CN104945332B - The preparation method of Erlotinib - Google Patents

The preparation method of Erlotinib Download PDF

Info

Publication number
CN104945332B
CN104945332B CN201410127452.6A CN201410127452A CN104945332B CN 104945332 B CN104945332 B CN 104945332B CN 201410127452 A CN201410127452 A CN 201410127452A CN 104945332 B CN104945332 B CN 104945332B
Authority
CN
China
Prior art keywords
preparation
erlotinib
bis
methoxy ethoxies
quinazoline
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.)
Expired - Fee Related
Application number
CN201410127452.6A
Other languages
Chinese (zh)
Other versions
CN104945332A (en
Inventor
丁克
陆小云
李伟华
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.)
Guangzhou Institute of Biomedicine and Health of CAS
Original Assignee
Guangzhou Institute of Biomedicine and Health of CAS
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 Guangzhou Institute of Biomedicine and Health of CAS filed Critical Guangzhou Institute of Biomedicine and Health of CAS
Priority to CN201410127452.6A priority Critical patent/CN104945332B/en
Publication of CN104945332A publication Critical patent/CN104945332A/en
Application granted granted Critical
Publication of CN104945332B publication Critical patent/CN104945332B/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/94Nitrogen atoms

Abstract

The invention discloses a kind of preparation method of Erlotinib, belong to field of medicine preparing technology.In the preparation method, with 4,5 two(2 methoxy ethoxies)2 p-nitriles are raw material, through reduction, the amido 4,5 two of intermediate 2 obtained by hydrolysis(2 methoxy ethoxies)Benzamide directly obtains Erlotinib key intermediate 6,7 2 with triethyl orthoformate cyclization(2 methoxy ethoxies)The ketone of 3H quinazolines 4, the chloro-product of the quinazoline and an amido phenylacetylene react, and obtain Erlotinib.The preparation method of the present invention, not only overcomes nitro reduction in prior synthesizing method and uses expensive catalyst, and temperature higher defect during cyclization, also avoid, by carbonamidine intermediate, shortening reactions steps, reduce reaction cost, improve yield.And all reaction conditions are very gentle in the preparation method, are particularly suitable for industrialized production.

Description

The preparation method of Erlotinib
Technical field
The invention belongs to field of medicine preparing technology, more particularly to a kind of preparation method of Erlotinib.
Background technology
Erlotinib(Erlotinib), trade name Erlotinib(Tarceva), it is the one of Osi Pharm Inc. of U.S. research and development Plant EGF EGFR EGFR-TK type small molecular inhibitor cancer therapy drugs.Listed first in the U.S. within 2004, mainly For treating non-small cell lung cancer.The chemical name of Erlotinib is N- (3- acetylene phenyl) (2- methoxyl group ethoxies of -6,7- two Base) -4- quinazoline amine, mechanism of action is to be combined suppression EGFR phosphorylations with substrate competition in the cell, blocks tumour cell letter Number transduction, so as to suppress the growth of tumour cell, induce its dead.The structure of Erlotinib is shown as a formula V.
Key step is related to key intermediate 6 in Erlotinib synthetic method, and 7- is double-(2- methoxy ethoxies)- 3H- quinolines The synthesis of oxazoline -4- ketone.In the prior art, have in the document report synthetic method of the intermediate, these methods and mainly deposit The problem of be that nitrification yield and purity are relatively low, nitro reduction uses expensive catalyst, and temperature is higher etc. during cyclization.These Factor have impact on the yield and purity of Erlotinib, be unfavorable for industrialized production.
In view of the above-mentioned problems, also document report uses 4,5- double(2- methoxy ethoxies)- 2- nitrobenzonitriles are Raw material, through palladium charcoal/hydrazine hydrate reduction nitro, cyan-hydrolysis obtains 4,5- pairs simultaneously(2- methoxy ethoxies)- 2- aminobenzoyls Amine intermediate, the intermediate obtains carbonamidine intermediate with DMF-DMA additions, then obtains Erlotinib key intermediate 6 through cyclization, 7- pairs-(2- methoxy ethoxies)- 3H- quinazoline-4-ones.Synthetic route is as follows:
This method synthesis 6,7- pairs-(2- methoxy ethoxies)Though -3H- quinazoline-4-ones can carry out industrialized production, Cyclization need to pass through carbonamidine intermediate, and reactions steps are long, add reaction cost, while also influenceing the yield of Erlotinib.
The content of the invention
Based on this, it is an object of the invention to overcome the defect of prior art there is provided a kind of preparation method of Erlotinib, The characteristics of this method has short synthetic reaction step, low cost and high yield.
To achieve the above object, the present invention takes following technical scheme:
A kind of preparation method of Erlotinib, comprises the following steps:
1)With 4,5- bis-(2- methoxy ethoxies)- 2- p-nitriles are raw material, in the presence of reducing agent, through reducing, Hydrolysis obtains 2- amidos -4,5- two(2- methoxy ethoxies)Benzamide;
2)2- amidos-the 4,5- two that above-mentioned steps are obtained(2- methoxy ethoxies)Benzamide and triethyl orthoformate Reacted under lewis acidic catalysis, direct cyclization obtains intermediate 6,7- bis-(2- methoxy ethoxies)- 3H- quinoline azoles Quinoline -4- ketone;
3)The 6,7- bis- that above-mentioned steps are obtained(2- methoxy ethoxies)- 3H- quinazoline-4-ones are carried out with POCl3 Reaction, obtains 6,7- bis-(2- methoxy ethoxies)- quinazoline -4- chlorine;
4)6, the 7- bis- that above-mentioned steps are obtained(2- methoxy ethoxies)- quinazoline -4- chlorine and an amido phenylacetylene are anti- Should, obtain Erlotinib.
The preparation method of the Erlotinib of the present invention, with 4,5- bis-(2- methoxy ethoxies)- 2- p-nitriles are raw material, Through reduction, intermediate 2- amidos -4,5- bis- obtained by hydrolysis(2- methoxy ethoxies)Benzamide directly with orthoformic acid three Ethyl ester cyclization obtains Erlotinib key intermediate 6,7- bis-(2- methoxy ethoxies)- 3H- quinazoline-4-ones, the quinazoline Chloro-product and amido phenylacetylene react, obtain Erlotinib.The preparation method avoids, by carbonamidine intermediate, shortening Reactions steps, reduce reaction cost, and improve yield.
In wherein some embodiments, step 1)In, reaction dissolvent is water, water-dioxane or water-ethanol;Reducing agent For sodium dithionite, iron, zinc, palladium carbon or Raney Ni- hydrazine hydrates;Reaction temperature is 0 DEG C -40 DEG C.Wherein, Raney Ni are Raney's nickel, is a kind of machine metallic compound as hydrogen activity catalytic reducer;Above-mentioned water-dioxane represents water and dioxy six The mixed solvent of ring;Equally, water-ethanol represents the mixed solvent of water and ethanol;Raney Ni- hydrazine hydrates represent Raney Ni and The mixing reducing agent of hydrazine hydrate.Using above-mentioned process conditions, overcome nitro reduction in prior synthesizing method and use expensive catalysis Agent, and the defect such as temperature is higher during reaction.
In wherein some embodiments, step 1)In, reaction dissolvent is water;Reducing agent is Raney Ni- hydrazine hydrates;Reaction Temperature is 20 DEG C -40 DEG C.
In wherein some embodiments, step 2)In, the lewis acid is trifluoroacetic acid or BFEE;Reaction Solvent is ethyl acetate, dichloromethane, ethanol, at least one of methanol.Ethyl acetate.
In wherein some embodiments, step 2)In, the 2- amidos -4,5- bis-(2- methoxy ethoxies)Benzoyl Amine, triethyl orthoformate, lewis acidic consumption mol ratio is 1:1-1.5:0.05-0.15, preferably 1:1.2:01;Reaction temperature For 70-80 DEG C, preferably 77 DEG C.Under the above-described reaction conditions, the progress that reacts fully can be promoted, the wave to raw material can be avoided again Take, meanwhile, the reaction condition is gentle, is particularly suitable for industrialized production.
In wherein some embodiments, step 3)In, reaction dissolvent is ethyl acetate, dichloromethane, in dioxane It is at least one.Ethyl acetate.
In wherein some embodiments, step 3)In, 6, the 7- bis-(2- methoxy ethoxies)- 3H- quinazoline-4-ones Consumption mol ratio with POCl3 is 1:2-4, preferably 1:3;Reaction temperature is 60-80 DEG C, preferably 70 DEG C.In above-mentioned reaction bar Under part, the progress that reacts fully can be promoted, the waste to raw material can be avoided again.
In wherein some embodiments, step 4)In, reaction dissolvent is methanol, ethanol, isopropanol, in n-butanol at least It is a kind of.Preferred alcohol.
In wherein some embodiments, step 4)In, 6,7- bis-(2- methoxy ethoxies)- quinazoline -4- chlorine and an amine The consumption mol ratio of base phenylacetylene is 1:1-1.3, preferably 1:1.1;Reaction temperature is 60-80 DEG C, preferably 70 DEG C.It can promote anti- It should fully carry out, the waste to raw material can be avoided again.
In wherein some embodiments, step 4)In, using the method purifying erlotinib of recrystallization, the recrystallization Solvent is ethanol, isopropanol, n-butanol, one kind in normal propyl alcohol.It is preferred that n-butanol.Using above-mentioned solvent, purity can be obtained Higher recrystallization product.
Compared with prior art, the invention has the advantages that:
The preparation method of a kind of Erlotinib of the present invention, with 4,5- bis-(2- methoxy ethoxies)- 2- p-nitriles are Raw material, through reduction, intermediate 2- amidos -4,5- bis- obtained by hydrolysis(2- methoxy ethoxies)Benzamide is directly and primitive nail Triethylenetetraminehexaacetic acid ester cyclization obtains Erlotinib key intermediate 6,7- bis-(2- methoxy ethoxies)- 3H- quinazoline-4-ones, it is to avoid By carbonamidine intermediate, reactions steps are shortened, reaction cost are reduced, and improve yield.
Also, the preparation method also overcomes nitro reduction in prior synthesizing method and uses expensive catalyst, and cyclization The defect such as temperature is higher during reaction.Meanwhile, all reaction conditions are very gentle in the preparation method, are particularly suitable for industrialization Production.
Embodiment
Describe the present invention in detail below in conjunction with specific embodiment, but any limitation is not caused to the present invention.
Following examples are with 4,5- bis-(2- methoxy ethoxies)- 2- p-nitriles are raw material, and through reduction, hydrolysis is obtained Intermediate 2- amidos -4,5- two(2- methoxy ethoxies)Benzamide directly obtains 6,7- bis- with triethyl orthoformate cyclization (2- methoxy ethoxies)- 3H- quinazoline-4-ones, the chloro-product of the quinazoline and an amido phenylacetylene react, and obtain angstrom sieve For Buddhist nun.Synthetic line is as follows:
Embodiment 1
(1)Compound ii(2- amidos -4,5- two(2- methoxy ethoxies)Benzamide)Synthesis.
At 0 DEG C, by chemical compounds I(4,5- bis-(2- methoxy ethoxies)- 2- p-nitriles)214g, Raney Ni50g It is suspended in about 2L water, mechanical agitation, the aqueous solution 500mL of the hydrazine hydrate containing 141mL is added dropwise, completion of dropping is warmed to room temperature(About 20-30℃), 3h is reacted, TLC is monitored to reaction and finished.Adding sodium hydroxide solution adjusts Ph values between 11-12, adds about 2L Dichloromethane is extracted 2 times, and organic phase saturated common salt washing, anhydrous sodium sulfate drying, revolving obtains the thick of 170g compound iis Product, yield is 83%.
The characterize data of the compound ii is:1H-NMR(400MHz,DMSO-d6):7.15(1H,s),6.40(2H,s), 6.27(1H,s),5.73(1H,s),4.01-4.03(2H,J=4.8Hz,t),3.95-3.98(2H,J=4.8Hz,t),3.64- 3.66(2H,J=4.8Hz,t),3.58-3.60(2H,J=4.8Hz,t),3.30(3H,s),3.31(3H,s)。
(2)Compound III(6,7- bis-(2- methoxy ethoxies)- 3H- quinazoline-4-ones)Synthesis.
By compound ii(2- amidos -4,5- two(2- methoxy ethoxies)Benzamide)170g, triethyl orthoformate 106.3g, trifluoroacetic acid 7g, add into about 1.7L ethyl acetate, are heated to reflux, about 4h, TLC is monitored to reaction and finished, and stop Reaction.Reaction solution is cooled to room temperature, there are a large amount of solids to separate out, suction filtration collects solid, and vacuum drying obtains compound III and consolidated Body 158g, carries out purity testing, purity is 99%, and yield is 90% with HPLC.
The characterize data of the compound III is:1H-NMR(400MHz,DMSO-d6):12.02(1H,brs),7.97(1H, s),7.46(1H,s),7.14(1H,s),4.23-4.25(2H,J=4.8Hz,t),4.17-4.20(2H,J=4.8Hz,t), 3.69-3.72(4H,J=4.8Hz,t),3.32(6H,s)。
(3)Compounds Ⅳ(6,7- bis-(2- methoxy ethoxies)- quinazoline -4- chlorine)Synthesis.
By compound III(6,7- bis-(2- methoxy ethoxies)- 3H- quinazoline-4-ones)5g is added to about 50mL acetic acid second In ester, nitrogen protection adds about 4.7mL POCl3s, 1gDMF is added dropwise(N,N-dimethylformamide), 70 DEG C are then heated to, About 2h is reacted, TLC is monitored to reaction and finished.Reaction solution is cooled to room temperature, 100mL frozen water is added dropwise, point liquid, aqueous phase acetic acid second Ester extracts 2 × 100mL, collects ethyl acetate phase, and saturated common salt washing, anhydrous sodium sulfate drying rotates ethyl acetate, and vacuum is done It is dry, the compounds Ⅳ solid 4.4g of yellowish is obtained, yield is 83%, purity testing is carried out with HPLC, purity is 98%.
The characterize data of the compounds Ⅳ is:1H-NMR(400MHz,DMSO-d6):8.85(1H,s),7.44(1H,s), 4.34-4.36(4H,br),3.75-3.77(4H,br),3.34(6H,s)。
(4)Erlotinib(Compound V)Synthesis.
Compounds Ⅳ 2.02g is added into 20mL absolute ethyl alcohols, 40 DEG C are heated to, homogeneous suspension is formed, it is warm herein The lower ethanol solution 10mL for adding 3-aminophenylacetylene of degree, is to slowly warm up to after 70 DEG C, about 1h, solution clarification, after about 2h, TLC Raw material disappearance is monitored, stops reaction.
Reaction solution is cooled to room temperature, has a large amount of solids to separate out, solid is collected by filtration, be dried in vacuo white solid about 2.6g, n-butanol recrystallization, obtains Erlotinib sterling, yield is 95.2%, and purity testing, purity 99.89% are carried out with HPLC.
Compound V characterize data is:1H-NMR(400MHz,D2O-d6):8.43(1H,s),7.42-7.42(2H,J= 9.6Hz,d),7.34(1H,s),7.08-7.12(1H,J=6.8Hz,t),6.99-7.00(1H,J=6.8Hz,d),6.89(1H, s),4.20-4.25(4H,m),3.86-3.88(4H,m),3.55(1H,s)3.50(6H,s)。
Embodiment described above only expresses the several embodiments of the present invention, and it describes more specific and detailed, but simultaneously Therefore the limitation to the scope of the claims of the present invention can not be interpreted as.It should be pointed out that for one of ordinary skill in the art For, without departing from the inventive concept of the premise, various modifications and improvements can be made, these belong to the guarantor of the present invention Protect scope.Therefore, the protection domain of patent of the present invention should be determined by the appended claims.

Claims (10)

1. a kind of preparation method of Erlotinib, it is characterised in that comprise the following steps:
1) with 4,5- bis- (2- methoxy ethoxies) -2- p-nitriles for raw material, in the presence of reducing agent, while being reduced And hydrolysis, obtain 2- amidos -4,5- bis- (2- methoxy ethoxies) benzamide;The reducing agent is hydrated for RaneyNi- Hydrazine;
2) (2- methoxy ethoxies) benzamides of 2- amidos -4,5- two for obtaining above-mentioned steps and triethyl orthoformate are on road Reacted under the catalysis of Lewis acid, direct cyclization obtains intermediate 6, (2- the methoxy ethoxies) -3H- quinazolines of 7- bis- -4- Ketone;
3) (2- the methoxy ethoxies) -3H- quinazoline-4-ones of 6,7- bis- obtained above-mentioned steps carry out anti-with POCl3 Should, obtain 6,7- bis- (2- methoxy ethoxies)-quinazoline -4- chlorine;
4) obtain above-mentioned steps 6,7- bis- (2- methoxy ethoxies)-quinazoline -4- chlorine and an amido phenylacetylene react, and obtain To Erlotinib.
2. the preparation method of Erlotinib according to claim 1, it is characterised in that step 1) in, reaction dissolvent is water, Water-dioxane or water-ethanol;Reaction temperature is 0 DEG C -40 DEG C.
3. the preparation method of Erlotinib according to claim 2, it is characterised in that step 1) in, reaction dissolvent is water; Reaction temperature is 20 DEG C -40 DEG C.
4. the preparation method of Erlotinib according to claim 1, it is characterised in that step 2) in, the lewis acid For trifluoroacetic acid or BFEE;Reaction dissolvent is ethyl acetate, dichloromethane, ethanol, at least one of methanol.
5. the preparation method of Erlotinib according to claim 1, it is characterised in that step 2) in, 2- amido -4, 5- bis- (2- methoxy ethoxies) benzamide, triethyl orthoformate, lewis acidic consumption mol ratio is 1:1-1.5:0.05- 0.15;Reaction temperature is 70-80 DEG C.
6. the preparation method of Erlotinib according to claim 1, it is characterised in that step 3) in, reaction dissolvent is second At least one of acetoacetic ester, dichloromethane, dioxane.
7. the preparation method of Erlotinib according to claim 1, it is characterised in that step 3) in, (the 2- of 6,7- bis- Methoxy ethoxy) the consumption mol ratio of -3H- quinazoline-4-ones and POCl3 is 1:2-4;Reaction temperature is 60-80 DEG C.
8. the preparation method of Erlotinib according to claim 1, it is characterised in that step 4) in, reaction dissolvent is first At least one of alcohol, ethanol, isopropanol, n-butanol.
9. the preparation method of Erlotinib according to claim 1, it is characterised in that step 4) in, 6,7- bis- (2- methoxies Base oxethyl) the consumption mol ratio of-quinazoline -4- chlorine and an amido phenylacetylene is 1:1-1.3;Reaction temperature is 60-80 DEG C.
10. the preparation method of Erlotinib according to claim 1, it is characterised in that step 4) in, using recrystallization Method purifying erlotinib, the solvent of the recrystallization is ethanol, isopropanol, n-butanol, one kind in normal propyl alcohol.
CN201410127452.6A 2014-03-31 2014-03-31 The preparation method of Erlotinib Expired - Fee Related CN104945332B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201410127452.6A CN104945332B (en) 2014-03-31 2014-03-31 The preparation method of Erlotinib

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201410127452.6A CN104945332B (en) 2014-03-31 2014-03-31 The preparation method of Erlotinib

Publications (2)

Publication Number Publication Date
CN104945332A CN104945332A (en) 2015-09-30
CN104945332B true CN104945332B (en) 2017-10-17

Family

ID=54160463

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201410127452.6A Expired - Fee Related CN104945332B (en) 2014-03-31 2014-03-31 The preparation method of Erlotinib

Country Status (1)

Country Link
CN (1) CN104945332B (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105646374B (en) * 2015-12-31 2018-11-27 山东罗欣药业集团恒欣药业有限公司 A kind of preparation method of erlotinib Hydrochloride
CN107337648B (en) * 2016-05-03 2020-04-17 南京理工大学 Method for synthesizing erlotinib
CN106279585A (en) * 2016-08-26 2017-01-04 宁波圣达精工智能科技有限公司 A kind of preparation method of fire-retardant intelligent compact bookshelf
CN106361007A (en) * 2016-08-26 2017-02-01 宁波圣达精工智能科技有限公司 Smart compact bookshelf
CN108047144A (en) * 2018-01-26 2018-05-18 中国科学院化学研究所 The preparation method of Tarceva
CN113603650B (en) * 2021-08-03 2023-03-10 杭州职业技术学院 Catalytic cyclization preparation method and application of erlotinib key intermediate

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999067202A1 (en) * 1998-06-19 1999-12-29 Syngenta Participations Ag . Process for preparation of pyrimidinone derivatives
WO2003064377A1 (en) * 2002-01-29 2003-08-07 Ube Industries, Ltd. 4,5-substituted-2-formylaminobenzamides and processes for their production and conversion
CN101463013A (en) * 2007-12-21 2009-06-24 上海百灵医药科技有限公司 Preparation of erlotinid hydrochloride
CN102659629A (en) * 2012-02-14 2012-09-12 杭州华东医药集团生物工程研究所有限公司 Compound and application thereof in preparing erlotinib
CN103145629A (en) * 2013-04-02 2013-06-12 朱志成 Synthetic process of tandutinib for treating solid tumors

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999067202A1 (en) * 1998-06-19 1999-12-29 Syngenta Participations Ag . Process for preparation of pyrimidinone derivatives
WO2003064377A1 (en) * 2002-01-29 2003-08-07 Ube Industries, Ltd. 4,5-substituted-2-formylaminobenzamides and processes for their production and conversion
CN101463013A (en) * 2007-12-21 2009-06-24 上海百灵医药科技有限公司 Preparation of erlotinid hydrochloride
CN102659629A (en) * 2012-02-14 2012-09-12 杭州华东医药集团生物工程研究所有限公司 Compound and application thereof in preparing erlotinib
CN103145629A (en) * 2013-04-02 2013-06-12 朱志成 Synthetic process of tandutinib for treating solid tumors

Also Published As

Publication number Publication date
CN104945332A (en) 2015-09-30

Similar Documents

Publication Publication Date Title
CN104945332B (en) The preparation method of Erlotinib
CN101163698B (en) Method for the production of 5-(4-[4-(5-cyano-3-indolyl)-butyl]-1-piperazinyl)-benzofuran-2-carboxamide
CN109020881B (en) Preparation method of apatinib
CN103570633B (en) The preparation method of Gefitinib
CN102557977A (en) Synthesis intermediate of erlotinib and preparation method thereof
CN105837502A (en) Synthesis method of Vadadustat
CN109053625A (en) A kind of preparation method replacing benzothiazole C2 alkyl derivative
CN109206317B (en) Preparation process of amantadine nitrate derivative
CN103304492B (en) The synthetic method of a kind of EGFR inhibitor Dacomitinib
WO2022088573A1 (en) Method for preparing pyrotinib
CN109928890B (en) Preparation method of tolvaptan intermediate 2-methyl-4-N- (2-methylbenzoyl) benzoic acid
CN111333543B (en) Synthesis method of rilpivirine intermediate
CN101463011B (en) Process for synthesizing 3,4-dihydropyrimidine-2-keto
CN101367767B (en) Room temperature solvent-free synthesis of 3,4-dihydropyrimidine-2-ketone
CN103773360B (en) Schiff base fluorescent polymer and preparation method thereof
CN102153518B (en) Preparation method of Gefitinib
JP2017503020A (en) Process for the preparation of (E) -4-N, N-dialkylaminocrotonic acid in the form of an HX salt and its use for the synthesis of EGFR tyrosine kinase inhibitors
CN100378073C (en) Dindoly methylane and preparation process thereof
CN103804373A (en) Synthesis process of azasetron hydrochloride
CN111533742A (en) Method for synthesizing 2-methoxy trimethylpurine diketone by taking cyanamide as raw material
CN105175400A (en) Preparation method of Afatinib intermediate
CN102584636A (en) Synthetic method of kreatine
CN106883185B (en) Preparation method of 4-chloro-2-trifluoromethylpyrimidine
CN109232434A (en) A kind of new method synthesizing flibanserin
CN103242244B (en) Canertinib preparation method

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
CF01 Termination of patent right due to non-payment of annual fee

Granted publication date: 20171017

CF01 Termination of patent right due to non-payment of annual fee