis in the cells is evaluated using a fluorescence microscope by an evaluator blinded to the method of treatment. The results are shown in FIG. 2.
  • The apoptosis induced by depletion of serum is blocked by lyophilized α1-antitrypsin and by agent CE-2072 (a synthetic inhibitor of serine protease). The latter is formally known as benzyloxcarbonyl-L-valyl-N-[1-(2-(5-(3-methylbenzyl)-1,3,4-oxadiazolyl]carbonyl)-2-(S)-Methylpropyl]-L-prolinamide.
  • 6.6. Amelioration of Ischemia in Donor Organs During Transport and Transplant
  • Human donor organs, including kidneys, are subject to ischemia during transport, which can last up to several hours. Biopsies (3 mm) are removed from the top medial surface of donor kidneys undergoing transport prior to implantation, and grouped by time after removal from the donor: 1-2 hours, 2-4 hours, and greater than 4 hours. Donor kidneys transplanted within one hour serve as the first control and the contralateral kidney serves as the second control. Half of the donor kidneys are treated with α1-antitrypsin (10 mg/ml fluid) upon removal from the donor to inhibit apoptosis.
  • 6.7. Therapy with Oxidation-Resistant Recombinant α1-Antitrypsin Variants
  • Val358-antitrypsin and Ile358-antitrypsin are produced from the appropriate nucleotide sequences by methods well known in the art, including construction of a plasmid, transfection of the host E. coli, selection of transfected colonies, expansion of the culture, and isolation and purification of the mutant gene product. Amounts of the recombinant agents effective in inhibiting apoptosis, excessive clotting, neutrophil extravasation, ischemia-reperfusion injury, or myocardial damage are applied in an experimental model of myocardial infarction (see Example 6.1, supra). Effective amounts are between 0.03 and 7 g/kg body weight, for example, about 0.5 g/kg. In some experiments the amount of variant antitrypsin is measured in the blood or other biological fluid. In those tests sufficient variant antitrypsin is administered to provide a concentration of about 1 μM to about 100 μM in the blood or biological fluid.
  • 6.8. Effect of α1-antitrypsin on apoptosis Primary rat brain granule cells are pretreated for one hour in the absence or presence of α1-antitrypsin (3.0 mg/ml), followed by replacement of the cell culture medium with either control medium containing 10% (vol/vol) fetal bovine serum, medium devoid of fetal bovine serum, or medium devoid of fetal bovine serum but containing α1-antitrypsin. After 24 hours of culture the level of apoptosis is measured. α1-Antitrypsin completely reverses the apoptosis associated with serum depletion, which results in cell death.
  • Throughout this application various publications and patents are referenced. The disclosures of these publications and patents in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this invention pertains. While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications, and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure has come within known or customary practice within the art to which the invention pertains and as can be applied to the essential features here before set forth and as follows in the scope of the appended claims.

Claims (17)

1-28. (canceled)
29. A method of treating arthritis, autoimmune disease or ischemia-reperfusion injury in a subject in need of such a treatment, said method comprising administering a composition comprising a therapeutically effective amount of α1-antitrypsin (AAT), an oxidation-resistant α1-antitrypsin Met358 variant, a free radical-resistant α1-antitrypsin M358 variant, or a combination thereof to the subject.
30. The method of claim 29, wherein the composition further comprises at least one free radical scavenger or inhibitor.
31. The method of claim 29, wherein the therapeutically effective amount is sufficient to provide at least 10 μM and no greater than 2 mM of the inhibitor in a biological fluid of the subject.
32. The method of claim 29, wherein the autoimmune disease comprises Ankylosing Spondylitis, Chagas disease, chronic obstructive pulmonary disease, Crohns Disease, Dermatomyositis, Endometriosis, Goodpasture's syndrome, Graves' disease, Guillain-Barré syndrome, Hashimoto's disease, Hidradenitis suppurativa, Kawasaki disease, IgA nephropathy, idiopathic thrombocytopenic purpura, interstitial cystitis, lupus erythematosus, mixed connective tissue disease, morphea, myasthenia gravis, narcolepsy, neuromyotonia, pemphigus vulgaris, pernicious anaemia, psoriasis, psoriatic Arthritis, polymyositis, primary biliary cirrhosis, relapsing polychondritis, rheumatoid arthritis, schizophrenia, scleroderma, Sjögren's syndrome, stiff person syndrome, temporal arteritis, ulcerative colitis, vasculitis, vitiligo, or Wegener's granulomatosis.
33. The method of claim 29, wherein the ischemia-reperfusion injury comprises ischemia-reperfusion injury associated with thrombolytic therapy, organ transplantation, coronary angioplasty, aortic cross-clamping, or cardiopulmonary bypass surgery.
34. The method of claim 29, in which the therapeutically effective amount is sufficient to provide at least 0.5 μM and no greater than 2000 μM in a biological fluid of the subject.
35. The method of claim 29, wherein the composition is administered parenterally, orally, vaginally, rectally, nasally, buccally, intravenously, intramuscularly, subcutaneously, intrathecally, epidurally, transdermally, intracerebroventricularly, by osmotic pump, by inhalation, or combinations thereof.
36. The method of claim 29, wherein the composition is administered orally.
37. The method of claim 29, wherein the composition is administered at least once daily.
38. The method of claim 29, wherein the composition comprises AAT.
39. The method of claim 38, wherein the AAT is substantially purified from a wild type, mutant, or transgenic mammalian source.
40. The method of claim 38, wherein the AAT is isolated from a culture of wild type, mutant, or transformed cells.
41. The method of claim 29, wherein the composition further comprises a pharmaceutically acceptable carrier.
42. The method of claim 29, wherein α1-antitrypsin, an oxidation-resistant α1-antitrypsin Met358 variant, a free radical-resistant α1-antitrypsin M358 variant, or a combination thereof are derivatized by esterification, acetylation, or amidation.
43. A method for ameliorating ischemia in a donor organ during transport, transplant, or a combination thereof, said method comprising treating the donor organ with a composition comprising α1-antitrypsin (AAT), an oxidation-resistant α1-antitrypsin Met358 variant, a free radical-resistant α1-antitrypsin M358 variant, or a combination thereof upon removal of the donor organ from a subject.
44. The method of claim 43, wherein the composition comprises AAT.
US12/767,858 1999-03-05 2010-04-27 Methods For Treating Arthritis, Autoimmune Disease, and Ischemia-Reperfusion Injury Abandoned US20110319330A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/767,858 US20110319330A1 (en) 1999-03-05 2010-04-27 Methods For Treating Arthritis, Autoimmune Disease, and Ischemia-Reperfusion Injury

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US12316799P 1999-03-05 1999-03-05
US09/518,081 US7704958B1 (en) 1999-03-05 2000-03-03 Methods and compositions for inhibiting apoptosis using serine protease inhibitors
US12/767,858 US20110319330A1 (en) 1999-03-05 2010-04-27 Methods For Treating Arthritis, Autoimmune Disease, and Ischemia-Reperfusion Injury

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/518,081 Division US7704958B1 (en) 1999-03-05 2000-03-03 Methods and compositions for inhibiting apoptosis using serine protease inhibitors

Publications (1)

Publication Number Publication Date
US20110319330A1 true US20110319330A1 (en) 2011-12-29

Family

ID=22407091

Family Applications (4)

Application Number Title Priority Date Filing Date
US09/518,081 Expired - Lifetime US7704958B1 (en) 1999-03-05 2000-03-03 Methods and compositions for inhibiting apoptosis using serine protease inhibitors
US12/051,373 Expired - Fee Related US8071551B2 (en) 1999-03-05 2008-03-19 Methods and compositions for treating diabetes
US12/767,858 Abandoned US20110319330A1 (en) 1999-03-05 2010-04-27 Methods For Treating Arthritis, Autoimmune Disease, and Ischemia-Reperfusion Injury
US13/281,311 Abandoned US20120040913A1 (en) 1999-03-05 2011-10-25 Methods and compositions for treating diabetes

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/518,081 Expired - Lifetime US7704958B1 (en) 1999-03-05 2000-03-03 Methods and compositions for inhibiting apoptosis using serine protease inhibitors
US12/051,373 Expired - Fee Related US8071551B2 (en) 1999-03-05 2008-03-19 Methods and compositions for treating diabetes

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/281,311 Abandoned US20120040913A1 (en) 1999-03-05 2011-10-25 Methods and compositions for treating diabetes

Country Status (3)

Country Link
US (4) US7704958B1 (en)
AU (1) AU3731400A (en)
WO (1) WO2000051624A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090298747A1 (en) * 1999-03-05 2009-12-03 Bio Holding, Inc. Methods and Compositions For Treatment of Nitric Oxide-Induced Clinical Conditions
US10300119B2 (en) 2013-05-15 2019-05-28 Mor Research Applications Ltd. Compositions and methods for preventing injury during or resulting from cardiac surgery
US10531655B2 (en) 2011-12-02 2020-01-14 The Regents Of The University Of California Reperfusion protection solution and uses thereof
US10980866B2 (en) 2014-09-22 2021-04-20 Hadasit Medical Research Services And Development Ltd. Alpha-1 anti-trypsin for treating liver diseases

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000051624A2 (en) * 1999-03-05 2000-09-08 The Trustees Of University Technology Corporation Methods and compositions useful in inhibiting apoptosis
DE10148553A1 (en) * 2001-10-01 2003-04-17 Protagen Ag Proteinase inhibitors for the therapy and diagnosis of neurodegenerative diseases
US6670325B2 (en) 2001-10-19 2003-12-30 Alpha Med Pharmaceuticals Corp. Treatment of osteocarcinoma with alpha-1—antitrypsin or secretory leucocyte protease inhibitor
WO2003060466A2 (en) * 2001-12-21 2003-07-24 Immunochemistry Technologies, Llc Affinity labeling of serine proteases for simultaneous detection of multiple serine protease activity levels
US7850970B2 (en) 2003-08-26 2010-12-14 The Regents Of The University Of Colorado Inhibitors of serine protease activity and their use in methods and compositions for treatment of bacterial infections
EP1697331A4 (en) * 2003-12-19 2010-08-04 Merck Sharp & Dohme Mitotic kinesin inhibitors
GB0411145D0 (en) * 2004-05-19 2004-06-23 Imp College Innovations Ltd Protease inhibitors and their therapeutic applications
CA2654446C (en) * 2005-06-07 2014-02-25 The Regents Of The University Of Colorado Inhibitors of serine protease activity and their use in methods and compositions for treatment of graft rejection and promotion of graft survival
US9457070B2 (en) 2005-06-07 2016-10-04 The Regents Of The University Of Colorado, A Body Corporate Compositions, methods and uses of alpha 1-antitrypsin for early intervention in bone marrow transplantation and treatment of graft versus host disease
CA2671322A1 (en) * 2005-12-02 2007-07-12 Regents Of The University Of Colorado Compositions and methods for treating actin-mediated medical conditions by administering an actin modulating agent
EP2066174B1 (en) * 2006-09-12 2017-11-08 Beth Israel Deaconess Medical Center, Inc. Compositions containing alpha-1-antitrypsin and methods for use
EP2089065B1 (en) 2006-10-27 2015-01-21 E.I. Du Pont De Nemours And Company Method for prion decontamination
US20110002909A1 (en) * 2007-11-02 2011-01-06 Vikram Arora Method, composition, and article of manufacture for providing alpha-1 antitrypsin
US20110237496A1 (en) * 2008-09-10 2011-09-29 Ilana Nathan Antinecrotic activity of alpha 1-antitrypsin
US20130131192A1 (en) 2009-11-03 2013-05-23 Grifols Therapeutics Inc. Composition, method, and kit for alpha-1 proteinase inhibitor
US9522179B2 (en) * 2010-03-04 2016-12-20 Virginia Commonwealth University Compositions and methods for modulating cardiac conditions
WO2011116220A2 (en) * 2010-03-17 2011-09-22 Arbonne International Llc Oral supplement
US20120225813A1 (en) * 2010-06-11 2012-09-06 Lewis Eli C Compositions, methods and uses for treatment of type 1 diabetes
US9518107B2 (en) 2010-08-31 2016-12-13 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. Pharmaceutical compositions containing polypeptides derived from α-1 antitrypsin and methods of use thereof
WO2012029061A2 (en) 2010-08-31 2012-03-08 Uri Wormser POLYPEPTIDES DERIVED FROM α-1 ANTITRYPSIN AND METHODS OF USE THEREOF
KR20200044138A (en) 2011-06-24 2020-04-28 더 리젠츠 오브 더 유니버시티 오브 콜로라도, 어 바디 코포레이트 Compositions, methods and uses for alpha-1 antitrypsin fusion molecules
JP2015504675A (en) 2012-01-10 2015-02-16 ザ リージェンツ オブ ザ ユニバーシティ オブ コロラド,ア ボディー コーポレイトTHE REGENTS OF THE UNIVERSITY OF COLORADO,a body corporate Compositions, methods, and uses of alpha-1 antitrypsin fusion molecules
CA2982387A1 (en) * 2015-04-16 2016-10-20 Prime Bio-Drug Development Limited Therapeutic peptides for cerebrovascular diseases
JP2020063311A (en) * 2020-01-31 2020-04-23 プライム・バイオ‐ドラッグ・ディヴェロップメント・リミテッドPrime Bio‐Drug Development Limited Therapeutic peptides for cerebrovascular diseases

Citations (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5114917A (en) * 1986-12-24 1992-05-19 John Lezdey Treatment of inflammation using alpha 1-antichymotrypsin
US5187089A (en) * 1990-06-21 1993-02-16 Incyte Pharmaceuticals, Inc. Protease nexin-i variants which inhibit elastase
US5508031A (en) * 1986-11-21 1996-04-16 Cetus Oncology Corporation Method for treating biological damage using a free-radial scavenger and interleukin-2
US5652237A (en) * 1994-09-09 1997-07-29 Warner-Lambert Company 2-substituted-4H-3, 1-benzoxazin-4-ones and benzthiazin-4-ones as inhibitors of complement C1r protease for the treatment of inflammatory processes
US5851987A (en) * 1997-04-14 1998-12-22 Incyte Pharmaceuticals, Inc. Human tumor-associated Kazal inhibitor-like polypeptides and encoding polynucleotides
US5872124A (en) * 1996-07-31 1999-02-16 Thomas Jefferson University Treatment of diseases of the central nervous system using uric acid as a scavenger of peroxynitrite
US5928883A (en) * 1996-11-13 1999-07-27 Mayo Foundation For Medical Education Eosinophil granole proteins as indicators of inflammatory bowel disorders
US6008196A (en) * 1994-06-02 1999-12-28 Hoechst Marion Roussel, Inc. Perfluoroalkyl ketone inhibitors of elastase and processes for making the same
US6126933A (en) * 1995-06-27 2000-10-03 Genetics Institute Methods of treating inflammatory bowel diseases by administering IL-11
US6271436B1 (en) * 1996-10-11 2001-08-07 The Texas A & M University System Cells and methods for the generation of transgenic pigs
US6303629B1 (en) * 1997-05-13 2001-10-16 Octamer, Inc. Methods for treating inflammation, inflammatory diseases, arthritis and stroke using pADPRT inhibitors
US6323219B1 (en) * 1998-04-02 2001-11-27 Ortho-Mcneil Pharmaceutical, Inc. Methods for treating immunomediated inflammatory disorders
US6342373B1 (en) * 1983-11-21 2002-01-29 Ucp Gen-Pharma Ag Process for preparing recombinant eglin, protease inhibitor
US6489308B1 (en) * 1999-03-05 2002-12-03 Trustees Of University Of Technology Corporation Inhibitors of serine protease activity, methods and compositions for treatment of nitric-oxide-induced clinical conditions
US6653350B1 (en) * 1998-09-08 2003-11-25 G.D. Searle & Co. Methods of treating osteoarthritis with inducible nitric oxide synthase inhibitors
US6849605B1 (en) * 1999-03-05 2005-02-01 The Trustees Of University Technology Corporation Inhibitors of serine protease activity, methods and compositions for treatment of viral infections
US20080261869A1 (en) * 2007-04-20 2008-10-23 Leland Shapiro Compositions and methods of use for alpha-1 antitrypsin having no significant serine protease inhibitor activity
US20090118162A1 (en) * 2005-06-07 2009-05-07 Regents Of The University Of Colorado Inhibitors of serine protease activity and their use in methods and compositions for treatment of graft rejection and promotion of graft survival
US20090203580A1 (en) * 2005-06-07 2009-08-13 Dinarello Charles A Compositions and methods of use for alpha-1 antitrypsin having no significant serine protease inhibitor activity
US20090298747A1 (en) * 1999-03-05 2009-12-03 Bio Holding, Inc. Methods and Compositions For Treatment of Nitric Oxide-Induced Clinical Conditions
US7704958B1 (en) * 1999-03-05 2010-04-27 Bio Holding, Inc. Methods and compositions for inhibiting apoptosis using serine protease inhibitors
US7850970B2 (en) * 2003-08-26 2010-12-14 The Regents Of The University Of Colorado Inhibitors of serine protease activity and their use in methods and compositions for treatment of bacterial infections
US7879846B2 (en) * 2006-09-21 2011-02-01 Kyorin Pharmaceutical Co.., Ltd. Serine hydrolase inhibitors
US20110201587A1 (en) * 2010-02-16 2011-08-18 Bio Holding, Inc. Hsp90 inhibitors and methods of use
US20120225813A1 (en) * 2010-06-11 2012-09-06 Lewis Eli C Compositions, methods and uses for treatment of type 1 diabetes

Family Cites Families (87)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5874424A (en) 1995-12-20 1999-02-23 Vertex Pharmaceuticals Incorporated Inhibitors of interleukin-1β converting enzyme
US4711848A (en) * 1984-03-14 1987-12-08 Zymogenetics, Inc. Site specific mutagenesis in alpha-1-antitrypsin
US5247084A (en) 1985-11-12 1993-09-21 Ono Pharmaceutical Co., Ltd. Derivatives of p-guanidinobenzoic acid
US4629567A (en) * 1986-03-07 1986-12-16 Smithkline-Rit Alpha-1-antiprotease purification
US5322775A (en) 1986-06-30 1994-06-21 Pharmaceutical Proteins Ltd. Peptide production
US4829054A (en) * 1987-04-13 1989-05-09 Miles Laboratories, Inc. Method of decreasing lung damage in a host following the onset of gram negative septicemia/endotoxemia
GB8826446D0 (en) 1988-11-11 1988-12-14 Agricultural & Food Res Peptide production
US5665589A (en) 1988-12-14 1997-09-09 The United States Of America As Represented By The Department Of Health And Human Services Human liver epithelial cell lines
US5529920A (en) 1988-12-14 1996-06-25 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Human liver epithelial cell line and culture media therefor
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
US5612194A (en) 1989-06-23 1997-03-18 Trustees Of The University Of Pennsylvania Methods of producing effective recombinant serine protease inhibitors and uses of these inhibitors
US5040545A (en) * 1989-11-02 1991-08-20 Possis Medical, Inc. Releasable lock assembly
US5663416A (en) 1990-05-22 1997-09-02 Cortech Inc. Oxidant sensitive and insensitive aromatic esters as inhibitors of human neutrophil elastase
US5134119A (en) * 1990-10-16 1992-07-28 Lezdey John Treatment of inflammation using 358 substituted alpha-antitrypsin
US5240956A (en) 1990-11-07 1993-08-31 Cortech, Inc. Ester inhibitors
US5470970A (en) 1991-02-28 1995-11-28 Dana-Farber Cancer Institute, Inc. Maspin, a serpin with tumor suppresing activity
US5416191A (en) 1991-04-01 1995-05-16 Cortech, Inc. Bradykinin antagonists
US5863899A (en) 1991-04-01 1999-01-26 Cortech, Inc. Bradykinin antagonists
US5843900A (en) 1991-04-01 1998-12-01 Cortech, Inc. Bradykinin antagonists
JPH06508116A (en) 1991-04-01 1994-09-14 コルテク,インコーポレイテッド bradykinin antagonist
US5610140A (en) 1991-04-01 1997-03-11 Cortech, Inc. Bradykinin receptor antagonists with neurokinin receptor blocking activity
JPH06509327A (en) 1991-04-18 1994-10-20 ザ ユー エー ビー リサーチ ファンデーション Composition and method for inhibiting elastase
US5478727A (en) 1991-05-24 1995-12-26 Arch Development Corporation Methods and compositions for the preparation and use of a herpes protease
FR2679920A1 (en) 1991-08-02 1993-02-05 Rhone Poulenc Rorer Sa Highly stable recombinant yeasts for the production of recombinant proteins, their preparation and their use
US5641670A (en) 1991-11-05 1997-06-24 Transkaryotic Therapies, Inc. Protein production and protein delivery
US5216022A (en) 1991-12-19 1993-06-01 Cortech, Inc. Aromatic esters of phenylenedialkanoates as inhibitors of human neutrophil elastase
WO1993018794A1 (en) * 1992-03-26 1993-09-30 Gensia, Inc. In vivo peptide therapy
GB9209882D0 (en) * 1992-05-07 1992-06-24 Glaxo Lab Sa Compositions
US5432178A (en) 1992-09-18 1995-07-11 Ono Pharmaceutical Co., Ltd. Amidinophenol derivatives
US5376633A (en) 1992-09-30 1994-12-27 Lezdey; John Method for deactivating viruses in blood component containers
EP0677099B1 (en) 1992-12-31 2005-11-30 Dana-Farber Cancer Institute, Inc. Enhancer sequence for modulating expression in epithelial cells
US5604201A (en) 1993-01-08 1997-02-18 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education On Behalf Of The Oregon Health Sciences University, A Non-Profit Organization Methods and reagents for inhibiting furin endoprotease
JP2768557B2 (en) 1993-05-18 1998-06-25 コリア・インスティチュート・オブ・サイエンス・アンド・テクノロジー Heat resistant α1-antitrypsin mutein
US5750506A (en) 1993-06-18 1998-05-12 Cortech, Inc. Bradykinin antagonists with extended hydrophobic side chains
US5346886A (en) * 1993-11-15 1994-09-13 John Lezdey Topical α-1-antitrypsin, non-aqueous lipid miscible, benzalkonium chloride compositions for treating skin
US5759548A (en) 1993-11-30 1998-06-02 Lxr Biotechnology Inc. Compositions which inhibit apoptosis, methods of purifying the compositions and uses thereof
DE69409471T2 (en) 1993-12-03 1998-08-27 Ono Pharmaceutical Co Amidinophenol derivatives with phospholipase A2 inhibitory activity
AU696429B2 (en) 1994-03-09 1998-09-10 Cortech, Inc. Bradykinin antagonist peptides incorporating n-substituted glycines
EP0764151A2 (en) 1994-06-10 1997-03-26 Universitaire Instelling Antwerpen Purification of serine protease and synthetic inhibitors thereof
US5486470A (en) 1994-07-21 1996-01-23 Merck & Co., Inc. Purified herpes simplex virus protease and methods of purification
US5610285A (en) 1994-08-24 1997-03-11 Bayer Corporation Purification of α-1 proteinase inhibitor using novel chromatographic separation conditions
US5514653A (en) 1994-09-09 1996-05-07 Washington University Method of blocking the SEC receptor
JPH10506390A (en) * 1994-09-23 1998-06-23 アリス ファーマシューティカル コーポレイション Compositions and methods for treating mast cell inflammatory conditions
US6090916A (en) 1994-10-18 2000-07-18 Corvas International, Inc. Nematode-extracted serine protease inhibitors and anticoagulant proteins
US5498616A (en) 1994-11-04 1996-03-12 Cephalon, Inc. Cysteine protease and serine protease inhibitors
US5618792A (en) 1994-11-21 1997-04-08 Cortech, Inc. Substituted heterocyclic compounds useful as inhibitors of (serine proteases) human neutrophil elastase
US5780009A (en) 1995-01-20 1998-07-14 Nexia Biotechnologies, Inc. Direct gene transfer into the ruminant mammary gland
US5712117A (en) 1995-02-08 1998-01-27 Zymogenetics, Inc. Cytoplasmic antiproteinase-2 and coding sequences
US5798442A (en) 1995-04-21 1998-08-25 Merck Frosst Canada, Inc. Peptidyl derivatives as inhibitors of pro-apoptotic cysteine proteinases
US6255091B1 (en) 1995-04-28 2001-07-03 Axys Pharmaceuticals, Inc. Potentiating metal mediated serine protease inhibitors with cobalt or zinc ions
AU6458396A (en) 1995-07-17 1997-02-18 Cephalon, Inc. Phosphorous-containing cysteine and serine protease inhibitors
US5834431A (en) 1995-09-08 1998-11-10 Cortech, Inc. Des-Arg9 -BK antagonists
US5849863A (en) 1995-09-08 1998-12-15 University Of Colorado Cytolytic bradykinin antagonists
US5811241A (en) 1995-09-13 1998-09-22 Cortech, Inc. Method for preparing and identifying N-substitued 1,4-piperazines and N-substituted 1,4-piperazinediones
WO1997010231A1 (en) 1995-09-14 1997-03-20 Cephalon, Inc. Ketomethylene group-containing cysteine and serine protease inhibitors
ES2293651T3 (en) 1995-11-28 2008-03-16 Cephalon, Inc. INHIBITORS OF CYSTEINE AND SERINE PROTEASES DERIVED FROM AMINO ACIDS D.
WO1997024339A1 (en) 1995-12-27 1997-07-10 Ono Pharmaceutical Co., Ltd. Tetrazole derivatives and drugs containing the same as the active ingredient
TW442452B (en) 1996-03-01 2001-06-23 Akzo Nobel Nv Serine protease inhibitors having an alkynylamino side chain
DE69735996T2 (en) 1996-03-11 2007-02-15 Bayer Corp. HUMAN BIKININE
WO1997037969A1 (en) 1996-04-10 1997-10-16 Ono Pharmaceutical Co., Ltd. Tryptase inhibitor and novel guanidino derivatives
EP0915086A4 (en) 1996-05-24 2001-01-17 Ono Pharmaceutical Co Phenylsulfonamide derivatives
US5780440A (en) * 1996-06-17 1998-07-14 Protease Sciences Inc. Treatment of pulmonary disease with protease inhibitors
WO1997048706A1 (en) 1996-06-18 1997-12-24 Warner-Lambert Company Pyrrolo[1,2-a]pyrazine-1,4-dione serine protease inhibitors
KR20000022532A (en) 1996-06-27 2000-04-25 오노 야꾸힝 고교 가부시키가이샤 Aryl (sulfide, sulfoxide and sulfone) derivatives and drugs containing the same as the active ingredient
US5616693A (en) 1996-07-01 1997-04-01 Alpha Therapeutic Corporation Process for seperating alpha-1-proteinase inhibitor from COHN IV1 +1V4 paste
WO1998006417A1 (en) 1996-08-16 1998-02-19 Cortech, Inc. Methods to treat cerebral ischemic injury and other neuronal diseases
JP3447749B2 (en) 1996-08-29 2003-09-16 富士通株式会社 An equipment failure diagnosis method and apparatus, and a recording medium storing a program for causing a computer to execute processing according to the method
US6372887B1 (en) 1996-11-07 2002-04-16 Heska Corporation Flea serine protease inhibitor proteins
JP2002514192A (en) 1996-11-13 2002-05-14 セフアロン・インコーポレーテツド Cysteine and serine protease inhibitors containing benzothiazo and related heterocyclic groups
US6180402B1 (en) 1996-11-20 2001-01-30 Qlt Inc. Method for inhibiting apoptosis induced by photodynamic therapy using a cysteine or serine protease inhibitor
TW499412B (en) 1996-11-26 2002-08-21 Dimensional Pharm Inc Aminoguanidines and alkoxyguanidines as protease inhibitors
RU2217436C2 (en) * 1996-12-06 2003-11-27 Кортеч, Инк. Derivatives of ox diazole, thiadiazole or triazole as inhibitors of serine proteases and pharmaceutical compositions containing thereof
WO1998046597A1 (en) * 1997-04-14 1998-10-22 Emory University Serine protease inhibitors
US6004933A (en) 1997-04-25 1999-12-21 Cortech Inc. Cysteine protease inhibitors
PT979240E (en) 1997-05-02 2004-08-31 Akzo Nobel Nv SERINE-PROTEASE INHIBITORS
HUP0002951A3 (en) 1997-05-08 2002-10-28 Smithkline Beecham Corp Bis-aminomethylcarbonyl compounds as protease inhibitors and pharmaceutical compositions containing them
AT407114B (en) 1997-06-10 2000-12-27 Immuno Ag ALPHA 1-ANTITRYPSIN PREPARATION AND METHOD FOR THE PRODUCTION THEREOF
US6124257A (en) * 1997-08-28 2000-09-26 Lezdey; John Method of treatment
EP1078917A4 (en) 1998-02-17 2002-11-06 Ono Pharmaceutical Co Amidino derivatives and drugs containing the same as the active ingredient
WO1999043308A2 (en) 1998-02-27 1999-09-02 Marlene Rabinovitch Treating pulmonary hypertension through tenascin suppression and elastase inhibition
DE19828113A1 (en) * 1998-06-24 2000-01-05 Probiodrug Ges Fuer Arzneim Prodrugs of Dipeptidyl Peptidase IV Inhibitors
EP1150699A4 (en) * 1999-02-01 2004-06-30 John Lezdey Treatment of bladder and gastrointestinal mastocytosis
AU3719100A (en) 1999-03-05 2000-09-21 Trustees Of University Technology Corporation, The Inhibitors of serine protease activity, methods and compositions for treatment of viral infections
US6110949A (en) * 1999-06-24 2000-08-29 Novartis Ag N-(substituted glycyl)-4-cyanothiazolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
ES2264937T3 (en) * 1999-07-30 2007-02-01 Boehringer Ingelheim Pharmaceuticals Inc. NEW COMPOUNDS DERIVED FROM SUCCINATE, USEFUL AS INHIBITORS OF CISTEINA-PROTEASA.
US6420364B1 (en) * 1999-09-13 2002-07-16 Boehringer Ingelheim Pharmaceuticals, Inc. Compound useful as reversible inhibitors of cysteine proteases
US20040220242A1 (en) * 2003-05-02 2004-11-04 Leland Shapiro Inhibitors of serine protease activity, methods and compositions for treatment of nitric oxide induced clinical conditions

Patent Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6342373B1 (en) * 1983-11-21 2002-01-29 Ucp Gen-Pharma Ag Process for preparing recombinant eglin, protease inhibitor
US5508031A (en) * 1986-11-21 1996-04-16 Cetus Oncology Corporation Method for treating biological damage using a free-radial scavenger and interleukin-2
US5667776A (en) * 1986-11-21 1997-09-16 Chiron Corporation Treatment for biological damage using tumor necrosis factor and a free-radical scavenger
US5702697A (en) * 1986-11-21 1997-12-30 Chiron Corporation Treatment for biological damage using a colony stimulating factor and a biological modifier
US5114917A (en) * 1986-12-24 1992-05-19 John Lezdey Treatment of inflammation using alpha 1-antichymotrypsin
US5187089A (en) * 1990-06-21 1993-02-16 Incyte Pharmaceuticals, Inc. Protease nexin-i variants which inhibit elastase
US6008196A (en) * 1994-06-02 1999-12-28 Hoechst Marion Roussel, Inc. Perfluoroalkyl ketone inhibitors of elastase and processes for making the same
US5652237A (en) * 1994-09-09 1997-07-29 Warner-Lambert Company 2-substituted-4H-3, 1-benzoxazin-4-ones and benzthiazin-4-ones as inhibitors of complement C1r protease for the treatment of inflammatory processes
US6126933A (en) * 1995-06-27 2000-10-03 Genetics Institute Methods of treating inflammatory bowel diseases by administering IL-11
US5872124A (en) * 1996-07-31 1999-02-16 Thomas Jefferson University Treatment of diseases of the central nervous system using uric acid as a scavenger of peroxynitrite
US6271436B1 (en) * 1996-10-11 2001-08-07 The Texas A & M University System Cells and methods for the generation of transgenic pigs
US5928883A (en) * 1996-11-13 1999-07-27 Mayo Foundation For Medical Education Eosinophil granole proteins as indicators of inflammatory bowel disorders
US5851987A (en) * 1997-04-14 1998-12-22 Incyte Pharmaceuticals, Inc. Human tumor-associated Kazal inhibitor-like polypeptides and encoding polynucleotides
US6303629B1 (en) * 1997-05-13 2001-10-16 Octamer, Inc. Methods for treating inflammation, inflammatory diseases, arthritis and stroke using pADPRT inhibitors
US6323219B1 (en) * 1998-04-02 2001-11-27 Ortho-Mcneil Pharmaceutical, Inc. Methods for treating immunomediated inflammatory disorders
US6653350B1 (en) * 1998-09-08 2003-11-25 G.D. Searle & Co. Methods of treating osteoarthritis with inducible nitric oxide synthase inhibitors
US8071551B2 (en) * 1999-03-05 2011-12-06 BioHolding, Inc. Methods and compositions for treating diabetes
US6489308B1 (en) * 1999-03-05 2002-12-03 Trustees Of University Of Technology Corporation Inhibitors of serine protease activity, methods and compositions for treatment of nitric-oxide-induced clinical conditions
US7807781B2 (en) * 1999-03-05 2010-10-05 The Regents Of The University Of Colorado Inhibitors of serine protease activity and their use in methods and compositions for treatment of viral infections
US6849605B1 (en) * 1999-03-05 2005-02-01 The Trustees Of University Technology Corporation Inhibitors of serine protease activity, methods and compositions for treatment of viral infections
US20090298747A1 (en) * 1999-03-05 2009-12-03 Bio Holding, Inc. Methods and Compositions For Treatment of Nitric Oxide-Induced Clinical Conditions
US7704958B1 (en) * 1999-03-05 2010-04-27 Bio Holding, Inc. Methods and compositions for inhibiting apoptosis using serine protease inhibitors
US20120040913A1 (en) * 1999-03-05 2012-02-16 Leland Shapiro Methods and compositions for treating diabetes
US7850970B2 (en) * 2003-08-26 2010-12-14 The Regents Of The University Of Colorado Inhibitors of serine protease activity and their use in methods and compositions for treatment of bacterial infections
US20120071545A1 (en) * 2003-08-26 2012-03-22 Leland Shapiro Compositions and methods for treatment of bacterial and mycobacterial infections
US20090220518A1 (en) * 2005-06-07 2009-09-03 Regents Of The University Of Colorado Methods and compositions for treatment of graft rejection and promotion of graft survival
US20090118162A1 (en) * 2005-06-07 2009-05-07 Regents Of The University Of Colorado Inhibitors of serine protease activity and their use in methods and compositions for treatment of graft rejection and promotion of graft survival
US20120045417A1 (en) * 2005-06-07 2012-02-23 Lewis Eli C Methods and compositions for islet cell preservation
US20120045460A1 (en) * 2005-06-07 2012-02-23 Lewis Eli C Methods and compositions for treatment of graft rejection and promotion of graft survival
US20090203580A1 (en) * 2005-06-07 2009-08-13 Dinarello Charles A Compositions and methods of use for alpha-1 antitrypsin having no significant serine protease inhibitor activity
US7879846B2 (en) * 2006-09-21 2011-02-01 Kyorin Pharmaceutical Co.., Ltd. Serine hydrolase inhibitors
US20080261869A1 (en) * 2007-04-20 2008-10-23 Leland Shapiro Compositions and methods of use for alpha-1 antitrypsin having no significant serine protease inhibitor activity
US20110201587A1 (en) * 2010-02-16 2011-08-18 Bio Holding, Inc. Hsp90 inhibitors and methods of use
US20120225813A1 (en) * 2010-06-11 2012-09-06 Lewis Eli C Compositions, methods and uses for treatment of type 1 diabetes

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
Adeyemi, E. O., et al., 1992, "Faecal elastase reflects disease activity in active ulcerative colitis", Scandinavian Journal of Gastroenterology, Vol. 27, No. 2, pages 138-142 *
Al-Bekairi, A. M., et al., 1992, "A STUDY OF URIC ACID PRETREATMENT FOR THE PROTECTION OF RAT GASTRIC MUCOSA AGAINST TOXIC DAMAGE", Food and Chemical Toxicology, Vol. 30, No. 6, pages 525-531. *
Arndt, B., et al., 1994, "Fecal parameters for assessing the activity of Crohn's disease", Klinisches Labor, Vol. 40, No. 1-2, pages 51-60. *
Hooper, D. C., et al., 1997, "Prevention of experimental allergic encephalomyelitis by targeting nitric oxide and peroxynitrite: Implications for the treatment of multiple sclerosis", Proceedings of the National Academy of Sciences, U.S.A., Vol. 94, No. 6, pages 2528-2533 *
Hooper, D. C., et al., 1998, "Uric acid, a natural scavenger of peroxynitrite, in experimental allergic encephalomyelitis and multiple sclerosis", Proceedings of the National Academy of Sciences, U.S.A., Vol. 95, No. 2, pages 675-680. *
Kastenbauer, S., et al., 1999, "Role of peroxynitrite as a mediator of pathophysiological alterations in experimental pneumococcal meningitis", Journal of Infectious Diseases, Vol. 180, No. 4, pages 1164-1170. *
Lichtenstein, G. R., 1992, "Clinical advances in inflammatory bowel disease", Current Opinion in Gastroenterology, Vol. 8, No. 4, pages 655-662. *
Skinner, K.A., et al., 1998, "Nitrosation of uric acid by peroxynitrite: Formation of a vasoactive nitric oxide donor", The Journal of Biological Chemistry, Vol. 273, No. 38, pages 24491-24497. *
Smith, R.C., et al., 1987, "ANTIOXIDANT PROPERTIES OF 2 IMIDAZOLONES AND 2 IMIDAZOLETHIONES", Biochemical Pharmacology, Vol. 36, No. 9, pages 1457-1460 *
Stadnicki, A., et al., 1997, "Hemostasis in inflammatory bowel disease: current data and literature review", Pages 137-145 in Cytokines, Cholera, and the Gut, [Papers from the Joint Meeting of the United States-Japan Cooperative Medical Sciences Program Panels on Malnutrition and Cholera], 1995, Editors: Keusch, G.T., et al., IOS Press, Amsterdam. *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090298747A1 (en) * 1999-03-05 2009-12-03 Bio Holding, Inc. Methods and Compositions For Treatment of Nitric Oxide-Induced Clinical Conditions
US10531655B2 (en) 2011-12-02 2020-01-14 The Regents Of The University Of California Reperfusion protection solution and uses thereof
US10300119B2 (en) 2013-05-15 2019-05-28 Mor Research Applications Ltd. Compositions and methods for preventing injury during or resulting from cardiac surgery
US10617745B2 (en) 2013-05-15 2020-04-14 Mor Research Applications Ltd. Methods for preventing post-operative complications of cardiopulmonary surgery
US10980866B2 (en) 2014-09-22 2021-04-20 Hadasit Medical Research Services And Development Ltd. Alpha-1 anti-trypsin for treating liver diseases

Also Published As

Publication number Publication date
AU3731400A (en) 2000-09-21
US7704958B1 (en) 2010-04-27
US20080261868A1 (en) 2008-10-23
WO2000051624A2 (en) 2000-09-08
US8071551B2 (en) 2011-12-06
WO2000051624A3 (en) 2000-12-28
US20120040913A1 (en) 2012-02-16

Similar Documents

Publication Publication Date Title
US8071551B2 (en) Methods and compositions for treating diabetes
Meriin et al. Role of molecular chaperones in neurodegenerative disorders
Powell The ubiquitin-proteasome system in cardiac physiology and pathology
Carragher Calpain inhibition: a therapeutic strategy targeting multiple disease states
US7795298B2 (en) IAP BIR domain binding compounds
DE19834591A1 (en) Use of substances that decrease the activity of dipeptidyl peptidase IV to increase blood sugar levels, e.g. for treating hypoglycemia
US20050222387A1 (en) Smac-peptides as therapeutics against cancer and autoimmune diseases
US20200222516A1 (en) Compositions and Methods for Modulating Cardiac Conditions
CA2138124A1 (en) Use of calpain inhibitors in the inhibition and treatment of medical conditions associated with increased calpain activity
US7374898B2 (en) Peptide inhibitors against seprase
US20090227521A1 (en) Use of compounds in the treatment of ischemia and neurodegeneration
WO2003086470A2 (en) Smac-peptides as therapeutics against cancer and autoimmune diseases
ES2621672T3 (en) 1- [1- (Benzoyl) -pyrrolidin-2-carbonyl] -pyrrolidin-2-carbonitrile derivatives
KR20000016700A (en) Depsipeptides and drugs containing the same as the active ingredient
US20100040607A1 (en) Combination Therapy with Inhibitors of HMGB and Caspase for the Treatment of Inflammatory Diseases
US20030133927A1 (en) Conjugates useful in the treatment of prostate cancer
EP1354952A1 (en) Smac-peptides as therapeutics against cancer and autoimmune diseases
US20200172574A1 (en) Inhibition of cardiac fibrosis in myocardial infarction
JP5996190B2 (en) Methods, systems and compositions for calpain inhibition
Powell Running Title: The ubiquitin-proteasome system in heart.
Chao et al. Kallikrein Gene Transfer in Hypertension, Cardiovascular and Renal Disease, and Stroke
Chao et al. THE TISSUE KALLIKREIN-KININ SYSTEM COMPONENTS TISSUE KALLIKREIN IN HYPERTENSION SOMATIC GENE DELIVERY OF TISSUE KALLIKREIN IN HYPERTENSIVE ANIMAL MODELS KALLIKREIN/KININ AND CARDIAC PROTECTION

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION