CN105367477A - 1-methyl tryptophan synthesis method - Google Patents

1-methyl tryptophan synthesis method Download PDF

Info

Publication number
CN105367477A
CN105367477A CN201410391864.0A CN201410391864A CN105367477A CN 105367477 A CN105367477 A CN 105367477A CN 201410391864 A CN201410391864 A CN 201410391864A CN 105367477 A CN105367477 A CN 105367477A
Authority
CN
China
Prior art keywords
methyl
reaction
tryptophan
solid
methyl tryptophan
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
CN201410391864.0A
Other languages
Chinese (zh)
Other versions
CN105367477B (en
Inventor
汪志勇
张胜
郑小琦
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.)
University of Science and Technology of China USTC
Original Assignee
University of Science and Technology of China USTC
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 University of Science and Technology of China USTC filed Critical University of Science and Technology of China USTC
Priority to CN201410391864.0A priority Critical patent/CN105367477B/en
Publication of CN105367477A publication Critical patent/CN105367477A/en
Application granted granted Critical
Publication of CN105367477B publication Critical patent/CN105367477B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Abstract

The present invention provides a direct and efficient 1-methyl tryptophan synthesis method, and specifically relates to a method for synthesizing 1-methyl tryptophan represented by the following formula I. The method comprises that: in an inert solvent, in the presence of an alkaline agent, tryptophan and a methylating agent CH3X (X is defined in the specification) are subjected to a reaction, and separation is performed to obtain the desired product 1-methyl tryptophan. According to the present invention, the organic solvent is adopted as the reaction medium, the suitable alkaline reagent is adopted as the dehydrogenating agent, the tryptophan reacts with the methyl halide, and the crude product is subjected to extraction, washing, re-crystallization and other operations so as to obtain the high purity 1-methyl tryptophan; and the method has characteristics of good yield, strong selectivity, simple operation, safety, and easy control. The formula I is defined in the specification.

Description

A kind of method of synthesizing 1-methyl tryptophan
Technical field
The invention belongs to the field of chemical synthesis, relate more specifically to the method for synthesizing 1-methyl tryptophan.
Background technology
The disease of a lot of serious threat human health is all weakened with human immune system relevant, and such as cancer cells will destroy the effective immunne response of patient gradually, and the transmissible diseases such as HIV and HCV are also cause viral longer-term persistence because human immunological competence weakens.In recent years research finds, the enzyme indoles amine-2,3-dioxygenase (indoleamine-2,3-dioxygenase, IDO) of catalysis tryptophan degradation is in regulation and control immunosuppression.This enzyme is long term activation in a lot of cancer patients, and the activity of this fermentoid is higher, then cancer patients's prognosis is poorer.Preclinical study shows, suppresses IDO effectively can improve cancer therapy drug chemotherapeutic treatment effect common at present.Verified in experiment in vitro, 1-methyl tryptophan (1-MT) is competitive inhibitor (Cady, the S.G. of IDO; Sono, M., Arch.Biochem.Biophys.1991,291,326-333.), its chemical structural formula is:
Recently, 1-methyl tryptophan and a series ofly gone up clinical chemotherapeutics as endoxan, cis-platinum, Zorubicin, or paclitaxel plus uses, and demonstrates splendid synergistic effect, become a kind of get a good chance of chemistry, immune conjoint therapy medicine (MullerA.J., DuhadawayJ.B., DonoverP.S., Sutanto-WardE., PrendergastG.C., Nat.Med., 2005,11,312 – 9).1-methyl tryptophan is expected to play a significant role in anticancer therapy, and its synthetic method exploitation will bring favourable Social benefit and economic benefit.
Japanese scholars Yamada, S.; Shioiri, T.; Itaya, T.; Hara, T.; The people such as Matsueda, R. at Chem.Pharm.Bull.1965,13, report a kind of synthetic method of 1-methyl tryptophan on 88.The method sodium Metal 99.5 makes sodium amide in liquefied ammonia, then adds tryptophane and iodomethane reaction, and the purifying through complexity obtains 1-methyl tryptophan.Use sodium Metal 99.5 in this method, this material habitually under very easily burn, and general industry product are again large bulks, need to shred to feed intake under protection of inert gas during reaction.Operation also needs at low temperatures, and require high to production unit, operative employee's personnel safety is also subject to very large threat.In addition, the leakage of ammonia also may have extreme influence to surrounding enviroment, industrial very difficult realization.
As mentioned above, Japanese scholars Yamada, S.; Shioiri, T.; Itaya, T.; Hara, T.; The method that the people such as Matsueda, R. report on Chem.Pharm.Bull. uses sodium Metal 99.5, also needs special cyrogenic equipment, requires high to operative employee, dangerous large, and the security of suitability for industrialized production is difficult to ensure.In the report of published 1-methyl tryptophan synthetic method, the method effectively do not addressed these problems.
Summary of the invention
In view of above-mentioned, the object of this invention is to provide that productive rate is good, selectivity is strong, simple to operate, safe and be easy to the method for the synthesis 1-methyl tryptophan controlled.
In one aspect, the invention provides the method for a kind of synthesis by the 1-methyl tryptophan shown in following formula I,
Described method comprises: in inert solvent, under alkaline reagents exists, makes the tryptophane shown in Formula Il and methylating reagent CH 3x reacts, through being isolated required product 1-methyl tryptophan, at described methylating reagent CH 3in X, X represents Cl, Br, I, OSO 2(OCH 3), OPO (OCH 3) 2, OSO 2cF 3or OSO 2c 6h 4cH 3,
Preferably, described inert solvent is one or more in toluene, tetrahydrofuran (THF), acetonitrile, acetone, butanols, DMF and dimethyl sulfoxide (DMSO).
Preferably, described alkaline reagents is one or more in Quilonum Retard, sodium carbonate, salt of wormwood, sodium phosphate, potassiumphosphate, sodium amide, lithium hydride, sodium hydride, potassium hydride KH, diisopropylamine lithium, hexamethyldisilazane lithium, hmds base sodium, hmds base potassium and butyllithium.
Preferably, described reaction is carried out under protection of inert gas.
Preferably, described tryptophane: alkaline reagents: methylating reagent CH 3the mol ratio of X is 1:1 ~ 10:0.5 ~ 2.5.
Preferably, described separation is included in after completion of the reaction, to go out reaction with shrend, in recrystallization solvent, then carrying out recrystallization and obtaining solid by filtering, finally carrying out drying.
Further preferably, described recrystallization solvent is one or more in toluene, ether, acetone, methyl-phenoxide, ethanol and t-butyl methyl ether.
Further preferably, described recrystallization carries out at the temperature of-20-5 DEG C.
Further preferably, by filtering the solid that obtains before the drying, form the aqueous solution by water dissolution, and with acid by pH regulator to 5.5 ~ 7.5 of the described aqueous solution to separate out solid, by filtering the acquisition solid of separating out, after washing, carry out drying.
Further preferably, the product obtained is pure 1-methyl tryptophan.
The present invention is by using organic solvent as reaction medium, and at suitable alkaline reagents as under dehydrogenating agent, tryptophane and methyl halogenated thing react, and crude product can obtain highly purified 1-methyl tryptophan through operations such as extraction, washing, recrystallizations.
Accompanying drawing explanation
Fig. 1 is the 1-methyl tryptophan synthesized according to the embodiment of the present invention 1 1h-NMR nmr spectrum.
Fig. 2 is the 1-methyl tryptophan synthesized according to the embodiment of the present invention 1 13c-NMR nmr spectrum.
The infrared spectrogram of the 1-methyl tryptophan that Fig. 3 synthesizes according to the embodiment of the present invention 1.
Fig. 4 is the mass spectrum of the 1-methyl tryptophan according to the embodiment of the present invention 1 synthesis.
Embodiment
In order to provide, productive rate is good, selectivity is strong, simple to operate, safe and be easy to the method for synthesis 1-methyl tryptophan that controls, the present invention's organic solvent is as reaction medium, suitable alkaline reagents is as dehydrogenating agent, tryptophane can react with methyl halogenated thing, the 1-methyl tryptophan crude product generated, through operations such as extraction, washing, recrystallizations, just can obtain highly purified 1-methyl tryptophan.
In one embodiment, the invention provides the method for a kind of synthesis by the 1-methyl tryptophan shown in following formula I,
Described method comprises: in inert solvent, under alkaline reagents exists, makes the tryptophane shown in Formula Il and methylating reagent CH 3x reacts, through being isolated required product 1-methyl tryptophan, at described methylating reagent CH 3in X, X represents Cl, Br, I, OSO 2(OCH 3), OPO (OCH 3) 2, OSO 2cF 3or OSO 2c 6h 4cH 3,
Preferably, described inert solvent is one or more in toluene, tetrahydrofuran (THF), acetonitrile, acetone, butanols, DMF and dimethyl sulfoxide (DMSO).
Preferably, described alkaline reagents is one or more in Quilonum Retard, sodium carbonate, salt of wormwood, sodium phosphate, potassiumphosphate, sodium amide, lithium hydride, sodium hydride, potassium hydride KH, diisopropylamine lithium, hexamethyldisilazane lithium, hmds base sodium, hmds base potassium and butyllithium.In the present invention, adopt commercial alkaline reagents to prepare without the need to original position, no longer need directly to use sodium Metal 99.5, as with sodium amide just without the need to sodium and liquefied ammonia original position preparation; The present invention's organic solvent replaces liquefied ammonia to make solvent in addition, just without the need to using low temperature pressure-resistant equipment on slave unit, operation being got up more safe and simple, reaction can be made industrially well to apply.
Preferably, described reaction is carried out under protection of inert gas.
Preferably, described tryptophane: alkaline reagents: methylating reagent CH 3the mol ratio of X is 1:1 ~ 10:0.5 ~ 2.5.
Preferably, described separation is included in after completion of the reaction, to go out reaction with shrend, in recrystallization solvent, then carrying out recrystallization and obtaining solid by filtering, finally carrying out drying.
Further preferably, described recrystallization solvent is one or more in toluene, ether, acetone, methyl-phenoxide, ethanol and t-butyl methyl ether.
Further preferably, described recrystallization carries out at the temperature of-20-5 DEG C.
Further preferably, by filtering the solid that obtains before the drying, form the aqueous solution by water dissolution, and with acid by pH regulator to 5.5 ~ 7.5 of the described aqueous solution to separate out solid, by filtering the acquisition solid of separating out, after washing, carry out drying.
By aforesaid method, the present invention can obtain pure 1-methyl tryptophan product.
In a more particular embodiment, first, in reaction vessel such as there-necked flask, cool in the suspension of 1 weight part tryptophane in 50-150mL inert solvent at low temperatures such as 0 DEG C of ice-water bath, afterwards preferred at rare gas element as N 2under protective atmosphere, in such as 30-50min, add the alkaline reagents of 1-10 equivalent part in batches.After adding alkaline reagents, preferably reaction system is at room temperature stirred 0.5-2 hour, reaction system can become muddy lilac solution from settled solution during this period.After stirring completes, again reaction system to be moved under low temperature in such as 0 DEG C of ice-water bath, by the methylating reagent of 0.5-2.5 equivalent part as the solution of methyl iodide in 10-50mL inert solvent, be added dropwise within reaction system within such as 20-40min, the purple of reaction system can take off gradually, and the opacity of reaction mixture solution also can reduce.After dropwising, more at room temperature continue reaction 2 ~ 10h.After completion of the reaction, by carrying out recrystallization in recrystallization solvent, filtering also dry afterwards and obtaining required product.
More preferably, after completion of the reaction, add about 2-10mL water and react with cancellation.Gained mixed solution is poured into 300-600mL in the recrystallization solvent of such as about 0 DEG C, a large amount of solid is separated out, direct suction filtration.Further preferably, the pH of this aqueous solution, to obtain the aqueous solution, is adjusted between 5.5 ~ 7.5 with acid such as concentrated hydrochloric acid, separates out a large amount of solid by gained solid 30-100mL water dissolution, by the solid suction filtration of separating out.The solid product of gained preferably washs with the frozen water of 50-200mL again, optionally uses 50-200mL at the recrystallization solvent wash of about 0 DEG C afterwards.White solid powder product 8.5g (productive rate 78%) is obtained, the specific rotatory power [α] of product with anhydrous sodium sulfate drying d 20+ 7.5 (c=1.0,2MHCl) show that the optical purity of product is maintained.
Be noted that in the inventive method, to reaction raw materials add or order by merging is not particularly limited.Such as, in one embodiment, tryptophane can first be dissolved in inert solvent, then adds alkaline reagents, finally reacts with methylating reagent again.In another embodiment, tryptophane and methylating reagent can first be dissolved in inert solvent, add alkaline reagents afterwards again and react.
The following example is for detailed description of the present invention, and unrestricted the present invention.
Embodiment 1
In the there-necked flask that ice-water bath is housed; 10.2g tryptophane is added the tetrahydrofuran (THF) (THF) of 100mL; under nitrogen protection condition; within 50min; add the commercial sodium amide of 5g in batches; remove ice-water bath after adding sodium amide to stir 2 hours in room temperature (about 25 DEG C), then with ice-water bath, temperature of reaction system is down to 0 DEG C.The solution of 7.46g methyl iodide in 50mLTHF is added dropwise in above-mentioned mixing solutions with constant pressure funnel in 30min, dropwises rear continuation stirring reaction 15 hours.After completion of the reaction, the water at room temperature adding 30mL reacts with quencher.Reaction mixture is poured in 400mL ether, observe a large amount of solid to separate out, use Büchner funnel suction filtration, the solid 100mL water dissolution obtained obtains solution, adds concentrated hydrochloric acid 25mL and regulates pH to 6.5, and by the solid of separating out, suction filtration is out again, with anhydrous sodium sulfate drying, obtain white solid powder product, 8.5g (productive rate 78%, m.p.269 DEG C).
To the nuclear-magnetism test compounds of the product Bruker400MHz obtained by aforesaid method 1h-NMR and 13c-NMR spectrogram, infrared and mass spectroscopy.Fig. 1 to Fig. 4 is respectively the product obtained according to the present embodiment 1 1h-NMR nmr spectrum, 13c-NMR nmr spectrum, infrared spectrogram and mass spectroscopy figure.As can be seen from the figure, the product obtained 1h-NMR nmr spectrum has following characteristics and absorbs: 1h-NMR (400MHz, CDCl 3): δ=2.96 (t, J=8.8Hz, 1H), 3.73 (s, 3H), 7.02 (d, J=8Hz, 1H), 7.14 (t, J=8Hz, 2H), 7.38 (d, J=8Hz, 1H), 7.57 (d, J=8Hz, 1H); 13c-NMR nmr spectrum has following characteristics and absorbs: 13c-NMR (100MHz, CDCl 3) δ (ppm)=25.3,32.0,52.9,104.6,109.9,118.2,119.1,121.7,126.7,129.4,129.5,136.7,171.3; Infrared spectrum has following characteristics and absorbs: ν max(KBr) cm -1: 3423,3066,2569,2077,1593,1408,1132,918,732,557; And mass spectral characteristic peak is as follows: 217.0973,218.1006.Can find out clearly from above result, the solid product obtained is 1-methyl tryptophan, and its purity is 100%.
Embodiment 2
10.2g tryptophane is added the dimethyl sulfoxide (DMSO) (DMSO) of 130mL, under nitrogen protection condition, within 30min, add 5g sodium hydroxide in batches, to add after sodium hydroxide stirring at room temperature one hour, then with ice, system temperature is down to 0 DEG C, the solution of the 30mLDMSO of 7.46g methyl iodide is instilled tryptophane within 30min, the DMSO solution of sodium amide, dropwise rear continuation stirring reaction 15h, make tryptophane complete reaction, the water quencher reaction of 50mL is added in system, reaction mixture is poured in 400mL ether, a large amount of solid is had to separate out, suction filtration, the solid water dissolution obtained obtains solution, adding concentrated hydrochloric acid adjusts between pH to 6 ~ 7, by the solid suction filtration of precipitation out, drying obtains white solid powder product 8.7g (productive rate 80%, m.p.269 DEG C).
Identical with embodiment 1, obtained product is carried out 1h-NMR and 13c-NMR nucleus magnetic resonance, infrared and mass spectroscopy.Result (not shown) shows, the solid product obtained is 1-methyl tryptophan, and its purity is 100%.
Embodiment 3
Tryptophane (10.2g) and methyl iodide (7.46g) are put into toluene and acetonitrile (1:1, mixed solvent 50mL) dissolves, add KOH (8.4g) within 2h in batches, solution becomes brown, room temperature for overnight, solution colour is colorless cleared solution by brown stain, add frozen water (150mL), acetonitrile is revolved and removes, extract with propyl carbinol (3 × 100mL), obtained organic phase anhydrous sodium sulfate drying is spent the night, then the organic phase being spin-dried for gained is filtered, obtain the 1-methyl tryptophan (7.7g that white is pure, 71%, m.p.269 DEG C).
Identical with embodiment 1, obtained product is carried out 1h-NMR and 13c-NMR nucleus magnetic resonance, infrared and mass spectroscopy.Result (not shown) shows, the solid product obtained is 1-methyl tryptophan, and its purity is 100%.
The above; be only the preferred embodiments of the disclosure; but protection scope of the present invention is not limited thereto; any those skilled in the art are in the technical scope of present disclosure; the change that can expect easily or replacement; all should be encompassed in protection scope of the present invention, therefore, the protection domain that protection scope of the present invention should limit with claim is as the criterion.

Claims (10)

1. synthesis is by a method for the 1-methyl tryptophan shown in following formula I,
Described method comprises: in inert solvent, under alkaline reagents exists, makes the tryptophane shown in Formula Il and methylating reagent CH 3x reacts, through being isolated required product 1-methyl tryptophan, at described methylating reagent CH 3in X, X represents Cl, Br, I, OSO 2(OCH 3), OPO (OCH 3) 2, OSO 2cF 3or OSO 2c 6h 4cH 3,
2. method according to claim 1, is characterized in that, described inert solvent is one or more in toluene, tetrahydrofuran (THF), acetonitrile, acetone, butanols, DMF and dimethyl sulfoxide (DMSO).
3. method according to claim 1, it is characterized in that, described alkaline reagents is one or more in Quilonum Retard, sodium carbonate, salt of wormwood, sodium phosphate, potassiumphosphate, sodium amide, lithium hydride, sodium hydride, potassium hydride KH, diisopropylamine lithium, hexamethyldisilazane lithium, hmds base sodium, hmds base potassium and butyllithium.
4. method according to claim 1, is characterized in that, described reaction is carried out under protection of inert gas.
5. method according to claim 1, is characterized in that, described tryptophane: alkaline reagents: methylating reagent CH 3the mol ratio of X is 1:1 ~ 10:0.5 ~ 2.5.
6. the method according to any one of claim 1-5, is characterized in that, described separation is included in after completion of the reaction, to go out reaction with shrend, in recrystallization solvent, then carrying out recrystallization and obtaining solid by filtering, finally carrying out drying.
7. method according to claim 6, is characterized in that, described recrystallization solvent is one or more in toluene, ether, acetone, methyl-phenoxide, ethanol and t-butyl methyl ether.
8. method according to claim 6, is characterized in that, described recrystallization carries out at the temperature of-20-5 DEG C.
9. method according to claim 6, is characterized in that, by filtering the solid of acquisition before the drying, the aqueous solution is formed by water dissolution, and with acid by pH regulator to 5.5 ~ 7.5 of the described aqueous solution to separate out solid, obtaining the solid of separating out by filtering, after washing, carrying out drying.
10. method according to claim 9, is characterized in that, the product obtained is pure 1-methyl tryptophan.
CN201410391864.0A 2014-08-11 2014-08-11 A method of synthesis 1- methyl tryptophan Active CN105367477B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201410391864.0A CN105367477B (en) 2014-08-11 2014-08-11 A method of synthesis 1- methyl tryptophan

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201410391864.0A CN105367477B (en) 2014-08-11 2014-08-11 A method of synthesis 1- methyl tryptophan

Publications (2)

Publication Number Publication Date
CN105367477A true CN105367477A (en) 2016-03-02
CN105367477B CN105367477B (en) 2018-11-27

Family

ID=55370170

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201410391864.0A Active CN105367477B (en) 2014-08-11 2014-08-11 A method of synthesis 1- methyl tryptophan

Country Status (1)

Country Link
CN (1) CN105367477B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109400517A (en) * 2017-08-17 2019-03-01 上海时莱生物技术有限公司 1- methyl-tryptophan class compound and its preparation method and application
CN110790696A (en) * 2018-08-03 2020-02-14 田海滨 11Preparation method and preparation system of C- α -methyl-L-tryptophan

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101121717A (en) * 2007-05-17 2008-02-13 四川大学 Synthesis for natural medicament physostigmine for resisting senile dementia disease and phenylaminoformic acid ester phenserine
CN102167766A (en) * 2011-03-21 2011-08-31 北京化工大学 Vinyl amino acid (ester) polymer and preparation method thereof
CN103570603A (en) * 2012-07-19 2014-02-12 杭州中肽生化有限公司 Novel synthetic method of hemiasterlin

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101121717A (en) * 2007-05-17 2008-02-13 四川大学 Synthesis for natural medicament physostigmine for resisting senile dementia disease and phenylaminoformic acid ester phenserine
CN102167766A (en) * 2011-03-21 2011-08-31 北京化工大学 Vinyl amino acid (ester) polymer and preparation method thereof
CN103570603A (en) * 2012-07-19 2014-02-12 杭州中肽生化有限公司 Novel synthetic method of hemiasterlin

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
HE, BIN ET AL: "Total Synthesis of (-)-Ardeemin", 《JOURNAL OF ORGANIC CHEMISTRY》 *
SUN, TING ET AL: "Synthesis and biological evaluation of novel 1-alkyltryptophan analogs as potential antitumor agents", 《MOLECULES》 *
YAMADA, SHUNICHI ET AL: "Ind.-N-alkylation of tryptophan and synthesis of 1-alkyl-tryptophan hydrazides", 《CHEMICAL & PHARMACEUTICAL BULLETIN》 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109400517A (en) * 2017-08-17 2019-03-01 上海时莱生物技术有限公司 1- methyl-tryptophan class compound and its preparation method and application
CN110790696A (en) * 2018-08-03 2020-02-14 田海滨 11Preparation method and preparation system of C- α -methyl-L-tryptophan

Also Published As

Publication number Publication date
CN105367477B (en) 2018-11-27

Similar Documents

Publication Publication Date Title
CN103910679A (en) Method for preparing enzalutamide
KR20140079770A (en) Production method for 4,4-difluoro-3,4-dihydroisoquinoline derivative
AU2011281421A1 (en) Process for preparing aminobenzoylbenzofuran derivatives
CN102993102A (en) Synthetic method of [1-methyl-2-(8'-octyl hydroxamic acid group)-5-N,N-bi(2'-chloroethyl)]-1H-benzimidazole
CN102746210A (en) Synthesis method for key intermediate of silodosin
JP2023524626A (en) Method for synthesizing roxadustat and intermediates thereof and intermediates thereof
CN104045602A (en) Improved method for preparing tetrazole for valsartan
CN105367477A (en) 1-methyl tryptophan synthesis method
CN107200707B (en) Preparation method of pimavanserin
CN111100128A (en) Synthetic method of Ribocini intermediate product and intermediate compound thereof
CN105859686A (en) Preparation technology of high-purity dabigatran etexilate
CN103242251B (en) Preparation method of letrozole
CN104744336B (en) A kind of Silodosin intermediate and preparation method thereof, and the method for preparing with the intermediate silodosin
CN106674281B (en) A kind of Rosuvastatin midbody compound, preparation method and its usage
NO179517B (en) Process for the preparation of 8-chloroquinolone derivatives
CN107325078B (en) Preparation method of cilostazol
Qiu et al. Microwave-assisted synthesis and antibacterial activity of novel chenodeoxycholic acid thiosemicarbazone derivatives
KR20140028433A (en) Proces for purifying fluvoxamine free base and process for preparing high purity fluvoxamine maleate using the same
CN107759603B (en) Preparation method of heterocyclic compound
US20080194822A1 (en) Imiquimod production process
AU2008358758A1 (en) A process for preparing atovaquone and associate intermediates
CN104496900A (en) Method for preparing 2-methoxy-6-one-5,7,8-trihydro-quinoline
Shahroudi et al. Synthesis of 2-selanylidene-1, 9a-dihydro-2H-pyrido [1, 2-a] pyrimidine derivatives
CN102050709B (en) Method for preparing 2-[2-(2,2,2-trifluoro-ethyoxyl) phenoxyl] alcohol
CN106866774B (en) For antitumor compound and its preparation method and application

Legal Events

Date Code Title Description
C06 Publication
PB01 Publication
C10 Entry into substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant