US20100324107A1 - USE OF PHTHALIMIDE AND/OR SULPHONAMIDE DERIVATIVES IN THE TREATMENT OF DISEASES WHICH REQUIRE REDUCING THE TNF-alpha LEVELS AND AN EXOGENOUS SOURCE OF NITRIC OXIDE, PHTHALIMIDE DERIVATIVES, SULPHONAMIDE DERIVATIVES, AND A METHOD FOR OBTAINING A SULPHONAMIDE DERIVATIVE - Google Patents

USE OF PHTHALIMIDE AND/OR SULPHONAMIDE DERIVATIVES IN THE TREATMENT OF DISEASES WHICH REQUIRE REDUCING THE TNF-alpha LEVELS AND AN EXOGENOUS SOURCE OF NITRIC OXIDE, PHTHALIMIDE DERIVATIVES, SULPHONAMIDE DERIVATIVES, AND A METHOD FOR OBTAINING A SULPHONAMIDE DERIVATIVE Download PDF

Info

Publication number
US20100324107A1
US20100324107A1 US12/747,589 US74758908A US2010324107A1 US 20100324107 A1 US20100324107 A1 US 20100324107A1 US 74758908 A US74758908 A US 74758908A US 2010324107 A1 US2010324107 A1 US 2010324107A1
Authority
US
United States
Prior art keywords
compound
treatment
furan
thiophene
tnf
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/747,589
Other languages
English (en)
Inventor
Jean Leandro Dos Santos
Chin Chung Man
Lidia Moreira Lima
Fernando Ferreira Costa
Carolina Lanaro
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Universidade Federal do Rio de Janeiro UFRJ
Universidade Estadual de Campinas UNICAMP
Universidade Estadual Paulista Julio de Mesquita Filho UNESP
Original Assignee
Universidade Federal do Rio de Janeiro UFRJ
Universidade Estadual de Campinas UNICAMP
Universidade Estadual Paulista Julio de Mesquita Filho UNESP
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 Universidade Federal do Rio de Janeiro UFRJ, Universidade Estadual de Campinas UNICAMP, Universidade Estadual Paulista Julio de Mesquita Filho UNESP filed Critical Universidade Federal do Rio de Janeiro UFRJ
Publication of US20100324107A1 publication Critical patent/US20100324107A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41961,2,4-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • 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

Definitions

  • the present invention describes the use of to phthalimide derivatives with nitric oxide donor properties, which have important activities in increasing the gamma-globin gene expression and anti-inflammatory and analgesic activities, effective in the treatment of hematologic diseases which require reduced TNF- ⁇ levels and an exogenous source of nitric oxide. More particularly, the present invention describes the use of such phthalimide derivatives for the treatment of sickle-cell disease.
  • the sickle-cell disease is the most prevalent hematologic genetic disease known, and is characterized by a point mutation in the ⁇ -globin gene, more specifically a single nucleotide change (GTG into GAG) in the sixth codon of the ⁇ -globin gene, resulting in the substitution of a glutamic acid with valine on the surface of the ⁇ -globin chain variant ( ⁇ s -globin) (SAFO, M. K et al. J. Med. Chem. v. 47, pp. 4665-4676, 2004).
  • valine which is present in the chain, carries out hydrophobic interactions with the pocket, comprised of hydrophobic amino acids, from a neighboring Hb S molecule, which is not possible in the oxygenated state of hemoglobin, since the hydrophobic pocket is inaccessible in this condition (ADACHI, K. et al. J. Biol. Chem. v. 263, n. 12, pp. 5607-5610, 1988).
  • Hb S The polymerization of Hb S is the central process of vaso-occlusion, a characteristic of the sickle-cell disease (BUNK, H. F. N Engl J Med v. 337, pp. 762-769, 1997; KAUL D. K. et al. Blood Rev. v. 10, pp. 29-44, 1996; a) FERRONE, F. A. et al. J. Mol. Biol. v. 183, pp. 591-610, 1985. b) FERRONE, F. et al. J. Mol. Biol. v. 183, pp. 611-631, 1985; SAMUEL, R. E., et al. Blood. v. 82, pp. 3474-3481, 1993).
  • the cells containing Hb S take on a sickle shape.
  • the sickle red blood cells showed a normal adherence to the vascular endothelium, monocyte, and macrophages (DUITS, A. J. et al. Clin Immunol Immunopathol v. 81, pp. 96-98, 1996; OKPALA, I. et al. J. Eur. J. Haematol. v. 69, pp. 135-144, 2002.)
  • Vascular occlusion is the main event responsible for the clinical picture of sickle-cell disease, being the cause of pain crises and organ failure.
  • Vaso-occlusive crises initiate at the venular microcirculation, as the sickle cells become trapped.
  • the primary event that is critical for vaso-occlusion includes the adhesion of red blood cells (reticulocytes and deformed dense cells) to the venular endothelium. This adhesion leads to the formation of heterocellular aggregates (white blood cells and sickle cells), which also contribute to obstruction, resulting in local hypoxia, increase on formation of Hb S polymers, and propagation of the occlusion of the neighboring vasculature.
  • Sickle red blood cell masses repeatedly clog the vessels of the microcirculation, leading to painful vascular occlusion crises. 5% to 10% among children or young adults with sickle-sell disease show, owing to the clogging of the microcirculation vessels, symptomatic pictures of stroke, effusion or hemorrhage resulting from stenosis or aneurismal dilatation of important cerebral arteries.
  • TNF- ⁇ exerts pro-inflammatory effects, increasing the chemiotactic properties, the adherence of neutrophils to the vascular endothelium, due to the increase of adhesion molecules, stimulating the production of free radicals and the synthesis of other inflammatory mediators, such as IL-1 and PGE2.
  • TNF- ⁇ also induces changes on the coagulation and anticoagulation properties and increases the hepatic synthesis of some acute-phase reagents. Furthermore, it is an important mediator of septic syndrome and endotoxic shock, being able to suppress the biosynthesis of lipoprotein lipases and lipogenic enzymes in adipose tissue, impairing the storage of lipids on adipocytes.
  • TNF- ⁇ to change the anticoagulation properties of the vascular endothelium and to induce the pro-coagulation activity on the cellular surface of endothelium, stimulating the production of the platelet-activating factor (PAF), and increasing the leukocyte adhesion to the vascular endothelium cells, results in a increase of the resistance to the blood flow, making circulation difficult and, thus, aggravating the microvascular stasis and the deoxygenation of Hb S.
  • PAF platelet-activating factor
  • Hb F fetal hemoglobin
  • TNF- ⁇ has a major role on peripheral hyperalgesia, and its inhibition has been associated with the reduction of chronic and acute pain, which accounts for the analgesic effect of thalidomide, the first anti-TNF- ⁇ drug introduced in therapeutics (RIBEIRO, R. A. et al. Eur. J. Pharmacol., v. 391, pp. 97-103, 2000).
  • TNF- ⁇ has been shown as an important strategy for preventing vascular and inflammatory complications related with sickle-cell disease.
  • TNF- ⁇ tumor necrosis factor-alpha converting enzyme
  • infliximab neutralizing antibodies
  • HU Hydroxyurea
  • HU is the main drug available for the treatment of sickle-cell disease approved by the Food and Drug Administration (FDA) agency
  • FDA Food and Drug Administration
  • various adverse effects are associated with its prolonged use, many of which are due to its ability to interrupt the cell cycle in S and G1 phases (BUCHANAN, G. R. et al. Hematology pp. 35-47, 2004; STUART, M. J. and NAGEL, R. L. Lancet. v. 364, pp. 1343-1360, 2004.), which actions characterize it as a cytotoxic and antineoplastic agent.
  • HU hydroxyurea
  • Hb F fetal hemoglobin
  • Hb S deoxygenated sickle-cell hemoglobin
  • HU Besides inhibiting the ribonucleotide reductase, HU also exerts its action mechanism as a nitric oxide (NO) donator drug, an important mediator in maintaining the normal blood flow and pressure. It is known that HU reacts with oxy- and deoxyhemoglobin to form methemoglobin, which then reacts with another HU molecule in order to form iron-nitrosyl-hemoglobin (HbNO). The formation of HbNO involves a number of reactions of the hydroxylamine moiety in order to form NO (COKIC, V. P. et al. Blood. v. 108, n. 1. pp. 184-191, 2006).
  • NO nitric oxide
  • nitric oxide (NO) in the treatment of sickle-cell disease is based on its ability to stimulate the production of fetal hemoglobin (Hb F) through the soluble guanylate cyclase (sGC) pathway.
  • sGC soluble guanylate cyclase pathway.
  • the activation of sGC increases the expression of ⁇ -globin in erythroleukemic cells and primary human erythroblasts.
  • the inhibition of sGC prevents this increase, which suggests that the sGC pathway regulates the expression of ⁇ -globin, and consequently, the synthesis of fetal hemoglobin (Hb F).
  • NO has vasodilator effects, which aggregates beneficial effects in physiopathology and in the treatment of sickle-cell disease. (KING, S. B. Free Rad. Biol. Med. v. 37, n 6, pp. 737-744, 2004).
  • the present invention relates to the novel use of some phthalimide derivatives and sulphonamide derivatives in the preparation of alternative drugs for the treatment of diseases which involve the need of reducing the levels of the TNF- ⁇ factor and the need of an exogenous source of nitric oxide.
  • the invention described herein discloses a solution for the major limitations associated with the drug therapy of diseases which involve the need of reducing the levels of the TNF- ⁇ factor and the need of an exogenous source of nitric oxide, providing an alternative for the reduction of side and adverse effects of commonly-used compounds.
  • This invention also provides two new phthalimide derivatives which are used in the preparation of drugs for the treatment of said diseases, as well as a new process for obtaining a specific sulphonamide derivative also used in the preparation of drugs for the treatment of diseases which involve the need of reducing the levels of the TNF- ⁇ factor and the need of an exogenous source of nitric oxide.
  • the present invention overcomes the problems related with the major limitations and complications associated with the drug therapy conventionally used for the treatment of sickle-cell disease, thus improving the quality of life of the patient with sickle-cell disease.
  • the present invention provides alternatives for the treatment of diseases in which there is an involvement of the increase of the TNF- ⁇ levels and the need of an exogenous source of nitric oxide for treatment.
  • the major limitations and complications associated with the drug therapy usually employed in the treatment of sickle-cell disease could be overcome or minimized with the use of the nitric oxide donor and TNF- ⁇ modulatory phthalimide and sulphonamide derivatives, thus improving the quality of life of the patient with sickle-cell disease.
  • the present invention refers to the use of a compound of general formula (I)
  • W ⁇ H, halogen, NO 2 , NH 2 , OH, C 1 -C 6 alcoxy, C 1 -C 6 haloalcoxy, C 1 -C 6 haloalkyl;
  • R corresponds to C 1 -C 7 alkyl, 2-phenyl, 3-phenyl, 4-phenyl, 2-benzyl, 3-benzyl, 4-benzyl, 2-ethylbenzyl, 3-ethylbenzyl, 4-ethylbenzyl, benzyl, thiophene, furan, pyrrole, 2-pyridine, 3-pyridine, 4-pyridine, pyrazine, pyrimidine, benzothiophene, benzofuran, indole, quinoline, isoquinoline, naphthalene, CH 2 -2-thiophene, CH 2 -3-thiophene, CH 2 -2-furan, CH 3 CH 2 -2-thiophene, CH 3 CH
  • the invention also refers to the use of a compound of general formula (II)
  • the invention still refers to a pharmaceutical composition for the treatment of diseases which require reducing the levels of the TNF- ⁇ factor and an exogenous source of nitric oxide comprising said composition, said compound being selected among those resulting from formulae I and/or II or combinations thereof in a pharmaceutically acceptable carrier.
  • the invention also refers to a method for obtaining the compound of formula IIA
  • step b) adding ethanol to the mixture obtained in step a
  • FIG. 1 Effect of derivatives (300 ⁇ mol/Kg), via i.p, in a mouse ear edema assay induced by capsaicin. Values represent the mean and standard error of the average of 5 animals. (*p ⁇ 0.05 was considered significant at the 95% confidence level using Student's t test)
  • FIG. 2 Effect of derivatives (300 ⁇ Mol/Kg), administered orally, in a peritonitis assay induced by 3% thioglycolate in mouse. Values represent the mean and standard error of the average of 4 animals. (*p ⁇ 0.05 was considered significant at the 95% confidence level using Student's t test)
  • FIG. 3 Example of gamma-globin mRNA in the presence of example 7 at different concentrations, in the absence of hemin, at times of 24 h, 48 h, 72 h and 96 h.
  • FIG. 4 Dose-response curve of compound IIA at concentrations of 5 ⁇ M, 30 ⁇ M, 60 ⁇ M and 100 ⁇ M at times 24 h, 48 h, 72 h and 96 h in the absence of hemin.
  • FIG. 5 Cell viability of the designed compounds.
  • FIG. 6 Dosing of nitric oxide by indirect pathway (nitrite)
  • the present invention has as its main novel characteristic the use of functionalized phthalimide and/or sulphonamide derivatives in the preparation of drugs for the treatment of diseases which require reduced levels of the TNF- ⁇ factor and an exogenous source of nitric oxide.
  • the invention also has as a novel characteristic the disclosure of new functionalized phthalimide derivatives designed from the prototypes thalidomide and hydroxyurea, and designed rationally through the strategy of molecular hybridization for the treatment of said diseases.
  • the invention also comprises, as another novel characteristic, a new method for obtaining a specific sulphonamide derivative which can be used in the preparation of a drug for the treatment of diseases which require reducing the levels of the TNF- ⁇ factor and an exogenous source of nitric oxide.
  • the new derivative was obtained with good to excellent chemical yields, by employing a methodology characterized by having a few synthetic steps, from commercially-available compounds, which qualifies this methodology for industrial use.
  • the present invention refers to the use of a compound of general formula (I)
  • W ⁇ H, halogen, NO 2 , NH 2 , OH, C 1 -C 6 alcoxy, C 1 -C 6 haloalcoxy, C 1 -C 6 haloalkyl;
  • R corresponds to C 1 -C 7 alkyl, 2-phenyl, 3-phenyl, 4-phenyl, 2-benzyl, 3-benzyl, 4-benzyl, 2-ethylbenzyl, 3-ethylbenzyl, 4-ethylbenzyl, benzyl, thiophene, furan, pyrrole, 2-pyridine, 3-pyridine, 4-pyridine, pyrazine, pyrimidine, benzothiophene, benzofuran, indole, quinoline, isoquinoline, naphthalene, CH 2 -2-thiophene, CH 2 -3-thiophene, CH 2 -2-furan, CH 3 CH 2 -2-thiophene, CH 3 CH
  • the compound designated by (1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)methyl nitrate is used in the preparation of a drug for the treatment of diseases which require reducing the levels of the TNF- ⁇ factor and an exogenous source of nitric oxide.
  • Said compound has the structural formula (IA):
  • the compound designated by 2-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)ethyl nitrate is used in the preparation of a drug for the treatment of diseases which require reducing the levels of the TNF- ⁇ factor and an exogenous source of nitric oxide.
  • Said compound has the structural formula shown as follows (IB):
  • Another preferred embodiment of the invention uses the compound designated by 3-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)benzyl nitrate in the preparation of a drug for the treatment of diseases which require reducing the levels of the TNF- ⁇ factor and an exogenous source of nitric oxide.
  • Said compound has the structural formula (IC):
  • the compound designated by 4-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)benzyl nitrate is used in the preparation of a drug for the treatment of diseases which require reducing the levels of the TNF- ⁇ factor and an exogenous source of nitric oxide.
  • Said compound has the structural formula (ID):
  • the compound designated by 2-[4-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl)phenyl]ethyl nitrate is used in the preparation of a drug for the treatment of diseases which require reducing the levels of the TNF- ⁇ factor and an exogenous source of nitric oxide.
  • Said compound has the structural formula (IE):
  • the present invention also refers to the use of a compound of general formula (II)
  • R 1 corresponds to 2-phenyl, 3-phenyl, 4-phenyl, 2-benzyl, 3-benzyl, 4-benzyl, 2-ethylbenzyl, 3-ethylbenzyl, 4-ethylbenzyl, benzyl, thiophene, furan, pyrrole, 2-pyridine, 3-pyridine, 4-pyridine, pyrazine, pyrimidine, benzothiophene, benzofuran, indole, quinoline, isoquinoline, naphthalene, CH 2 -2-thiophene, CH 2 -3-thiophene, CH 2 -2-furan, CH 2 -3-furan, CH 3 CH 2 -2-thiophene, CH 3 CH 2 -3-thiophene, CH 3 CH 2 -3-thiophene
  • the compound designated by 4-amino-N-hydroxybenzenesulphonamide is used in the preparation of a drug for the treatment of diseases which require reducing the levels of the TNF- ⁇ factor and an exogenous source of nitric oxide.
  • Said compound has the structural formula (IIA):
  • the subject invention still refers to pharmaceutical compositions for the treatment of diseases which require reducing levels of the TNF- ⁇ factor and an exogenous source of nitric oxide comprising said compositions, said compounds being selected among those resulting from formulae I and/or II or combinations thereof.
  • the compositions described in the subject invention comprise compounds of general formula I and/or II or pharmaceutically acceptable salts thereof, in association with a pharmaceutically acceptable excipient.
  • compositions of the present invention may be administered in a variety of dosage forms, such as orally, in the form of tablets, capsules, sugar or tablets covered with a film, liquid solutions or suspensions; rectally in the form of suppositories; parenterally, i.e., intramuscularly, or by infusion or intravenous and/or intrathecal and/or intraspinal injection.
  • compositions to which this invention relates are usually prepared according to conventional methods and administered in a suitable pharmaceutical form.
  • Solid oral pharmaceutical forms may contain, together with the active compound, different diluents, such as lactose, dextrose, saccharose, cellulose, corn starch, potato starch, or other suitable diluents; lubricants, such as silica, talc, stearic acid, magnesium or calcium stearate, and/or polyethylene glycols or other pharmaceutically acceptable lubricants; binding agents such as starches, gum arabic, gelatin, methylcellulose, carboxymethylcellulose, polyvinylpyrrolidone, or other suitable binding agents; disaggregating agents, such as starch, alginic acid, alginates or starch or sodium glycolate, or other suitable disaggregating agents; effervescent mixtures; dyes; sugary materials; wetting agents such as lectin, polysorbates, laurylsulphates; and, generally, non-toxic pharmacologically inactive substances used in pharmaceutical formulations.
  • diluents such as lactose, dextrose
  • Liquid dispersions for oral administration may include, for example, syrups, emulsions and suspensions.
  • Syrups may contain, as a carrier, for example, saccharose or saccharose with glycerine and/or mannitol and/or sorbitol or another pharmaceutically acceptable carrier.
  • Suspensions and emulsions may contain, as a carrier, among others, a natural gum, agar, sodium alginate, pectin, methylcellulose, carboxymethylcellulose, polyvinyl alcohol or other suitable carriers.
  • Suspensions or solutions for intramuscular injection may contain, together with the active compound, a pharmaceutically acceptable carrier, i.e., sterile water, olive oil, ethyl oleate, glycols, i.e., polyethylene glycol, or other pharmaceutically acceptable carrier, and, if desired, a suitable amount of lidocaine hydrochloride.
  • Solutions for intravenous injections or infusions may contain, as a carrier, for example, sterile water, or preferably, they may be in the form of sterile salt, aqueous or isotonic solutions, or may contain, as a carrier, propylene glycol or another pharmaceutically acceptable carrier.
  • the suppositories may contain, together with the active compound, a pharmaceutically acceptable carrier, such as cocoa butter, polyethylene glycol, sorbitan polyoxyethylene, fatty acid ester surfactant, lecithin, or other pharmaceutically suitable carriers.
  • a pharmaceutically acceptable carrier such as cocoa butter, polyethylene glycol, sorbitan polyoxyethylene, fatty acid ester surfactant, lecithin, or other pharmaceutically suitable carriers.
  • the present invention also refers to a novel method for obtaining the compound designated by 4-amino-N-hydroxybenzenesulphonamide of formula (IIA)
  • step b) adding ethanol to the mixture obtained in step a
  • the synthesis of 4-amino-N-hydroxybenzenesulphonamide is carried out by adding, into a 10 mL round-bottom flask, 21.6 mg of hydroxylamine hydrochloride (0.31 mmol), 26.1 mg of sodium bicarbonate (0.31 mmol), and 0.1 ml of distilled water. Upon ceasing the elimination of CO 2 , 2 mL of ethanol was added. Next, 100 mg of 4-(1,3-dioxo-1,3-dihydro-2H-isoindol-2-yl) benzenesulphonyl chloride (0.31 mmol) were added.
  • the compound obtained (4-amino-N-hydroxybenzenesulphonamide) through the described method is used, as detailed previously, in the treatment of diseases which require reducing the levels of the TNF- ⁇ factor and an exogenous source of nitric oxide.
  • the obtained compound (4-amino-N-hydroxybenzenesulphonamide) is used in the preparation of a drug for the treatment of sickle-cell disease.
  • the present invention still refers to a new phthalimide derivative designated by 3-(1-3-dioxo-1,3-dihydro-2H-isoindol-2-yl)benzyl nitrate represented by the formula shown as follows (IC)
  • the invention also refers to another phthalimide derivative designated by 2-[4-(1-3-dioxo-1,3-dihydro-2H-isoindol-2-yl)phenyl]ethyl nitrate of general formula shown as follows (IE)
  • the compounds described in the present invention were subjected to a number of tests in order to ensure the intended activities in the treatment of diseases which require reducing the TNF- ⁇ levels and an exogenous source of nitric oxide. Particularly, the compounds were subjected to tests in order to ensure their activity as auxiliary agents in the treatment of symptoms of sickle-cell disease. The tests carried out and the results obtained are described in the following.
  • the compounds were evaluated with the AMES test in order to identify a possible mutagenicity. This test is important to obtain compounds having a lower genotoxic profile, and also guides molecular changes to obtain more safe compounds.
  • the tests were conducted with the previously described compounds of general formula IA, IB, IC, ID, IE, and IIA and are shown in tables 1, 2 and 3 below.
  • the compound of formula IA showed mutagenicity ratios (RM) of 2.45 and 2.34; in the TA100 strain, the absence of metabolic activation (112 nmol/plate), and in the TA102 strain, in the presence of metabolic activation (56 nmol/plate), respectively. At concentrations higher than 56 nmol in TA 102 (+S9), toxicity can be observed, with a reduction on the number of revertants per plate (Table 01).
  • Compound IA is an alkyl derivative which has reactive methylene carbon, i.e., the carbon atom has a positive partial charge by removal of the electron density due to the more electronegative a moieties.
  • the compound of formula IB shows, at the concentrations used, mutagenicity in TA100 strain in the presence of metabolic activation, at all tested concentrations and with the following mutagenicity ratios (MR): 0.01 ⁇ mol (3.4); 0.021 ⁇ mol (3.22); 0.042 ⁇ mol (3.8); 0.085 ⁇ mol (3.8) e 0.17 ⁇ mol (2.5) (Table 02).
  • MR mutagenicity ratios
  • the compound IC when the compound IC is compared with the compound IB, it can be observed a lower mutagenicity of the interphenylene derivative.
  • the aryl derivatives that is, those having an aromatic ring bonded to the phthalimide moiety (compounds IC, ID, IE and IIA)
  • the alkyl derivatives that is, those in which the alkyl chain is directly bonded to the phthalimide moiety (compounds IA and IB); supporting the hypothesis that steric factors hinder the access of the bionucleophile to the reactive site, modulating the mutagenicity of the synthesized derivatives.
  • the compound IIA is a sulphonamide derivative which does not have the nitrate moiety, common to all other compounds.
  • the hydroxylamine moiety has been pointed out as one of the main metabolites, generated in the reduction of the nitro group, responsible for the mutagenic activity of nitro compounds (e.g., chloramphenicol, metronidazole, and nitrofurans).
  • the compound IIA containing the hydroxylamine moiety was evaluated in order to verify the toxicophoric contribution from this moiety in the synthesized compound.
  • This derivative was subsequently reacted with phthalic anhydride to obtain a phthalimide derivative.
  • the compound ID showed in the TA100 strain, in the presence of metabolic activation and at a concentration of 3.58 ⁇ mol/plate, a mutagenicity ratio of 2.03; while in the absence of metabolic activity, at this same concentration, it showed mutagenicity signs with MR values of 1.94. At a concentration of 1.8 ⁇ mol/plate in the absence of S9, in the TA102 strain, it showed mutagenicity signs, with a MR of 1.87 (Table 03).
  • Table 03 Table 03
  • the compound IE an interphenylene derivative of compound IB, in the TA100 strain and in the presence of metabolic activation, showed, as well as compound IB, mutagenesis at concentrations of 0.12; 0.25; 0.5; 1 and 2 ⁇ mol/plate, with MR values of 2.8 ; 3.7; 4.86; 5.72 e 5.79, respectively (Table 03).
  • MR values are higher in compound IE with respect to compound IB, the latter is at a lower molar concentration.
  • the need for higher concentrations for compound IE to show mutagenesis confirms, in structural terms, what had already been observed between compounds IA and IC: the presence of phenyl bonded to the phthalimide moiety reduces the mutagenicity of the compounds.
  • Alkyl derivatives show higher mutagenicity expressed by the average of the number of revertants/plate than aryl derivatives (compounds IIA, IC and ID);
  • a mutagenicity ratio of 2 is considered low if compared to drugs used in therapeutics, such as metronidazole, which has a MR of 14.9 when tested at 58.4 ⁇ mol in TA 100 without metabolic activation (SILVA, A. T. A. et al. Mini Rev. Med. Chem., v. 5, pp. 893-914, 2005). This allows us to conclude that although there are signs of mutagenicity for the compounds, it is too low, and these results may not reflect in eukaryotic cells.
  • This assay is characterized by an acute inflammatory response of the ear, with development of edema, and it was performed in order to evaluate the anti-inflammatory activity of the synthesized compounds.
  • indomethacin was used as a control at 100 ⁇ mol/Kg, and the phthalimide derivatives were firstly evaluated at 300 ⁇ mol/Kg via i.p. From table 4, it can be observed that compounds IC and IE show an ear edema inhibition percentage higher than 64%. Compound IC showed an inhibition percentage of about 64.09% in the performed assay. The other compounds, however, show a similar activity when compared to indomethacin ( FIG. 01 ), taking into consideration the standard error.
  • an abdominal writhing assay induced by acetic acid (Table 6) was carried out.
  • compound ID has an important analgesic activity, inhibiting abdominal writhing by 66%.
  • Other compounds, such as IA, IIA and IE also have significant inhibitions of the abdominal writhing induced by acetic acid, demonstrating the analgesic potential of these compounds.
  • the analgesic activity may be related to the ability of these compounds to inhibit the cytokine TNF ⁇ , since it is known that this would be one of the mechanisms that explain the analgesia of molecules such as thalidomide.
  • Compound IE has activity in this model, increasing the gene expression of gamma-globin, in the presence or absence of hemin;
  • compound IE did not show a significantly higher activity than in the absence of hemin, when compared to the control ( FIG. 3 );
  • compound IE has an effect on the expression of gamma-globin at low concentration (5 ⁇ M e 30 ⁇ M);
  • the compound IE showed high percentages of cell viability (higher than 90%) in assays with and without hemin, demonstrating the absence of toxic effects at the concentrations used;
  • Compound IIA showed higher activity than the control in the expression of gamma-globin ( FIG. 4 ).
  • the cell viability at 0 h was 97%, and this pattern was maintained during the realization of the assay, demonstrating the absence of toxicity of the compound IIA ( FIG. 5 ).
  • TA97, TA98, TA100 and TA102 There are many genetically modified strains of Salmonella typhimurium in order to detect a prevalent type of mutation, which include: TA97, TA98, TA100 and TA102.
  • TA100 and TA102 detect mutations which cause base pair substitutions, while TA 98 and TA 97 detect changes where there is a gap in the DNA reading frame (MARON & AMES, 1983).
  • Salmonella typhimurium TA100 and TA102 strains from the mutagenicity laboratory of the “Faculdade de Ciências Farmacêuticas—UNESP Araraquara” were used. Such strains have the following characteristics: (AMES, 1983)
  • Salmonella typhimurium strains were stored in a freezer at ⁇ 80° C., in flasks for freezing with 0.9 mL of culture and 0.1 mL of DMSO as a cryoprotector agent, so as to maintain all their genetic characteristics unchanged.
  • magnesium sulphate 0.25 g of magnesium sulphate, 2.5 g of citric acid, 12.5 g of dibasic potassium phosphate, and 4.375 g of sodium and ammonium phosphate were dissolved into 16.75 mL of distilled water at 45° C. (amounts enough for 25 mL of VB solution). The solution was sterilized in an autoclave for 15 minutes at 121° C.
  • Glycosylated Minimum Agar GAA
  • agar 7.5 g was dissolved into 465 mL of distilled water, and then the solution was sterilized in an autoclave for 15 minutes at 121° C.
  • Oxoid medium 0.75 g was dissolved into 30 mL of distilled water. The solution was sterilized in an autoclave for 15 minutes at 121° C.
  • the negative control is the solvent used to dissolve the sample, using as a standard volume, the highest volume of the tested sample 100 ⁇ L, which is also the amount that is required to dissolve the maximum used concentration of the drug.
  • the positive controls are mutagenic compounds specific for each strain and test condition, with 25 ⁇ L/plate of sodium azide (1.25 ⁇ g/plate) and 100 ⁇ L of mitomycin (0.5 ⁇ g/plate) being the controls for TA100 and TA102, respectively, in the absence of metabolic activation.
  • the positive control for TA100 is 50 ⁇ L 2-antramine (1.25 ⁇ g/plate) and for TA102 it is 50 ⁇ L 2-aminofluorene (1.25 ⁇ g/plate).
  • the preincubation method was used.
  • the GMA distributed to the plates was left under rest for 48 hours in an oven at 37° C. for subsequent use.
  • Salmonella typhimurium (TA100 and TA 102) strains were inoculated individually with a platinum loop, in the respective nutrient broths and maintained at 37° C., under constant stirring (160 rpm) during 14 hours, in order to achieve a density of 1 to 2 ⁇ 10 9 bacteria/mL.
  • the final data obtained from the assay was analyzed using the statistical software Salanal ( Salmonella Assay Analysis) version 1.0 from the Research Triangle Institute, RTP, North Carolina, USA.
  • Salanal Salmonella Assay Analysis
  • Such software allows the dose-response effect to be evaluated by means of analysis of variance (ANOVA-test F) computations between the measurement of the number of revertants at the different tested concentrations (doses) and the negative control, followed by a linear regression.
  • the software model chosen for analyzing the data was Bernstein's (BERNSTEIN, L. et al. Mutat. Res. v. 97, p. 267-281, 1982.).
  • the slope of the straight line from the linear portion of the dose-response curve is also provided by this software and corresponds to the number of revertants induced per measurement unit of the analyzed sample.
  • MR mutagenicity ratio
  • MR average number of revertants per test plate (spontaneous+induced revertants)
  • the spontaneous growing means that the number of revertants which developed on the plate, regardless of being induced or not, wherein values higher or equal to 2 are considered as a positive response (VALENT, G. V. et al. Env. Toxicol. Water Quality. v. 8, p. 371-381, 1993.).
  • the mutagenicity test with metabolic activation system was performed with a microsomal fraction S9 (S9 mix) prepared from a liver homogenizate from Sprague Dawley rats, previously treated with Aroclor 1254, acquired in the freeze-dried form.
  • the S9 mixture has a viability of 4 hours from preparation when put on ice.
  • the plates are then incubated for 48 hours at 37° C. After the required time has elapsed, the counting of the revertant colonies was carried out. All tested concentrations, positive and negative controls were run in triplicate.
  • the human leukemia cell line K562 ATCC American Type Culture Collection), Philadelphia, Pa., USA was used.
  • the cells were cultured in DMEM medium (Dulbecco's Modified Eagle Medium, Invitrogen, USA) containing 10% of fetal bovine serum and glutamine.
  • DMEM medium Dulbecco's Modified Eagle Medium, Invitrogen, USA
  • the cells were maintained at 37° C. under a 5% CO 2 atmosphere.
  • the cells were incubated at a density of 1 ⁇ 10 5 cells/mL. In order to carry out the culture with hemin (30 uM), it was added 72 hours before the beginning of the experiment with the desired compound.
  • the 0-hour time consisted in removing non-treated K562 cells. From this point, the respective compounds were added at the desired concentrations (5, 30, 60 and 100 uM), the cells were then maintained for 7 days under culture, without a to new addition of any compound or substitution of the culture medium. Cell collections were performed at the following times: 0, 24, 48, 72 and 96 hours. The morphology of the cells was analyzed at these points through cytospin slides stained with Leishman and the cell viability was performed by staining with trypan blue in a Neuberger's chamber.
  • RNA from K562 The extraction method with TRIzol reagent (Gibco-BRL, Gaithersburg, Md.) according to the manufacturer's instructions was used in order to obtain the RNA from K562.
  • the sample containing K562 and TRIzol was incubated for 5 minutes at room temperature in order to achieve complete dissociation of the nucleoproteic complexes, 200 ⁇ L of chloroform (CHCl 3 ) was added and thoroughly stirred, and incubation was again performed by 5 minutes at room temperature. After centrifugation for 15 minutes at 19,000 g at a temperature of 4° C., the supernatant was obtained and stored in another tube, proceeding immediately to the step of precipitation with 500 ⁇ L of cold isopropanol.
  • chloroform CHCl 3
  • RNA precipitate was left to dry for 10 minutes at room temperature, and then resuspended in sterile water containing diethyl pyrocarbonate (DEPC) and incubated at 55° C. for 10 minutes and subsequently put on ice for total solubilization of the RNA.
  • DEPC diethyl pyrocarbonate
  • the sample was checked as to its integrity by electrophoresis on a 1.2% denaturing agarose gel.
  • the samples having a suitable amount of RNA showed integrity on both ribosomal subunits: 18S e 28S.
  • the RNA samples were stored in a freezer at ⁇ 80° C.
  • RNA samples obtained were subjected to the complementary DNA (cDNA) synthesis using the Superscript III RTTM kit (Invitrogen, Life Technologies). After reading in a spectrophotometer (Gene Quant-Pharmacia, USA) and quantification, 3 ⁇ g of RNA were treated with the enzyme DNase I (Invitrogen, Life Technologies), for removal of contaminant DNA.
  • 1.0 ⁇ L of 1 u/ ⁇ L DNase I, 1.0 ⁇ L of 10 ⁇ DNase I Reaction Buffer (200 mM Tris-HCl, 20 mM MgCl2, 500 mM KCl2) and water sufficient for a final volume of 10.0 ⁇ L of reaction were added. The reaction was carried out for 15 minutes at room temperature and stopped with 1.0 ⁇ L of 25 mM EDTA, and incubated for 10 minutes at 65° C.
  • RNA synthesis 1.0 ⁇ L of 50 ⁇ M oligo (dT) 20 and 1.0 ⁇ L of 10 mM dNTP's were then added. The samples were incubated for 5 minutes at 65° C., followed by 1 minute at 4° C. To each sample, 10.0 ⁇ L of the following reaction mixture were added: 2 ⁇ L of 10 ⁇ RT buffer, 4.0 ⁇ L of 25 mM MgCl2, 2.0 ⁇ L of 0.1 M DTT, 1.0 ⁇ L of 40 U/ ⁇ L RNase OUTTM and 1.0 ⁇ L of 200 U/ ⁇ L, Superscript III RTTM. The reaction occurred for 50 minutes at 50° C., followed by 5 minutes at 85° C. Thereafter, 1.0 ⁇ l of 2 U/ ⁇ L E. coli RNase H was added for 20 minutes at 37° C.
  • BAC beta-actin
  • Reactions were carried out with: 5.0 ⁇ L of 10 ⁇ PCR buffer (20 mM Tris-HCl, 500 mM KCl), 1.5 ⁇ l of 50 mM MgCl2, 1.0 ⁇ L of 10 mM dNTP's, 1.0 ⁇ L of 10 mM of BACF primer (5′-AAGAGATGGCCACGGCTGCT-3′), 1.0 ⁇ L, of 10 mM of BACR primer (5′- TCGCTCCAACCGACTGCTGT-3′), 0.5 ⁇ L of Taq DNA polymerase, 1.0 ⁇ l of cDNA and 39 ⁇ L of water, for a final volume of 50 ⁇ L.
  • the program was started for 2 minutes at 94° C., followed by 35 cycles: 94° C./30 seconds-58° C./45 seconds-72° C./40 seconds, being terminated by 72° C./7 minutes.
  • the products were subjected to electrophoresis on 1% agarose gel to verify the amplification of 640 pb.
  • the primers used in Quantitative Real-Time PCR reactions were designed with the software “Primer Express” (Applied Biosystems), analyzed in the program Blast (www.ncbi.nlm.nih.gov/blast) to verify the conditions for formation of structures, such as hairpins and dimers.
  • the optimum concentration of primer to be used in quantitative real-time PCR should be sufficiently low to allow duplication of all copies of the gene present in the sample.
  • reactions containing each of the primers were performed at the final concentration of 150 nM, 300 nM, 600 nM e 900 nM.
  • the cycle in which fluorescence is detected above the established threshold is called threshold cycle or Tc. Since the same amount of sample was used in all reactions, the Tc should not change. If the increase on the primer concentration caused a reduction in Tc, so the amount of this reagent in the reaction was still insufficient. Thus, the optimum concentration chosen was the minimum, associated with the lower Tc.
  • the primer concentrations used in the amplification of the studied genes and the amplification efficiency obtained is shown in Table 9. The concentrations were defined by the amplification efficiency generated under the tested conditions.
  • amplification efficiency is obtained from formula 10 ( ⁇ 1/slope), wherein slope means the slope value of the curve. Optimization occurs using the optimum primer concentration with 7 known sample amounts, in logarithmic scale: 2 ng (2 ⁇ 100), 6.32 ng (2 ⁇ 100.5), 20 ng (2 ⁇ 101), 63.2 ng (2 ⁇ 101.5), 120 ng, 200 ng (2 ⁇ 102) e 632 ng (2 ⁇ 102.5). The results are used to construct a standard curve Tc versus sample amount.
  • cDNA aliquots were used as template in quantitative real-time PCR reactions.
  • the technique consists in optically monitoring the fluorescence emitted during the PCR reaction, by means of the binding of a is specific probe or dye to the newly synthesized strand.
  • the reactions always run in duplicate, were performed using the reagent SYBERGreen PCR Master Mix® (Applied Biosystems), which in addition to containing all reagents required for PCR (dNTP's, MgCL2, buffer, Taq Ampli-Gold), also contains the SYBERGreen dye, a double-stranded intercalating agent necessary for the detection of the reaction from cycle to cycle. Further, cDNA samples and specific primers for the analyzed gene were used.
  • the real-time amplification detection was performed in the apparatus ABI 5700 Sequence Detector System® (Applied Biosystems) in fluorescence versus cycle number graphs.
  • the reactions carried out contained 12.5 ⁇ L of the reagent SYBERGreen PCR Master Mix®, 25 ng of cDNA sample and the optimum determined primer concentration, making up a final volume of 25 ⁇ L. In all cases, negative controls were made containing sterile water in place of the sample. The reactions were prepared in 96-well plates (Sorenson, BioScience Inc.) with plastic caps which allow the passage of light. The program was started at 95° C./10 minutes, followed by 45 cycles: 95° C./15 seconds-60° C./1 minute. At the end of a normal amplification, a degradation step is added during which the temperature rises gradually from 60° C. to 95° C.
  • the florescence signal of SYBR Green decreases.
  • the resulting graph allows verifying if there is one or more PCR products present in each reaction, due to melting temperature differences between amplified PCR products, this difference being caused by the number and composition of bases of each product.
  • the Q values of the calibrator genes of to each sample were subjected to the Gnorm program, which computes the geometric mean between the same, which value is called sample Normalization Factor.
  • sample Normalization Factor The normalized expression of a given gene of interest in a certain sample is given by the ration between value Q of the gene of interest of the sample and the sample Normalization Factor.
  • the obtained data is expressed in arbitrary units or absolute expression value.
  • the antinociceptive profile was evaluated through the abdominal writhing assay induced by acetic acid in mice.
  • Swiss mice from both genders were used, weighing from 21 to 28 grams, fasted for a time period of about 8 hours.
  • the test substance was administered orally and had, as a carrier, 5% gum arabic.
  • acetic acid 0.1N 0.1 mL/10 g weight
  • the administration of acetic acid 0.1N was performed in the peritoneal cavity of the animals.
  • Ten minutes after the injection of acetic acid writhings were counted during 20 minutes. Controls were made for the carrier (gum arabic) and it does not show pharmacological activity.
  • This assay was performed using Swiss mice from both genders weighing from 18 to 30 grams. The animals were fasted for 8 hours with free access to water. This assay consists in locally administering (right ear) 20 ⁇ l of a capsaicin solution (250 ⁇ g/ear) diluted in acetone, 1 hour after the i.p. administration (diluted in 0.5% gum arabic). The left ear (control) received the carrier in which capsaicin was diluted (acetone) and the right ear received capsaicin. This assay is characterized by an acute inflammatory response of the ear, with development of edema. The animals were sacrificed and their ears were weighted to obtain the inflammation index. A biopsy (8 mm in diameter) of the ear was carried out.
  • the weights of the inflamed ears were compared against the weights of the contralateral ear (control ear) which were not treated with the phlogistic agent.
  • the edema inhibition percentage was calculated by subtracting the ear treated with the carrier by that treated with capsaicin from each group of animals treated with the test substances, and then it was divided by the difference between the groups of the irritating agents and the control groups. The result was subtracted by 1 and multiplied by 100, being shown in Table 4.
  • mice were treated with the substances being analyzed or carrier and after 1 h of oral administration, they were simultaneously subjected to the peritonitis assay, by administering intraperitoneally 1 ml of a 3% thioglycolate solution. 4 h after the administration of thioglycolate, the peritoneal cavity was washed with 3 ml of a HANKS solution (Balanced salt solution, free of Ca 2+ and Mg 2+ ). Next, the peritoneal wash was analyzed and the total counting of white blood cells was made in a Newbauer's chamber under an optical microscope with a 40 ⁇ objective. Results are shown in Table 5.
  • the determination of cell viability ( FIG. 5 ) to was performed by suspending peritoneal macrophages in a RPMI solution, set at a concentration of 5 ⁇ 10 6 cells/mL, 100 ⁇ L of which were added to each cavity of 96-cavity tissue culture plates, being incubated with drugs and prodrugs at concentrations of 10 ⁇ 4 , 10 ⁇ 5 , 10 ⁇ 6 , 10 ⁇ 7 , 10 ⁇ 8 mM during 24 hours at 37° C. and 5% of CO 2 .
  • the absorbance reading was performed in a UV/Visible spectrophotometer at 540 nm with reference filter at 620 nm (Microplate Reader-Model 550-BIORAD).
  • mice were previously inoculated with 3.0 mL of 3% thioglycolate intraperitoneally in order to stimulate the macrophages of this cavity. After 3 days of stimulation, the animals were sacrificed and the peritoneal macrophages collected. The cells were then washed from 2 to 3 times per centrifugation at 358 g (Centrifuge Fanem Excelsa II 206MP) during 5 minutes in sterile PBS and, then, resuspended in 1 mL of RPMI-1640 for counting in a Neubauer's chamber. After counting, the concentration was set at 5 ⁇ 10 6 cells/ml and these cells were distributed to disposable sterile plates with 96 cavities.
  • the plates thus containing the suitable concentration of cells were taken to incubation for 24 hours in a oven at 37° C., containing 95% of moisture and 5% of CO 2 in the presence of the anti-inflammatory agents and taurine derivatives at the respective concentrations: 10 ⁇ 8 , 10 ⁇ 7 , 10 ⁇ 6 , 10 ⁇ 5 and 10 ⁇ 4 mM or even in the presence of the RPMI-1640 medium only.
  • LPS was used as a positive control and the RPMI-1640 medium as a cell control.
  • the culture supernatants, upon the end of the incubation period, were collected in order to determine the nitric oxide levels thereof.
  • the RPMI-1640 used during this whole process was supplemented with 2 mM /-glutamine, penicillin (100 U/ml), streptomycin (100 ug/ml), 5% of bovine fetal serum and 2-mercaptoethanol 5 ⁇ 10 ⁇ 2 M(RPMI-1640-C).
  • the concentration of nitric oxide was evaluated. This evaluation was made by measuring the concentration of accumulated nitrite (a stable degradation product of nitric oxide) through a diazotization reaction with the Griess reagent (1% sulfanilamide, 0.1% naphthylenediamine dihydrochloride, in 5% of phosphoric acid), according to the method of Green et al. (1982).

Landscapes

  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Dermatology (AREA)
  • Hematology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US12/747,589 2007-12-12 2008-12-12 USE OF PHTHALIMIDE AND/OR SULPHONAMIDE DERIVATIVES IN THE TREATMENT OF DISEASES WHICH REQUIRE REDUCING THE TNF-alpha LEVELS AND AN EXOGENOUS SOURCE OF NITRIC OXIDE, PHTHALIMIDE DERIVATIVES, SULPHONAMIDE DERIVATIVES, AND A METHOD FOR OBTAINING A SULPHONAMIDE DERIVATIVE Abandoned US20100324107A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
BRPI0705396-7 2007-12-12
BRPI0705396-7A BRPI0705396B1 (pt) 2007-12-12 2007-12-12 Compostos derivados ftalimídicos
PCT/BR2008/000386 WO2009073940A2 (en) 2007-12-12 2008-12-12 USE OF PHTHALIMIDE AND/OR SULPHONAMIDE DERIVATIVES IN THE TREATMENT OF DISEASES WHICH REQUIRE REDUCING THE TNF-α LEVELS AND AN EXOGENOUS SOURCE OF NITRIC OXIDE, PHTHALIMIDE DERIVATIVES, SULPHONAMIDE DERIVATIVES, AND A METHOD FOR OBTAINING A SULPHONAMIDE DERIVATIVE

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/BR2008/000386 A-371-Of-International WO2009073940A2 (en) 2007-12-12 2008-12-12 USE OF PHTHALIMIDE AND/OR SULPHONAMIDE DERIVATIVES IN THE TREATMENT OF DISEASES WHICH REQUIRE REDUCING THE TNF-α LEVELS AND AN EXOGENOUS SOURCE OF NITRIC OXIDE, PHTHALIMIDE DERIVATIVES, SULPHONAMIDE DERIVATIVES, AND A METHOD FOR OBTAINING A SULPHONAMIDE DERIVATIVE

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/404,777 Division US8338474B2 (en) 2007-12-12 2012-02-24 Use of phthalimide and/or sulphonamide derivatives in the treatment of diseases which require reducing the TNF-α levels and an exogenous source of nitric oxide, phthalimide derivatives, sulphonamide derivatives, and a method for obtaining a sulphonamide derivative
US13/404,634 Division US8314255B2 (en) 2007-12-12 2012-02-24 Use of phthalimide and/or sulphonamide derivatives in the treatment of diseases which require reducing the TNF- α levels and an exogenous source of nitric oxide, phthalimide derivatives, sulphonamide derivatives, and a method for obtaining a sulphonamide derivative

Publications (1)

Publication Number Publication Date
US20100324107A1 true US20100324107A1 (en) 2010-12-23

Family

ID=40755919

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/747,589 Abandoned US20100324107A1 (en) 2007-12-12 2008-12-12 USE OF PHTHALIMIDE AND/OR SULPHONAMIDE DERIVATIVES IN THE TREATMENT OF DISEASES WHICH REQUIRE REDUCING THE TNF-alpha LEVELS AND AN EXOGENOUS SOURCE OF NITRIC OXIDE, PHTHALIMIDE DERIVATIVES, SULPHONAMIDE DERIVATIVES, AND A METHOD FOR OBTAINING A SULPHONAMIDE DERIVATIVE
US13/404,634 Expired - Fee Related US8314255B2 (en) 2007-12-12 2012-02-24 Use of phthalimide and/or sulphonamide derivatives in the treatment of diseases which require reducing the TNF- α levels and an exogenous source of nitric oxide, phthalimide derivatives, sulphonamide derivatives, and a method for obtaining a sulphonamide derivative
US13/404,777 Expired - Fee Related US8338474B2 (en) 2007-12-12 2012-02-24 Use of phthalimide and/or sulphonamide derivatives in the treatment of diseases which require reducing the TNF-α levels and an exogenous source of nitric oxide, phthalimide derivatives, sulphonamide derivatives, and a method for obtaining a sulphonamide derivative

Family Applications After (2)

Application Number Title Priority Date Filing Date
US13/404,634 Expired - Fee Related US8314255B2 (en) 2007-12-12 2012-02-24 Use of phthalimide and/or sulphonamide derivatives in the treatment of diseases which require reducing the TNF- α levels and an exogenous source of nitric oxide, phthalimide derivatives, sulphonamide derivatives, and a method for obtaining a sulphonamide derivative
US13/404,777 Expired - Fee Related US8338474B2 (en) 2007-12-12 2012-02-24 Use of phthalimide and/or sulphonamide derivatives in the treatment of diseases which require reducing the TNF-α levels and an exogenous source of nitric oxide, phthalimide derivatives, sulphonamide derivatives, and a method for obtaining a sulphonamide derivative

Country Status (8)

Country Link
US (3) US20100324107A1 (pt)
EP (1) EP2224922A4 (pt)
KR (1) KR20100114880A (pt)
CN (1) CN101951901B (pt)
BR (1) BRPI0705396B1 (pt)
CO (1) CO6290640A2 (pt)
WO (1) WO2009073940A2 (pt)
ZA (1) ZA201108106B (pt)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8282967B2 (en) 2005-05-27 2012-10-09 The University Of North Carolina At Chapel Hill Nitric oxide-releasing particles for nitric oxide therapeutics and biomedical applications
US8591876B2 (en) 2010-12-15 2013-11-26 Novan, Inc. Methods of decreasing sebum production in the skin
US8981139B2 (en) 2011-02-28 2015-03-17 The University Of North Carolina At Chapel Hill Tertiary S-nitrosothiol-modified nitric—oxide-releasing xerogels and methods of using the same
WO2015108835A1 (en) * 2014-01-14 2015-07-23 Sarmont Llc 3,3-disubstituted-1-hydroxytriaz-1-ene 2-oxides and wound-healing compositions using them
US9526738B2 (en) 2009-08-21 2016-12-27 Novan, Inc. Topical gels and methods of using the same
US9919072B2 (en) 2009-08-21 2018-03-20 Novan, Inc. Wound dressings, methods of using the same and methods of forming the same

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6137766A (ja) * 1984-07-31 1986-02-22 Sankyo Co Ltd フタルイミド誘導体およびその製造法
JPS6242967A (ja) * 1985-08-21 1987-02-24 Sankyo Co Ltd フタルイミド化合物およびその製造法
US5248785A (en) * 1990-02-12 1993-09-28 Virginia Commonwealth University Using allosteric hemoglobin modifiers to decrease oxygen affinity in blood
NZ336035A (en) * 1996-11-05 2002-03-28 Childrens Medical Center Angiogenesis inhibitory composition comprising an inhibitory compound and antiinflammatory drug
CA2381205C (en) * 1999-08-02 2010-05-11 Duke University Method for determining physiological effects of hemoglobin
BR0316256A (pt) * 2002-11-18 2005-10-04 Celgene Corp Métodos de inibir a produção de tnf-alfa e a atividade de pde4, de tratar ou prevenir uma doença ou um distúrbio, de controlar os nìveis de camp em uma célula e de produzir um composto, composição farmacêutica e composto
CA2518513C (en) * 2003-03-12 2014-05-20 Celgene Corporation N-alkyl-hydroxamic acid-isoindolyl compounds and their pharmaceutical uses
MXPA06006061A (es) * 2003-12-02 2006-08-11 Celgene Corp Metodos y composiciones para el tratamiento y manejo de la hemoglinopatia y la anemia.

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Gladwin et al., 2011, http://www.ncbi.nlm.nih.gov/pubmed/21364138 *
SickeCellAnemiaNIH2011, 2011, http://public.nhlbi.nih.gov/newroom/home/GetPressRelease.aspx?id=2759 *
SickleCellAnemiaNYTimes2012, 2012, http://health.nytimes.com/health/guides/disease/sickle-cell-anemia/treatment.html *
Ware, 2010, Blood, vol 115, no 26, 5300-5311 *

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9403851B2 (en) 2005-05-27 2016-08-02 The University Of North Carolina At Chapel Hill Nitric oxide-releasing particles for nitric oxide therapeutics and biomedical applications
US8282967B2 (en) 2005-05-27 2012-10-09 The University Of North Carolina At Chapel Hill Nitric oxide-releasing particles for nitric oxide therapeutics and biomedical applications
US8956658B2 (en) 2005-05-27 2015-02-17 The University Of North Carolina At Chapel Hill Nitric oxide-releasing particles for nitric oxide therapeutics and biomedical applications
US8962029B2 (en) 2005-05-27 2015-02-24 The University Of North Carolina At Chapel Hill Nitric oxide-releasing particles for nitric oxide therapeutics and biomedical applications
US11691995B2 (en) 2005-05-27 2023-07-04 The University Of North Carolina At Chapel Hill Nitric oxide-releasing particles for nitric oxide therapeutics and biomedical applications
US9403852B2 (en) 2005-05-27 2016-08-02 The University Of North Carolina At Chapel Hill Nitric oxide-releasing particles for nitric oxide therapeutics and biomedical applications
US9737561B2 (en) 2009-08-21 2017-08-22 Novan, Inc. Topical gels and methods of using the same
US9526738B2 (en) 2009-08-21 2016-12-27 Novan, Inc. Topical gels and methods of using the same
US9919072B2 (en) 2009-08-21 2018-03-20 Novan, Inc. Wound dressings, methods of using the same and methods of forming the same
US10376538B2 (en) 2009-08-21 2019-08-13 Novan, Inc. Topical gels and methods of using the same
US11583608B2 (en) 2009-08-21 2023-02-21 Novan, Inc. Wound dressings, methods of using the same and methods of forming the same
US8591876B2 (en) 2010-12-15 2013-11-26 Novan, Inc. Methods of decreasing sebum production in the skin
US9713652B2 (en) 2011-02-28 2017-07-25 The University Of North Carolina At Chapel Hill Nitric oxide-releasing S-nitrosothiol-modified silica particles and methods of making the same
US8981139B2 (en) 2011-02-28 2015-03-17 The University Of North Carolina At Chapel Hill Tertiary S-nitrosothiol-modified nitric—oxide-releasing xerogels and methods of using the same
WO2015108835A1 (en) * 2014-01-14 2015-07-23 Sarmont Llc 3,3-disubstituted-1-hydroxytriaz-1-ene 2-oxides and wound-healing compositions using them

Also Published As

Publication number Publication date
US8314255B2 (en) 2012-11-20
EP2224922A2 (en) 2010-09-08
CN101951901A (zh) 2011-01-19
ZA201108106B (en) 2013-07-31
BRPI0705396B1 (pt) 2021-10-05
WO2009073940A2 (en) 2009-06-18
BRPI0705396A2 (pt) 2009-12-22
KR20100114880A (ko) 2010-10-26
US20120157690A1 (en) 2012-06-21
US20120245364A1 (en) 2012-09-27
EP2224922A4 (en) 2010-12-22
WO2009073940A3 (en) 2009-11-12
CN101951901B (zh) 2012-11-28
US8338474B2 (en) 2012-12-25
CO6290640A2 (es) 2011-06-20

Similar Documents

Publication Publication Date Title
US8314255B2 (en) Use of phthalimide and/or sulphonamide derivatives in the treatment of diseases which require reducing the TNF- α levels and an exogenous source of nitric oxide, phthalimide derivatives, sulphonamide derivatives, and a method for obtaining a sulphonamide derivative
Cheng et al. Therapeutic targeting of autophagy in disease: biology and pharmacology
JP5598775B2 (ja) 癌の介入治療および根絶のためのiteとその類似体を含む医薬組成物
Ye et al. Hydroxysafflor yellow A protects neuron against hypoxia injury and suppresses inflammatory responses following focal ischemia reperfusion in rats
JP2012523239A (ja) 新規抗老化剤及びそれらを同定する方法
EP1888074B1 (en) Agent for the prevention and treatment of liver diseases containing pyrazolopyrimidine derivative
JP2019218379A (ja) 肝障害の治療方法
KR20190038507A (ko) 글루코코르티코이드계 화합물을 포함하는 비소세포성 폐암 치료용 약학 조성물
CA2479676A1 (en) Dexanabinol and dexanabinol analogs regulate inflammation related genes
WO2017155053A1 (ja) 非アルコール性脂肪性肝疾患/非アルコール性脂肪性肝炎の治療剤
US8383656B2 (en) Thiazolidinedione energy restriction-mimetic agents
US20050137251A1 (en) Dexanabinol and dexanabinol analogs regulate inflammation related genes
Lian et al. RJT-101, a novel camptothecin derivative, is highly effective in the treatment of melanoma through DNA damage by targeting topoisomerase 1
JP6672173B2 (ja) 進行した非アルコール性脂肪性肝炎の治療方法
US11058650B2 (en) Fatty acid amides for the prevention and/or treatment of steatohepatitis
Lai et al. Potent and orally active purine-based fetal hemoglobin inducers for treating β-thalassemia and sickle cell disease
CN116942834B (zh) 包含核仁应激诱导剂的抗肿瘤药物
KR20240007258A (ko) 리실 옥시다제의 억제제로서 비티아졸 유도체
JP2020132626A (ja) サフラナール製剤によるcdc25bの抑制および阻害
Wimmer et al. En Route to Targeted Ribosome Editing to Replenish Skin Anchor Protein LAMB3 in Junctional Epidermolysis Bullosa
Kang et al. DMB1285 [cyclopropyl-{4-[4-(4-fluorophenyl)-2-piperidin-4-yl-thiazol-5-yl] pyrimidin-2-yl} amine] suppresses tumor necrosis factor-α production by blocking p38 mitogen-activated protein kinase/mitogen-activated protein kinase-activated protein kinase 2 signaling pathway
JPWO2004093858A1 (ja) Mmp発現抑制剤
WO2023159048A2 (en) Small molecule stat3 inhibitor for treating triple negative breast cancer
EP4333843A1 (en) Bithiazol deratives as inhibitors of lysyl oxidases
JP5496232B2 (ja) Il−8発現抑制剤

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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