WO2009076665A1 - Materials and methods for inhibiting mammalian s-nitrosoglutathione reductase - Google Patents

Materials and methods for inhibiting mammalian s-nitrosoglutathione reductase Download PDF

Info

Publication number
WO2009076665A1
WO2009076665A1 PCT/US2008/086738 US2008086738W WO2009076665A1 WO 2009076665 A1 WO2009076665 A1 WO 2009076665A1 US 2008086738 W US2008086738 W US 2008086738W WO 2009076665 A1 WO2009076665 A1 WO 2009076665A1
Authority
WO
WIPO (PCT)
Prior art keywords
disease
compound
gsnor
systemic
compounds
Prior art date
Application number
PCT/US2008/086738
Other languages
French (fr)
Inventor
Paresh Sanghani
Original Assignee
Indiana University Research And Technology Corporation
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 Indiana University Research And Technology Corporation filed Critical Indiana University Research And Technology Corporation
Priority to JP2010538218A priority Critical patent/JP2011507811A/en
Priority to EP08859998A priority patent/EP2229451A4/en
Publication of WO2009076665A1 publication Critical patent/WO2009076665A1/en
Priority to US12/813,721 priority patent/US9198909B1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4021-aryl substituted, e.g. piretanide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • 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
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/902Oxidoreductases (1.)
    • G01N2333/904Oxidoreductases (1.) acting on CHOH groups as donors, e.g. glucose oxidase, lactate dehydrogenase (1.1)

Definitions

  • GSNOR S-nitrosoglutathione reductase
  • SNOs S-nitrosothiols
  • SNO concentrations range from nM to ⁇ M concentrations
  • thiol S- nitrosylation and NO transferase reactions are involved in virtually all classes of cell signaling, ranging from regulation of ion channels and G-protein coupled reactions, to reception stimulation and activation of nuclear regulatory proteins.
  • Regulatory signaling exists for the synthesis, transport, activation and metabolism of SNOs. (Gaston BM, Carver J, Doctor A, Palmer LA. S-nitrosylation signaling in cell biology.
  • S-nitrosoglutathione one of the major forms of SNO that is formed endogenously, is catabolized by the enzyme S-nitrosoglutathione reductase (GSNOR).
  • SNOR S-nitrosoglutathione reductase
  • the regulation of GSNOR activity is believed to be one of the mechanisms by which cellular NO is regulated.
  • GSNOR catalyzes the denitrosylation of intracellular proteins by the reduction of s- nitrosoglutathione (GSNO).
  • GSNOR Because of its role in the regulation of the s-nitrosylation of intracellular proteins, GSNOR has become an important target for the development of new therapeutic agents targeting this enzyme, since inhibition of the activity of GSNOR should increase NO concentrations, which in turn should lead to clinically significant changes in end-organ function.
  • GSNOR GSNOR's role in the modulation of GSNO concentrations, which in turn alters cellular concentrations of NO
  • therapeutic compounds that inhibit GSNOR activity have the potential to treat a wide variety of diseases whose common pathophysiology is abnormally low NO concentrations.
  • GSNOR inhibitors could lead to increased concentrations of GSNO and NO and have the potential to impact these diseases.
  • Nitric oxide and the s-nitrosylation/de-nitrosylation cycle play an essential role in many pathologic disease states due largely to dysfunction of endothelial cells.
  • Various vascular disorders such as heart disease, heart failure, heart attack, hypertension, atherosclerosis, and restenosis can be directly linked to abnormal NO activity and/or concentrations, and subsequent changes in the s-nitrosylation of target proteins.
  • Other conditions such as asthma and impotence are also linked to abnormal levels of NO bioactivity (Gaston BM, Carver J, Doctor A, Palmer LA. S-nitrosylation signaling in cell biology. MoI Interv 2003,3:253-63; brieflyig MT, Kojda G.
  • Knockout mice lacking the genes for GSNOR were found to resist airway hyperresponsivity due to higher GSNO concentrations in bronchial fluids and demonstrated diminished tachyphylaxis to ⁇ - agonists because of the s-nitrosylation of G-protein coupled receptor kinases (Que LG, Liu L, Yan Y, et al. Protection from experimental asthma by an endogenous bronchodilator. Science 2005;308: 1618-21). Cystic fibrosis patients have also been shown to have abnormally low levels of GSNO which is thought to play a role in the pathophysiology of this disease; therapies that inhibit GSNOR may offer these patients a new therapeutic option. (Zeitlin PL.
  • One aspect of the invention is a compound and/or a method for using a compound to alter enzyme activity, comprising the steps of: providing at least one compound selected from the group consisting of:
  • R ._ H CH 3 , F, Cl, Br, OCH 3 , C -CH-.
  • R 2 -H, -Cl, -F.. -Br
  • R 3 -C 2 H 5 , -C 4 H 9 , ⁇ J
  • R 4 -OC 2 H 5 , -OCH 3 , -C 2 H 5 , -F, -Cl, -Br, -OCH 3 , -COOC 2 H 5
  • R 1 -H, -CH 3 , -C 2 H 5 , -C 3 H 7
  • R 2 -H, -CN, -OH 3 -CH 2 OH .
  • R 1 -H, -CH 3 , -C 2 H 5 , -C 3 H 7
  • One embodiment is a method for treating a disease or a condition, comprising the steps of: providing at least one compound according to any of the compounds provided herein including those referred to above, and administering a therapeutically effective dose of said compound to a patient in need thereof.
  • Still another embodiment is a method for treating a disease as outlined above, wherein the disease or condition is selected from the group consisting of: Cardiovascular Diseases, including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease; Pulmonary Diseases, including but not limited to asthma, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis; Urogenital Diseases, including but not limited to impotence and abnormalities of ejaculation.
  • Cardiovascular Diseases including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure
  • Inherited genetic diseases including but not limited to cystic fibrosis and sickle cell anemia; Neurologic Diseases, including but not limited to Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS); Inflammatory Diseases, including but not limited to systemic inflammatory response syndrome, rheumatoid arthritis, Sjogren's syndrome, ulcerative colitis, Crohn's disease, glomerulonephritis, psoriasis, cutaneous lupus erythematosus, systemic lupus erythematosus, systemic sclerosis, dermatitis, and other inflammatory diseases; Oncologic Diseases, including both hematologic and solid tumors. In one embodiment the therapeutically effective amount of said compound in on the range of about 0.01 mg/kg per of body mass day to 1000 mg/kg of body mass per day.
  • Yet another embodiment is a method for diagnosing or studying a disease or a condition, comprising the steps of: providing at least one compound as disclosed above and contacting the compound with s-nitrosoglutathione reductase; an observing a change in conformity with a change in s-nitrosylation.
  • the disease or condition being diagnosed is selected from the group consisting of: Cardiovascular Diseases, including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease; Pulmonary Diseases, including but not limited to asthma, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis; Urogenital Diseases, including but not limited to impotence and abnormalities of ejaculation.
  • Cardiovascular Diseases including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome
  • Inherited genetic diseases including but not limited to cystic fibrosis and sickle cell anemia; Neurologic Diseases, including but not limited to Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS); Inflammatory Diseases, including but not limited to systemic inflammatory response syndrome, rheumatoid arthritis, Sjogren's syndrome, ulcerative colitis, Crohn's disease, glomerulonephritis, psoriasis, cutaneous lupus erythematosus, systemic lupus erythematosus, systemic sclerosis, dermatitis, and other inflammatory diseases; Oncologic Diseases, including both hematologic and solid tumors.
  • Still another embodiment is a method of studying protein nitrosylation comprising the steps of: providing at least one compound according disclosed herein, including those name in the above and contacting said compound with s-nitrosoglutathione reductase; and observing a change in s-nitrosylation.
  • Another embodiment provides a kit for altering the activity of s- nitrosoglutathione reductase comprising at least one compound named in the above.
  • Still another embodiment provides a method for treating a disease or condition comprising the steps of providing at least one compound selected from the group consisting of:
  • the disease or condition is selected from the group consisting of: Cardiovascular diseases, including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease; Pulmonary Diseases, including but not limited to asthma, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis; Urogenital Diseases, including but not limited to impotence and abnormalities of ejaculation; Inherited Genetic Diseases, including but not limited to cystic fibrosis and sickle cell anemia; Neurologic Diseases, including but not limited to Parkinson's
  • Still another embodiment provides a method for diagnosing or studying a disease, condition or chemical and/or enzymatic reaction including the steps of providing at least one compound selected from the group consisting of: NC' ⁇ , or a pharmaceutically acceptable or ester thereof; contacting said compound with s-nitrosoglutathione reductase; and observing a change in conformity with a change in s-nitrosylation.
  • the disease and/or condition being diagnosed and/or studied is selected from the group consisting of: Cardiovascular Diseases, including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease; Pulmonary Diseases, including but not limited to asthma, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis; Urogenital Diseases, including but not limited to impotence and abnormalities of ejaculation.
  • Cardiovascular Diseases including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, s
  • Inherited genetic diseases including but not limited to cystic fibrosis and sickle cell anemia; Neurologic Diseases, including but not limited to Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS); Inflammatory Diseases, including but not limited to systemic inflammatory response syndrome, rheumatoid arthritis, Sjogren's syndrome, ulcerative colitis, Crohn's disease, glomerulonephritis, psoriasis, cutaneous lupus erythematosus, systemic lupus erythematosus, systemic sclerosis, dermatitis, and other inflammatory diseases; Oncologic Diseases, including both hematologic and solid tumors. Still another embodiment is a kit for altering the activity of s-nitrosoglutathione reductase, comprising at least one of the compounds described in the above.
  • One aspect of the present invention provides compounds that can be used to inhibit the enzyme S-nitrosoglutathione reductase (GSNOR). These compounds include Compound A, Compound B, Compound C and Compound D defined below, as well as all physiologically acceptable salts thereof.
  • GSNOR S-nitrosoglutathione reductase
  • One embodiment is a method of treating human or animal patients comprising the steps of providing at least one compound selected from the group comprising Compound A, Compound B, Compound C and Compound D or a physiological salt thereof and administering a therapeutically effective dose of the compound to a human or animal patient.
  • a method of diagnosing a disease in a human or an animal comprising the steps of providing at least one compound, selected from the group comprising Compound A, Compound B, Compound C and Compound D or a physiological salt thereof and contacting said compound with at least one enzyme in the human or animal.
  • Yet another embodiment is a method of studying the mechanism, chemistry or role in physiology of the enzyme s-nitrosoglutathione reductase (GSNOR) either in vitro or in vivo comprising the steps of providing at least one compound selected from the group comprising Compound A, Compound B, Compound C and Compound D or a physiological salt thereof and contacting said compound with the enzyme s-nitrosoglutathione reductase (GSNOR).
  • GSNOR s-nitrosoglutathione reductase
  • One embodiment is a method of treating, diagnosing, curing, controlling or otherwise affecting a disease, defect or other medical condition treated by administering a therapeutically effective dose of at least one of the following compounds selected from the group including Compound A, Compound B, Compound C and Compound D or a physiological salt thereof.
  • condition affected is selected from the group comprising the activation of elements of the immune system including, but not limited to, macrophages, thymocytes, lymphocytes, or intercellular networks that involve NO signaling networks, cellular processes such as apoptosis, the activity of endothelial cells, cardiovascular disorders, including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease.
  • elements of the immune system including, but not limited to, macrophages, thymocytes, lymphocytes, or intercellular networks that involve NO signaling networks, cellular processes such as apoptosis, the activity of endothelial cells, cardiovascular disorders, including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery
  • the condition affected is selected from pulmonary diseases including but not limited to asthma, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis.
  • pulmonary diseases including but not limited to asthma, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis.
  • the condition affected is selected from diseases of the urogenital system, including but not limited to impotence and abnormalities of ejaculation.
  • the condition affected is selected from inherited genetic diseases, including but not limited to cystic fibrosis and sickle cell anemia.
  • the condition affected is selected from neurologic diseases, including but not limited to Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS).
  • the condition affected is selected from inflammatory diseases, including but not limited to systemic inflammatory response syndrome, rheumatoid arthritis, Sjogren's syndrome, ulcerative colitis, Crohn's disease, glomerulonephritis, psoriasis, cutaneous lupus erythematosus, systemic lupus erythematosus, systemic sclerosis, dermatitis, and other inflammatory diseases.
  • the condition affected is selected from oncologic diseases, including both hematologic and solid tumors.
  • FIG. 1 is a diagram illustrating the Kinetic mechanism of GSNOR and the types of complexes that an inhibitor binding to the GSNO site could form along the kinetic pathway of GSNOR.
  • FIG. 2 illustrates the effect of compound 8 on the fluorescence of GSNOR bound
  • FIG. 3 illustrates data consistent with an increase in the accumulation of nitroso species in RAW 264.7 cells in presence of GSNOR inhibitors.
  • FIG. 4 illustrates the effects of Compound 8 on the nitrosylation of cellular proteins.
  • FIG. 5 illustrates a dose-response relationship of Compound 8 and sodium nitroprusside.
  • FIG. 6 illustrates that inhibition of GSNOR increases cGMP production.
  • Table 1 represents structures of some inhibitors of GSNOR.
  • Table 2 represents data illustrating inhibition of various alcohol dehydrogenase isozymes by compounds that inhibit the enzyme GSNOR.
  • Table 3 represents a summary of kinetic data consistent with mechanism of GSNOR inhibition by various compounds.
  • Table 4 represents data summarizing kinetic contrasts measured with various exemplary compounds thought to inhibit GSNOR.
  • GSNOR is generally categorized as a member of the alcohol dehydrogenase family of enzymes; its physiological substrates are thought to be nitrosylated proteins, S-nitrosoglutathione (GSNO) and NADH. Nitrosylated intercellular proteins are often the result of NO bioactivity and these proteins account for much of NO's physiological effects. Accordingly, the denitrosylation reaction catalyzed by GSNOR helps healthy, normal cells to maintain a balance between nitrosylated and de-nitrosylated proteins and functions as an integral part of the cycle that modulates the effects of NO and its attendant effects.
  • the inventors have identified a number of compounds including substituted pyrroles, indoles, thiophenes and aromatic rings that affect the activity of GSNOR and therefore may be used to modulate NO's effect on intercellular proteins and on physiology.
  • One aspect includes molecules having the structure below generally referred to as Compound A and using the same to treat and/or diagnose various diseases and/or conditions or to study various enzyme catalyzed reactions; these molecules have the following generic structure and include pharmaceutically acceptable salts and/or esters thereof:
  • R 5 -H, -CH 3 , -C 2 H 5 , -C 3 H 7
  • Another aspect includes molecules having the structure generally referred to as Compound B and using the same to treat and/or diagnose various diseases and/or conditions or to study various enzyme catalyzed reactions; these molecules have the following generic structure and include pharmaceutically acceptable salts and/or esters thereof:
  • R 2 -H, -Cl, -F, -Br
  • R 3 -C 2 H 5 , -C 4 H 9 , -Qf
  • R 4 -OC 2 H 5 , -OCH 3 , -C 2 H 5 , -F, -Cl, -Br, -OCH 3 , -COOC 2 H 3
  • Still another aspect includes molecules having the structure generally referred to as Compound C and using the same to treat and/or diagnose various diseases and/or conditions or to study various enzyme catalyzed reactions; these molecules have the following generic structure and include pharmaceutically acceptable salts and/or esters thereof:
  • R 2 -H, -CN, -OH, -CH 2 OH
  • Yet another aspect includes molecules having the structure generally referred to as Compound D and using the same to treat and/or diagnose various diseases and/or conditions or to study various enzyme catalyzed reactions; these molecules have the following generic structure and include pharmaceutically acceptable salts and/or esters thereof:
  • the compounds useful in the invention may be delivered not only as single agents by oral, inhalation or parenteral routes, but also in the form of cocktails which are mixtures of other appropriate compounds to treat a particular disease.
  • cocktail in the treatment of asthma, cardiovascular disease and other diseases treated by the invention is routine.
  • a common administration vehicle e.g., pill, tablet, implant, injectable solution, etc.
  • a therapeutically effective amount will be determined by the parameters discussed below; but it is that amount which establishes a level of the drug(s) in the area of the blood stream or tissue to be treated, such as the lung or vascular smooth muscle, which is effective in causing a therapeutic benefit.
  • the formulations of the invention are applied in pharmaceutically acceptable amounts and in pharmaceutically acceptable compositions.
  • Such preparations may routinely contain salts, buffering agents, preservatives, compatible carriers, and optionally other therapeutic ingredients.
  • the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically acceptable salts thereof and are not excluded from the scope of the invention.
  • Such pharmacologically and pharmaceutically acceptable salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulfonic, tartaric, citric, methane sulfonic, formic, malonic, succinic, naphthalene-2-sulfonic, and benzene sulfonic.
  • pharmaceutically acceptable salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts.
  • included along with the Compounds A, B, C, and D are their pharmaceutically acceptable salts, including base addition salts.
  • pharmaceutically-acceptable salts includes salts commonly used to form alkali metal salts and to form addition salts of free bases. The nature of the salt is not critical, provided that it is pharmaceutically acceptable.
  • Suitable pharmaceutically-acceptable base addition salts of the compounds of the invention may be prepared from an inorganic base or an organic base.
  • Suitable pharmaceutically-acceptable base addition salts include metallic salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts made from N 5 N'- dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N- methylglucamine, and procaine. All of these salts may be prepared by conventional means from the corresponding compounds of the invention by reacting, for example, the appropriate base with one of the compounds of the invention.
  • esters of compounds of the invention are also included in the invention. These esters may be prepared by an acid catalyzed reaction between the compounds of the invention and an alcohol like methanol, ethanol, isopropyl, butanol and other alkyl and aryl alcohols.
  • Suitable buffering agents include, for example, acetic acid and a salt (1-2% W/V); citric acid and a salt (1-3% W/V); boric acid and a salt (0.5-2.5% W/V); and phosphoric acid and a salt (0.8-2% W/V).
  • Suitable preservatives include benzalkonium chloride (0.003-0.03% W/V); chlorobutanol (0.3-0.9% W/V); parabens (0.01-0.25% W/V) and thimerosal (0.004-0.02%
  • the invention is also directed to pharmaceutical compositions and/or formulations and uses for the same comprising, for example, at least one of the following Compound 6, 7 or 8 (Table 1) or the like and pharmaceutically acceptable salts and/or esters thereof and at least one pharmaceutically acceptable carrier or diluent, as well as methods for using the same to treat and or diagnose various diseases of conditions a well as methods for studying various chemical and biological processes.
  • the formulations of the present invention can be solutions, suspensions, syrups, tablets, capsules, and the like.
  • the compositions may contain a suitable carrier, diluent, or excipient, such as a medium chain triglyceride oil or magnesium stearate.
  • a medium chain triglyceride oil and magnesium stearate is present in an approximately 1 : 1 ratio. Standard texts, such as Remington's Pharmaceutical Science, 18th
  • One preferred carrier is polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • One more preferred is a mixture of polyethylene glycol having a high molecular weight, for example of greater than 900 (most preferably, about 1,000), and polyethylene glycol having a low molecular weight, for example of less than 500 preferably about 400).
  • One particularly preferred carrier is PEG in the ratio of about one part PEG with a MW of 100 to about two parts PEG with a MW of about 400.
  • Preferred emulsifiers include phosphatidylcholine emulsifiers, such as dilauroylphosphatidylcholine.
  • the formulations can include powdered carriers, such as lactose, sucrose, mannitol, starch, cellulose derivatives, xanthum gum, magnesium stearate, stearic acid, and the like.
  • the composition may also comprise a penetration enhancer. Suitable penetration enhancers include glycerol, glycerol monolaureate, dimethyl sulfoxide or oils, such as a mineral oil or medium chain triglyceride oil.
  • Antioxidants such as, for example, butylated hydroxytoluene (BHT), sodium bisulfate, sodium sulfite, sodium EDTA, ascorbic acid, and the like can be used either alone or in combination with other suitable antioxidants or stabilizing agents typically employed in pharmaceutical compositions.
  • BHT butylated hydroxytoluene
  • sodium bisulfate sodium bisulfate
  • sodium sulfite sodium EDTA
  • ascorbic acid ascorbic acid
  • the formulations can also include any of the commonly used disintegrants, lubricants, plasticizers, colorants, and dosing vehicles.
  • Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, a standard reference text in this field. Suitable formulations typically contain from about 1 to about 1000 mg of active ingredient per dosage unit. In these pharmaceutical compositions, the active ingredient will ordinarily be present in an amount of about 0.5 to about 95%, by weight, based on the total weight of the composition.
  • compositions of the invention may be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.
  • the active compounds of the present invention may be a pharmaceutical composition having a therapeutically effective amount of a conjugate of the invention optionally included in a pharmaceutically-acceptable carrier.
  • pharmaceutically-acceptable carrier means one or more compatible solid or liquid fillers, diluents, or encapsulating substances which are suitable for administration to a human or other animal.
  • carrier denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application.
  • the components of the pharmaceutical compositions are capable of being commingled with the molecules of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy.
  • compositions suitable for parenteral administration conveniently comprise a sterile preparation of the compounds of the invention.
  • This preparation may be formulated according to known methods.
  • the sterile preparation thus may be a sterile solution or suspension in a non-toxic parenterally-acceptable diluent or solvent.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono or di-glycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Carrier formulations suitable for oral, subcutaneous, intravenous, intramuscular, etc. can be found in, for example, Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa.
  • a subject or a patient may mean a human, primate, horse, cow, pig, sheep, goat, dog, cats, rodent and the like.
  • the term 'about' used in conjunction with a numerical value or range generally means plus or minus 20 percent; e.g. 'about 1.0' includes values from 0.8 to 1.2.
  • an effective amount is an amount necessary to delay the onset of, inhibit the progression of or halt altogether the onset of, or progression of, to diagnose the particular condition being treated.
  • an effective amount for treating asthma will be that amount necessary to open airways and decrease airway inflammation such that a therapeutic benefit results.
  • effective amounts will depend, of course, on the particular condition being treated; the severity of the condition; individual patient parameters including age, physical condition, size and weight; concurrent treatment; frequency of treatment; and the mode of administration. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is preferred generally that a maximum dose be used, that is, the highest safe dose according to sound medical judgment.
  • Dosage may be adjusted appropriately to achieve desired drug levels, locally or systemically.
  • daily oral doses of active compounds will be from about 0.01 mg/kg per day to 1000 mg/kg per day. It is expected that IV doses in the range of about 1 to 1000 mg/m 2 per day will be effective. In the event that the response in a subject is insufficient at such doses, even higher doses (or effective higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits. Continuous IV dosing over, for example 24 hours or multiple doses per day are also contemplated as necessary to achieve appropriate systemic levels of the compounds.
  • preferred dosing schedules including concentration, length of administration, and the like are described herein.
  • a variety of administration routes are available. The particular mode selected will depend, of course, upon the particular drug selected, the severity of the disease state being treated and the dosage required for therapeutic efficacy.
  • the methods of this invention may be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of the active compounds without causing clinically unacceptable adverse effects.
  • modes of administration include, for example, oral, rectal, sublingual, topical, nasal, transdermal, intradermal or parenteral routes.
  • parenteral includes subcutaneous, intravenous, intramuscular, or infusion. In some instances intravenous routes may be preferred.
  • the compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy.
  • compositions suitable for oral administration may be presented as discrete units such as capsules, cachets, tablets, or lozenges, each containing a predetermined amount of the active compound.
  • Other compositions may include, for example, suspensions in aqueous liquors or non-aqueous liquids such as a syrup, an elixir, or an emulsion.
  • Other delivery systems can include time-release, delayed release or sustained release delivery systems.
  • release delivery systems can avoid repeated administrations of the active compounds of the invention, increasing convenience to the subject and the physician.
  • release delivery systems include polymer based systems such as polylactic and polyglycolic acid, polyanhydrides and polycaprolactone; nonpolymer systems that are lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono, di and triglycerides; hydrogel release systems; silastic systems; peptide based systems; wax coatings, compressed tablets using conventional binders and excipients, partially fused implants and the like.
  • a pump-based hardware delivery system can be used, some of which are adapted for implantation.
  • a long-term sustained release implant also may be used.
  • Long-term release as used herein, may mean that the implant is constructed and arranged to deliver therapeutic levels of the active ingredient for at least 30 days, and in some instances 60 days or longer.
  • Long-term sustained release implants are well known to those of ordinary skill in the art and include some of the release systems described above. Such implants can be particularly useful in treating chronic medical conditions.
  • the compounds of the invention are also useful, in general, for treating disease or conditions including, but not limited to, Cardiovascular Diseases, including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease; Pulmonary Diseases, including but not limited to asthma, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis; Urogenital Diseases, including but not limited to impotence and abnormalities of ejaculation; Inherited Genetic Diseases, including but not limited to cystic fibrosis and sickle cell anemia; Neurologic Diseases, including but not limited to Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS
  • GSNOR is an alcohol dehydrogenases, and like other members of ADH family, is capable of oxidizing and reducing primary alcohols and aldehydes with the help of a molecule of NAD(H).
  • Octanol was chosen as the substrate in the initial screening assay as it is bulky like GSNO, but has 200-fold higher K M value (1 mM) and a k cat value (200 min "1 ) that is more than 2 order of magnitude lower than that of GSNO. This allows the initial velocity conditions to prevail for a period up to 10 min (data not shown) and it enables conducting a large number of concurrent assays in a 384 well plate format.
  • the pH of the assay was adjusted to pH 10 in order to obtain significant rate of oxidation of octanol in the absence of compounds.
  • the screening for GSNOR inhibitors was performed in the presence of, saturating concentration OfNAD + and an un-saturating concentration of octanol, thereby increasing the probability of identifying compounds that bind, only or primarily in the GSNO binding site of GSNOR. This was deemed desirable as there are many dehydrogenases inside the cells, and compounds binding in the coenzyme binding site are more likely to be nonspecific inhibitors of GSNOR than compounds that bind primarily to the form of GSNOR that binds GSNO.
  • GSNOR may have a preferred kinetic pathway (shown by the bold lines) through GSNOR'NADH complex (EA) in its random mechanism during the reduction of aldehyde.
  • EA GSNOR'NADH complex
  • the aldehyde (B) can bind to the free form of GSNOR (E), or preferentially to the GSNOR'NADH complex (EA) to form the competent ternary complex (EAB).
  • the EAB complex undergoes catalysis to form the products, NAD + (Q) and alcohol (P). After catalysis, either of the products can leave the enzyme.
  • a GSNO inhibitor can bind to the free form of GSNOR (step 1), GSNOR'NADH (step 2) and GSN0R » NAD + (step 3) binary complexes to form EI, EAI and EQI ternary complexes, respectively.
  • dodecanoic acid was found to be a noncompetitive inhibitor against varied GSNO concentration, even though it binds at the GSNO binding site.
  • the noncompetitive inhibition of GSNOR by dodecanoic acid can be explained on the basis of the kinetic mechanism of GSNOR (shown in Fig. 1) during the reduction of aldehyde, 12- oxododecanoic acid (12-ODDA).
  • GSNOR has a preferred kinetic pathway through the E'NADH complex during the reduction of 12-ODDA.
  • dodecanoic acid would act as a noncompetitive inhibitor against varied GSNO levels if it binds to GSNOR at more than one place in the kinetic pathway; e.g., one where it competes with GSNO to bind to the enzyme (steps 1 and 2 in Fig. 1) and one where GSNO does not normally bind in the kinetic pathway (step 3 in Fig. 1).
  • Competition with GSNO for binding to GSNOR would involve binding the inhibitor to the E « NADH complex (step 1) and to a small extent, to the free enzyme (step 2) and give rise to the slope effect closured in the double reciprocal plot of the kinetic data.
  • the fluorescence of NADH increases and shifts to a lower wavelength as NADH transfers from the polar environment in the solvent to the less polar environment within the GSNOR active site.
  • the addition of Compound 8 decreased the fluorescence of NADH but interestingly, there is a blue shift in the emission peak indicating that NADH is still in the nonpolar environment of the active site (compare curve a and c in Fig. 2a).
  • Such a quenching of the fluorescence of the dihydropyridine ring has been observed when the amide inhibitors bind to Horse liver ADH'NADH complex.
  • Compounds 6 and 7 also quenched the NADH fluorescence while moving the emission maxima to a shorter wavelength (data not shown).
  • RAW cells have been used extensively as a model system for examining NO and s-nitrosothiol biochemistry. Briefly, RAW cells were treated with the inhibitors alone or in combination with GSNO and the intracellular nitrosylated species were quantitated using the triiodide-based chemiluminescence method. Referring now to Fig. 3 A, no significant amounts of nitroso species were detected in untreated cells and cells exposed only to the compounds at concentrations used in these assays. A significant buildup of nitrosylated compounds was evident in cells treated with 500 ⁇ M GSNO.
  • nitrosylated compounds appeared to reach an equilibrium level within 1 hour of exposure to GSNO and remained fairly constant for the duration of 6 hours. In contrast, nitrosylated compounds continued to accumulate in cells treated with GSNO and 33 ⁇ M concentrations of either Compound 6 or 8. At 6 hours the levels of nitrosylated species inside the cells treated with GSNO and Compounds 6 or 8 were 3 to 4 fold higher than the level of nitrosylated species found in cells treated with GSNO alone. Still longer exposure of cells to Compounds 6 or 8 (for up to 24 hours) resulted in an 80% decrease in the amounts of nitrosylated species measured at 4 hours (data not shown).
  • Compound 7 (Table 1) was not as effective as Compounds 6 or 8 in inhibiting GSNOR inside the cells. This is evidenced by only a 1.3 to 1.7 fold increase in the levels of nitrosylated species and the shorter duration of its effect as judged from an insignificant difference in the levels of nitrosylated species measured at 6 hrs.
  • An analysis of the molecular size of the nitrosylated species inside the treated cells showed that more than 95% of the nitrosylated species were greater than 5 kDa in size.
  • Compound 8 (GSNORi in the figure) potentiates the effect of GSNO by 2.5 fold. Without being limited by any specific explanation or hypothesis, these results suggest that one mechanism by which Compound 8 may exert its biochemical effects is due to its inhibition of GSNOR, which then potentiates the effects of GSNO on cGMP production resulting in higher cGMP levels. These data may explain at a biochemical level the ability of at least some of these compounds that inhibit GSNOR to relax isolated aortic rings as demonstrated elsewhere herein. [0083] The increase in the nitrosylation of proteins upon GSNOR inhibition may suggest that GSNOR inhibition should increase the bioactivity of NO stemming from s-nitrosylation of cellular proteins in organs as well as in cells in tissue culture.
  • these teachings reveal novel inhibitors of s-nitrosoglutathione reductase and many related compounds that inhibit GSNOR perhaps by binding into the GSNO binding site. At least some of these compounds bind to GSNOR at multiple places in the kinetic pathway, thereby affording a type of inhibition not easily overcome by the up-regulation of GSNO and NADH. Data collected using these compounds support the assertion that GSNOR is one of the primary enzymes involved in regulating the nitrosylation of intracellular proteins. [0087] In addition to being structurally diverse, each of the Compounds 6-8 has a free carboxyl group like many of the excellent substrates of GSNOR, including GSNO and 12- hydroxydodecanoic acid.
  • Compoundsl2-14, 43-53, 72, 84, 86 and 72-83 are related to Compound 6 of Table 3; Compounds 24, 56, 58, 59, 62, 60-62, 64, 66, 67, 69, 70 in Table 4 and are related to Compound 7 of Table 1.
  • a number of explanations and experiments are provided by way of explanation and not by limitation. No theory of how the novel technology operates is to be considered limiting, whether proffered by virtue of description, comparison, explanation or example. Accordingly, the following examples and discussion are presented by way of guidance and explanation and not limitation.
  • the 1, 2- diarylpyrroles disclosed herein can be synthesized using the general methodology disclosed in Scheme I.
  • the general synthetic strategy entailed the preparation of suitable 1,4-diketones followed by heating with appropriate amines in the Paal-Knorr condensation, cyclization to yield the targets.
  • the analogs having an alkyl group (R 3 ), Me or Et) at position 5 in the pyrrole ring were synthesized following Scheme 1.
  • the Stetter reaction 16 of substituted benzaldehydes with R, ⁇ -unsaturated ketones using the thiazolium salt catalyst proved very versatile and high yielding (NEt 3 , EtOH, reflux, 60-90%).
  • RAW 264.7 cells were cultured in DMEM containing 10% heat-inactivated serum. Cells were treated with 33 ⁇ M of Compound 8 for 0, 2, 4, 8, or 24 h alone or in combination with 1 mM NAME for 4 h (lane 4+N). At indicated times, the cells were quenched and the lysate was analyzed for s-nitrosothiol content by the biotin switch assay. Equal amounts of proteins were loaded in each lane and the degree of biotinylation (and hence s-nitrosylation) determined using an anti-biotin antibody. [0096] Referring now to FIG.
  • mice aorta segments were equilibrated in oxygenated PSS (95% O 2 and 5% CO 2 ) at 37 0 C. Following equilibration, l ⁇ M phenylephrine was added to each ring for submaximal contraction. After stabilization, increasing concentrations (10 ""9 M to 10 "4 M) of either Compound 8 or sodium nitroprusside (SNP) was added to the rings and the tone of the rings determined.
  • SNP sodium nitroprusside
  • L-NAME 500 micromolar was added to the bath and allowed to incubate for 30 minutes. After 30 minutes, Compound 8 (50 micromolar) was added and the tone of the rings determined as described above. Each experiment was performed using two rings from three different mice and the mean ( ⁇ SEM) for each response determined.
  • the dissociation constant was measured at 25 0 C in 50 mM potassium phosphate pH 7.5.
  • Each K D value is an average of three independent experiments and is shown with the associated standard error.
  • GSNOR inhibitors were performed using a library of 60,000 compounds from ChemDiv Inc in the Chemical Genomics Core facility at Indiana University. Screening was conducted in 384 well plates and involved incubating GSNOR with 12.5 ⁇ M compound, 1 mM each OfNAD + and octanol in 0.1 M sodium glycine pH 10. Enzyme activity was determined by measuring the rate of production of NADH spectrophotometrically at 340 nm. Inhibition of GSNOR was calculated from the ratio of enzyme activity in the presence of compounds to that in no compound controls performed on the same assay plate.
  • ⁇ Fi the change in the fluorescence at 455 nm upon the addition of inhibitor.
  • AF M is the maximum fluorescence change that was obtained from curve fitting.
  • Ej and L T are the concentrations of GSNOR and inhibitor, respectively.
  • K D is the equilibrium dissociation constant for the formation of GSNOR » NADH*Inhibitor complex. The data were fitted using the Graphpad Prizm 4.0.
  • RAW 264.7 cells were cultured in DMEM medium supplemented with 10 % FBS, 200 U/ml of penicillin and 200 ⁇ g/mL of streptomycin. The cells were incubated at 37 0 C in an atmosphere containing 5% CO 2 and 95% air. For the experiments, 1-2 X 10 6 cells were plated in six- well plates a day before the experiment. On the day of the assay the medium was replaced with 3 ml of fresh medium and the cells were treated with compounds for a predetermined length of time.
  • lysis buffer 50 mM potassium phosphate pH 7.0 containing 50 mM NEM and ImM EDTA.
  • Cells were lysed by sonication using a micro tip probe (three pulses of 30% duty cycle; 2 output control on a Fisher Sonicator).
  • Cell debris was pelleted by centrifugation (10 min at 16,00Og) and the cell lysate was analyzed for protein concentration using the Bio-Rad dye-binding protein assay. The concentration of nitroso compounds in the cell lysate was determined using the triiodide based chemiluminescence method using a Sievers 280 NO analyzer.
  • the amount of s-nitrosothiols in the cell lysate was determined by initially passing the cell lysate through a microspin column and treating the eluate with 5 mM HgCl 2 before determining the nitroso compound concentration using chemiluminescence.
  • the cells were pretreated with the compounds for 16 hours prior to the day of experiment. Later experiments showed that this pretreatment had no effect on the rate of accumulation of nitroso species inside the cells.
  • RAW 264.7 cells were cultured in 10% heat-inactivated serum containing DMEM. Cells were treated with 33 micromolar Compound 8 for varied lengths of time alone or in combination with 1 mM NAME for 4 h (4+N). At indicated times, the cells were quenched and the lysate was analyzed for s-nitrosothiol content by the biotin switch assay as described by Jaffiey et al, Methods Enzymol. 396, 105-118 (2005) with modifications suggested by Wang et al, Free Radic. Biol. Med. 44, 1362-1372 (2008) and Zhang et al, Free Radic. Biol. Med. 38, 874-881 (2005).
  • mice were anesthetized with diethyl ether.
  • a thoracotomy was performed to expose thoracic and abdominal aorta.
  • a 25 gauge syringe was inserted into the apex of left ventricle and perfused free of blood with oxygenated Krebs Henseleit buffer. The right atrium was cut to provide an exit for blood.
  • the aorta was removed and cleaned of fat and adventitia.
  • the aorta was cut into 2-mm-long segments and mounted on a four-channel wire myograph (AD Instruments). Vessel rings were maintained in 10-ml organ baths with oxygenated PSS (95% O 2 and 5% CO 2 ) at 37 0 C.
  • Rings were allowed to equilibrate for 80 minutes with the buffer in each organ bath changed every 20 min. One gram pretension was placed on each aortic ring (appropriate starting tension for optimal vasomotor function as determined in previous experiments). An eight-channel octal bridge (Powerlab) and data-acquisition software (Chart version 5.2.2) were used to record all force measurements. After equilibration for 80 min, l ⁇ M 27 phenylephrine was added to each ring for submaximal contraction. After stabilization, either Compound 8 or sodium nitroprusside (SNP) was added to the rings and the tone of the rings determined.
  • SNP sodium nitroprusside

Abstract

Novel inhibitors of s-nitrosoglutathione reductase (GSNOR) are disclosed that specifically inhibit GSNOR among the alcohol dehydrogenases; methods of using these and related molecules are also disclosed. These inhibitors may interact with the GSNO binding site and inhibit GSNOR at multiple places in its kinetic pathway. These molecules inhibit GSNOR in a dose-dependent manner and demonstrate that GSNOR actively regulates the s-nitrosylation of proteins against incoming low molecular weight nitrosothiols. These compounds are useful in methods of treating, diagnosing and studying various conditions and diseases, including but not limited to Cardiovascular Diseases, Pulmonary Diseases, Urogenital Diseases, Neurologic Diseases, Inflammatory Diseases, and Oncologic Diseases, as well as various genetic conditions. Innovative uses for these compounds include using them to study enzymes involving the s-nitrosylation of proteins and various cellular processes that involve or that are thought to involve the activity of these enzymes.

Description

TITLE
MATERIALS AND METHODS FOR INHIBITING MAMMALIAN S- NITROSOGLUTATHIONE REDUCTASE
STATEMENT OF GOVERNMENTAL RIGHTS
[0001] Part of the work during the development of this invention was made with government support provided by the United States Government under NIH Grant No. R21 HL087816. The U.S. Government has certain rights in the invention.
PRIORITY CLAIM
[0002] This application claims the benefit of US Provisional Patent Application Number 61/013,522 filed on December 13, 2007 and US Provisional Patent Application Number 61/021,781 filed on January 17, 2008, both of these Provisional Patent Applications are incorporated herein by reference in their entireties as if each were individually incorporated herein by reference in their entirety.
TECHNICAL FIELD
[0003] Various aspects and embodiments relate generally to materials and methods for inhibiting the enzyme S-nitrosoglutathione reductase (GSNOR) and for diagnosing, studying and treating various conditions and diseases related to the activity of this enzyme.
BACKGROUND
[0004] Endogenous S-nitrosothiols (SNOs) are naturally occurring proteins in which a sulfur atom from cysteine or homocystine reacts with NO to form an S-NO bond. Within mammalian cells, SNO concentrations range from nM to μM concentrations; thiol S- nitrosylation and NO transferase reactions are involved in virtually all classes of cell signaling, ranging from regulation of ion channels and G-protein coupled reactions, to reception stimulation and activation of nuclear regulatory proteins. Regulatory signaling exists for the synthesis, transport, activation and metabolism of SNOs. (Gaston BM, Carver J, Doctor A, Palmer LA. S-nitrosylation signaling in cell biology. MoI Interv 2003;3:253-63). S-nitrosoglutathione (GSNO), one of the major forms of SNO that is formed endogenously, is catabolized by the enzyme S-nitrosoglutathione reductase (GSNOR). The regulation of GSNOR activity is believed to be one of the mechanisms by which cellular NO is regulated. GSNOR catalyzes the denitrosylation of intracellular proteins by the reduction of s- nitrosoglutathione (GSNO). Because of its role in the regulation of the s-nitrosylation of intracellular proteins, GSNOR has become an important target for the development of new therapeutic agents targeting this enzyme, since inhibition of the activity of GSNOR should increase NO concentrations, which in turn should lead to clinically significant changes in end-organ function.
[0005] S-nitrosylation of cellular proteins has emerged as the key reaction through which NO exerts its numerous effects inside the body. The growing list of proteins whose activities are regulated by s-nitrosylation include ion channel proteins, kinases, proteolytic enzymes, transcription factors and proteins involved in energy transduction. (Gewaltig MT, Kojda G. Vasoprotection by nitric oxide: mechanisms and therapeutic potential. Cardiovasc Res 2002;55:250-60). In conjunction with s-nitrosylation of proteins, NO has been shown to regulate processes and proteins involved in apoptosis, G-protein-coupled receptor based signaling, vascular tone, and inflammatory responses.
[0006] Given GSNOR' s role in the modulation of GSNO concentrations, which in turn alters cellular concentrations of NO, therapeutic compounds that inhibit GSNOR activity have the potential to treat a wide variety of diseases whose common pathophysiology is abnormally low NO concentrations. GSNOR inhibitors could lead to increased concentrations of GSNO and NO and have the potential to impact these diseases.
[0007] Nitric oxide and the s-nitrosylation/de-nitrosylation cycle play an essential role in many pathologic disease states due largely to dysfunction of endothelial cells. Various vascular disorders such as heart disease, heart failure, heart attack, hypertension, atherosclerosis, and restenosis can be directly linked to abnormal NO activity and/or concentrations, and subsequent changes in the s-nitrosylation of target proteins. Other conditions such as asthma and impotence are also linked to abnormal levels of NO bioactivity (Gaston BM, Carver J, Doctor A, Palmer LA. S-nitrosylation signaling in cell biology. MoI Interv 2003,3:253-63; Gewaltig MT, Kojda G. Vasoprotection by nitric oxide: mechanisms and therapeutic potential. Cardiovasc Res 2002,55 :250-60;Gerard C. Biomedicine. Asthmatics breathe easier when it's SNO-ing. Science 2005;308:1560-l). [0008] The therapeutic potential of preventing the breakdown of s-nitrosothiols via inhibition of GSNOR has been demonstrated in a mouse model for asthma. Knockout mice lacking the genes for GSNOR were found to resist airway hyperresponsivity due to higher GSNO concentrations in bronchial fluids and demonstrated diminished tachyphylaxis to β- agonists because of the s-nitrosylation of G-protein coupled receptor kinases (Que LG, Liu L, Yan Y, et al. Protection from experimental asthma by an endogenous bronchodilator. Science 2005;308: 1618-21). Cystic fibrosis patients have also been shown to have abnormally low levels of GSNO which is thought to play a role in the pathophysiology of this disease; therapies that inhibit GSNOR may offer these patients a new therapeutic option. (Zeitlin PL. Is it go or NO go for S-nitrosylation modification-based therapies of cystic fibrosis transmembrane regulator trafficking? MoI Pharmacol 2006; 70:1155-8; Childers M, Eckel G, HimmelA, CaldwellJ. A new model of cystic fibrosis pathology : lack of transport of glutathione and its thiocyanate conjugates. Med Hypotheses 2007;68:101-12). [0009] Abnormal NO concentrations and abnormal GSNO activity may also play a role in other diseases including Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, stroke, septic shock, cardiogenic shock, endotoxic shock, toxic shock syndrome, systemic inflammatory response syndrome, and other inflammatory diseases {Zeitlin PL. Is it go or NO go for S-nitrosylation modification-based therapies of cystic fibrosis transmembrane regulator trafficking? MoI Pharmacol 2006, 70:1155-8; Heiss C, Lauer T, Dejam A, et al. Plasma nitroso compounds are decreased inpatients with endothelial dysfunction. JAm Coll Cardiol 2006;47:573-9; Murad F. Shattuck Lecture. Nitric oxide and cyclic GMP in cell signaling and drug development. N EnglJ Med 2006; 355:2003-11; Kroncke KD1 Fehsel K, Kolb-Bachofen V. Inducible nitric oxide synthase in human diseases. Clin Exp Immunol 1998, 113:147-56).
SUMMARY
[0010] One aspect of the invention is a compound and/or a method for using a compound to alter enzyme activity, comprising the steps of: providing at least one compound selected from the group consisting of:
Figure imgf000005_0001
Figure imgf000005_0002
R ._ H, CH3 , F, Cl, Br, OCH3, C -CH-.
R5 H, -CH3, ~C2H5, -C3H7
Figure imgf000005_0003
K.| C2H5, -H, -OXI3, -O3Xi7
R2 = -H, -Cl, -F.. -Br
/==y ^4
R3= -C2H5, -C4H9, \J
R4= -OC2H5, -OCH3, -C2H5, -F, -Cl, -Br, -OCH3, -COOC2H5
R5= =H, -CH3, -C2H5, -C3H7
Figure imgf000006_0001
R1 = -H, -CH3, -C2H5, -C3H7 R2 = -H, -CN, -OH3 -CH2OH . and
Figure imgf000006_0002
R1 = -H, -CH3, -C2H5, -C3H7
Figure imgf000006_0003
Vn-CH3I Vn-C2H5 Vn-C3H7
or a physiologically acceptable salt thereof; and contacting said compound with s- nitrosoglutathione reductase.
[0011] One embodiment is a method for treating a disease or a condition, comprising the steps of: providing at least one compound according to any of the compounds provided herein including those referred to above, and administering a therapeutically effective dose of said compound to a patient in need thereof. Still another embodiment is a method for treating a disease as outlined above, wherein the disease or condition is selected from the group consisting of: Cardiovascular Diseases, including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease; Pulmonary Diseases, including but not limited to asthma, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis; Urogenital Diseases, including but not limited to impotence and abnormalities of ejaculation. Inherited genetic diseases, including but not limited to cystic fibrosis and sickle cell anemia; Neurologic Diseases, including but not limited to Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS); Inflammatory Diseases, including but not limited to systemic inflammatory response syndrome, rheumatoid arthritis, Sjogren's syndrome, ulcerative colitis, Crohn's disease, glomerulonephritis, psoriasis, cutaneous lupus erythematosus, systemic lupus erythematosus, systemic sclerosis, dermatitis, and other inflammatory diseases; Oncologic Diseases, including both hematologic and solid tumors. In one embodiment the therapeutically effective amount of said compound in on the range of about 0.01 mg/kg per of body mass day to 1000 mg/kg of body mass per day.
[0012] Yet another embodiment is a method for diagnosing or studying a disease or a condition, comprising the steps of: providing at least one compound as disclosed above and contacting the compound with s-nitrosoglutathione reductase; an observing a change in conformity with a change in s-nitrosylation. In one embodiment, the disease or condition being diagnosed is selected from the group consisting of: Cardiovascular Diseases, including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease; Pulmonary Diseases, including but not limited to asthma, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis; Urogenital Diseases, including but not limited to impotence and abnormalities of ejaculation. Inherited genetic diseases, including but not limited to cystic fibrosis and sickle cell anemia; Neurologic Diseases, including but not limited to Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS); Inflammatory Diseases, including but not limited to systemic inflammatory response syndrome, rheumatoid arthritis, Sjogren's syndrome, ulcerative colitis, Crohn's disease, glomerulonephritis, psoriasis, cutaneous lupus erythematosus, systemic lupus erythematosus, systemic sclerosis, dermatitis, and other inflammatory diseases; Oncologic Diseases, including both hematologic and solid tumors. Still another embodiment is a method of studying protein nitrosylation comprising the steps of: providing at least one compound according disclosed herein, including those name in the above and contacting said compound with s-nitrosoglutathione reductase; and observing a change in s-nitrosylation. Another embodiment provides a kit for altering the activity of s- nitrosoglutathione reductase comprising at least one compound named in the above. [0013] Still another embodiment provides a method for treating a disease or condition comprising the steps of providing at least one compound selected from the group consisting of:
Figure imgf000008_0001
a pharmaceutically acceptable salt or ester thereof; and administering a therapeutically effective dose of said compound to a patient in need thereof. In one embodiment, the disease or condition is selected from the group consisting of: Cardiovascular diseases, including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease; Pulmonary Diseases, including but not limited to asthma, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis; Urogenital Diseases, including but not limited to impotence and abnormalities of ejaculation; Inherited Genetic Diseases, including but not limited to cystic fibrosis and sickle cell anemia; Neurologic Diseases, including but not limited to Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS); Inflammatory Diseases, including but not limited to systemic inflammatory response syndrome, rheumatoid arthritis, Sjogren's syndrome, ulcerative colitis, Crohn's disease, glomerulonephritis, psoriasis, cutaneous lupus erythematosus, systemic lupus erythematosus, systemic sclerosis, dermatitis, and other inflammatory diseases; Oncologic Diseases, including both hematologic and solid tumors. In one embodiment the therapeutically effective amount of the compound used to treat a disease or condition in the range of about 0.01 mg/kg per of body mass day to 1000 mg/kg of body mass per day.
[0014] Still another embodiment provides a method for diagnosing or studying a disease, condition or chemical and/or enzymatic reaction including the steps of providing at least one compound selected from the group consisting of:
Figure imgf000009_0001
Figure imgf000009_0002
NC'^, or a pharmaceutically acceptable or ester thereof; contacting said compound with s-nitrosoglutathione reductase; and observing a change in conformity with a change in s-nitrosylation. In one embodiment the disease and/or condition being diagnosed and/or studied is selected from the group consisting of: Cardiovascular Diseases, including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease; Pulmonary Diseases, including but not limited to asthma, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis; Urogenital Diseases, including but not limited to impotence and abnormalities of ejaculation. Inherited genetic diseases, including but not limited to cystic fibrosis and sickle cell anemia; Neurologic Diseases, including but not limited to Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS); Inflammatory Diseases, including but not limited to systemic inflammatory response syndrome, rheumatoid arthritis, Sjogren's syndrome, ulcerative colitis, Crohn's disease, glomerulonephritis, psoriasis, cutaneous lupus erythematosus, systemic lupus erythematosus, systemic sclerosis, dermatitis, and other inflammatory diseases; Oncologic Diseases, including both hematologic and solid tumors. Still another embodiment is a kit for altering the activity of s-nitrosoglutathione reductase, comprising at least one of the compounds described in the above.
[0015] One aspect of the present invention provides compounds that can be used to inhibit the enzyme S-nitrosoglutathione reductase (GSNOR). These compounds include Compound A, Compound B, Compound C and Compound D defined below, as well as all physiologically acceptable salts thereof.
[0016] One embodiment is a method of treating human or animal patients comprising the steps of providing at least one compound selected from the group comprising Compound A, Compound B, Compound C and Compound D or a physiological salt thereof and administering a therapeutically effective dose of the compound to a human or animal patient. [0017] In still another embodiment is a method of diagnosing a disease in a human or an animal comprising the steps of providing at least one compound, selected from the group comprising Compound A, Compound B, Compound C and Compound D or a physiological salt thereof and contacting said compound with at least one enzyme in the human or animal.
[0018] Yet another embodiment is a method of studying the mechanism, chemistry or role in physiology of the enzyme s-nitrosoglutathione reductase (GSNOR) either in vitro or in vivo comprising the steps of providing at least one compound selected from the group comprising Compound A, Compound B, Compound C and Compound D or a physiological salt thereof and contacting said compound with the enzyme s-nitrosoglutathione reductase (GSNOR).
[0019] One embodiment is a method of treating, diagnosing, curing, controlling or otherwise affecting a disease, defect or other medical condition treated by administering a therapeutically effective dose of at least one of the following compounds selected from the group including Compound A, Compound B, Compound C and Compound D or a physiological salt thereof. In one embodiment the condition affected is selected from the group comprising the activation of elements of the immune system including, but not limited to, macrophages, thymocytes, lymphocytes, or intercellular networks that involve NO signaling networks, cellular processes such as apoptosis, the activity of endothelial cells, cardiovascular disorders, including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease. [0020] In another embodiment of the invention, the condition affected is selected from pulmonary diseases including but not limited to asthma, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis.
[0021] In another embodiment of the invention, the condition affected is selected from diseases of the urogenital system, including but not limited to impotence and abnormalities of ejaculation. In another embodiment of the invention, the condition affected is selected from inherited genetic diseases, including but not limited to cystic fibrosis and sickle cell anemia. [0022] In another embodiment of the invention, the condition affected is selected from neurologic diseases, including but not limited to Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS).
[0023] In another embodiment of the invention, the condition affected is selected from inflammatory diseases, including but not limited to systemic inflammatory response syndrome, rheumatoid arthritis, Sjogren's syndrome, ulcerative colitis, Crohn's disease, glomerulonephritis, psoriasis, cutaneous lupus erythematosus, systemic lupus erythematosus, systemic sclerosis, dermatitis, and other inflammatory diseases. In another embodiment of the invention the condition affected is selected from oncologic diseases, including both hematologic and solid tumors.
BRIEF DESCRIPTION OF THE FIGURES
[0024] FIG. 1 is a diagram illustrating the Kinetic mechanism of GSNOR and the types of complexes that an inhibitor binding to the GSNO site could form along the kinetic pathway of GSNOR.
10025] FIG. 2 illustrates the effect of compound 8 on the fluorescence of GSNOR bound
NADH in the presence or absence of 12-HDDA.
[0026] FIG. 3 illustrates data consistent with an increase in the accumulation of nitroso species in RAW 264.7 cells in presence of GSNOR inhibitors.
[0027] FIG. 4 illustrates the effects of Compound 8 on the nitrosylation of cellular proteins.
[0028] FIG. 5 illustrates a dose-response relationship of Compound 8 and sodium nitroprusside.
[0029] FIG. 6 illustrates that inhibition of GSNOR increases cGMP production.
[0030] Table 1 represents structures of some inhibitors of GSNOR.
[0031] Table 2 represents data illustrating inhibition of various alcohol dehydrogenase isozymes by compounds that inhibit the enzyme GSNOR.
[0032] Table 3 represents a summary of kinetic data consistent with mechanism of GSNOR inhibition by various compounds.
[0033] Table 4 represents data summarizing kinetic contrasts measured with various exemplary compounds thought to inhibit GSNOR.
DETAILED DESCRIPTION
[0034] For the purposes of promoting an understanding of the principles of the novel technology, reference will now be made to the preferred embodiments thereof, and specific language will be used to describe the same. It will nevertheless be understood that no limitation of the scope of the novel technology is thereby intended, such alterations, modifications, and further applications of the principles of the novel technology being contemplated as would normally occur to one skilled in the art to which the novel technology relates. [0035] The enzyme S-nitrosoglutathione reductase (GSNOR) catalyzes the removal of nitro groups from nitrosylated proteins. GSNOR is generally categorized as a member of the alcohol dehydrogenase family of enzymes; its physiological substrates are thought to be nitrosylated proteins, S-nitrosoglutathione (GSNO) and NADH. Nitrosylated intercellular proteins are often the result of NO bioactivity and these proteins account for much of NO's physiological effects. Accordingly, the denitrosylation reaction catalyzed by GSNOR helps healthy, normal cells to maintain a balance between nitrosylated and de-nitrosylated proteins and functions as an integral part of the cycle that modulates the effects of NO and its attendant effects.
[0036] The inventors have identified a number of compounds including substituted pyrroles, indoles, thiophenes and aromatic rings that affect the activity of GSNOR and therefore may be used to modulate NO's effect on intercellular proteins and on physiology. One aspect includes molecules having the structure below generally referred to as Compound A and using the same to treat and/or diagnose various diseases and/or conditions or to study various enzyme catalyzed reactions; these molecules have the following generic structure and include pharmaceutically acceptable salts and/or esters thereof:
COMPOUND A
Figure imgf000013_0001
R5 = -H, -CH3, -C2H5, -C3H7
[0037] Another aspect includes molecules having the structure generally referred to as Compound B and using the same to treat and/or diagnose various diseases and/or conditions or to study various enzyme catalyzed reactions; these molecules have the following generic structure and include pharmaceutically acceptable salts and/or esters thereof:
COMPOUND B
Figure imgf000013_0002
Figure imgf000013_0003
-C3H.7 R2 = -H, -Cl, -F, -Br
,R4
R3= -C2H5, -C4H9, -Qf
R4= -OC2H5, -OCH3, -C2H5, -F, -Cl, -Br, -OCH3, -COOC2H3
R5= =H, -CH3, -C2H5, -C3H7 [0038] Still another aspect includes molecules having the structure generally referred to as Compound C and using the same to treat and/or diagnose various diseases and/or conditions or to study various enzyme catalyzed reactions; these molecules have the following generic structure and include pharmaceutically acceptable salts and/or esters thereof:
COMPOUND C
Figure imgf000014_0001
R1 — JtI, -UH3, -O2JIj, -C3H7
R2 = -H, -CN, -OH, -CH2OH
[0039] Yet another aspect includes molecules having the structure generally referred to as Compound D and using the same to treat and/or diagnose various diseases and/or conditions or to study various enzyme catalyzed reactions; these molecules have the following generic structure and include pharmaceutically acceptable salts and/or esters thereof:
COMPOUND D
Figure imgf000014_0002
RJ — -H, -CH3, -C2H5, -C3H7
Figure imgf000014_0003
[0040] Synthesis of the backbone of compounds such as Compound 6 in Table 1 and the like is given in general form in Scheme I. Additional details concerning the synthesis of this and related molecules can be found in the publication, {Journal of Medicinal Chemistry, 1997, vol. 40, No. 11).
[0041] The compounds useful in the invention may be delivered not only as single agents by oral, inhalation or parenteral routes, but also in the form of cocktails which are mixtures of other appropriate compounds to treat a particular disease. The use of cocktails in the treatment of asthma, cardiovascular disease and other diseases treated by the invention is routine. In this embodiment, a common administration vehicle (e.g., pill, tablet, implant, injectable solution, etc.) would contain both a compound of this invention and at least one additional therapeutic drug and/or supplementary potentiating agent.
[0042] The compounds described herein when used alone or in cocktails are administered in therapeutically effective amounts. A therapeutically effective amount will be determined by the parameters discussed below; but it is that amount which establishes a level of the drug(s) in the area of the blood stream or tissue to be treated, such as the lung or vascular smooth muscle, which is effective in causing a therapeutic benefit.
[0043] When administered, the formulations of the invention are applied in pharmaceutically acceptable amounts and in pharmaceutically acceptable compositions. Such preparations may routinely contain salts, buffering agents, preservatives, compatible carriers, and optionally other therapeutic ingredients. When used in medicine the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically acceptable salts thereof and are not excluded from the scope of the invention. Such pharmacologically and pharmaceutically acceptable salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulfonic, tartaric, citric, methane sulfonic, formic, malonic, succinic, naphthalene-2-sulfonic, and benzene sulfonic. Also, pharmaceutically acceptable salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts. Furthermore, included along with the Compounds A, B, C, and D are their pharmaceutically acceptable salts, including base addition salts. The term pharmaceutically-acceptable salts, includes salts commonly used to form alkali metal salts and to form addition salts of free bases. The nature of the salt is not critical, provided that it is pharmaceutically acceptable.
[0044] Suitable pharmaceutically-acceptable base addition salts of the compounds of the invention may be prepared from an inorganic base or an organic base. Suitable pharmaceutically-acceptable base addition salts include metallic salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts made from N5N'- dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N- methylglucamine, and procaine. All of these salts may be prepared by conventional means from the corresponding compounds of the invention by reacting, for example, the appropriate base with one of the compounds of the invention.
[0045] Also included in the invention are the pharmaceutically acceptable esters of compounds of the invention. These esters may be prepared by an acid catalyzed reaction between the compounds of the invention and an alcohol like methanol, ethanol, isopropyl, butanol and other alkyl and aryl alcohols.
[0046] Suitable buffering agents include, for example, acetic acid and a salt (1-2% W/V); citric acid and a salt (1-3% W/V); boric acid and a salt (0.5-2.5% W/V); and phosphoric acid and a salt (0.8-2% W/V).
[0047] Suitable preservatives include benzalkonium chloride (0.003-0.03% W/V); chlorobutanol (0.3-0.9% W/V); parabens (0.01-0.25% W/V) and thimerosal (0.004-0.02%
W/V).
[0048] The invention is also directed to pharmaceutical compositions and/or formulations and uses for the same comprising, for example, at least one of the following Compound 6, 7 or 8 (Table 1) or the like and pharmaceutically acceptable salts and/or esters thereof and at least one pharmaceutically acceptable carrier or diluent, as well as methods for using the same to treat and or diagnose various diseases of conditions a well as methods for studying various chemical and biological processes.
[0049] The formulations of the present invention can be solutions, suspensions, syrups, tablets, capsules, and the like. The compositions may contain a suitable carrier, diluent, or excipient, such as a medium chain triglyceride oil or magnesium stearate. In preferred formulations, a medium chain triglyceride oil and magnesium stearate is present in an approximately 1 : 1 ratio. Standard texts, such as Remington's Pharmaceutical Science, 18th
Ed., 1990, incorporated herein by reference, may be consulted to prepare suitable preparations, without undue experimentation.
[0050] One preferred carrier is polyethylene glycol (PEG). One more preferred is a mixture of polyethylene glycol having a high molecular weight, for example of greater than 900 (most preferably, about 1,000), and polyethylene glycol having a low molecular weight, for example of less than 500 preferably about 400). One particularly preferred carrier is PEG in the ratio of about one part PEG with a MW of 100 to about two parts PEG with a MW of about 400. [0051] Preferred emulsifiers include phosphatidylcholine emulsifiers, such as dilauroylphosphatidylcholine.
[0052] The formulations can include powdered carriers, such as lactose, sucrose, mannitol, starch, cellulose derivatives, xanthum gum, magnesium stearate, stearic acid, and the like. The composition may also comprise a penetration enhancer. Suitable penetration enhancers include glycerol, glycerol monolaureate, dimethyl sulfoxide or oils, such as a mineral oil or medium chain triglyceride oil.
[0053] Antioxidants such as, for example, butylated hydroxytoluene (BHT), sodium bisulfate, sodium sulfite, sodium EDTA, ascorbic acid, and the like can be used either alone or in combination with other suitable antioxidants or stabilizing agents typically employed in pharmaceutical compositions.
[0054J The formulations can also include any of the commonly used disintegrants, lubricants, plasticizers, colorants, and dosing vehicles. Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, a standard reference text in this field. Suitable formulations typically contain from about 1 to about 1000 mg of active ingredient per dosage unit. In these pharmaceutical compositions, the active ingredient will ordinarily be present in an amount of about 0.5 to about 95%, by weight, based on the total weight of the composition.
[0055] The magnitude of the therapeutic dose of the compounds of the invention will vary with the nature and severity of the condition to be treated and with the particular route of administration. Although the most suitable route in any given case will depend largely on the nature and severity of the condition being treated and on the nature of the active ingredient, the compositions of the invention may be conveniently presented in unit dosage form and prepared by any of the methods well known in the art of pharmacy.
[0056| The active compounds of the present invention may be a pharmaceutical composition having a therapeutically effective amount of a conjugate of the invention optionally included in a pharmaceutically-acceptable carrier. The term "pharmaceutically-acceptable carrier" as used herein means one or more compatible solid or liquid fillers, diluents, or encapsulating substances which are suitable for administration to a human or other animal. The term "carrier" denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. The components of the pharmaceutical compositions are capable of being commingled with the molecules of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy. [0057] Compositions suitable for parenteral administration conveniently comprise a sterile preparation of the compounds of the invention. This preparation may be formulated according to known methods. The sterile preparation thus may be a sterile solution or suspension in a non-toxic parenterally-acceptable diluent or solvent. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono or di-glycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. Carrier formulations suitable for oral, subcutaneous, intravenous, intramuscular, etc. can be found in, for example, Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa. [0058] A subject or a patient, as used herein, may mean a human, primate, horse, cow, pig, sheep, goat, dog, cats, rodent and the like. The term 'about' used in conjunction with a numerical value or range generally means plus or minus 20 percent; e.g. 'about 1.0' includes values from 0.8 to 1.2.
[0059] The compounds of the invention are administered in effective amounts. An effective amount is an amount necessary to delay the onset of, inhibit the progression of or halt altogether the onset of, or progression of, to diagnose the particular condition being treated. For example, in general, an effective amount for treating asthma will be that amount necessary to open airways and decrease airway inflammation such that a therapeutic benefit results. When administered to a subject, effective amounts will depend, of course, on the particular condition being treated; the severity of the condition; individual patient parameters including age, physical condition, size and weight; concurrent treatment; frequency of treatment; and the mode of administration. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is preferred generally that a maximum dose be used, that is, the highest safe dose according to sound medical judgment.
[0060] Dosage may be adjusted appropriately to achieve desired drug levels, locally or systemically. Generally, daily oral doses of active compounds will be from about 0.01 mg/kg per day to 1000 mg/kg per day. It is expected that IV doses in the range of about 1 to 1000 mg/m2 per day will be effective. In the event that the response in a subject is insufficient at such doses, even higher doses (or effective higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits. Continuous IV dosing over, for example 24 hours or multiple doses per day are also contemplated as necessary to achieve appropriate systemic levels of the compounds. Likely, preferred dosing schedules, including concentration, length of administration, and the like are described herein.
[0061] A variety of administration routes are available. The particular mode selected will depend, of course, upon the particular drug selected, the severity of the disease state being treated and the dosage required for therapeutic efficacy. The methods of this invention, generally speaking, may be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of the active compounds without causing clinically unacceptable adverse effects. Such modes of administration include, for example, oral, rectal, sublingual, topical, nasal, transdermal, intradermal or parenteral routes. The term "parenteral" includes subcutaneous, intravenous, intramuscular, or infusion. In some instances intravenous routes may be preferred. [0062] The compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the conjugates of the invention into association with a carrier which may constitute one or more accessory ingredients. In general, the compositions may be prepared by uniformly and intimately bringing the compounds into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product. [0063] Compositions suitable for oral administration may be presented as discrete units such as capsules, cachets, tablets, or lozenges, each containing a predetermined amount of the active compound. Other compositions may include, for example, suspensions in aqueous liquors or non-aqueous liquids such as a syrup, an elixir, or an emulsion. [0064] Other delivery systems can include time-release, delayed release or sustained release delivery systems. Such systems can avoid repeated administrations of the active compounds of the invention, increasing convenience to the subject and the physician. Many types of release delivery systems are available and known to those of ordinary skill in the art. They include polymer based systems such as polylactic and polyglycolic acid, polyanhydrides and polycaprolactone; nonpolymer systems that are lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono, di and triglycerides; hydrogel release systems; silastic systems; peptide based systems; wax coatings, compressed tablets using conventional binders and excipients, partially fused implants and the like. In addition, a pump-based hardware delivery system can be used, some of which are adapted for implantation.
[0065] A long-term sustained release implant also may be used. "Long-term" release, as used herein, may mean that the implant is constructed and arranged to deliver therapeutic levels of the active ingredient for at least 30 days, and in some instances 60 days or longer. Long-term sustained release implants are well known to those of ordinary skill in the art and include some of the release systems described above. Such implants can be particularly useful in treating chronic medical conditions.
[0066] The compounds of the invention are also useful, in general, for treating disease or conditions including, but not limited to, Cardiovascular Diseases, including systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease; Pulmonary Diseases, including but not limited to asthma, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis; Urogenital Diseases, including but not limited to impotence and abnormalities of ejaculation; Inherited Genetic Diseases, including but not limited to cystic fibrosis and sickle cell anemia; Neurologic Diseases, including but not limited to Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS); Inflammatory Diseases, including but not limited to systemic inflammatory response syndrome, rheumatoid arthritis, Sjogren's syndrome, ulcerative colitis, Crohn's disease, glomerulonephritis, psoriasis, cutaneous lupus erythematosus, systemic lupus erythematosus, systemic sclerosis, dermatitis, and other inflammatory diseases; Oncologic Diseases, including both hematologic and solid tumors. Those skilled in the art will be able to recognize with no more than routine experimentation numerous equivalents to the specific products and processes described above. Such equivalents are intended to be included within the scope of the appended claims. All references disclosed herein are incorporated by reference as if each were incorporated individually. Results and Discussion
Identification of compounds that inhibit GSNOR at physiological pH: [0067] GSNOR is an alcohol dehydrogenases, and like other members of ADH family, is capable of oxidizing and reducing primary alcohols and aldehydes with the help of a molecule of NAD(H). Octanol was chosen as the substrate in the initial screening assay as it is bulky like GSNO, but has 200-fold higher KM value (1 mM) and a kcat value (200 min"1) that is more than 2 order of magnitude lower than that of GSNO. This allows the initial velocity conditions to prevail for a period up to 10 min (data not shown) and it enables conducting a large number of concurrent assays in a 384 well plate format. The pH of the assay was adjusted to pH 10 in order to obtain significant rate of oxidation of octanol in the absence of compounds. The screening for GSNOR inhibitors was performed in the presence of, saturating concentration OfNAD+ and an un-saturating concentration of octanol, thereby increasing the probability of identifying compounds that bind, only or primarily in the GSNO binding site of GSNOR. This was deemed desirable as there are many dehydrogenases inside the cells, and compounds binding in the coenzyme binding site are more likely to be nonspecific inhibitors of GSNOR than compounds that bind primarily to the form of GSNOR that binds GSNO.
[0068] Compounds that inhibited the rate of octanol oxidation by 40% or more in the initial screening assay were selected for further analysis. The GSNOR inhibitory activity of the initially identified compounds was confirmed at pH's 10 and 7.5 using 12- hydroxydodecanoinc acid and s-nitrosoglutathione as substrates, respectively. Table 1 lists representative compounds from each of the eight different classes of compounds identified in the initial screening experiment. Each of the identified compounds in Table 1 was a better inhibitor of GSNOR than the existing GSNOR inhibitor, dodecanoic acid, at inhibiting GSNOR at pH 10. Still referring to Table 1, Compounds 6-8 inhibited GSNOR more effectively at pH 10 (53-97 % inhibition) than at pH 7.5 (1.7-35 % inhibition). The decrease in the affinity of these Compounds at pH 7.5 is likely due to the change in the ionization status of their ionizable groups at lower pH. Both the imidamide group in Compound 1 and the pyridinyl nitrogen in Compound 2 would be more protonated at pH 7.5 than at pH 10 and would be repelled by a similarly charged residue in the GSNOR active site. Compound 8 also displays higher GSNOR inhibition at pH 10 than at pH 7.5. However, it has a phenolic hydroxyl group whose ionization state would be suppressed significantly at pH 7.5. Without wishing to be bound by theory, it is possible that the ionized phenolic hydroxyl group is involved in an important interaction with an oppositely charged residue within the GSNOR active site. It is likely that Compounds 6-8 could serve as effective GSNOR inhibitors at physiological pH only if the charge on their ionizable group is reversed or their pKa values perturbed significantly; accordingly, Compounds 6-8 were not characterized further. [0069] Compounds 4-8 appear to be especially promising inhibitors of GSNOR as they appeared to maintain their affinity for the GSNOR active site at both of the pHs assayed. These Compounds also exhibited hundred-fold lower IC50 values than does dodecanoic acid measured at physiological pH. Referring now to Compound 5 (Table 1), the removal of the ester group significantly improved the affinity of the inhibitor for GSNOR as evidenced by a three-fold higher inhibition of GSNOR by the hydrolyzed form of the Compound (Table 1). Compounds 4-8 meet Lipinski's five point rule for small molecules with drug-like properties and appeared to be good candidates for further investigation as inhibitors of GSNOR. Selectivity of GSNOR inhibition
[0070] The ability of Compounds 4-8 to selectively inhibit GSNOR as opposed to other alcohol dehydrogenases (ADHs) was deemed to be important, as most ADHs have a similar overall structure, and broad substrate specificity. Inhibition of three additional isozymes of ADH family, specifically, β2 β 2-, π-, σ- ADHs by the same concentration of Compounds 4-8 was examined along with that of GSNOR. In each case, the inhibition assay was conducted in the presence of saturating or KM concentrations of the coenzyme and the alcohol substrate. Referring now to Table 2, Compounds 6-8 are highly specific inhibitors of GSNOR. At concentrations that inhibit more than 70% of GSNOR activity, there is no significant inhibition of any of the other ADH isozymes tested with Compounds 6-8. Compound 4 inhibited π-ADH more than GSNOR and it was not studied any further. Compound 5, while being a modestly good inhibitor of GSNOR, also showed significant inhibition of σ-ADH. Accordingly, further modifications of Compound 5 may be necessary to improve its selectivity for GSNOR over the other ADH isozymes. Compounds 6-8 show promise as highly effective and selective inhibitors of GSNOR and were selected as lead compounds for further investigation.
Determining the mechanism of inhibition of GSNOR by compounds such as 6-8 and the like
[0071] Dead-end inhibition and fluorescence studies were performed to identify the binding site of Compounds 6-8 as they appeared to have very little similarity with any of the known GSNOR substrates or inhibitors. Referring now to Table 3, the inhibitors tested exhibited noncompetitive and uncompetitive inhibition against varied concentrations of either GSNO or NADH. This may indicate that neither of these substrates can completely prevent Compounds 6-8 from binding to GSNOR. This would occur if, for example, Compounds 6-8 were binding to multiple enzyme complexes occurring in the kinetic pathway (shown in Figurel). Inhibition by binding to a site outside the active site in GSNOR is unlikely since the type of inhibition by the compounds against NADH and GSNOR would have been similar. An additional dead-end inhibition study involving dodecanoic acid as inhibitor against varied GSNO was performed to determine the type of complexes that an inhibitor binding in the substrate binding site would form in the kinetic pathway of GSNO reduction. [0072] Referring now to FIG. 1, GSNOR may have a preferred kinetic pathway (shown by the bold lines) through GSNOR'NADH complex (EA) in its random mechanism during the reduction of aldehyde. In this proposed mechanism, the aldehyde (B) can bind to the free form of GSNOR (E), or preferentially to the GSNOR'NADH complex (EA) to form the competent ternary complex (EAB). The EAB complex undergoes catalysis to form the products, NAD+ (Q) and alcohol (P). After catalysis, either of the products can leave the enzyme. A GSNO inhibitor can bind to the free form of GSNOR (step 1), GSNOR'NADH (step 2) and GSN0R»NAD+ (step 3) binary complexes to form EI, EAI and EQI ternary complexes, respectively.
[0073] Referring to Table 3, dodecanoic acid was found to be a noncompetitive inhibitor against varied GSNO concentration, even though it binds at the GSNO binding site. The noncompetitive inhibition of GSNOR by dodecanoic acid can be explained on the basis of the kinetic mechanism of GSNOR (shown in Fig. 1) during the reduction of aldehyde, 12- oxododecanoic acid (12-ODDA). GSNOR has a preferred kinetic pathway through the E'NADH complex during the reduction of 12-ODDA. Consistent with this mechanism dodecanoic acid would act as a noncompetitive inhibitor against varied GSNO levels if it binds to GSNOR at more than one place in the kinetic pathway; e.g., one where it competes with GSNO to bind to the enzyme (steps 1 and 2 in Fig. 1) and one where GSNO does not normally bind in the kinetic pathway (step 3 in Fig. 1). Competition with GSNO for binding to GSNOR would involve binding the inhibitor to the E«NADH complex (step 1) and to a small extent, to the free enzyme (step 2) and give rise to the slope effect closured in the double reciprocal plot of the kinetic data. Binding to GSNOR in the kinetic pathway where GSNO does not normally bind would involve binding the inhibitor to ErNAD+ complex and will give rise to the intercept effect in the double reciprocal plot. The noncompetitive inhibition of GSNOR by Compounds 6-8 against varied concentrations of GSNO can also be explained by their forming complexes with E'NADFH and E»NADI. The uncompetitive inhibition by Compounds 6 and 8 and almost uncompetitive inhibition by Compound 7 (although inhibition by Compound 7 statistically fits noncompetitive mechanism better, the Kis value is five fold higher than the Ka value and has high standard errors) against varied NADH can be explained by the compounds binding to the E'NADH complex in the nearly ordered kinetic mechanism of GSNOR during aldehyde reduction and the high affinity of NADH (KD = 0.05 μM) for GSNOR. Both these factors would make the contribution of E«I very small in the inhibition of the enzyme under the experimental conditions and make the inhibition uncompetitive. The inhibition of GSNOR caused by binding Compounds 6-8 to the enzyme at more than one place in the kinetic pathway is similar to that shown by sulfoxide and amide inhibitors of horse liver alcohol dehydrogenase. [0074] Equilibrium binding studies were conducted to test the hypothesis formulated to account for the dead-end inhibition studies, namely, that Compounds 6-8 (Table I) were binding in the substrate (GSNO or any alcohol and aldehyde) binding site. If Compounds 6-8 were to bind in the substrate binding site, they should exclude only the substrate and not the coenzyme from the GSNOR active site.
[0075] Referring now to FIG. 2, (A) Changes in the fluorescence of NADH (curve a) upon sequential addition of GSNOR (curve b) and Compound 8 (curve c). To a solution of 1.7 μM NADH were added 2 μM GSNOR and 50 μM Compound 8 in sequence and the fluorescence of the solution measured each time (λexc = 350 run; λemm = 375-550 nm); (B) Changes in the fluorescence of NADH (curve a) upon sequential addition of GSNOR (curve b), 12-HDDA (curve c) and Compound 8 (curve d). To a solution of 1.7 μM NADH were added 2 μM GSNOR, 810 μM 12-HDDA and 50 μM Compound 8 in sequence and the fluorescence measured each time (λexc = 350 nm; λemm = 375-550 nm); (C) Binding of Compound 8 to GSNOR'NADH complex. The change in fluorescence of a 1.7 μM NADH and 2 μM GSNOR mixture (λexc = 350 nm; λemm = 455 nm) with increasing concentrations of Compound 8 was fitted to a single site binding model (equation 1 ; see materials and methods) using the Graphpad Prizm 4. All of the fluorescence studies were conducted at room temperature in 50 niM potassium phosphate pH 7.5.
[0076] Still referring to Fig. 2a, the fluorescence of NADH increases and shifts to a lower wavelength as NADH transfers from the polar environment in the solvent to the less polar environment within the GSNOR active site. The addition of Compound 8 decreased the fluorescence of NADH but interestingly, there is a blue shift in the emission peak indicating that NADH is still in the nonpolar environment of the active site (compare curve a and c in Fig. 2a). Such a quenching of the fluorescence of the dihydropyridine ring has been observed when the amide inhibitors bind to Horse liver ADH'NADH complex. Compounds 6 and 7 also quenched the NADH fluorescence while moving the emission maxima to a shorter wavelength (data not shown). This suggests that Compounds 6-8 do not exclude NADH from its binding site and are forming an E*NADH»Inhibitor complex. [0077] In order to determine the effect of Compounds 6-8 on the binding of the substrate, binding studies were conducted in the presence of the alcohol substrate, 12- hydroxydodecanoic acid (12-HDDA). The formation of GSNOR-NADH* 12-HDDA abortive ternary complex has been reported earlier. Referring to Fig. 2b curve c, 12-HDDA binds to GSNOR'NADH complex with a dissociation constant of 170 μM and increases the fluorescence of NADH in the ternary complex as shown in the figure. The addition of the same amount of Compound 8 as used in the assay reported in Fig. 2a quenches the NADH fluorescence and results in a spectrum (Fig. 2b curve d). Shown in Fig. 2b curve d, it is similar to results described with E»NADH«Compound 8 complex formation suggesting that Compound 8 has displaced 12-HDDA from the active site to form GSNOR»NADH»Compound 8 complex (i.e., curve c has higher fluorescence than that observed in Fig 2a curve c because not all of the 12-HDDA bound to the enzyme has been displaced by Compound 8). Compounds 6 and 7 also exhibit a similar effect on the fluorescence of GSNOR'NADH* 12-HDDA complex. These binding experiments demonstrate that Compounds 6-8 exclude only the alcohol/aldehyde substrate from binding into the active site.
[0078] Referring now to Fig. 2a, the fluorescence change observed upon the formation of the GSNOR»NADH»Inhibitor complex was used to determine the equilibrium dissociation constant of the inhibitors for the GSNOR'NADH complex. The equilibrium dissociation constant of Compounds 6-8 is less than 10 μM, indicating that these Compounds have high affinity for the GSNOR'NADH complex. Compounds 6 and 8 have significantly higher affinity for the GSNOR'NADH complex than Compound 7 as evidenced by their 3 to 5 fold lower equilibrium dissociation constant. Inhibition of GSNOR inside the cells
[0079] The ability of Compounds 6-8 (Table 1) to inhibit GSNOR inside the cells was tested in rat macrophages (RAW 264.7 cells). RAW cells have been used extensively as a model system for examining NO and s-nitrosothiol biochemistry. Briefly, RAW cells were treated with the inhibitors alone or in combination with GSNO and the intracellular nitrosylated species were quantitated using the triiodide-based chemiluminescence method. Referring now to Fig. 3 A, no significant amounts of nitroso species were detected in untreated cells and cells exposed only to the compounds at concentrations used in these assays. A significant buildup of nitrosylated compounds was evident in cells treated with 500 μM GSNO. The nitrosylated compounds appeared to reach an equilibrium level within 1 hour of exposure to GSNO and remained fairly constant for the duration of 6 hours. In contrast, nitrosylated compounds continued to accumulate in cells treated with GSNO and 33 μM concentrations of either Compound 6 or 8. At 6 hours the levels of nitrosylated species inside the cells treated with GSNO and Compounds 6 or 8 were 3 to 4 fold higher than the level of nitrosylated species found in cells treated with GSNO alone. Still longer exposure of cells to Compounds 6 or 8 (for up to 24 hours) resulted in an 80% decrease in the amounts of nitrosylated species measured at 4 hours (data not shown). (These results are consistent with temporary inhibition of the GSNOR rather than permanent inhibition of the enzyme.) Compound 7 (Table 1) was not as effective as Compounds 6 or 8 in inhibiting GSNOR inside the cells. This is evidenced by only a 1.3 to 1.7 fold increase in the levels of nitrosylated species and the shorter duration of its effect as judged from an insignificant difference in the levels of nitrosylated species measured at 6 hrs. An analysis of the molecular size of the nitrosylated species inside the treated cells showed that more than 95% of the nitrosylated species were greater than 5 kDa in size. Furthermore, 21 to 28 % of the nitrosylated species in treated cells were resistant to mercury pretreatment, suggesting that N-nitrosothiolated proteins were also forming inside the cells. These observations indicate that Compounds 6-8 inhibit GSNOR inside the cells. It is also evident that GSNOR regulates the degree of nitrosylation of intracellular proteins by exogenously derived nitrosylating species.
[00801 The effect of varied concentrations of compounds on the accumulation of nitrosylated compounds was examined to compare the effectiveness of Compounds 6-8 in inhibiting intracellular GSNOR. The level of intracellular nitrosylation increased with increasing concentration of the compounds in the medium. Compounds 6 and 8 are more effective at inhibiting GSNOR inside the cells than is Compound 7 as evident from the 3 fold higher nitrosylation observed at the 33 μM initial concentration. Although Compound 7 is less effective at inhibiting GSNOR inside the cells the either Compound 6 or 8, it is nevertheless capable of raising the levels of nitroso compounds to the same extent as the other compounds. [0081] The effect of GSNOR inhibition on the nitrosylation of cellular proteins was also examined using the biotin switch assay technique developed by Jeffrey et al, Nat. Cell Biol. 3, 193-197 and modified by Wang et al, Free Radic. Biol. Med. 44, 1362-1372 (2008). Referring now to FIG. 4, Compound 3 increased the nitrosylation of cellular proteins with time in RAW cells. The effects of GSNOR inhibition on the nitrosylation of cellular proteins appeared to peak around 8 hours before decreasing to normal levels within 24 hours. The accumulation of SNOs was less when cells were simultaneously treated with Compound 8 and NO synthase inhibitor, L-NAME (Fig. 4). By way of explanation and not limitation, these results suggest that the accumulation of SNOs in GSNOR inhibited cells occurred from the reaction of constitutively produced NO (by NOSs) with cellular proteins. [0082] The compound cGMP plays key roles in vascular biology. To test whether inhibition of GSNOR increases cGMP production, RAW 264.7 cells were incubated with 50μM GSNO ±, both with and without Compound 8, one of the subject GSNOR inhibitors of this invention. Next, the amount of cGMP accumulation after 10 minutes was measured. Referring now to FIG. 6, GSNO activates soluble Guanylate Cyclase as previously described {Mayer et a,. U Biol Chem, Vol. 273, Issue 6, 3264-3270, February 6, 1998). Compound 8 (GSNORi in the figure) potentiates the effect of GSNO by 2.5 fold. Without being limited by any specific explanation or hypothesis, these results suggest that one mechanism by which Compound 8 may exert its biochemical effects is due to its inhibition of GSNOR, which then potentiates the effects of GSNO on cGMP production resulting in higher cGMP levels. These data may explain at a biochemical level the ability of at least some of these compounds that inhibit GSNOR to relax isolated aortic rings as demonstrated elsewhere herein. [0083] The increase in the nitrosylation of proteins upon GSNOR inhibition may suggest that GSNOR inhibition should increase the bioactivity of NO stemming from s-nitrosylation of cellular proteins in organs as well as in cells in tissue culture. This hypothesis was tested by determining the effects of Compound 8 on the vascular tone of organ cultures of murine aorta. Compound 8 (50μM) completely relaxed the vessels within 15 min. A complete concentration response curve revealed an EC50 of 5μM for Compound 8 (Fig. 5A), with as little as 300 nM giving ~10% relaxation. Direct comparison with sodium nitroprusside (SNP) revealed that although Compound 8 was less potent than SNP at relaxing the vascular smooth muscle (Fig. 5A) and that the relaxation occurred much more slowly, the duration of the effect was much longer than with SNP. The vascular effects of SNP are immediate, which explains its clinical utility for hypertensive crises. The GSNORi mediated relaxation took around 3 minutes to begin relaxation, but at a 50μM concentration it sustained vessel relaxation for up to 2 hours, after which the integrity of the vessel preparations began to decline.
[0084] Preincubation of aortic rings with 500μM L-NAME for 30 minutes inhibited the vasorelaxation to Compound 8 by roughly 58% (Fig. 5B). The partial inhibition of Compound 8 induced relaxation of the aorta by L-NAME suggests that NO produced by the NOSs mediates the relaxation of smooth muscles observed during GSNOR inhibition. Thus, GSNOR actively regulates the bioactivity of NO by regulating the nitrosylation of cellular proteins, and this confirms the notion that RSNOs are involved in vasorelaxation in an isolated organ.
[0085] Therefore, these three compounds help define the cellular effects of inhibiting GSNOR, and potentially harness the beneficial effects of GSNOR inhibition without completely knocking-out its activity.
[0086] The accumulation of nitrosothiols by the inhibition of GSNOR is consistent with the studies of Stamler and others showing that GSNOR is the primary enzyme involved in regulating levels of s-nitrosylated proteins inside cells. In light of the debate around the sensitivity of the triiodide based quantitation of nitrosothiols, it was concluded that GSNOR inhibition per se would not lead to a large increase in the nitrosylation level of cellular proteins, at least without stimulation of NO synthases. Without being bound by any specific theory or explanation, these results suggest that partial down-modulation of GSNOR activity, rather than abrogation, may prove the most efficacious. In summary, these teachings reveal novel inhibitors of s-nitrosoglutathione reductase and many related compounds that inhibit GSNOR perhaps by binding into the GSNO binding site. At least some of these compounds bind to GSNOR at multiple places in the kinetic pathway, thereby affording a type of inhibition not easily overcome by the up-regulation of GSNO and NADH. Data collected using these compounds support the assertion that GSNOR is one of the primary enzymes involved in regulating the nitrosylation of intracellular proteins. [0087] In addition to being structurally diverse, each of the Compounds 6-8 has a free carboxyl group like many of the excellent substrates of GSNOR, including GSNO and 12- hydroxydodecanoic acid. Given the importance of Arg 115 at the base of GSNOR active site in the binding of GSNO and HMGSH, it is very likely that the free carboxyl group in Compounds 6-8 is interacting with Argl 15. By virtue of not binding in the coenzyme binding site, Compounds 6-8 will have a high probability of specifically inhibiting GSNOR among NAD(H) binding dehydrogenases. These compounds may also serve as good lead compounds for obtaining other highly potent cell-permeable GSNOR inhibitors. [0088] In order to identify additional compounds that inhibit GSNOR, and may have diagnostic research or therapeutic utility, analogues of specific compounds identified in the original assays were tested. The compounds are disclosed in Table 4. Briefly, Compoundsl2-14, 43-53, 72, 84, 86 and 72-83 are related to Compound 6 of Table 3; Compounds 24, 56, 58, 59, 62, 60-62, 64, 66, 67, 69, 70 in Table 4 and are related to Compound 7 of Table 1. [0089] A number of explanations and experiments are provided by way of explanation and not by limitation. No theory of how the novel technology operates is to be considered limiting, whether proffered by virtue of description, comparison, explanation or example. Accordingly, the following examples and discussion are presented by way of guidance and explanation and not limitation.
EXAMPLES Materials and Methods;
[0090] All the chemicals used in the experiments were purchased from Sigma- Aldrich Chemical Company. RAW 264.7 cells, DMEM medium, and fetal bovine serum were purchased from American Tissue and Cell Culture. Recombinant human GSNOR, β2 β 2-, π-, and σ- ADHs were expressed in E. coli and purified as described earlier. Synthesis of Compound 6
[0091] Depending upon the substitution pattern, the 1, 2- diarylpyrroles disclosed herein can be synthesized using the general methodology disclosed in Scheme I. The general synthetic strategy entailed the preparation of suitable 1,4-diketones followed by heating with appropriate amines in the Paal-Knorr condensation, cyclization to yield the targets. The analogs having an alkyl group (R3), Me or Et) at position 5 in the pyrrole ring were synthesized following Scheme 1. The Stetter reaction 16 of substituted benzaldehydes with R, ά-unsaturated ketones using the thiazolium salt catalyst proved very versatile and high yielding (NEt3, EtOH, reflux, 60-90%). The condensation of VII with aryl amines (Scheme I) proceeded smoothly to give good yields (50-80%) of the desired pyrroles. For additional information the reader is directed to see, for example, Journal of Medical Chemistry, 1997, Vol. 40. 40, No. 11. Synthesis of Compound 7
[0092] Depending upon the substitution pattern, Compound 7 can be synthesized by the general synthetic approach described in Scheme II. For additional information the reader is directed to see, for example, Trofimov, F. A. et al, Khimiya Geterotsiklichoskikh Soedinenii, (10) 1343-6; 1975. Synthesis of Compound 8
[0093] Depending upon the substitution pattern Compound 8 can be synthesized by the general synthetic approach described in Scheme III. For additional information the reader is directed to see, for example, J Comb. Chem. 2004, 6, 573-583. High throughput Screening: [0094] Referring now to FIG. 3, briefly, RAW 264.7 cells were incubated with 500 μM GSNO alone (T) or in presence of 33 μM Compound 6 (o) or 7 (Δ) or 8 (■). At indicated times or at 4 hr (in case of B), the cells were lysed and the lysate was analyzed for protein and nitroso species concentration by Bradford and chemiluminescence assay, respectively. For details, see Materials and methods. Data represents mean ± SE (n=3-12). [0095] Referring now to FIG. 4, RAW 264.7 cells were cultured in DMEM containing 10% heat-inactivated serum. Cells were treated with 33 μM of Compound 8 for 0, 2, 4, 8, or 24 h alone or in combination with 1 mM NAME for 4 h (lane 4+N). At indicated times, the cells were quenched and the lysate was analyzed for s-nitrosothiol content by the biotin switch assay. Equal amounts of proteins were loaded in each lane and the degree of biotinylation (and hence s-nitrosylation) determined using an anti-biotin antibody. [0096] Referring now to FIG. 5, mouse aorta segments were equilibrated in oxygenated PSS (95% O2 and 5% CO2) at 37 0C. Following equilibration, lμM phenylephrine was added to each ring for submaximal contraction. After stabilization, increasing concentrations (10""9 M to 10"4 M) of either Compound 8 or sodium nitroprusside (SNP) was added to the rings and the tone of the rings determined. (B) Inhibition of Compound 8 induced relaxation of aortal rings by NO synthase inhibitor, L-NAME. To pre-equilibrated and submaximally contracted aortal rings with phenylephrine (1 micromolar), L-NAME (500 micromolar) was added to the bath and allowed to incubate for 30 minutes. After 30 minutes, Compound 8 (50 micromolar) was added and the tone of the rings determined as described above. Each experiment was performed using two rings from three different mice and the mean (±SEM) for each response determined.
[0097] Referring now to FIG. 6, RAW 264.7 cells were incubated with 50 μM GSNO ± Compound 8 the subject GSNOR inhibitor of this invention and then measured the amount of cGMP accumulation after 10 minutes. As shown in the figure GSNO activates soluble Guanylate Cyclase as previously described (Mayer et a., J Biol Chem, Vol. 273, Issue 6, 3264-3270, February 6, 1998). One mechanism by which Compound 8 exerts its biochemical effects is due to its inhibition of GSNOR, which then potentiates the effects of GSNO on cGMP production resulting in higher cGMP levels. These data likely explain at a biochemical level the ability of all these compounds that inhibit GSNOR to relax isolated aortic rings.
[0098] Referring now to Table 1 , inhibition was studied at pH 10. These assays were performed in 0.1 M sodium glycine containing 1 mM octanol, 1 mMNAD+, 0.1 mM EDTA and 50 μM inhibitor. Inhibition studies at pH 7.5 were performed in 50 mM potassium phosphate pH 7.5 that included 15 μM NADH, 10 μM GSNO, 0.1 mM EDTA and 50 μM inhibitor. The data fit to inhibition curve consistent with partial inhibition, and a fit of the data to this model returned a hill coefficient of 2.4.
[0099] Referring now to Table 2, inhibition studies were performed in presence or absence of 5 μM inhibitor. These studies were carried out at 25 0C in 50 mM potassium phosphate pH 7.5 including 0.1 mM EDTA. The enzymes activities were measured by following the changes in absorbance at 340 nm. The values show the percent reduction in the enzyme activity (from a minimum of two measurements) caused by the inhibitor. The standard errors for this data are below 15% of the averages shown, except when the inhibition was below 20%. Studies with β2 β 2-, σσ-, π-ADH and involving Compounds 5-8 were performed in 0.05 % DMSO. Studies with GSNOR were performed in presence of 1 % DMSO, except when Compound 4 was the inhibitor. Studies with Compound 4 were performed in 0.36 % DMSO. DMSO at 0.36 % inhibited β2 β 2-, σσ-, π-ADH by 24, 18, and 9 %, respectively. Studies with β 2 β 2-ADH involved adding 3.5 μg of the enzyme to the assay mixture including 2 mM NAD+, 1 mM ethanol and the inhibitor.
[00100] Studies with σσ -ADH involved adding 0.5 μg of enzyme to the assay mixture containing 2 mM NAD+, 30 mM ethanol and the inhibitor. Studies with π-ADH involved adding 19.5 μg of the enzyme to the assay mixture including 1 mM NAD+, 35 mM ethanol and the inhibitor. Studies with GSNOR involved adding 0.1 μg of the enzyme to an assay mixture including 15 μM NADH, 5 μM GSNO and the inhibitor.
[00101] Referring now to Table 3, inhibition experiments were performed at 25 0C in 50 mM potassium phosphate (pH 7.5) including 0.1 mM EDTA. A minimum of five concentrations of the varied substrate and three inhibitor concentrations were used for each experiment. NADH and GSNO concentrations were held at 15 or 10 μM, respectively when present as a constant substrate in the assay. The K15 and Kn values are respectively the slope and intercept inhibition constants and are listed along with their associated standard errors. All data were fit to competitive (C), noncompetitive (NC), or uncompetitive (UC) inhibition models. The type of inhibition shown in the Table represents the best fit of the data to the given model as judged from F statistics analysis. The Kp value is the equilibrium dissociation constant of the inhibitor for binding to the GSNOR'NADH complex, obtained by measuring the changes in the fluorescence of GSNOR bound NADH with the addition of Inhibitor (λeXc= 350 nm; λemm = 455 nm). The dissociation constant was measured at 25 0C in 50 mM potassium phosphate pH 7.5. Each KD value is an average of three independent experiments and is shown with the associated standard error.
[00102] The screening for GSNOR inhibitors was performed using a library of 60,000 compounds from ChemDiv Inc in the Chemical Genomics Core facility at Indiana University. Screening was conducted in 384 well plates and involved incubating GSNOR with 12.5 μM compound, 1 mM each OfNAD+ and octanol in 0.1 M sodium glycine pH 10. Enzyme activity was determined by measuring the rate of production of NADH spectrophotometrically at 340 nm. Inhibition of GSNOR was calculated from the ratio of enzyme activity in the presence of compounds to that in no compound controls performed on the same assay plate. Following their identification from the high-throughput screening, the GSNOR inhibitory properties of the initial hits were confirmed at the pH 10 using 12- hydroxydodecanoic acid as the substrate and at pH 7.5 using GSNO as the substrate (see the brief description of Table 1 for details on how the assay was carried out.) Inhibition of ADH isozymes by various compounds:
[00103] Inhibition of β2 β 2-, π-, σ-, -ADH was evaluated by determining the inhibitory effect of GSNOR inhibitors on the rate of oxidation of ethanol by each of these ADH isozymes. The assay mixtures included saturating amount OfNAD+ (1-2 mM) and ethanol at its KM concentration for each of the respective enzyme. All the assays were performed at 25 0C in 50 mM potassium phosphate pH 7.5 including 0.1 mM EDTA and involved determining the rate of formation of NADH spectrophotometrically at 340 nm. Specific assay conditions for each isozyme are described in the legend of Table 2. Dead-end inhibition studies:
[00104] Inhibition experiments with the GSNOR inhibitors were conducted at 25 0C in 3 ml of 50 mM potassium phosphate (pH 7.5) containing 0.1 mM EDTA. Five different concentrations of GSNO or NADH were used when they were the varied substrates and maintained at 10 and 15 μM, respectively, when present as the nonvaried substrate. A minimum of three inhibitor concentrations were used in these assays and the rate of NADH and GSNO consumption was determined spectrophotometrically by following change in absorbance at 340 nm. The data were fit to the competitive, noncompetitive and uncompetitive inhibition models and the model that the data was chosen on the basis of F- statistics performed using the Graphpad Prizm 4.0 program. Fluorescence studies:
[00105] Fluorescence studies were conducted in 50 mM potassium phosphate pH 7.5, at room temperature using a Fluoromax-2 fluorescence spectrometer (Instruments S.A., Inc., Edison, NJ). The equilibrium dissociation constant of GSNOR inhibitors was determined by measuring the changes in the fluorescence of GSNOR bound NADH (λexc = 350 run; λemm = 455 nm) upon the addition of inhibitor. During the experiment, increasing amounts of inhibitor were added to a solution including 2 μM GSNOR and 1.7 μM NADH. The decrease in fluorescence at 455 nm with each addition of inhibitor was plotted against the final concentration of inhibitor and the data were fitted to equation 1 using nonlinear regression to obtain the dissociation constant of the inhibitor for GSNOR-NADH complex,
Figure imgf000033_0001
In equation 1, ΔFis the change in the fluorescence at 455 nm upon the addition of inhibitor. AFM is the maximum fluorescence change that was obtained from curve fitting. Ej and LT, are the concentrations of GSNOR and inhibitor, respectively. KD is the equilibrium dissociation constant for the formation of GSNOR»NADH*Inhibitor complex. The data were fitted using the Graphpad Prizm 4.0. Cell culture studies:
[00106] RAW 264.7 cells were cultured in DMEM medium supplemented with 10 % FBS, 200 U/ml of penicillin and 200 μg/mL of streptomycin. The cells were incubated at 37 0C in an atmosphere containing 5% CO2 and 95% air. For the experiments, 1-2 X 106 cells were plated in six- well plates a day before the experiment. On the day of the assay the medium was replaced with 3 ml of fresh medium and the cells were treated with compounds for a predetermined length of time. Following the incubation period, the cells were washed three times with PBS and scraped off the plate in 250 μl of lysis buffer (50 mM potassium phosphate pH 7.0 containing 50 mM NEM and ImM EDTA). Cells were lysed by sonication using a micro tip probe (three pulses of 30% duty cycle; 2 output control on a Fisher Sonicator). Cell debris was pelleted by centrifugation (10 min at 16,00Og) and the cell lysate was analyzed for protein concentration using the Bio-Rad dye-binding protein assay. The concentration of nitroso compounds in the cell lysate was determined using the triiodide based chemiluminescence method using a Sievers 280 NO analyzer. Cell lysates were treated with 15% v/v of a sulfanilamide solution (5 % w/v in 0.2 M HCl) and kept at room temperature for 5 min to remove nitrite. The triodide mixture was prepared fresh every day as described earlier and kept at 60 0C in the reaction vessel. The concentration of nitroso species was derived from a standard curve generated using GSNO. For determining the amount of the small size nitroso compounds in the cell lysate, a 5 kDa-cutoff Amicon ultra filtration unit was used according to the supplier's instructions. The amount of s-nitrosothiols in the cell lysate was determined by initially passing the cell lysate through a microspin column and treating the eluate with 5 mM HgCl2 before determining the nitroso compound concentration using chemiluminescence. For some experiments, the cells were pretreated with the compounds for 16 hours prior to the day of experiment. Later experiments showed that this pretreatment had no effect on the rate of accumulation of nitroso species inside the cells.
Determination s-nitrosothiol accumulation in RAW 264.7 cells using the biotin switch assay method:
[00107] RAW 264.7 cells were cultured in 10% heat-inactivated serum containing DMEM. Cells were treated with 33 micromolar Compound 8 for varied lengths of time alone or in combination with 1 mM NAME for 4 h (4+N). At indicated times, the cells were quenched and the lysate was analyzed for s-nitrosothiol content by the biotin switch assay as described by Jaffiey et al, Methods Enzymol. 396, 105-118 (2005) with modifications suggested by Wang et al, Free Radic. Biol. Med. 44, 1362-1372 (2008) and Zhang et al, Free Radic. Biol. Med. 38, 874-881 (2005). Briefly, free sulfydryls in -200 μg of cell lysate were blocked with 20 mM MMTS in 1 ml of HEN buffer (250 mM HEPES pH 7.7 containing 1 mM EDTA and 0.1 mM Neocupronine) containing 2% SDS at 50 0C for 20 min. Free MMTS was removed by gel-filtration spin columns and the blocked proteins were labeled with 1 mM Biotin-HPDP (Pierce) in presence or absence of 30 mM ascorbate and 2 μM CuCl for 2.5 hours. Equal amounts of proteins were loaded in each lane and the degree of biotinylation (and hence s- nitrosylation) determined using an anti-biotin antibody (SIGMA). Wire myography:
[00108] Mice were anesthetized with diethyl ether. A thoracotomy was performed to expose thoracic and abdominal aorta. A 25 gauge syringe was inserted into the apex of left ventricle and perfused free of blood with oxygenated Krebs Henseleit buffer. The right atrium was cut to provide an exit for blood. The aorta was removed and cleaned of fat and adventitia. The aorta was cut into 2-mm-long segments and mounted on a four-channel wire myograph (AD Instruments). Vessel rings were maintained in 10-ml organ baths with oxygenated PSS (95% O2 and 5% CO2) at 37 0C. Rings were allowed to equilibrate for 80 minutes with the buffer in each organ bath changed every 20 min. One gram pretension was placed on each aortic ring (appropriate starting tension for optimal vasomotor function as determined in previous experiments). An eight-channel octal bridge (Powerlab) and data-acquisition software (Chart version 5.2.2) were used to record all force measurements. After equilibration for 80 min, lμM 27 phenylephrine was added to each ring for submaximal contraction. After stabilization, either Compound 8 or sodium nitroprusside (SNP) was added to the rings and the tone of the rings determined. For the determination of SNP and ACh dose-response relationships, aortic rings were precontracted with 10-6 M PE, and SNP or Compound 8 was then added in increasing concentrations from 10-9 M to 10^4- M. In a subset of experiments, L-NAME was added (500μM final) to the bath and allowed to incubate for 30 minutes. After 30 minutes, Compound 8 was added and the tone of the rings determined as described above. [00109] While the novel technology has been illustrated and described in detail in the figures and foregoing description, the same is to be considered as illustrative and not restrictive in character, it being understood that only the preferred embodiments have been shown and described and that all changes and modifications that come within the spirit of the novel technology are desired to be protected. As well, while the novel technology was illustrated using specific examples, theoretical arguments, accounts, and illustrations, these illustrations and the accompanying discussion should by no means be interpreted as limiting the novel technology. All patents, patent applications, and references to texts, scientific treatises, publications, and the like referenced in this application are incorporated herein by reference in their entirety.

Claims

CLAIMS WHAT IS CLAIMED IS:
1. A method of altering enzyme activity, comprising the steps of: providing at least one compound selected from the group consisting of:
Figure imgf000036_0001
, H ;
Figure imgf000036_0002
H, CH3 , F, Cl, Br, OCH3, C -CH,
R5 = -H, -CH3, -C2H5, -C3H7
Figure imgf000036_0003
R3= -C2H5, -C4H9, \J
R4= -OC2H5, -OCH3, -C2H5, -F, -Cl, -Br, -OCH3, -COOC2H5
R-5~ ~H.S -0x13, -C2AX5., -O3H7
Figure imgf000037_0001
R1 - -H, -CH3, -C2H5, -C3H7 R2 = -H, -CN, -OH, -CH2OH . and
Figure imgf000037_0002
"v^-CH^ \^C2Hg V^CsH7
R^ -H- S S o or a physiologically acceptable salt thereof; and contacting said compound with s-nitrosoglutathione reductase.
2. A method of treating a disease or a condition, comprising the steps of: providing at least one compound according to claim 1 , and administering a therapeutically effective dose of said compound to a patient in need thereof.
3. The method of treating a disease or a condition according to claim 1, wherein said disease or condition is selected from the group consisting of: systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis, impotence and abnormalities of ejaculation, cystic fibrosis and sickle cell anemia, Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, multiple sclerosis, systemic inflammatory response syndrome, rheumatoid arthritis, Sjogren's syndrome, ulcerative colitis, Crohn's disease, glomerulonephritis, psoriasis, cutaneous lupus erythematosus, systemic lupus erythematosus, systemic sclerosis, dermatitis, and other inflammatory diseases, hematologic tumors and solid tumors.
4. The method according to claim 2, wherein the therapeutically effective amount of said compound is in the range of about 0.01 mg/kg per of body mass day to about 1000 mg/kg of body mass per day.
5. A method of diagnosing a disease or a condition, comprising the steps of: providing at least one compound according to claim 1 ; contacting said compound with s-nitrosoglutathione reductase; and observing a change in conformity with a change in s-nitrosylation.
6. The method of diagnosing, according to claim 5, wherein the disease or condition being diagnosed is selected from the group consisting of: systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery restenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease, exercise- induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis, impotence and abnormalities of ejaculation, cystic fibrosis and sickle cell anemia, Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, multiple sclerosis, systemic inflammatory response syndrome, rheumatoid arthritis, Sjogren's syndrome, ulcerative colitis, Crohn's disease, glomerulonephritis, psoriasis, cutaneous lupus erythematosus, systemic lupus erythematosus, systemic sclerosis, dermatitis, and other inflammatory diseases, hematologic tumors and solid tumors.
7. A method of studying protein nitrosylation comprising the steps of: providing at least one compound according to claim 1; contacting said compound with s-nitrosoglutathione reductase; and observing a change in s-nitrosylation.
8. A kit for altering the activity of s-nitrosoglutathione reductase, comprising: at least one compound according to claim 1.
9. A method of treating a disease or a condition, comprising the steps of: providing at least one compound selected from the group consisting of:
Figure imgf000039_0001
a pharmaceutically acceptable salt or ester thereof; and administering a therapeutically effective dose of said compound to a patient in need thereof.
10. The method of treating a disease or a condition according to claim 9, wherein said disease or condition is selected from the group consisting of: systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery re-stenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease, exercise-induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis, impotence and abnormalities of ejaculation, cystic fibrosis and sickle cell anemia, Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, multiple sclerosis, systemic inflammatory response syndrome, rheumatoid arthritis, Sjogren's syndrome, ulcerative colitis, Crohn's disease, glomerulonephritis, psoriasis, cutaneous lupus erythematosus, systemic lupus erythematosus, systemic sclerosis, dermatitis, and other inflammatory diseases, hematologic tumors and solid tumors.
11. The method according to claim 9, wherein the therapeutically effective amount of said compound is in the range of about 0.01 mg/kg per of body mass day to about 1000 mg/kg of body mass per day.
12. A method of diagnosing a disease or a condition, comprising the steps of: providing at least one compound according to claim 9; contacting said compound with s-nitrosoglutathione reductase; and observing a change in conformity with a change in s-nitrosylation.
13. The method of diagnosing, according to claim 12, wherein the disease or condition being diagnosed is selected from the group consisting of: systemic hypertension, pulmonary artery hypertension, coronary atherosclerosis, systemic atherosclerosis, coronary artery restenosis, cardiac failure, congestive heart failure, cardiogenic shock, cardiomyopathy, septic shock, toxic shock syndrome, cerebrovascular accidents (CVAs), embolic disease, exercise- induced asthma, bronchiectasis, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, and lung disease due to cystic fibrosis, impotence and abnormalities of ejaculation, cystic fibrosis and sickle cell anemia, Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, multiple sclerosis, systemic inflammatory response syndrome, rheumatoid arthritis, Sjogren's syndrome, ulcerative colitis, Crohn's disease, glomerulonephritis, psoriasis, cutaneous lupus erythematosus, systemic lupus erythematosus, systemic sclerosis, dermatitis, and other inflammatory diseases, hematologic tumors and solid tumors.
14. A method of studying protein nitrosylation comprising the steps of: providing at least one compound according to claim 9; contacting said compound with s-nitrosoglutathione reductase; and observing a change in s-nitrosylation.
15. A method of studying protein nitrosylation comprising the steps of: providing at least one compound according to claim 9; contacting said compound with s-nitrosoglutathione reductase; and observing a change in s-nitrosylation.
16. A kit for altering the activity of s-nitrosoglutathione reductase, comprising: at least one compound according to claim 9.
PCT/US2008/086738 2007-12-13 2008-12-13 Materials and methods for inhibiting mammalian s-nitrosoglutathione reductase WO2009076665A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP2010538218A JP2011507811A (en) 2007-12-13 2008-12-13 Materials and methods for inhibiting mammalian S-nitrosoglutathione reductase
EP08859998A EP2229451A4 (en) 2007-12-13 2008-12-13 Materials and methods for inhibiting mammalian s-nitrosoglutathione reductase
US12/813,721 US9198909B1 (en) 2007-12-13 2010-06-11 Materials and methods for inhibiting mamalian S-nitrosoglutathione reductase

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US1352207P 2007-12-13 2007-12-13
US61/013,522 2007-12-13
US2178108P 2008-01-17 2008-01-17
US61/021,781 2008-01-17

Publications (1)

Publication Number Publication Date
WO2009076665A1 true WO2009076665A1 (en) 2009-06-18

Family

ID=40755911

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/086738 WO2009076665A1 (en) 2007-12-13 2008-12-13 Materials and methods for inhibiting mammalian s-nitrosoglutathione reductase

Country Status (4)

Country Link
US (1) US9198909B1 (en)
EP (2) EP2229451A4 (en)
JP (1) JP2011507811A (en)
WO (1) WO2009076665A1 (en)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010019910A1 (en) 2008-08-15 2010-02-18 N30 Pharmaceuticals, Llc Novel pyrrole inhibitors of s-nitrosoglutathione reductase as therapeutic agents
WO2010107476A1 (en) * 2009-03-19 2010-09-23 Duke University Inhibiting gsnor
WO2011038204A1 (en) * 2009-09-25 2011-03-31 N30 Pharmaceuticals, Llc Novel dihydropyrimidin-2(1h)-one compounds as s-nitrosoglutathione reductase inhibitors
WO2011100433A1 (en) * 2010-02-12 2011-08-18 N30 Pharmaceuticals, Llc Novel s-nitrosoglutathione reductase inhibitors
JP2012500217A (en) * 2008-08-15 2012-01-05 エヌサーティー・ファーマシューティカルズ,エルエルシー Novel pyrrole inhibitors of S-nitrosoglutathione reductase as therapeutic agents
WO2012039718A1 (en) * 2010-09-24 2012-03-29 N30 Pharmaceuticals, Llc Novel dihydropyrimidin-2(1h)-one compounds as neurokinin-3 receptor antagonists
EP2512247A1 (en) * 2009-12-16 2012-10-24 N30 Pharmaceuticals, LLC Novel thiophene inhibitors of s-nitrosoglutathione reductase
EP2533637A1 (en) * 2010-02-12 2012-12-19 N30 Pharmaceuticals, Inc. Chromone inhibitors of s-nitrosoglutathione reductase
JP2013542937A (en) * 2010-10-08 2013-11-28 エヌ30 ファーマシューティカルズ,インコーポレイテッド Novel substituted quinoline compounds as S-nitrosoglutathione reductase inhibitors
US8642628B2 (en) 2008-08-15 2014-02-04 N30 Pharmaceuticals, Inc. Pyrrole inhibitors of S-nitrosoglutathione reductase
US8785643B2 (en) 2010-12-16 2014-07-22 N30 Pharmaceuticals, Inc. Substituted bicyclic aromatic compounds as S-nitrosoglutathione reductase inhibitors
US8946434B2 (en) 2010-07-16 2015-02-03 N30 Pharmaceuticals, Inc. Dihydropyridin-2(1H)-one compound as S-nirtosoglutathione reductase inhibitors and neurokinin-3 receptor antagonists
CN105121437A (en) * 2013-03-15 2015-12-02 发现生物医药公司 Coumarin derivatives and methods of use in treating cystic fibrosis, chronic obstructive pulmonary disease, and misfolded protein disorders
WO2017077535A1 (en) * 2015-11-02 2017-05-11 Carmel-Haifa University Economic Corporation Ltd. Apoptosis related protein in the tgf-beta signaling pathway (arts) mimetic compounds, compositions, methods and uses thereof in induction of differentiation and/or apoptosis of premalignant and malignant cells, thereby restoring their normal-like phenotype
WO2018208793A1 (en) * 2017-05-08 2018-11-15 Musc Foundation For Research Development S-nitrosoglutathiome (gsno) and gsno reductase inhibitors for use in therapy
US10369145B2 (en) 2013-03-15 2019-08-06 Discoverybiomed, Inc. Coumarin derivatives and methods of use in treating hyperproliferative diseases
US10399946B2 (en) 2015-09-10 2019-09-03 Laurel Therapeutics Ltd. Solid forms of an S-Nitrosoglutathione reductase inhibitor
WO2021209689A1 (en) * 2020-04-17 2021-10-21 Helsingin Yliopisto Compounds and compositions for treating sweet potato against sweet potato pathogenic viruses

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112574193B (en) * 2020-12-31 2022-05-17 南京医科大学 Oral GSNOR inhibitor and pharmaceutical application thereof
CN115804829B (en) * 2022-11-11 2023-12-12 广州国家实验室 Use of S-nitrosylated glutathione reductase inhibitors for improving pulmonary fibrosis angiogenesis

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4792568A (en) * 1986-04-14 1988-12-20 Rorer Pharmaceutical Corporation Aryl pyrroles as useful antiallergy compounds
US20050014697A1 (en) * 2003-06-04 2005-01-20 Stamler Jonathan S. Compositions and methods for modulating S-nitrosoglutathione reductase

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5137910A (en) * 1989-05-05 1992-08-11 C.D. Searle & Co. Compositions containing indole-2-carboxylate compounds for treatment of CNS disorders
US5229413A (en) 1989-05-05 1993-07-20 G. D. Searle & Co. Compositions containing indole-2-carboxylate compounds for treatment of CNS disorders
PT93943A (en) * 1989-05-05 1991-02-08 Searle & Co PROCESS FOR THE PREPARATION OF COMPOUNDS CONTAINING INDOLE-2-CARBOXYLATE COMPOUNDS FOR TREATMENT OF CNS DISTURBACTIONS
US7179791B2 (en) * 2001-01-11 2007-02-20 Duke University Inhibiting GS-FDH to modulate NO bioactivity
US20070135454A1 (en) * 2003-11-14 2007-06-14 Tracy Bayliss Bicyclic pyrimidin-4(3h)-ones and analogues and derivatives thereof which modulate the function of the vanilloid-1-receptor(vr1)
EP2139855A1 (en) * 2007-03-22 2010-01-06 NeuroSearch A/S Medicaments useful as potassium channel modulators
EP2134176A4 (en) * 2007-03-30 2012-08-29 Brigham & Womens Hospital Compounds and methods for enhancing mhc class ii therapies

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4792568A (en) * 1986-04-14 1988-12-20 Rorer Pharmaceutical Corporation Aryl pyrroles as useful antiallergy compounds
US20050014697A1 (en) * 2003-06-04 2005-01-20 Stamler Jonathan S. Compositions and methods for modulating S-nitrosoglutathione reductase

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2229451A4 *

Cited By (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3069721A1 (en) * 2008-08-15 2016-09-21 Nivalis Therapeutics, Inc. Novel pyrrole inhibitors of s-nitrosoglutathione reductase as therapeutic agents
EP2318006A1 (en) * 2008-08-15 2011-05-11 N30 Pharmaceuticals, LLC Novel pyrrole inhibitors of s-nitrosoglutathione reductase as therapeutic agents
US8642628B2 (en) 2008-08-15 2014-02-04 N30 Pharmaceuticals, Inc. Pyrrole inhibitors of S-nitrosoglutathione reductase
JP2015071617A (en) * 2008-08-15 2015-04-16 エヌサーティー・ファーマシューティカルズ・インコーポレーテッド Novel pyrrole inhibitors of s-nitrosoglutathione reductase as therapeutic agents
EP2315591A4 (en) * 2008-08-15 2013-01-02 N30 Pharmaceuticals Inc Novel pyrrole inhibitors of s-nitrosoglutathione reductase as therapeutic agents
US9814700B2 (en) 2008-08-15 2017-11-14 Nivalis Therapeutics, Inc. Pyrrole inhibitors of S-nitrosoglutathione reductase as therapeutic agents
EP2318006A4 (en) * 2008-08-15 2011-12-28 N30 Pharmaceuticals Llc Novel pyrrole inhibitors of s-nitrosoglutathione reductase as therapeutic agents
JP2012500217A (en) * 2008-08-15 2012-01-05 エヌサーティー・ファーマシューティカルズ,エルエルシー Novel pyrrole inhibitors of S-nitrosoglutathione reductase as therapeutic agents
US9498466B2 (en) 2008-08-15 2016-11-22 Nivalis Therapeutics, Inc. Pyrrole inhibitors of S-nitrosoglutathione reductase as therapeutic agents
US8957105B2 (en) 2008-08-15 2015-02-17 N30 Pharmaceuticals, Inc. Pyrrole inhibitors of S-nitrosoglutathione reductase as therapeutic agents
US8846736B2 (en) 2008-08-15 2014-09-30 N30 Pharmaceuticals, Inc. Pyrrole inhibitors of S-nitrosoglutathione reductase as therapeutic agents
US9029402B2 (en) 2008-08-15 2015-05-12 Nivalis Therapeutics, Inc. Pyrrole inhibitors of S-nitrosoglutathione reductase
EP2315591A1 (en) * 2008-08-15 2011-05-04 N30 Pharmaceuticals, LLC Novel pyrrole inhibitors of s-nitrosoglutathione reductase as therapeutic agents
US8691816B2 (en) 2008-08-15 2014-04-08 N30 Pharmaceuticals, Inc. Pyrrole inhibitors of S-nitrosoglutathione reductase as therapeutic agents
US8686015B2 (en) 2008-08-15 2014-04-01 N30 Pharmaceuticals, Inc. Pyrrole inhibitors of S-nitrosoglutathione reductase as therapeutic agents
US8673961B2 (en) 2008-08-15 2014-03-18 N30 Pharmaceuticals, Inc. Pyrrole inhibitors of S-nitrosoglutathione reductase as therapeutic agents
US9180119B2 (en) 2008-08-15 2015-11-10 Nivalis Therapeutics, Inc. Pyrrole inhibitors of S-nitrosoglutathione reductase as therapeutic agents
US8470857B2 (en) 2008-08-15 2013-06-25 N30 Pharmaceuticals, Inc. Pyrrole inhibitors of S-nitrosoglutathione reductase as therapeutic agents
WO2010019910A1 (en) 2008-08-15 2010-02-18 N30 Pharmaceuticals, Llc Novel pyrrole inhibitors of s-nitrosoglutathione reductase as therapeutic agents
US9138427B2 (en) 2008-08-15 2015-09-22 Nivalis Therapeutics, Inc. Pyrrole inhibitors of S-nitrosoglutathione reductase as therapeutic agents
WO2010107476A1 (en) * 2009-03-19 2010-09-23 Duke University Inhibiting gsnor
US9067893B2 (en) 2009-09-25 2015-06-30 Nivalis Therapeutics, Inc. Dihydropyrimidin-2(1H)-one compounds as S-nitrosoglutathione reductase inhibitors
JP2013505949A (en) * 2009-09-25 2013-02-21 エヌサーティー・ファーマシューティカルズ・インコーポレーテッド Novel dihydropyrimidin-2 (1H) -one compounds as S-nitrosoglutathione reductase inhibitors
US8741915B2 (en) 2009-09-25 2014-06-03 N30 Pharmaceuticals, Inc. Dihydropyrimidin-2(1H)-one compounds as S-nitrosoglutathione reductase inhibitors
US9283229B2 (en) 2009-09-25 2016-03-15 Nivalis Therapeutics, Inc. Dihydropyrimidin-2(1H)-one compounds as S-nitrosoglutathione reductase inhibitors
WO2011038204A1 (en) * 2009-09-25 2011-03-31 N30 Pharmaceuticals, Llc Novel dihydropyrimidin-2(1h)-one compounds as s-nitrosoglutathione reductase inhibitors
US8859611B2 (en) 2009-12-16 2014-10-14 N30 Pharmaceuticals, Inc. Thiophene inhibitors of S-nitrosoglutathione reductase
US8586624B2 (en) 2009-12-16 2013-11-19 N30 Pharmaceuticals, Inc. Thiophene inhibitors of S-nitrosoglutathione reductase
EP2512247A4 (en) * 2009-12-16 2013-05-22 N30 Pharmaceuticals Inc Novel thiophene inhibitors of s-nitrosoglutathione reductase
JP2013514363A (en) * 2009-12-16 2013-04-25 エヌサーティー・ファーマシューティカルズ・インコーポレーテッド Novel thiophene inhibitor of S-nitrosoglutathione reductase
EP2512247A1 (en) * 2009-12-16 2012-10-24 N30 Pharmaceuticals, LLC Novel thiophene inhibitors of s-nitrosoglutathione reductase
US8481590B2 (en) 2010-02-12 2013-07-09 N30 Pharmaceuticals, Inc. Chromone inhibitors of S-nitrosoglutathione reductase
AU2011215833B2 (en) * 2010-02-12 2015-07-09 Nivalis Therapeutics, Inc. Novel S-nitrosoglutathione reductase inhibitors
WO2011100433A1 (en) * 2010-02-12 2011-08-18 N30 Pharmaceuticals, Llc Novel s-nitrosoglutathione reductase inhibitors
RU2585763C2 (en) * 2010-02-12 2016-06-10 Нивалис Терапьютикс,Инк. Novel s-nitrosoglutathione reductase inhibitors
US9717706B2 (en) 2010-02-12 2017-08-01 Nivalis Therapeutics, Inc. Chromone inhibitors of S-nitrosoglutathione reductase
US8759548B2 (en) 2010-02-12 2014-06-24 N30 Pharmaceuticals, Inc. S-nitrosoglutathione reductase inhibitors
US9707212B2 (en) 2010-02-12 2017-07-18 Nivalis Therapeutics, Inc. S-nitrosoglutathione reductase inhibitors
US8669381B2 (en) 2010-02-12 2014-03-11 N30 Pharmaceuticals, Inc. Chromone inhibitors of S-nitrosoglutathione reductase
CN102869255B (en) * 2010-02-12 2016-08-31 内瓦利斯治疗公司 The novel inhibitors of GSNO reductase
CN102869255A (en) * 2010-02-12 2013-01-09 N30医药品公司 Novel S-nitrosoglutathione reductase inhibitors
EP2533637A4 (en) * 2010-02-12 2013-07-17 N30 Pharmaceuticals Inc Chromone inhibitors of s-nitrosoglutathione reductase
EP2533637A1 (en) * 2010-02-12 2012-12-19 N30 Pharmaceuticals, Inc. Chromone inhibitors of s-nitrosoglutathione reductase
JP2013519680A (en) * 2010-02-12 2013-05-30 エヌサーティー・ファーマシューティカルズ・インコーポレーテッド Novel S-nitrosoglutathione reductase inhibitor
US9187447B2 (en) 2010-02-12 2015-11-17 Nivalis Therapeutics, Inc. S-nitrosoglutathione reductase inhibitors
US8946434B2 (en) 2010-07-16 2015-02-03 N30 Pharmaceuticals, Inc. Dihydropyridin-2(1H)-one compound as S-nirtosoglutathione reductase inhibitors and neurokinin-3 receptor antagonists
US9283213B2 (en) 2010-07-16 2016-03-15 Nivalis Therapeutics, Inc. Dihydropyridin-2(1H)-one compounds as S-nitrosoglutathione reductase inhibitors and neurokinin-3 receptor antagonists
US8906933B2 (en) 2010-09-24 2014-12-09 N30 Pharmaceuticals, Inc. Dihydropyrimidin-2(1H)-one compounds as neurokinin-3 receptor antagonists
WO2012039718A1 (en) * 2010-09-24 2012-03-29 N30 Pharmaceuticals, Llc Novel dihydropyrimidin-2(1h)-one compounds as neurokinin-3 receptor antagonists
US9139528B2 (en) 2010-10-08 2015-09-22 Nivalis Therapeutics, Inc. Substituted quinoline compounds as S-nitrosoglutathione reductase inhibitors
US9433618B2 (en) 2010-10-08 2016-09-06 Nivalis Therapeutics, Inc. Substituted quinoline compounds as S-nitrosoglutathione reductase inhibitors
US9315462B2 (en) 2010-10-08 2016-04-19 Nivalis Therapeutics, Inc. Substituted quinoline compounds as S-nitrosoglutathione reductase inhibitors
JP2013542937A (en) * 2010-10-08 2013-11-28 エヌ30 ファーマシューティカルズ,インコーポレイテッド Novel substituted quinoline compounds as S-nitrosoglutathione reductase inhibitors
US9856219B2 (en) 2010-10-08 2018-01-02 Nivalis Therapeutics, Inc. Substituted quinoline compounds as S-nitrosoglutathione reductase inhibitors
US8921562B2 (en) 2010-10-08 2014-12-30 N30 Pharmaceuticals, Inc. Substituted quinoline compounds as S-nitrosoglutathione reductase inhibitors
US8785643B2 (en) 2010-12-16 2014-07-22 N30 Pharmaceuticals, Inc. Substituted bicyclic aromatic compounds as S-nitrosoglutathione reductase inhibitors
US9249132B2 (en) 2010-12-16 2016-02-02 Nivalis Therapeutics, Inc. Substituted bicyclic aromatic compounds as S-nitrosoglutathione reductase inhibitors
US9364481B2 (en) 2010-12-16 2016-06-14 Nivalis Therapeutics, Inc. Substituted bicyclic aromatic compounds as S-nitrosoglutathione reductase inhibitors
US9221810B2 (en) 2010-12-16 2015-12-29 Nivalis Therapeutics, Inc. Substituted bicyclic aromatic compounds as S-nitrosoglutathione reductase inhibitors
US9012646B2 (en) 2010-12-16 2015-04-21 Nivalis Therapeutics, Inc. Substituted bicyclic aromatic compounds as S-nitrosoglutathione reductase inhibitors
CN105121437A (en) * 2013-03-15 2015-12-02 发现生物医药公司 Coumarin derivatives and methods of use in treating cystic fibrosis, chronic obstructive pulmonary disease, and misfolded protein disorders
US9815825B2 (en) 2013-03-15 2017-11-14 Discoverybiomed, Inc. Coumarin derivatives and methods of use in treating cystic fibrosis, chronic obstructive pulmonary disease, and misfolded protein disorders
EP2970248A4 (en) * 2013-03-15 2017-01-11 Discoverybiomed Inc. Coumarin derivatives and methods of use in treating cystic fibrosis, chronic obstructive pulmonary disease, and misfolded protein disorders
CN105121437B (en) * 2013-03-15 2018-12-04 发现生物医药公司 Coumarin derivative and method for treating cystic fibrosis, chronic obstructive pulmonary disease and misfolded protein matter illness
US10369145B2 (en) 2013-03-15 2019-08-06 Discoverybiomed, Inc. Coumarin derivatives and methods of use in treating hyperproliferative diseases
US10399946B2 (en) 2015-09-10 2019-09-03 Laurel Therapeutics Ltd. Solid forms of an S-Nitrosoglutathione reductase inhibitor
WO2017077535A1 (en) * 2015-11-02 2017-05-11 Carmel-Haifa University Economic Corporation Ltd. Apoptosis related protein in the tgf-beta signaling pathway (arts) mimetic compounds, compositions, methods and uses thereof in induction of differentiation and/or apoptosis of premalignant and malignant cells, thereby restoring their normal-like phenotype
US11866409B2 (en) 2015-11-02 2024-01-09 Carmel-Haifa University Economic Corporation Ltd. Apoptosis related protein in the tgf-beta signaling pathway (ARTS) mimetic compounds, compositions, methods and uses thereof in induction of differentiation and/or apoptosis of premalignant and malignant cells, thereby restoring their normal-like phenotype
WO2018208793A1 (en) * 2017-05-08 2018-11-15 Musc Foundation For Research Development S-nitrosoglutathiome (gsno) and gsno reductase inhibitors for use in therapy
CN110891586A (en) * 2017-05-08 2020-03-17 Musc研究发展基金会 S-nitrosoglutathione (GSNO) and GSNO reductase inhibitors for use in therapy
US10925858B2 (en) 2017-05-08 2021-02-23 The US Government as represented by the Department of Veterans Affairs S-nitrosoglutathione (GSNO) and GSNO reductase inhibitors for use in therapy
WO2021209689A1 (en) * 2020-04-17 2021-10-21 Helsingin Yliopisto Compounds and compositions for treating sweet potato against sweet potato pathogenic viruses

Also Published As

Publication number Publication date
US9198909B1 (en) 2015-12-01
EP2229451A1 (en) 2010-09-22
JP2011507811A (en) 2011-03-10
EP2581451A1 (en) 2013-04-17
EP2229451A4 (en) 2012-06-13
EP2581451B1 (en) 2016-03-09

Similar Documents

Publication Publication Date Title
EP2581451B1 (en) Compounds for inhibiting mammalian s-nitrosoglutathione reductase
KR100394157B1 (en) Treatment for atherosclerosis and other cardiovascular and inflammatory diseases
US9289415B2 (en) Treatment of cancer
Heitzer et al. Increased NAD (P) H oxidase-mediated superoxide production in renovascular hypertension: evidence for an involvement of protein kinase C
US7414139B2 (en) Catalytic antioxidants and methods of use
JP3188715B2 (en) Hexahydro-5-imino-1,4-heteroazepine derivatives as inhibitors of nitric oxide synthase
KR20080069231A (en) Pharmaceutical combination comprising atorvastatin derivatives
Stykel et al. Nitrosative stress in Parkinson’s disease
KR20210031931A (en) Use of sGC stimulants for the treatment of mitochondrial disorders
Oberle et al. Heme oxygenase-1 induction may explain the antioxidant profile of pentaerythrityl trinitrate
Sjakste et al. Endothelium-and nitric oxide-dependent vasorelaxing activities of gamma-butyrobetaine esters: possible link to the antiischemic activities of mildronate
US9012492B2 (en) Apocynin-lipoic acid conjugates and uses thereof
WO2023086561A1 (en) Combination of a ssao inhibitor and thr-beta agonist for use in the treatment of liver disorders
Fuchi et al. Artificial host molecules to covalently capture 8-Nitro-cGMP in neutral aqueous solutions and in cells
US20070010477A1 (en) Acyl homoserine lactones for inhibition of cell growth
US6485941B1 (en) Inhibition of the carboxyltransferase component of acetyl-CoA carboxylase, and the use of such inhibition in anti-cancer and anti-lipogenic therapies
WO2020117942A1 (en) Choline metabolism inhibitors
Zhang et al. Berberine Reverses Nitroglycerin Tolerance through Suppressing Protein Kinase C Alpha Activity in Vascular Smooth Muscle Cells
JP2007191451A (en) Prolyl hydroxylase inhibitor
Gendron et al. Redox‐Active Agents in Reactions Involving the Trypanothione/Trypanothione Reductase‐based System to Fight Kinetoplastidal Parasites
Marks The 1986 Upjohn award lecture. Interaction of chemicals with hemoproteins: implications for the mechanism of action of porphyrinogenic drugs and nitroglycerin
Ogunrombi et al. Neurotoxicity studies with the monoamine oxidase B substrate 1-methyl-3-phenyl-3-pyrroline
US20210393677A1 (en) Compounds for modulation and as functional replacement of alphaketoglutaric acid (2og)-dependent oxygenases
AU733198B2 (en) Treatment for atherosclerosis and other cardiovascular and inflammatory diseases
Liao Discovery Of Piperlongumine Derivatives As Anti-Leukemic And Anti-Prostate Cancer Agents

Legal Events

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

Ref document number: 08859998

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2010538218

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2008859998

Country of ref document: EP