WO2007124617A1 - Dérivés de coumarine, leurs procédés de préparation, compositions pharmaceutiques et utilisations - Google Patents

Dérivés de coumarine, leurs procédés de préparation, compositions pharmaceutiques et utilisations Download PDF

Info

Publication number
WO2007124617A1
WO2007124617A1 PCT/CN2006/000839 CN2006000839W WO2007124617A1 WO 2007124617 A1 WO2007124617 A1 WO 2007124617A1 CN 2006000839 W CN2006000839 W CN 2006000839W WO 2007124617 A1 WO2007124617 A1 WO 2007124617A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
compound
cisplatin
substituted
independently selected
Prior art date
Application number
PCT/CN2006/000839
Other languages
English (en)
French (fr)
Inventor
Ping Xie
Xiaoguang Chen
Shiping Xu
Hongyan Li
Lanmin Li
Yanli Zhou
Yue Liu
Zhigang Luo
Xiaozhen Jiao
Xuguang Zheng
Furong Zhang
Original Assignee
Institute Of Mataria Medica, Chinese Academy Of Medical Sciences
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 Mataria Medica, Chinese Academy Of Medical Sciences filed Critical Institute Of Mataria Medica, Chinese Academy Of Medical Sciences
Priority to PCT/CN2006/000839 priority Critical patent/WO2007124617A1/zh
Publication of WO2007124617A1 publication Critical patent/WO2007124617A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/22Oxygen atoms attached in position 2 or 4
    • C07D215/227Oxygen atoms attached in position 2 or 4 only one oxygen atom which is attached in position 2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/22Oxygen atoms attached in position 2 or 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/48Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • C07D215/50Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen attached in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/48Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • C07D215/54Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen attached in position 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms

Definitions

  • the invention discloses a new structural type of coumarin and dihydroquinolinone derivatives and pharmaceutically acceptable salts thereof, a preparation method of the same, a pharmaceutical composition containing the same, and particularly for preparing chronic renal failure , diabetes, sputum blood pressure and cardiovascular and cerebrovascular diseases as well as cirrhosis and prostatic hypertrophy drugs. Background technique
  • the compound of the present invention has significant effects of inhibiting transforming growth factor ⁇ ⁇ (TGF P 1 ) and reducing angiotensin n (An g II) and renin, and thus the compound of the present invention has potential treatment for chronic renal failure, hypertension, The role of diabetes, cirrhosis and prostatic hypertrophy and pulmonary fibrosis.
  • Renal protection, hypotension, and reduction of diabetic nephropathy urinary protein levels may differ from angiotensin-converting enzyme inhibitors (ACEI) and angiotensin II ATI-type receptor antagonists (AT1RA). It is related to the inhibition of TGF- ⁇ 1 and the decrease of TGF- ⁇ 1 mRNA expression in renal tissues and the inhibition of matrix metalloproteinase activity secreted by cells. Thereby inhibiting glomerular sclerosis and renal interstitial fibrosis.
  • ACEI angiotensin-converting enzyme inhibitors
  • AT1RA angiotensin II ATI-type receptor antagonists
  • Cisplatin tumor chemotherapy will likely be used to alleviate the renal toxicity of Cisplatin tumor chemotherapy; for the treatment of chronic renal insufficiency caused by hypertension and diabetes; prevention and treatment of glomerular sclerosis and renal fibrosis caused by various causes; Treatment of type 2 diabetes to prevent further development of diabetic nephropathy.
  • the protective effect of the drug on cisplatin-induced nephrotoxicity is used here to initially evaluate the activity and efficacy of the compound.
  • a further technical problem to be solved by the present invention is to provide a new class of pharmaceutical compositions containing the compounds of the present invention and carriers conventionally used in the pharmaceutical field.
  • a further technical problem to be solved by the present invention is to provide the use of such novel compounds for the preparation of a medicament for the treatment of chronic renal failure, diabetes, hypertension and cardiovascular and cerebrovascular diseases, as well as cirrhosis and prostatic hypertrophy.
  • the present invention uses the following technical solutions.
  • X is selected from 0, NH;
  • W is selected from C0, CH 2 ;
  • R is selected from a linear or branched - 6 alkyl group
  • R 6 , R 7 and R 8 are independently selected from the group consisting of H, 0H, N0 2 , carboxyl, halogen, .8 alkyl, .8 methoxy; R 3 is selected from
  • a substituted or unsubstituted phenyl.4 alkyl group (the substituent on the phenyl ring is R 12 , the substituent on the fluorenyl group is R"); a substituted or unsubstituted N-pyrrolyl group, (a substituent on the pyrrole ring) For R 13 );
  • R" is selected from C 1-4 oxiranyloxy
  • R 13 represents a mono- or poly-substituent, and the substituent is independently selected from the group consisting of H, 0H, halogen, N0 2 , C0 2 H, C 1-8 mercapto, .8 haloalkyl, C s carbonylalkyl, C 8 alkoxy a group, a CL 8 carboxyalkoxy group, a d. 8 acyl group, a d- 8 alkoxycarbonyl group, a d- 8 aryl group, a tetrazolyl group, an unsubstituted or substituted phenylcarbonyl group, and a substituent on the benzene ring is R 14 ;
  • R 15 , Ri 6 and R 17 each represent a mono- or poly-substitution, and the substituents are independently selected from the group consisting of H, 0H, halogen, N0 2 , C0 2 H, d- 8 alkyl, - 8 haloalkyl, d. 8 carbonyl fluorenyl, C 1-8 alkoxy, C 1-8 ;
  • Preferred compounds include, but are not limited to, as shown in formula (IA)
  • RX is selected from the group consisting of 0, NH, NCH 3 and NCH(CH 3 ) 2 ;
  • R 6 , R 7 and independently selected from the group consisting of H, OH, Cl, N0 2 , C 14 alkyl, C 1-4 fluorenyloxy;
  • Rn is mono- or poly-substituted, and the substituents are independently selected from the group consisting of H, OH, Cl, N0 2 , C 1-4 alkyl. 1-4 alkoxy, CF 3
  • RX is selected from the group consisting of O, should, NCH 3 , NCH(CH 3 ) 2 ;
  • R 7 and R 8 are independently selected from the group consisting of H, OH, Cl, N0 2 , C M alkyl, C M alkoxy;
  • R" is selected from the group consisting of C0 2 H, C0 2 CH 3 , C0 2 Et ;
  • R 12 is mono- or poly-substituted, and the substituents are independently selected from the group consisting of H, OH, Cl, N0 2 , CM alkyl, C methoxy, CF 3 , OCH 2 C0 2 H, formyl, acetyl, C0. 2 H, C0 2 CH 3 , C0 2 Et, ( ⁇ ,, 2,, 3", 4,,-tetrazole-5,,).
  • Preferred compounds include, but are not limited to, as shown by the general formula (IC)
  • RX is selected from the group consisting of O, NH, NCH 3 , NCH(CH 3 ) 2;
  • R 13 is mono- or poly-substituted, and the substituents are independently selected from the group consisting of H, OH, Cl, N0 2 , d. 4 alkyl, C M alkoxy, CF 3 ,
  • Compounds include, but are not limited to, as shown in formula (IC1)
  • RX is selected from the group consisting of 0, NH, NCH 3 and NCH(CH 3 ) 2 ;
  • R 14 is mono- or poly-substituted, and the substituents are independently selected from the group consisting of H, OH, Cl, N0 2 , C w alkyl, C 1-4 alkoxy, CF 3 ,
  • Preferred compounds include, but are not limited to, as shown by the general formula (ID)
  • RX is selected from the group consisting of O, NH, NCH 3 , NCH(CH 3 ) 2;
  • R 6 , R 7 and R 8 are independently selected from the group consisting of H, OH, Cl, N0 2 , C M alkyl, _ 4 methoxy;
  • R 15 is mono- or poly-substituted, and the substituents are independently selected from the group consisting of H, OH, Cl, N0 2 , C M alkyl, C M methoxy, CF:
  • Preferred compounds include, but are not limited to, as shown by the general formula (IE)
  • RX is selected from the group consisting of 0, NH, NCH 3 and NCH(CH 3 ) 2 ;
  • R 6 , R 7 and R 8 are independently selected from the group consisting of H, OH, Cl, N0 2 , C M alkyl, C 1-4 alkoxy;
  • R 16 is mono- or poly-substituted, and the substituents are independently selected from the group consisting of H, OH, Cl, N0 2 , Q 4 alkyl, C M alkoxy, CF 3
  • Preferred compounds include, but are not limited to, as shown in formula (IF)
  • RX is selected from the group consisting of O, NH, NCH 3 and NCH(CH 3 ) 2 ;
  • R 6 , R 7 and R 8 are independently selected from the group consisting of H, OH, Cl, N0 2 , C M decyl, Q. 4 alkoxy;
  • R 17 is mono- or poly-substituted, and the substituents are independently selected from the group consisting of H, OH, Cl, N0 2 , C 1-4 fluorenyl, C 1-4 decyloxy, CF 3
  • More preferred compounds include, but are not limited to, one of the groups selected from the group consisting of the following compounds
  • halogen means fluorine, chlorine, bromine or iodine.
  • the compound of the present invention may exist in the form of an isomer, and generally, the “compound of the present invention” includes an isomer of the compound.
  • the compound of the present invention may exist as a cis-trans isomer of a double bond having an asymmetric configuration having an S configuration or an R configuration, and the present invention encompasses all possible stereoisomers and mixtures of two or more isomers. If cis/trans isomers are present, the invention relates to cis form and trans form as well as mixtures of these forms, if desired, a single foreign object can be isolated according to conventional methods or by stereoselective synthesis.
  • the compound of the present invention further includes a pharmacologically acceptable salt thereof, a hydrate of a salt, an ester or a prodrug.
  • a process for the preparation of a compound of the present invention which is prepared by reacting various substituted coumarins of 3-carboxyl groups with corresponding various substituted amino compounds. The amidation reaction is carried out under the appropriate reactants, catalysts and suitable solvents.
  • Another class of compounds is prepared by reacting various substituted aniline compounds with chloroacetyl chloride to provide an intermediate of a portion of the compounds of the invention.
  • the preferred reactants are phosphorus pentachloride, phosphorus oxychloride, ruthenium, osmium-dimethylformamide, phosphorus trichloride and thionyl chloride, oxalyl chloride, more preferably phosphorus pentachloride, phosphorus oxychloride. , thionyl chloride.
  • the catalyst used in the preparation of the compound of the present invention includes a tertiary amine, pyridine, 4-dimethylaminopyridine, 4-pyrrolidinopyridine and the like. Among them, preferred are tertiary amines and pyridines. More preferably, it is pyridine.
  • the reaction is carried out in a suitable solvent or in the above condensing agent, such as dichloromethane, 1,2-dichloroethane, ethylene glycol dimethyl ether, tetrahydrofuran and hydrazine, hydrazine-dimethylformamide (DMF), etc. .
  • reaction temperature is 10 to 110 ° C, preferably 20 to 90 ° C, more preferably 30 to 80 ° C, and particularly preferably 50 to 70. C.
  • the following reaction equations are specified
  • the invention therefore also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising as an active ingredient a compound of the invention and a conventional pharmaceutical excipient or adjuvant.
  • the pharmaceutical composition of the present invention usually contains 0.1 to 95% by weight of the compound of the present invention.
  • compositions of the compounds of the invention can be prepared according to methods well known in the art.
  • the compounds of the invention may be combined with one or more solid or liquid pharmaceutical excipients and/or adjuvants to provide a suitable administration form or dosage for use as a human or veterinary drug. form.
  • the compound of the present invention or a pharmaceutical composition containing the same may be administered in a unit dosage form, which may be enterally or parenterally, such as orally, muscle, subcutaneous, nasal, oral mucosa, skin, peritoneum or rectum.
  • injections include intravenous, intramuscular, subcutaneous, intradermal, and acupoint injections.
  • the dosage form can be a liquid dosage form or a solid dosage form.
  • the liquid dosage form may be a true solution, a colloid, a microparticle dosage form, an emulsion dosage form, or a suspension dosage form.
  • Other dosage forms such as tablets, capsules, pills, aerosols, pills, powders, solutions, suspensions, emulsions, granules, suppositories, frozen powders, and the like.
  • the compounds of the present invention can be formulated into common preparations, sustained release preparations, controlled release preparations, targeted preparations, and various microparticle delivery systems.
  • a carrier for example, a diluent and an absorbent such as starch, dextrin, calcium sulfate, lactose, mannitol, sucrose, sodium chloride, glucose, urea, calcium carbonate, kaolin, microcrystalline cellulose, silicic acid.
  • a diluent and an absorbent such as starch, dextrin, calcium sulfate, lactose, mannitol, sucrose, sodium chloride, glucose, urea, calcium carbonate, kaolin, microcrystalline cellulose, silicic acid.
  • wetting agents and binders such as water, glycerin, polyethylene glycol, ethanol, propanol, starch syrup, dextrin, syrup, honey, glucose solution, gum arabic, gelatin syrup, sodium carboxymethyl cellulose , shellac, methylcellulose, potassium phosphate, polyvinylpyrrolidone, etc.
  • disintegrating agents such as dried starch, alginate, agar powder, brown algae starch, sodium bicarbonate and citric acid, calcium carbonate, polyoxyethylene sorbus Sugar alcohol fatty acid ester, sodium dodecyl sulfonate, methyl cellulose, ethyl cellulose, etc.
  • disintegration inhibitors such as sucrose, glyceryl tristearate, cocoa butter, hydrogenated oil, etc.
  • absorption enhancer For example, quaternary ammonium salts, sodium lauryl sulfate, etc.
  • lubricants such as talc, silica, corn starch, stearate,
  • a carrier for example, a diluent and an absorbent such as glucose, lactose, starch, cocoa butter, hydrogenated vegetable oil, polyvinylpyrrolidone,
  • Gelucire, kaolin, talcum powder, etc. binders such as acacia, tragacanth, gelatin, ethanol, honey, liquid sugar, rice cereal or batter; etc.; disintegrating agents, such as agar powder, dried starch, alginate, Sodium dodecyl sulfate, methyl cellulose, ethyl cellulose, and the like.
  • the active ingredient compound of the present invention is mixed with the various carriers described above, and the resulting mixture is placed in a hard gelatin capsule or soft capsule.
  • Active Ingredient The compound of the present invention can also be formulated into a microcapsule, suspended in an aqueous medium to form a suspension, or can be enclosed in a hard capsule or used as an injection.
  • the compound of the present invention is formulated into an injectable preparation such as a solution, a suspension solution, an emulsion, or a lyophilized powder injection, which may be aqueous or non-aqueous, and may contain one and/or more drugs.
  • a pharmaceutically acceptable carrier, diluent, binder, lubricant, preservative, surfactant or dispersing agent may be selected from the group consisting of water, ethanol, polyethylene glycol, 1,3-propanediol, ethoxylated isostearyl alcohol, polyoxylated isostearyl alcohol, polyoxyethylene sorbitan fatty acid ester, and the like.
  • an appropriate amount of sodium chloride, glucose or glycerin may be added to the preparation for injection, and a conventional cosolvent, a buffer, a pH adjuster or the like may be added. These excipients are commonly used in the field.
  • a coloring agent e.g., a preservative, a flavor, a flavoring agent, a sweetener or the like may be added to the pharmaceutical preparation.
  • the pharmaceutical or pharmaceutical composition of the present invention can be administered by any known administration method for the purpose of enhancing the therapeutic effect for the purpose of administration.
  • the activity and effect of the compounds and/or compositions of the present invention are determined by known in vitro and in vivo assay methods, and the continued synthesis of coumarin and dihydroquinoline compounds has significant renal protective effects.
  • the compound of the present invention is cisplatin (Cisplatin). It can protect rat renal mesangial cells and human renal tubular epithelial cells from injury in vitro; it can significantly reduce serum urea nitrogen (BUN) and creatinine (Scr) levels and urine in acute rat kidney injury induced by Cisplatin in vivo.
  • UP Protein (UP) level, reduce renal edema caused by Cisplatin; also reduce serum BU level, Scr level and urinary protein (UP) level in 5/6 nephrectomized rats; reduce streptozotocin-induced rat diabetes Kidney disease urinary protein levels.
  • the compound 31 of the present invention is neuroprotective, hypotensive, and reduces the level of urinary protein in diabetic nephropathy unlike angiotensin converting enzyme inhibitor (ACEI) and angiotensin ⁇ type 1 receptor antagonist (AT1RA). It is related to the inhibition of TGF- ⁇ 1 and the decrease of TGF- ⁇ 1 mRNA expression in renal tissues and the inhibition of matrix metalloproteinase activity secreted by cells. The main route may be attributed to the inhibition of glomerular sclerosis and renal interstitial fibrosis.
  • ACEI angiotensin converting enzyme inhibitor
  • AT1RA angiotensin ⁇ type 1 receptor antagonist
  • the compounds of the present invention may be useful for relieving renal toxicity of Cisplatin tumor chemotherapy; for treating chronic renal insufficiency caused by hypertension and diabetes; preventing and treating glomerular sclerosis and renal fibrosis caused by various causes; Treatment of ⁇ -type diabetes to prevent further development of diabetic nephropathy.
  • the compounds of the present invention can be used for the preparation of a transforming growth factor-P l CTGF- ⁇ ⁇ inhibitor, an angiotensin II ( ⁇ ) converting enzyme receptor antagonist, which can be used for the treatment of cardiovascular and cerebrovascular diseases, kidney diseases, Diabetes, cirrhosis, and enlarged prostate.
  • cardiovascular and cerebrovascular diseases described above are hypertension, heart, cerebral embolism, myocardial infarction, and stroke.
  • the dosage of the pharmaceutical composition of the compound of the present invention depends on a number of factors, such as the nature and severity of the disease to be prevented or treated, the sex, age, weight, personality and individual response of the patient or animal, the route of administration, the number of administrations, For therapeutic purposes, the therapeutic dose of the present invention can vary widely. In general, the dosage of the pharmaceutical ingredient of the present invention is well known to those skilled in the art.
  • the prophylactic or therapeutic effect of the present invention can be accomplished by appropriately adjusting the amount of the actual drug contained in the final formulation of the compound composition of the present invention to achieve its therapeutically effective amount.
  • Suitable daily dosage ranges for the compounds of the invention are from 0.001 to 150 mg/kg body weight, preferably from 0.1 to 100 mg/kg body weight, more preferably from 1 to 60 mg/kg body weight, most preferably from 5 to 45 mg/kg body weight. body weight.
  • the above dosages may be administered in a single dosage form or divided into several, e.g., two, three or four dosage forms, which are limited by the clinical experience of the administering physician and include dosage regimens employing other therapeutic means.
  • the total dose required for each treatment can be divided into multiple or single dose administrations.
  • the compounds or compositions of this invention may be administered alone or in combination with other therapeutic or symptomatic agents and adjusted in dosage.
  • BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 Protective effect of compound 31 of the invention on cisplatin-induced damage to rat mesangial cells (rMC).
  • Figure 3 Morphological observation of the effect of compound 31 of the present invention on apoptosis of human renal tubular epithelial cells (HKC)
  • Example 1 3-[4,-(1",2"3",4"-tetrazole-5")phenylaminocarbonyl1-6-nitro-7-hydroxy-8-methylcoumarin (31 Preparation After the reaction of 3-carboxy-6-nitro-7-hydroxy-8-methylcoumarin 10.6 g (0.04 Mol) with 30 ml of thionyl chloride, the excess thionyl chloride was removed and 20 ml was added. Dimethylformamide (DMF), 40 ml of pyridine and 7 g (0.043 Mol) of 4-aminophenyl (hydrazine, 2,3,4,-tetrazole-5 were stirred well and heated at 50 ° C for 4 hours. , washed with ethanol, water, dried
  • Example 2 3-[3'-(1",2"3",4"-tetrazole-5")phenylaminocarbonyl]-6-nitro-7-hydroxy-8-methylcoumarin (32 Preparation According to the preparation method of the compound of Example 31, the preparation of the compound 32 was carried out by reacting 3-aminophenyl (hydrazine, 2', 3', 4'-tetrazole-5 s ) to obtain a compound 32.
  • the preparation of the compound 33 was carried out by using 3-carboxy-6-nitro-7-hydroxy-8-butylcoumarin and 3-aminophenyl (oxime, 2', 3',4'-tetrazole-5!
  • the preparation of the compound 28 was carried out by using 1-isopropyl-3-carboxy-6-nitrodihydroquinolinone-2 and 4-( ⁇ , 2'3', 4'-tetrazole-5') benzene ammonia is reacted to obtain compound 28
  • the preparation of the compound 29 was carried out by using 1-isopropyl-3-carboxy-6-nitrodihydroquinolinone-2 and 3-(1',2'3'. , 4'-tetrazole-5') benzene ammonia is reacted to obtain compound 29
  • the compound 67 was prepared by reacting 2-ethoxycarbonylpyrrole with 2-chloro-3-chloromethyldihydroquinoline to obtain the compound 67.
  • the compound 69 was prepared by reacting 3-trifluoromethyl-4-ethoxycarbonylpyrazole with 2-chloro-3-chloromethyldihydroquinoline to obtain a compound. 69
  • the compound 64 was prepared by reacting 2-formyl-pyrazole with 2-chloro-3-chloromethyldihydroquinoline to obtain a compound 64.
  • the compound 70 was prepared by reacting 3-n-propyl-4-ethoxycarbonylpyrazole with 2-chloro-3-chloromethyldihydroquinoline to obtain a compound 70.
  • the compound 66 was prepared by reacting 4-ethoxycarbonyl 5'-methylimidazole with 2-chloro-3-chloromethyldihydroquinoline to obtain a compound 66.
  • the compound 72 was prepared by reacting 2-carboxypyrrole with 2-chloro-3-chloromethyldihydroquinoline to obtain a compound 72.
  • the compound 68 was prepared by reacting 2-carboxyimidazole with 2-chloro-3-chloromethyldihydroquinoline to obtain a compound 68.
  • the compound 65 was prepared by reacting 4-carboxy 5'-methylimidazole with 2-chloro-3-chloromethyldihydroquinoline to obtain a compound 65.
  • the compound 51 was prepared by reacting 2-acetylpyrrole with 3-chloromethyl-7-methoxydihydroquinolinone-2 to obtain a compound 51.
  • the compound 53 was prepared by using 3-(4'-methoxyphenylcarbonyl)pyrrole and 2-chloro-3-chloromethyl-7-methoxydihydroquine. The morpholine is reacted to obtain compound 53
  • mice Male Kunming (KM) mice, 16g ⁇ 22g, were randomly divided into the vehicle control group and the cisplatin model group and the drug-administered group, with 8 rats in each group.
  • the control group was intraperitoneally injected with normal saline, cisplatin was dissolved in physiological saline, and intraperitoneally injected, according to 7 mg/kg.
  • the above dosages were all 0.4ml/20g, and the administration was started 2 days before the injection of cisplatin.
  • the blood was taken from the eyeballs on the 3rd, 5th, and 7th days after the injection of cisplatin.
  • the serum BU and Scr were detected by the kit. And weigh the weight.
  • Cisplatin 7 1 ⁇ 54 ⁇ 0 ⁇ 32 7.9 39.71 ⁇ 5.86 30.1 ⁇
  • Rat mesangial cells (rMC) with exponential growth phase were added with appropriate amount of 0.02% EDTA.
  • the cells were digested with 0.25% trypsin solution to cause the adherent cells to fall off.
  • a cell suspension having a concentration of 10 4 /ml was prepared using RPMI1640 medium (containing 20% calf serum), and 0.1 ml was inoculated per well in a 96-well plate. The plates were placed in a 37 ° C, 5% CO 2 incubator, and different concentrations of drug were added after 24 hours.
  • the compound 31 of the present invention has different concentrations of cisplatin at 0, 33, 100 ⁇ mol/L, respectively.
  • Combination group, each set has 3 parallel holes.
  • the medium was discarded, and 0.1 ml of serum-free RPMI1640 medium-prepared MTT (0.5 mg/ml) was added to each well, and incubated at 37 ° C for 4 hours, and the living cells were able to reduce MTT to formazan.
  • the supernatant was discarded, and the formazan was dissolved in 150 ⁇ l of DMSO and shaken well on a plate shaker.
  • the absorbance (OD) was measured at a wavelength of 570 nm using a microplate reader at a reference wavelength of 450 nm.
  • the principle of the MTT experiment is based on the ability of living cells to reduce tetrazolium bromide (MTT) to a blue-violet DMSO-soluble formazan compound, whereas dead cells do not.
  • Formazan dissolved in DMSO has a strong absorption peak at 570 nm, and this absorption value has a good linear relationship with the number of living cells.
  • the protective effect of the compound 31 of the present invention on rat mesangial cells under the action of cisplatin was observed by MTT method.
  • the compound 31 of the present invention has a protective effect against rMC toxicity caused by cisplatin.
  • the inhibitory rates of 0.12 ⁇ mol/L cisplatin on the growth of the control group, 33 ⁇ mol/L of the compound of the present invention group 31, and 100 ⁇ mol/L of the compound of the present invention group 31 were 10.7%, 8.3%, 3.8%, respectively; 0.37 mol/
  • the inhibitory rates of L cisplatin on the cell growth of the control group, 33 ⁇ mol/L of the compound of the present invention group 31, and 100 ⁇ mol/L of the compound of the invention group 31 were 19.0%, 12.8%, and 7.6%, respectively; 1 ⁇ mol/L cisplatin pair
  • the inhibition rate of cell growth of 33 ⁇ mol/L of the compound of the present invention group 31 and 100 ⁇ mol/L of the compound of the present invention group was 25.4%, 18.2%, and 12.0%, respectively.
  • the compound 31 of the present invention showed no significant protective effect on
  • Cisplatin Inhibition Cisplatin Inhibition
  • control group (n 6), cisplatin 0.12, 0.5, 2.5 ⁇ mol/L in different concentration groups, the compound 31 of the present invention in each group of 0, 2, 10, 50 u mol/L and cisplatin respectively.
  • MTT 0.5 mg/m
  • serum-free RPMI1640 medium 0.1 ml
  • Live cells can reduce MTT to formazan.
  • the supernatant was discarded, and the formazan was dissolved in 150 ⁇ l of DMSO and shaken well on a plate shaker.
  • the absorbance (OD) was measured at a wavelength of 570 nm using a microplate reader at a reference wavelength of 450 nm.
  • the protective effect of the compound 31 of the present invention on rat HKC cells under the action of cisplatin was observed by the MTT method.
  • the compound 31 of the present invention has a protective effect against HKC toxicity caused by cisplatin.
  • the inhibitory rate of 0.12 ⁇ mol/L cisplatin against the control group, 2 ⁇ mol/L of the compound of the present invention group 31, 10 ⁇ mol/L of the compound of the present invention 31, 50 ⁇ mol/L of the compound of the present invention group 31 was 5.9%, respectively.
  • Coomassie Brilliant Blue G-250 is brownish-red (free) in an acidic solution. When it is combined with a protein by hydrophobic interaction, it turns blue and has a maximum absorption peak at 595 nm. A standard curve was established using calf serum albumin (BSA) as the standard protein. The urine protein concentration was estimated based on the standard curve.
  • BSA calf serum albumin
  • Standard curve Take standard protein (lmg/ml) 0, 5, 10, 20, 40, 60, 80 ⁇ ⁇ , make up to ⁇ ⁇ ⁇ with double distilled water, so that the final concentration is 0, 50, 100, 200, 400 , 600, SOO g / mL force ⁇ 5ml G-250 dye solution, mixed hook, placed at room temperature for 15min, measured OD value at 595nm.
  • Determination Take a urine sample 100 ⁇ 1 and operate as above.
  • 50 male Wistar rats were randomly divided into 5 groups according to body weight, 10 in each group, respectively, solvent control group, cisplatin model group, cisplatin combined with Benazapril 10 mg/kg group, cisplatin combined with compound of the invention 31 10 mg/kg group, cisplatin combined Compound 31 of the invention 31 30 mg/kg group.
  • the solvent control group received intraperitoneal injection of 10 ml/kg normal saline; the cisplatin model group received intraperitoneal injection of cisplatin 6 mg/kg-time; cisplatin combined with benazepril 10 mg/kg group was intraperitoneally injected with cisplatin 6 mg/kg for 3 days before administration of benazepril 10 mg/kg for 3 days.
  • benapapril 10 mg/kg was administered once daily for 4 days for a total of 7 days; cisplatin was combined with the compound of the present invention 31 10 mg/kg group and cisplatin in combination with the compound of the invention 31 30 mg/kg group was intraperitoneally injected with cisplatin 6 mg
  • the compound of the present invention, 31 mg/kg, 30 mg/kg was administered orally for 3 days in the first 3 days. Thereafter, the compound of the present invention 31 10 mg/kg and 30 mg/kg-times were administered intragastrically daily for a total of 4 days, for a total of 7 days.
  • Each of the above administration volumes was 10 ml/kg.
  • the body weight was weighed daily, and the urine volume of the rats was collected for 24 hours after the last administration, and the urine protein amount was measured by the G-250 method for 24 hours. Twenty-four hours after the last administration, the rats were anesthetized with sodium pentobarbital 30 mg/kg, and blood was taken from the eyelids. Serum BUN, Scr, TGF- ⁇ i and plasma ANGII concentrations were measured using a kit. ⁇
  • kidney tissue was taken, weighed, and the kidney coefficient was calculated.
  • 10% tissue homogenate was prepared from 1/2 left kidney with phosphate buffer (pH 7.4) to measure lipid peroxidation and glutathione levels; the rest was prepared with Trizol 10% tissue homogenate to extract total RNA.
  • RT-PCR analysis of renal tissue TGF- ⁇ ! mRNA level.
  • the right kidney was treated with a neutral formalin for pathological examination.
  • the observation model group cisplatin combined with bennazapril group, cisplatin in combination with the compound of the present invention 31 10 m g / kg group, cisplatin combined with the compound of the invention 31 30 mg / kg group of rats, serum biochemistry and body weight, urine protein, The organ index was changed, and the rats were sacrificed 4 days after administration, and the kidneys were taken for pathological examination.
  • Cisplatin model 5.65 ⁇ 3.43 m 354.6
  • the body weight of the rats was significantly reduced, the kidney tissue was hypertrophied, and the color was dark red or gray.
  • the three groups had less weight loss and less kidney index. Wherein cisplatin was combined with the compound of the present invention 31.
  • the kidney index of the 30 mg/kg group was statistically significant compared with the cisplatin model group (p ⁇ 0.05) (see Table 12).
  • the serum biochemical test BU and Src kits are produced by Beijing Beihua Fine Chemicals Co., Ltd.; the plasma ANG II radioimmunoassay kit is the product of Beijing North Biotechnology Research Institute; TGF- ⁇ 1 ELISA kit is Biotech product Shanghai Senxiong Technology Industrial Co., Ltd. Dispense.
  • Urine protein assay Rats were placed in metabolic cages for 24 hours of urine, and urine protein content was determined using the Coomassie Brilliant Blue G-250 method. A standard curve was established using calf serum albumin (BSA) as the standard protein. Calculate the urine protein concentration based on the standard curve and calculate the 24-hour urine protein amount (UP ay- ⁇
  • Compound 31 of the present invention is administered by intragastric administration 6 times a week for 16 weeks (12 weeks of administration); benazepril and losartaii are administered intragastrically 6 times a week for 16 weeks ( Dosing for 12 weeks).
  • the sham operation group and the model group were intragastrically administered with a suspension of 0.5% sodium carboxymethylcellulose (CMC) in a volume of 10 ml.kg" 1 and the solvent control was continued for 16 weeks (12 weeks of administration).
  • CMC carboxymethylcellulose
  • Plasma TGF- ⁇ was determined by ELISA! Level, the rats were placed in a metabolic cage to collect urine for 24 hours, and the amount of urine protein was measured (UP ay); the growth of the rats was observed by weighing weekly; after 16 weeks (12 weeks of administration), in addition to measuring the above indicators, Animals were sacrificed in each group, the weight of the heart was weighed, the heart index was calculated, and the kidneys were taken for pathology.
  • the losartan group, the benazepril group, the compound of the present invention 31 10 mg/kg, and the compound of the present invention 31 30 mg/kg decreased by 21.4% (P ⁇ 0.05), 14.9%, 20.3% > 34.9%, respectively. ⁇ 0 ⁇ 01).
  • the results are shown in Table 17. Compared with the sham operation group, the heart index of the model group increased by 17.8% (P ⁇ 0.01).
  • the losartan group, the benazepril group, the compound of the present invention 31 10 mg/kg, and the compound 30 30 mg/kg of the present invention decreased by 18.0% (P ⁇ 0.01), 6.4% 2.4%, and 13.7% (P ⁇ 0.01), respectively, compared with the model group.
  • the losartan group and the compound of the present invention 31 30 mg/kg group had a significantly decreased cardiac index, which was similar to the sham operation group.
  • Urine protein assay Rats were placed in metabolic cages for 24 hours of urine and urine protein levels were determined using the Coomassie Brilliant Blue G-250 method. A standard curve was established using calf serum albumin (BSA) as the standard protein. Calculate the urine protein concentration based on the standard curve and calculate the 24-hour urine protein amount (UP ay ⁇
  • a control group a model group, a losartan (10 mg/kg) group, and a compound of the present invention 31 10 mg/kg were administered. Simultaneous administration of the mold.
  • Compound 31 of the present invention is administered intragastrically, 6 times a week for 24 weeks; losartan is administered intragastrically, 6 times a week for 24 weeks.
  • the control group and the model group were intragastrically administered with a suspension of 0.5% sodium carboxymethylcellulose (CMC) in a volume of 10 ml.kg" for 24 weeks.
  • CMC sodium carboxymethylcellulose
  • the animals were anesthetized with ether, blood was taken from the posterior venous plexus, serum BUN, Scr, Glu levels were measured, plasma ANGII levels were determined by radioimmunoassay, and the rats were placed in metabolic cages.
  • the urine volume was collected for 24 hours, and the amount of urine protein (UP a 1 ) was measured.
  • the growth of the rats was observed by weighing the rats every week. After 16 weeks and 24 weeks, the animals were sacrificed and the kidneys were taken for pathology.
  • the Glu level in the model group increased by 473.7%; the BUN level increased by 61.8% (P ⁇ 0.05), and the Scr. level increased by 18.1% (P ⁇ 0.05), indicating that the diabetic rat model was established.
  • the model was successful and the kidney function of the rat had been damaged.
  • Glu and BUN Scr. were also elevated in the rats in each administration group, and no significant improvement in renal function was observed.
  • the ANGII level of the rats in the 31 group was lower than that in the model group.
  • the Glu level in the model group increased by 409.9% (P ⁇ 0.05); the BUN level increased by 65.9% (P ⁇ 0.05), and the Scr. level increased by 25.9% (P ⁇ 0.05).
  • the Glu and Scr. in the rats in the drug-administered group also increased.
  • the BU level in the rats in the losarta group and the compound group 31 decreased, which was 25.8% and 17.0% lower than that in the model group.
  • the Glu level in the model group was increased by 484.2% (P ⁇ 0.05); the BUN level was increased by 88.2% (P ⁇ 0.05), and the Scr. level was increased by 21.1%.
  • Glu, BUN, and Scr. were also elevated in the rats in each administration group.
  • the urine protein level of the model group was increased by 63.9%, and the rats of the l 0 sart an group and the compound of the present invention group 31 were decreased by 23.4% and 47.5%, respectively.
  • Table 20 Effect of Compound 31 of the present invention on various indexes of rat diabetic nephropathy model induced by streptozotocin (12 weeks after modeling)
  • the Glu level in the model group increased by 512.5% (P ⁇ 0.05); the BUN level increased by 127.7% (P ⁇ 0.05), and the Scr. level increased by 42.6%.
  • the Glu. of the rats in each administration group also increased.
  • the BU level of the rats in the 31st group of the present invention decreased, which was 19.1% lower than that of the model group; the Scr. level of the losartan group decreased, which was 27.1% lower than that of the model group; plasma ANGII Level, no difference in each group.
  • the kidney organ index of the model group increased by 105.5% (P ⁇ 0.05), and the rats of each administration group also increased.
  • Table 22 Compound 31 of the present invention caused by streptozotocin Effects of various indicators of rat diabetic nephropathy model (16 weeks after modeling)
  • GROUP n UP (mg .day") Increase rate (%) Inhibitory rate (%)
  • HEC Human renal tubular epithelial cells in the exponential growth phase were digested with appropriate amount of 0.25% trypsin solution containing 0.02% EDTA to cause adherent cells to fall off.
  • HKC cells were collected, washed once with PBS, centrifuged, supernatant removed, fixed cells at 4 °C, 4% paraformaldehyde for 20 minutes, washed once with PBS, added with Hoechst 33342 (final concentration 10 ⁇ g/ml), stained at 37 °C 5- 10 minutes. The dye solution was removed by centrifugation, and the droppings were observed under an Olympus fluorescence microscope and photographed.
  • 3 ⁇ 4 nuclear morphological changes are the most typical features of apoptotic cells, and are also the basic parameters for determining apoptotic cells.
  • the main morphological changes of apoptotic cells include nucleus pyknosis, extensive cleavage of chromosomal DNA, and fragmentation along the nuclear membrane into a polymorphic high-density particle region; the nuclear membrane shrinks and the fragment is wrapped in cytoplasm after disintegration. The cells are fully collapsed, and the cell membrane shrinks and contracts, but the cell membrane envelops the organelle or nuclear fragment to form apoptotic bodies.
  • Hoechst 33342 is a lipophilic dye that stains DNA across cells into cells.
  • HEC Human renal tubular epithelial cells in the exponential growth phase were digested with appropriate amount of 0.25% trypsin solution containing 0.02% EDTA to cause adherent cells to fall off.
  • the most important biochemical marker of apoptosis is the activation of endogenous calcium-magnesium-dependent endonuclease, which selectively degrades DNA between chromosomal bodies, resulting in extensive cleavage of cellular DNA, formation of DNA fragments of varying sizes, and even Monomeric or oligonuclear nucleus, thus presenting a regular "DNA ladder"-like strip at 180-200 bp apart on agarose gel electrophoresis.
  • cisplatin was used as a control drug, and cisplatin was applied to HKC cells for 36 hours with different concentrations of compound 31 (2-50 u mol/L) of the present invention, and subjected to agarose gel electrophoresis.
  • the increase in the cisplatin-induced "DNA ladder” spline is gradually reduced in a dose-dependent manner.
  • the compound 31 of the present invention has a significant inhibitory effect on the apoptosis of HKC induced by 10 ⁇ mol/L cisplatin. (See Figure 4)
  • Method - Take rat liver microsomes (protein content is about 15mg/ml) 0. 1ml; add different concentrations of drugs and reagents (1) different drug concentrations ⁇ ⁇ ⁇ ; (2) lmmol / L FeS0 4 50 ⁇ 1; (3) 10 mmol/L L-cysteine 20 ⁇ l; (4) PBS (pH 7.4) 0.82 ml. The total is 1ml.
  • the reaction was carried out for 30 min at 37 ° C; the reaction was terminated by adding 20% TCA 0.3 ml; centrifugation at 2000 rpm for 15 min; the supernatant was taken up with 1.0 ml of 0. 67% thiobarbituric acid (TBA) 0. 6 ml, boiling water was heated for 10 min. After cooling, the OD value was measured at 532 nm, and the inhibition rate was calculated.
  • Inventive compound 31 has a certain inhibitory effect on Fe 2+ -L-cysteine-induced liver microsomal lipid peroxidation in vitro, and reaches a maximum inhibitory concentration at 10 ug/ml, and the inhibition rate is 32.3%.
  • the effect is weaker than the phenolic hydroxyl compound S-3-l. (See Table 25, Figure 5)
  • kidney tissue homogenate (10 ml of homogenate per gram of kidney tissue) was taken, and each method was determined as follows. The lipid peroxidation level of the kidney tissue of the rats was compared, and the differences between the administration groups were compared.
  • the kidney tissue homogenate malondialdehyde (MDA, malondialdehyde) was determined as follows: Take 0.1ml homogenate, add 0.1ml 10% SDS, let stand for 20min at room temperature; add 2ml 0.1N HC1 and 1.0ml 1% TBA, mix, 100 ° C water bath, 40 min; after cooling, add 4 ml of n-butanol, shake for 3 to 5 min, extract, centrifuge at 3000 rpm for 10 min; take 0.2 ml of the upper n-butanol solution and add to 96-well plate, and measure the OD value at 532 nm with a microplate reader; Standard curve: Tetraethoxypropane (TEP) 0, 20, 40, 60, 80, 100 ⁇ mol L is used as a standard.
  • TBA Tetraethoxypropane
  • the lipid peroxidation level of renal tissue was significantly higher than that of the control group (P ⁇ 0.05) after a single injection of 6 mg/kg cisplatin in the peritoneal cavity of the rats.
  • the level of peroxidation decreased, and the lipid peroxidation level of cisplatin combined with compound 31 (30 mg/kg) in the kidney of rats decreased by 45.2% compared with cisplatin group (P ⁇ 0.05).
  • kidney tissue homogenate thiol (-SH) content determination is as follows:
  • T-SH Total GSH
  • tissue homogenate was added to 1.5 ml of 0.2 mol/L Tris buffer (pH 8.2), 0.1 ml of 0.01 mol/L DTNB, and 7.9 ml of absolute ethanol to make the total volume. 10 ml; reagent blanks are also prepared; standard curve: Take reduced glutathione (GSH) 0, 125, 250, 500, 1000 ⁇ mol / L as standard; the above color reaction 15min, centrifuged at 3000g for 15min at room temperature; 0.2 ml of the supernatant was added to a 96-well plate, and the OD value was measured at 410 nm using a microplate reader.
  • GSH reduced glutathione
  • Non-protein-bound GSH 2.5ml tissue homogenate was added with 2ml double distilled water, 0.5ml 50 TCA, continuously shaken for 10 ⁇ 15min, centrifuged at 3000g for 15min; reagent blank was also prepared; standard curve: same as above; Add 1.0ml of 0.4mol/L Tris (pH8.9), 0.1ml 0.01 mol/L DTOB to 1.0ml filtrate or supernatant, and shake well. Add 5ml of supernatant to 96-well plate within 5min after DTNB is added. , OD value was measured at 412 nm using a microplate reader.
  • PB-SH Protein-bound GSH
  • the total GSH (T-GSH) and protein-bound GSH (PB-GSH) levels in renal tissues were lower than those in the control group (P ⁇ 0.01), and non-protein binding type, 4 days after a single injection of 6 mg/kg cisplatin in the abdominal cavity of rats. There was no significant change in GSH (NB-GSH) levels.
  • Combination of cisplatin Benazapril group and cisplatin combined with the compound of the present invention The total GSH (T-GSH) and protein-bound GSH (PB-GSH) levels in the renal tissue of the 31 groups were also decreased, and there was no significant change compared with the cisplatin group. (See Table 27) Table 27. Effect of Compound 31 of the Invention on Glutathione Level in Kidney Tissue of Rats with Acute Renal Injury Induced by cisplatin (n-10)
  • Cisplatin + 31 (10mg/kg) 15.82 ⁇ 2.49
  • Cisplatin + 31 (30mg/kg) 15.24 ⁇ 2.85
  • Cisplatin model 2.96 ⁇ 0.25
  • each drug concentration is set to the control tube, and the following table is added:
  • HHL (lOmM) Drug or water IN HC1 ACE (80mU/ml)
  • each tube (experimental tube and control tube) was added with 1.5 ml of ethyl acetate and shaken to extract 1.0 ml of the upper layer of ethyl acetate; 120 ° C, 30 min volatile acetic acid
  • the ethyl ester was dissolved in 1 ml of water, and the absorption value was measured at 228 nm.
  • ACE 6x10 The inhibition rate of each dosing tube was calculated by using a water supply pipe as a control.
  • the compound 31 of the present invention has a weak inhibitory effect on angiotensin converting enzyme at a concentration of 1 (T 9 niol/L, ⁇ ⁇ /L in vitro) (Table 28).
  • Compound 31 of the present invention is angiotensin in vitro. Inhibition of invertase (ACE)
  • Balb/c 3T3 or NIH 3T3 cells were seeded into 96-well plates at 37 ° C, 5% CO 2 , DMEM medium (containing 10% fetal bovine serum). After 2-4 days of culture, when the cells are close to fusion, the culture medium is replaced with binding buffer (50 mmol/L HEPES containing NaCl, KC1, MgS0 4 and CaCl 2 ), and 100 pmol/L [ 125 I]TGF- ⁇ is added. 1 Excitation test, while adding the test compound. After 4 hours of cell culture, the medium was discarded and the cells were washed with ice-cold binding buffer. Non-specific binding of lOnmol/LTGF- ⁇ 1 was determined. The cells were dissolved in Triton X-100 buffer and the radioactivity was measured.
  • Standard curve preparation Set 8 wells of standard wells, add 100 ⁇ of sample dilution solution to each well, add 100 ⁇ of the standard to the first well, mix and aspirate 100 ⁇ 1 with the sampler, and move to the second well. This was repeatedly diluted to the seventh well, and finally, the ⁇ was aspirated from the seventh well, so that the volume was 100 ⁇ l.
  • the eighth hole was a blank control.
  • Cisplatin + 31 (30mg/kg) 5.52 ⁇ 1.84 61.4
  • Precipitation is the total R A required. After the supernatant is decanted, add 1.5 ml of 75 % ethanol, shake once and carefully pour off the ethanol. Centrifuge at 1.2000 rpm for a few seconds and blot the supernatant with a Tip. Then, 200 ⁇ l of DEPC-treated water was added to dissolve the precipitate, which was stored at -20 ° C until use.
  • RNA is estimated by RNA concentration.
  • Anti-sense primer 5' CCAAGG TAA CGC CAG GAA T
  • Anti-sense primer 5'CTT CCT TAA TGT CAC GCA CGA TTT C 3 '
  • Angiotensin II (ANGII) radioimmunoassay assay Rats were anesthetized with intraperitoneal injection of 35 mg/kg sodium pentobarbital. Blood was taken from the fundus venous plexus with a capillary glass tube and placed in an ice water bath to cool the enzyme inhibitor anticoagulant tube. Shake well and immediately place it in an ice water bath for cooling. Remove it when it is centrifuged. Centrifuge for 5 min at 4 °C lOOOO rpm and separate the blood paddle (can be stored at -20 °C for 2 months).
  • N2006/000839 takes the common logarithm of the standard concentration as the abscissa and the corresponding logit value as the ordinate as the standard curve.
  • the ANGII concentration of the sample to be tested can be obtained from the standard curve. result:
  • Substrate zymography analysis of the effect of compound 31 of the present invention on the ability of HT-1080 cells to secrete matrix metalloproteinases It was improved according to the method of Heussen et al.
  • the HT-1080 cells in the logarithmic growth phase were counted, and the cells were counted after digestion, and seeded in a 24-well culture plate at a density of IX 10 5 /well, and cultured overnight.
  • the medium containing a certain concentration of the drug and the control solvent was added to each well for the next day for 12 hours.
  • the culture supernatant was discarded, washed three times with PBS, and then cultured for 10 hours with a serum-free dosing medium of 300 ⁇ l.
  • the cell culture supernatant was collected, and the cell debris was removed by centrifugation at 200 g for 10 min at 4 ° C, and the supernatant was stored at -20 ° C for use, and the cells were digested and counted.
  • SDS-polyacrylamide gel electrophoresis is carried out in the literature. Prepare a volume of 8% separation gel and 5% concentrated gel. The separation gel contains 0.1% (w/v) gelatin. The volume of the culture supernatant corresponding to the same number of cells was calculated by the number of cells, and electrophoresis was carried out according to the volume (DTT was not contained in the loading buffer).
  • the gel was peeled off, rinsed with distilled water, and transferred to 100 ml of a 2.5% Triton X-100 solution, and shaken at a low speed on a shaker to elute the SDS. After 30 min, the new Triton X-100 solution was allowed to continue to elute for 30 min. The gel was transferred to 100 ml of gelatinase buffer (50 mmol/L Tris-HCl, pH 7.5, 10 mmol/L CaCl 2 , 200 mmol/L NaCl, ⁇ mol/L ZnCl 2 ), and incubated at 37 ° C for 16 hours.
  • gelatinase buffer 50 mmol/L Tris-HCl, pH 7.5, 10 mmol/L CaCl 2 , 200 mmol/L NaCl, ⁇ mol/L ZnCl 2
  • Type IV collagen is an important component of extracellular matrix. Gelatinase or type IV collagenase can degrade type IV collagen, which is one of the important factors affecting the degradation of type IV collagen.
  • SDS-polyacrylamide gel electrophoresis can be used to separate the gelatinase secreted by HT1080 cells into the culture supernatant and their active forms by molecular weight. After removal of the SDS by protein and protein binding by Triton X-100, gelatinase can restore its protein. Degradation activity. After the drug acts on the cells, if it affects the regulatory pathway of gelatinase expression and secretion, the content of various forms of gelatinase secreted into the culture supernatant will change.
  • gelatinase can degrade the gelatin around it under suitable reaction conditions.
  • the protein degradation zone cannot be stained by Coomassie blue, so in gelatinase A negative dye band can appear near the active area.
  • the more gelatinase secreted by tumor cells the greater the brightness and width of the band.
  • the effect of compound 31 of the present invention on the ability of HT-1080 cells to secrete matrix metalloproteinase was observed by this substrate zymography (Zymography).
  • the cells were changed to the rat mesangial cell (rMC) method and the substrate of the present invention was analyzed by substrate zymography.
  • rMC rat mesangial cell
  • mice Female SD rats, weighing 180-200 g, were injected intraperitoneally with 60 mg/kg streptozotocin (STZ) in addition to the normal control group, which caused diabetes in rats after 3 days.
  • the normal control group was intraperitoneally injected with 0.1 mol/L citrate buffer (pH 7.4).
  • 0.1 mol/L citrate buffer pH 7.4
  • the normal control group, the model control group, the losartan (10 mg/kg) positive drug group, and the compound of the present invention were administered in three dose groups of 5, 10, and 20 mg/kg.
  • Three days after modeling, the blood glucose levels were grouped and administered simultaneously. All were administered by gavage, 6 times a week, and continued for 20 weeks.
  • a 0.5% sodium carboxymethylcellulose (CMC) suspension was administered by gavage in the normal control group and the model control group.
  • CMC sodium carboxymethylcellulose
  • diabetic rats were injected subcutaneously with insulin 3 U/2 twice a week to maintain their body weight.
  • the rats were weighed weekly to observe the growth status of the rats. The following indexes were measured at 16 and 20 weeks after modeling. a. Anesthetized animals, blood was taken from the posterior venous plexus, and serum urea nitrogen (BUN), creatinine (Scr), and blood glucose were measured. Glu), and serum TGF- ⁇ levels;
  • the rats were placed in a metabolic cage to collect urine for 24 hours, and the amount of urinary albumin (1 Mb) and urine creatinine (Ucr) were measured, and the 24-hour urinary albumin amount and creatinine clearance rate (C.L) were calculated.
  • Serum TGF- ⁇ is detected by ELISA kit produced by Shanghai Senxiong Technology Industrial Co., Ltd.;
  • Specimens were fixed with 10% formaldehyde, embedded in paraffin, stained with HE and PASM, light microscopy, and histological photographs. Glomerular lesions: 30 small balls per animal count, no abnormalities; segmental sclerosis; diffuse sclerosis and balloon exudation classification (%). Renal tubular vacuolar degeneration with "one; +; ++; +++" indicates no abnormalities; mild, moderate, and severe lesions.
  • Diabetic rats were significantly lighter than normal control rats.
  • the blood glucose level of diabetic rats has been maintained at a high level, and the average blood glucose level is greater than 300 mg/dL.
  • the results are shown in Table 1.
  • the BUN level of the model group began to increase at 8 weeks after model establishment, and increased by 128.2% and 137.0% at 16 and 20 weeks after model establishment (both P ⁇ 0.01).
  • Losartan can alleviate the increase of BU in diabetic rats.
  • the levels of BUN decreased by 18.5% (P>0.05) and 31.4% (P ⁇ 0.05) at 16 and 20 weeks after treatment.
  • the three doses of Compound 56 of the present invention can alleviate the increase of BU in diabetic rats in different degrees, and the effect of 20 mg/kg treatment group is obvious.
  • the BU level at 16 weeks and 20 weeks after treatment is 27.0% lower than that of the model group ( ⁇ 0.05). 31.3% ( ⁇ 0 ⁇ 05).
  • the compound of the present invention 56 20 mg/kg can significantly inhibit the increase of TGF- ⁇ level in the blood of diabetic rats.
  • Table 38 Effect of Compound 56 on the Level of TGF- ⁇ in Blood of STZ-induced Diabetic Nephropathy Rats Administration Time, TGF-pi(ng/ml)

Description

香豆素衍生物及其制法和其药物组合物与用途
技术领域
本发明公开了新结构类型的香豆素及二氢喹啉酮衍生物及其药用盐、 这类化合物的制备 方法、 含有这类化合物的药物组合物, 特别是用于制备治疗慢性肾衰、 糖尿病、 髙血压和心 脑血管疾患以及肝硬化和前列腺肥大药物中的应用。 背景技术
在以前的研究中合成了一系列香豆素衍生物 (中国专利——徐世平等, 申请号:
02155525.7, 申请日: 2002年 12月 5日; 国际专利 (PCT)——国际申请号: PCT/CN03/01046, 国际申请日: 2003年 12月 5日, 优先权日: 2002年 12月 5日), 具有较好的抗肾衰和降压 作用, 对某些器官的纤维化有治疗作用。 基于上述结果我们在继续研究中证明, 本发明合成 的一系列新香豆素, 和一系列二氢喹啉酮衍生物。本发明化合物具有显著的抑制转化生长因 子 β ΐ (TGF P 1 )和降低血管紧张素 n(AngII)及肾素的作用, 因此本发明的化合物具有潜在 的治疗慢性肾衰、 高血压、 糖尿病、 肝硬化和前列腺肥大及肺纤维化的作用。
药理试验表明, 在我们研究的新一代的化合物中, 部分化合物对顺铂 (Cisplatin) 弓 |起 体外培养的大鼠肾系膜细胞、 人肾小管上皮细胞损伤具有保护作用; 在体内可显著地降低 Cisplatin所致急性大鼠肾损伤血清尿素氮(BI )和肌酐(Scr)水平及尿蛋白 (UP)水平, 减小 Cisplatin引起的肾水肿; 同时亦可降低 5/6肾切除模型大鼠血清 BUN水平、 Scr水平及 尿蛋白 (UP) 水平; 降低链脲霉素所致大鼠糖尿病肾病尿蛋白水平。 其肾保护、 降压以及 降低糖尿病肾病尿蛋白水平可能不同于血管紧张素转化酶抑制剂 (ACEI) 及血管紧张素 II ATI型受体拮抗剂( AT1RA)。而与抑制 TGF- β 1和降低肾组织 TGF- β 1 mRNA表达以及抑 制细胞分泌的基质金属蛋白酶活性有关。从而抑制肾小球硬化和肾间质纤维化。这些化合物 将可能用于缓解 Cisplatin肿瘤化疗的肾脏毒性;用于治疗由高血压和糖尿病所导致的慢性肾 功能不全; 预防和治疗各种原因引起的肾小球硬化和肾脏纤维化; 用于 II型糖尿病的治疗以 预防进一步糖尿病肾病的发生。
为了活性测定方面的便利, 在这里采用了, 以药物对顺铂所致的肾毒性的保护作用, 来 初步评价化合物的活性和疗效。
为了进一步评价化合物的疗效, 也釆用了 5/6肾切除模型大鼠, 作进一步研究, 以确定 本发明化合物的疗效。 发明内容
本发明要解决的技术问题是提供一类新的香豆素及二氢喹啉酮衍生物其异构体和其药用 本发明要解决的另一个技术问题是提供这类化合物的制备方法;
本发明要解决的又一个技术问题是提供一类新的药物组合物, 其含有本发明的化合物及 制药领域常用的载体。
本发明要解决的再一个技术问题是提供这类新的化合物在制备治疗慢性肾衰、 糖尿病、 高血压和心脑血管疾患以及肝硬化和前列腺肥大药物中的应用。 为解决本发明的技术问题, 本发明釆用如下的技术方案,
具体讲, 本发明的化合物如通式 (I)所示
Figure imgf000004_0001
(I)
其中:
X选自 0、 NH;
W选自 C0、 CH2;
R选自直链或支链的 -6烷基;
R6 、 R7 和 R8独立地选自 H、 0H、 N02、 羧基、 卤素、 .8烷基、 .8垸氧基; R3选自
取代或未取代的苯基 (苯环上的取代基为 Ru );
取代或未取代的苯基 .4烷基 (苯环上的取代基为 R12, 垸基上的取代基为 R" ); 取代或未取代的 N-吡咯基, (吡咯环上的取代基为 R13);
取代或未取代的咪唑基 (咪唑基上的取代基为 R15);
取代或未取代的吡唑基 (吡唑基上的取代基为 Rl6);
取代或未取代的 N-吲哚基 (吲哚基上的取代基为 R17);
并且, R" 选自 C1-4垸氧羰基;
R13表示单取代或多取代, 取代基独立的选自 H、 0H、 卤素、 N02、 C02H、 C1-8垸基、 .8卤代烷基、 C s羰烷基、 C 8烷氧基、 C L8羧基烷氧基、 d.8酰基、 d_8烷氧羰基、 d_8酯 基、 四氮唑基 , 未取代或取代的苯羰基、 苯环上的取代基为 R14 ;
Ru、 R12、 R14. R15, Ri6 和 R17均表示单取代或多取代, 取代基独立的选自 H、 0H、 卤素、 N02、 C02H、 d-8烷基、 -8卤代烷基、 d.8羰垸基、 C1-8烷氧基、 C 1-8 ;
酰基、 C 8烷氧羰基、 -8酯基、 四氮唑基。 优选的化合物包括但不限定于如通式 (IA) 所示
Figure imgf000004_0002
IA
其中, R-X选自 0 、 NH、 NCH3, NCH(CH3)2;
R6、 R7和 独立的选自 H、 OH 、 Cl、 N02、 C14烷基、 C1-4垸氧基;
Rn为单取代或多取代, 取代基独立的选自 H、 OH、 Cl、 N02、 C1-4烷基、 。1-4烷氧基、 CF3
OCH2C02H 、 甲酰基、 乙酰基、 C02H、 C02CH3、 C02Et、 (Γ,,2,,3,,,4,,-四唑 -5")。 优选的化合物包括但不限定于如通式 (IB ) 所示
Figure imgf000005_0001
IB
其中, R-X选自 O 、 應、 NCH3, NCH(CH3)2 ;
、 R7和 R8独立的选自 H、 OH 、 Cl、 N02、 CM烷基、 CM烷氧基;
R"选自 C02H、 C02CH3、 C02Et;
R12为单取代或多取代, 取代基独立的选自 H、 OH、 Cl、 N02、 CM烷基、 C^垸氧基、 CF3, OCH2C02H 、 甲酰基、 乙酰基、 C02H、 C02CH3、 C02Et、 (Γ,,2,,3",4,,-四唑 -5,,)。 优选的化合物包括但不限定于如通式 (IC) 所示
Figure imgf000005_0002
IC
其中, R-X选自 O 、 NH、 NCH3, NCH(CH3)2;
R6、 R7和 独立的选自 H、 OH、 Cl、 N02、 Cw烷基、 CM烷氧基;
R13为单取代或多取代, 取代基独立的选自 H、 OH、 Cl、 N02、 d.4烷基、 CM烷氧基、 CF3
OC¾C02H 、 甲酰基、 乙酰基、 C02H、 C02CH3、 C02Et、 (1",2"3,,,4,,-四唑 -5")、 取代的苯羰 更优选的化合物包括但不限定于如通式 (IC1 ) 所示
Figure imgf000005_0003
IC1
其中, R-X选自 0 、 NH、 NCH3, NCH(CH3)2
R6、 R7和 独立的选自 H、 OH 、 Cl、 N02、 CM烷基、 C1-4垸氧基;
R14为单取代或多取代, 取代基独立的选自 H、 OH、 Cl、 N02、 Cw烷基、 C1-4烷氧基、 CF3
OCH2C02H 、 甲酰基、 乙酰基、 C02H、 C02CH3、 C02Et。 优选的化合物包括但不限定于如通式 (ID) 所示
Figure imgf000006_0001
ID
其中, R-X选自 O 、 NH、 NCH3, NCH(CH3)2;
R6、 R7和 R8独立的选自 H、 OH 、 Cl、 N02、 CM烷基、 _4垸氧基;
R15为单取代或多取代, 取代基独立的选自 H、 OH、 Cl、 N02、 CM烷基、 CM垸氧基、 CF:
OCH2C02H 、 甲酰基、 乙酰基、 C02H、 C02CH3、 C02Et、 (Γ,2,,3,,,4,,-四唑 -5")。
优选的化合物包括但不限定于如通式 (IE) 所示
Figure imgf000006_0002
IE
其中, R-X选自 0 、 NH、 NCH3, NCH(CH3)2;
R6、 R7和 R8独立的选自 H、 OH 、 Cl、 N02、 CM烷基、 C1-4垸氧基;
R16为单取代或多取代, 取代基独立的选自 H、 OH、 Cl、 N02、 Q_4烷基、 CM烷氧基、 CF3
OCH2C02H 、 甲酰基、 乙酰基、 C02H、 C02CH3、 C02Et、 (Γ,,2"3",4,,-四唑 -5,,)。
优选的化合物包括但不限定于如通式 (IF) 所示
Figure imgf000006_0003
IF
其中, R-X选自 O 、 NH、 NCH3, NCH(CH3)2;
R6、 R7和 R8独立的选自 H、 OH 、 Cl、 N02、 CM垸基、 Q.4烷氧基;
R17为单取代或多取代, 取代基独立的选自 H、 OH、 Cl、 N02、 C1-4垸基、 C1-4垸氧基、 CF3
OCH2C02H 、 甲酰基、 乙酰基、 C02H、 C02CH3、 C02Et、 (1",2,,3,,,4"-四唑 -5")。
更优选的化合物包括但不限定于化合物选自下列化合物的群组之一
Figure imgf000007_0001
Figure imgf000008_0001
C8000/900ZN3/X3d
Figure imgf000009_0001
在本发明中, 术语 "卤素"是指氟、 氯、 溴、 碘。 根据本发明, 本发明化合物可以异构体 的形式存在, 而且通常所述的 "本发明化合物"包括该化合物的异构体。
本发明化合物可存在双键的顺反异构体, 不对称中心具有 S构型或 R构型, 本发明包括所 有可能的立体异构体以及两种或多种异构体的混合物。如果存在顺 /反异构体, 本发明涉及顺 式形式和反式形式以及这些形式的混合物, 如果需要单一异物体可根据常规方法分离或通过 立体选择合成制备。
根据本发明的实施方案, 所述的本发明化合物还包括其药效学上可接受的盐、 盐的水合 物、 酯或前体药物。 根据本发明还涉及制备本发明化合物的方法, 3-羧基的各种取代香豆素、 与相应的各种 取代氨类化合物反应制备。 酰氨化反应是在合适的反应剂、 催化剂及合适的溶剂条件下进行 的。 另外一类化合物的制备是, 用各种取代的苯胺类化合物与氯代乙酰氯进行反应, 得部分 本发明化合物的中间体。 以这些中间体与三氯氧憐及二甲基甲酰胺反应, 生成取代的喹啉, 然后与相应的各种取代吡咯化合物反应, 所得产物水解后得各种目的化合物。 这些化反应是 在合适的反应剂、 催化剂以及合适的溶剂条件下进行的。 这些反应剂包括三氯化磷、 三氯氧 磷、五氯化磷、二氯亚砜、草酰氯、乙酸酐以及硫酸二甲酯、碘甲烷、 1,3-二环己基亚胺 (DCC)、 二吡啶碳酸酯 (2-DPC)、 1,3-二异丙基碳酰亚胺 (DIPC)、 1-(3-二甲胺丙基) -3-乙基碳酰亚胺 (EDCI)等。 其中优选的反应剂为五氯化磷、 三氯氧磷、 Ν, Ν-二甲基甲酰胺、 三氯化磷和二氯 亚砜、 草酰氯, 更优选五氯化磷、 三氯氧磷、 二氯亚砜。 制备本发明化合物所使用的催化剂 包括三级胺、 吡啶、 4-二甲氨基吡啶和 4-吡咯垸基吡啶等。 其中优选为三级胺和吡啶。 更优 选为吡啶。 反应在适宜的溶剂中或上述缩合剂中进行, 如二氯甲垸、 1 , 2—二氯乙烷、 乙二 醇二甲醚、 四氢呋喃和 Ν,Ν-二甲基甲酰胺 (DMF)等。其中优选为甲苯、 DMSO和 DMF, 更优选 甲苯和 DMF。 反应温度为 10—110°C, 优选为 20— 90°C, 更优选为 30— 80°C, 特别优选为 50 — 70。C。 下列反应方程式具体说明
Figure imgf000009_0002
Figure imgf000010_0001
Figure imgf000010_0002
(lid)
Figure imgf000010_0003
(He)
Figure imgf000010_0004
(Hf)
Figure imgf000010_0005
dig)
Figure imgf000011_0001
(Ilh)
Figure imgf000011_0002
(Hi)
Figure imgf000011_0003
a=C orN: b=C orN
(Hj)
Figure imgf000011_0004
本发明因此还涉及含有作为活性成份的本发明化合物和常规药物赋形剂或辅剂的药物组 合物。 通常本发明药物组合物含有 0. 1-95重量%的本发明化合物。
本发明化合物的药物组合物可根据本领域公知的方法制备。 用于此目的时, 如果需要, 可将本发明化合物与一种或多种固体或液体药物赋形剂和 /或辅剂结合,制成可作为人药或兽 药使用的适当的施用形式或剂量形式。
本发明化合物或含有它的药物组合物可以单位剂量形式给药, 给药途径可为肠道或非肠 道, 如口服、 肌肉、 皮下、 鼻腔、 口腔粘膜、 皮肤、 腹膜或直肠等。
本发明化合物或含有它的药物组合物的给药途径可为注射给药。 注射包括静脉注射、 肌 肉注射、 皮下注射、 皮内注射和穴位注射等。
给药剂型可以是液体剂型、 固体剂型。 如液体剂型可以是真溶液类、 胶体类、 微粒剂型、 乳剂剂型、 混悬剂型。 其他剂型例如片剂、 胶囊、 滴丸、 气雾剂、 丸剂、 粉剂、 溶液剂、 混 悬剂、 乳剂、 颗粒剂、 栓剂、 冻千粉针剂等。 本发明化合物可以制成普通制剂、 也可以是缓释制剂、 控释制剂、 靶向制剂及各种微粒 给药系统。
为了将单位给药剂型制成片剂, 可以广泛使用本领域公知的各种载体。 关于载体的例子 是, 例如稀释剂与吸收剂, 如淀粉、 糊精、 硫酸钙、 乳糖、 甘露醇、 蔗糖、 氯化钠、 葡萄糖、 尿素、 碳酸钙、 白陶土、 微晶纤维素、 硅酸铝等; 湿润剂与粘合剂, 如水、 甘油、 聚乙二醇、 乙醇、 丙醇、 淀粉浆、 糊精、 糖浆、 蜂蜜、 葡萄糖溶液、 阿拉伯胶浆、 明胶浆、 羧甲基纤维 素钠、 紫胶、 甲基纤维素、 磷酸钾、 聚乙烯吡咯烷酮等; 崩解剂, 例如干燥淀粉、 海藻酸盐、 琼脂粉、 褐藻淀粉、 碳酸氢钠与枸橡酸、 碳酸钙、 聚氧乙烯山梨糖醇脂肪酸酯、 十二垸基磺 酸钠、 甲基纤维素、 乙基纤维素等; 崩解抑制剂, 例如蔗糖、 三硬脂酸甘油酯、 可可脂、 氢 化油等; 吸收促进剂, 例如季铵盐、 十二烷基硫酸钠等; 润滑剂, 例如滑石粉、 二氧化硅、 玉米淀粉、 硬脂酸盐、 硼酸、 液体石蜡、 聚乙二醇等。 还可以将片剂进一步制成 包衣片, 例 如糖包衣片、 薄膜包衣片、 肠溶包衣片, 或双层片和多层片。
例如为了将给药单元制成丸剂, 可以广泛使用本领域公知的各种载体。 关于载体的例子 是, 例如稀释剂与吸收剂, 如葡萄糖、 乳糖、 淀粉、 可可脂、 氢化植物油、 聚乙烯吡咯烷酮、
Gelucire, 高岭土、 滑石粉等; 粘合剂, 如阿拉伯胶、 黄蓍胶、 明胶、 乙醇、 蜂蜜、 液糖、 米糊或面糊等; 崩解剂, 如琼脂粉、 干燥淀粉、 海藻酸盐、 十二烷基磺酸钠、 甲基纤维素、 乙基纤维素等。
例如为了将给药单元制成胶囊, 将有效成分本发明化合物与上述的各种载体混合, 并将 由此得到的混合物置于硬的明胶胶囊或软胶囊中。也可将有效成分本发明化合物制成微囊剂, 混悬于水性介质中形成混悬剂, 亦可装入硬胶囊中或制成注射剂应用。
例如, 将本发明化合物制成注射用制剂, 如溶液剂、 混悬剂溶液剂、 乳剂、 冻干粉针剂, 这种制剂可以是含水或非水的, 可含一种和 /或多种药效学上可接受的载体、 稀释剂、 粘合 剂、 润滑剂、 防腐剂、 表面活性剂或分散剂。 如稀释剂可选自水、 乙醇、 聚乙二醇、 1 , 3- 丙二醇、 乙氧基化的异硬脂醇、 多氧化的异硬脂醇、 聚氧乙烯山梨醇脂肪酸酯等。 另外, 为 了制备等渗注射液, 可以向注射用制剂中添加适量的氯化钠、 葡萄糖或甘油, 此外, 还可以 添加常规的助溶剂、 缓冲剂、 pH调节剂等。 这些辅料是本领域常用的
此外, 如需要, 也可以向药物制剂中添加着色剂、 防腐剂、 香料、 矫味剂、 甜味剂或其 它材料。
为达到用药目的, 增强治疗效果, 本发明的药物或药物组合物可用任何公知的给药方法 给药。 用已知体内外试验方法测定本发明化合物和 /或组合物的活性和效果, 继续合成的香豆素 和二氢喹啉类化合物, 具有明显地肾保护作用, 本发明化合物对顺铂(Cisplatin)引起体外培 养的大鼠肾系膜细胞、 人肾小管上皮细胞损伤具有保护作用; 在体内可显著地降低 Cisplatin 所致急性大鼠肾损伤血清尿素氮(BUN)和肌酐(Scr)水平及尿蛋白(UP)水平,减小 Cisplatin 引起的肾水肿; 同时亦可降低 5/6肾切除模型大鼠血清 BU 水平、 Scr水平及尿蛋白 (UP) 水平; 降低链脲霉素所致大鼠糖尿病肾病尿蛋白水平。
初步作用机制研究表明, 本发明化合物 31其肾保护、 降压以及降低糖尿病肾病尿蛋白水 平不同于血管紧张素转化酶抑制剂 (ACEI) 及血管紧张素 ΠΑΤ1 型受体拮抗剂 (AT1RA)。 而与抑制 TGF- β 1和降低肾组织 TGF- β 1 mRNA表达以及抑制细胞分泌的基质金属蛋白酶活 性有关。 其主要途径可能归结为对 的抑制, 从而抑制肾小球硬化和肾间质纤维化。
本发明化合物将可能用于缓解 Cisplatin肿瘤化疗的肾脏毒性; 用于治疗由高血压和糖尿 病所导致的慢性肾功能不全; 预防和治疗各种原因引起的肾小球硬化和肾脏纤维化; 用于 π 型糖尿病的治疗以预防进一步糖尿病肾病的发生。
另外的研究结果表明, 本发明化合物给药 20周在 STZ诱发的雌性 SD大鼠糖尿病动物模 型, 可明显降低血尿素氮 (BUN) ,血肌酐 (Scr) 和血中 TGF-βΙ水平, 同时增加肌酐清除 率(C.L), 肾脏组织病理学结果表明, 本发明化合物 56对于 STZ诱发大鼠糖尿病肾病所致的 肾小球硬化、 肾小管空泡变性和肾间质病变均具有明显的治疗作用。 说明本发明化合物具有 明显延缓或治疗糖尿病肾病的作用。
总而言之, 本发明的化合物作可以用于制备转化生长因子- P l CTGF- β ΐ)抑制剂, 血管紧 张素 II (ΑΠ)转化酶受体拮抗剂, 可用于治疗心脑血管疾患、 肾脏疾患、 糖尿病、 肝硬化以 及前列腺肥大。 上述所述的心脑血管疾患是高血压、 心、 脑栓塞、 心肌梗塞、 脑中风。
本发明化合物药物组合物的给药剂量取决于许多因素, 例如所要预防或治疗疾病的性质 和严重程度, 患者或动物的性别、 年龄、 体重、 性格及个体反应, 给药途径、 给药次数、 治 疗目的, 因此本发明的治疗剂量可以有大范围的变化。 一般来讲, 本发明中药学成分的使用 剂量是本领域技术人员公知的。 可以根据本发明化合物组合物中最后的制剂中所含有的实际 药物数量, 加以适当的调整, 以达到其治疗有效量的要求, 完成本发明的预防或治疗目的。 本发明化合物的每天的合适剂量范围本发明的化合物的用量为 0.001— 150mg/Kg体重,优选为 0.1— 100mg/Kg体重, 更优选为 1一 60mg/Kg体重, 最优选为 5~45mg/Kg体重。 上述剂量可以 单一剂量形式或分成几个, 例如二、 三或四个剂量形式给药这受限于给药医生的临床经验以 及包括运用其它治疗手段的给药方案。
每一种治疗所需总剂量可分成多次或按一次剂量给药。 本发明的化合物或组合物可单独 服用, 或与其他治疗药物或对症药物合并使用并调整剂量。 附图说明 图 1本发明化合物 31对 cisplatin引起大鼠肾系膜细胞 (rMC) 损伤的保护作用。
图 2 本发明化合物 31对 cisplatin引起人肾小管上皮细胞 (HKC)损伤的保护作用
图 3 本发明化合物 31对人肾小管上皮细胞 (HKC)凋亡影响的形态学观察
A control
B cisplatin 2 mol/L
C cisplatin 2 μ mol/L+31 2 μ mol/L
D cisplatin 2 μ mol/L+31 10 μ mol/L
E cisplatin 2 μ mol/L+31 50 μ mol/L 图 4 本发明化合物 31对 cisplatin诱导的 HKC细胞染色体 DNA断裂的影响
1 control
2 cisplatin 10 μ mol/L +31 50 mol/L
3 cisplatin 10 μ mol/L +31 10 μ mol/L
4 cisplatin 10 μ mol/L +31 2 μ mol/L
5 cisplatin 10 μ mol/L
6 marker 图 5. 本发明化合物 31对 Fe2+-L-cys诱发肝微粒体脂质过氧化的影响 图 6. 本发明化合物 31对 cisplatin所致急性肾损伤大鼠肾组织 TGF- β! mR A表达的影响
Lane 1. Control
Lane 2. Cisplatin+31 lOmg/kg
Lane 3. Cisplatin+31 30mg/kg
Lane 4 .Cisplatin+ Benazapril lOmg/kg Lane 5. Cisplatin model
Lane 6. Marker 图 7. 本发明化合物 31对 cisplatin所致急性肾损伤大鼠肾组织 TGF- β! mRNA表达的影响统 计分析 图 .8. 本发明化合物 31对 HT-1080细胞分泌基质金属蛋白酶能力的影响
1 cisplatin 5 mol/L
2 cisplatin 5 μιηοΙ/L +31 3.12^moVL
3 cisplatin 5 mol/L +31 12.5μηιο1/ί
4 cisplatin 5μηιο1/ί +31 50 mol/L
5 control 图 9. 本发明化合物 31对大鼠肾系膜细胞 (rMC) 分泌基质金属蛋白酶能力的影响
1 control
2 cisplatin 5μηιο1 Τ,
3 31 3.12μηιο1/Ε
4 31 12.5μιηο1/ί
5 31 50 μπιοΙ/L 具体实施方式
下面的实施例用来进一步说明本发明, 但是这并不意味着对本发明的任何限制。
实施例:
实施例 1 3-[4,-(1",2"3",4"-四唑 -5")苯氨羰基 1-6-硝基 -7-羟基 -8-甲基香豆素 (31 ) 的制备 将 3-羧基 -6-硝基 -7-羟基 -8-甲基香豆素 10.6g(0.04Mol)与 30ml二氯亚砜反应完成后, 除 去多余的二氯亚砜, 加入 20ml二甲基甲酰胺 (DMF), 40ml吡啶和 7g(0.043Mol)4-氨基苯基 ( Γ,2,,3,,4,-四唑 -5 搅拌均匀, 在 50°C加热 4小时。 过滤, 用乙醇、 水洗涤、 干燥
1HNMR(300MHz,DMSO),5(ppm): 10.78(s,lH,NH),8.8(s,lH,NH), 8.6 (b,2H, 4,5-H),8.0(d,2H,J= 8.7Hz,Ar'H),7.9(d,2H,J=8.7Hz,Ar'H),2.23(s,3H,CH3)
实施例 2 3-[3'-(1",2"3",4"-四唑 -5")苯氨羰基】 -6-硝基 -7-羟基 -8-甲基香豆素 (32) 的制备 根据实施例化合物 31的制备方法, 化合物 32的制备, 不同点在于以 3-氨基苯基 (Γ,2', 3',4'-四唑 -5s) 进行反应得化合物 32
1HNMR(300MHz,DMSO),5(ppm): 10.70(s,lH,丽), 8.92(s,lH,5-H),8.67(s,lH, 4-H),8.4(s,lH,Ar2, -H),7.89(d,lH5J=7.8Hz,Ar6'-H),7.70(d,lH,J=7.8Hz, Ar4,-H), 7.59(t,lH,J=7.8Hz,Ar5'-H),2.27 (s,3H,CH3)
实施例 3 3-[3,-(l",2"3",4,,-四唑 -5")苯氨羰基】 -6-硝基 -7-羟基 -8-丁基香豆素 (33)
根据实施例化合物 31 的制备方法, 化合物 33的制备, 不同点在于以 3-羧基 -6-硝基 -7- 羟基 -8-丁基香豆素与 3-氨基苯基 (Γ,2',3',4'-四唑 -5!) 进行反应得化合物 33
1HNMR(300MHz5DMSO),6(ppm): 8.914(s,lH,4-H),8.708(s,lH,5-H),8.483n (s,lH,Ar-H-2,),7.833 ει
-乙-愛 7-9-¾¾-ε
Figure imgf000015_0001
(οε)峯 碁 聲 2-9- [奪 8 m^
(ε
HOO'HE's)乙 8'£'( O'H£'s)96'£XH-9'zH6=r'HZ'P)0 η 6=^1^)0 U r S' ' Y^liZ 'P)06 '(H-'S''£'zHt ·8=ί¾Κ'Ρ)00·8'(Η寸' ΗΓ8)08·8'(ΗΝΗΓ3)ε8·(Η:(κκίφ'(θ8ίΜα'ζΗ )0ε)Ή兩
Li
Figure imgf000015_0002
峯互暴愛 峯 彔
Figure imgf000015_0003
Figure imgf000015_0004
H6=JT'm'P)9i / (H-JV-S'zH8K'Hri)(H9'Z (H-S'zH8'乙 =f'HI'P)68 '<¾_ -'9' ^«)1乙8'//" 6' (H-JV-nn's)0/ '8 '(H-t7'HI's)888'8 '(HNtHrs)^8 i:(∞dd)g'(oSi aiZHM00£)"aiA[NHI
Figure imgf000015_0005
峯亙暴 ¾丄-
(9£) 峯 ¾暴¾丄 -8-¾¾ϋ- [¾^M*(«s-iii63-«fr'tte«z'«T)-t£]-£ 9\ Μ^
(¾ _"17'ZHSK'H£1)J6O '(¾0_"£ ¾¾'乙=1"'1¾ )½£'1'(¾ -' ¾¾'乙=1:¾^ 1^(¾:)-"1¾¾ =:¾3'&)09//^(¾ 0- £'8)/^6 'ε'(Η-9'ζΗΖ,·8=Γ'Ηΐ'Ρ)8ΐ3'Ζ,£(Η-$¾"8-Γ¾ΐ£Ρ)806'Ζ, '(H_JV_"S'"£'zH '8=f'iK'P)9W/ (H-JV-"9 '"Ζ'ζΗ 8=Γ'ίΚ'Ρ)Κ0·8 '(H 7'Hrs)068'8' (HN'Hl's) 188ΌΪ :(∞dd)§'(os a'zHW00£)¾0 IMHi
Figure imgf000015_0006
峯 暴 ¾丄- (se 峯 碁聲丄 -8-¾¾ώ- 奪繞籩 ¾("s-¾ffl-" £"z'"i W-£ s m^
(£HO-c^£zH3 =f¾e£l)0£6O£(¾D-£'zH2 ■L=[ 'HZ'b)£ n'(¾; "Z'ZHSK'HZ¾6Wr(¾:)-" zHr/;HZ £ 8'Z(H-JV-'S''£ZH;8=fHZ'P)0l76' H-JV-'9 Z' U =mZ^) \ S0-8'(H-S'HIS) £69'8 '(H寸 'H I £s)0 ΐ 6·8'(ΗΜ-'Ηΐ 's)/,KOl : (uid^g'CoSIAia'zHlAIOOe^IAINH, p£
Figure imgf000015_0007
峯亘暴愛丄 -8-¾¾ (f£ 峯萏暴 ¾丄-8-¾¾- ¾貤-9-【奪凝 t m^.
Figure imgf000015_0008
S =fiK SSS'r(¾ -'J' H:'H l)8 8' '(H-'S-JV'ZH8K' HI SI9' (H-'9' 'ZHSK'IK'«I)
6f8000/900ZN3/XDd L\9tl\ILmi OAV 焉 τ愛鲤 -9-耷篛 -ε -ί^'Μ ' m ^嗨导 w s m\ χε ^im Mn
(H S'ZH 6=fm'P) 6'9
'(H-t9¾K'6=i'HrP)0 ' /(H-8'zH6=jrm'P)I 2'Z/(H-t m's)£6r8'(H-/zH6=r'HrP)i£ 8'(H-S 'ΗΓ5)ζ60·6'(Η 'H s)g00'6XHN£Hlcs)/,i79 l HN¾rs)£90^i:(∞dd)'(oS]ia£zH 00£)"aHNHI
z 醒 in
(H-^'Z.-8=lr ,c=,rcHrPP)90-Z,'(H- 6 r^r'HrP)9S- (H-8czHe'6=f'HlP)6S-Z,e(H-t9ZH
Figure imgf000016_0001
焉二 ¾¾-9-¾¾-ε r士 ¾ '^ ε ' ^rnrn ιε ^mm^
(ε) 醒棚 n¾二-耷 -9- (聲^ ¾¾篛- n mm^
'(HJV - ' ZHS =f¾I¾W '(H-JV'S'c£'zHSK'iK 6£ '(H-8
Figure imgf000016_0002
¾ P)090'6'(H-l7'Hrs) LLY6 '(ΗΝΗ ΐ '^9f I ΐ '(ΗΝΗΐ'δ)^01 ' £ ϊ :(radd)'(oS a£zH OOe)¾PiNHj τ {^^ ^i n^ ∑;-醒
Figure imgf000016_0003
•9=r'HZ'P)l0-££(¾3O¾ecs)99-£'(HO£Hrtw)0 i(HJV'zHr8=r£H2'P)09-9£(HJV'zHr8-reH3<P)06 ·9'(Η-5'Η 8)ε6·/(Η寸' H s)£9'8'(HO'HI'jq)08'8'(HN'Hrjq)9 6:(radd)'(osiAia'z腦 00ε)ΉΗΝΗ
-L- -9- -i
Figure imgf000016_0004
(8£)峯亙暴聲 ώ-8-¾ -τ耷融 -9- (¾篛篱 2¾¾- 愛凝 -£
Figure imgf000016_0005
(¾D£S-/.=ri£ srii(¾DcZHS'A=r¾£b)8ST'(¾DO ¾£' ε6·ε'(Η- ' zH K'HI) ' (H-8'H s)8rA '(H-'9' 'ZHSK¾K 9£' '(H-'S' ¾¾9' =ί ΉΖ'ν)69·ί'(Η-ζ 'Hi's) LL-LXB- nV^m' (ΗΝ¾ΐ¾) ,9,0i:(∞dd)£(OSWa'zHII00£)¾]A[NHl
6e8000/900ZN3/X3d L19UI/L00Z O -醒擁11焉二¾篛-£ 士¾^^ ' ζι ιε ^ rn
Figure imgf000017_0001
¾η)80 Η-ν'9' ' zH//8=fHZ;'P)8£8'Z/(H-JV'S''£'zH;8= iK'P)96'乙' (Η ΖΗ8·Η:'ΗΙ'Ρ)9Ι0 ·8'(Η ' HI's)i766'8 ΗΜ£ΖΗ9'ε=ΓίΗϊ'Ρ)ε2ΐ7 ΐ ΗΜ¾Γ8)8ΐ ·εΐ:(^ θ8ΙΑ[α£ζΗΙΑΙ00ε)Ή^ΗΙ
-醒称攀霄二¾篛-£ ^ ¾ ^ 'w ^ '?^ Ι¾ u^ m
(ετ) z-m ^- ( ^ -^ " m^
(HJV'£"A=f Ηΐϊ) ΐ ΐ 'LXWYU
'∞)εε·Ζ/(ΙΡν'ζΗΐ7·8=Γ'Η Ρ)/^·Ζ;(ΗΡν 'ΖΗ8·乙 =Γ'Ηΐ'Ρ)乙 9·/ (Ην'ζΗΐ·8=ΐ:¾Κ'Ρ)ΐΖ/ ίΡν'ΖΗΓ8 ^['lll'V)66'LXU- cHl's)z,6-8 H ¾ s)nTl HMiHrs)829^i:(ra<I^ OS a'zHH00e)¾HNHI
(n) z-m m ~- ^m ^ 9i画
(Η-8£6=Γ'Ηΐ'Ρ)ΐ85" XWY- ' ^ mYZ^i'HVVV^L U-^H ^·8=Γ'Ηΐ£Ρ)Λ8·,'(Η·ΐν-^'ζΗ8Ί=Γ ¾ΓΡ).0·8£(Η-Ζί'ζΗ6=εΓ'ζΗΐ =ΙΓ¾ΐ'ΡΡ)9^·8'(Η-$¾ =Γ£ΗΙ
'Ρ)90·6
Figure imgf000017_0002
9 醒報拳
Figure imgf000017_0003
(H-8'ffV_'9
' ' m'^es'L-L&L u- 'n ^ws '(H-L '£-6=ζ['ίτ=ΗΉι'νν)ίν ' (u-iefz= 'p) 乙 ο·6
' ((H-17 Ήΐ 's) 6 - -9- -
Figure imgf000017_0004
(s) 醒糊 ¾:-¾ -9- 籙焉 ^("S-^H-"^"^^"^)-^]^ PI m^
Figure imgf000017_0005
·6=ΓΗΐ'ρ)ΐ9·,£(Η-^ν-¾ι52*'Η^υ)½'^ο·8'(Η-, ^β=Η^ί =ί' ννν)βγ \Ή.-^ηγζ=ί'ι Ρ)980·6'(Η寸 ¾I's)S0 6 '(HM'HI cs)££0X ΐ>(Η 'Ηΐ£δ) Ϊ91 ' £1 ^^^'(oSI^tfzHMOOE^l^NH,
6C8000/900ZN3/X3d L\9 ZllL00Z OAV (-¾0-£ΖΗ3· =¾£ζΗ3Ά=ΙΓ'Η2¾·2'(¾0-Μ'ζΗ2-εΐ=¾£ΖΗ2·εΐ=ΙΓ¾'ΐ') £0 £·ε'(Η-„ζΗ/8=Γ¾Ζ'Ρ 9'9'(¾ 9 ' ' ζΗΙ^^'ίΚ'Ρ^ΟΟ'ΖΛΗ-Ρ'ΖΗ /^Γ'ΖΗΓ ^Γ'ΗΓ ΖΤ乙
'(Η-^ζΗ6=ί'ΗΓρ)96ε·ζ Η-/ζΗ6·9=ζι: ^Υ2=Η^ι)ί£9 η- ^ι^=[^ν) ζ6 ϊΐ- ϊΐι^)
918·8'(-0-ΗΜ" 'ζΗΙ ς^'ζΗΙ^^Γ'ΗΓΟδό/^'θΗϋ ΤΗΐ'δ^^Π υ^φ'Ο^ α'ζΗ Οθε)爾腿 !
(6ε) 醒糊攀焉二- (聲 *¾¾- )-ε zz m^
Figure imgf000018_0001
Figure imgf000018_0002
' K 6S0'6'(H寸' m's)Zr6'(HN¾rs 68'II:(∞dd)'(oswa'zHI I00£)¾HMHi
-醒柳¾霄二 s篛 I '^ irn χε ^m m
(H) 醒糊舉焉 (愛瀠 IZim^
(tPV'ZH8'A=f ΗΪ W/(H
•tV'zH8'«=J:'HI'P)W /(HJV'ZHZ;8=flK'P)£S'Z (HJV'zHS'A=f'H )0Z;Z/(HJV'zH/8=J:'Hrp)I86 •/H-l HI^WS'H-l Hl^^'S HM'm's^S'irHN'Hrs^ radd osi^a'ZH OOSyai^Ni^
-醒糊¾焉二¾篛-£^1±¾^^ ' m 9i c £ m i^^ m
(9i) 醒糊¾¾二- ( 襟 oz m^
(Η-' V'zHZ/8=f 'Η
I'P)l786'9'(H-9'zHSK'HI £ 'Z/(H-8'zHr8=Jf'HI'P)S9l7'Z (H-A 'ΖΗ 乙 =f Η Ρ)689·/ (Η 9ν c LZ= f'zH//8=^ :'Ηΐ'ΡΡ)98/;/ Η-ς'ζΗΓ8=:'Ηΐ'Ρ)686·Α'(Η·ΐν-'ζ'ζΗ;Ζ=ΐ:'ΗΓΡ)9ε 8'(Η-ΐ7¾
-醒柳¾焉二¾篛-£ ^士5!^'^ i m si ' ^ m τε ^m
(so 醒糊攀^: r- (愛镊颈 ¾¾ - -¾篛- 6Ϊ m^
(£H3-3O'H£¾S0£ ¾DO¾ ¾82 H-9£zH2,Z,=3rzH8^=Ir'Hr S6£' ,'(H-8'zHr8=r
Figure imgf000018_0003
'HZ'V) 6L&L '(H-S'ztK 'H P lO'S'OH-i H s^^ OHN'HI's i^WraddytoS a'zHlAIOOS^IAQMHt
6C8000/900ZN3/X3d (¾0-3'ZHri7=f'H£ ½' (¾0-M'iK's)S8'£'(¾0
Figure imgf000019_0001
HI'PP)
(6X)
Figure imgf000019_0002
-醒棚暴蓄二愛篛 -ετ? ' ΟΪ ' ^ χε ^mm^m
(oi) 醒 #i學焉二- (聲羅 1ί¾¾¾ώ2Κ¾¾-4ε)-ε 9Z m^
Figure imgf000019_0003
m^LVLXn-L^m-L^ ^L^ m^mL-L '(H-s'zHrs^'Hi^oo's'CH^v'zHrn^Wb)
-醒称擎¾二 r^^wM i m " ' ^ m ιε ^ m^
i "醒釉華焉: r-[¾籙籩 sz m^
(HPV¾ /^ ε·/τεο·8'(Η- (8T) r- ¾ij ti^^-[¾^^^(«s-©Ba-«t7' £^KT)-c£]-£ z
(¾DO'H£'s)sio (H-9'zH6=i:£H P)8A£ '(H-'9'^'zH6=i'IK'P)£S6'/ (H-'S''£'zH6=r'HZ'P) SS0'8e(H-S'zH-6=f 'HrP)£ (H ' m's)686'8'(HN'm's)I08'6:(rodd)'(osi^a'z頭 Οί^ΉΜΝΙ^
cop 醒糊攀霄:耷鋤 -8-¾¾n-[聲籙籩 *("5-¾53-" "ε" '"ι) - 】 -ε ιτ m^
6e8000/900ZN3/X3d ZJ9 /■ΟΟΖ OAV 霄二
(sz)
Figure imgf000020_0001
聊 ¾
(¾DK'Hes) ΐ 8 · e'(H-9¾- c S'H- , e
'H-i3eH-8¾S<ra)l8,,-S6,-£(H--ie,6=zn7-3=Ir'H p p)0S-8e(H-S'zHA =r 'ΗΙ'Ρ)乙 0'6'(H- m's)t7r6' (顯 'Hl wiWradc 'oswa'zHMoo ^aHNi^
11 ^ ws mm^ 醒 貤
'ΗΪ'ΡΡ)乙 or乙
Figure imgf000020_0002
¾I'P)9乙;乙 '(κμν¾ε'∞)9Α8·乙 -εε6·ζ(Η-乙 ¾Κ·6
=Μ [czuVZ=^ f 'ΗΙ'ΡΡ)Ζες·8'(ΗΟ¾ Ρ 60'6'(HzO 'HI's)8Sr6:(mdd)'(osi^a'z丽 Οθε^ΗΙ^ΝΗ!
-9-聲篛 -ε-¾ώ-ΐ ^^ ^ m z 导 陣 ^ χε ^nm m
(oz) z-ffi#iiti¾ ¾¾-9-(¾^¾f¾¾¾ -¾¾-4ε)-ε-¾ώ-χ ιε m^
( -M'H£'s)908 · £'(H- ' S'H -' £'?H /8+f¾KS)9/9'(H-9'H- 'ZHZ;8=f'IK'P)£ /(H-8'zH9'6=f'H P)/^Z/(H-;ziK'6=z zHi^=ii:'H
Figure imgf000020_0003
ζ ^nws m ^ 腿糊擎霄二耷貤
Figure imgf000020_0004
ο£ m
(¾D-N'H£'s) 18· ε'(Η - 'ζΗ ΖΗ:¾ε ΐ ·Ζ/(Η- ^'Η-' ε i Y =ii) i-L BrSin- i V =iiYiZiV)iL-L £(H-8£zH£,6=r'P)/,8,Z.'(H-.eZH£-6=zf'zHZ.X=Ir'HI 'ΡΡ) ·8'(Η-ς'ζΗ/Ζ=Γ'ΗΓΡ)80·6'(Η寸' HI's rS'HN'Hrs mraddytosi^a'ZHI^OOSyaRNHj
zz ^^ ^ m^ ^鱷柳攀焉二
(¾D-M¾£'s) 18 ·
Figure imgf000020_0005
Η,τ=ΙΓ'Ηΐ'ΡΡ),/.· Η-8'ζΗε^=ΓίΗΐ'Ρ) 8·Α'(Η-,9'ζΗΖ<-;=Γ'Ηΐ'Ρ)9ζ·8£(Η-,'ζΗε·6=ζΓ'ζΗΐ7·3=ιΓ'Ηΐ
Figure imgf000020_0006
6£8000/900ZN3/X3d 1HNMR(300MHz,DMSO),(ppm):11.985(s,lH,NH),9.20(s,lH,4-H),9.129(s,lH, 5-H),8.56(d,lH,J= 9.3,7-H),7.94(m,5H,ArH)), 3.84(s,3H,NCH3)
实施例 34 1-异丙基 -3-〖4,-(1,,,2,,3,,,4"-四唑 -5")苯氨羰基] -6-硝基 -二氢喹啉酮 -2 (28)
根据实施例化合物 31的制备方法, 化合物 28的制备, 不同点在于以 1-异丙基 -3-羧基 -6 -硝基二氢喹啉酮 -2与 4-(Γ,2'3',4'-四唑 -5')苯氨进行反应得化合物 28
1HNMR(300MHz,DMSO)5(ppm):9.00(s,lH,5-H),8.89(d,lH,J=9.3Hz,7-H),8.47 (s,lH,4-H),8.07(d, 2H,J=7.2Hz,Ar'H),7.9(d,2H,J=7.2Hz,Ar'H),7.59 (d,lH, J=9.3, 8-H),5.55(m,lH, 异丙基 -CH),1.4 l(d,6H, 异丙基 -(C )2)
实施例 35 1-异丙基 -3-[3,-(1",2"3",4"-四唑 -5")苯氨羰基】 -6-硝基 -二氢喹啉酮 -2 (29)
根据实施例化合物 31的制备方法, 化合物 29的制备, 不同点在于以 1-异丙基 -3-羧基 -6 -硝基二氢喹啉酮 -2与 3-(1',2'3',4'-四唑 -5')苯氨进行反应得化合物 29
1HNMR(300MHz,DMSO),(ppm):9.00(d,lH,J=2.4Hz,5-H),8.80(m,lH,7-H),8.50 (s,lH,4-H),8.47(d, J=2.4,2'-H),7.96(m,lH,4'-H),7.79(m,2H,5'56'-H),7.60(m,lH,8-H),5.59(m,lH, 异丙基 -CH),1.40(d, 6H, 异丙基 -(C¾)2:>
化合物 7、 8、 9、 26、 和 27等按以上方法制备
实施例 36 3-[2,-(4,,-硝基苯羰基) -吡咯 -N-亚甲基] -二氢喹啉酮 -2 (41)
将 0.464克 (2.20mmol)2-氯 -3-氯甲基二氢喹啉置于 10ml圆底瓶中,加入 0.475克 (2.37mmo 1) 2-对硝苯羰基吡咯及 0.910克 (6.60mmol)无水碳酸钾和 4毫升 Ν,Ν-二甲基甲酰胺 (分子筛干 燥)于外浴 55°C反应, TLC监测 (石油醚: 乙酸乙酯 = 5: 1)5.5小时后反应完全。
后处理:过滤,滤饼用乙酸乙酯充分洗漆,后用水溶解,乙酸乙酯提取,合并有机相,饱和食盐水洗, 无水硫酸镁干燥,浓缩后用石油醚\乙酸乙酯重结晶的 0.397克产物, 2-氯 -3-[2'-(4"-硝基苯羰基) -吡咯 -N-亚甲基] -二氢喹啉,产率 52.24%, mp.l59.0〜159.9°C
1HNMR(300MHz,CDCL3),a(ppm): 8.292(d,2H,J=8.4Hz3,5ArH )8.032(d,lH,J=8.4Hz,5H) 7.896(d,2H,J=8. 7Hz,2,6-ArH),7.7447.478(m,4H),7.210(s,lH,QU4-H)6.862(d,lH,PY5-H)6.388(t,lH,PY3-H)5.879(s,2H,-CH2N)
ESI-MS m/z(Intensity)392.5(100,M+)
将 0.350克 (0.895mmol) 2-氯 -3-[2,-(4"-硝基苯羰基) -吡咯 -N-亚甲基] -二氢喹啉置于 10ml 圆底瓶中,加入 2ml醋酸和一滴水于外浴 120°C回流反应,原料在加热后全部溶解, TLC监测 (石 油醚: 乙酸乙酯 = 1 : 1, 2滴醋酸),二天后反应完全,反应液冷却后, 有灰色结晶析出。
后处理:过滤,用二氯甲烷充分洗涤灰色针晶, 干燥得 0.275克浅绿色针晶, ,产率 82.58%, mp. 275.0〜276.9°C
1HNMR(300MHz,DMSO),S(ppm): 8.301(d,2H,J=9Hz53,5ArH)57.921(d32H, J=9Hz,2,6rH),7.5637.
Figure imgf000022_0001
- -^d ¾11'£=1:¾1 )0 9'(11- - ^d'zH ΐ " £=Γ¾ ΐ ^O^XU-. '^ I · 8=Γ'Η 'Ρ)06·9'(Η-£ e^d'H S'L'^^ m^OVL-OVL '(Η-"9'"ζ'ζΗΓ8=Γ'ΕΚ'Ρ)0// (ζΗΐ; '^dg 'ειοαθ eZHIA[00£) WmrHx
19啄导 W ^AiF峯亘暴 ¾ώ¾_ε ¾)¾ϋ
(19) ¾® [¾ώ2Κ-Μ-¾ϋ¾ϋ -^] -£ Of um^
(¾0-M¾
6's)£S-S'(H- l7
Figure imgf000022_0002
9'£+1¾1 )0/;9'(11-£ '«1) - '(11-"5;
(Z9) 峯亙暴 鹙-" 6ε mm^
(¾0-McH3's) gg-g '(H-'fr- d
Figure imgf000022_0003
ΐ'Ρ)08·9 '(H"JV 'H- ¾'HJV'H0I'ra)09'A-0 A %≡- e ^)Z9^^d)%i\ja iz 00£)^ ml
09 ^ ^ m ^
Figure imgf000022_0004
( L)09Z '(001)851 XiV+n)96Z :(%) S -I3
°(¾D¾00 - Z'L=i
¾ε 'υ iLv\ '(¾ο ο- Ήζ '∞) ςιν '(¾ Ήζ 's) esz-ς ' -Η ' τ= '6'£=Η 'Η ΐ LZV9 ' -Η 'Ηΐ 'S) 998·9 'ζ^-Η 'ζ'\=ζ[ ΐ=ιί ¾ΐ 'ΡΡ) 196*9 '(ΖτΗ ί=Η e 'L= ι£ ¾ΐ 'υ L VL '(9_Η 'Γ8=Γ 'HI Ί) 2 ί '(^-H ¾l 'S) ZIZ'L '(8"Η c 'L=i 'Ηΐ 'Ρ) WL '(S-H 'Γ8=Γ 'Ηΐ 'Ρ) £6VL '(Η_Ν 'Ηΐ 'S) 9S6'll '(zHf '∞ddg '^ο θ 顯00£) ΉΗΜΉ!
(M-¾O¾Scs)Z,6i7-Sc(H-^Jd¾r«i)^£-9£(H-e^d¾Iira),08,9(H9<ra)060 6C8000/900ZN3/X3d 191^動0 OAV
/ OAVsi s〕/:d/zl>ioo9i 6
Figure imgf000023_0001
r)寸 XHsHSCTTHp)^9 X¾HsH)9<T 16169 XH4¾r)寸 X900HVHrp),H6rsZss-Z=-----.,.,. 根据实施例化合物 41的制备方法, 化合物 67的制备, 不同点在于以 2-乙氧羰基吡咯与 2-氯 -3-氯甲基二氢喹啉进行反应得化合物 67
1H-NMR (300MHz, CDC13, δρριη, JHz), 10.11 (S, 1H, N-H), 7.847 (S, 1H, H-4), 7.508- 7.687 (m, 4H, H-5, H-6, H-7, H-8), 7.334 (d, 1H, J=8.4, H-5 , 7.217 (d, 1H, J=6.9, H-4 , 5.676 (S, 2H, -CHa-), 4.476 (m, 2H, -OCH^CH3 ), 1.465 (t, 3H, J尸 7.2, J2=6.9, - OCH2C
EI-MS ( % ): 297(1^,100), 158(77)
实施例 47 3-(3,-三氟甲基 -4'-乙氧羰基吡唑 -N-亚甲基)二氢喹啉酮 -2 (69)
根据实施例化合物 41的制备方法, 化合物 69的制备, 不同点在于以 3-三氟甲基 -4-乙氧 羰基吡唑与 2-氯 -3-氯甲基二氢喹啉进行反应得化合物 69
1H-NMR (300MHz, CDC13, δρριη, JHz), 10.278 (S, 1H, N-H), 8.329 (S, 1H, H-5 , 7.889 (S, 1H, H-4), 7.635-7.552 (m, 2H, H-5, H-8), 7.26-7.376 (m, 2H, H-6, H-7), 5.36 (S, 2H, -CH2-), 4.298 (m, 2H, -OCH^CHb), 1.325 (t, 3H, ^=7.2, J2=6.9, - OCH2CH3
EI-MS ( % ): 365(M+,100), 158(77)
实施例 48 3-(2,-甲酰基吡咯 -N-亚甲基)二氢喹啉酮 -2 (64)
根据实施例化合物 41的制备方法, 化合物 64的制备, 不同点在于以 2-甲酰基-吡唑与 2 -氯 -3-氯甲基二氢喹啉进行反应得化合物 64
1H-NMR (300MHz, CDC13, δρρηι, JHz), 10.412 (S, 1H, N-H), 9.55 (S, 1H, -CHO), 7.74 1 (S, 1H, H-4), 7.563-7.493 (m, 2H, H-5, H-8), 7.343-7.237 (m, 2H, H-6, H-7), 7.224 (d, 1H, J=7.8, H-5 , 6.989 (d, 1H, J=3.9, H-4,, 6.287 ( S, 1H, H-3 。
ESI-MS ( % ): 253[M+1]+
实施例 49 3-(3,-正丙基 -5'-乙氧羰基吡唑 -N-亚甲基)二氢喹啉酮 -2 (70)
根据实施例化合物 41的制备方法, 化合物 70的制备, 不同点在于以 3-正丙基 -4-乙氧羰 基吡唑与 2-氯 -3-氯甲基二氢喹啉进行反应得化合物 70
1H- MR (300MHz, CDCI3, δρριη, JHz), 10.904 ( S, 1H, N-H), 7.563-7.480 (m, 2H, H-5, H-8), 7.340-7.219 (m, 3H, H-6, H-7, H-4), 6.706 (S, 1H, H-4,, 5.407 (S, 2H, - - ), 4. 421 (m, 2H, -OCH2CH3), 2.612 (t, 2H, J尸 6.9, J2=7.8, - CH2CH3 ), 1.684 (m, 2H, 圍 C H2CH¾CH3 ), 1.406 (t, 3H, Ji=7.8, J2=6.9, - OCH2CH3), 0.971 (t, 3H, Ji=6.9, J2=7.8, -CH2 C¾ )。
EI-MS ( % ): 339(M+,92), 158(100), 135(89) 实施例 50 3-(4,-乙氧羰基 -5'-甲基咪唑 -N-亚甲基)二氢喹啉酮 -2 (66)
根据实施例化合物 41的制备方法,化合物 66的制备, 不同点在于以 4-乙氧羰基 5'-甲基 咪唑与 2-氯 -3-氯甲基二氢喹啉进行反应得化合物 66
1H-NMR (300MHz, CDC13, Sppm, JHz), 8.022 (S, IH, H-2, 7.605-7.496 (m, 3H, H-4, H-
5, H-8), 7.260-7.147 (m, 2H, H-6, H-7), 5.643 (S, 2H, -CHs-), 4.407 (m, 2H, -OCHaCH ), 2.689 (S, 3H, -CH3), 1.397 (t, 3H, J=7.2 - OCH2CH
EI-MS (%): 311(^,48), 158(100), 265(80)
实施例 51 3-(2,-羧基吡咯 -N-亚甲基)二氢喹啉酮 -2 (72)
根据实施例化合物 41的制备方法, 化合物 72的制备, 不同点在于以 2-羧基吡咯与 2-氯 -3-氯甲基二氢喹啉进行反应得化合物 72
1H-NMR (300MHz, CDC13, δρριη, JHz), 11.947 (S, IH, -COOH), 7.498-7.427 (m, 2H, H- 5, H-6), 7.300 (d, IH, J=8.45 H-8), 7.226 (t, IH, J尸 2.1, J2=5.1, H-5), 7.109 (t, 1H, Ji= 7.5, J2=7.8, H-7), 6.920 (dd, IH, J尸 1.8, J2=2.1, H-4!), 6.851 (S, IH, H-4), 6.198 (t, IH, J!=3.9, J2=2.7, H-3 , 5.397 (S, 2H, -CH
EI-MS (%): 268(M+,23), 158(100), 224(70)
实施例 52 3-(2,-羧基咪唑 -N-亚甲基)二氢喹啉酮 -2 (68)
根据实施例化合物 41的制备方法, 化合物 68的制备, 不同点在于以 2-羧基咪唑与 2-氯 -3-氯甲基二氢喹啉进行反应得化合物 68
Ή-NMR (300MHz, CDC13, 5ppm, JHz), 11.973 (S, IH, -COOH), 7.755 (S, IH, H-4), 7. 604 (d, IH, J=5.7, H-5), 7.487 (t, IH, ^=7.8, J2=6.9, H-6), 7.305 (d, IH, J=8.1, H-8), 7. 226 (S, 1H, H-5 7.164 (t, IH, J尸 7.5, J2=7.8, H-7), 6.922 (S, IH, H-4 5.073 (S, 2H, -CHr)。
ESI-MS (%): 270[M+1]+
实施例 53 3-(4,-羧基 5'-甲基咪唑 -N-亚甲基)二氢喹啉酮 -2 (65)
根据实施例化合物 41的制备方法, 化合物 65的制备, 不同点在于以 4-羧基 5'-甲基咪唑 与 2-氯 -3-氯甲基二氢喹啉进行反应得化合物 65
1H-NMR (300MHz, CDC13, Sppm, JHz), 12.035 (S, IH, -COOH), 9.068 (S, IH, N-H), 7. 696 (S, IH, H-2), 7.643 (d, 1H, J=7.8, H-5), 7.519 (m, IH, H-6), 7.336 (d, IH, J=8.1, H-8), 7.189 (t, IH, J尸 7.8, J2=7.5, H-7), 5.483 (S, 2H, -CHa-), 2.501 (S, 3H, -CH EI-MS (%): 283(M+,10), 158(100), 239(65) (m-+
Figure imgf000026_0001
'S3 £) 9£: ¾isii啊) z/ui:s]A[-IS3
(N¾D-¾3's)080,Si(H-^d[£zHr2=r'Hlel)8l7g-9c(H-S^ <Ι£ΖΗΐ =Γ£Ηΐ¾630· (Η-9η0ίζΗ5·-=Γ¾ΐί 651·-(Η-8η 'ζΗ^·8=Γ£Ηΐ'Ρ)20ε·-(Η-Λα0<ΖΗΓ8=ΓίΗ
Figure imgf000026_0002
(9P) 醒糊攀 亚 -Ν[-¾ϋ¾ϋ (鞭 LS
(+W '93£ Ϊ)£·ε9£:( 啊) z/ui:siAi-IS3 (N¾0-¾2's)99t7- g(H-^d'zH£=r 'm'08I£'9(zH£=fHI'P) 6/;9(H-&^dm's)8W/ H-9n0'zHSK'Hn)90r8(H-8ni)'ZHr8=f'H rp)£6S-/,'(HScra)
Figure imgf000026_0003
(£8Z-+ '9^5)¥^1%+¥1 'SaO'S)9"6S£:(^suajui)z/ui:s]^-lsa (¾30- 'HE'S)/, I8 £(N¾D-'H6's)8Z,0,S(H-^d'zHI =f'H
(H-8n0'zHr8=f'H P) ^ (H-/l0'zHSK'HI¾l78l7'Z/(H-&^drHrs)£9 /(H-Sn0zH8 =f'H 1'P)919"Z,'(H-I7n0
Figure imgf000026_0004
(PP) Ζ-||#ΐϋι1 :-[¾ώ 2Ε-Ν-¾ιι¾ϋ(¾ώ -«^-4£]-£ SS ΜΜ^
+[ι+ηςί£ :(%) S -isa • (- ¾- ¾κ 's) ιςς·ς S-H 'HI 'S) 96Γ9 m 'HI 'S) 6 ·9
'ζε-Η 'Ηΐ 'S) 806'9 '(8'乙' 9'S-H 'Ht 9097τ96Γ '( -R 'HI 'S) ZOL'L '(9t_H_JV '8 'L=i ¾K 'P) 6 'L '(S'£-H-JV 'S'L=[ 'HZ 'Ρ) 0£·8 '(ζΗΓ '^ddg '^OQJ 'ΖΗΙΑΙΟθε) ΉΙΑΙΝ-Hj
£9啄 秦亙彔 ¾ώ¾-ε 聲^ 敏-' -z
Figure imgf000026_0005
6C8000/900ZN3/X3d ζΖ
6Ρ ^U ^im 華霄二 ¾»ώ
Figure imgf000027_0001
£9aO-9)S- -6Z:(^su9jui)z/ui:s^-lsa (¾DO -'H£'s)I6i^ (¾30-¾£'s)8S£'£(M¾0-¾K's)n£'S (H_
Figure imgf000027_0002
(¾000-'H£'s)I9£ (N¾D-'HZ:
:s)£9£'S(H-Z 9'S' ui'Hi n)乙 60·乙〜 0 £·乙 (Η^Γώ'ΖΗΓδ^'ΗΓ^^'ζ Η-δ^ηό'ΖΗΓδ:!:^!^^ , H-ZP¾¾I cs)e09'/,(H-m0'HI¾06r8(H-l7n iHl<s),gi7-8
Figure imgf000027_0003
(6S) 腿拗攀 ^二 (奪 ¾-Ν-¾|&ι¾2 ε)-ε 09 m^
(+η
Figure imgf000027_0004
(¾ O0-'H£'s) 9£'£'(M¾ -'H∑:'s)U£'S(H-t^<I'ZH6'£
Figure imgf000027_0005
S 导 暴霄二 -¾ώϋ-ε-鹙 -3
(M¾3- cH^s)08l7-s(H-^d'zH£=r¾r 0ie'9(H-£^<l'zHZ't7=feHlip)S9Z,,9(H-l7n0iHlcs)8£0^(zHr8=fiH l)90YL(ULCm) ϊ83'Ζ,~86$· ,'(Η-8£$η0£ζΗ6·9=Γ£Ηζ'Ρ)δΙ ,·8 : (∞tid)9<(oS^a£zHIAI00£)¾DA[NH1
6f8000/900ZN3/X3d Ll9tll/L00Z OAV 1HNMR(300MHz!DMSO),6(ppm): 8.303(d,2H,J=9Hz,3,5-ArH),7.919(d,2H,J=9Hz, 2,6-ArH),7.52
0(s,lH,Pyr5-H)7.483(d,lH3J=8.7Hz,QU5-H)7.119(s,lH,QU4-H), 6.795(d,lH,J=2.7Hz,QU8-H),6.7
73(s,lH,Pyr3-H),6.739(dd,lH,J邻 =8.7Hz,J间 =2.7Hz,QU6-H)6.320(dd,lH,J邻 =4.2Hz,J问 =2.7Hz,Pyr4
-H) 5.452(s,2H,-CH2N) 3.771(s,3H,-OC¾)
ESI-MS:m/z(Intensity):404.6(5.6E5, M^l 9.4(1.3E5,)
实施例 63 3-(2,-乙酰吡咯 -N-亚甲基 )-7甲氧基二氢喹啉酮 -2 (51)
根据实施例化合物 41的制备方法, 化合物 51的制备, 不同点在于以 2-乙酰吡咯与 3-氯 甲基 -7甲氧基二氢喹啉酮 -2进行反应得化合物 51
1HNMR(300MHz5DMSO),6(ppm): 8.409(d,lH,J=8.7Hz,QU5-H)7.273(t,lH, J=1.8Hz,Pyr4-H)7.17 4(dd,lH,J 邻 =3.6Hz,J fm=1.5Hz,Pyr5-H)6.885(s,lH,QU4-H) 6.792(d,lH5J=2.7Hz,QU8-H)6.726(dd,J 邻 =8.7Hz,J间 =2.1Hz,QU6-H)6.219 (dd, 1H,J邻 =2.1Hz,J间 =3.3HzPyr3-H)5.330(s,2H,-CH2N)3.771(s, 3H,-OCH3) 2.341 (s,3H,-COCH3)
ESI-MS:m/z(Intensity):297.5(1.8E6, M+)
实施例 64 3-【3,-(4,,-甲氧苯羰基)吡咯 -N-亚甲基】-7-甲氧基 -二氢喹啉酮 -2 (53)
根据实施例化合物 41的制备方法,化合物 53的制备,不同点在于以 3-(4'-甲氧基苯羰基) 吡咯与 2-氯 -3-氯甲基 -7-甲氧基二氢喹啉进行反应得化合物 53
1HNMR(300MHz,DMSO),5(ppm): 8.763(d,2H,J=8.7Hz,2,6-ArH),7.646(s,lH, QU4-H)7.555〜7.5 23(m,2H),7.024 (d,2H,J=9Hz,3.,5-ArH) ,6.980(t,lH,Pyr5-H) 6.7996.765(m,2H,QU6,8-H),6.502(t, lH,Pyr4-H)5.021(s,2H,-CH2N)3.819(s,3H,QU-OCH3)3.782(s,3H,Ar-OCH3)
EI-MS:m/z(100%):388.5(M+)
实施例 65 3-[3,-(4"-氯苯羰基)吡咯 -N-亚甲基】-7甲氧基 -二氢喹啉酮 -2 (57)
根据实施例化合物 41的制备方法, 化合物 57的制备, 不同点在于以 3-(4'-氯苯羰基)吡 咯与 2-氯 -3-氯甲基 -7-甲氧基二氢喹啉进行反应得化合物 57
1HNMR(300MHz,DMSO),5(ppm): 8.752(d,2H,J=8.1Hz2,6-ArH),7.640 (s,lH, QU4-H),7.569〜7.
522(m,4H),7.003(t,lH,J=2.1Hz,Pyr5-H)6.797〜6.773 (m,2H),6.527(t,lH,J=2.1Hz,Pyr4-H)5.023(s,
2H,-CH2N)3.782(s,3H,-OCH3)
£1^8(%):392(27,^),188.2(100, M+-204)
化合物 42和 43按以上方法制备 药理实验: 实验例 1 本发明化合物对 cisplatin所致小鼠急性肾损伤的保护作用 方法
取雄性昆明 (KM)小鼠, 16g〜22g, 按体重随机分为溶剂对照组和顺铂模型组、 给药组, 每组 8只。 对照组腹腔注射生理盐水, 顺铂以生理盐水溶解, 腹腔注射, 按 7mg/kg。 以上各给药 体积均为 0.4ml/20g, 于注射顺铂前 2天开始给药, 注射顺铂后第 3天、 5天、 7天分别眼球 取血, 用试剂盒检测血清 BU 、 Scr, 并称体重。
结果见表 1、 2、 3、 4、 5、 6和 7。
表 1化合物对顺铂造成小鼠肾损伤的保护作用 (造模后 3天)
浓度 体重 (g) Scr BUN WBC 样 品
(mg/kg) 开始 结束 (mg dL) 降低 (%) (mg/dL) 降低 (%) (xl03/mm3) 阴性对照组 22.7 ±1.2 28·7±2.7 1.59±0.17 28.11 ±5.24 7.1 ±0.52 顺铂 7 21.3±1.0 18·4±2.4 2.55±1.06 60.56 t 32.94±31.53 17.19† 3.0± 1.25
Losartan 25 21.5±1.9 17.8±2.0 2.13+0.68 16.37 49.23 ±37.99 - 2.8 ±0.92
50 21.2±1.7 18.8 ±1.3 1.93 +0.40 24.56 30.93 ± 19.40 6.10 5.1 ±1.33 秦皮乙酸乙酯 50 21.2+1.2 18.5±2.1 2.31 ±0.93 9.65 35.16+ 18.63 - ND
100 20.3 ±1.6 17.8± 1·7 2.52±0.51 1.40 28.17+6.43 14.47 D 秦皮正丁醇 50 22.0±1.1 17·8±2.5 2.70±0.82 - 37.40±22.56 - ND
100 21.0+1.3 18.2±1.2 2.34+0.99 8.33 26.2$ ±12.42 20.31 ND
25 25 20.5 ±1.8 19.5±1.4 2.15±0.57 15.79 54.58±33.43 - D
50 21·7±2.6 18.5±2.7 2.79 ±0.55 - 24.55+9.79 25.47 ND
26 25 21.5±1.5 18·8±1.7 2.25+0.59 11.69 21.98+9.45 24.16 ND
50 21.7±0.5 18·7±1·8 2.57+0.62 - 53.66+45.02 , ' D
27 25 21.5±1.0 18·8±0.4 2.35 ±0.40 7.96 21.21 ±4.04 35.61 4.2 ±2.05
50 20·8± 1·5 19.3 ±0.8 2.51 ±0.82 1.49 18.13±3.44 44.95 3.6±0.77
32 25 20.7±2·0 18.5±1.4 2.21 ±0.67 13.24 22.64±4.86 31.28 4.1 ±1.18
50 20.8 ±1.2 19.3 ±2.4 2.33+0.48 8.58 21.94±8.00 33.39 4.9 ±0.61
31 25 21·0±1.7 19.2±1.6 1.86±0.31 27.23 16.57±5.31 49.69 4.4±1.34
50 21.5±1.0 19.0±1.7 1.98 ±0.62 22.38 16.35+2.73 50.36 5.0 ±1.47
表 2化合物对顺铂造成小鼠肾损伤的保护作用 (造模后 5天)
浓度 体重 (g) Scr BUN 样 品
(mg/kg) 开始 结束 (mg/dL) 降低 (%) (mg/dL) 降低 (%) 阴性对照组 22.7 ± 1.2 28.7 ±1.9 1.71 ±0.49 35.15±6.63
顺 铂 7 21·3 ±1.0 17.3±1.2 2.48+0.75 44.94 ί 72.10± 13.63 105.08 t
Losartan 25 21.5± 1·9 18.7±4.2 2.37 ±1.05 4.65 36.29±2.30 49.67
50 21.2±1·7 18·4±3·0 2.21 ±0.67 11.05 46.70±9.01 35.22 秦皮乙酸乙酯 50 21.2±1·2 15·8±3.8 2.39 ±1.02 3.88 80.89±21.77 -
100 20.3 ± 1.6 19.0±3.6 2.39±0·84 3.88 39.45 ±11.73 45.28 秦皮正丁醇 50 22.0±1.1 16.8±3·8 2.86±0.45 - 33.92±0.44 52.96
100 21.0±1.3 16.2±2.2 2.22+0.04 10.47 75.46+42.28 -
25 25 20.5 ±1.8 15.7±0.6 2.54 ±0.74 - 95.77± 52.92 -
50 21.7±2.6 17.3 ±3.8 2.27+0.99 8.53 56.80±33.68 21.21
26 25 21.5±1.5 17.0±2.4 1.68+0.76 32.38 30.21 ±6.95 59.10
50 21.7 ±0.5 18.0±4.0 2.57+0.76 - 25.57±4.67* 64.54
27 25 21·5±1·0 16.8± 1.2 1.28±0·33 48.58 24.26 ±3.16* 66.35
50 20.8 ±1.5 18·2±2.0 1.45+0.76 41.76 31.60±6.12 56.17
32 25 20.7 ±2.0 16.0±2.0 0.98±1.13* 60.51 37.42±3.65 48.09
50 20.8 ±1.2 19.3±1.3 1·30±0·0.73 47.56 30.58±3.92* 57.58
31 25 21.0±1.7 17.0± 1.7 0.89+0.65* 64.21 22.47 ±2.04* 68.83
50 21.5土 1.0 18.6±3.0 1.85 + 1.13 25.34 20.31 ±2.77* 71.83
表 3化合物对顺铂造成小鼠肾损伤的保护作用 (造模后 3天) 浓度 Scr BUN 样 品
( s/ e) (mg/dL) 降低(%;) (mg/dL) 降低(%) 阴性对照组 1.68 ±0.29 30.52±2.52
顺铂 7 1·54±0·32 7.9 39.71 ±5.86 30.1 †
Losartan 25 1.38±0.35 10.3 15.17±2.86 61.8
50 1.45 ±0.49 5.9 16.80±8.28 57.7
XLF-III-43 12.5 1.54±0.84 - 17.17±3.44 56.8
25 1.76±0.62 - 17.91 ±3.46 54.9
31 12.5 1.67±0.80 - 17.55±2.70 55.8
25 1.42 ±0.32 7.9 19.79±4.56 50.2
32 12.5 2.20±0.25 - 26.38±4.50 33.6
25 1.92 ±0.48 - 21.02±5.37 47.1 表 4化合物对顺铂造成小鼠肾损伤的保护作用 (造模后 5天) 浓度 Scr BUN 样 品
(mg/kg) (mg/dL) 降低(%) (mg/dL) 降低(%) 阴性对照组 1·26±0.21 28.15±4.13 顺铂 7 1·13±0·15 10.4 32.02±5.65 13.7 t
Losartan 25 1.45 ±0.21 - 34.86± 10.08 50 1.42 ±0.38 42.68± 17.80
XLF-III-43 12.5 1.14±0·07 - Ι8.87±3·28 41.1
25 1.69 ±0.79 - 22.06±3.87 31.1
31 12.5 1.21 ±0.20 - 18.57±0.80 42.0
25 1.21 ±0.27 - 19.60±4.69 38.8
32 12.5 1.33 ±0.42 - 30.98±7.87 3.2
25 1.45 ±0.30 - 27.41 ±3.13 14.4 表 5化合物对顺铂造成小鼠肾损伤的保护作用 (造模后 7天) 浓度 Scr BUN 样 品
(mg/kg) (mg/dL) 降低 (%) (mg/dL) 降低(%) 阴性对照组 0·85 ±0.14 26.27± 5.55 顺铂 7 0.78±0.17 8.8| 47.44 ±7.38 80.6 t
Losartan 25 1.15±0.26 - 61.98± 17.95 -
50 1.17±0.30 - 40.50士 19.03 14.6
XLF-III-43 12.5 0.91 ±0.42 - 26.46 ±6.32 44.2
25 0.76±0·09 2.2 25.91 ±5.68 45.4
31 12.5 0.78±0.18 - 25.29±2.46 46.7
25 0.83 ±0.21 - 25.65±6.16 45.9
32 12.5 0·58±0.14 25.0 35.65 ±5.79 22.7
25 0.68±0.32 12.4 28.70+3.98 39.5
表 ό化合物对顺铂造成小鼠肾损伤的保护作用 (造模后 5天) 浓度 Scr BUN
样 品
(mg/kg) (mg/dL) 降低 (%) (mg/dL) 降低 (%) 阴性对照组 0.22±0·15 25.56± 0.77 顺铂 7 0· 15 ±0.08 - 28·32± 1 ·89 10.8 t
45 12.5 0.16±0·19 - 24.70± 8.28 12.78
25 0.21 ±0.19 - 26·77± 8.11 5.49
41 12.5 0· 19±0.13 - 51.43 ±24.77 -
25 0.29±0.10 - 26.32士 5.01 7.06
46 12.5 0.59 ±0.24 - 33.96± 7.91 -
25 1.15±0.51 - 25.12±3.93 11.31 06 000839
51 12.5 0·54±0.18 - 28.99士 6.18 -
25 0.38±0.11 - 26.91 ±7.08 4.96
58 12.5 0.69 ±0.14 - Π7.85 ± 53.96 -
25 0.56 ±0.20 - 96·71 ± 88.08 -
47 12.5 0.83 ±0.92 - 41.43 ±30.09 -
25 0.59±0.37 - 64.35 ±47.70 -
60 12.5 0.28 ±0.17 - 31.11 ±3. Π -
25 0.40 ±0.23 - 29.31 ±9.84 -
61 12.5 0.58 ±0.20 - 72.27 ±41.03 -
25 0.33 ±0.11 - 86.86 ±44.42 - 7化合物对顺铂造成小鼠肾损伤的保护作用 (造模后 7天) 浓度 Scr BUN 样 品
(mg/kg) (mg/dL) 降低(%) (mg/dL) 降低(%) 阴性对照组 0.53 ±0.18 24.39± 1.52 顺铂 7 0.40 ±0.24 - 48.61 ± 17.22##
45 12.5 0.33 ±0.11 17.68 46.39 ±20.96 4.57
25 .0.46 ±0.16 - 21.15 + 6.69** 56.49
41 12.5 - - - 一
25 - - - -
46 12.5 0.52±0.06 - 42.88±21.80 11.78
25 · 0.46±0.12 - 58.81 ±39.88 一
51 12.5 0.43 ±0.11 - 25.04±4.61 48.48
25 0.50+0.04 - 63.71 ±40.78
58 12.5 - - - -
25 0.44士 0.12 - 24.72 ±7.29* 49.14 47 12.5 0.22±0·12 44.07 42.48 ± 17.31 12.61
25 0.28+0.06 30.51 24.17±6.45* 50.27
60 12.5 0.20 ±0.05 49.15 26.78±5.92* 44.90
25 0.48 ±0.20 - 26.85±5.03* 44.76
61 12.5 0.45 ±0·21 - 16.70±4.43* 65.65
25 0.20+0.06 49.15 48.17±0.25 0.89
#: Ρ<0.05, 与对照相比; ##: Ρ<0.01, 与对照相比
*: Ρ<0.05, 与模型相比; **: Ρ<0.01, 与模型相比 实验例 2 本发明化合物 31在体外对 cisplathi所引起肾脏细胞损伤的保护作用
(一) MTT法观察本发明化合物 31与 cisplatin合用对大鼠肾系膜细胞 rMCs生长的影响 取指数生长期的大鼠肾系膜细胞 (rat mesangial cell, rMC), 加入适量含 0.02%EDTA的 0.25%胰蛋白酶液消化细胞, 使贴壁细胞脱落。 用 RPMI1640培养基 (含 20%小牛血清)制备成 浓度为 104/ml的细胞悬液, 于 96孔板中每孔接种 0.1ml。 将平板置于 37°C、 5%C02培养箱, 24小时后加不同浓度药物。 设对照组 (n=6), cisplatin 0.12、 0.37、 1.0、 3.0、 10.0、 30 μ mol/L 不同浓度组, 本发明化合物 31在 0、 33、 100 μ mol/L分别与顺铂上述不同浓度合用组, 每 组设 3个平行孔。
继续培养 72或 96小时, 弃去培养基, 每孔加入 0.1ml无血清 RPMI1640培养液配制的 MTT( 0.5mg/ml ), 37°C温育 4小时,活细胞可将 MTT还原为甲簪。弃上清液,加入 150 μ 1 DMSO 溶解甲簪, 于平板振荡器上充分振摇。用酶标仪以 450nm作参比波长, 于 570nm检测波长测 定吸光度 (OD)。
细胞生长抑制率%=1— (加药组细胞 OD值 /对照组细胞 OD值) X 100%
结果如下:
MTT 实验的原理是根据活细胞能将溴化四氮唑蓝 (MTT) 还原为一蓝紫色的可溶于 DMSO的甲簪化合物, 而死细胞则无此能力。 溶于 DMSO的甲簪在 570nm处有较强的吸收 峰,且此吸收值与活细胞数成较好的线性关系。用 MTT法观察了本发明化合物 31对 cisplatin 作用下大鼠肾系膜细胞的保护作用。
在 cisplatin浓度较低时,本发明化合物 31对于 cisplatin造成的 rMC毒性具有保护作用。 0.12 μ mol/L cisplatin对 对照组、 33 μ mol/L本发明化合物 31组、 100 μ mol/L本发明化合物 31组细胞生长的抑制率分别为 10.7%、 8.3%、 3.8%; 0.37 mol/L cisplatin对 对照组、 33 μ mol/L本发明化合物 31组、 100 μ mol/L本发明化合物 31组细胞生长的抑制率分别为 19.0%、 12.8%、 7.6%; 1 μ mol/L cisplatin对 对照组、 33 μ mol/L本发明化合物 31组、 100 μ mol/L本 发明化合物 31组细胞生长的抑制率分别为 25.4%、 18.2%、 12.0%。 本发明化合物 31对于 3 μ mol/L cisplatin^ 10 μ mol/L cisplatin以及 30 μ mol/L cisplatin造成的 rMC毒性未见明显的保 护作用。 (见表 8, 附图 1)
表 8. 本发明化合物 31对 cisplatin弓 1起大鼠肾系膜细胞 (rMC) 损伤的保护作用 Control 33u μ mol/L 100 μ mol/L
31 Cisplatin Inhibition Cisplatin Inhibition Cisplatin Inhibition
(umol/L) (%) (umol/L) (%) ( umol/L) (%)
0.12 10.7 0.12 8.3 0.12 3.8
0.37 19.0 0.37 12.8 0.37 7.6
1.0 25.4 1.0 18.2 1.0 12.0
3.0 41.4 3.0 31.8 3.0 35.9
10 63.7 10 59.7 10 64.5
30 89.3 30 89.3 30 90.1
(二) MTT法观察本发明化合物 31与 cisplatin合用对人肾小管上皮细胞 (HKC)生长的影响 取指数生长期的人肾小管上皮细胞 (HKC), 加入适量含 0.02%EDTA的 0.25%胰蛋白酶液 消化细胞, 使贴壁细胞脱落。 用 DMEM/F12培养基 (含 20%小牛血清)制备成浓度为 104/ml的 细胞悬液, 于 96孔板中每孔接种 0.1ml。 将平板置于 37°C、 5%C02培养箱, 24小时后加不 同浓度药物。 设对照组 (n=6), cisplatin 0.12、 0.5、 2.5 μ mol/L不同浓度组, 本发明化合物 31 在 0、 2、 10、 50 u mol/L分别与顺铂上述不同浓度合用组每组设 3个平行孔。
继续培养 72或 96小时, 弃去培养基, 每孔加入 0.1ml 1无血清 RPMI1640培养液配制的 MTT(0.5mg/m), 37°C温育 4小时,活细胞可将 MTT还原为甲簪。弃上清液,加入 150 μ 1 DMSO 溶解甲簪, 于平板振荡器上充分振摇。用酶标仪以 450nm作参比波长, 于 570nm检测波长测 定吸光度 (OD)。
细胞生长抑制率%=1 - (加药组细胞 OD值 /对照组细胞 OD值) X 100% 结果如下:
用 MTT法观察了本发明化合物 31对 cisplatin作用下大鼠 HKC细胞的保护作用本发明化 合物 31对于 cisplatin造成的 HKC毒性具有保护作用。 0.12 μ mol/L cisplatin对对照组、 2 μ mol/L本发明化合物 31组、 10 μ mol/L本发明化合物 31、 50 μ mol/L本发明化合物 31组细胞 生长的抑制率分别为 5.9%、 3.8%、 0.1%、 0.1%; 0.50 μ mol/L cisplatin对对照组、 2 μ mol/L 本发明化合物 31组、 10 μ mol/L本发明化合物 31、 50 μ mol/L本发明化合物 31组细胞生长的 抑制率分别为 11.1%、 5.5%、 3.5%、 6.7%; 2.5 μ mol/L cisplatin对对照组、 2 μ mol/L本发明 化合物 31组、 10 μ mol/L本发明化合物 31组、 50 μ mol/L本发明化合物 31组细胞生长的抑 制率分别为 78.0%、 69.6%、 47.7%、 43.1%。 (见表 9, 图 2)
表 9. 本发明化合物 31对 cisplatin引起人肾小管上皮细胞 (HKC)损伤的保护作用
Control 2 μ mol/L 10 μ mol/L 50 u mol/L
31
Cisplatin Inhibition Cisplatin Inhibition Cisplatin Inhibition Cisplatin Inhibition ( μ mol/L) (%) (umol L) (%) ( mol/L) (%) ( μ mol/L) (%)
0.12 5.9 0.12 . 3.8 0.12 0.1 0.12 0.1
0.5 11.1 0.5 5.5 0.5 3.5 0.5 6.7
2.5 78.0 2.5 69.6 2.5 47.7 2.5 43.1 实验例 3 本发明化合物 31对 Cisplatin.所致大鼠急性肾损伤的保护作用
(一) 检测方法
1. 血清生化指标- 血清尿素氮 (BUN)、 肌酐 (Scr) 釆用北京化工厂生产的临床诊断用试剂盒捡测。
2. 考马氏亮蓝 G— 250法测定尿蛋白含量
2.1 原理
考马氏亮蓝 G— 250在酸性溶液中呈棕红色(游离态),当它与蛋白通过疏水作用结合后, 变为蓝色, 最大吸收峰在 595nm。 以小牛血清白蛋白 (BSA)作为标准蛋白, 建立标准曲线。 根据标准曲线推测尿蛋白浓度。
2.2试剂配制
100^^ 0-250溶解于501^ 95%乙醇中, 加入 85% (w/v)的磷酸 100ml, 加水定容至 1L。 2.3 方法与操作步骤
标准曲线: 取标准蛋白 ( lmg/ml) 0、 5、 10、 20、 40、 60、 80 μ ΐ, 用双蒸水补至 ΙΟΟ μ Ι, 使得终浓度为 0、 50、 100、 200、 400、 600、 SOO g/mL 力卩 5ml G-250染液, 混勾, 室温 放置 15min, 在 595nm处测量 OD值。
测定: 取尿液样本 100 μ 1, 同上操作。
(二) 实验方法
50只雄性 Wistar大鼠按体重随机分为 5组, 每组 10只, 分别为溶剂对照组、 cisplatin模 型组、 cisplatin合用 Benazapril 10mg/kg组、 cisplatin合用本发明化合物 31 10mg/kg组、 cisplatin 合用本发明化合物 31 30mg/kg组。 溶剂对照组腹腔注射 10ml/kg 生理盐水; cisplatin模型组 腹腔注射 cisplatin 6mg/kg—次; cisplatin合用 benazepril 10mg/kg组于腹腔注射 cisplatin 6mg/kg 前 3天灌胃给予 benazepril 10mg/kg共计 3天, 以后每日灌胃给予 benazapril 10mg/kg一次, 共计 4日, 总计给药 7天; cisplatin合用本发明化合物 31 10mg/kg组及 cisplatin合用本发明 化合物 31 30mg/kg 组分别于腹腔注射 cisplatin 6mg/kg 前 3 天灌胃给予本发明化合物 31 10mg/kg、 30mg/kg共计 3天。 以后每日分别灌胃给药本发明化合物 31 10mg/kg、 30mg/kg— 次, 共计 4日, 总计给药 7天。 以上各给药体积均为 10ml/kg。
每日称体重, 末次给药后收集大鼠 24小时尿量, 用 G-250法检测 24小时尿蛋白量。 末 次给药后 24小时, 以戊巴比妥钠 30mg/kg麻醉大鼠, 于眼眶取血。 分别用试剂盒检测血清 BUN、 Scr、 TGF- β i和血浆 ANGII浓度。 ·
处死动物, 取肾组织, 称重, 计算肾脏器系数。 将 1/2左侧肾脏用磷酸缓冲液 (pH7.4)制 备 10%组织匀浆, 测其脂质过氧化及谷胱甘肽水平; 其余部分用 Trizol制备 10%组织匀浆, 提取总 RNA, 进行 RT—PCR, 分析肾组织 TGF- β! mRNA水平。 右侧肾用中性福尔马林固 定做病理检査。
(三) 检测指标
与对照组相比较, 观察模型组、 cisplatin合用 bennazapril组、 cisplatin合用本发明化合物 31 10mg/kg组、 cisplatin合用本发明化合物 31 30mg/kg组大鼠各组间血清生化及体重、 尿蛋 白、 脏器指数的变化, 并于给药后 4天处死大鼠, 取肾脏作病理检查。
(四) 实验结果
1. 本发明化合物 31对 cisplatin所致急性肾损伤大鼠血清生化 (BUN及 Scr)水平的影 响
给药后 7天, cisplatin模型组大鼠 BU 及 Scr水平较溶剂对照组明显升高,分别增加 747.3 %及 285.4% (P<0.01 ); cisplatin合用 bennazapril组、 cisplatin合用本发明化合物 31 10mg/kg 组、 cisplatin合用本发明化合物 31 30mg/kg组大鼠与 cisplatin模型组相比, BU 水平显著降 低, 分别降低 23.2% (P<0.01 )、 40.0% (PO.01 )及 46.9% (P<0.01 )。 Scr水平显著降低, 分 别降低 24.3% (P<0.05)、 32.3% (P<0.05) 及 35.8% (P<0.05 ) (见表 10) 表 10. 本发明化合物 31对 cisplatin所致急性肾损伤大鼠血清生化 (BUN及 Scr)水平的影响 (n=10)
Increase Inhibitory rate
Parameters Group X ±s . rate (%) (%)
BUN Control 27.98±4.01
(mg/dL) Cisplatin model 237.10±24.34 ## 747.3
Cisplatin +Benazapril 182.18±62.43*' 23.2
Cisplatin + 31(10mg/kg) 142·19±53.06*' 40.0
Cisplatin + 31(30mg/kg) 125.84±67.06" 46.9
Scr Control 1.14±0.56
(mg/dL) Cisplatin model 4.41 ±0.73 m 285.4
Cisplatin + Benazapril 3.33 ± 1.02' 24.3
Cisplatin + 31(10mg/kg) 2.96 ± 1.20' 32.3
Cisplatin + 31(30mg/kg) 2.83 ± 1·16' 35.8
##·· P<0.01 vs control, *: P<0.05 vs cisplatin model, **: P<0.01 vs cisplatin model
2. 本发明化合物 31对 cisplatin所致急性肾损伤大鼠尿蛋白 (UP) 水平的影响 给药后 7天, cisplatin模型组大鼠 24小时尿蛋白量较对照组明显升高 354.6% (P<0.01 ), cisplatin合用 bennazapril组、 cisplatin合用本发明化合物 3110mg/kg组、 cisplatin合用本发明 化合物 31 30mg/kg组大鼠与 cisplatin模型组相比, 24小时尿蛋白量明显下降,分别下降 59.8% (Ρ<0·05)、 48.8% (Ρ<0.05 )、 63.6% (Ρ<0,05)。 (见表 11 )
表 11. 本发明化合物 31对 cisplatin所致急性肾损伤大鼠尿蛋白 (UP) 水平的影响 (n=10)
Uric-protein Increase rate Inhibitory rate
Group
(mg/day, 士 s) (% ) (% )
Control 1.24±0·72
Cisplatin model 5.65±3.43 m 354.6
Cisplatin + Benazapril 2.27± 1.40# 59.8
Cisplatin + 31 (lOmg/kg) 2.89 ±0.60* 48.8
Cisplatin + 31 (30mg/kg) 2.06± 1.4 63.6
##: P<0.01 vs control, *: P<0.05 vs cisplatin model
3. 本发明化合物 31对 cisplatin所致急性肾损伤大鼠体重及肾脏指数的影响
给药后 7天, 大鼠体重明显降低, 肾组织肥大, 色暗红或灰白。 cisplatin合用 bennazapril 组、 cisplatin合用本发明化合物 31 10mg/kg组、 cisplatin合用本发明化合物 31 30mg/kg组大 鼠, 体重亦明显降低, 肾组织肥大较轻, 色鲜红或暗红。 三组体重减轻较小, 肾脏指数增加 较少。 其中 cisplatin合用本发明化合物 31 30mg/kg组大鼠肾脏指数与 cisplatin模型组大鼠相 比有统计学意义 (p<0.05) (见表 12)
表 12. 本发明化合物 31对 cisplatin所致急性肾损伤大鼠体重及肾脏指数的影响 (n=10)
Body weight (g) Kidney/body
Group - weight ratio
Initial Increased (mg/lOOg)
Control 254.50± 10.66 6.50±4.12 0/73 ±0.08
Cisplatin model 251.50±4.12 -41.67± 16.93## 0,93 ±0.08 Cisplatin + Benazapril 254·50±8.96 -27.14± 14.68 0,85 ±0·11
Cisplatin + 31(10mg/kg) 266·00±9.66 -33.50± 14.15 0.87±0.09
Cisplatin + 31(30mg/kg) 259.50±4.38 -27·00± 14.18 0·83 ±0.06*
##: Ρ<0.01 vs control, *: Ρ<0.05 vs cisplatin model
实验例 4 本发明化合物 31对大鼠 5/6肾切除所致慢性肾功能不全的影响
(一) 检测方法
血清生化检测 BU 、 Src试剂盒为北京北化精细化学品有限责任公司生产; 血浆 ANG II 放免试剂盒为北京北方生物技术研究所产品; TGF- β 1 ELISA kit为 Biotech产品上海森 雄科技实业有限公司分装。
尿蛋白测定 将大鼠置于代谢笼中收集 24 小时尿,采用考马氏亮蓝 G— 250法测定尿蛋白 含量。 以小牛血清白蛋白 (BSA) 作为标准蛋白, 建立标准曲线。 根据标准曲线推算尿蛋白 浓度,计算 24小时尿蛋白量 (UP ay—^
(二) 实验方法
参照 《新药临床前研究指导原则》 建立大鼠部分肾脏切除引起的慢性肾功能不全模型
1.模型建立
取体重 200g-240g雄性 Wistar大鼠, 腹腔注射戊巴比妥钠 35mg/kg, 待麻醉后, 手术摘 除右肾, 切除左肾上下极肾实质, 止血, 关闭腹腔, 缝合。
2. 分组给药
实验共设 6 组, 每组 10只鼠, 设假手术组、 模型组、 benazepril(4mg/kg)阳性对照组, losartan(10mg/kg)阳性对照组, 本发明化合物 31 10mg/kg、 30mg/kg两个剂量给药组。
手术 4周后, 检测大鼠血清尿素氮、 肌酐指标, 依据血清尿素氮、 肌酐水平随机分组, 并开始给药。本发明化合物 31为灌胃给药,每周 6次,持续给药到 16周(给药 12周); benazepril 和 losartaii, 均为灌胃给药每周 6次, 持续给药至 16周 (给药 12周)。 假手术组、 模型组灌 胃 0. 5%羧甲基纤维素钠 (CMC) 混悬液, 给予体积为 10ml .kg"1, 持续给予溶剂对照至 16周 (给药 12周)。
3. 检测指标
于术后 8、 12、 16周, 乙醚麻醉动物, 眼球后静脉丛取血, 检测血清 BUN、 Scr水平, 放免法测定血浆 ANG II水平, ELISA法测定血浆 TGF- β!水平, 将大鼠置于代谢笼中收集 24小时尿, 测尿蛋白量 (UP ay ) ; 每周称体重观察大鼠生长状况; 术后 16周 (给药 12 周), 除测定上述指标, 各组分别处死动物, 称量心脏重量, 计算心脏指数, 取肾脏做病理。
(三) 实验结果
1. 本发 ^化合物 31对 5/6肾切除模型大鼠 BUN水平的影响
结果见表 13。 与假手术组相比, 模型组大鼠 BU 水平在术后一直维持较髙水平, 在术后 4周升高 46.5%(P<0.01), 术后 8周升高 71.7%(P<0.01), 术后 12周升高 127·7%(Ρ<0.01), 术后 16 周升髙 84.2%(Ρ<0.01)。 各给药组术后 8周与模型组相比无明显差别。 术后 12周, losartan组与 模型组相比下降 26.8% , benazepril组 、 本发明化合物 31 10mg/kg、 30mg/kg两个剂量与模型 组相比无明显差别; 术后 16周 losartan组、 benazepril组、 本发明化合物 31 10mg/kg、 本发明化 合物 31 30mg/kg组与模型组相比分别下降 22.1% (P<0.01 )、 14.7%、 13·2%、 19.9%(Ρ<0·05)。 表 13.本发明化合物 31对 5/6肾切除模型大鼠 BUN水平的影响 Weeks after Increase Inhibitory
Group BUN (mg ·ά1-1)
operation rate (%) rate (%)
4 SM 31.13 ±2.73
Model 45.61 ±3.10## 46.5
Losartan 47.88± 3.89## 53.8
Benazepril 47.68 ±2.51## 53.2
31 lOmg/kg 45.99± 8.53## 47.7
31 30mg/kg 45.41 ±3.51## 45.8
8 SM 16.43 ± 1.68
Model 28.21 ± 1.58## 71.7
Losartan 29.81 ±6.59 -5. 7
Benazepril 27.93 ± 3.88 1. 0
31 lOmg/kg 32.12 ± 6.87 -13. 9
31 30mg/kg 27.72 ± 6.23 1. 7
12 SM 17.07± 2.44
Model 38.85 ± 4.41## 127.7
Losartan 28.44 ±2.93" 26.8
Benazepril 41.95 ± 5.24 -8.0
31 lOmg/kg 32.13 ± 14.27 17.3
31 30mg/kg 40.02± 7.74 -3.0
16 SM 25.34± 4.52 ·
Model 46.66±9.38## 84.2
Losartan 36.36±4.90" 22.1
Benazepril 39.78± 8.26 14.7
31 lOmg/kg 40.49± 6.38 13.2
31 30mg/kg 37.38± 6.67' 19.9
##: P<0.01 vs SM, *: P<0.05 vs model group, **: P<0.01 vs model group 2. 本发明化合物 31对 5/6肾切除模型大鼠 Scr水平的影响
结果见表 14。与假手术组相比, 模型组大鼠 Scr水平在术后 4周、术后 8周无明显变化, 术后 12周升高 104.0%(P<0.01), 术后 16周升高 68.2%(P<0.01)。 与模型组相比, 术后 12周 losartan组、 benazepril组、 本发明化合物 31 10mg/kg组、 本发明化合物 31 30mg/kg组与模型 组相比分别下降 13.2%、 15.5%、 7.3%、 39.0%(P<0.01)。 术后 16周 losartan组、 benazepril组、 本发明化合物 31 10mg/kg、 本发明化合物 31 30mg/kg 组与模型组相比分别下降 21.4% (P<0.05 )、 14.9%. 20.3% > 34.9%(Ρ<0·01)。
表 14.本发明化合物 31对 5/6肾切除模型大鼠 Scr水平的影响
Weeks Increase rate
Group Scr (mg ·άΓ') Inhibitory rate (%)
operation (%)
4 SM 1.35 ±0.38
Model 1.52土 0.25 13.0
Losartan 1.52±0.j7 12.9
Benazepril 1.52±0.21 13.2
31 lOmg/kg 1.47 + 0.25 9.3
31 30mg/kg 1.50± 0.16 11.6
8 SM 1.41 ±0.30
Model 1.76 ± 0.26 24.6 Losartan 1.80±0.49 -2.2
Benazepril 1.69 ±0.57 4.0
31 lOmg/kg 1·84±0·27 -4.7
3130mg/kg 1.55±0·30 11.9
SM 0.88±0.31
Model 1.79±0·62#* 104.0
Losartan 1.55±0·52 13.2
Benazepril 1.51 ±0.39 15.5
31 lOmg/kg 1.66±0.63 7.3
3130mg/kg 1.09±0.19" 39.0
SM 0.98±0.36
Model 1.65±0.27## 68.2
Losartan 1.29±0.37' 21.4
Benazepril 1.40 ±0.56 14.9
31 lOmg/kg 1.31 ±0.49 20.3
3130mg/kg 1.07±0.4( 34.9
##: P<0.01 vs SM, *: P<0.05 vs model group, **: P<0.01 vs model group
3. 本发明化合物 31对 5/6肾切除模型大鼠尿蛋白 (UP) 水平的影响
结果见表 15。 与假手术组相比, 模型组大鼠 UP*day-l水平明显升高, 术后 8周时升高 314.1%(P<0.01), 术后 12周升髙 280.3%(P<0.01), 术后 16周升高 816.9%(P<0.01)。 术后 8周 losartan组、 benazepril组、本发明化合物 3130mg/kg组与模型组相比分别下降 49.1%(P<0.05)、 48.2% (P<0.05)、 46.7% (P<0.05), 本发明化合物 31 10mg/kg组与模型组相比未见下降; 术后 12周 losartan组、 benazepril组、本发明化合物 3110mg/kg组、本发明化合物 3130mg/kg 组与模型组相比分别下降 27.3%(P<0.05)、 26.6%、 30.3%(P<0.05)> 36.6%(P<0.05); 术后 16 周 losartan组、 benazepril组、 本发明化合物 31 10mg/kg组、 本发明化合物 31 30mg/kg组与 模型组相比分别下降 61.0%(Ρ<0.01)、 51·4%(Ρ<0.05)、 40.1%(Ρ<0.05)、 72.3%(Ρ<0.01)ο
表 15.本发明化合物 31对 5/6肾切除模型大鼠尿蛋白 (UP)水平的影响
Weeks after -, TT1 , .κ τ , n.s Inhibitory rate
operation Gr0Up UP(mg.day ) Increase rate (%) (%)
SM 6·70±4·16.
Model 27.74 ± 11.88## 314.1
Losartan 14.12±6.94' 49.1
Benazepril 14.37±6.6 48.2
31 lOmg/kg 29.22 ±22.93 -5.3
3130mg/kg 14.78±3.72' 46.7
SM 14·56±3·91
Model 55.00±10.59## 280.3
Losartan 39.97± 18.76* 27.3
Benazepril 40.35± 19.16 26.6
31 lOmg/kg 38.33 ±12.70* 30.3
3130mg/kg 34.87± 10.25* 36.6
SM 11.49±5.12
Model 105.38±38.61'"' 816.9
Losartan 41.06+21.61" 61.0
Benazepril 51.19±32.68* 51.4
31 lOmg/kg 63.08± 19.44* 40.1
3130mg/kg 29.21 ±13.70*' 72.3 ##: PO.01 vs SM, *: P<0.05 vs model group, **: P<0.01 vs model group 4. 本发明化合物 31对 5/6肾切除模型大鼠体重的影响
结果见表 16。 术后 4周假手术组、 模型组、 losartan组、 benazepril组、 本发明化合物 31 10mg/kg组、本发明化合物 31 30mg/kg组体重未见明显差异。与假手术组相比, 术后 16周模 型组大鼠、 losartan组、 benazepril组体重下降约 35g(10%); 本发明化合物 31 10mg/kg、 本发 明化合物 31 30mg/kg组下降约 10g(2%)。本发明化合物 31 10mg/kg、本发明化合物 31 30mg/kg 组体重下降幅度较小。
表 16.本发明化合物 31对 5/6肾切除模型大鼠体重的影响
Weeks after operation Group Body weight(g)
SM 272.5 ± 6.9
Model 284.1 ± 16.5
Losartan 282.0± 18.1
Benazepril 284.3 ± 15.6
31 lOmg/kg 278.8 ±28.5
31 30mg/kg 271.3 ± 19.6
SM 389.0± 25.6
Model 352.7 ± 19.8#
Losartan 359.3 ± 33.8
Benazepril 353· 1 ± 48·8
31 l Omg/kg 378.3 ± 49.6
31 30mg/kg 382.5 ± 28·5*
#: P<0.05 vs SM , *: P<0.05 vs model group
5. 本发明化合物 31对 5/6肾切除模型大鼠心脏指数的影响
结果见表 17。 与假手术组相比, 术后 16 周模型组大鼠心脏指数升高 17.8%(P<0.01)。 losartan组、 benazepril组、 本发明化合物 31 10mg/kg、 本发明化合物 31 30mg/kg组与模型组 相比分别下降 18.0%(P<0.01)、 6.4% 2.4%、 13.7%(P<0.01)。 其中 losartan组、 本发明化合物 31 30mg/kg组心脏指数下降较为明显, 与假手术组相近。
表 17.本发明化合物 31对 5/6肾切除模型大鼠心脏指数的影响
Weeks after -, Heart /Body weight Index Increase rate Inhibitory
Group
operation r (g/lOOg Body weight) (%) rate (%)
16 SM 0.293 ± 0.015
Model 0.345 ± 0.030## 17.8
Losartan 0.283 ± 0.037" 18.0
Benazepril 0.323土 0細 6.4
31 lOmg/kg 0·337 ± 0.076 2.4
31 30mg/kg 0.298 ±0.032" 13.7
##'· P<0.01 vs SM, **: P<0.01 w model group 实验例 5 本发明化合物 31 对链脲霉素所致大鼠糖尿病肾病模型的影响 (一) 检测方法: 血清生化检测 采用荷兰威同 selecrta-E全自动生化分析仪测定
尿蛋白测定 将大鼠置于代谢笼中收集 24 小时尿液,采用考马氏亮蓝 G— 250法测定尿蛋 白含量。 以小牛血清白蛋白 (BSA) 作为标准蛋白, 建立标准曲线。 根据标准曲线推算尿蛋 白浓度,计算 24小时尿蛋白量 (UP ay^
(二) 实验方法:
1. 模型建立
取体重 200g-240g雄性 Wistar大鼠, 腹腔注射链脲霉素( streptozotocin STZ Sigma公司 产品) 60mg/kg (以 0.1mol/L柠檬酸缓冲液 (pH=7.4) 配成所需要的浓度)
2. 分组给药
实验共设 4组, 设对照组、 模型组、 losartan(10mg/kg)组, 本发明化合物 31 10mg/kg给药 组。 造模同时给药。 本发明化合物 31为灌胃给药, 每周 6次, 持续给药到 24周; losartan 为灌胃给药, 每周 6次, 持续给药至 24周。对照组、 模型组灌胃 0. 5%羧甲基纤维素钠(CMC) 混悬液, 给予体积为 10ml .kg", 持续给予至 24周。
3. 检测指标
于造模后 4、 8、 12、 16、 24周, 乙醚麻醉动物, 眼球后静脉丛取血, 检测血清 BUN、 Scr、 Glu水平, 放免法测定血浆 ANGII水平, 将大鼠置于代谢笼中收集 24小时尿, 测定尿 蛋白量 (UP a 1 ) ; 每周称体重观察大鼠生长状况; 术后 16周、 24周, 除测定上述指标, 各组分别处死动物, 取肾脏做病理。
(三) 实验结果
1. 造模后 4周 (见表 18)
与对照组大鼠相比, 模型组大鼠 Glu水平升高 473.7%; BUN水平升高 61.8%(P<0.05), Scr.水平升髙 18.1%(P<0.05), 说明糖尿病大鼠模型造模成功,且大鼠肾功能已出现损伤。各给 药组大鼠 Glu、 BUN Scr.亦升高, 肾功能未见明显改善。 本发明化合物 31组大鼠 ANGII水 平均较模型组低。
表 18.本发明化合物 31对链脲霉素所致大鼠糖尿病肾病模型的各项指标的影响(造模后 4周 n=6)
Group Glu BUN (mmol/L) Scr All
(mmol/L) (umol/L) (pg/ml)
Control 5.7±1.2 8.7±0.6 94.0±11.8 367.7±43
Model 32.7±1.8 † 473.7% 14±0.7# † 61.8% 111.0±8.5# † 18.1% 257.6±43.9
Losartan 26.4±4.9 11.6±1.4 106.0±15.8 308.1±79
31 27±6.3 12.9±2.6 107.8±1 1.9 191.6±35.4
#·· P<0.05, vs control group
2. 造模后 8周 (见表 19)
与对照组大鼠相比, 模型组大鼠 Glu 水平升高 409.9%(P<0.05) ; BUN 水平升高 65.9%(P<0.05), Scr.水平升髙 25.9%(P<0.05),各给药组大鼠 Glu、 Scr.亦升高, losarta 组、 本发明化合物 31组大鼠 BU 水平有所下降, 分别较模型组降低 25.8%及 17.0%。
表 19.本发明化合物 31对链脲霉素所致大鼠糖尿病肾病模型的各项指标的影响(造模后 8周 n=6)
Group Glu BUN (mmol/L) Scr
(mmol/L) (umol/L)
Control 6.5±0.5 8.8±0.4 80.1±7.2
Model 33.3±1.6#† 409.9% 14.7±1.8# t 65.9% 100.8±5.7# t 25.9%
Losartan 30.8±1.6 10.9±0.5 125.8% 111.6±2.4 31 28.9±4.9 12.2壬 2.6 ' |17.0% 96.8±8.5
#: P<0.05, vs control group
3. 造模后 12周 (见表 20,21)
与对照组大鼠相比, 模型组大鼠 Glu 水平升高 484.2%(P<0.05) ; BUN 水平升髙 88.2%(P<0.05), Scr.水平升高 21.1%。 各给药组大鼠 Glu、 BUN、 Scr.亦升高。 与对照组大鼠 相比, 模型组大鼠尿蛋白水平升高 63.9%, l0sartan组、 本发明化合物 31组大鼠分别较模型组 降低 23.4%及 47.5%。 表 20.本发明化合物 31 对链脲霉素所致大鼠糖尿病肾病模型的各项指标的影响 (造模后 12周)
Group Glu BUN (mmol/L) Scr All
(mmo (umol/L) (Pg/ml)
Control 6 5.5±0.9 8.5±0.5 88.1±23.8 144.4±75.2 Model 5 32.3±1.8# t 484.2% 15.9±1.1*† 88.2% 106.7±5.8 t 21.1% 172.3±69.2 Losartan 6 28.9±4.1 110.5% 14.8±2.0 |6.9% 105.6±5.9 137.1±53.7 31 6 30.4±9.8 i 5.9% 13.8士 2.8丄 13.2% 113.9±13.5 156.8±32.5
#: P<0.05, vs control group
表 21. 本发明化合物 31 对链脲霉素所致大鼠糖尿病肾病模型尿蛋白的影响 (造模后 12周)
Group n UP (mg•day"1) Increase rate (%) Inhibitory rate (%)
Control 6 8.6±1.7
Model 5 14.1±5.1 63.9
Losartan 6 10.8±3.2 23.4
31 6 7.4±2.0* 47.5
#: P<0.05, vs control group *·· P<0.05, vs model group
4. 造模后 16周 (见表 22)
与对照组大鼠相比, 模型组大鼠 Glu水平升高 512.5%(P<0.05); BUN水平 升高 127.7%(P<0.05), Scr.水平升高 42.6%。 各给药组大鼠 Glu.亦升高, 本发 明化合物 31组大鼠 BU 水平有所下降, 较模型组降低 19.1%; losartan组 Scr. 水平有所下降, 较模型组降低 27.1%; 血浆 ANGII水平, 各组无差异。与对照组 大鼠相比,模型组大鼠肾脏脏器指数升高 105.5%(P<0.05),各给药组大鼠亦升高, 表 22.本发明化合物 31 对链脲霉素所致大鼠糖尿病肾病模型的各项指标的影响 (造模后 16周)
Group n Glu (mmol/L) BUN(mmol/L) Scr(umol/L) All (pg/ml)
Control 6 4.4±1.2 5.6±2.4 161.2±41.2 214.5±59.1
Model 5 27.4±1.1# 12.8±0.7# † 229.9±66.1 † 195.2±41.0
t 512.5% 127.7.7% 42.6%
Losartan 4 25.0±6.0 12.3±0.2 167.5±40.6 218.3±76.5
1 27.1%
31 6 28.4±3.9 10.4±1.3 199.7±45.4 226.8±43.5
4 19.1% I 13.1%
#: Ρ<0·05, vs control group
5. 造模后 24周 (见表 23, 24) 与对照组大鼠相比, 模型组大鼠 Glu水平升高 28.0%; BU 水平升高 251.0%(P<0.05), Scr. 水平升高 12.6%。 各给药组大鼠 Glu、 BUN、 Scr.亦升高; 血浆 ANGII水平, 各组无差异; 与 对照组大鼠相比, 模型组大鼠肾脏脏器指数升高 85%, 各给药组大鼠亦升高。 与对照组大鼠 相比, 模型组大鼠尿蛋白水平升高 119.3%, losartan组、 本发明化合物 31组大鼠分别较模型 组降低 34.9%及 47.0%。
表 23.本发明化合物 31 对链脲霉素所致大鼠糖尿病肾病模型的各项指标的影响 (造模后 24周)
Group n Glu (mmol/L) BUN(mmol/L) Scr(umol/L) All (pg/ral)
Control 5 5.3±0.6 4.2±1.6 94.8±8.1 155.1±77.3
Model 5 6.8±3.3 † 17.6±7.1s t 106.8±16.5 † 122.6±59.3
28.0% 251.0% 12.6%
Losartan 5 5.4±2.6 20.3±6.1 106.0±7.1 158.8±100.9
31 6 6.5±3.7 16.3±12.9 119.3±49.5 130.6±82.7
# P<0.05, vs control group
表 24.本发明化合物 31 对链脲霉素所致大鼠糖尿病肾病模型尿蛋白的影响 (造模后 24周)
GROUP n UP (mg .day") Increase rate (%) Inhibitory rate (%)
Control 5 6.11±1.02
Model 5 13.4±10.2# 119.3
Losartan 5 8.72±1.75 34.9
31 6 7.1±3.2* 47.0
#: P<0.05, vs control group, * : P<0.05, vs model group 本发明化合物 31抗肾功能不全的作用机制研究
(一)本发明化合物 31对细胞凋亡的影响
1. 荧光显微镜检测凋亡细胞
方法:
取指数生长期的人肾小管上皮细胞 (HKC), 加入适量含 0.02%EDTA的 0.25%胰蛋白酶液 消化细胞, 使贴壁细胞脱落。 用 DMEM/F12培养基 (含 20%小牛血清)制备成浓度为 104/ml的 细胞悬液, 于 24孔板中每孔接种 0.5ml。 将平板置于 37Ό、 5%C02培养箱, 24小时后加不 同浓度药物。 继续作用所需时间。 收集 HKC细胞, PBS洗一次, 离心, 去上清, 4°C, 4%多 聚甲醛固定细胞 20分钟, PBS洗一次, 加入 Hoechst 33342 (终浓度为 10 μ g/ml), 37°C染色 5- 10分钟。 离心除去染液, 滴片, Olympus荧光显微镜下观察并照相。
结果:
¾胞核形态变化为凋亡细胞最典型的特征, 也是判断凋亡细胞的基本参数。凋亡细胞 的主要形态学变化包括, 细胞核固缩, 染色体 DNA广泛断裂, 断片沿核膜浓聚成多型性 高密度颗粒区; 核膜皱缩, 崩解后包裹染色体片段弥散于细胞浆。 细胞全面皱缩, 细胞膜 皱缩外突, 但细胞膜包裹细胞器或核片段形成凋亡小体。 Hoechst 33342是一种亲脂性染 料, 可跨膜进入细胞对 DNA进行染色。 本研究釆用荧光染色方法观察了 cisplatin合用不 同浓度本发明化合物 31 ( 2-50μη ο1/ί) 作用 24小时的 HKC细胞形态变化。 用荧光染料 Hoechst 33342对细胞进行染色, 在紫外光的激发下, 发出蓝色荧光。 从图可见 cisplatin 组凋亡细胞特征性形态明显, 染色体 DNA断裂并聚集成细小的凝聚块, 并可见到凋亡小 体。 随着本发明化合物 31药物浓度的加大, 镜下凋亡细胞比例逐渐渐少, (见图 3) 2. 本发明化合物 31对 cisplatin诱导的 HKC细胞染色体 DNA断裂的影响 方法:
取指数生长期的人肾小管上皮细胞 (HKC), 加入适量含 0.02%EDTA的 0.25%胰蛋白酶液 消化细胞, 使贴壁细胞脱落。 用 DMEM/F12培养基 (含 20%小牛血清)制备成浓度为 104/ml的 细胞悬液, 于培养瓶中接种 4ml。 将培养瓶置于 37°C、 5%C02培养箱, 24小时后加不同浓度 药物。 继续作用所需时间。 收集 2X 106个细胞, 用 PBS洗涤两次, 重悬于 0.5ml细胞消化液 中(lOOmmol/LNaCl, 10mmol/L Tris-HCl pH8.0, 25mmol/L EDTApH8.0, O.lmg/ml Proteinase K), 混匀后, 50°C保温 12小时, 消化后, 细胞用等体积苯酚 /氯仿 /异戊醇 (25:24:1)抽提一次, 水相 再用氯仿 /异戊醇 (24:1 ) 抽提两次除蛋白, 所得水相用 2.5倍体积无水乙醇沉淀 DNA,用醋 酸钠 (3mol/L)或醋酸铵 (2.5mol/L)助沉, 12000g/min取各管离心, 取上清 18 μ 1, 力卩 5 μ 1上样缓 冲液, 在 1.5%琼脂糖凝胶上电泳, 紫外光下观察并照相。
结果:
细胞凋亡最主要的生化标志是内源性钙-镁离子依赖性核酸内切酶被激活,选择性降解染 色体核小体间 DNA,导致细胞 DNA广泛断裂, 形成大小不一的 DNA片段, 甚至单聚或寡聚 核小体, 从而在琼脂糖凝胶电泳上呈现规则的、 间隔 180~200bp的 " DNA梯子"样条带。 本 文以 cisplatin作为对照药, 用不同浓度的本发明化合物 31 (2-50 u mol/L)合同 cisplatin作用 于 HKC细胞 36小时, 进行琼脂糖凝胶电泳, 由图可见随着本发明化合物 31浓度的加大, cisplatin诱导的 "DNA梯子"样条带逐渐减弱, 呈一定的剂量依赖性。 本发明化合物 31对 10 μ mol/L cisplatin诱导的 HKC凋亡具有明显的抑制作用。 (见图 4)
(二) 抗氧化作用
1.本发明化合物 31对 Fe2+-L-Cys诱发肝微粒体脂质过氧化的影响
方法- 取大鼠肝微粒体 (蛋白含量约为 15mg/ml ) 0. 1ml ; 加入不同浓度的药物和试剂 (1 ) 不 同药物浓度 ΙΟ μ Ι; (2) lmmol/L FeS04 50 μ 1; (3) 10mmol/L L-半胱氨酸 20 μ 1; (4) PBS (pH7.4)0.82ml。合计为 1ml。在 37°C环境中反应 30min;加 20%TCA 0. 3ml终止反应; 2000rpm 离心 15min; 取上清 1. 0ml加 0. 67%硫代巴比妥酸 (TBA) 0. 6ml, 沸水加热 lOmin; 冷却后, 在 532nm处测 0D值, 计算抑制率。
结果:
发明化合物 31在体外对 Fe2+-L-半胱氨酸诱发的肝微粒体脂质过氧化作用具有一定的 抑制作用,且在 10 ug/ml达到最大抑制浓度,抑制率为 32.3%。作用弱于酚羟基化合物 S-3-l。 (见表 25, 图 5)
表 25.本发明化合物 31对 Fe2+-L-cyS诱发肝微粒体脂质过氧化的影响
Concentration
Group Inhibitory rate (%)
O g OD value (x士 s)
/ml)
31 control 0.297 ±0.002
0.312 0.297 ±0.009 0.0%
0.625 0.296 ±0.002 0.4%
1.25 0.294 + 0.003 0.9%
2.5 0.285士 0.003 4.0%
5 0.252 ±0.005 15.2%
10 0.201 ±0.005 32.3%
20 0.200 ±0.003 32.7% 06 000839
S-3-1 control 0.297 ±0.002
0.25 0.149 ± 0002 49.8%
0.5 0.046± 0.001 84.4%
1 0.025 ± 0.002 91.5%
2 0.021 ±0.001 92.9%
4.00 0.024 ± 0.002 91.8%
2. 本发明化合物 31对 cisplatin所致急性肾损伤大鼠肾组织脂质过氧化水平的影响 方法:
上述药效学实验 (本发明化合物 31对顺铂所致大鼠急性肾损伤的保护实验) 中大鼠, 取 肾组织匀浆(每克肾组织制备匀浆 10ml), 按下述方法测定各组大鼠肾组织脂质过氧化水平, 比较各给药组间差异。 肾组织匀桨丙二醛 (MDA, malondialdehyde) 测定如下: 取 0.1ml匀 浆, 加入 0.1ml 10%SDS, 室温静置 20min; 加 2ml 0.1N HC1和 1.0ml 1 %TBA, 混匀, 100 °C水浴, 40min; 冷却后加 4ml正丁醇, 振荡 3〜5min, 萃取, 3000rpm离心 lOmin; 取上层 正丁醇液 0.2ml加至 96孔板中, 用酶标仪 在 532nm处测定 OD值; 标准曲线: 以四乙氧基 丙烷 (TEP) 0、 20、 40、 60、 80、 100 μ mol L作为标准。
结果:
给大鼠腹腔单次注射 6mg/kg cisplatin后 4天, 肾组织脂质过氧化水平明显高于对照组 (P<0.05), cisplatin合用本发明化合物 31组、合用 Benazapril组大鼠肾组织脂质过氧化水平降 低, 其中 cisplatin合用本发明化合物 31 (30mg/kg)组大鼠肾组织脂质过氧化水平与 cisplatin 组相比下降 45.2%(P<0.05)。本发明化合物 31在体内具有一定的抗脂质过氧化作用。 (见表 26) 表 26.本发明化合物 31对 cisplatin所致急性肾损伤大鼠肾组织脂质过氧化水平的影响 (n=10)
MDA
Group Increase rate (%) Inhibitory rate (%)
( μ mol/L)
Control 30.74± 13.51
Cisplatin 53.42±22.57# 73.8
Cisplatin +Benazapril 41.67 ±23.05 22.0
Cisplatin + 31 (lOmg/kg) 42.03 ± 18.84 21.3
Cisplatin + 31 (30mg/kg) 29.28 ± 8.91* 45.2
#: P<0.05 vs control, *: P<0.05 v<y cisplatin model
3. 本发明化合物 31对 cisplatin所致急性肾损伤大鼠肾组织谷胱甘肽水平的影响 方法:
上述药效学实验(本发明化合物 31对 cisplatin所致大鼠急性肾损伤的保护实验)中大鼠, 取肾组织匀浆(每克肾组织制备勾浆 10ml), 按下述方法测定各组大鼠肾组织谷胱甘肽水平, 比较各给药组间差异。 肾组织匀浆巯基 (一 SH) 含量测定 (DT B法) 如下:
( 1 )总 GSH(T-SH): 0.5ml组织匀浆加入 1.5ml的 0.2mol/L Tris缓冲液( pH8.2 ), 0.1ml 0.01mol/L DTNB, 7.9ml无水乙醇, 使得总体积为 10ml; 试剂空白同样制备; 标准曲线: 取 还原型谷胱甘肽 (GSH) 0、 125、 250、 500、 1000 μ mol/L 作为标准; 上述颜色反应 15min 后, 在室温下 3000g离心 15min; 取上清 0.2ml加至 96孔板中, 用酶标仪在 410nm处测 OD 值。
(2) 非蛋白结合型 GSH (NP-SH): 2.5ml组织匀浆加入 2ml双蒸水, 0.5ml 50 TCA, 不断摇动 10〜15min, 3000g离心 15min; 试剂空白同样制备;标准曲线: 同上; 1.0ml滤液 或上清液中加入 2.0ml 0.4mol/L Tris (pH8.9), 0.1ml 0.01 mol/L DTOB, 摇匀; DTNB加入后 5min内, 取上清 0.2ml加至 96孔板中, 用酶标仪在 412nm处测 OD值。 (3 ) 蛋白结合型 GSH (PB-SH): 其值即为 T-SH测量值减去 P-SH测量值。
结果:
给大鼠腹腔单次注射 6mg/kg cisplatin后 4天, 肾组织总 GSH (T-GSH)、 蛋白结合型 GSH(PB-GSH)水平均低于对照组 (P<0.01), 非蛋白结合型 GSH(NB-GSH)水平无明显变化。 cisplatin合用 Benazapril组、 cisplatin合用本发明化合物 31组大鼠肾组织总 GSH (T-GSH)、 蛋白结合型 GSH(PB-GSH)水平亦下降, 与 cisplatin组相比无明显变化。 (见表 27) 表 27.本发明化合物 31对 cisplatin所致急性肾损伤大鼠肾组织谷胱甘肽水平的影响 (n-10)
GSH
Parameters Group
( μ mol/g kidney)
T-GSH Control 21.32±2.64
、 Cisplatin model 16.61 ± 3.43*#
Cisplatin +Benazapril 16.56 ± 2.75
Cisplatin + 31(10mg/kg) 15.82±2.49
Cisplatin + 31(30mg/kg) 15.24±2.85
NB-GSH Control 2.78 + 0.38
Cisplatin model 2.96±0.25
Cisplatin +Benazapril 3.86 ± 1.51
Cisplatin + 31(10mg/kg) 4.05 ± 1.84
Cisplatin + 31(30mg/kg) 2.80 ±0.34
PB-GSH Control 18.54±2.49
Cisplatin model 13.65 ± 3.42##
Cisplatin +Benazapril 12.89 ± 4.21
Cisplatin + 3 l(10mg/kg) 1 1.84± 3·33
Cisplatin + 31(30mg/kg) 12.43 ±2.83
##: P<0.01 vs control
(三) 与肾素 -血管紧张素 -TGF- β通路相关的机制
1. 体外检测本发明化合物 31对血管紧张素转化酶 (ACE) 的抑制作用
方法:
反应底物 HHL以缓冲液 (HEPES 50mM, NaCl 300mM, PH=8. 3) 配成 lOraM, ACE以缓冲液配成
80mU/ml , 每一药物浓度均设对照管, 依次如下表加药:
HHL(lOmM) Drug or water IN HC1 ACE(80mU/ml)
Test tube 125ul 10.4ul 125ul
Control tube 125ul 10.4ul 250ul 125ul
37°C,振摇反应 30min后加 125ullN HCl终止反应; 各管 (实验管和对照管) 加入 1.5ml乙酸 乙酯振摇萃取, 取上层乙酸乙酯 1.0ml; 120°C、 30min挥发干乙酸乙酯, 加水 lml溶解, 紫 外 228nm处测吸收值。 ACE
Figure imgf000048_0001
6x10 以加水管为对照计算各 加药管的抑制率。
结果:
^外通过检测在血管紧张素转化酶作用下 HHL反应生成马尿酸 (Hippuric acid) 的量 P T/CN2006/000839 来判断血管紧张素转化酶的活性。 本发明化合物 31体外在 l(T9niol/L、 Ιθ ηοΙ/L浓度下对血 管紧张素转化酶具有微弱的抑制作用。 (见表 28) 表 28.本发明化合物 31在体外对血管紧张素转化酶(ACE) 的抑制作用
Drugs Concentration(mol/L) Inhibitory rate (%)
Control
Figure imgf000049_0001
2. 本发明化合物 31对 TGF- β 1受体结合试验的拮抗作用
方法:
将 Balb/c 3T3或 NIH 3T3 细胞接种到 96孔板, 培养条件为 37 °C, 5%C02, DMEM培养 基 (含 10%胎牛血清)。 培养 2—4 天后,在细胞接近融合时, 将培养液换成结合缓冲液 (50mmol/L HEPES中含有 NaCl, KC1, MgS04和 CaCl2),加入 100 pmol/L [125I]TGF- β 1 激 发试验, 同时加入受试化合物。 细胞培养 4小时后, 弃去培养基, 用冰冷的结合缓冲液洗漆 细胞。测定 lOnmol/LTGF-β 1的非特异性结合。细胞溶解在 Triton X-100缓冲液中, 测定放 射性。
结果:
实验结果见表 29, 本发明化合物 31对 TGF-β!受体结合具有一定的抑制作用。 表 29.本发明化合物 31在体外对 TGF-β 1受体结合试验的拮抗作用
Cell Lines Concentration^ moIL) Inhibitory rate (%)
NIH-3T3 0.1 15.0
1.0 20.0
10.0 25.6
Balb 3T3 0.1 0
1.0 5.7
10.0 12.2
3.本发明化合物 31对 cisplatin致急性肾损伤大鼠血浆 TGF- β ι升髙的抑制作用 方法:
(1)标准曲线制作: 设标准孔 8孔, 每孔中各加入样品稀释液 100 μΐ, 第一孔加标准品 100 μΐ, 混匀后用加样器吸出 100μ1, 移至第二孔。 如此反复作对倍稀释至第七孔, 最后, 从第七孔中吸出 ΙΟΟμΙ弃去, 使之体积均为 100μ1。 第八孔为空白对照。
(2)加样:待测品孔中每孔各加入已激活的待测样品 100μ1。将反应板置 37°C 120min。 洗板: 用洗涤液将反应板充分洗涤 4〜6次, 在滤纸上印干。 06 000839
(3) 每孔中加入第一抗体工作液 50 μ 1。 将反应板充分混匀后置于 37°C 60min。 洗板: 用洗涤液将反应板充分洗涤 4〜6次, 在滤纸上印干。
(4) 每孔加酶标抗体工作液 100 μ 1。 将反应板置 37°C 60min。 洗板: 用洗涤液将反应 板充分洗涤 4〜6次, 在滤纸上印干。
(5) 每孔加入底物工作液 100 μ 1, 置于 37°C暗处反应 5〜10inin。
(6) 每孔加入 1滴终止液混匀。
(7) 在 492nm处测定 OD值。
( 8) 结果计算: 所有 OD值均减去空白值后计算。 以标准品 2000、 1000、 500、 250、 125、 62.5、 32 pg/ml对 OD值作标准曲线, 根据该标准曲线计算相应的 TGF- β i值。
结果:
上述 cisplatin所致急性肾损伤大鼠模型, 腹腔注射 6mg/kg cisplatin后 4天, 血浆 TGF- β 1水平较对照组动物增加 72.8%(Ρ<0.05),与 cisplatin模型组相比, cisplatin合用 bennazapril 组、 cisplatin合用本发明化合物 31 10mg/kg组、 cisplatin合用本发明化合物 31 30mg/kg组大 鼠血浆 TGF- β 1水平分别下降 44.5% (P<0.05 )、 60.8% (P<0.01 )、 61.4% (P<0.01 ) (见表 30) 表 30.本发明化合物 31对 cisplatin致急性肾损伤大鼠血浆 TGF- β i水平的影响 (n=10)
Increase rate Inhibitory rate
Group TGF- P i(ng/ml)
(%) (%)
Control 8.26±3.94
Cisplatin model 14.28 + 6.35" 72.8
Cisplatin +Benazapril 7.92 ±3.70' 44.5
Cisplatin + 31(10mg/kg) 5.60 +2.03 60.8
Cisplatin + 31(30mg/kg) 5.52± 1.84 61.4
#: P<0.05 vs control , *: P<0.05 vs cisplatin model, **: P<0.01 vs cisplatin model
4. 本发明化合物 31对 5/6肾切除模型大鼠血浆 TGF-βΙ水平的影响
方法:
同本发明化合物 31对 cisplatin致急性肾损伤大鼠血浆 TGF- β 1升高的抑制作用 结果:
结果见表 31。 与假手术组相比, 模型组大鼠血浆 TGF-βΙ水平在术后 12周无明显变化; 术后 16周开始升高, 较假手术组升高 40.7%。 术后 16周 losartan组、 benazepril组、 本发明 化合物 31 10mg/kg、本发明化合物 31 30mg/kg组与模型组相比分别下降 35.7%、 9.3%、 16.7%、 39.9%。 表 31.本发明化合物 31对 5/6肾切除模型大鼠血浆 TGF-βΙ水平的影响
Weeks after Increase rate' Inhibitory rate
GROUP TGF- β 1 (ng «πιΓ1)
operation (%) (%)
12 SM 13·28± 3.53
Model 12.24±4.44 -7.9%
Losartan 10.51 ± 6.16
Benazepril 9.65 +4.23
31 lOmg/kg 12.57±4.61
31 30mg/kg 10.94±2.58
16 SM 11.05 ±5.27
Model 15.54± 5.67 40.7% Losartan 9.99+5.94 35.7%
Benazepril 14.10±6.07 9.3%
31 lOmg/kg 12.94 ±3.44 16.7%
31 30mg/kg 9.34 ± 1.07" 39.9%
**: P<0.01 vs model group
5. RT-PCR观察本发明化合物 31对 cisplatin所致急性肾损伤大鼠肾组织 TGF- β! mRNA表达的影响
方法:
肾组织总 RNA提取
( 1 ) 在经过 DEPC处理过的 50ml离心管中放置 0.5g液氮冻存的肾组织,加入 5ml Trizol试剂, 用勾浆器充分匀浆约 l〜2min, 随后 Vortex振荡混匀, 冰浴 lOmino
(2) 加入 1.75ml氯仿, 充分振荡或上下 ^¾回颠倒数次, 稍静止后出现分层, 随即于 4 °C, 12000rpm离心 15min。
(3) 将上清 (水相) 转移于另一支干净 50ml离心管中, 注意不要吸到中间层 的蛋白沉淀。加入等体积的异丙醇 (4 °C预冷)混匀。 4 °C, 12000rpm离心 20min。
(4) 沉淀即所需要的总 R A, 倒掉上清后, 加入 1.5ml 75 %乙醇, 晃荡洗涤 一次并小心地倒掉乙醇。 再 1.2000rpm离心几秒钟, 用 Tip头吸干上清。 然后加 入 200 μ 1 DEPC处理过的水溶解沉淀, 储存于 -20°C待用。
(5 ) 吸 20 μ 1 RNA提取液加 DEPC处理水至 400 μ 1, 于 260nm及 280nm测
OD值。 若 OD260nm: OD28nm>2, 则所测 RNA较纯。 由 1 OD26()nm= 40 μ g/ml
RNA推算 RNA浓度。
大鼠 TGF- 引物
Sense primer: 5' ATG GTG GAC CGC AAC AAC 3 '
Anti-sense primer: 5' CCAAGG TAA CGC CAG GAA T
大鼠 P -actin 引物
Sense primer: 5'GTG GGG CGC CCC AGG CAC CA 3'
Anti-sense primer: 5'CTT CCT TAA TGT CAC GCA CGA TTT C 3 '
RT-PCR
反应体系 (Add nuclease-free water to total volume 50 μ 1 ) :
Figure imgf000051_0001
反应条件:
1 cycle 48。C for 45min
1 cycle 94。C for 2min
35 cycles 94 °C for 30second
55。C for lmin
68。C for 2min
Figure imgf000052_0001
电泳
取 RT- PCR产物 40μ1, 加 6X上样缓冲液 8μ1, 于 1.7%琼脂糖凝胶电泳 (恒压 70V), 拍照。
结果:
上述 效学实验 (本发明化合物 31对 cisplatin所致大鼠急性肾损伤的保护实验) 中大鼠肾组织 RT-PCR产物经琼脂糖凝胶电泳, EB染色, 观察 DNA条带强弱。 结果 cisplatin组大鼠的 DNA条带强于对照组; cisplatin合用 benazapril组、 cisplatin合用本发明化合物 31组大鼠的 DNA条带弱于 cisplatin组大鼠,通过半 定量分析分别下降 42.0%、 14.2%、 44.5% (见图 6,7)。 benazapril和本发明化合 物 31均能不同程度地抑制 cisplatin诱导的大鼠肾组织 TGF- β 1 mRNA表达增 高。
6. 本发明化合物 31对 cisplatin所致急性肾损伤大鼠血浆 Angll水平的影 响
方法:
血管紧张素 II (ANGII) 放免分析测定: 大鼠腹腔注射 35mg/kg戊巴比妥 钠麻醉后,用毛细玻璃管于眼底静脉丛取血 lml,置于冰水浴冷却的酶抑制剂抗 凝管中, 摇匀, 即刻再置入冰水浴中冷却, 待离心时取出。 4°C lOOOrpm离心 5min, 分离血桨 (在 -20°C可保存 2月)。
步骤:
在冰水浴中按下表加样:
总管 NSB管 标准管 样本管
缓冲液 150 ΐ ― ―
ANGII标准 ― 100 Ul ―
样本血浆 ― ― 100 U l
125卜 ANGII 50μ1 50U1 ' 50U1 50ul
ANGII抗体 ― 50ul 50 Ul
摇匀, 4'C放置 15小时以上
分离剂 250 U 1 250 μΐ 250 ΐ
摇匀, 室温放置 15min、 3500rpm离心 15min, 吸去上清, 再测定各管沉淀物 的放射性 Bi (cpm)o
结果计算: 结合百分率计算: 设 SQ管计数为 BQ, 各标准管或样品管计数为
B, 则计算公式如下: H xioo%
B
Logit计算:
Figure imgf000052_0002
N2006/000839 以标准浓度取常用对数值为横坐标, 对应的 logit值为纵坐标作标准曲线。 待测样品 ANGII浓度即可根据标准曲线得出。 结果:
上述 cisplatin所致急性肾损伤大鼠模型, 腹腔注射 6mg/kg cisplatin后 4天, 血浆 ANG II水平较对照组动物增加 279.6%(P<0.01), 与 cisplatin单药组相比, cisplatin合用 bennazapril组、 cisplatin合用本发明化合物 31 10mg/kg组、 cisplatin 合用本发明化合物 31 30mg/kg组大鼠血浆 Ang ll水平分别下降 44.3%(P<0.05 )、 45.0% (P<0.01 )、 60.2% (P<0.01 ) (见表 32)。 cisplatin与本发明化合物 31合用 组大鼠的血桨 Ang II水平亦升高, 各实验组间血桨 Ang II水平无显著性差异。 表 32.本发明化合物 31对 cisplatin所致急性肾损伤大鼠血浆 Ang ll水平的影响 (n=10)
Group Ang II (ng/ml) Increase rate (%) Inhibitory rate (%)
Control 138.82± 83.46
Cisplatin model 527.02 ± 179.32 ## 279.6
Cisplatin + Benazapril 293.41 ± 91.97 44.3
Cisplatin + 31(10mg/kg) 289.67± 158.89*' 45.0
Cisplatin + 31(30mg/kg) 209.65 ±66.08*' 60.2
##: P<0.01 vs control, **: P<0.01 vs cisplatin model
7. 本发明化合物 31对 5/6肾切除模型大鼠血浆 Angll水平的影响 方法:
同本发明化合物 31对 cisplatin所致急性肾损伤大鼠血浆 ANGII水平的影 响。
结果:
结果见表 33。与假手术组相比,模型组 ANGII水平在术后 16周升高 23.9%, 术后 16周 benazepril组、本发明化合物 31 10mg/kg组、本发明化合物 31 30mg/kg 组与模型组相比分别下降 18.3%、 7.7%、 39.8%; losartan组与模型组相比有所升 表 33. 本发明化合物 31对 5/6肾切除模型大鼠血浆 Angll水平的影响
Weeks after _ Inhibitory rate
. GROUP Ang II (pg ·ηΊ " Increase rate (%)
operation (%)
16 SM 106.78 ± 32.53
Model 140.32± 66.86 23.9
Losartan 245.91 ±73.88 -75.3
Benazepril 1 14.70±26.94 18.3
31 lOmg/kg 129.46±37.22 7.7
31 30mg/kg 84.45 ± 17.62 39.8
(四) 与胶原形成和分解相关的机制
1. 底物酶谱法分析本发明化合物 31对 HT-1080细胞分泌基质金属蛋白酶能力的影响 方法: 根据 Heussen等的方法加以改进。取对数生长期的 HT-1080细胞,消化后进行细胞计数, 以 I X 105/孔的密度接种于 24孔培养板中, 培养过夜。 次日每孔加入含有一定浓度药物和对 照溶剂的培养基培养 12h。 弃培养上清, PBS洗三次, 然后换无血清加药培养基 300μ1继续 培养 12小时。 收集细胞培养上清, 4°C, 200g离心 lOmin去除细胞碎片, 上清液于 -20°C储 存备用, 细胞消化计数。 SDS-聚丙烯酰胺凝胶电泳参考文献进行。 配制一定体积的 8% 分离 胶和 5%浓缩胶, 分离胶中含 0.1% (w/v)明胶。按细胞数折算出相同细胞数所对应的培养上清 体积, 并按此体积加样电泳 (上样缓冲液中不含 DTT)。 电泳完毕后, 剥离凝胶, 以蒸馏水 漂洗后, 移入 100ml 2.5 %TritonX-100溶液中, 在摇床上低速摇动以洗脱 SDS。 30min后, 换 新的 TritonX-100溶液继续洗脱 30min。 凝胶移入 100ml明胶酶缓冲液 (50mmol/L Tris-HCl, pH7.5, 10mmol/L CaCl2,200mmol/LNaCl,^mol/L ZnCl2) 中, 37°C恒温温育 16小时。 蒸馏水 漂洗后, 凝胶以 0.1% 考马斯亮蓝 R-250染液染色 4小时, 蒸馏水漂洗后, 在脱色液 (冰醋 酸: 甲醇: 7K: =10: 45: 45 ) 中脱色 l-2h, 至对照组出现明显、 清晰的负染条带。 凝胶扫描 照相, 负染条带的宽度和亮度反映明胶酶的活性, 用 Gel-Pro Analyzer 3.1软件对负染条带密 度扫描并进行半定量分析。
结果:
IV型胶原是细胞外基质的重要组成成分, 明胶酶 (gelatinase) 或称 IV型胶原酶能降解组 成基质膜的 IV型胶原, 其活性是影响 IV型胶原降解重要因素之一。用 SDS-聚丙烯酰胺凝胶电 泳可以将 HT1080细胞分泌到培养上清中的明胶酶和它们的活性形式按分子量大小分开, 经 TritonX-100去除和蛋白结合的 SDS后, 明胶酶能恢复其蛋白降解活性。药物作用细胞后, 如 果影响到明胶酶表达和分泌的调节通路, 则分泌到培养上清中的各种形式的明胶酶的含量将 发生变化。 如果把明胶惨入到凝胶液中使之与丙烯酰胺共价聚合, 在适宜的反应条件下, 明 胶酶就能降解其周围的明胶, 蛋白降解区不能被考马斯亮蓝染色, 所以在明胶酶的活性区域 附近就能出现一条负染带。 肿瘤细胞分泌的明胶酶越多, 带的亮度和宽度也就越大。 用这种 底物酶谱法 (Zymography) 观察本发明化合物 31对 HT-1080细胞分泌基质金属蛋白酶能力 的影响, 结果表明在 cisplatin作用下 HT1080细胞分泌明胶酶比对照组明显减少; cisplatin合 用本发明化合物 31组明胶酶分泌增加, 且具有一定的浓度依赖关系。 (见图 8)
2. 底物酶谱法分析本发明化合物 31对大鼠肾系膜细胞 (rMC) 分泌基质金属蛋白酶能 力的影响
方法:
细胞改用大鼠肾系膜细胞(rat mesangial cell, rMC)方法同底物酶谱法分析本发明化合物
31对 HT-1080细胞分泌基质金属蛋白酶能力的影响。
结果:
用底物酶谱法 (Zymography) 观察本发明化合物 31 对大鼠肾系膜细胞分泌基质金属蛋 白酶能力的影响, 结果表明在 cisplatin作用下 HT1080细胞分泌明胶酶比对照组减少; 本发 明化合物 31组可使 rMC明胶酶分泌增加, 且具有一定的浓度依赖关系。 (见图 9) 实验例 6 本发明化合物 56对链脲霉素致雌性 SD大鼠糖尿病肾病模型的影响
实验方法
模型建立及分组给药
取雌性 SD 大鼠, 体重 180-200g, 除正常对照组外, 腹腔注射 60mg/kg 链脲霉素 (Streptozotocin, STZ), 3天后可造成大鼠糖尿病。 正常对照组腹腔注射 0.1mol/L柠檬酸缓 冲液 (pH7.4)。 00839 实验共设 6组, 每组 10-12只。 设正常对照组、模型对照组、 氯沙坦 (10mg/kg)阳性药组, 本发明化合物 56 5、 10、 20mg/kg三个剂量组。 造模后 3天按血糖水平分组, 同时给药。 均 为灌胃给药, 每周 6次, 持续给药至 20周。 正常对照组及模型对照组同时灌胃给予 0.5%羧 甲基纤维素钠 (CMC) 混悬液。
于造模后 4周, 给糖尿病大鼠皮下注射胰岛素 3U/只, 每周 2次, 以维持其体重。
检测指标
每周称体重观察大鼠生长状况; 于造模后 16、 20周测定如下指标- a. 乙醚麻醉动物, 眼球后静脉丛取血, 检测血清尿素氮 (BUN)、 肌酐 (Scr)、 血糖 (Glu), 及血清 TGF-βΙ水平;
b. 将大鼠置于代谢笼中收集 24小时尿液, 测定尿白蛋白量 (1Mb)、 尿肌酐 (Ucr) 水平, 计算 24小时尿白蛋白量及肌酐清除率 (C.L);
c 术后 20周处死动物, 计算各组动物肾脏指数、 心脏指数, 取肾脏做组织病理学检查。 检测方法:
a. BUN, Glu, 采用北京北化康泰临床试剂有限公司检测试剂盒检测;
b. 血清 TGF-βΙ采用上海森雄科技实业有限公司生产的 ELISA试剂盒检测;
c Scr、 Ucr和 1Mb, 采用北京利德曼生化技术有限公司试剂盒检测。
肾组织病理学检查
标本经 10%甲醛固定, 石蜡包埋, HE和 PASM染色, 光镜检査, 组织学照片。 肾小球 病变:每只动物计数 30个小球,按未见异常;节段性硬化;弥漫性硬化和球囊渗出分类(%)。 肾小管空泡变性以 "一; +; ++; +++"号表示未见异常; 轻、 中、 重病变程度。
统计学处理 一
实验结果用均值土标准差(;土 SD)表示,在 SPSS软件中统计。多组间差异采用 One Way ANOVA进行统计处理, 肾组织病理学检验中肾小管空泡变性经 X2检验比较。 当?<0.05时 认为组间差异有统计学意义。 ' 实验结果
1. 大鼠生长及血糖水平
与正常对照组大鼠相比, 糖尿病大鼠体重明显较轻。糖尿病大鼠血糖水平一直维持较高 水平, 平均血糖值大于 300mg/dL。
2.本发明化合物 56对 STZ所致糖尿病肾病大鼠血 BUN水平的影响
结果见表 1。与对照组相比, 模型组大鼠 BUN水平在造模后 8周开始升高, 于造模后 16 周、 20周分别上升了 128.2%、 137.0% (均 P<0.01)。 Losartan可缓解糖尿病大鼠 BU 升高, 治疗后 16周、 20周 BUN水平较模型组下降 18.5%(P>0.05)、 31.4%(P<0.05)。 本发明化合物 56三个剂量治疗组均可不同程度缓解糖尿病大鼠 BU 升高, 20mg/kg 治疗组作用较明显, 治疗后 16周、 20周 BU 水平较模型组下降 27.0%(Ρ<0.05)、 31.3%(Ρ<0·05)。
3.本发明化合物 56对 STZ所致糖尿病肾病大鼠血 Scr水平的影响
结果见表 2。 与对照组相比, 模型组大鼠 Scr水平升高不明显。 在治疗 20周时, 模型组 Scr水平上升了 14.1%(P>0.05), 本发明化合物 56 20mg/kg组大鼠 Scr较模型组降低 25.3%, 但无显著性差异。 4.本发明化合物 56对 STZ所致糖尿病肾病大鼠肌酐清除率 (C.L)的影响
结果见表 3。与对照组相比,模型组大鼠 C丄在造模后 20周降低 (-19.8%,P>0.05)。LOSartan 及本发明化合物 56 20mg/kg可升高糖尿病肾病大鼠的 C丄, C丄分别较模型组大鼠升高 62.4% 及 44.3% (均 P>0.05)。
5.本发明化合物 56对 STZ所致糖尿病肾病大鼠尿白蛋白 (1Mb)水平的影响
结果见表 4。与对照组相比, 模型组大鼠 Ualb水平在造模后 8周开始升高, 于造模后 16 周、 20周分别上升了 163.3%(P<0.05)、 491.3%(P<0.05)。 Losartan于治疗 16周、 20周时可缓 解糖尿病大鼠 Ualb升高, Ualb水平较模型组下降 65.7%(P<0.05)、 47.2%(P<0.05)o本发明化 合物 56 20mg/kg治疗组亦可缓解糖尿病大鼠 Ualb升高, 于治疗 16周、 20周时 Ualb水平 较模型组下降 75.7%(P<0.01)和 35.3%(P>0.05)。
表 34.化合物 56对 STZ致糖尿病肾病大鼠血 BUN水平的影响 给药时间 BUN (rag/dL)
组别 抑制率(o/。)
(周) 1±SD
Figure imgf000056_0001
注: 0. 05, ##P<0. 01 , 与正常对照组比较; *Ρ<0. 05, **Ρ<0· 01, 分别与模型组比较 t 表 35化合物 56对 STZ致糖尿病肾病大鼠血 Scr水平的影响 给药时间 Scr. (mg/dL)
组别
(周) 一
X+SD
16 正常对照组 0. 26+0. 03
模型组 0. 26±0. 10 0
Losartan
0. 28±0. 05 -10. 0
lOmg/kg
56 5mg/kg 0. 24+0. 05 5. 8
10mg/kg 0. 27+0. 09 6. 3 20mg/kg 0. 14±0. 04* 43. 5
20 正常对照组 0. 26+0. 06
模型组 0. 29±0. 09 -14. 1
Losartan
0. 18+0. 04 38. 1
10mg/kg
56 5mg/kg 0. 25+0. 10 14. 6
lOmg/kg 0. 24±0. 07 17. 5
20mg/kg 0. 22+0. 04* 25. 3 注: #P<0. 05, 難〈0. 01, 与正常对照组比较; *P<0. 05, **P<0. 01, 分别与模型组比较 c 表 36.化合物 56对 STZ致糖尿病肾病大鼠肌酐清除率 (CL)的影响
C丄 (ml/min) 增加率
X±SD (%)
16 正常对照组 1.47 ±0.56
模型组 3.43 ±2.16 134.0
Losartan 10mg/kg 3.24 ±1.89 -5.6
56 5mg/kg 3.19 ±1.41 -7.1
10mg/kg 2.68 +1.46 -21.9
20mg/kg 3.76 ±2.36 9.6
20 正常对照组 1.88±0.95
模型组 1.51+0.71 -19.8
Losartan 10mg/kg 2.45±1.14 62.4
56 5mg/kg 1.48+1.11 -1.5
56 lOmg/kg 1.86+1.01 23.2
56 20mg/kg 2.18±1.66 44.3 注: #P<0. 05, ##P<0. 01 , 与正常对照组比较; *P〈0. 05, **P〈0. 01 , 分别与模型组比较 c 表 37化合物 56对 STZ致糖尿病肾病大鼠尿白蛋白 (Ualb)水平的影响 给药时间 Ualb(mg/day)
组别 抑制率(%) (周) X±SD
16 正常对照组 0.40±0.17
模型组 . 1.06±0.77 163.3
Losartan 10mg/kg 0.36+0.31 * 65.7
56 5mg/kg 1.21+0.83 14.0
10mg/lcg 1.04±0.61 1.9
20mg/kg 0.26 ±0.14 75.7
20 正常对照组 0.42+0.16
模型组 2.49±1.22# 491.3 Losartanl Omg/kg 1.31+0.90* 47.2
56 5mg/kg 1.94+0.79 21.8
1 Omg/kg 2.16±0.70 13.1
20mg/kg 1.61+0.84* 35.3
注: #P<0. 05, ##P<0. 01 , 与正常对照组比较; *P<0. 05, **P〈0. 01, 分别与模型组比较 t
6. 本发明化合物 56对 STZ所致糖尿病肾病大鼠 TGF-pi水平的影响
本发明化合物 56 20mg/kg可明显抑制糖尿病大鼠血中 TGF-βΙ水平增髙。 表 38.化合物 56对 STZ致糖尿病肾病大鼠血中 TGF-βΙ水平的影响 给药时间 , TGF-pi(ng/ml)
H fill 抑制率 (:%)
16 正常对照组 35.2±8.7
模型组 37.1+9.6# -5.2
Losartanl Omg/kg 33.9+12.0 8.6
56 5mg/kg 35.4+10.5 4.5
1 Omg/kg 36.5±8.9 1.5
20mg/kg 26.9±11.9 27.3
20 正常对照组 34.4±2.3
模型组 40.3±10.9# -17.2
Losartanl Omg/kg 32.4±8.2 19.6
56 5mg/kg 40.6+7.1 -0.6
1 Omg/kg 27.9±4.9 * 30.9
20mg/kg 29.9+6.6 * 25.9
注: #P<0. 05, 〈0. 01, 与正常对照组比较; *P〈0. 05, **P<0. 01, 分别与模型组比较 t
7.本发明化合物 56化合物对 STZ所致糖尿病肾病大鼠心脏、 肾脏肥大的缓解作用
结果见表 6 和表 7。 与对照组相比, 模型组大鼠肾脏指数、 心脏指数分别升高 102.9%(P<0.05)、 25.0%(P<0.05)。 Losartan治疗组大鼠肾脏指数、 心脏指数较模型组降低, 分 别降低 11.3%(P>0.05)和 7.5%(P>0.05)。 本发明化合物 56治疗组大鼠肾脏指数亦较模型组降 低, 低、 中、 高三个剂量分别降低 11.3%、 9.9%、 9.9% (均 P >0.05 ); 本发明化合物 56心脏 指数较模型组低,低、中、高三个剂量分别下降 7.5%(P>0.05)、12.5%(P<0.05)和 10.0%(P<0.05 )o 表 39.化合物 56对 STZ致糖尿病肾病大鼠肾脏指数的影响
肾指数(g/100g体重) 抑制率
组别
X+SD (%)
正常对照组 0.70+0.06 模型组 1.42+0.19# 102.9
LosartanlOmg/kg 1.26+0.17 11.3
56 5mg/kg 1.26+0.09 11.3
lOmg/kg 1.28+0.02 9.9
20mg/kg 1.28±0.03 9.9
注: #P<0. 05, ##P<0. 01 , 与正常对照组比较; *P<0. 05, **P〈0. 01, 分别与模型组比较。 表 40. 化合物 56对 STZ致糖尿病肾病大鼠心脏指数的影响
心脏指数 (g/lOOg体重) 抑制率
组别 ―
X±SD (%)
正常对照组 0.32+0.03
模型组 0.40+0.03# 25.0
Losartan 10mg/kg ' 0.37+0.05 7.5
56 5mg/kg 0.37±0.03 7.5
10mg/kg 0.35±0.02* 12.5
20mg/kg 0.36±0.03* 10.0 注: #P<0. 05, ##P<0. 01 , 与正常对照组比较; *P〈0. 05, **P<0. 01, 分别与模型组比较。
8.本发明化合物 56对 STZ所致糖尿病肾病大鼠白内障的缓解作用
造模 20周时白内障发生率, 模型组大鼠为 80%, Losartan组为 62.5%,本发明化合物 56三剂量治疗组分别为 85.7%、 62.5%和 50%。
9.本发明化合物 56对 STZ所致糖尿病肾病大鼠肾脏病理的影响
①正常对照组: 96. 3%肾小球未见异常改变, 个别肾小球发生节段性坏死, 肾小管和肾间质未 见异常改变。
②糖尿病动物模型: 该动物模型肾脏主要病变在肾小球和肾小管以及肾间质。 肾小球病变是 以小球血管袢节段性硬化为主, 即小球轻度系膜区增宽, 系膜细胞增生, 毛细血管基底膜 增厚。 肾小球弥漫性硬化, 即小球系膜基质增多, 系膜区增宽, 毛细血管基底膜弥漫性增 厚、 硬化。 这类病变偶见; 在小球囊内未见纤维素样和小滴状物渗出。 肾小管病变主要表 现在近端小管上皮空泡变性(糖原沉积), 胞核固缩, 染色深。 严重者满视野均可见这种病 变。 肾间质炎细胞浸润和肾盂黏膜上皮细胞增生等也明显增加。 上述病变检查结果分别见 表 8和表 9。模型组肾小管和间质病变积分为 5. 33+1. 80。有 4例动物肾小动脉硬化, 中膜 增厚, 局部向血管腔内突出, 管腔缩窄等。
③氯沙坦组: 正常肾小球比例多于模型组 (P〈0. 05), 肾小管空泡变性和间质病变也明显轻于 模型组(P〈0. 05)。
④ 56给药组: 本发明化合物 56各剂量组肾小球节段性硬化、 系膜细胞增生的肾小球比例明 显少于模型组 (P<0. 01), 而肾小管空泡变性和间质病变积分中剂量组与模型组比较有非常 显著性差异 (P<0. 01) ,大、 小剂量组亦有些好转, 但与模型组比较无统计学差异 (P>0. 05)。 结果见表 8和 9。 结论: 本发明化合物 56各剂量组对 STZ诱导的雌性 SD大鼠糖尿病动物模型肾小球硬化、 肾 小管空泡变性和肾间质病变具有明显的治疗作用。
表 41 化合物 56对 STZ致糖尿病肾病大鼠肾小球病变的影响 (%) 未见异常 节段性硬化
正常对照 9 96. 30±3. 52 3. 70±3. 52
模型组 9 62. 20+9. 57s 37. 76+9. 57""
氯沙坦组 8 74. 76+10. 68* 25. 24±10. 68**
56 (5mg/kg) 8 77. 14±8. 91* 22. 91+8. 99**
56 (10mg/kg) 8 87. 49+6. 61** 12. 51+6. 61**
56 (20mg/kg) 8 90. 00±6. 16** 9. 17±6. 09**
注: #P<0. 05, 離〈0. 01, 与正常对照组比较; *P<0. 05, **P<0. 01, 分别与模型组比较。 表 42化合物 56对 STZ致糖尿病肾病大鼠肾小管空泡变性和肾间质病变积分 组别 例数 积分 (X±SD)
正常对照组 9 0
模型组 9 5. 33+1. 80
Losartan 8 3· 42±1. 81*
56 (5mg/kg) 8 4. 57+1. 62
56 (10mg/kg) 8 2. 50±1. 93**
56 (20mg/kg) 8 3. 75+1. 83
注: *P<0. 05 ; **P〈0. 01, 分别与模型组比较。

Claims

权 利 要 求
1、 如通式 (I)所示的化合物
Figure imgf000061_0001
其中:
X选自 0、 NH;
W选自 CO、 CH2;
R选自直链或支链的 d-6烷基;
R6 、 R7和 独立地选自 H、 OH、 N02、 羧基、 卤素、 C^烷基、 d.8烷氧基; R3选自
取代或未取代的苯基 (苯环上的取代基为 Ru );
取代或未取代的苯基 CM烷基 (苯环上的取代基为 R12, 烷基上的取代基为 R" ),· 取代或未取代的 N-吡咯基, (吡咯环上的取代基为 R13);
取代或未取代的咪唑基 (咪唑基上的取代基为 R15);
取代或未取代的吡唑基 (吡唑基上的取代基为 Rl6);
取代或未取代的 N-吲哚基 (吲哚基上的取代基为 R17);
并且, R" 选自 C1-4烷氧羰基;
R13表示单取代或多取代, 取代基独立的选自 H、 OH、 卤素、 N02、 C02H、 C1-8烷基、 C1-8卤代烷基、 .8羰烷基、 d.8烷氧基、 C I-8羧基烷氧基、 酰基、 C1-8垸氧羰基、 C1-8酯 基、 四氮唑基 , 未取代或取代的苯羰基、 苯环上的取代基为 R14 ;
Ru、 R12、 R14, R15、 R16 和 Rn均表示单取代或多取代, 取代基独立的选自 H、 OH、 卤素、 N02、 C02H、 C 8烷基、 d.8卤代烷基、 d.8羰烷基、 C1-8垸氧基、
C M酰基、 d.8垸氧羰基、 C1-8酯基、 四氮唑基。
2、 根据权利要求 1所述的化合物, 其特征在于, 如通式 (IA) 所示
Figure imgf000061_0002
其中, R-X选自 0 、 NH、 NCH3, NCH(CH3)2;
R6、 R7 和 独立的选自 H、 OH、 Cl、 N02、 C1-4烷基、 C1-4垸氧基;
为单取代或多取代, 取代基独立的选自 H、 OH、 Cl、 N02、 CM烷基、 C14垸氧基、 CF3、 OCH2C02H 、 甲酰基、 乙酰基、 C02H、 C02CH3、 C02Et、 (1",2"3,,,4,,-四唑 -5")。
3、 根据权利要求 1所述的化合物, 其特征在于, 如通式 (IB) 所示
Figure imgf000062_0001
其中, R-X选自 O 、 NH、 NCH3, NCH(CH3)2
R6、 R7和 R8独立的选自 H、 OH 、 Cl、 N02、 .4烷基、 .4烷氧基;
R"选自 C02H、 C02C¾、 C02Et;
R12为单取代或多取代, 取代基独立的选自 H、 OH、 Cl、 N02、 Cw垸基、 Cw烷氧基、 CF3、 OCH2C02H 、 甲酰基、 乙酰基、 C02H、 C02C¾、 C02Et、 (Γ,,2"3",4,,-四唑 -5")。
4、 根据权利要求 1所述的化合物, 其特征在于, 如通式 (IC) 所示
Figure imgf000062_0002
其中, R-X选自 0 、 NH、 NCH3, NCH(CH3)2;
R6、 R7和 独立的选自 H、 OH 、 C N02、 d_4垸基、 C1-4烷氧基;
R13为单取代或多取代, 取代基独立的选自 H、 OH、 Cl、 N02、 C1-4烷基、 C1-4垸氧基、 CF3
OCH2C02H 、 甲酰基、 乙酰基、 C02H、 C02CH3、 C02Et、 (1",2"3",4"-四唑 -5")、 取代的苯羰
5、 根据权利要求 4所述的化合物, 其特征在于, 如通式 (IC1 ) 所示
Figure imgf000062_0003
ici
其中, R-X选自 O 、 NH、 NCH3, NCH(CH3)2;
R6、 R7和 独立的选自 H、 OH 、 CU N02、 CM垸基、 C1-4烷氧基;
RI4为单取代或多取代, 取代基独立的选自 H、 OH、 Cl、 N02、 C1-4烷基、 C1-4烷氧基、 CF3
OCH2C02H 、 甲酰基、 乙酰基、 C02H、 C02CH3、 C02Et。
6、 根据权利要求 1所述的化合物, 其特征在于, 如通式 (ID〉 所示
Figure imgf000063_0001
ID
其中, R-X选自 O NH NCH3, NCH(CH3)2;
R7和 R8独立的选自 H OH Cl N02 C1-4烷基、 .4烷氧基;
R15为单取代或多取代, 取代基独立的选自 H OH Cl N02 CM烷基、 d.4烷氧基、 CF3
OCH2CO2H 、 甲酰基、 乙酰基、 C02H C02C¾ C02Et (Γ',2 3",4,,-四唑 -5,,)。
7、 根据权利要求 1所述的化合物, 其特征在于, 如通式 (IE) 所示
Figure imgf000063_0002
其中, R-X选自 O 、 丽、 NCH3, NCH(CH3)2 ;
R6 R7和 独立的选自 H OH Cl N02 .4烷基、 C1-4垸氧基;
R16为单取代或多取代, 取代基独立的选自 H OH Cl N02 CM垸基、 C1-4垸氧基、 CF3
OCH2C02H 、 甲酰基、 乙酰基、 C02H C02C¾ C02Et (1",2',3,,,4,'-四唑-5")。
8、 根据权利要求 1所述的化合物, 其特征在于, 如通式 (IF) 所示
Figure imgf000063_0003
其中, R-X选自 0 NH NCH3, NCH(CH3)2;
R6 R7和 独立的选自 H OH Cl N02 CM烷基、 C1-4垸氧基;
R17为单取代或多取代, 取代基独立的选自 H OH Cl N02 CM垸基、 CM烷氧基、 CF3
OCH2C02H 、 甲酰基、 乙酰基、 C02H C02C¾ C02Et (1 ,2 3 ,4,,-四唑-5,,)。
9、 如权利要求 1所述的化合物, 其特征在于, 所述的化合物选自下列化合物的群组之一 化合物 W-R3
XR R6— Rs CH2N^ CH;h R,b CH2 QRd MP-C 编号 or Ri°or R' a or Rc
Figure imgf000064_0001
、 根据权利要求 1-9所述的化合物, 其特征在于, 该化合物还包括其药用盐、 水合物、 酯 或前体药物。
11、 制备如权利要求 1-10所述的化合物的方法, 其特征在于,
i) 用取代的 3-羧基香豆素、 与相应的取代胺类化合物缩合; 或
ii) 用各种取代的苯胺类化合物与氯代乙酰氯进行反应, 得到的中间体与三氯氧磷及二甲 基甲酰胺反应, 生成取代的 3-氯甲基喹啉, 然后其氯甲基与相应的各种取代吡咯类化 合物 1-位氮反应, 所得产物水解后得各种目的化合物。
12、 根据权利要求 11中任一制备方法, 其特征在于, 所述的反应所用的反应试剂包括三氯化 磷、 三氯氧磷、 五氯化磷、 二氯亚砜、 草酰氯、 乙酸酐、 1,二环己基碳化亚胺、 二吡啶碳酸 酯、 1,3-二异丙基碳酰亚胺、 1-(3-二甲胺丙基) -3-乙基碳酰亚胺、 乙酸酐、 三氯氧磷和二甲基 甲酰氨; 所用的催化剂包括三级胺、 吡啶、 4-二甲氨基吡啶和 4-吡咯烷基吡啶; 所用的有机 溶剂包括二甲基亚砜、 甲苯、 二氯甲垸、 乙二醇二甲醚, 1,2-二氯乙烷、 四氢呋喃和 Ν, Ν -二 甲基甲酰胺。
13、 一种药物组合物, 其特征在于, 含有药物有效剂量的如权利要求 1-10所述的任一化合物 及药用载体。
14、 根据权利要求 13的药物组合物, 其特征在于, 所述的药物组合物可以是片剂、 胶囊、 液 体口服液、 丸剂、 注射剂、 缓释制剂、 控释制剂及各种微粒给药系统。
15、 如权利要求 1-10任一化合物作为制备转化生长因子 - β lCTGF- β 1)抑制剂中的应用。
16、 如权利要求 1-10任一化合物作为制备血管紧张素 II (ΑΠ)转化酶受体拮抗剂中的应用。
17、 如权利要求 1-10任一化合物在制备治疗心脑血管疾患的药物中的应用。
18、 如权利要求 1-10任一化合物在制备治疗肾脏疾患的药物中的应用。
19、 如权利要求 1-10任一化合物在制备治疗糖尿病的药物中的应用。 0、 根据权利要求 13的应用, 其特征在于, 所述的心脑血管疾患是高血压、 心、 脑栓塞、 心 肌梗塞、 脑中风。 1、 如权利要求 1-10任一化合物在制备治疗肝硬化、 前列腺肥大的药物中的应用。
PCT/CN2006/000839 2006-04-28 2006-04-28 Dérivés de coumarine, leurs procédés de préparation, compositions pharmaceutiques et utilisations WO2007124617A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/CN2006/000839 WO2007124617A1 (fr) 2006-04-28 2006-04-28 Dérivés de coumarine, leurs procédés de préparation, compositions pharmaceutiques et utilisations

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2006/000839 WO2007124617A1 (fr) 2006-04-28 2006-04-28 Dérivés de coumarine, leurs procédés de préparation, compositions pharmaceutiques et utilisations

Publications (1)

Publication Number Publication Date
WO2007124617A1 true WO2007124617A1 (fr) 2007-11-08

Family

ID=38655032

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2006/000839 WO2007124617A1 (fr) 2006-04-28 2006-04-28 Dérivés de coumarine, leurs procédés de préparation, compositions pharmaceutiques et utilisations

Country Status (1)

Country Link
WO (1) WO2007124617A1 (zh)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009080821A3 (en) * 2007-12-21 2010-01-14 Giuliani International Limited Multitarget compounds active at a ppar and cannabinoid receptor
US20110021522A1 (en) * 2008-01-11 2011-01-27 The Regents Of The University Of California Activators of executioner procaspases 3, 6 and 7
JP2013543861A (ja) * 2010-11-16 2013-12-09 サントル ナショナル ドゥ ラ ルシェルシュ シアンティフィク キノリノン誘導体
CN107935976A (zh) * 2017-11-13 2018-04-20 山东大学 香豆素酰胺衍生物及其应用
WO2018174288A1 (ja) 2017-03-24 2018-09-27 大正製薬株式会社 2(1h)-キノリノン誘導体
CN110963988A (zh) * 2018-09-29 2020-04-07 泰州医药城国科化物生物医药科技有限公司 一种可作为gpr35受体激动剂的香豆素衍生物、制备方法及其应用
CN111533729A (zh) * 2020-05-07 2020-08-14 盐城工学院 一种四氮唑取代的喹啉酮衍生物及其制备方法与应用
CN113493433A (zh) * 2020-03-18 2021-10-12 泰州医药城国科化物生物医药科技有限公司 一种可作为gpr35受体激动剂的苯并吡喃酮衍生物、制备方法及其应用
CN114699405A (zh) * 2022-03-15 2022-07-05 四川轻化工大学 化合物在制备治疗非酒精性脂肪肝药物中的应用

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4547511A (en) * 1981-03-03 1985-10-15 Aktiebolaget Leo Heterocyclic carboxamides, compositions containing such compounds, processes for their preparation and methods of treatment therewith
WO1989007939A2 (en) * 1988-03-01 1989-09-08 The Upjohn Company Coumarins to inhibit reverse transcriptase in humans
WO2004050082A1 (fr) * 2002-12-05 2004-06-17 Institute Of Materia Medica, Chinese Academy Of Medical Sciences Nouveaux derives coumarine-amide et leur preparation, composition medicamenteuse associee et son utilisation
CN1199963C (zh) * 2000-07-03 2005-05-04 奥赖恩公司 具comt抑制活性的香豆素衍生物

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4547511A (en) * 1981-03-03 1985-10-15 Aktiebolaget Leo Heterocyclic carboxamides, compositions containing such compounds, processes for their preparation and methods of treatment therewith
WO1989007939A2 (en) * 1988-03-01 1989-09-08 The Upjohn Company Coumarins to inhibit reverse transcriptase in humans
CN1199963C (zh) * 2000-07-03 2005-05-04 奥赖恩公司 具comt抑制活性的香豆素衍生物
WO2004050082A1 (fr) * 2002-12-05 2004-06-17 Institute Of Materia Medica, Chinese Academy Of Medical Sciences Nouveaux derives coumarine-amide et leur preparation, composition medicamenteuse associee et son utilisation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DATABASE CA [online] 1984, "3-Quinolinecarboxamide, 1,2-dihydro-2-oxo-N-phenyl", Database accession no. (101:38327) *

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009080821A3 (en) * 2007-12-21 2010-01-14 Giuliani International Limited Multitarget compounds active at a ppar and cannabinoid receptor
JP2011506581A (ja) * 2007-12-21 2011-03-03 ジュリアーニ インターナショナル リミテッド Ppar及びカンナビノイド受容体において活性である多標的化合物
US20110021522A1 (en) * 2008-01-11 2011-01-27 The Regents Of The University Of California Activators of executioner procaspases 3, 6 and 7
US8642788B2 (en) * 2008-01-11 2014-02-04 The Regents Of The University Of California Activators of executioner procaspases 3, 6 and 7
JP2013543861A (ja) * 2010-11-16 2013-12-09 サントル ナショナル ドゥ ラ ルシェルシュ シアンティフィク キノリノン誘導体
WO2018174288A1 (ja) 2017-03-24 2018-09-27 大正製薬株式会社 2(1h)-キノリノン誘導体
KR20190133667A (ko) 2017-03-24 2019-12-03 다이쇼 세이야꾸 가부시끼가이샤 2(1h)-퀴놀리논 유도체
CN107935976A (zh) * 2017-11-13 2018-04-20 山东大学 香豆素酰胺衍生物及其应用
CN110963988A (zh) * 2018-09-29 2020-04-07 泰州医药城国科化物生物医药科技有限公司 一种可作为gpr35受体激动剂的香豆素衍生物、制备方法及其应用
CN110963988B (zh) * 2018-09-29 2023-08-22 泰州医药城国科化物生物医药科技有限公司 一种可作为gpr35受体激动剂的香豆素衍生物、制备方法及其应用
CN113493433A (zh) * 2020-03-18 2021-10-12 泰州医药城国科化物生物医药科技有限公司 一种可作为gpr35受体激动剂的苯并吡喃酮衍生物、制备方法及其应用
CN113493433B (zh) * 2020-03-18 2023-12-01 泰州医药城国科化物生物医药科技有限公司 一种可作为gpr35受体激动剂的苯并吡喃酮衍生物、制备方法及其应用
CN111533729A (zh) * 2020-05-07 2020-08-14 盐城工学院 一种四氮唑取代的喹啉酮衍生物及其制备方法与应用
CN111533729B (zh) * 2020-05-07 2022-07-26 盐城工学院 一种四氮唑取代的喹啉酮衍生物及其制备方法与应用
CN114699405A (zh) * 2022-03-15 2022-07-05 四川轻化工大学 化合物在制备治疗非酒精性脂肪肝药物中的应用

Similar Documents

Publication Publication Date Title
WO2007124617A1 (fr) Dérivés de coumarine, leurs procédés de préparation, compositions pharmaceutiques et utilisations
AU2016219906B2 (en) Fluorinated tetrahydronaphthyridinyl nonanoic acid derivatives and uses thereof
CN104546846B (zh) 1H‑吡唑并[3,4‑b]吡啶及其治疗应用
JP5755417B2 (ja) 肝細胞増殖因子(分散因子)活性のピラゾール誘導体モジュレーター
US10899750B2 (en) Methods and compositions of small molecule modulators of hepatocyte growth factor (scatter factor) activity
CN101678214B (zh) 芳基醚哒嗪酮衍生物
WO2003093238A1 (fr) Derives de quinoleine et derives de quinazoline inhibiteurs de l&#39;autophosphorylation du recepteur du facteur stimulant la proliferation des macrophages
CN105873913B (zh) 靶向截短的腺瘤性结肠息肉(apc)蛋白的治疗法
EA025393B1 (ru) Пиразолилмочевины в качестве ингибиторов киназы
US11834663B2 (en) Fibrotic treatment
CN101679401A (zh) 用于治疗肿瘤的作为met激酶抑制剂的2-氧代-3-苄基-苯并唑-2-酮衍生物及相关化合物
CN105753858A (zh) 杂环化合物及p27Kip1分解抑制剂
US7105503B2 (en) Pyrazole compounds
CN101528728A (zh) 6-1h-咪唑并-喹唑啉和喹啉衍生物,新颖的有效镇痛剂和抗炎剂
CN110946854A (zh) 超纯的四氢大麻酚-11-羧酸
TW200904409A (en) Azetidines
CA2771234A1 (en) Substituted hydrazide compounds and use thereof
CN102781914A (zh) 吲哚衍生物
JP2014509297A (ja) 肝細胞増殖因子(散乱因子)活性の小分子モジュレーターの使用方法
CN106661060B (zh) 菲罗啉膦酸类衍生物及其制备方法和应用
CN102046605B (zh) 作为酪氨酸激酶调节剂用于治疗肿瘤的二氢吡唑衍生物
CN108456165B (zh) 磺酰脲类化合物及其制备方法和应用
JP2000500138A (ja) 耐性腫瘍の治療方法
CN105998016A (zh) 吡非尼酮衍生物在制药中的应用

Legal Events

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

Ref document number: 06741755

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 06741755

Country of ref document: EP

Kind code of ref document: A1