US20090227809A1 - New 2-(alpha-hydroxypentyl) benzoates, their preparations and their uses - Google Patents

New 2-(alpha-hydroxypentyl) benzoates, their preparations and their uses Download PDF

Info

Publication number
US20090227809A1
US20090227809A1 US12/468,929 US46892909A US2009227809A1 US 20090227809 A1 US20090227809 A1 US 20090227809A1 US 46892909 A US46892909 A US 46892909A US 2009227809 A1 US2009227809 A1 US 2009227809A1
Authority
US
United States
Prior art keywords
hydroxypentyl
solution
benzoate
general formula
ion
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
US12/468,929
Inventor
Jinghua Yang
Xiaoliang Wang
Zhibin Xu
Ying Peng
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.)
Institute of Materia Medica of CAMS
Original Assignee
Institute of Materia Medica of CAMS
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 Institute of Materia Medica of CAMS filed Critical Institute of Materia Medica of CAMS
Priority to US12/468,929 priority Critical patent/US20090227809A1/en
Publication of US20090227809A1 publication Critical patent/US20090227809A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F1/00Compounds containing elements of Groups 1 or 11 of the Periodic System
    • C07F1/06Potassium compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C65/00Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C65/01Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing hydroxy or O-metal groups
    • C07C65/03Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing hydroxy or O-metal groups monocyclic and having all hydroxy or O-metal groups bound to the ring
    • 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
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C65/00Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C65/01Compounds having carboxyl groups bound to carbon atoms of six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups containing hydroxy or O-metal groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F1/00Compounds containing elements of Groups 1 or 11 of the Periodic System
    • C07F1/04Sodium compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F3/00Compounds containing elements of Groups 2 or 12 of the Periodic System
    • C07F3/04Calcium compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F3/00Compounds containing elements of Groups 2 or 12 of the Periodic System
    • C07F3/06Zinc compounds

Definitions

  • This invention relates to new chemically synthetic 2-( ⁇ -hydroxypentyl) benzoates, their preparations and pharmaceutical compositions containing the salts as active ingredients.
  • This invention also relates to the uses of the compounds in the prevention and treatment of the diseases such as cardioischemia, cerebroischemia, heart and brain arterial occlusions, etc.
  • Acute ischemic cerebral apoplexy is a common disease with high incidence (12-18 cases per ten thousands) and high death rate (6-12 cases per ten thousands) which seriously endangers human's health, which often leaves sequela in the survivals and imposes a heavy burden on the patients' family and society. Therefore, it is very valuable of developing drug for the prevention and treatment of the disease.
  • Many researchers have been studying the mechanism of the disease (acute ischemic cerebral apoplexy) since the 1980's, and have proposed theories such as energy metabolism, excitatory poison, oxidative injury, calcium overload and many other theories for the purpose of developing high effective and low toxic drugs. However, an ideal therapeutic drug is still under developing.
  • Drugs such as calcium antagonists, excitatory receptor antagonists, free radical scavengers are being clinically used, but the effects are uncertain.
  • Thrombolytic drugs such as t-PA are being used to treat acute ischemic cerebral apoplexy (within 6 hrs from incidence), and is effective, but the hazard of hemorrhage has not been solved. Thus it is still a focus of developing new drugs to treat ischemic cerebral apoplexy.
  • Coronary heart disease is also a severe disease which harms human's health. Due to the coronary atherosclerosis and the formation of thrombus, ischemic cardiac muscle trauma is induced. For this reason, it has been being a leading work of developing new drugs to prevent and treat coronary atherosclerosis, prevent thrombus formation and dilatate coronary artery.
  • the object of the present invention is to provide new 2-( ⁇ -hydroxypentyl) benzoates which can significantly inhibit platelet aggregation and ameliorate cerebral microcirculation and effective to cardioischemia, cerebroischemia, heart and brain arterial occlusions.
  • Another object of the present invention is to provide a synthetic method of 2-( ⁇ -hydroxypentyl) benzoates.
  • Yet another object of the present invention is to provide a pharmaceutical composition which can prevent and treat cardioischemia, cerebroischemia, heart and brain arterial occlusions.
  • the fourth object of the present invention is to provide use of the above compounds and pharmaceutical compositions in the prevention and treatment of cardioischemia, cerebroischemia, heart and brain arterial occlusions, and amelioration of cerebral microcirculation.
  • the present invention provides a compound of the following general formula (I):
  • M is a monovalent metal ion, such as K + , Na + , Li + ; a divalent metal ion, such as Ca 2+ , Mg 2+ , Zn 2+ ; or an organic basic group, such as anilino, benzyl amino, morpholinyl or diethylamino.
  • the preparation method of present invention is as following:
  • the solvent for the hydrolysis and ring-opening reaction may be any one of methanol, ethanol, acetone, isopropanol, water or mixture of H 2 O-alcohol (or ketone).
  • the crystallization solvent may be any one of methanol, ethanol, propanol, isopropanol, acetone, acetyl acetate, chloroform, ether, dichloromethane, benzene, toluene, petroleum ether, or a mixture of two or three above solvents with different proportions; monovalent base may be an inorganic base, such as a chemically pure inorganic base, for example sodium hydroxide, potassium hydroxide, or lithium hydroxide, etc; or a chemically pure organic base, such as sodium (or potassium) methoxide, sodium (or potassium) ethoxide, etc.
  • inorganic base such as a chemically pure inorganic base, for example sodium hydroxide, potassium hydroxide, or lithium hydroxide, etc
  • a chemically pure organic base such as sodium (or potassium) methoxide, sodium (or potassium) ethoxide, etc.
  • M is a monovalent metal ion (e.g., sodium or potassium)
  • the reaction solvent medium is methanol, ethanol, acetone, isopropanol, water or a mixture of water-alcohol (or ketone); divalent metal salt may be magnesium chloride, calcium chloride, or zinc chloride; the crystallization solvent may be methanol, ethanol, isopropanol, acetyl acetate, chloroform, ether, dichloromethane, or a mixture of two or three above solvents with different proportions.
  • M a monovalent metal ion (e.g., sodium or potassium)
  • step (2) a solution which contains an equivalent or slightly excess amount of divalent base or a divalent metal salt to the 2-( ⁇ -hydroxypentyl) benzoic acid, for instance an alcohol solution of calcium hydroxide, is added, under the temperature of ⁇ 10 ⁇ 0° C., to afford the 2-( ⁇ -hydroxypentyl) benzoate of present invention wherein M is a divalent metal ion, such as calcium.
  • the compound of present invention i.e., 2-( ⁇ -hydroxylpentyl) benzoate wherein M is divalent metal ion is prepared.
  • step (2) To the solution obtained in step (2), a solution which contains an equivalent or slightly excess amount of organic base to the 2-( ⁇ -hydroxypentyl) benzoic acid, for instance chemically pure aniline, is added, under the temperature of ⁇ 10 ⁇ 0° C., to afford the 2-( ⁇ -hydroxypentyl) benzoate of present invention wherein M is an organic base, such as aniline.
  • the compound of present invention i.e., 2-( ⁇ -hydroxylpentyl) benzoate wherein M is divalent metal ion is prepared.
  • the acid used in the acidify reaction may be any one of concentrated or diluted hydrochloric acid or sulfuric acid; the temperature should be controlled within the range of ⁇ 20 ⁇ +20° C.; the organic solvent to extract 2-( ⁇ -hydroxypentyl) benzoic acid may be any one of ether, ethyl acetate, chloroform, dichloromethane, benzene, toluene, petroleum ether, n-hexane, or cyclohexane; the monovalent base is selected from potassium hydroxide, sodium hydroxide, lithium hydroxide, sodium (or potassium) methoxide, or sodium (or potassium) ethoxide; the divalent inorganic metal salt or divalent inorganic base is selected from MgCl 2 , MgCO 3 , CaCl 2 , CaCO 3 , ZnCl 2 , ZnCO 3 , MgSO 4 , Zn(OH) 2 , Mg(OH) 2 or Ca(OH) 2 ; the organic
  • the present compounds show good effects on prevention and therapeutics to cardioischemia and cerebroischemia, further, the present compounds also have pharmacological effects of anti-platelet aggregation, arterial occlusion of heart and brain therapeutic effects and cerebral microcirculation amelioration effect, etc.
  • the present compounds have shown from animal tests excellent effects on protecting ischemic injury of heart, anti-platelet aggregation and alleviation of injury due to cerebral-arterial occlusion, moreover, no side effects such as exciting or hemorrhage have not been found.
  • the pharmaceutical composition of the prevent invention comprises an treatment effective amount of the compound of the present invention as active ingredient and a pharmaceutically acceptable carrier.
  • the compounds and pharmaceutical compositions of the prevent invention can be used for the preparation of drugs which can prevent and treat cardioischemia and cerebroischemia, arterial occlusion of heart and brain, amelioration of cardiac-cerebral microcirculations.
  • pharmaceutically acceptable carrier means the ordinary drug carriers such as diluents, excipients, fillers such as starch, saccharide; binders such as cellulose derivatives, alginates, gelatin, and polyvinylpyrrolidone; moisturizing agents such as glycerol; disintegrating agents such as agar, calcium carbonate and sodium bicarbonate; resorption accelerators such as quaternary ammonium compounds; surface active agents such as cetyl alcohol; adsorptive carriers such as kaolin and bentonite; lubricants such as talc, calcium or magnesium stearate, and polyethyleneglycols; further, other assistants such as flavors and sweeteners can also be added.
  • drug carriers such as diluents, excipients, fillers such as starch, saccharide; binders such as cellulose derivatives, alginates, gelatin, and polyvinylpyrrolidone; moisturizing agents such as glycerol; disintegrating agents such as agar
  • the compounds of this invention can be administered orally and intravenously to the patients in need of such treatment in a form of pharmaceutical formulation.
  • a form of pharmaceutical formulation When administrated orally, it can be administrated in the dosage form such as tablets, particles, or capsules. And it can be in the form of solutions or oily/aqueous suspensions for injection.
  • Preferred dosage forms are tablet, capsule and injection.
  • compositions can be prepared by common procedures in the art.
  • the compounds of this invention can be admixed with one or more carriers and formed into a desired dosage form.
  • the present pharmaceutical composition preferably contains active ingredients in a weight ratio of 3:2, most preferably 1:1.
  • the dose of present compound may vary based on administration route, age, weight, disease type and severeness of disease of the patient being treated.
  • a typical daily dose may be from 50 mg to 600 mg per day, preferably 100 ⁇ 200 mg per day, which can be administered once or more times.
  • the potassium dl-2-( ⁇ -hydroxypentyl) benzoate as prepared in the above Examples 1 till 4 procedures is a white granular crystal.
  • the sodium dl-2-( ⁇ -hydroxypentyl) benzoate as prepared in the above Examples 5 till 8 procedures is a foamed white solid.
  • the ether extract was combined and dried with anhydrous Na 2 SO 4 at a low temperature and then filtered.
  • Tablets Ingredient Amount (mg/tablet) Active ingredient 50 ⁇ 200 Medicinal starch 20 ⁇ 50 Microcrystalline cellulose 15 ⁇ 35 Magnesium stearate 0.5 Talc 0.5 ⁇ 1 Sodium carboxymethyl cellulose 2 ⁇ 5
  • the active ingredient, starch, microcrystalline cellulose and sodium carboxymethyl cellulose were crushed and mixed.
  • the mixture was moisturized homogenously and powdered, which were then sieved and dried and then sieved again.
  • Magnesium stearate and talc were mixed with the above mixture and the mixture was compressed to tablets, and the tablets were coated with a film coating (which may be hydroxypropylmethyl cellulose and the like). Each tablet contains 50 ⁇ 200 mg of active ingredient.
  • Capsules Ingredient Amount (mg/capsule) Active ingredient 50 ⁇ 200 Medicinal starch or mannitose 20 ⁇ 50 Methyl cellulose 3 ⁇ 4 Cross-linked PVP 0.5 ⁇ 1
  • the active ingredient and excipients were mixed and sieved.
  • the so-obtained mixture was filled into stomach-soluble hard capsules with determine amounts.
  • Each capsule contains 50 ⁇ 200 mg of active ingredient.
  • Intravenous solution Ingredient Amount Active ingredient 50 ⁇ 100 mg/bottle Sodium hydroxide Appropriate Water for injection or saline 5 ⁇ 50 ml
  • the active ingredient was dissolved in an appropriate amount of water for injection or isotonic saline and then filtered. Adjusting pH to 10.5 (which may vary from 9.0 to 10.5) with appropriate amount of NaOH. The intravenous solution was filled into bottles under sterilizing condition.
  • the active ingredient was dissolved in an appropriate amount of water for injection and the pH was adjusted to 9.0 ⁇ 10.0 with NaOH. The solution was filtered and freeze-dried to afford a cake or powder. The lyophilized injection can be injected and transfused intravenously after it is solved in the 0.9% NaCl solution for injection or 5% glucose injection.
  • dl-PHPB was dissolved in double distilled water.
  • TTC 2,3,5-triphenyltetrazolium chloride
  • Middle cerebral artery occlusion the rats were anesthetized by trichloroacetaldehyde monohydrate (350 mg/kg, ip). The left CCA was exposed through a middle neck incision and was carefully dissected free from surrounding nerves. The ICA and ECA were isolated. Then a 4 cm long 0.26 mm diameter nylon suture was inserted through proximal ECA into ICA for a length of 2.0 mm from the bifurcation. The wounds were sutured and the rats were released. The room temperature was kept 24-25° C. during the test.
  • MCAO Middle cerebral artery occlusion
  • Animals were divided into two groups, 1) administration group: dl-PHPB (200 mg/kg) and HCl (pH 1.6) 0.5 ml (to mimic the acidic condition of the human stomach) were administrated per os (p.o) 30 minutes prior to ischemia; 2) Control group: double-distilled water and HCl (pH 1.6) 0.5 ml were administrated per os (p.o) 30 minutes prior to ischemia.
  • the anaesthetized animals were decapitated 24 hours after MCAO. Each brain were rapidly removed and kept in ice-cold saline (0-4° C.). After 10 minutes, the coronal section was sliced into five pieces after the removal of bulbus olfactus, cerebellum, and low brain stem. Upon cutting, the first cut was at the center point of the connection line of the polus anterior of cerebrum and the optic chiasm, the second cut was at the site of optic chiasm, the third was at the site of infundibulum stalk, and the fourth was between the infundibulum stalk and the caudal pole of posterior lobe.
  • the sliced brain was kept in a 5 ml solution of TTC (4%) and K 2 HPO 4 (IM), shaded and incubated at 37° C. for 30 minutes. During the incubation, slices were turned over every 7-8 minutes. After staining, the normal cerebral tissue showed rosy color, but the infarcted tissue showed white color. The infarcted tissue was separated from the normal tissue and weighed. The infarction area is calculated from the weight percentage of the infarct tissue to the total cerebral tissue.
  • Table 1 shows the infarct area, the area of the dl-PHBP treated group is 19.83 ⁇ 3.53%, and that of the control is 26.99 ⁇ 3.51%.
  • the infarction is significantly reduced (P ⁇ 0.01) in the treated group compared to the control.
  • Platelet Aggregometer type: PAT-4A MEGURO-KU, TOKYO, JAPAN.
  • Control group double-distilled water (400 mg/kg) was administrated per os (p.o) 30 minutes prior blood sampling;
  • Table 2 shows the inhibitory effect on platelet aggregation induced by ADP.
  • Potassium 2-(1-hydroxypentyl)-benzoate, dl-PHPB) significantly inhibits platelet aggregation compared to the control group.
  • Table 3 shows the protective effects of dl-PHPB on cardiac arrhythmia induced by ischemia-reperfusion in isolated hearts on rats
  • Dl-PHPB can significantly shorten the duration of arrhythmia and VT induced by ischemia-reperfusion in isolated hearts on rats, decreases the accidence of spasmic ventricular tachycardia, and shows significant protections on the ischemic injuries of hearts at the concentration of 10 ⁇ 4 mol/L.
  • novel 2-( ⁇ -hydroxypentyl) benzoates can be used for the preparation of pharmaceutical compositions, which is used for the prevention and treatment of the diseases such as cardiac ischemia, cerebral ischemia, arterial occlusion (obstruction) of heart and brain, etc.

Abstract

The invention relates to new synthetic 2-(α-hydroxypentyl) benzoates, their preparation and pharmaceutical compositions containing such salts as active ingredients. The invention also relates to the use of the compounds for preventing and treating cardioischemia, cerebroischemia and cardiac or cerebral arterial occlusion (obstruction), etc.

Description

    TECHNICAL FIELD
  • This invention relates to new chemically synthetic 2-(α-hydroxypentyl) benzoates, their preparations and pharmaceutical compositions containing the salts as active ingredients. This invention also relates to the uses of the compounds in the prevention and treatment of the diseases such as cardioischemia, cerebroischemia, heart and brain arterial occlusions, etc.
  • BACKGROUND OF THE INVENTION
  • Acute ischemic cerebral apoplexy is a common disease with high incidence (12-18 cases per ten thousands) and high death rate (6-12 cases per ten thousands) which seriously endangers human's health, which often leaves sequela in the survivals and imposes a heavy burden on the patients' family and society. Therefore, it is very valuable of developing drug for the prevention and treatment of the disease. Many researchers have been studying the mechanism of the disease (acute ischemic cerebral apoplexy) since the 1980's, and have proposed theories such as energy metabolism, excitatory poison, oxidative injury, calcium overload and many other theories for the purpose of developing high effective and low toxic drugs. However, an ideal therapeutic drug is still under developing. Drugs such as calcium antagonists, excitatory receptor antagonists, free radical scavengers are being clinically used, but the effects are uncertain. Thrombolytic drugs such as t-PA are being used to treat acute ischemic cerebral apoplexy (within 6 hrs from incidence), and is effective, but the hazard of hemorrhage has not been solved. Thus it is still a focus of developing new drugs to treat ischemic cerebral apoplexy.
  • Coronary heart disease is also a severe disease which harms human's health. Due to the coronary atherosclerosis and the formation of thrombus, ischemic cardiac muscle trauma is induced. For this reason, it has been being a leading work of developing new drugs to prevent and treat coronary atherosclerosis, prevent thrombus formation and dilatate coronary artery.
  • SUMMARY OF THE INVENTION
  • The object of the present invention is to provide new 2-(α-hydroxypentyl) benzoates which can significantly inhibit platelet aggregation and ameliorate cerebral microcirculation and effective to cardioischemia, cerebroischemia, heart and brain arterial occlusions.
  • Another object of the present invention is to provide a synthetic method of 2-(α-hydroxypentyl) benzoates.
  • Yet another object of the present invention is to provide a pharmaceutical composition which can prevent and treat cardioischemia, cerebroischemia, heart and brain arterial occlusions.
  • The fourth object of the present invention is to provide use of the above compounds and pharmaceutical compositions in the prevention and treatment of cardioischemia, cerebroischemia, heart and brain arterial occlusions, and amelioration of cerebral microcirculation.
  • The present invention provides a compound of the following general formula (I):
  • Figure US20090227809A1-20090910-C00001
  • wherein n=1,2; M is a monovalent metal ion, such as K+, Na+, Li+; a divalent metal ion, such as Ca2+, Mg2+, Zn2+; or an organic basic group, such as anilino, benzyl amino, morpholinyl or diethylamino.
  • The preparation method of present invention is as following:
  • 1. Preparation of 2-(α-hydroxypentyl) benzoates of general formula (I) wherein M is a monovalent metal ion:
  • Solving an equivalent racemic 3-n-butyl-isobenzofuran-1-(3H)-one in a hydrolysis and ring-opening reaction solvent medium, adding an equivalent or slightly excess amount of monovalent base. After that hydrolysis and ring-opening reaction is conducted under the temperature of 10-100° C., for 0.5-6 hours, to afford the 2-(α-hydroxypentyl) benzoates of general formula (I) wherein M is a monovalent metal ion.
  • The solvent for the hydrolysis and ring-opening reaction may be any one of methanol, ethanol, acetone, isopropanol, water or mixture of H2O-alcohol (or ketone).
  • The crystallization solvent may be any one of methanol, ethanol, propanol, isopropanol, acetone, acetyl acetate, chloroform, ether, dichloromethane, benzene, toluene, petroleum ether, or a mixture of two or three above solvents with different proportions; monovalent base may be an inorganic base, such as a chemically pure inorganic base, for example sodium hydroxide, potassium hydroxide, or lithium hydroxide, etc; or a chemically pure organic base, such as sodium (or potassium) methoxide, sodium (or potassium) ethoxide, etc.
  • 2. Preparation of 2-(α-hydroxypentyl) benzoates of general formula (I) wherein M is a divalent metal ion:
  • 2-(α-Hydroxypentyl) benzoate of general formula (I) wherein M is a monovalent metal ion (e.g., sodium or potassium) is solved in a solvent, then an equivalent or slightly excess amount of chemically pure divalent metal salt is added and an ion-exchange reaction is conducted under the temperature of 10-100° C. for 0.5-10 hours to afford the 2-(α-hydroxypentyl) benzoates of general formula (I) wherein M is a divalent metal ion.
  • The reaction solvent medium is methanol, ethanol, acetone, isopropanol, water or a mixture of water-alcohol (or ketone); divalent metal salt may be magnesium chloride, calcium chloride, or zinc chloride; the crystallization solvent may be methanol, ethanol, isopropanol, acetyl acetate, chloroform, ether, dichloromethane, or a mixture of two or three above solvents with different proportions.
  • 3. Preparation of the 2-(α-hydroxypentyl) benzoates of general formula (I) wherein M is an organic base:
  • (1) 2-(α-hydroxypentyl) benzoate of general formula (I) wherein M is a monovalent metal ion (e.g., sodium or potassium) is solved in a solvent, then a inorganic acid is added to the solution to adjust pH value to 6.0-2.0 at the temperature of −20-20° C., preferably −20-0° C., to acidify the 2-(α-hydroxy pentyl) benzoate and 2-(α-hydroxypentyl) benzoic acid is obtained.
  • (2) After the reaction is completed, an organic extraction solvent is added to the solution and the free acid 2-(α-hydroxypentyl) benzoic acid is extracted with common extraction method at a temperature of −20-0° C. An organic solution containing 2-(α-hydroxypentyl) benzoic acid is obtained and the solution is kept for use under the temperature of −20-10° C., preferably −20˜0° C.
  • (3) To the solution obtained in the above step, a solution which contains an equivalent or slightly excess amount of monovalent base to the 2-(α-hydroxypentyl) benzoic acid, for example an alcohol solution of potassium hydroxide, is added, at the temperature of −10˜0° C., to afford the 2-(α-hydroxylpentyl) benzoate of present invention wherein M is a monovalent metal ion, such as potassium. After the reaction is sufficiently conducted, with a same purification method with that of preparation method 1, the compound of present invention, i.e., 2-(α-hydroxylpentyl) benzoate wherein M is a monovalent metal ion is prepared.
  • (4) To the solution obtained in step (2), a solution which contains an equivalent or slightly excess amount of divalent base or a divalent metal salt to the 2-(α-hydroxypentyl) benzoic acid, for instance an alcohol solution of calcium hydroxide, is added, under the temperature of −10˜0° C., to afford the 2-(α-hydroxypentyl) benzoate of present invention wherein M is a divalent metal ion, such as calcium. After the reaction is sufficiently conducted, with a same purification method with that of preparation method 2, the compound of present invention, i.e., 2-(α-hydroxylpentyl) benzoate wherein M is divalent metal ion is prepared.
  • (5) To the solution obtained in step (2), a solution which contains an equivalent or slightly excess amount of organic base to the 2-(α-hydroxypentyl) benzoic acid, for instance chemically pure aniline, is added, under the temperature of −10˜0° C., to afford the 2-(α-hydroxypentyl) benzoate of present invention wherein M is an organic base, such as aniline. After the reaction is sufficiently conducted, with a same purification method with that of preparation method 2, the compound of present invention, i.e., 2-(α-hydroxylpentyl) benzoate wherein M is divalent metal ion is prepared.
  • The acid used in the acidify reaction may be any one of concentrated or diluted hydrochloric acid or sulfuric acid; the temperature should be controlled within the range of −20˜+20° C.; the organic solvent to extract 2-(α-hydroxypentyl) benzoic acid may be any one of ether, ethyl acetate, chloroform, dichloromethane, benzene, toluene, petroleum ether, n-hexane, or cyclohexane; the monovalent base is selected from potassium hydroxide, sodium hydroxide, lithium hydroxide, sodium (or potassium) methoxide, or sodium (or potassium) ethoxide; the divalent inorganic metal salt or divalent inorganic base is selected from MgCl2, MgCO3, CaCl2, CaCO3, ZnCl2, ZnCO3, MgSO4, Zn(OH)2, Mg(OH)2 or Ca(OH)2; the organic base is selected from aniline, benzyl amine, morpholine or diethylamine; the solvent to solve the 2-(α-hydroxylpentyl) benzoate wherein M is a monovalent metal salt may be any one of H2O, MeOH—H2O, EtOH—H2O, acetone-H2O, isopropanol-H2O.
  • It has been found that the present compounds show good effects on prevention and therapeutics to cardioischemia and cerebroischemia, further, the present compounds also have pharmacological effects of anti-platelet aggregation, arterial occlusion of heart and brain therapeutic effects and cerebral microcirculation amelioration effect, etc.
  • The present compounds have shown from animal tests excellent effects on protecting ischemic injury of heart, anti-platelet aggregation and alleviation of injury due to cerebral-arterial occlusion, moreover, no side effects such as exciting or hemorrhage have not been found.
  • The pharmaceutical composition of the prevent invention comprises an treatment effective amount of the compound of the present invention as active ingredient and a pharmaceutically acceptable carrier.
  • The compounds and pharmaceutical compositions of the prevent invention can be used for the preparation of drugs which can prevent and treat cardioischemia and cerebroischemia, arterial occlusion of heart and brain, amelioration of cardiac-cerebral microcirculations.
  • The above mentioned “pharmaceutically acceptable carrier” means the ordinary drug carriers such as diluents, excipients, fillers such as starch, saccharide; binders such as cellulose derivatives, alginates, gelatin, and polyvinylpyrrolidone; moisturizing agents such as glycerol; disintegrating agents such as agar, calcium carbonate and sodium bicarbonate; resorption accelerators such as quaternary ammonium compounds; surface active agents such as cetyl alcohol; adsorptive carriers such as kaolin and bentonite; lubricants such as talc, calcium or magnesium stearate, and polyethyleneglycols; further, other assistants such as flavors and sweeteners can also be added.
  • The compounds of this invention can be administered orally and intravenously to the patients in need of such treatment in a form of pharmaceutical formulation. When administrated orally, it can be administrated in the dosage form such as tablets, particles, or capsules. And it can be in the form of solutions or oily/aqueous suspensions for injection. Preferred dosage forms are tablet, capsule and injection.
  • The dosage forms of present pharmaceutical compositions can be prepared by common procedures in the art. For example, the compounds of this invention can be admixed with one or more carriers and formed into a desired dosage form.
  • The present pharmaceutical composition preferably contains active ingredients in a weight ratio of 3:2, most preferably 1:1.
  • The dose of present compound may vary based on administration route, age, weight, disease type and severeness of disease of the patient being treated. A typical daily dose may be from 50 mg to 600 mg per day, preferably 100˜200 mg per day, which can be administered once or more times.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The following examples are only illustrative and are not intended to limit the scope of the present invention.
  • Example 1 Preparation of Racemic Potassium 2-(α-hydroxypentyl) benzoate (Herein after Also Referred as Potassium dl-2-(α-hydroxypentyl) benzoate, or dl-PHPB)
  • Dissolving dl-3-n-Butyl-isobenzofuran-1-(3H)-one (8.5 g, 0.045 mol) in 20 ml methanol and a 10 ml methanol solution of KOH (2.6 g, 0.046 mol) was added. The reaction solution was stirred under reflux for one hour. After that, TLC analysis (petroleum ether-acetone=10:1) and I2 vapor coloration showed that the starting material was disappeared. The reaction solution was concentrated under reduced pressure to afford a sticky yellow residue, which was allowed to crystallize in the refrigerator after addition of 20 ml of chloroform. The crude product was recrystallized in MeOH—CHCl3, and a white granular crystal (10.07 g, yield=91.50%) was obtained.
  • Example 2 Preparation of Potassium dl-2-(α-hydroxypentyl) benzoate
  • Dissolving dl-3-n-Butyl-isobenzofuran-1-(3H)-one (0.63 g, 3.3 mmol) in 10 ml methanol and a 10 ml methanol solution of KOH (0.19 g, 3.4 mmol) was added. The reaction solution was stirred under reflux for one hour. After that, TLC analysis (petroleum ether-acetone=10:1) and I2 vapor coloration showed that the starting material was disappeared. The reaction solution was concentrated under reduced pressure to afford a sticky yellow residue, which was allowed to crystallize in the refrigerator after addition of 5 ml of chloroform. The crude product was recrystallized in MeOH—CHCl3, and a white granular crystal (0.6 g, yield=73.56%) was obtained.
  • Example 3 Preparation of Potassium dl-2-(α-hydroxypentyl) benzoate
  • Dissolving sodium dl-2-(α-hydroxypentyl) benzoate (1.96 g, 8.5 mmol) in 10 ml of H2O and the solution was cooled to about 0° C. in an ice-salt bath. The pH was adjusted to 2.0˜3.0 with 1N HCl and the solution was extracted quickly with cold ether (3×20 ml). The ether extract was combined and dried with anhydrous Na2SO4 at a low temperature for 3 hours and then filtered quickly under the low temperature. To the filtrate a 20 ml methanol solution of anhydrous K2CO3 (0.58 g, 4.2 mmol) was added and the mixture was stirred fastly to ambient temperature. A white solid appeared in the ether solution and the solution was kept for over 24 hours, then the white solid was filtered and dried (1.4 g, yield=66.67%).
  • Example 4 Preparation of Potassium dl-2-(α-hydroxypentyl) benzoate
  • Dissolving sodium dl-2-(α-hydroxypentyl) benzoate (1.78 g, 7.7 mmol) in about 10 ml of H2O and the solution was cooled to about 0° C. in an ice-salt bath. The pH was adjusted to 2.0˜3.0 with 1N HCl and the solution was extracted quickly with cold ether (3×20 ml). The ether layer was combined and dried with anhydrous Na2SO4 at a low temperature for 2 hours and then filtered quickly under the low temperature. To the filtrate a 10 ml methanol solution of KOH (0.43 g, 7.7 mmol) was dropped in and the mixture was kept in the ice-salt bath to ambient temperature. After concentration under reduced pressure, the residue was recrystallized with MeOH-ether. A white solid was obtained (1.3 g, yield=68.42%).
  • The potassium dl-2-(α-hydroxypentyl) benzoate as prepared in the above Examples 1 till 4 procedures is a white granular crystal.
  • mp. 151-152° C.
  • IR (KBr)
  • 3198 cm−1 OH), 2933 cm−1 CH3), 1577, 1561 (νCOO)
  • 1H-NMR (300 MHz, DMSO) δ (ppm)
  • 7.65 (dd, J=6.3 Hz, 2.7 Hz, 1H), 7.17-7.05 (m, 3H), 4.32 (t, 1H), 3.40 (s, 1H), 1.80-1.55 (m, 2H), 1.38-1.04 (m, 4H), 0.81 (t, 3H)
  • Elemental Analysis C12H15O3K (FW246.35)
  • C(%) H(%) K(%)
    Calculated 58.51 6.14 15.87
    Found 58.24 6.01 15.84
    Figure US20090227809A1-20090910-C00002
  • Example 5 Preparation of Sodium dl-2-(α-hydroxypentyl) benzoate
  • Dissolving dl-3-n-Butyl-isobenzofuran-1-(3H)-one (4.6 g, 0.024 mol) in 20 ml methanol and a 10 ml aqueous solution of NaOH (0.86 g, 0.022 mol) was added. The reaction solution was stirred under reflux for 2 hours. After that, TLC analysis (petroleum ether-acetone=10:1) and I2 vapor coloration showed that the starting material was disappeared. The reaction solution was concentrated under reduced pressure to afford a sticky yellow residue, which did not crystallize in the solvents such as chloroform, acetone, ether or methanol, etc. A small amount of residue was solidified with ether-ethyl acetate (10:1) to afford a white solid, which was very hydroscopic and became sticky over filtration. The yellow residue was washed with ether for several times and dehydrated with anhydrous benzene, dried under reduced pressure to afford a white foamed solid (3.67 g, yield=65.91%).
  • Example 6 Preparation of Sodium dl-2-(α-hydroxypentyl) benzoate
  • Dissolving dl-3-n-Butyl-isobenzofuran-1-(3H)-one (7.6 g, 0.04 mol) in 20 ml methanol and a fresh methanol solution (20 ml) of sodium methoxide (0.92 g, 0.04 mol of Na) was added. The reaction solution was stirred under reflux for 2 hours. After that, TLC analysis (petroleum ether-acetone=10:1) and I2 vapor coloration showed that the starting material was disappeared. The reaction solution was concentrated under reduced pressure to afford a sticky yellow residue, which did not crystallize in the solvents such as chloroform, acetone, ether or methanol, etc. The yellow residue was washed with ether for several times and dehydrated with anhydrous benzene, dried under reduced pressure to afford a white foamed solid (6.4 g, yield=69.56%).
  • Example 7 Preparation of Sodium dl-2-(α-hydroxypentyl) benzoate
  • Dissolving dl-PHPB (1.0 g, 0.004 mol) in 10 ml of H2O and the solution was cooled to about 0° C. in an ice-salt bath. The pH was adjusted to 2.0˜3.0 with 1N HCl and the solution was extracted quickly with cold ether (3×20 ml). The ether extract was combined and dried with anhydrous Na2SO4 at a low temperature for 2 hours and then filtered quickly under the low temperature. To the filtrate a 20 ml methanol solution of anhydrous Na2CO3 (0.58 g, 4.2 mmol) was added and the solution became turbid and return clear after 3 hours of standing with ice-salt bath. The reaction solution was concentrated under reduced pressure to afford a sticky yellow residue, which was washed with ether for several times and dehydrated with anhydrous benzene, dried under reduced pressure to afford a white foamed solid (0.4 g, yield=42.78%).
  • Example 8 Preparation of dl-Sodium-2-(α-hydroxypentyl) benzoate
  • Dissolving dl-PHPB (2.4 g, 0.01 mol) in 20 ml of H2O and the solution was cooled to about 0° C. in an ice-salt bath. The pH was adjusted to 2.0˜3.0 with 1N HCl and the solution was extracted quickly with cold ether (3×20 ml). The ether extract was combined then a 20 ml methanol solution of NaOH (0.39 g, 0.01 mol), the resulted solution was kept over night in the ice-salt bath. After concentration under reduced pressure, a sticky yellow residue was afforded, which was processed as above, and a white foamed solid (1.2 g, yield==53.48%) was afforded.
  • The sodium dl-2-(α-hydroxypentyl) benzoate as prepared in the above Examples 5 till 8 procedures is a foamed white solid.
  • IR (film) 3398 cm−1 OH), 2969 cm−1 CH3), 1558, 1394 cm−1 COO)
  • 1H-NMR (300 MHz, DMSO) δ (ppm) 7.70 (dd, J=6.9 Hz, 1.8 Hz, 1H), 7.17-7.07 (m, 3H), 4.39 (t, 1H), 1.65-1.48 (m, 2H), 1.39-1.10 (m, 4H), 0.83 (t, 3H)
  • Example 9 Preparation of Lithium dl-2-(α-hydroxypentyl) benzoate
  • Dissolving dl-3-n-Butyl-isobenzofuran-1-(3H)-one (1.2 g, 0.006 mol) in 15 ml methanol and LiOH.H2O (0.26 g, 0.006 mol) was added. The reaction solution was stirred under reflux for 2 hours. After that, TLC analysis (petroleum ether-acetone=10:1) and I2 vapor coloration showed that the starting material was disappeared. The reaction solution was concentrated under reduced pressure to afford a white crystal (1.1 g, yield=81.38%).
  • mp. 134-136° C.
  • IR (KBr)
  • 3323 cm−1 OH), 2931 cm−1 CH3), 1604, 1414 cm−1 COO)
  • 1H-NMR (300 MHz, DMSO) δ (ppm)
  • 7.66 (dd, 1H), 7.17-7.16 (m, 3H), 4.32 (t, 1H), 1.78-1.54 (m, 2H), 1.27-1.02 (m, 4H), 0.84 (t, 3H)
  • Elemental Analysis C12H15O3Li (FW214.19)
  • C(%) H(%) Li(%)
    Calculated 67.29 7.06 3.24
    Found 67.34 6.87 3.26
  • Example 10 Preparation of Calcium dl-2-(α-hydroxypentyl) benzoate
  • Dissolving dl-3-n-Butyl-isobenzofuran-1-(3H)-one (1.4 g, 7.1 mmol) in 15 ml methanol and a 20 ml aqueous solution of NaOH (0.28 g, 7.0 mmol) was added. The reaction solution was stirred under reflux for 2 hours. After that, TLC analysis (petroleum ether-acetone=10:1) and I2 vapor coloration showed that the starting material was disappeared. Calcium chloride (0.4 g, 4.5 mmol) was solved in 40 ml of H2O, and the solution was dropped into the above reaction solution. The reaction was carried out in a 60° C. water bath for 2 hours, and the pH was adjusted to about 7 with 1N HCl and then reaction solution was filtered. The filtrate was concentrated to 10 ml under reduced pressure and a white solid appeared. The solution with the white solid appeared was kept for 30 minutes and then filtered. The filter cake was washed several times with water and H2O-MeOH (1:1), respectively, until no chloride ion can be detected. Heating the filter cake to dry to afford a white solid (1.08 g, yield=33.23%).
  • Example 11 Preparation of Calcium dl-2-(α-hydroxypentyl) benzoate
  • Dissolving dl-3-n-Butyl-isobenzofuran-1-(3H)-one (1.1 g, 5.8 mmol) in 20 ml anhydrous ethanol and a 10 ml aqueous solution of NaOH (0.39 g, 9.8 mmol) was added. The reaction solution was stirred under reflux for 2 hours. After that, TLC analysis (petroleum ether-acetone=10:1) and I2 vapor coloration showed that the starting material was disappeared. The reaction solution was cooled to about 0° C. in an ice-salt bath, and acidified to pH 5.0˜6.0 with 1N HCl, additional 20 ml ethanol was added to solve the precipitated white solid. Calcium carbonate powder (0.33 g, 3.3 mmol) was added and the reaction solution was stirred vigorously under low temperature and kept over night. A white solid appeared from the reaction solution and the reaction solution was filtered. The filter cake was washed several times with H2O-MeOH. Heating the filter cake to dry to afford a white solid (0.65 g, yield=24.73%).
  • Example 12 Preparation of Calcium dl-2-(α-hydroxypentyl) benzoate
  • Calcium chloride (0.12 g, 0.002 mmol) was dissolved in 20 ml H2O, and heated to about 60° C. To the solution a 10 ml aqueous solution of dl-PHPB (0.5 g, 0.001 mol) was added. After a while, a white solid appeared and the reaction was continued for another 3 hours. After that, the reaction solution was filtered and the filter cake was washed with hot water and dried under heating to obtain a white solid (0.21 g) and the filtrate was concentrated under reduced pressure and washed with hot water and heated to dry, additional 0.15 g of white solid was obtained. The combined total white solid was 0.36 g (yield=78.02%).
  • Calcium dl-2-(α-hydroxypentyl) benzoate as prepared in the above Examples 9 till 12 procedures is a white solid, which decomposes above a temperature of 252° C.
  • IR (KBr)
  • 3323 cm−1 OH), 2931 cm−1 CH3), 1604, 1401 cm−1 COO)
  • 1H-NMR (300 MHz, DMSO) δ (ppm)
  • 7.56 (d, 1H), 7.24-7.05 (m, 3H), 4.55 (t, 1H), 1.71-1.52 (m, 2H), 1.26-1.04 (m, 4H), 0.80 (t, 3H)
  • Elemental Analysis C24H30O6Ca (FW454.57)
  • C(%) H(%) Ca(%)
    Calculated 63.41 6.65 8.82
    Found 63.20 6.61 9.02
  • Example 13 Preparation of dl-2-(α-hydroxypentyl) benzoic Acid Benzyl Amine Salt
  • Dissolving dl-3-n-Butyl-isobenzofuran-1-(3H)-one (1.4 g, 7.1 mmol) in 15 ml methanol and a 20 ml aqueous solution of NaOH (0.28 g, 7.0 mmol) was added. The reaction solution was stirred continuously under reflux for 2 hours. After that, TLC analysis (petroleum ether-acetone=10:1) and I2 vapor coloration showed that the starting material was disappeared. The reaction solution was cooled to about 0° C. in an ice-salt bath and acidified with 1N HCl to pH 3.0-4.0, and the solution was extracted quickly with cold ether (3×20 ml). The ether extract was combined and dried with anhydrous Na2SO4 at a low temperature and then filtered. Benzyl amine (0.9 ml, 8.24 mmol) was added to the reaction solution and the reaction solution was kept over night in the ice-salt bath. After concentration under reduced pressure, a yellow sticky residue was obtained and a white solid (1.07 g) separated under the addition of petroleum ether-ether, which was recrystallized in ethyl acetate to afford a white crystal (0.61 g, yield=26.28%).
  • mp. 86-88° C.
  • IR (KBr) 3396 cm−1 NH), 2927 cm−1 (br, νOH), 1637 cm−1 C═C) 1515 cm−1 COO)
  • 1HNMR (300 MHz, DMSO) δ (ppm)
  • 7.65-7.13 (m, 9H), 4.67 (t, 1H, CH), 3.95 (d, 2H, CH2), 1.58-1.66 (m, 2H, CH2), 1.14-1.34 (m, 4H, CH2CH2), 0.82 (t, 3H, CH3)
  • Elemental Analysis C19H25NO3(FW315.41)
  • C(%) H(%) N(%)
    Calculated 72.35 7.99 4.44
    Found 72.32 7.98 4.76
    Figure US20090227809A1-20090910-C00003
  • Example 14 Preparation of Pharmaceutical Compositions
  • Tablets
    Ingredient Amount (mg/tablet)
    Active ingredient  50~200
    Medicinal starch 20~50
    Microcrystalline cellulose 15~35
    Magnesium stearate 0.5
    Talc 0.5~1  
    Sodium carboxymethyl cellulose 2~5
  • The active ingredient, starch, microcrystalline cellulose and sodium carboxymethyl cellulose were crushed and mixed. The mixture was moisturized homogenously and powdered, which were then sieved and dried and then sieved again. Magnesium stearate and talc were mixed with the above mixture and the mixture was compressed to tablets, and the tablets were coated with a film coating (which may be hydroxypropylmethyl cellulose and the like). Each tablet contains 50˜200 mg of active ingredient.
  • Example 15 Preparation of Pharmaceutical Compositions
  • Capsules
    Ingredient Amount (mg/capsule)
    Active ingredient  50~200
    Medicinal starch or mannitose 20~50
    Methyl cellulose 3~4
    Cross-linked PVP 0.5~1  
  • The active ingredient and excipients were mixed and sieved. The so-obtained mixture was filled into stomach-soluble hard capsules with determine amounts. Each capsule contains 50˜200 mg of active ingredient.
  • Example 16 Preparation of Pharmaceutical Compositions
  • Intravenous solution
    Ingredient Amount
    Active ingredient 50~100 mg/bottle
    Sodium hydroxide Appropriate
    Water for injection or saline 5~50 ml
  • The active ingredient was dissolved in an appropriate amount of water for injection or isotonic saline and then filtered. Adjusting pH to 10.5 (which may vary from 9.0 to 10.5) with appropriate amount of NaOH. The intravenous solution was filled into bottles under sterilizing condition.
  • Example 17 Preparation of Pharmaceutical Compositions
  • Lyophilized intravenous injection
    Ingredient Amount
    Active ingredient 50~100 mg/bottle
    Sodium hydroxide Appropriate
  • The active ingredient was dissolved in an appropriate amount of water for injection and the pH was adjusted to 9.0˜10.0 with NaOH. The solution was filtered and freeze-dried to afford a cake or powder. The lyophilized injection can be injected and transfused intravenously after it is solved in the 0.9% NaCl solution for injection or 5% glucose injection.
  • Test Example 1 Effect of the Present Compounds on the Infarction Area after Local Cerebral Ischemia in Rats
  • (1) Materials and Methods
  • Animal: male Wistar rats (250-280 g), from the Animal Center of the Chinese Academy of Medical Sciences.
  • Drugs:
  • dl-PHPB was dissolved in double distilled water.
  • 2,3,5-triphenyltetrazolium chloride (TTC) was purchased from Beijing Chemical Plant.
  • Methods: Middle cerebral artery occlusion (MCAO): the rats were anesthetized by trichloroacetaldehyde monohydrate (350 mg/kg, ip). The left CCA was exposed through a middle neck incision and was carefully dissected free from surrounding nerves. The ICA and ECA were isolated. Then a 4 cm long 0.26 mm diameter nylon suture was inserted through proximal ECA into ICA for a length of 2.0 mm from the bifurcation. The wounds were sutured and the rats were released. The room temperature was kept 24-25° C. during the test.
  • Groups:
  • Animals were divided into two groups, 1) administration group: dl-PHPB (200 mg/kg) and HCl (pH 1.6) 0.5 ml (to mimic the acidic condition of the human stomach) were administrated per os (p.o) 30 minutes prior to ischemia; 2) Control group: double-distilled water and HCl (pH 1.6) 0.5 ml were administrated per os (p.o) 30 minutes prior to ischemia.
  • Infarction Measurement:
  • The anaesthetized animals were decapitated 24 hours after MCAO. Each brain were rapidly removed and kept in ice-cold saline (0-4° C.). After 10 minutes, the coronal section was sliced into five pieces after the removal of bulbus olfactus, cerebellum, and low brain stem. Upon cutting, the first cut was at the center point of the connection line of the polus anterior of cerebrum and the optic chiasm, the second cut was at the site of optic chiasm, the third was at the site of infundibulum stalk, and the fourth was between the infundibulum stalk and the caudal pole of posterior lobe. The sliced brain was kept in a 5 ml solution of TTC (4%) and K2HPO4 (IM), shaded and incubated at 37° C. for 30 minutes. During the incubation, slices were turned over every 7-8 minutes. After staining, the normal cerebral tissue showed rosy color, but the infarcted tissue showed white color. The infarcted tissue was separated from the normal tissue and weighed. The infarction area is calculated from the weight percentage of the infarct tissue to the total cerebral tissue.
  • (2) Result:
  • Effect of dl-PHBP to the cerebral infarct areas in the rats of permanent MCAO.
  • Table 1 shows the infarct area, the area of the dl-PHBP treated group is 19.83±3.53%, and that of the control is 26.99±3.51%. The infarction is significantly reduced (P<0.01) in the treated group compared to the control.
  • TABLE 1
    Control group (%) Treated group(%)
    30.50 26.01
    27.08 20.24
    27.92 18.24
    30.64 22.74
    24.87 16.84
    27.44 15.93
    20.46 18.79
    X ± SD 26.99 ± 3.51 19.83 ± 3.53**
    **P < 0.01 vs control group
  • (3) Conclusion:
  • Potassium 2-(α-hydroxypentyl)-benzoate, dl-PHPB) significantly reduced the cerebral tissue injury induced by MCAO and decreased the infarction areas.
  • Test Example 2 The Effect of the Present Compounds on Platelet Aggregation
  • (1) Materials and Methods
  • Animal: male Wistar rats (260-280 g), from the Animal Center of the Chinese Academy of Medical Sciences.
  • Drugs:
  • Potassium 2-(1-hydroxypentyl)-benzoate, dl-PHPB),
  • ADP, obtained from Shanghai Institute of Biochemistry, Acedemia Sinica.
  • Equipment: Platelet Aggregometer (type: PAT-4A MEGURO-KU, TOKYO, JAPAN.
  • Groups: Animals were divided into four groups:
  • 1) Control group: double-distilled water (400 mg/kg) was administrated per os (p.o) 30 minutes prior blood sampling;
  • 2) Dl-PHPB (400 mg/kg) was administrated per os (p.o) 30 minutes prior to the blood sampling;
  • 3) Dl-PHPB (400 mg/kg) was administrated per os (p.o) 60 minutes prior to the blood sampling;
  • 4) Dl-PHPB (200 mg/kg) was administrated per os (p.o) 30 minutes prior to the blood sampling;
  • Method:
  • After oral administration of dl-PHPB, blood was drawn from the carotid of rats at the time points of 30 and 60 minutes, respectively. Platelet rich plasma (PRP) and platelet poor plasma (PPP) were prepared upon common procedure. According to the method described by of Born, PRP (200 μl) was put on the Platelet Aggregometer and pre-incubated at 37° C. for 5 minutes and then ADP with the final concentration of 5 μmol/L was added to induce platelet aggregation. The maximum aggregation was measured 5 minutes after the addition of ADP.
  • (2) Results
  • Table 2 shows the inhibitory effect on platelet aggregation induced by ADP.
  • TABLE 2
    Rate of platelet Rate of
    Groups N aggregation (%) inhibition (%)
    Control 7 0.5556 ± 0.0287
    dl-PHPB (400 mg/kg 30 min) 7 0.4483 ± 0.0610** 19.31
    dl-PHPB (400 mg/kg 60 min) 9 0.4989 ± 0.0562* 10.21
    dl-PHPB (200 mg/kg 30 min) 8 0.5021 ± 0.0622 6.62
    **P < 0.01,
    *P < 0.05 vs Control group
  • (3) Conclusion:
  • Potassium 2-(1-hydroxypentyl)-benzoate, dl-PHPB) significantly inhibits platelet aggregation compared to the control group.
  • Test Example 3 The Protective Effects of dl-PHPB on Cardiac Arrhythmia Induced by Ischemia-Reperfusion in Isolated Hearts on Rats
  • (1) Materials and Methods
  • Animals: male Wistar rats, 250-300 g, from the Animal Center of the Chinese Academy of Medical Sciences, randomly grouped.
  • Drugs:
  • dl-PHPB,
  • NaCl, from the Beijing Chemical Reagent Factory No. 2,
  • KCl and MgSO4, Beijing Shuanghuan Chemical Reagents Factory,
  • KH2PO4, Beijing Yili Fine Chemicals Co., Ltd.
  • NaHCO3, Beijing Chemical Reagents Company
  • Glucose, Beijing Guohua Chemical Reagents Factory
  • CaCl2, Sigma.
  • Apparatus:
  • Langendoff perfusion system;
  • XD-7100 ECG, Shanghai Medical Electronic Devices Factory
  • Methods:
  • (1) remove the rat heart quickly after decapitation and put it into K—H solution at 4° C., fix the aorta to the perfusion system;
  • (2) perfuse the heart with K—H solution, 6-8 ml/min, 37±0.5° C., under the pressure of about 60 mm H2O;
  • (3) connect the two copper electrodes to the cardiac apex and the bottom of the right atria and record the cardiogram;
  • (4) perporate under the left anterior descending branch of the coronary artery with 3/0 line;
  • (5) perfuse the heart for 10 minutes and record the normal cardiogram;
  • (6) ligate the left anterior descending branch of the coronary artery to make myocardial ischemia for 15 minutes;
  • (7) nip the line and resume the perfusion, record the change of cardiogram for 30 minutes (mainly on VF and Spasmic VT);
  • (8) dissolve the dl-PHPB at pH 1.5 to a desired concentration, add it into K—H solution to do the experiments.
  • (2) Results:
  • Table 3 shows the protective effects of dl-PHPB on cardiac arrhythmia induced by ischemia-reperfusion in isolated hearts on rats
  • TABLE 3
    Spasmic
    Duration of VT VT VF VE
    Arrhythmia Incidence Duration Incidence Incidence Incidence
    Group N (min) (%) (min) (%) (%) (%)
    Control 12 22.5 ± 9.5 100 8.2 ± 7.9   66.7 33.3 83.3
    dl-PHPB 6 23.3 ± 5.8 100 5.0 ± 6.5 50 33.3 66.7
    (10−5 mol/L)
    dl-PHPB 7  1.7 ± 0.6*** 100 1.1 ± 0.6**  0* 0 42.9
    (10−4 mol/L)
    VT: ventricular tachycardia
    VF: ventricular fibrillation
    VE: ventricular ectopic beats
    *P < 0.05,
    **P < 0.01,
    ***P < 0.001 compared with the control group
  • (3) Conclusion:
  • Dl-PHPB can significantly shorten the duration of arrhythmia and VT induced by ischemia-reperfusion in isolated hearts on rats, decreases the accidence of spasmic ventricular tachycardia, and shows significant protections on the ischemic injuries of hearts at the concentration of 10−4 mol/L.
  • INDUSTRIAL AVAILABILITY
  • The novel 2-(α-hydroxypentyl) benzoates can be used for the preparation of pharmaceutical compositions, which is used for the prevention and treatment of the diseases such as cardiac ischemia, cerebral ischemia, arterial occlusion (obstruction) of heart and brain, etc.

Claims (10)

1. A compound of the following general formula (I):
Figure US20090227809A1-20090910-C00004
wherein M is a monovalent metal ion, a divalent metal ion, or an organic base group and n is 1 or 2.
2. The compound according to claim 1, wherein M is a potassium ion, a sodium ion, or a lithium ion.
3. The compound according to claim 1, wherein M is a calcium ion, a magnesium ion, or a zinc ion.
4. The compound according to claim 1, wherein M is an anilino group, a benzyl amino group, a morpholinyl group or a diethylamino group.
5. A preparation method of the compound of general formula (I) according to claim 1 wherein M is a monovalent metal ion, comprising:
solving an equivalent racemic 3-n-butyl-isobenzofuran-1-(3H)-one in a hydrolysis and ring-opening reaction solvent medium, adding an equivalent or slightly excess amount of monovalent base, conducting hydrolysis and ring-opening reaction under the temperature of 10˜100° C., to afford the 2-(α-hydroxypentyl) benzoate of general formula (I) wherein M is a monovalent metal ion.
6. The preparation method according to claim 5, wherein the solvent for the hydrolysis and ring-opening reaction is any one of methanol, ethanol, acetone, isopropanol, water or mixture of water-alcohol (ketone),
the monovalent base is an inorganic base, such as sodium hydroxide, potassium hydroxide, or lithium hydroxide; or a chemically pure organic base, such as sodium (potassium) methoxide, sodium (potassium) ethoxide.
7. A preparation method of the compound of general formula (I) according to claim 1 wherein M is a divalent metal ion, comprising:
solving 2-(α-Hydroxypentyl) benzoate of general formula (I) wherein M is a monovalent metal ion (sodium or potassium) in a solvent, adding an equivalent or slightly excess amount of chemically pure divalent metal salt and conducting an ion-exchange reaction under the temperature of 10-100° C., to afford the 2-(α-hydroxylpentyl) benzoate of general formula (I) wherein M is a divalent metal ion.
8. The preparation method according to claim 7, wherein the reaction solvent medium is any one of methanol, ethanol, acetone, isopropanol, water or a mixture of water-alcohol (ketone); and the divalent metal salt is any one of magnesium chloride, calcium chloride, or zinc chloride.
9. A preparation method of the compound of general formula (I) according to claim 1, comprising:
(1) solving 2-(α-hydroxypentyl) benzoate of general formula (I) wherein M is a monovalent metal ion in a solvent medium, adding an inorganic acid to the solution and adjusting the pH value to 6.0˜2.0, allowing the 2-(α-hydroxypentyl) benzoate to react with the acid to afford 2-(α-hydroxypentyl) benzoic acid, at a temperature of −20˜20° C., preferably −20˜0° C.;
(2) adding an organic extraction solvent to the so-obtained benzoic acid solution and extracting free 2-(α-hydroxypentyl) benzoic acid to obtain an organic solution containing 2-(α-hydroxypentyl) benzoic acid, keeping the solution for use under the temperature of −20˜10° C., preferably −20˜0° C.;
(3) adding a solution which contains an equivalent or slightly excess amount of monovalent base to the 2-(α-hydroxypentyl) benzoic acid solution obtained in the above step under the temperature of −10˜0° C., to afford a 2-(α-hydroxylpentyl) benzoate of general formula (I) wherein M is a monovalent metal ion;
(4) adding a solution which contains an equivalent or slightly excess amount of divalent base or a divalent metal salt solution to the 2-(α-hydroxypentyl) benzoic acid solution obtained in step (2) under the temperature of −10˜0° C., to afford a 2-(α-hydroxypentyl) benzoate of general formula (I) wherein M is a divalent metal ion;
(5) adding a solution which contains an equivalent or slightly excess amount of organic base to the 2-(α-hydroxypentyl) benzoic acid solution obtained in step (2) under the temperature of −10˜0° C., to afford a 2-(α-hydroxypentyl) benzoate of general formula (I) wherein M is an organic base.
10. The preparation method according to claim 9, wherein the acid used in the acidification reaction is any one of concentrated or diluted hydrochloric acid or sulfuric acid; the acidification reaction temperature is controlled within the range of −20˜+20° C.; the organic solvent to extract 2-(α-hydroxypentyl) benzoic acid is any one of ether, ethyl acetate, chloroform, dichloromethane, benzene, toluene, petroleum ether, n-hexane, or cyclohexane; the monovalent base is any one of sodium hydroxide, potassium hydroxide, lithium hydroxide, sodium (or potassium) methoxide, or sodium (or potassium) ethoxide; the divalent inorganic metal salt or divalent inorganic base is any one of magnesium chloride, magnesium carbonate, calcium chloride, calcium carbonate, zinc chloride, zinc carbonate, magnesium sulfate, magnesium hydroxide, or calcium hydroxide; the organic base is any one of aniline, benzyl amine, morpholine or diethylamine; the solvent is any one of methanol, ethanol, acetone, isopropanol, water or water-alcohol (ketone) or any one of ether, acetyl acetate, chloroform, dichloromethane, benzene, toluene, petroleum ether, n-hexane, or cyclohexane.
US12/468,929 2002-05-09 2009-05-20 New 2-(alpha-hydroxypentyl) benzoates, their preparations and their uses Abandoned US20090227809A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/468,929 US20090227809A1 (en) 2002-05-09 2009-05-20 New 2-(alpha-hydroxypentyl) benzoates, their preparations and their uses

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
PCT/CN2002/000320 WO2003095412A1 (en) 2002-05-09 2002-05-09 2-(α-HYDROXYPENTYL) BENZOATE AND ITS PREPARATION AND USE
US10/512,792 US7550507B2 (en) 2002-05-09 2002-05-09 2-(alpha-hydroxypentyl) benzoate and its preparation and use
US12/468,929 US20090227809A1 (en) 2002-05-09 2009-05-20 New 2-(alpha-hydroxypentyl) benzoates, their preparations and their uses

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/CN2002/000320 Division WO2003095412A1 (en) 2002-05-09 2002-05-09 2-(α-HYDROXYPENTYL) BENZOATE AND ITS PREPARATION AND USE
US10/512,792 Division US7550507B2 (en) 2002-05-09 2002-05-09 2-(alpha-hydroxypentyl) benzoate and its preparation and use

Publications (1)

Publication Number Publication Date
US20090227809A1 true US20090227809A1 (en) 2009-09-10

Family

ID=33569588

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/512,792 Expired - Lifetime US7550507B2 (en) 2002-05-09 2002-05-09 2-(alpha-hydroxypentyl) benzoate and its preparation and use
US12/468,929 Abandoned US20090227809A1 (en) 2002-05-09 2009-05-20 New 2-(alpha-hydroxypentyl) benzoates, their preparations and their uses

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/512,792 Expired - Lifetime US7550507B2 (en) 2002-05-09 2002-05-09 2-(alpha-hydroxypentyl) benzoate and its preparation and use

Country Status (11)

Country Link
US (2) US7550507B2 (en)
EP (2) EP2186792B1 (en)
JP (1) JP5058439B2 (en)
KR (1) KR100756470B1 (en)
CN (1) CN1243541C (en)
AU (1) AU2002252949A1 (en)
BR (1) BR0215777B1 (en)
CA (1) CA2485479C (en)
DE (1) DE60233825D1 (en)
IL (1) IL164845A (en)
WO (1) WO2003095412A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104414981A (en) * 2013-08-28 2015-03-18 云南昊邦制药有限公司 Freeze-dried hydroxyl-pentyl benzoate preparation for injection and preparation method of freeze-dried hydroxyl-pentyl benzoate preparation for injection

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6904402B1 (en) * 1999-11-05 2005-06-07 Microsoft Corporation System and iterative method for lexicon, segmentation and language model joint optimization
CN1243541C (en) 2002-05-09 2006-03-01 中国医学科学院药物研究所 2-(alpha-hydroxypentyl) benzoate and its preparing process and usage
CN100422133C (en) * 2003-09-01 2008-10-01 北京天衡药物研究院 Novel 2-(alpha-hydroxyl amyl) and its preparing method and use
CN100422134C (en) 2004-03-12 2008-10-01 北京天衡药物研究院 2-(alpha-n-pentanone) phenmethyl and its preparation process and application
US20070203233A1 (en) * 2004-06-04 2007-08-30 Team Academy Of Pharmaceutical Science NOVEL 2-(alpha-N-PENTANONYL)BENZOATES, THEIR PREPARATION AND USE
CN100540523C (en) * 2004-06-17 2009-09-16 北京天衡药物研究院 L-2-(Alpha-hydroxy amyl group) benzoate and method for making and purposes
JP2011527290A (en) * 2008-07-08 2011-10-27 インスティトゥート オブ マタリア メディカ,チャイニーズ アカデミー オブ メディカル サイエンシズ Application to role in prevention and / or treatment of Alzheimer's disease using potassium 2- (α-hydroxy-pentyl) benzoate
CN102397272B (en) * 2010-09-17 2013-08-14 石药集团恩必普药业有限公司 Application of butylphthalide and derivatives thereof in preparation of medicines for preventing and treating ALS
EA022988B9 (en) * 2010-10-27 2016-08-31 Прометик Биосайнсиз Инк. Phenylketone carboxylate compounds and pharmaceutical uses thereof
CN109394720A (en) * 2012-11-21 2019-03-01 石药集团中奇制药技术(石家庄)有限公司 A kind of composition and preparation method thereof of 3-N-butylphthalide derivative
CN103113210A (en) * 2013-02-19 2013-05-22 石药集团中奇制药技术(石家庄)有限公司 Hydroxyl amyl benzoate potassium crystal and preparation method thereof
CN103127025B (en) * 2013-03-06 2016-03-09 石家庄鸯星科技有限公司 The preparation method of racemization 2-(Alpha-hydroxy amyl group) benzoate sheet
CN105343014B (en) * 2013-03-25 2018-08-24 贵州贵安新区协生元医药科技有限公司 Injection racemization 2- (Alpha-hydroxy amyl) benzoate freeze-dried powder and preparation method thereof
CN104086399B (en) 2013-07-17 2016-08-24 浙江奥翔药业股份有限公司 5-bromo-2-(Alpha-hydroxy amyl group) different crystal forms and preparation method thereof of benzoic acid sodium salt
CN104546827B (en) * 2013-10-09 2019-12-27 石药集团恩必普药业有限公司 Application of butylphthalide or derivatives thereof in preparing medicine for treating or preventing diabetes
CN104546828B (en) * 2013-10-09 2017-11-14 石药集团恩必普药业有限公司 Application of the butylphthalide or derivatives thereof in the medicine for treating or preventing diabetic complication is prepared
CN103896763B (en) * 2014-03-27 2015-11-18 云南昊邦制药有限公司 A kind of 2-(Alpha-hydroxy amyl group) potassium benzoate polymorphic and preparation method thereof, preparation and application
CN103989651A (en) * 2014-03-27 2014-08-20 云南昊邦制药有限公司 2-(alpha-hydroxy pentyl) benzoate controlled-release tablet and preparation method thereof
CN104510721A (en) * 2014-12-25 2015-04-15 云南昊邦制药有限公司 2-(alpha-hydroxypentyl) benzoate dropping pill and preparation method thereof
CN104510720A (en) * 2014-12-25 2015-04-15 云南昊邦制药有限公司 2-(alpha-hydroxypentyl) benzoate dispersible tablet and preparation method thereof
CN106146297A (en) * 2015-04-03 2016-11-23 米文君 A kind of new compound and application thereof
CN105130934B (en) * 2015-08-15 2018-11-23 石药集团恩必普药业有限公司 A kind of butylphenyl phthaleine bulk pharmaceutical chemicals product and preparation method thereof
CN105859670B (en) * 2016-04-19 2019-04-05 丽珠医药集团股份有限公司 A kind of preparation method of high purity butylene phthalide
WO2018126897A1 (en) * 2017-01-06 2018-07-12 郑州大学 APPLICATION OF SODIUM 5-BROMO-2-(α-HYDROXYPENTYL) BENZOATE IN DRUGS TREATING CARDIOVASCULAR DISEASE
CN109776466B (en) * 2018-03-19 2023-01-31 河南真实生物科技有限公司 Benzoic acid compound and preparation method and application thereof
CN108715579B (en) 2018-05-17 2020-03-24 四川大学 Organic amine ester derivative medicine of 2- (α hydroxypentyl) benzoic acid
CN109223749A (en) * 2018-11-14 2019-01-18 郑州大学 The bromo- 2-(α-Hydroxy pentyl of 5-) medicinal usage of the benzoic acid sodium salt in treatment myocardial hypertrophy and heart failure
CN109678715B (en) * 2018-11-28 2019-11-12 中国药科大学 Salt, the preparation method and the usage that 2- (1- acyl-oxygen n-pentyl) benzoic acid and basic amino acid or aminoguanidine are formed
CN111943898A (en) * 2019-05-17 2020-11-17 浙江大学 Benzotetrazole derivatives, preparation method thereof, pharmaceutical composition containing same and application thereof
KR20220156597A (en) 2020-03-20 2022-11-25 씨에스피씨 엔비피 파머슈티컬 캄파니 리미티드 Uses of Butylphthalide and Its Derivatives
CN112457265A (en) * 2020-11-06 2021-03-09 浙江大学 Tetrazole derivative, preparation method thereof, pharmaceutical composition containing tetrazole derivative and application of pharmaceutical composition

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1283621A (en) * 1999-07-05 2001-02-14 中国医学科学院药物研究所 Process for preparing optically active 3-n-butyl phenylphthaleine

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1243541C (en) 2002-05-09 2006-03-01 中国医学科学院药物研究所 2-(alpha-hydroxypentyl) benzoate and its preparing process and usage

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1283621A (en) * 1999-07-05 2001-02-14 中国医学科学院药物研究所 Process for preparing optically active 3-n-butyl phenylphthaleine

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Lower, Introduction to Acid-Base Chemistry,1999, Simon Fraser University, p.1-19. *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104414981A (en) * 2013-08-28 2015-03-18 云南昊邦制药有限公司 Freeze-dried hydroxyl-pentyl benzoate preparation for injection and preparation method of freeze-dried hydroxyl-pentyl benzoate preparation for injection

Also Published As

Publication number Publication date
BR0215777A (en) 2005-03-01
EP1508561B1 (en) 2009-09-23
KR100756470B1 (en) 2007-09-07
CN1382682A (en) 2002-12-04
IL164845A0 (en) 2005-12-18
JP5058439B2 (en) 2012-10-24
US7550507B2 (en) 2009-06-23
WO2003095412A1 (en) 2003-11-20
EP1508561A4 (en) 2006-07-19
CA2485479C (en) 2010-06-22
BR0215777B1 (en) 2017-10-24
AU2002252949A1 (en) 2003-11-11
DE60233825D1 (en) 2009-11-05
EP2186792A8 (en) 2010-08-04
KR20040106457A (en) 2004-12-17
EP2186792A1 (en) 2010-05-19
CA2485479A1 (en) 2003-11-20
CN1243541C (en) 2006-03-01
US20050288263A1 (en) 2005-12-29
IL164845A (en) 2011-08-31
EP1508561A1 (en) 2005-02-23
EP2186792B1 (en) 2015-11-25
JP2005524715A (en) 2005-08-18

Similar Documents

Publication Publication Date Title
US7550507B2 (en) 2-(alpha-hydroxypentyl) benzoate and its preparation and use
EP3838887B1 (en) 2-(1-acyloxypentyl) benzoic acid salt formed by basic amino acid or aminoguanidine, preparation method therefor and uses thereof
US5190947A (en) Codeine salt of a substituted carboxylic acid, its use and pharmaceutical compositions thereof
EP0318330A3 (en) Glutathione esters for treating ventricular arrhythmia
JPH02270855A (en) Anti-inflammatory aryl compound
WO2005123651A1 (en) L-2-(α-HYDROXYPENTYL)BENZOATES, THE PREPARATION AND THE USE THEREOF
US3697563A (en) (3,4,5-trimethoxy-benzamido)-alkanoic acids for prophylaxis and treatment of cardiac disorders
RU2305092C2 (en) Novel 2-(alpha-hydroxypentyl)benzoates, their preparing and using
SE453387B (en) 2-AMINO-3- (HALOGENEBENZOYL) -METHYLPHENYLETIC ACIDS, ESTARS AND SALTS THEREOF
JP2010189418A (en) NEW 2-(alpha-HYDROXYPENTYL)BENZOATE, METHOD FOR PRODUCING THE SAME AND USE THEREOF
US4356306A (en) Water-soluble derivatives of 6,6&#39;-methylene-bis-(2,2,4-trimethyl-1,2-dihydroquinoline)
US4510147A (en) Compositions for and medical use of water-soluble derivatives of 6,6-methylene-bis-(2,2,4-trimethyl-1,2-dihydroquinoline)
US3269909A (en) Analgesic methods and compositions of benzoic acid derivatives
US3966960A (en) 3-(4-Biphenylcarbonyl)propionic acid as an inhibitor of platelet aggregation
JPS632988A (en) 1,3-dithiol-2-ylidenesulfonylacetic acid derivative
JPH05112463A (en) Therapeutic agent for cataract
US3726913A (en) Derivatives of aminoalkanoic acids
KR100344099B1 (en) Novel biphenyldicarboxylate derivative and method for manufacturing the same
US3849572A (en) Biphenyleneacetic acid anti-inflammatory agents
HU203839B (en) Process for producing pharmaceutical composition containing acrylic acid derivatives
GB1579839A (en) Chlorophenoxybenzene derivatives
US4254162A (en) Phenoxy acetic acid derivative, its preparation and therapeutic use
US3852467A (en) Antiinflammatory compositions
JP2007528878A (en) Novel 2- (α-n-pentanonyl) benzoate and production method and use thereof
MXPA03007192A (en) Novel benzoylguanidine salt

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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