CN111072665A - Cyclic-trans-4-L-hydroxyprolyl-L-serine-O-amino acid esters and salts thereof - Google Patents

Cyclic-trans-4-L-hydroxyprolyl-L-serine-O-amino acid esters and salts thereof Download PDF

Info

Publication number
CN111072665A
CN111072665A CN201811218423.5A CN201811218423A CN111072665A CN 111072665 A CN111072665 A CN 111072665A CN 201811218423 A CN201811218423 A CN 201811218423A CN 111072665 A CN111072665 A CN 111072665A
Authority
CN
China
Prior art keywords
compound
serine
trans
hydroxyprolyl
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN201811218423.5A
Other languages
Chinese (zh)
Other versions
CN111072665B (en
Inventor
何仲贵
姜琪坤
张天虹
孙进
王永军
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Suzhou Yutai Pharmaceutical Technology Co ltd
Zhejiang Shifangji Pharmaceutical Technology Co.,Ltd.
Original Assignee
Shenyang Pharmaceutical University
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 Shenyang Pharmaceutical University filed Critical Shenyang Pharmaceutical University
Priority to CN201811218423.5A priority Critical patent/CN111072665B/en
Publication of CN111072665A publication Critical patent/CN111072665A/en
Application granted granted Critical
Publication of CN111072665B publication Critical patent/CN111072665B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Abstract

The invention belongs to the technical field of medicine, and relates to cyclo-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester, pharmaceutically acceptable salts thereof and a preparation method thereof. The ring-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester prodrug and the pharmaceutically acceptable salt thereof have the structure shown in formula (I) or (II), wherein R is1And R2Is an amino acid residue. The compound can remarkably improve the intestinal membrane permeability of the cyclo-trans-4-L-hydroxyprolyl-L-serine and improve the bioavailability of the medicament.

Description

Cyclic-trans-4-L-hydroxyprolyl-L-serine-O-amino acid esters and salts thereof
Technical Field
The invention belongs to the technical field of medicine, and relates to cyclo-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester, pharmaceutically acceptable salts thereof and a preparation method thereof.
Background
The cyclo-trans-4-L-hydroxyprolyl-L-serine has a liver protection effect on autoimmune liver injury caused by canavalin A (Con A), and the action mechanism is as follows:
1. enhancing the free radical scavenging ability of liver tissues and inhibiting lipid peroxidation damage of liver cells;
2. attenuation of cytotoxic effects due to over-expression of NO;
3. reducing the accumulation of inflammatory cells in areas of inflammation and tissue damage;
4. inhibiting the release of inflammatory cytokines;
5. inhibiting hepatocyte apoptosis.
Because of the large polarity and poor membrane permeability of the small intestine of the cyclo-trans-4-L-hydroxyprolinyl-L-serine molecule, which results in low bioavailability (about 15%), a route to increase the membrane permeability of cyclo-trans-4-hydroxyprolinyl-L-serine, and thus its oral bioavailability, is needed. In recent years, specific transporters present on biological membranes of many tissues play a crucial role in mediating drug transport across membranes, and intestinal oligopeptide transporter (PEPT1) is studied more deeply as a target. The human oligopeptide transporter has two subtypes, PepT1 and PepT2, wherein PepT1 contains 708 amino acid residues and twelve transmembrane polypeptide chains, is mainly distributed in the brush border membrane of small intestine epithelial cells, and has increasing expression level in the direction from the proximal end to the distal end of the small intestine. According to the literature report, the drugs for improving the small intestinal membrane permeability based on the PepT1 target spot and further improving the oral bioavailability of the drugs are mainly concentrated on nucleosides, such as cytarabine, gemcitabine, fluorouracil and the like. There is no report in the prior art relating to the preparation of a prodrug from cyclo-trans-4-L-hydroxyprolinyl-L-serine.
Disclosure of Invention
The present invention aims to provide a cyclic-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester and a salt thereof, particularly to cyclic-trans-4-L-hydroxyprolyl-L-serine-3-CH2-O-amino acid ester prodrugs and ring-trans-4-L-hydroxyprolyl-L-serine-7-O-amino acid ester prodrugs and their pharmaceutically acceptable salts, and the use of said prodrugs for increasing the intestinal membrane permeability and bioavailability of ring-trans-4-L-hydroxyprolyl-L-serine.
The invention is realized by the following technical scheme:
a ring-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester prodrug having the structure:
Figure BDA0001834140370000021
wherein R is1And R2Is an amino acid residue;
the amino acid residues are selected from: l-alanyl, L-valyl, L-isoleucyl, L-phenylalanyl, L-glycyl, L-leucyl, L-tryptophyl, L-tyrosyl, L-histidyl, L-seryl, L-lysyl, L-arginyl, L-threonyl.
Wherein R is1Preferably L-valyl, L-isoleucyl, L-phenylalanyl, R2Preferably L-valyl is used as the starting material,
Figure BDA0001834140370000022
Cyclic-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester prodrugs and pharmaceutically acceptable salts thereof include cyclic-trans-4-L-hydroxyprolyl-L-serine-3-CH2-O-amino acid ester prodrug (I) or ring-trans-4-L-hydroxyprolyl-L-serine-7-O-amino acid ester prodrug (II) and their pharmaceutically acceptable salts.
The ring-trans-4-L-hydroxyprolyl-L-serine-3-CH2The general synthetic route for the-O-amino acid esters and pharmaceutically acceptable salts thereof is as follows:
Figure BDA0001834140370000023
wherein R is1Is an amino acid residue;
a: basic substances (sodium carbonate, sodium hydroxide, diethylamine, triethylamine, sodium bicarbonate, ammonia water), reaction solvents (dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane, N-dimethylhexanamide);
b: basic substances (diethylamine, triethylamine, sodium hydroxide, sodium bicarbonate, ammonia water), reaction solvents (dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane, N-dimethylhexanamide);
c: EDCI, HOBT, reaction solvent (dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane, N-dimethylhexanamide);
d, basic substances (diethylamine, triethylamine, sodium hydroxide, sodium bicarbonate and ammonia water) and reaction solvents (dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane and N, N-dimethyl hexanamide);
e: dicyclohexylcarbodiimide (DCC), 4-Dimethylaminopyridine (DMAP), reaction solvent (dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane, N-dimethylhexanamide); f: acids (hydrochloric acid, acetic acid, formic acid, maleic acid, lactic acid, carbonic acid, trifluoroacetic acid, phosphoric acid, p-toluenesulfonic acid)
The first step is as follows: dissolving a compound I, a compound II and an alkaline substance in a reaction solvent, filling nitrogen, reacting for 1-10 h, and performing rotary evaporation to obtain a compound III, wherein the alkaline substance comprises sodium carbonate, diethylamine, triethylamine, sodium hydroxide, sodium bicarbonate and ammonia water, the reaction solvent comprises dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane and N, N-dimethylhexanamide, the reaction temperature is 0-boiling point of the solvent, and the molar ratio of the compound I to the compound II is 0.1-10.
The second step is that: dissolving the compound III in a reaction reagent containing an alkaline substance, and reacting at room temperature for 1-10 h to obtain a compound IV; wherein the alkaline substance comprises diethylamine, triethylamine, sodium hydroxide, sodium bicarbonate and ammonia water, the reaction solvent comprises dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane and N, N-dimethylhexanamide, and the reaction temperature is 0 ℃ to the boiling point of the solvent.
The third step: dissolving a compound IV, a compound V, 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI) and 1-Hydroxybenzotriazole (HOBT) in a reaction solvent, introducing nitrogen, reacting for 1-10 h, and performing rotary evaporation to obtain a compound VI, wherein the reaction solvent comprises dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane and N, N-dimethylhexanamide, the reaction temperature is 0-the boiling point of the solvent, and the molar ratio of the compound IV to the compound V is 0.1-10.
The fourth step: dissolving the compound VI in a reaction reagent containing an alkaline substance, and reacting at room temperature for 1-10 h to obtain a compound VII; wherein the alkaline substance comprises diethylamine, triethylamine, sodium hydroxide, sodium bicarbonate and ammonia water, the reaction solvent comprises dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane and N, N-dimethylhexanamide, and the reaction temperature is 0 ℃ to the boiling point of the solvent.
The fifth step: dissolving a compound VII, N-tert-butyloxycarbonyl-amino acid, 1-ethyl- (3-dimethylaminopropyl) carbonyldiimine hydrochloride (EDCI) and 1-Hydroxybenzotriazole (HOBT) in a reaction solvent, filling nitrogen, reacting for 1-10 h, and performing rotary evaporation to obtain a compound IX, wherein the reaction solvent comprises dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane and N, N-dimethylhexanamide, the reaction temperature is 0-the boiling point of the solvent, and the molar ratio of the compound VII to the N-tert-butyloxycarbonyl-amino acid is 0.1-10.
And a sixth step: dissolving compound IX in different acids to obtain compound X, wherein the acids include hydrochloric acid, acetic acid, formic acid, maleic acid, lactic acid, carbonic acid, trifluoroacetic acid, phosphoric acid, and p-toluenesulfonic acid.
Wherein the N-tert-butyloxycarbonyl-amino acid in the fifth step is N-tert-butyloxycarbonyl-L-valine, N-tert-butyloxycarbonyl-L-isoleucine or N-tert-butyloxycarbonyl-L-phenylalanine, respectively.
The synthetic route for the cyclic-trans-4-L-hydroxyprolyl-L-serine-7-O-valine ester and the pharmaceutically acceptable salts thereof is as follows:
Figure BDA0001834140370000041
wherein R is2Is an amino acid residue;
a: 1-ethyl- (3-dimethylaminopropyl) carbonyldiimine hydrochloride (EDCI), 1-Hydroxybenzotriazole (HOBT), reaction solvents (dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane, N-dimethylhexanamide);
b: basic substances (diethylamine, triethylamine, sodium hydroxide, sodium bicarbonate, ammonia water), reaction solvents (dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane, N-dimethylhexanamide);
c: EDCI, HOBT, reaction solvent (dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane, N-dimethylhexanamide);
d: acid: hydrochloric acid, acetic acid, formic acid, maleic acid, lactic acid, carbonic acid, trifluoroacetic acid, phosphoric acid, p-toluenesulfonic acid
The first step is as follows: dissolving a compound I, a compound II and 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI) and 1-Hydroxybenzotriazole (HOBT) in a reaction solvent, introducing nitrogen, reacting at room temperature, and carrying out rotary evaporation to obtain a compound III, wherein the reaction solvent comprises dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane and N, N-dimethylhexanamide, the reaction temperature is 0 ℃ to the boiling point of the reaction solvent, and the molar ratio of the compound I to the compound II is 0.1-10.
The second step is that: dissolving the compound III in a reaction solvent containing an alkaline substance, and reacting at room temperature for 2h to obtain a compound IV; wherein the alkaline substance comprises diethylamine, triethylamine, sodium hydroxide, sodium bicarbonate and ammonia water, the reaction solvent comprises dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane and N, N-dimethylhexanamide, and the reaction temperature is 0 ℃ to the boiling point of the reaction solvent.
The third step: dissolving a compound IV, a compound V, EDCI and HOBT in a reaction solvent, introducing nitrogen, reacting at room temperature for 12h, and carrying out rotary evaporation to obtain a compound VI, wherein the reaction solvent comprises dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane and N, N-dimethylhexanamide, the reaction temperature is 0 ℃ to the boiling point of the reaction solvent, and the molar ratio of the compound IV to the compound V is 0.1-10.
The fourth step: dissolving compound VI in different acids to obtain compound VII, wherein the acids include hydrochloric acid, acetic acid, formic acid, maleic acid, lactic acid, carbonic acid, trifluoroacetic acid, phosphoric acid, and p-toluenesulfonic acid.
In the present invention, the structure of cyclo-trans-4-L-hydroxyprolyl-L-serine has two hydroxyl groups, i.e., a hydroxyl group at the 3-position methyl group and a hydroxyl group at the 7-position. Different amino acid types and steric hindrance near ester bonds have different influences on PEPT1 mediated transport, and the invention respectively connects the hydroxyl on the 3-methyl of the cyclo-trans-4-L-hydroxyprolinyl-L-serine with different amino acids through ester bonds or connects the hydroxyl on the 7-position with valine to form a peptide-like prodrug, thereby obviously improving the membrane permeability of the cyclo-trans-4-L-hydroxyprolinyl-L-serine and further improving the oral bioavailability.
The invention has the advantages that: the invention synthesizes the cyclic-trans-4-L-hydroxyprolyl-L-serine-3-CH for the first time2-O-amino acid ester and ring-trans-4-L-hydroxyprolyl-L-serine-7-O-amino acid ester prodrugs and pharmaceutically acceptable salts thereof, which significantly improve the small intestinal membrane permeability of ring-trans-4-L-hydroxyprolyl-L-serine, wherein ring-trans-4-L-hydroxyprolyl-L-serine-3-CH2The membrane permeability of the-O-valine ester is highest. When administered orally, cyclo-trans-4-L-hydroxyprolyl-L-serine-3-CH2After the-O-valine ester, the absolute bioavailability of the ring-trans-4-L-hydroxyprolyl-L-serine in rats is improved from 11.71 percent to 48.2 percent.
Description of the drawings:
FIG. 1 is a scheme showing cyclo-trans-4-L-hydroxyprolyl-L-serine-3-CH2Plasma concentration-time profile of (E) -O-valine ester and cyclo-trans-4-L-hydroxyprolyl-L-serine (n ═ 5)
a.
Figure BDA0001834140370000051
Oral compounds cyclo-trans-4-L-hydroxyprolyl-L-serine-3-CH2-O-valine ester (12mg/kg as ring-trans-4-L-hydroxyprolyl-L-serine)
b.
Figure BDA0001834140370000052
Oral administration of cyclo-trans-4-L-hydroxyprolyl-L-serine (12mg/kg)
c.
Figure BDA0001834140370000053
Intravenous injection of cyclo-trans-4-L-hydroxyprolyl-L-serine (6mg/kg)
The specific implementation mode is as follows:
according to the formula cyclo-trans-4-L-hydroxyprolyl-L-serine-3-CH2Synthesis routes for the-O-amino acid esters and the ring-trans-4-L-hydroxyprolyl-L-serine-7-O-valine esters, respectively, the following compounds are prepared.
Example 1:
the first step is as follows: and dissolving the compound I, the compound II and sodium carbonate in dichloromethane, introducing nitrogen, reacting at room temperature for 2 hours, and performing rotary evaporation to obtain a compound III.
The second step is that: dissolving the compound III in a tetrahydrofuran solution containing diethylamine, reacting for 2h at room temperature, evaporating the solvent under reduced pressure, diluting the residue with dichloromethane, sequentially washing with 5% phosphoric acid aqueous solution, aqueous solution and saturated salt solution, suction filtering, mixing the filtrate with silica gel, performing silica gel column chromatography, performing gradient elution with dichloromethane and methanol (100:1-1:100), collecting the column chromatography, and concentrating to obtain the compound IV.
The third step: dissolving the compound IV, the compound V, 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI) and 1-Hydroxybenzotriazole (HOBT) in dichloromethane, introducing nitrogen, reacting at room temperature for 2h, and evaporating the solvent under reduced pressure to obtain a compound VI.
The fourth step: dissolving the compound VI in a tetrahydrofuran solution containing diethylamine, reacting for 2h at room temperature, evaporating the solvent under reduced pressure, diluting the residue with dichloromethane, washing with a 5% phosphoric acid aqueous solution, an aqueous solution, a saturated sodium carbonate aqueous solution and a saturated salt solution in sequence, performing suction filtration, mixing the filtrate with silica gel, performing silica gel column chromatography, performing gradient elution on dichloromethane and methanol (100:1-1:100), collecting the column chromatography, and concentrating to obtain a compound VII.
The fifth step: dissolving a compound VII, N-tert-butyloxycarbonyl-L-valine, 1-ethyl- (3-dimethylaminopropyl) carbonyldiimine hydrochloride (EDCI) and 1-Hydroxybenzotriazole (HOBT) in dichloromethane, introducing nitrogen, reacting at room temperature for 2 hours, and performing rotary evaporation to obtain a compound IX.
And a sixth step: dissolving compound IX in small amount of ethanol, introducing hydrogen chloride gas for 5 hr under cooling in ice water bath, slowly dripping ethanol solution into glacial ethyl ether solution to precipitate white solid, vacuum filtering, and purifying with chloroformWashing with glacial ethyl ether, and oven drying to obtain cyclo-trans-4-L-hydroxyprolyl-L-serine-3-CH2-O-valine ester.
Example 2:
the first step is as follows: and dissolving the compound I, the compound II and sodium carbonate in dichloromethane, introducing nitrogen, reacting at room temperature for 2 hours, and performing rotary evaporation to obtain a compound III.
The second step is that: dissolving the compound III in a tetrahydrofuran solution containing diethylamine, reacting for 2h at room temperature, evaporating the solvent under reduced pressure, diluting the residue with dichloromethane, sequentially washing with a 5% phosphoric acid aqueous solution, an aqueous solution and saturated salt water, performing suction filtration, mixing the filtrate with silica gel, performing silica gel column chromatography, performing gradient elution with dichloromethane and methanol (100:1-1:100), collecting the column chromatography, and concentrating to obtain the compound IV.
The third step: dissolving the compound IV, the compound V and 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI) and 1-Hydroxybenzotriazole (HOBT) in dichloromethane, reacting for 2h at room temperature, and evaporating the solvent under reduced pressure to obtain a compound VI.
The fourth step: dissolving the compound VI in a tetrahydrofuran solution containing diethylamine, reacting for 2h at room temperature, evaporating the solvent under reduced pressure, diluting the residue with dichloromethane, sequentially washing with a 5% phosphoric acid aqueous solution, an aqueous solution, a saturated sodium carbonate aqueous solution and a saturated salt solution, performing suction filtration, mixing the filtrate with silica gel, performing silica gel column chromatography, performing gradient elution with dichloromethane and methanol (100:1-1:100), collecting the column chromatography, and concentrating to obtain the compound VII.
The fifth step: dissolving a compound VII, N-tert-butyloxycarbonyl-L-isoleucine, 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI) and 1-Hydroxybenzotriazole (HOBT) in dichloromethane, introducing nitrogen, reacting at room temperature for 2h, and performing rotary evaporation to obtain a compound IX.
And a sixth step: dissolving compound IX in small amount of ethanol, introducing hydrogen chloride gas for 5 hr under cooling in ice water bath, slowly dripping ethanol solution into glacial ethyl ether solution to precipitate white solid, vacuum filtering, washing with glacial ethyl ether, and oven drying to obtain cyclo-trans-4-L-hydroxyprolyl-L-serine-3-CH2-O-isoleucine ester.
Example 3:
the first step is as follows: and dissolving the compound I, the compound II and sodium carbonate in dichloromethane, introducing nitrogen, reacting at room temperature for 2 hours, and performing rotary evaporation to obtain a compound III.
The second step is that: dissolving the compound III in a tetrahydrofuran solution containing diethylamine, reacting for 2h at room temperature, evaporating the solvent under reduced pressure, diluting the residue with dichloromethane, sequentially washing with a 5% phosphoric acid aqueous solution, an aqueous solution and saturated salt water, performing suction filtration, mixing the filtrate with silica gel, performing silica gel column chromatography, performing gradient elution with dichloromethane and methanol (100:1-1:100), collecting the column chromatography, and concentrating to obtain the compound IV.
The third step: dissolving the compound IV, the compound V, 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI) and 1-Hydroxybenzotriazole (HOBT) in dichloromethane, introducing nitrogen, reacting at room temperature for 2h, and evaporating the solvent under reduced pressure to obtain a compound VI.
The fourth step: dissolving the compound VI in a tetrahydrofuran solution containing diethylamine, reacting for 2h at room temperature, evaporating the solvent under reduced pressure, diluting the residue with dichloromethane, sequentially washing with a 5% phosphoric acid aqueous solution, an aqueous solution, a saturated sodium carbonate aqueous solution and a saturated salt solution, performing suction filtration, mixing the filtrate with silica gel, performing silica gel column chromatography, performing gradient elution with dichloromethane and methanol (100:1-1:100), collecting the column chromatography, and concentrating to obtain the compound VII.
The fifth step: dissolving a compound VII, N-tert-butyloxycarbonyl-L-phenylalanine, 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI) and 1-Hydroxybenzotriazole (HOBT) in dichloromethane, introducing nitrogen, reacting at room temperature for 2h, and performing rotary evaporation to obtain a compound IX.
And a sixth step: dissolving compound IX in small amount of ethanol, introducing hydrogen chloride gas for 5 hr under cooling in ice water bath, slowly dripping ethanol solution into glacial ethyl ether solution to precipitate white solid, vacuum filtering, washing with glacial ethyl ether, and oven drying to obtain cyclo-trans-4-L-hydroxyprolyl-L-serine-3-CH2-O-phenylalanine ester.
Example 4
The first step is as follows: dissolving a compound I, a compound II, 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride (EDCI) and 1-Hydroxybenzotriazole (HOBT) in dichloromethane, introducing nitrogen, reacting at room temperature, and performing rotary evaporation to obtain a compound III;
the second step is that: dissolving the compound III in a tetrahydrofuran solution containing diethylamine, and reacting for 2h at room temperature to obtain a compound IV;
the third step: dissolving a compound IV, N-tert-butyloxycarbonyl-L-valine, EDCI and HOBT in dichloromethane, introducing nitrogen, reacting at room temperature for 12h, and performing rotary evaporation to obtain a compound VI;
the fourth step: dissolving the compound IX in a small amount of ethanol, introducing hydrogen chloride gas for 5 hours under the cooling of ice water bath, slowly dripping the ethanol solution into the glacial ethyl ether solution to precipitate white solid, carrying out suction filtration, washing with the glacial ethyl ether, and drying the obtained white powder to obtain the cyclo-trans-4-L-hydroxyprolyl-L-serine-7-O-valine ester.
The information identifying the ring-trans-4-L-hydroxyprolyl-L-serine and the compounds of examples 1 to 4 is shown in Table 1:
TABLE 1 information on cyclo-trans-4-L-hydroxyprolyl-L-serine and prodrugs thereof
Figure BDA0001834140370000081
Example 5: rat in vivo intestinal perfusion studies of cyclo-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester
By using rat in vivo and small intestine perfusion technology, 10cm rat jejunum is selected, two ends of the rat jejunum are inserted with tubes, the cyclo-trans-4-L-hydroxyprolinyl-L-serine and the compounds in examples 1 to 4 are respectively dissolved in Kreb-Ringers nutrient solution (pH 5.5) with the concentration of 0.5mM, and the drug-containing perfusion solution is passed through the rat jejunum at the flow rate of 0.2mL/min to obtain the membrane permeation rate of the cyclo-trans-4-L-hydroxyprolinyl-L-serine and the compounds in examples 1 to 4 in the jejunum.
TABLE 2 Membrane permeation rates for Ring-trans-4-L-hydroxyprolyl-L-serine and prodrugs thereof
Figure BDA0001834140370000091
Example 6: pharmacokinetic study in rats
From example 5, it can be seen that the compound of example 1 has the highest membrane permeability among the compounds of examples 1 to 4, and therefore the compound of example 1 and cyclo-trans-4-L-hydroxyprolyl-L-serine were subjected to in vivo pharmacokinetic studies.
Sprague-Dawley rats of the experimental group and the control group were gavaged with the compound of example 1 and an aqueous solution of cyclo-trans-4-L-hydroxyprolinyl-L-serine (each 12mg/kg as cyclo-trans-4-L-hydroxyprolinyl-L-serine) while intravenously injecting an aqueous solution of cyclo-trans-4-L-hydroxyprolinyl-L-serine (6mg/kg), respectively, and the plasma concentration of cyclo-trans-4-L-hydroxyprolinyl-L-serine was measured, and the results thereof are shown in Table 3 and FIG. 1.
TABLE 3 pharmacokinetic parameters of Ring-trans-4-L-hydroxyprolyl-L-serine in rats after oral administration of Ring-trans-4-L-hydroxyprolyl-L-serine and the Compound of example 1 (12mg/kg in Ring-trans-4-L-hydroxyprolyl-L-serine)
Figure BDA0001834140370000092
P <0.05 verses cyclo-trans-4-L-hydroxyprolyl-L-serine (p.o) as control
As can be seen from Table 3 and FIG. 1, the peak time after gavage of cyclo-trans-4-L-hydroxyprolyl-L-serine and the compound of example 1 was 1.4h and 0.56h, respectively, indicating that the absorption of the compound of example 1 was significantly accelerated and the area under the drug-time curve (AUC) of the compound of example 10-t) Significantly higher than cyclo-trans-4-L-hydroxyprolyl-L-serine; the oral bioavailability of cyclo-trans-4-L-hydroxyprolinyl-L-serine and its prodrug, calculated from the data for intravenous injection of cyclo-trans-4-L-hydroxyprolinyl-L-serine, was 11.71% and 48.2%, respectively, and the oral bioavailability of the compound of example 1 was significantly improved to achieve the intended purpose.

Claims (10)

1. A ring-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester prodrug having the structure:
Figure FDA0001834140360000011
wherein R is1And R2Is an amino acid residue.
2. The ring-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester prodrug of claim 1, and pharmaceutically acceptable salts thereof:
wherein R is1And R2Selected from: l-alanyl, L-valyl, L-isoleucyl, L-phenylalanyl, L-glycyl, L-leucyl, L-tryptophyl, L-tyrosyl, L-histidyl, L-seryl, L-lysyl, L-arginyl, L-threonyl;
wherein R is1Preferably L-valyl, L-isoleucyl, L-phenylalanyl, R2L-valyl is preferred.
3. The pro-drug of cyclo-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester and pharmaceutically acceptable salts thereof according to claim 1 or 2, wherein the pharmaceutically acceptable salts include hydrochloride, acetate, formate, maleate, lactate, carbonate, trifluoroacetate, phosphate, p-toluenesulfonate.
4. A process for the preparation of ring-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester prodrugs and pharmaceutically acceptable salts thereof according to claim 1, wherein: the preparation method of the formula (I) is as follows:
a: dissolving a compound I, a compound II and an alkaline substance in a reaction solvent, and reacting at room temperature to obtain a compound III;
b: dissolving the compound III in a reaction solvent containing an alkaline substance, and reacting to obtain a compound IV;
c: dissolving a compound IV, a compound V, 1-ethyl- (3-dimethylaminopropyl) carbonyldiimine hydrochloride and 1-hydroxybenzotriazole in a reaction solvent, and reacting to obtain a compound VI;
d: dissolving the compound VI in a reaction solvent containing an alkaline substance, and reacting to obtain a compound VII;
e: dissolving a compound VII, N-tert-butyloxycarbonyl-amino acid, dicyclohexylcarbodiimide and 4-dimethylaminopyridine in a reaction solvent, and reacting to obtain a compound IX;
f: dissolving the compound IX in acid, and reacting to obtain a compound X;
Figure FDA0001834140360000021
5. a process for the preparation of ring-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester prodrugs and pharmaceutically acceptable salts thereof according to claim 1, wherein: the preparation method of the formula (II) is as follows:
a: dissolving a compound I, a compound II, 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride and 1-hydroxybenzotriazole in a reaction solvent, and reacting at room temperature to obtain a compound III;
b: dissolving the compound III in a reaction solution containing an alkaline substance, and reacting at room temperature to obtain a compound IV;
c: dissolving the compound IV, the compound V, EDCI and HOBT in a reaction solvent, and reacting at room temperature to obtain a compound VI;
d: dissolving the compound VI in different acids, and reacting to obtain a compound VII;
Figure FDA0001834140360000022
6. a process for the preparation of a ring-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester prodrug and a pharmaceutically acceptable salt thereof according to claim 4 or 5, wherein the reaction solvent described in steps a, b, c, d, e of claim 4 and a, b, c of claim 5 is dichloromethane, N-dimethylformamide, tetrahydrofuran, dioxane or N, N-dimethylhexanamide; the basic substance in step a, b, d or step b of claim 5 is diethylamine, triethylamine, sodium hydroxide, sodium bicarbonate, ammonia water or sodium carbonate.
7. A process for the preparation of a ring-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester prodrug and a pharmaceutically acceptable salt thereof according to claim 4 or 5, wherein the acid in step f of claim 4 and step d of claim 5 is hydrochloric acid, acetic acid, formic acid, maleic acid, lactic acid, carbonic acid, trifluoroacetic acid, phosphoric acid or p-toluenesulfonic acid.
8. A pharmaceutical composition comprising a ring-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester prodrug of any one of claims 1-3 and pharmaceutically acceptable salts thereof.
9. Use of a ring-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester prodrug as claimed in any one of claims 1 to 3 and pharmaceutically acceptable salts thereof or a pharmaceutical composition as claimed in claim 8 for the manufacture of a medicament for the treatment of autoimmune hepatitis.
10. Use of a ring-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester prodrug as claimed in any one of claims 1 to 3 and pharmaceutically acceptable salts thereof or a pharmaceutical composition as claimed in claim 8 for the manufacture of a medicament for improving the intestinal membrane permeability and bioavailability of ring-trans-4-L-hydroxyprolyl-L-serine.
CN201811218423.5A 2018-10-19 2018-10-19 Cyclic-trans-4-L-hydroxyprolyl-L-serine-O-amino acid esters and salts thereof Active CN111072665B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201811218423.5A CN111072665B (en) 2018-10-19 2018-10-19 Cyclic-trans-4-L-hydroxyprolyl-L-serine-O-amino acid esters and salts thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201811218423.5A CN111072665B (en) 2018-10-19 2018-10-19 Cyclic-trans-4-L-hydroxyprolyl-L-serine-O-amino acid esters and salts thereof

Publications (2)

Publication Number Publication Date
CN111072665A true CN111072665A (en) 2020-04-28
CN111072665B CN111072665B (en) 2021-02-23

Family

ID=70308112

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201811218423.5A Active CN111072665B (en) 2018-10-19 2018-10-19 Cyclic-trans-4-L-hydroxyprolyl-L-serine-O-amino acid esters and salts thereof

Country Status (1)

Country Link
CN (1) CN111072665B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113999238A (en) * 2021-11-08 2022-02-01 上海高准医药有限公司 Preparation method of cyclic-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester hydrochloride
WO2023064991A1 (en) * 2021-10-22 2023-04-27 Neuren Pharmaceuticals Limited Process for preparing bicyclic glycine-proline compounds and monocyclic glycine-proline intermediates thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102552187A (en) * 2012-02-28 2012-07-11 大连医科大学 Oral hydroxyl silk peptide nanoparticles and preparation method thereof
CN102920653A (en) * 2012-10-22 2013-02-13 沈阳药科大学 Cyclo-trans-4-L-hydroxy prolyl-L-serine injection and preparation method thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102552187A (en) * 2012-02-28 2012-07-11 大连医科大学 Oral hydroxyl silk peptide nanoparticles and preparation method thereof
CN102920653A (en) * 2012-10-22 2013-02-13 沈阳药科大学 Cyclo-trans-4-L-hydroxy prolyl-L-serine injection and preparation method thereof

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023064991A1 (en) * 2021-10-22 2023-04-27 Neuren Pharmaceuticals Limited Process for preparing bicyclic glycine-proline compounds and monocyclic glycine-proline intermediates thereof
CN113999238A (en) * 2021-11-08 2022-02-01 上海高准医药有限公司 Preparation method of cyclic-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester hydrochloride
CN113999238B (en) * 2021-11-08 2022-12-16 上海高准医药有限公司 Preparation method of cyclic-trans-4-L-hydroxyprolyl-L-serine-O-amino acid ester hydrochloride

Also Published As

Publication number Publication date
CN111072665B (en) 2021-02-23

Similar Documents

Publication Publication Date Title
ES2408216T3 (en) Composition for proteasome inhibition
EP2641605B1 (en) Polymer derivative of cytidine metabolism antagonist
US7498035B2 (en) Compounds of hydrophilic polymer-polycarboxyl oligopeptide and medicines, medical composite comprising above compound and use of above compound in medicines
RU2321412C2 (en) Novel solid preparation comprising block-copolymer and anthracycline antitumor preparation and method for its preparing
JP4757633B2 (en) Micellar preparation containing poorly water-soluble anticancer agent and novel block copolymer
ES2659778T3 (en) Polyethylene glycol-amino acid-irinotecan pharmaceutical conjugate and pharmaceutical composition thereof
US11180615B2 (en) Cationic polyphosphazene compound, polyphosphazenes-drug conjugate compound and method for preparing same
CN111072665B (en) Cyclic-trans-4-L-hydroxyprolyl-L-serine-O-amino acid esters and salts thereof
WO2005092898A1 (en) The conjugates of a hydrophilic polymer-tripterygium&#39;s extracts and the pharmaceutical compositions thereof
CN104448295B (en) The rapamycin derivative of polyethylene glycol multijaw oligopeptides bonding
WO2018144991A1 (en) Self-assembled gels formed with anti-retroviral drugs, prodrugs thereof, and pharmaceutical uses thereof
AU744425B2 (en) 20(s) camptothecin glycoconjugates
JP6358797B2 (en) Use of umirolimus and its derivatives to treat cancer
US20050220753A1 (en) Hydrophilic polymers-flavoids conjugates and pharmaceutical compositions comprising them
CN110156736B (en) Daidzein carbamate prodrug, salt thereof, preparation method and application thereof
EP3378494A1 (en) Polymeric derivative of macrolide immunosuppressant
CN113332305B (en) Lipid composition of cytidine compound
CN103502214A (en) Prodrugs of D-isoglutamyl-[D/L]-tryptophan
CN101891801B (en) Saturated aliphatic chain alcohol RGD (Arginyl-Glycyl-Aspartic acid) pentapeptide diester compound, synthesis method and application thereof
CN115960162A (en) Membrane-disturbed anti-tumor nano lipopeptide and preparation method and application thereof
CN115109258A (en) 7-ethyl-10-hydroxycamptothecin polymer, preparation method and application thereof
CN102188716B (en) Macromolecule vinblastine conjugate adopting amino acids or oligopeptides as connexons
CN113827735A (en) Multi-targeting drug carrier loaded with SN-38 and application thereof
JPH0820533A (en) Bacterial shock-treating agent

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20201104

Address after: Unit e347, A1 / F, biomedical industrial park, 218 Xinghu street, Suzhou Industrial Park, Suzhou area, China (Jiangsu) pilot Free Trade Zone, Suzhou City, Jiangsu Province

Applicant after: Suzhou Yutai Pharmaceutical Technology Co.,Ltd.

Address before: 110016 No. 103, Wenhua Road, Shenhe District, Liaoning, Shenyang

Applicant before: SHENYANG PHARMACEUTICAL University

GR01 Patent grant
GR01 Patent grant
TR01 Transfer of patent right
TR01 Transfer of patent right

Effective date of registration: 20230105

Address after: 215125 Unit 604, Building B1, Biomedical Industrial Park, No. 218, Xinghu Street, Suzhou Industrial Park, China (Jiangsu) Pilot Free Trade Zone, Suzhou, Jiangsu Province

Patentee after: Suzhou Yutai Pharmaceutical Technology Co.,Ltd.

Patentee after: Zhejiang Shifangji Pharmaceutical Technology Co.,Ltd.

Address before: 215000 unit e347, A1 / F, biomedical industrial park, 218 Xinghu street, Suzhou Industrial Park, Suzhou area, China (Jiangsu) pilot Free Trade Zone, Suzhou City, Jiangsu Province

Patentee before: Suzhou Yutai Pharmaceutical Technology Co.,Ltd.