CN115340472A - Glutamic acid derivative and synthetic method and application thereof - Google Patents

Glutamic acid derivative and synthetic method and application thereof Download PDF

Info

Publication number
CN115340472A
CN115340472A CN202211135753.4A CN202211135753A CN115340472A CN 115340472 A CN115340472 A CN 115340472A CN 202211135753 A CN202211135753 A CN 202211135753A CN 115340472 A CN115340472 A CN 115340472A
Authority
CN
China
Prior art keywords
glutamic acid
acid derivative
tert
synthesis
reaction
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.)
Pending
Application number
CN202211135753.4A
Other languages
Chinese (zh)
Inventor
李宜明
王容天
王玉
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.)
Hefei University of Technology
Original Assignee
Hefei University of Technology
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 Hefei University of Technology filed Critical Hefei University of Technology
Priority to CN202211135753.4A priority Critical patent/CN115340472A/en
Publication of CN115340472A publication Critical patent/CN115340472A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/10Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C271/22Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms to carbon atoms of hydrocarbon radicals substituted by carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C269/00Preparation of derivatives of carbamic acid, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C269/06Preparation of derivatives of carbamic acid, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups by reactions not involving the formation of carbamate groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/04General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length on carriers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/06General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length using protecting groups or activating agents
    • C07K1/061General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length using protecting groups or activating agents using protecting groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/02Ortho- or ortho- and peri-condensed systems
    • C07C2603/04Ortho- or ortho- and peri-condensed systems containing three rings
    • C07C2603/06Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members
    • C07C2603/10Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members containing five-membered rings
    • C07C2603/12Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members containing five-membered rings only one five-membered ring
    • C07C2603/18Fluorenes; Hydrogenated fluorenes
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention discloses a glutamic acid derivative and a synthesis method and application thereof, wherein the molecular structure of the glutamic acid derivative is as follows:
Figure DDA0003851432490000011
the method comprises the steps of firstly carrying out substitution reaction on commercially purchased N- (9-fluorenylmethoxycarbonyl) -D-glutamic acid 1-tert-butyl ester and alpha-bromo-4-methoxyacetophenone, and then removing the protection of glutamic acid main chain carboxylThe glutamic acid derivative with photosensitive group on the side chain can be obtained by tert-butyl ester. The glutamic acid derivative can be compatible with polypeptide solid phase synthesis and used for preparing and producing various polypeptides needing light-operated protection groups.

Description

Glutamic acid derivative and synthetic method and application thereof
Technical Field
The invention relates to a glutamic acid derivative with a photosensitive p-methoxybenzoyl group on a side chain and a synthesis method and application thereof, which are compatible with solid phase synthesis, and belong to the technical field of protein synthesis.
Background
Photocage peptides and proteins, whose biological functions are inhibited by photocleavable protecting groups, can regain their activity under light irradiation. These compounds are widely used to detect and elucidate complex biological processes because the illumination can be precisely controlled in space, time and amplitude in a non-invasive manner. Side chain cage forms of serine, threonine, tyrosine, cysteine, lysine, arginine, glutamine, aspartic acid, and glutamic acid (Glu) have been reported. Among these functions, carboxylic acid groups play an important role in protein activity and in protein interactions with other biological macromolecules or small molecules. Peptide and protein derivatives capped with carboxylic acid moieties have been developed to study biochemical mechanisms and protein-protein/ligand interactions.
For example, an article (J.Am.chem.Soc.1991, 113, 2758-2760.) reports a method for triggering an enzymatic reaction using photoactivatable bacteriophage T4 lysozyme containing aspartyl b-nitrobenzyl ester in the active site Asp 20. Similarly, an article (J.Am.chem.Soc.2013, 135, 4580-4583.) reports the introduction of a photolabile blocking group into the basic C-terminal carboxylate of a PDZ domain ligand to exploit synaptic PDZ domain-mediated interactions.
Generally, caged peptides are synthesized by standard Solid Phase Peptide Synthesis (SPPS) methods, which provides the greatest flexibility in designing caged peptides and protein derivatives. The article (J.biomed.Mater.Res.PartA 2013,101A, 787-796) produced a photoactivatable peptide containing "RGD" by modifying the carboxylic acid of Asp with 2-nitrobenzyl (2-NB). However, the photochemical and photophysical properties of 2-NB limit its further use in biological processes. The article (Peptides 2007,28,1074-1082.) reported 4,5-Dimethoxynitrobenzyl (DMNB) has improved photochemical properties, but failed to incorporate into the side chain carboxylic acid during Fmoc SPPS due to side reactions of Asp (ODMNB) and Glu (ODMNB) to form asparagine and pyrrolidone. The article (Tetrahedron lett.,56 (2015), pp.4582-4585) reports a method for the synthesis of photocage peptides by modification of the derivative Asp and Glu with 4-methoxy-7-nitroindoline (MNI). MNI cages have excellent photochemical properties and fast photolysis kinetics, but the synthetic steps are cumbersome.
Thus, there remains a need for new Fmoc-compatible methods to produce side chain carboxy-blocked peptides/proteins with good photochemical properties simply and quickly.
Disclosure of Invention
Aiming at the defects of the prior art, the invention provides a glutamic acid derivative with a photosensitive p-methoxybenzoyl group on a side chain and a synthesis method and application thereof, wherein the glutamic acid derivative is compatible with solid phase synthesis.
The invention firstly carries out substitution reaction on commercially purchased N- (9-fluorenylmethoxycarbonyl) -D-glutamic acid 1-tert-butyl ester and alpha-bromo-4-methoxyacetophenone, and then removes the glutamic acid main chain carboxyl protecting group tert-butyl ester to obtain the glutamic acid derivative with the photosensitive group on the side chain. The glutamic acid derivative can be compatible with polypeptide solid phase synthesis and used for preparing and producing various polypeptides needing light-operated protection groups.
The glutamic acid derivative is abbreviated as Fmoc-Glu (cage) -OH, and the structural formula of the glutamic acid derivative is shown as follows:
Figure BDA0003851432470000021
the invention firstly carries out substitution reaction on commercially purchased N- (9-fluorenylmethoxycarbonyl) -D-glutamic acid 1-tert-butyl ester and alpha-bromo-4-methoxyacetophenone, and then removes the glutamic acid main chain carboxyl protecting group tert-butyl ester to obtain the glutamic acid derivative with a photosensitive group on a side chain. The synthetic route is as follows:
Figure BDA0003851432470000022
the invention relates to a synthesis method of a glutamic acid derivative Fmoc-Glu (cage) -OH, which comprises the following steps:
step 1: synthesis of Compound I
After the reaction was completed by dissolving commercially available 1-tert-butyl N- (9-fluorenylmethoxycarbonyl) -D-glutamate and alpha-bromo-4-methoxyacetophenone in dry Dichloromethane (DCM) and adding solid particles of potassium carbonate under an argon atmosphere, stirring at 40 ℃ for 24 hours, concentrating under vacuum and isolating by column chromatography compound I, 1- (tert-butyl) 5- (2- (4-methoxyphenyl) -2-oxoethyl) ((9H-fluoren-9-yl) methoxy) carbonyl) -L-glutamic acid.
In the step 1, the molar ratio of the N- (9-fluorenylmethoxycarbonyl) -D-glutamic acid 1-tert-butyl ester, the alpha-bromo-4-methoxyacetophenone and the potassium carbonate is 1:1.2:1.5.
step 2: synthesis of target product
Dissolving a compound I in Dichloromethane (DCM), placing in an ice bath, stirring, cooling to 0 ℃, then slowly dropwise adding a trifluoroacetic acid (TFA) solution into a reaction system, reacting for 1 hour, removing the ice bath, recovering the room temperature, and continuing stirring and reacting for 6 hours; after the reaction is finished, the solvent is removed by vacuum concentration, and the pure target product, namely- (S) -2- ((((((9H-fluoren-9-yl) methoxyl) carbonyl) amino) -5- (2- (4-methoxyphenyl) -2-oxoethoxy) -5-oxovaleric acid is obtained.
In step 2, the molar ratio of compound I to trifluoroacetic acid (TFA) is 1:60.
in step 2, the volume ratio of Dichloromethane (DCM) to trifluoroacetic acid (TFA) in the reaction system is 1:1.
the glutamic acid derivative is used as a special protective amino acid, and is connected to the polypeptide by a common Fmoc (Fmoc) solid-phase polypeptide synthesis method, so that various polypeptides requiring light-operated protective groups are obtained.
The reaction process is schematically shown as follows:
Figure BDA0003851432470000031
the invention has the following beneficial effects:
the invention discloses a glutamic acid derivative with a photosensitive p-methoxybenzoyl group on a side chain, a synthesis method and application thereof.
Drawings
FIG. 1 is the compound I-1- (tert-butyl) 5- (2- (4-methoxyphenyl) -2-oxoethyl) (((9H-fluoren-9-yl) methoxy) carbonyl) -L-glutamic acid hydrogen spectrum-CDCl 3 (Compound I) hydrogen spectrum of (C).
Figure 2 is a hydrogen spectrum of the compound (S) -2- ((((((9H-fluoren-9-yl) methoxy) carbonyl) amino) -5- (2- (4-methoxyphenyl) -2-oxoethoxy) -5-oxopentanoic acid (Fmoc-Glu (cage) -OH).
FIG. 3 is a carbon spectrum of the compound (S) -2- ((((((9H-fluoren-9-yl) methoxy) carbonyl) amino) -5- (2- (4-methoxyphenyl) -2-oxoethoxy) -5-oxopentanoic acid (Fmoc-Glu (cage) -OH).
FIG. 4 is the linear polypeptide ARKGKIKPKA-NH prior to attachment of Glu (cage) molecule 2 The high performance liquid chromatogram of (1).
FIG. 5 is a mass spectrum of the linear polypeptide ARKGKIKPKA prior to attachment of a Glu (cage) molecule.
FIG. 6 is a high performance liquid chromatogram of the linear polypeptide GLE (cage) ARKGKIKPKA following ligation of a Glu (cage) molecule.
FIG. 7 is a mass spectrum of the linear polypeptide GLE (cage) ARKGKIKPKA following ligation of a Glu (cage) molecule.
FIG. 8 is a high performance liquid chromatogram of solid crude peptide SKGLE (cage) ARKGKIKPKA with photo-controlled molecules.
FIG. 9 is a mass spectrum of solid crude peptide SKGLE (cage) ARKGKIKPKA with photo-controlled molecules.
FIG. 10 is a high performance liquid chromatogram of Photocaged peptide SKGLEARKGKIKPKA.
FIG. 11 is a mass spectrum of Photodelocalized peptide SKGLEARKGKIKPKA.
FIG. 12 is a high performance liquid chromatogram of solid crude peptide AE (cage) FGLKLDRIG with photo-control molecules.
FIG. 13 is a mass spectrum of solid crude peptide AE (cage) FGLKLDRIG with photo-controlled molecules.
FIG. 14 is a high performance liquid chromatogram of photodegainted peptide AEFGLKLDRIG.
FIG. 15 is a mass spectrum of Photodelocalized peptide AEFGLKLDRIG.
FIG. 16 is a comparison of HPLC chromatograms of solid crude peptide AE (cage) FGLKLDRIG before and after removal of the photo control molecule.
Detailed Description
For the purpose of facilitating an understanding of the present invention, reference will now be made to the following examples, which are provided to illustrate the features and advantages of the present invention, and are not intended to limit the scope of the appended claims.
Example 1:
1. commercially available 1-tert-butyl N- (9-fluorenylmethoxycarbonyl) -D-glutamate (852mg, 1.74mmol) and alpha-bromo-4-methoxyacetophenone (474mg, 2.08mmol) were dissolved in 10mL of dried Dichloromethane (DCM) and solid particles of potassium carbonate (312mg, 2.26mmol) were added simultaneously, the reaction was left under argon atmosphere and stirred at 40 ℃ for 24 hours, after completion of the reaction, the reaction solution was concentrated in vacuo and isolated by column chromatography to give compound I, 1- (tert-butyl) 5- (2- (4-methoxyphenyl) -2-oxoethyl) ((((9H-fluoren-9-yl) methoxy) carbonyl) -L-glutamate (897.6mg, 1.57mmol) in 90% yield.
2. Dissolving the compound I (897.6mg, 1.57mmol) in 7mL of Dichloromethane (DCM), placing in an ice bath, stirring, cooling to 0 ℃, slowly dropwise adding 7mL of trifluoroacetic acid (TFA) solution into the reaction system, reacting for 1 hour, removing the ice bath, recovering the room temperature, and continuing stirring for 6 hours. After the reaction was completed, the solvent was removed by vacuum concentration to obtain pure target product, i.e., (S) -2- ((((((9H-fluoren-9-yl) methoxy) carbonyl) amino) -5- (2- (4-methoxyphenyl) -2-oxoethoxy) -5-oxopentanoic acid (Fmoc-Glu (cage) -OH) (731mg, 1.42mmol) in 90% yield.
Example 2:
263.15mg (0.1 mmol) of amino resin with a degree of substitution of 0.38mmol/g was weighed, and 10ml of N, N-Dimethylformamide (DMF)/dichloromethane solution (DCM) was added to the resin to swell the resin, and the volume ratio of the added solution was: DCM =1:1, swelling time is 10 minutes, a diaphragm pump is used as a power source, and a swelled product is dried by pumping to obtain a swelled resin.
20% piperidine was added to the resin for 10 minutes to completely remove the Fmoc protecting group of the amino group on the resin, and then washed three times with DMF, DCM, DMF to remove the remaining piperidine after the reaction and the protecting group of the small molecule removed.
Dissolving the first amino acid Fmoc-Ala-OH (124.53mg, 0.4mmol, 4eq.) and the condensing agent 6-chlorobenzotriazole-1,1,3,3-tetramethylurea hexafluorophosphate (HCTU, 157.20mg,0.38mmol, 3.8eq.) in 4ml DMF, adding N, N-diisopropylethylamine (DIEA, 132. Mu.l, 0.8mmol, 8eq.) to activate for 1 min, adding into the amino resin, and placing in a shaking table and shaking at normal temperature for 30 min; after the reaction is finished, washing the mixture with DMF, DCM and DMF three times respectively, adding Fmoc-Ala-OH (124.53mg, 0.4mmol, 4eq.), 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate (HATU, 144.49mg,0.38mmol, 3.8eq.), N-hydroxy-7-azobenzotriazol (HOAt, 51.60mg,0.38mmol, 3.8eq), DIEA (132 mu.l, 0.8mmol, 8eq) into DMF, and reacting for 30 minutes; after the reaction is finished, the mixture is washed by DMF, DCM and DMF three times respectively, DMF solution containing 20% (volume fraction) piperidine is added into the resin, the mixture is washed by DMF, DCM and DMF three times respectively after 5 minutes of reaction, 20% piperidine is added into the resin again for reaction for 10 minutes so as to remove the Fmoc protecting group of the amino group on the resin completely, the mixture is washed by DMF, DCM and DMF three times respectively so as to remove the residual piperidine and the detached micromolecule protecting group after the reaction, and the resin is washed by DMF, DCM and DMF respectively. The condensation of the latter amino acids was carried out analogously until all sequences completed the solid phase condensation, finally obtaining linear Fmoc-S (tBu) K (Boc) GLE (cage) AR (Pbf) K (Boc) GK (Boc) IK (Boc) PK (Boc) A-NH 2 And (3) resin. The Fmoc protecting group at the N-terminus was removed in two steps with 20% piperidine in DMF, washed three times with DMF, DCM, DMF, DCM and then the residual DCM solvent was removed from the resin under reduced pressure to give a dry resin.
Example 3:
to the resin obtained in example 2, 10ml of a prepared cleavage reagent (a mixture of trifluoroacetic acid, phenol, water, and triisopropylsilane in a volume ratio of trifluoroacetic acid: water: phenol: triisopropylsilane = 5) was added, and the mixture was reacted at room temperature for 2.5 hours, and then the polypeptide chain was cleaved from the resin, and the filtrate was collected into a centrifuge tube, and the cleavage solution was concentrated by a nitrogen bubbling method. Finally, after the solution to be cut is concentrated to less than 5ml, 30ml of ethyl acetate is added for sedimentation, and a low-speed centrifuge (4500 rpm) is used for centrifugation, so that the crude peptide is settled to the bottom. After removal of the supernatant, glacial ethyl ether was added again, sonicated until a crude peptide suspension was formed, the cleaved small molecule impurities were dissolved in glacial ethyl ether and finally removed by centrifugation. After the two times of reaction, the solid sediment is placed in a shade place for air drying, and 150mg of solid crude peptide (SKGLE (cage) ARKGKIKPKA) with photo-controlled molecules is obtained.
A small amount of the crude peptide was dissolved in a pure water solution, and the solution was subjected to reverse phase high performance liquid chromatography (RP-HPLC) after passing through a membrane. The analytical gradient is 10% -70% acetonitrile concentration for 30min. ESI-MS identification of the main peak after chromatographic analysis verifies the correctness of the solid crude peptide. And (3) after the solid crude peptide is verified to be correct, separating and purifying the solid crude peptide by using a C18 semi-preparative column (the semi-preparative gradient is 10-70% of acetonitrile concentration, and the time is 30 min), collecting a correct product peak solution, and freeze-drying the correct product peak solution in a freeze-dryer to obtain a white flocculent SKGLE (cage) ARKGKIKPKA pure peptide product (112 mg).
Example 4:
the SKGLE (cage) ARKGKIKPKA pure peptide product obtained in example 3 is selected as the target short peptide for further light cage removal experiment. 1mgSKGLE (cage) ARKGKIKPKA pure peptide product was dissolved in 200uL buffer (6 MGn-HCl,200mM NaH) 2 PO 4 pH = 7.0), placed in an ultraviolet crosslinking chamber, on a flat ice surface, irradiated with 365nm light for 5min. The reaction was monitored by RP-HPLC. The gradient was analyzed for 30min at 10% -70% acetonitrile concentration. ESI-MS identification of the main peak after chromatographic analysis verifies the correctness of the product. The results show that SKGLE (cage) ARKGKIKPKA pure peptide (CagedForm) is mostly converted into SKGLEARKGKIKPKA (NativeForm), and prove that the side chain photosensitive protecting group of Fmoc-Glu (cage) -OH molecule can be well removed under 254nm illumination condition, and has good photocaging efficiency (see figure 10 and figure 11).
Example 5:
weighing 312.5mg (0.1 mmol) of chlorine resin with a substitution degree of 0.32mmol/g, adding 10ml of N, N-Dimethylformamide (DMF) solution into the resin to swell the resin for 20 minutes, and pumping the swelled product by using a diaphragm pump as a power source to obtain the swelled resin.
Dissolving a first amino acid Fmoc-Gly-OH (118.92mg, 0.4mmol, 4eq.) in 4ml of DMF, adding N, N-diisopropylethylamine (DIEA, 132 mu l,0.8mmol, 8eq.) into the amino resin after activation for 1 minute, and placing the amino resin in a shaker for shaking at normal temperature for 12 hours; after the reaction, the mixture was washed with DMF, DCM and DMF three times, DMF solution containing 20% (volume fraction) piperidine was added to the resin, the mixture was washed with DMF, DCM and DMF three times after 5 minutes, and 20% piperidine was added to the resin again and reacted for 10 minutes to completely remove the Fmoc protecting group of the amino group on the resin. After the reaction, DMF, DCM and DMF are used for washing for three times respectively, and then Fmoc-Ile-OH (141.36mg, 0.4mmol, 4eq.) and 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate (HATU, 144.49mg,0.38mmol, 3.8eq.) and N-hydroxy-7-azobenzotriazol (HOAt, 51.60mg,0.38mmol, 3.8eq.) and DIEA (132 mu.l, 0.8mmol, 8eq.) are added into DMF for reaction for 30 minutes; the subsequent condensation of amino acids was carried out analogously until all sequences completed the solid phase condensation, finally obtaining linear Fmoc-AE (cage) FGLK (Boc) LD (OtBu) R (Pbf) IG-COOH resins. The Fmoc protecting group at the N-terminus was removed in two steps with 20% piperidine in DMF, washed three times with DMF, DCM, DMF, DCM and then the residual DCM solvent was removed from the resin under reduced pressure to give a dry resin.
Example 6:
to the resin obtained in example 5, 10ml of a prepared cleavage reagent (a mixture of trifluoroacetic acid, phenol, water, and triisopropylsilane in a volume ratio of trifluoroacetic acid: water: phenol: triisopropylsilane = 5) was added, and the mixture was reacted at room temperature for 2.5 hours, and then the polypeptide chain was cleaved from the resin, and the filtrate was collected into a centrifuge tube, and the cleavage solution was concentrated by a nitrogen bubbling method. Finally, after the solution to be cut is concentrated to less than 5ml, 30ml of ethyl acetate is added for sedimentation, and a low-speed centrifuge (4500 rpm) is used for centrifugation, so that the crude peptide is settled to the bottom. After removing the supernatant, the crude peptide suspension was again added, sonicated to form a crude peptide suspension, and the cleaved small molecule impurities were dissolved in glacial ethyl ether and finally removed by centrifugation. After the two times of reaction, the solid sediment is placed in a shade place for air drying, and 118mg of solid crude peptide (AE (cage) FGLKLDRIG) with photo-controlled molecules is obtained.
A small amount of the crude peptide was dissolved in a pure water solution, and the solution was subjected to reverse phase high performance liquid chromatography (RP-HPLC) after passing through a membrane. The gradient was analyzed for 30min at 10% -70% acetonitrile concentration. ESI-MS identification of the main peak after chromatographic analysis verifies the correctness of the solid crude peptide. After the detection is correct, the solid crude peptide is separated and purified by using a C18 semi-preparative column (the semi-preparative gradient is 10 to 70 percent of acetonitrile concentration, and the time is 30 min), the correct product peak solution is collected and put into a freeze dryer for freeze drying, and a white flocculent AE (cage) FGLKLDRIG pure peptide product (76 mg) is obtained.
Example 7:
the AE (cage) FGLKLDRIG pure peptide product obtained in example 6 was selected as the target short peptide for further light degating experiment. 1mgAE (cage) FGLKLDRIG pure peptide product was dissolved in 200uL buffer (6M Gn-HCl,200mM NaH) 2 PO 4 pH = 7.0), placed in an ultraviolet crosslinking chamber, on a flat ice surface, irradiated with 365nm light for 5min. The reaction was monitored by RP-HPLC. The analytical gradient is 10% -70% acetonitrile concentration for 30min. ESI-MS identification of the main peak after chromatographic analysis verifies the correctness of the product. The results show that most of AE (cage) FGLKLDRIG pure peptide (CagedForm) is converted into AEFGLKLDRIG (NativeForm), and prove that the side chain photosensitive protecting group of Fmoc-Glu (cage) -OH molecule can be well removed under 254nm illumination condition, and has good photocaging efficiency (see figures 14, 15 and 16).
In conclusion, the invention discloses a glutamic acid derivative with a photosensitive p-methoxybenzoyl group on a side chain and a synthesis method and application thereof, which are compatible with solid phase synthesis.
While the invention has been described with reference to specific preferred embodiments, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the spirit and scope of the invention.

Claims (7)

1. A glutamic acid derivative, abbreviated as Fmoc-Glu (cage) -OH, characterized by the structural formula:
Figure FDA0003851432460000011
2. a method for synthesizing the glutamic acid derivative according to claim 1, wherein:
firstly, carrying out substitution reaction on N- (9-fluorenylmethoxycarbonyl) -D-glutamic acid 1-tert-butyl ester and alpha-bromo-4-methoxyacetophenone, and then removing glutamic acid main chain carboxyl protecting group tert-butyl ester to obtain a glutamic acid derivative with a photosensitive group on a side chain;
the synthetic route is as follows:
Figure FDA0003851432460000012
3. the synthesis method according to claim 2, characterized by comprising the following steps:
step 1: synthesis of Compound I
Dissolving 1-tert-butyl N- (9-fluorenylmethoxycarbonyl) -D-glutamate and alpha-bromo-4-methoxyacetophenone in dry dichloromethane, adding solid potassium carbonate particles, stirring at 40 ℃ for 24 hours under the protection of argon, after the reaction is finished, carrying out vacuum concentration, and carrying out column chromatography purification and separation to obtain a compound I, namely 1- (tert-butyl) 5- (2- (4-methoxyphenyl) -2-oxoethyl) ((9H-fluoren-9-yl) methoxy) carbonyl) -L-glutamic acid;
step 2: synthesis of target product
Dissolving a compound I in dichloromethane, placing the dichloromethane in an ice bath, stirring and cooling to 0 ℃, then slowly dropwise adding a trifluoroacetic acid solution into a reaction system, reacting for 1 hour, removing the ice bath, recovering the room temperature, and continuing stirring and reacting for 6 hours; after the reaction is finished, the solvent is removed by vacuum concentration, and the pure target product, namely- (S) -2- ((((((9H-fluoren-9-yl) methoxyl) carbonyl) amino) -5- (2- (4-methoxyphenyl) -2-oxoethoxy) -5-oxovaleric acid is obtained.
4. The method of synthesis according to claim 3, characterized in that:
in the step 1, the molar ratio of the N- (9-fluorenylmethoxycarbonyl) -D-glutamic acid 1-tert-butyl ester, the alpha-bromo-4-methoxyacetophenone and the potassium carbonate is 1:1.2:1.5.
5. the method of synthesis according to claim 3, characterized in that:
in step 2, the molar ratio of the compound I to trifluoroacetic acid is 1:60.
6. the method of synthesis according to claim 3, characterized in that:
in the step 2, the volume ratio of dichloromethane to trifluoroacetic acid in the reaction system is 1:1.
7. use of the glutamic acid derivative according to claim 1, wherein:
the glutamic acid derivative is connected to the polypeptide by an N-fluorenylmethyloxycarbonyl Fmoc solid-phase polypeptide synthesis method, so that various polypeptides needing light-operated protection groups are obtained.
CN202211135753.4A 2022-09-19 2022-09-19 Glutamic acid derivative and synthetic method and application thereof Pending CN115340472A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202211135753.4A CN115340472A (en) 2022-09-19 2022-09-19 Glutamic acid derivative and synthetic method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202211135753.4A CN115340472A (en) 2022-09-19 2022-09-19 Glutamic acid derivative and synthetic method and application thereof

Publications (1)

Publication Number Publication Date
CN115340472A true CN115340472A (en) 2022-11-15

Family

ID=83955404

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211135753.4A Pending CN115340472A (en) 2022-09-19 2022-09-19 Glutamic acid derivative and synthetic method and application thereof

Country Status (1)

Country Link
CN (1) CN115340472A (en)

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995000536A1 (en) * 1993-06-22 1995-01-05 Fujisawa Pharmaceutical Co., Ltd. Peptide compounds
US5559111A (en) * 1994-04-18 1996-09-24 Ciba-Geigy Corporation δ-amino-γ-hydroxy-ω-aryl-alkanoic acid amides
WO2005095332A1 (en) * 2004-03-23 2005-10-13 Neomps Amino acid derivatives used as agents bonding to a solid support
CN103906509A (en) * 2011-11-02 2014-07-02 施万制药 Neprilysin inhibitors
CN104003908A (en) * 2014-01-17 2014-08-27 宁波北仑大港科技创新服务中心 Preparing method of hydroxyl-protected serine
CN106831737A (en) * 2017-02-27 2017-06-13 上海众强药业有限公司 The preparation of Wei Patawei and its derivative
US20190015518A1 (en) * 2016-01-04 2019-01-17 University Of Kansas Drug delivery compositions and methods
CN113292456A (en) * 2021-06-17 2021-08-24 常州吉恩药业有限公司 Preparation method of N-fluorenylmethoxycarbonyl-L-aspartic acid-4-tert-butyl ester

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995000536A1 (en) * 1993-06-22 1995-01-05 Fujisawa Pharmaceutical Co., Ltd. Peptide compounds
US5559111A (en) * 1994-04-18 1996-09-24 Ciba-Geigy Corporation δ-amino-γ-hydroxy-ω-aryl-alkanoic acid amides
WO2005095332A1 (en) * 2004-03-23 2005-10-13 Neomps Amino acid derivatives used as agents bonding to a solid support
CN103906509A (en) * 2011-11-02 2014-07-02 施万制药 Neprilysin inhibitors
CN104003908A (en) * 2014-01-17 2014-08-27 宁波北仑大港科技创新服务中心 Preparing method of hydroxyl-protected serine
US20190015518A1 (en) * 2016-01-04 2019-01-17 University Of Kansas Drug delivery compositions and methods
CN106831737A (en) * 2017-02-27 2017-06-13 上海众强药业有限公司 The preparation of Wei Patawei and its derivative
CN113292456A (en) * 2021-06-17 2021-08-24 常州吉恩药业有限公司 Preparation method of N-fluorenylmethoxycarbonyl-L-aspartic acid-4-tert-butyl ester

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
JOHNC. SHEEHA,ET AL.: "Phenacyl Photosensitive Blocking Groups", 《J. ORG. CHEM.》, vol. 38, no. 21, pages 3771 - 3774, XP055049257, DOI: 10.1021/jo00961a027 *
SHAN TANG,ET AL.: "An Fmoc-compatible method for synthesis of peptides containing photocaged aspartic acid or glutamic acid", 《TETRAHEDRON LETTERS》, vol. 56, pages 4582 - 4585, XP029213868, DOI: 10.1016/j.tetlet.2015.06.016 *

Similar Documents

Publication Publication Date Title
JP4405594B2 (en) Improved solid phase peptide synthesis and reagents for use in such synthesis
CN107406480B (en) Peptide synthesis method
DK2757107T3 (en) A process for the solid phase synthesis of liraglutide
RU2515555C2 (en) Method of obtaining degarelix
EP3438121B1 (en) Liquid phase synthesis method for oxytocin using polypeptides
CN108218957B (en) Method for preparing AMG416 by combining solid phase and liquid phase
CN113150075B (en) Cyclic poly-arginine cell-penetrating peptide molecule and synthesis method and application thereof
CN109575109B (en) Method for preparing degarelix by fragment condensation
WO2005087794A1 (en) Process for octreotide synthesis
CN105408344B (en) Peptide-resin conjugates and uses thereof
CN103265620B (en) Somatostatin and preparation method thereof
CN112585153B (en) Compound or salt thereof, and preparation method and application thereof
CN103665115B (en) The chemical preparation process of cyclic decapeptide compound GG-110824
AU657534B2 (en) Peptide process
WO2017020568A1 (en) Coupling peptide chain capable of dissolving indissolvable polypeptide and application of same to separation and purification in liquid chromatogram
Tjoeng et al. Liquid-phase syntheses of protected peptides on the new 3-nitro-4-(bromomethyl) benzoylpoly (ethylene glycol) support
KR102061382B1 (en) Synthesis of beta-turn peptidomimetic cyclic compounds
CN112062829A (en) Preparation method of elcatonin
CN115340472A (en) Glutamic acid derivative and synthetic method and application thereof
JP2001342200A (en) Method for producing octreotide and its derivative
WO2006105199A2 (en) Compositions and methods for synthesis of peptide and related conjugate
JP4878031B2 (en) Solid phase peptide synthesis
KR101971417B1 (en) Process for the Preparation of Buserelin
KR101971418B1 (en) Process for the Preparation of Goserelin
CN112010945B (en) Preparation method of carbetocin impurity Gly9-OH

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