CN106068262A - Quinazoline derivant and preparation method thereof and in application pharmaceutically - Google Patents

Quinazoline derivant and preparation method thereof and in application pharmaceutically Download PDF

Info

Publication number
CN106068262A
CN106068262A CN201580012570.5A CN201580012570A CN106068262A CN 106068262 A CN106068262 A CN 106068262A CN 201580012570 A CN201580012570 A CN 201580012570A CN 106068262 A CN106068262 A CN 106068262A
Authority
CN
China
Prior art keywords
acid
compound
chloro
cancer
methoxy
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
CN201580012570.5A
Other languages
Chinese (zh)
Other versions
CN106068262B (en
Inventor
李瑶
徐波
陈雷
李升�
刘建余
钱枚琳
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.)
Haisco Pharmaceutical Group Co Ltd
Original Assignee
Sichuan Haisco Pharmaceutical Co Ltd
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 Sichuan Haisco Pharmaceutical Co Ltd filed Critical Sichuan Haisco Pharmaceutical Co Ltd
Publication of CN106068262A publication Critical patent/CN106068262A/en
Application granted granted Critical
Publication of CN106068262B publication Critical patent/CN106068262B/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Abstract

The present invention relates to a kind of quinazoline derivant and preparation method thereof and in application pharmaceutically, specifically the present invention relates to quinazoline derivant or its stereoisomer, hydrate, metabolite, solvate, pharmaceutically acceptable salt, eutectic or prodrug, relate to their preparation method, relate to the compound medicine compositions including its pharmaceutical composition and the present invention in purposes pharmaceutically, particularly as the purposes of the double target spot inhibitor of EGFR/HER2.

Description

Quinazoline derivant and preparation method thereof and in application pharmaceutically Technical field
The present invention relates to a kind of quinazoline derivant and preparation method thereof and in application pharmaceutically, specifically a kind of novel quinazoline derivant or its stereoisomer, hydrate, solvate, metabolite, pharmaceutically acceptable salt, eutectic or prodrug with the double target spot inhibitory action of EGFR/HER2, its pharmaceutical composition and its in application pharmaceutically.
Background technology
Receptor tyrosine kinase superfamily in cell surface receptor is played an important role by extracellular growth factors to the regulation of cell signal.Receptor tyrosine kinase can catalytic phosphatase group be transferred to from ATP on the tyrosine group of substrate.When no ligand activation receptor tyrosine kinase, these kinases are in unphosphorylated free state, and its kinase domain is in inactive structure.When part is combined with the extracellular fragment of receptor tyrosine kinase, oligomerization, and autophosphorylation occur for acceptor, the binding site of signal protein are formd while the catalytic activity for increasing kinases, signal protein is in connection, so as to activate many signal paths.These signal paths are connected each other, propagation, existence, differentiation, function, migration and the apoptosis of regulating cell.When receptor tyrosine kinase loses regulation and control, during abnormal activation, cell can change into tumour cell, propagation, growth ability and resistance ability are improved, with stronger into vessel patency, invasiveness and transfer ability (Yarden and Sliwkowski, 2001, Nat Rev Mol Cell Biol, 2,127-137).
EGF (EGF) acceptor is the receptor tyrosine enzyme that a class receives much concern.EGF receptor family includes 4 members:EGFR/ErbB1/HER1, ErbB2/HER2/Neu, ErbB3/HER3 and ErbB4/HER4.The signal path of normal EGFR activation can adjust the propagation of cell, into blood vessel, growth, migration and adhesion, in cell and (Wu, the Zhong etc. of being played an important role in intercellular interaction of Organogenesis Process or adult, 2007, J Thorac Oncol, 2,430-439).Many molecule times can result in the sustained activation of EGFR kinase activity, constantly trigger substantial amounts of downstream signal transduction path, including K-ras activation.Therefore EGFR families with it is closely related into knurl.EGFR overexpression or mutation can be detected in the kinds cancers such as head and neck cancer, oophoroma, carcinoma of urinary bladder, cervical carcinoma, cancer of the esophagus, stomach cancer, breast cancer, endometrial carcinomas, colon cancer, lung cancer and brain tumor, generally indicate this bad prognosis.In addition, HER2 is considered as a kind of stronger oncoprotein, its mutation is also visible in kinds of tumors, especially breast cancer, lung cancer and colon cancer etc. (Olayioye, Neve etc., 2000, EMBO J, 19,3159-3167).
At present, a variety of EGFR EGFR-TKs micromolecular inhibitors are developed, the Gefitinib and Erlotinib of such as first generation, it optionally can reversibly suppress EGFR, so as to prevent the growth of tumour, especially lack (such as del747-750) containing exons 19 and exon 21 is mutated tumour (Barker, the Gibson etc. of (such as L858R), 2001, Bioorg Med Chem Lett,11,1911-1914;Wakeling, Guy etc., 2002, Cancer Res, 62,5749-5754).However, acquired resistance much occurs using the patient of Gefitinib and Erlotinib, this acquired resistance has 50%-60% to be due to that the T790M mutation that extron 20 occurs are caused.T790M mutation can influence kinase catalytic domain, weaken targeted inhibition agent and the interaction of target spot, make treatment invalid (Pao and Chmielecki, 2010, Nat Rev Cancer, 10,760-774).
Research finds that irreversible inhibitor can form covalent bond with the cysteine residues played a decisive role in the avtive spot of kinases, as a kind of strategy of potential suppression T790M mutation.And in EGFR families, only EGFR and HER2 have cysteine in corresponding site.Therefore this irreversible inhibitor has specificity (Singh, Dobrusin etc., 1997, J Med Chem, 40,1130-1135 of height to EGFR and HER2;Fry, Bridges etc., 1998, Proc Natl Acad Sci U S A, 95,12022-12027).Many irreversible double inhibitors have been developed (such as Afatinib, HKI-272 and OK a karaoke club replace Buddhist nun), compared to reversible inhibitor, these medicines not only can effectively suppress EGFR sensitizing mutations (such as del747-750, L858R) at low concentrations, and can effectively overcome the resistance that T790M is caused.It is worth noting that, in preclinical study, irreversible double inhibitors are unlike Gefitinib and Erlotinib generation quick drug resistance (Ercan, Zejnullahu etc., 2010, Oncogene, 29,2346-2356;Chmielecki, Foo etc., 2011, Sci Transl Med, 3,90ra59).
In order to meet clinical demand, it is necessary to continue research and development in the double target spot inhibitor of the EGFR/HER2 of resistance and safety that can effectively overcome T790M mutation to produce.
CN10679384 describes the novel amide derivative for suppressing growth of cancer cells, and its structural formula is as follows:
Wherein, A isR4、R5、R6And R7Each stand alone as hydrogen, halogen, N-C1-6Alkyl or N- alcohol amides base or C-C1-6Anti- the acylamino- (- NHCOC of alkyl1-6), hydroxycarbonyl group (- COOH), C1-6Alkoxy carbonyl (- COOC1-6)、C1-6Alkyl, or for by hydroxyl, C1-6Dialkylamine or the alkyl of heterocyclic group substitution;R1It is by the 1-5 X aryl replaced or heterocyclic group, or the C being substituted with aryl1-6Alkyl;R2It is hydrogen, hydroxyl, C1-6Alkoxy, or by C1-6Alkoxy or the C of heterocyclic group substitution1-6Alkoxy;R3It is hydrogen ,-COOH, C1-6Alkoxy carbonyl, or the unsubstituted acylamino-s of N- or the substituted acylamino-s of N-;naAnd nbRespectively 0-6 integer;X is hydrogen, halogen, hydroxyl, cyano group, nitro, (halo, dihalo or three halos) methyl, sulfydryl, C1-6Alkylthio group, acrylamido, C1-6Alkyl, C1-6Alkenyl, C1-6Alkynyl, C1-6Alkoxy, aryloxy group, C1-6Dialkyl amido, or the C to be replaced by Z1-6Alkyl or C1-6Alkoxy, condition is that X group can be fused together to form ring structure when X quantity is two or more;Y is hydroxyl, C1-6Alkyl, or the C replaced by Z1-6Alkyl, the C1-6Alkyl contains 1-4 It is individual to be selected from N, O, S, SO and SO2Group;And Z is C1-6Alkyl, aryl or heterocyclic group, the aromatic yl group are C5-12Monocyclic or bicyclic aromatic group, the heterocyclic group is to be selected from N, O, S, SO and SO containing 1-42Group C5-12Monocyclic or bicyclic aromatics or nonaromatic, and the aryl and heterocyclic group are unsubstituted, or it is selected from halogen, hydroxyl, amino, nitro, cyano group, C1-6Alkyl, C1-6Alkenyl, C1-6Alkynyl, C1-6Alkoxy, C1-6Alkyl monosubstituted amino and C1-6The substituent substitution of dialkyl amido.It is not considered as that it is a part of the invention to be specifically described in this patent.
CN102731485 describes 4- (substitution phenylamino) quinazoline derivant, and its structural formula is as follows:
R1It is selected from R2Selected from hydrogen or N, N- dimethylamino methyl;R3Selected from methoxyethyl, tetrahydrofuran -3- bases, (S)-tetrahydrofuran -3- bases or (R)-tetrahydrofuran -3- bases, and each R4And R5Independently selected from one or more hydrogen, halogen, C1-C10Alkyl, C1-C10Alkoxy and halo C1-C10Alkyl;And if only if R2For hydrogen, R3During for methoxyethyl, R1It is notAnd if only if R2For N, N- dimethylamino methyls, R3During for tetrahydrofuran -3- bases, R1It is notIt is not considered as that it is a part of the invention to be specifically described in this patent.
WO2011029265 describes quinazoline derivant, and its structural formula is as follows:
Wherein, A is selected from carbon atom or nitrogen-atoms;When A is carbon atom, R1Selected from hydrogen atom or alkoxy, wherein described alkoxy is optionally further replaced by one or more substituents selected from halogen or alkoxy, R2Selected from cyano group;When A is nitrogen-atoms, R1Selected from hydrogen atom or alkoxy, wherein described alkoxy is optionally further replaced by one or more substituents selected from halogen or alkoxy, R2It is unsubstituted;R3With having structure:- D-T-L or-D;D is selected from aryl or heteroaryl, wherein described aryl or heteroaryl are optionally further selected from halogen, alkyl or three by one or more independently of one another The substituent of methyl fluoride is replaced;T is selected from-(CH2)r-、-O(CH2)r-、-NH(CH2) r- or-S (O) r (CH2)r-;L is selected from aryl or heteroaryl, wherein described aryl or heteroaryl are optionally further replaced by one or more halogens or alkyl independently of one another;R4And R5It is each independently selected from hydrogen atom, alkyl, alkoxy, hydroxyl, hydroxyalkyl, halogen, carbonyl, amino, cyano group, nitro, carboxylic acid or carboxylate;B is selected from carbon atom, oxygen atom or S (O) r groups;When B is carbon atom, R6And R7It is each independently selected from hydrogen atom, alkyl, alkoxy, hydroxyl, hydroxyalkyl, halogen, carbonyl, amino, cyano group, nitro, carboxylic acid or carboxylate;When B is oxygen atom or S (O) r groups, R6And R7It is unsubstituted;R8Selected from hydrogen atom or alkyl;R9Selected from hydrogen atom, alkyl, aryl, carboxyl or carboxylate;R is 0,1 or 2;And n is 1,2,3,4 or 5.It is not considered as that it is a part of the invention to be specifically described in this patent.
The content of the invention
The present invention provides a class formation novel compound, shows by research, and the compounds of this invention shows excellent drug activity as the double target spot inhibitor of EGFR/HER2.
The invention provides a kind of formula (II0) shown in compound or its stereoisomer, hydrate, metabolite, solvate, pharmaceutically acceptable salt, eutectic or prodrug,
Wherein:
R2Choosing:From C1-3Alkyl or 4 to 8 circle heterocycles bases, described heterocyclic radical are optionally further selected from F, Cl, hydroxyl, C by 0 to 21-3Alkyl or C1-3The substituent of alkoxy is replaced, and described heterocyclic radical contains 1 to 2 hetero atom for being selected from N or O;
Ring A is selected from
R3Selected from F, CF3、CHF2、CH2F, Cl, hydroxyl or C1-3Alkyl;
X1ForOr X1Formed together with ring A
R4、R5And R6Respectively H;
R9Selected from F, Cl, hydroxyl, C1-3Alkyl or C2-3Alkynyl;
N, m, t each are selected from 0,1,2,3 or 4;
Restrictive condition is, when n is 0, R2Selected from C1-3Alkyl, and m be 0 when, ring A is not
Preferably, compound of the invention is selected from the compound shown in logical formula (II):
Wherein:
R2Selected from methyl, ethyl or
Ring A is selected from
N is 0 or 1;
R3Selected from F;
M is 0 or 1;
R4、R5And R6It is H;
R9Selected from F or Cl;
T is 3;
Restrictive condition is, when n is 0, R2Selected from methyl or ethyl, and m, when being 0, ring A is not
Specifically, compound of the present invention or its stereoisomer, hydrate, metabolite, solvate, pharmaceutically acceptable salt, eutectic or prodrug are selected from, but are not limited to:
According to specific embodiments of the present invention, the compound or its stereoisomer of the present invention, hydrate, metabolite, solvate, pharmaceutically acceptable salt, eutectic or prodrug, wherein described salt is selected from hydrochloride, hydrobromate, sulfate, phosphate, acetate, trifluoroacetate, rhodanate, maleate, hydroxymaleic acid salt, glutarate, mesylate, esilate, benzene sulfonate, tosilate, benzoate, salicylate, phenylacetate, cinnamate, lactate, malonate, pivalate, succinate, fumarate, malate, mandelate, tartaric acid Salt, gallate, gluconate, laruate, palmitate, pectate, picrate, citrate or combinations thereof, it is preferred that, described salt is selected from hydrochloride, hydrobromate, sulfate, phosphate, acetate, maleate, mesylate, benzene sulfonate, trifluoroacetate, tosilate, benzoate, salicylate, cinnamate, lactate, malonate, succinate, fumarate, malate, tartrate, citrate or combinations thereof, more preferably hydrochloride and maleate.
Present invention also offers a kind of pharmaceutical composition, described composition includes:The compound of the present invention or its stereoisomer, hydrate, metabolite, solvate, pharmaceutically acceptable salt, eutectic or prodrug of effective dose, and pharmaceutically acceptable carrier, diluent, adjuvant, medium or excipient.
According to specific embodiments of the present invention, pharmaceutical composition of the invention still further comprises one or more other therapeutic agents.
According to specific embodiments of the present invention, the other therapeutic agents described in pharmaceutical composition of the invention include:Cis-platinum (cisplatin),Carboplatin (carboplatin),Oxaliplatin (oxaliplatin),Dacarbazine (dacarbazine),Temozolomide (temozolomide),Procarbazine (procarbazine),Methotrexate (MTX) (methotrexate),Fluorouracil (fluorouracil),Cytarabine (cytarabine),Gemcitabine (gemcitabine),Purinethol (mercaptopurine),Fludarabine (fludarabine),Vincaleukoblastinum (vinblastine),Vincristine (vincristine),Vinorelbine (vinorelbine),Taxol (paclitaxel),Docetaxel (docetaxel),TPT (topotecan),Irinotecan (irinotecan),Etoposide (etoposide),ET-743 (trabectedin),Dactinomycin D (dactinomycin),Doxorubicin (doxorubicin),Epirubicin (epirubicin),Daunomycin (daunorubicin),Mitoxantrone (mitoxantrone),Bleomycin (bleomycin),Mitomycin C (mitomycin),Ipsapirone (ixabepilone),TAM (tamoxifen),Flutamide (flutamide),Sirolimus (sirolimus),Afatinib(afatinib),alisertib,amuvatinib,A Pa replaces Buddhist nun (apatinib),Axitinib (axitinib),Bortezomib (bortezomib),SKI-606 (bosutinib),Bu Linibu (brivanib),Card is rich to replace Buddhist nun (cabozantinib),AZD2171 (cediranib),crenolanib,Ke Zhuo replaces Buddhist nun (crizotinib),Dabrafenib (Da Lafeini),dacomitinib,Da Lushe replaces (danusertib),Dasatinib (dasatinib),Many Weis replace Buddhist nun (dovitinib),Tarceva (erlotinib),foretinib,ganetespib,Gefitinib (Gefitinib),Buddhist nun (ibrutinib) is replaced according to Shandong,Conmana (icotinib),Imatinib (imatinib),iniparib,Lapatinib (lapatinib),lenvatinib,linifanib,linsitinib,Masitinib (masitinib),momelotinib,Not for husky Buddhist nun (motesanib),HKI-272 (neratinib),Nilotinib (nilotinib),niraparib,oprozomib,olaparib,Pazopanib (pazopanib),pictilisib,ponatinib,quizartinib,regorafenib,rigosertib,rucaparib,ruxolitinib,Saracatinib (saracatinib),saridegib,Sorafenib (sorafenib),Sutent (sunitinib),tasocitinib,telatinib,tivantinib,tivozanib,tofacitinib,trametinib,ZD6474 (vandetanib),veliparib,Wei Luofeini (vemurafenib),vismodegib,volasertib,Alemtuzumab (alemtuzumab),Bevacizumab (bevacizumab),brentuximab vedotin,Catumaxomab (catumaxomab), Cetuximab (cetuximab), ground promise monoclonal antibody (denosumab), lucky trastuzumab (gemtuzumab), her monoclonal antibody (ipilimumab), Buddhist nun's trastuzumab (nimotuzumab), difficult to understand (ofatumumab), Victibix (panitumumab), Rituximab (rituximab), tositumomab (tositumomab), Herceptin (trastuzumab) or combinations thereof.
The application of pharmaceutical preparation is being prepared as a kind of EGFR/HER2 receptor tyrosine kinase inhibitors present invention also offers described compound or its stereoisomer, hydrate, ester, metabolite, solvate, pharmaceutically acceptable salt, eutectic or prodrug or described pharmaceutical composition, particularly for preparing the application in being used to treat and/or prevent the pharmaceutical preparation of excess proliferative disease.
According to specific embodiments of the present invention, the compound or its stereoisomer of the present invention, hydrate, ester, metabolite, solvate, pharmaceutically acceptable salt, in the application of eutectic or prodrug or described pharmaceutical composition, the excess proliferative disease includes brain tumor, non-small cell lung cancer, epidermis squamous carcinoma, carcinoma of urinary bladder, cancer of pancreas, colon cancer, breast cancer, oophoroma, cervix cancer, carcinoma of endometrium, colorectal cancer, kidney, adenocarcinoma of esophagus, esophageal squamous cell carcinoma, solid tumor, NHL, liver cancer, lung cancer, stomach cancer, cutaneum carcinoma, thyroid cancer, head and neck cancer, prostate cancer, one or more in glioma and nasopharyngeal carcinoma, it is preferred that non-small cell lung cancer, breast cancer, epidermis squamous carcinoma, one or more in stomach cancer and colon cancer.
Detailed description of the invention
Unless there are opposite statement, the term used in the specification and in the claims has following implications.
Involved carbon, hydrogen, oxygen, sulphur, nitrogen or halogen include their isotope situation in group of the present invention and compound, and involved carbon, hydrogen, oxygen, sulphur or nitrogen is optionally further substituted by their one or more corresponding isotopes in group of the present invention and compound, the isotope of wherein carbon includes12C、13C and14C, the isotope of hydrogen includes protium (H), deuterium (D is called heavy hydrogen), tritium (T is called superheavy hydrogen), and the isotope of oxygen includes16O、17O and18O, the isotope of sulphur includes32S、33S、34S and36S, the isotope of nitrogen includes14N and15N, the isotope of fluorine includes17F and19F, the isotope of chlorine includes35Cl and37Cl, the isotope of bromine includes79Br and81Br。
" pharmaceutically acceptable salt " or " its pharmaceutically acceptable salt " refers to that the compounds of this invention keeps the biological effectiveness and characteristic of free alkali, and described free alkali passes through the salt with nontoxic inorganic acid or organic acid reaction acquisition.The non-limiting example of described inorganic acid includes hydrofluoric acid, hydrochloric acid, hydrobromate, sulfate, phosphoric acid, hypochlorous acid, perchloric acid, acid iodide, carbonic acid, nitrous acid, nitroxylic acid, metaboric acid, boric acid, metasilicic acid, silicic acid, metaphosphorous acid, metaphosphoric acid, pyrophosphoric acid, hydrosulphuric acid, sulfurous acid, thiosulfuric acid and permanganic acid;Described organic acid non-limiting example include formic acid, acetic acid, trifluoroacetic acid, thiocyanic acid, maleic acid, hydroxymaleic acid, glutaric acid, methanesulfonic acid, ethyl sulfonic acid, benzene sulfonic acid, to toluene sulphur Acid, benzoic acid, salicylic acid, phenylacetic acid, cinnamic acid, lactic acid, malonic acid, pivalic acid, butanedioic acid, fumaric acid, malic acid, mandelic acid, tartaric acid, gallic acid, gluconic acid, laurate, palmitic acid, pectic acid, picric acid and citric acid.
" carrier " refers to that obvious stimulation will not be produced to organism and will not eliminate the bioactivity of given compound and the material of characteristic.
" excipient " refers to be added in pharmaceutical composition to promote the inert substance that compound is administered.Non-limiting example includes calcium carbonate, calcium phosphate, sugar, starch, cellulose derivative (including microcrystalline cellulose), gelatin, vegetable oil, polyethylene glycols, diluent, granulating agent, lubricant, adhesive and disintegrant.
" adjuvant " is nonspecific immunity strengthening agent, when being injected together with antigen or being previously implanted body, can strengthen the former immune response of body fight or change type of immune response.
" diluent " is also named " filler ".When active compound is processed into pulvis, or in order to make it easy to spray the inert substance being diluted added.Such as:Clay, kaolin, clay, talcum powder etc..
" prodrug " refers to can be the compounds of this invention with bioactivity through biotransformationin vivo, and such conversion is hydrolyzed or is that precursor structure is influenceed through enzymatic conversion in blood tissues in blood by pro-drug.The prodrug of the present invention is prepared by modifying the functional group in the compounds of this invention, and the modification can be removed by conventional operation or in vivo, and obtain parent compound.
" eutectic " refers to the crystal that active pharmaceutical ingredient and eutectic formation are combined into the presence of hydrogen bond or other non-covalent bonds, and wherein API and CCF pure state are the presence of fixed stoichiometric proportion between solid, and each component at room temperature.Eutectic is a kind of multicomponent crystal, both comprising the binary eutectic formed between two kinds of neutral solids, also comprising neutral solid with or solvate formation multi-element eutectic.The non-limiting example of " the eutectic formation " includes alanine, valine, leucine, isoleucine, proline, phenylalanine, tryptophan, methionine, glycine, serine, threonine, cysteine, tyrosine, asparagine, glutamine, lysine, arginine, histidine, aspartic acid, L-aminobutanedioic acid, glutamic acid, pyroglutamic acid, sulfuric acid, phosphoric acid, nitric acid, hydrobromic acid, acid, formic acid, acetic acid, propionic acid, benzene sulfonic acid, benzoic acid, phenylacetic acid, salicylic acid, alginic acid, anthranilic acid, camphoric acid, citric acid, vinyl sulfonic acid, formic acid, fumaric acid, furancarboxylic acid, gluconic acid, glucuronic acid, glutamic acid, glycolic, isethionic acid, lactic acid, maleic acid, malic acid, mandelic acid, mucic acid, pamoic acid, pantothenic acid, stearic acid, butanedioic acid, sulfanilic acid, tartaric acid, p-methyl benzenesulfonic acid, malonic acid, 2 hydroxy propanoic acid, oxalic acid, glycolic, glucuronic acid, galacturonic acid, citric acid, cinnamic acid, p-methyl benzenesulfonic acid, methanesulfonic acid, ethyl sulfonic acid or trifluoromethanesulfonic acid, ammonia, isopropylamine, Trimethylamine, diethylamine, triethylamine, tripropylamine, diethanol amine, monoethanolamine, dimethylethanolamine, DMAE, 2- DEAE diethylaminoethanols, dicyclohexylamine, caffeine, procaine, choline, glycine betaine, Benethamine Penicillin, ethylenediamine, gucosamine, methylglucosamine, theobromine, triethanolamine, tromethamine, purine, piperazine, piperidines and N-ethylpiperidine.
" stereoisomer " refers to as the isomers produced by the spatially arrangement mode difference of atom in molecule, including cis-trans-isomer, enantiomter and rotamer.
" optional " or " optionally " or " selective " or " optionally " refer to then described event or situation can with but may not occur, the description includes the situation and wherein nonevent situation for wherein occurring the event or situation.For example, " optionally by alkyl-substituted heterocyclic radical " refer to the alkyl can with but may not exist, the description includes wherein heterocyclic radical by alkyl-substituted situation, and wherein heterocyclic radical is not by alkyl-substituted situation.
Embodiment
Below by way of specific embodiment describe in detail the present invention implementation process and produce beneficial effect, it is intended to help reader more fully understand the present invention essence and feature, not as to this case can practical range restriction.
The structure of compound by nuclear magnetic resonance (NMR) or (and) mass spectrum (MS) determines.NMR measure is to use (Bruker Avance III 400 and Bruker Avance 300) nuclear magnetic resonance spectrometer, and measure solvent is deuterated dimethyl sulfoxide (DMSO-d6), deuterochloroform (CDCl3), deuterated methanol (CD3OD), inside it is designated as tetramethylsilane (TMS).
MS measure uses (Agilent 6120B (ESI) and Agilent 6120B (APCI)).
HPLC measure uses Agilent 1260DAD high pressure liquid chromatographs (Zorbax SB-C18100 × 4.6mm).
Tlc silica gel plate uses Yantai Huanghai Sea HSGF254 or Qingdao GF254 silica gel plates, and the specification that the silica gel plate that thin-layered chromatography (TLC) is used is used is 0.15mm~0.20mm, and the specification that thin-layer chromatography isolates and purifies product use is 0.4mm~0.5mm.
Column chromatography is carrier typically using the mesh silica gel of Yantai Huanghai Sea silica gel 200~300.
Oneself initiation material known of the present invention can be used or synthesized according to methods known in the art, or be can purchase in the smooth science and technology of Thailand, pacified the companies such as silent resistance to Jilin Chemical, Shanghai moral, the imperial chemical industry of Chengdu section, splendid remote chemistry science and technology, lark prestige science and technology.
Blanket of nitrogen refers to that reaction bulb connects the nitrogen balloon of an about 1L volume.
Nitrogen atmosphere refers to that reaction bulb connects the hydrogen balloon of an about 1L volume.
Hydrogenation is generally vacuumized, and is filled with hydrogen, is operated 3 times repeatedly.
Without specified otherwise in embodiment, reaction is carried out under nitrogen atmosphere.
Without specified otherwise in embodiment, solution refers to the aqueous solution.
Without specified otherwise in embodiment, the temperature of reaction is room temperature.
Room temperature is optimum reaction temperature, is 20 DEG C~30 DEG C.
Other symbols used herein have following meaning:
Bn:Benzyl;Me:Methyl;Et:Ethyl;Ts:P-toluenesulfonyl;TBS:T-Butyldimethylsilyl;Boc: Tertbutyloxycarbonyl;Ac:Acetyl group.
Intermediate 1:The chloro- 7- methoxy-quinazolines -6- alcohol (1d) of 4-
4-chloro-7-methoxy-quinazolin-6-ol
The first step:(4- hydroxyl -7- methoxy-quinazoline -6- bases) acetic acid esters (1b)
(4-hydroxy-7-methoxy-quinazolin-6-yl)acetate
6- hydroxyl -7- methoxyl group -3H- quinazoline-4-ones 1a (40g, 208mmol, Kang Manlin) and acetic anhydride (296mL are added into reaction bulb, 211mmol), after stirring, add pyridine (67mL), rise to 100 DEG C stirring 4 hours after be cooled to room temperature.Reaction solution is poured into frozen water (500g), after stirring 5 minutes, suction filtration is depressurized, successively filter cake is washed with water (100mL × 2), petroleum ether (100mL × 2), filter cake is collected, drying obtains 1b (36g, yield of gray solid shape:73%).
Second step:(the chloro- 7- methoxy-quinazolines -6- bases of 4-) acetic ester hydrochloride (1c)
(4-chloro-7-methoxy-quinazolin-6-yl)acetate hydrochloride
1b (30g are added into reaction bulb, 130mmol) with thionyl chloride (205mL, 128mmol), POCl3 (38mL, 126mmol) and DMF (3mL) is slowly added dropwise, after stirring, rise to 120 DEG C to stir 4 hours, be cooled to room temperature, be concentrated under reduced pressure to obtain residue.Toluene is added into residual species, the rotation that is concentrated under reduced pressure removes solvent, repeats this step twice, is concentrated under reduced pressure to give the 1c crude products (289g) of sepia solid-like, is directly used in the next step.
3rd step:The chloro- 7- methoxy-quinazolines -6- alcohol (1d) of 4-
4-chloro-7-methoxy-quinazolin-6-ol
1c (325.5g, 100mmol) and methanolic ammonia solution (7mol/L, 560mL) are added into reaction bulb, is stirred at room temperature 1 hour.Suction filtration, is washed with methanol (100mL × 2), is collected filter cake, is dried to obtain 1d (17g, yield of gray solid shape:65.4%).
Intermediate 2:The fluoro- 4- hydroxy-piperdines -1- t-butyl formates (2b) of (3S) -3-
tert-butyl(3S)-3-fluoro-4-hydroxy-piperidine-1-carboxylate
3- fluorin-4-oxygens are added into reaction bulb for piperidines -1- t-butyl formates 2a (20g, 92mmol) and methanol (70mL), after stirring, it is cooled to 0 DEG C, sodium borohydride (7g, 184mmol) is added portionwise, reaction 1 hour is warmed to room temperature.It is concentrated under reduced pressure, ethyl acetate (200mL) is added into residue, saturated aqueous ammonium chloride (200mL), water (300mL), is extracted with ethyl acetate (300mL × 3), merges organic phase, anhydrous sodium sulfate drying, filtering, filtrate decompression is concentrated to give 2b (20g, yield of white solid:90%).
Fast purifying chromatograph (Biotage, Isolera one, eluant, eluent:5% ethyl acetate/petroleum ether (v/v)), obtain the 2b-1 and 2b-3 of white solid mixture (8g, yield:40%) with the 2b-2 and 2b-4 of white solid mixture (10g, yield:50%).
Intermediate 3:4- (the chloro- 2,4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- alcohol (3c)
4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-ol
The first step:[4- (the chloro- 2,4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- bases] acetic acid esters (3b)
[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]acetate
1c (34.0g, 0.135mol), 2,4 two fluoro- 3- chloroanilines (24.3g, 0.148mol) and isopropanol (400mL) are sequentially added into reaction bulb, 85 DEG C is heated to and reacts 3 hours.Room temperature is cooled to, suction filtration collects filter cake, is dried to obtain the 3b (51.0g) of gray solid shape.
1H NMR(400MHz,CDCl3)δ8.71(s,1H),8.37(td,1H),7.56(s,1H),7.36(s,1H),7.07(td,1H),3.98(s,3H),2.41(s,3H)。
Second step:4- (the chloro- 2,4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- alcohol (3c)
4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-ol
3b (51.0g, 0.135mol) and 7M methanolic ammonia solution (400mL) are added into reaction bulb, is stirred at room temperature 1 hour.Suction filtration, is washed with methanol (100mL × 2), is collected filter cake, is dried to obtain 3c (23.0g, yield of gray solid shape:50.0%).
MS m/z(ESI):338.0[M+1]。
Intermediate 4:(3aS, 6aR) -5- hydroxyls -3,3a, 4,5,6,6a- hexahydro -1H- pentamethylene [c] pyrroles -2- t-butyl formates (4d)
tertbutyl-(3aS,6aR)-5-hydroxy-3,3a,4,5,6,6a-hexahydro-1H-cyclopenta[c]pyrrole-2-carboxy late
The first step:N- pi-allyl-N- Propargyls t-butyl carbamates (4b)
tert-butyl N-allyl-N-prop-2-ynyl-carbamate
Under ice bath, N- tertbutyloxycarbonyl allylamine 4a (20g are added into reaction bulb, 127.39mmoL) and N, dinethylformamide (100mL), adding sodium hydride, (60% is scattered in paraffin oil, 7.6g, 191.08mmol), stirred 30 minutes at 0 DEG C after completion of dropping, naturally it is warmed to room temperature stirring 20 minutes, propargyl bromide (30.3g is added dropwise under ice bath, 245.78mmol), stirred 30 minutes at 0 DEG C, reaction solution is poured slowly into ice cube (200g), extracted with ethyl acetate (100mL × 3), merge organic phase, saturation drinking water (100mL × 3) is washed, anhydrous sodium sulfate drying, filtering, silica gel column chromatography separating-purifying (petrol ether/ethyl acetate (v/v)=100:1~50:1) 4b (21g, yield of yellow liquid are obtained:85%).
MS m/z(ESI):140.1[M+1]。
Second step:(3aS, 6aR) -5- oxos -1,3,3a, 4,6,6a- hexahydro pentamethylene simultaneously [c] pyrroles -2- t-butyl formates (4c)
tert-butyl(3aR,6aS)-5-oxo-1,3,3a,4,6,6a-hexahydrocyclopenta[c]pyrrole-2-carboxylate
4b (39g are sequentially added into reaction bulb, 202.2mmoL), methyl ether (275mL), cobalt octacarbonyl (83g, 242.7mmoL) with water (82.4mL), it is heated to reflux being stirred overnight, it is concentrated under reduced pressure, saturation drinking water (500mL) is added into residue, the hydrochloric acid (240mL) of ethyl acetate (250mL) and 1M, it is stirred at room temperature 5 minutes, stand a point liquid, aqueous phase is extracted with ethyl acetate (250mL × 3), merge organic phase, saturation drinking water (250mL × 2) is washed, organic phase anhydrous sodium sulfate drying, filtering, it is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (petrol ether/ethyl acetate (v/v)=3:1~2:1) 4c (27.6g, yield of faint yellow solid shape are obtained:61.3%).
MS m/z(ESI):170.1[M+1]。
1H NMR(400MHz,CDCl3):δ3.68-3.64(m,2H),3.24-3.21(d,2H),2.97-2.88(m,2H),2.52-2.45(dd,2H),2.20-2.14(dd,2H),1.46(s,9H)。
3rd step:(3aS, 6aR) -5- hydroxyls -3,3a, 4,5,6,6a- hexahydro -1H- pentamethylene simultaneously [c] pyrroles -2- t-butyl formates (4d)
tert-butyl-(3aS,6aR)-5-hydroxy-3,3a,4,5,6,6a-hexahydro-1H-cyclopenta[c]pyrrole-2-carbox ylate
4c (1.5g, 6.7mmol) is added into reaction bulb, after stirring, ice bath is cooled to 0 DEG C, adds sodium borohydride (0.38g, 10mmol), 0 DEG C is reacted 30 minutes after adding.Saturated ammonium chloride (20mL) is added dropwise into reaction solution, add ethyl acetate (50mL), aqueous layer with ethyl acetate extracts (40mL × 2), merge organic phase, washed (40mL × 1), organic phase anhydrous sodium sulfate drying, filtered with saturation drinking water, it is concentrated under reduced pressure, silica gel column chromatography separating-purifying (petrol ether/ethyl acetate (v/v)=1:1) 4d (1.4g, yield of colorless oil are obtained:93%).
1H NMR(400MHz,CDCl3)δ4.30(p,1H),3.50(dd,2H),3.35(dd,2H),2.63-2.58(m,2H),2.25–2.08(m,2H),1.51(dd,2H),1.46(s,9H)。
Intermediate 5:The fluoro- 4- hydroxy-piperdines -1- t-butyl formates (5c) of (3R) -3-
tert-butyl(3R)-3-fluoro-4-hydroxy-piperidine-1-carboxylate
The first step:(R)-(5- ethyl quinine -2- bases)-(6- methoxyl group -4- quinolyls) methylamine tri hydrochloride (5b)
(R)-(5-ethylquinuclidin-2-yl)-(6-methoxy-4-quinolyl)methanamine trihydrochloride
Hydroquinine is added into reaction bulb and determines 5a (25g, 76.7mmol) with triphenylphosphine (30g, 115mmol), add tetrahydrofuran (250mL), it is cooled to 0 DEG C, diisopropyl azodiformate (26.5g is added dropwise, 130.4mmol), 0 DEG C is reacted 10 minutes, diphenyl phosphate azide (36g is added dropwise, 130.4mmol), room temperature reaction is stayed overnight, triphenylphosphine (30g is added portionwise, 115mmol), 45 DEG C are reacted 4 hours, add water (25g), 45 DEG C of reactions are stayed overnight, it is concentrated under reduced pressure, add dichloromethane (250mL) and 2M hydrochloric acid (150mL), stirring 20 minutes, separate water layer, extracted with dichloromethane (250mL × 2), water layer is concentrated, with ethyl acetate (150mL), methanol (50mL) is beaten, with ethyl acetate (150mL), methanol (30mL) is washed, filter out solid, it is dried to obtain the 5b (26g of white solid, yield 77.8%).
1H NMR(400MHz,DMSO+D2O)δ9.04(d,1H),8.24(dt,2H),7.94(t,1H),7.71(dd,1H),6.06(d,1H),4.57(q,1H),4.09–4.01(m,3H),3.66–3.35(m,4H),1.96–1.74(m,4H),1.41–1.22(m,3H),1.07–0.96(m,1H),0.85(t,3H)。
Second step:The fluoro- 4- hydroxy-piperdines -1- t-butyl formates (5c) of (3R) -3-
tert-butyl(3R)-3-fluoro-4-hydroxy-piperidine-1-carboxylate
Take 5b (2.03g, 4.63mmol) it is dissolved in 46.3mL tetrahydrofuran, ice-water bath is cooled down, and saturated aqueous sodium carbonate is adjusted to neutrality, is rotated to dry, with toluene band water three times, 46.3mL tetrahydrofuran is added, filtering adds trichloroacetic acid (0.75g, 4.63mmol), water is configured to the tetrahydrofuran solution of 0.1M (R)-(5- ethyl quinine -2- bases)-(6- methoxyl group -4- quinolyls) methylamine list trichloroacetic acid monohydrate.Double benzsulfamide (the 3.77g of N- fluoro are added into reaction bulb, 11.6mmol) with sodium carbonate (1.85g, 17.3mmol), add tetrahydrofuran (33mL), it is cooled to -20 DEG C, add above-mentioned 0.1M (R)-((1S, 2R, 4S, 5R) -5- ethyls quinine -2- bases) (6- methoxy quinoline -4- bases) methylamine list trichloroacetic acid monohydrate tetrahydrofuran solution, -20 DEG C are reacted 10 minutes, add N- tertbutyloxycarbonyl -4- piperidones (4.63g, 23.1mmol), -20 DEG C of reactions are stayed overnight, add methyl tertiary butyl ether(MTBE) (50mL), stirring 30 minutes, filtering, filtrate decompression is concentrated, silica gel column chromatography separating-purifying (petrol ether/ethyl acetate (v/v)=5:1) 5c (1.9g, yield 74.8%) of white solid is obtained.
1H NMR(400MHz,CDCl3)δ4.84(ddd,1H),4.47(s,1H),4.27–4.10(m,1H),3.37–3.19 (m,2H),2.64–2.46(m,2H),1.50(s,9H)。
19F NMR(400MHz,CDCl3) δ -195.86~-196.43 (1F).
By 5c (1.9g, 8.7mmol) it is dissolved in methanol (8mL), it is cooled to 0 DEG C, plus sodium borohydride (0.5g, 13.0mmol), 0 DEG C is reacted 2.5 hours, adds saturated aqueous ammonium chloride (20mL), is stirred 20 minutes, it is concentrated under reduced pressure, water (45mL) is added, is extracted with ethyl acetate (25mL × 3), merges organic layer, with anhydrous sodium sulfate drying, filtering, filtrate decompression is concentrated, silica gel column chromatography separating-purifying (petrol ether/ethyl acetate (v/v)=7:1) 5c-1 (0.6g, yield 30%, ee of white solid, are obtained>99%) with 5c-2 (0.45g, yield 22.5%, ee>99%)
Intermediate 6:4- (the 3,4 2 chloro- fluoro- aniline of 2-) -7- methoxy-quinazoline -6- alcohol (6c)
4-((3,4-dichloro-2-fluoro-anilino)-7-methoxyquinazolin-6-ol
The first step:[4- (the 3,4 2 fluoro- chloro- aniline of 2-) -7- methoxy-quinazoline -6- bases] acetic acid esters (6b)
[4-(3,4-difluoro-2-chloro-anilino)-7-methoxy-quinazolin-6-yl]acetate
1c (32.4g, 0.129mol), 3,4 two fluoro- 2- chloroanilines (32.6g, 0.155mol) and isopropanol (250mL) are sequentially added into reaction bulb, 100 DEG C is heated to and reacts 4 hours.Room temperature is cooled to, suction filtration collects filter cake, is dried to obtain the 6b (51.1g) of gray solid shape, is directly used in the next step.
MS m/z(ESI):396.0[M+1]。
Second step:4- (the 3,4 2 fluoro- chloro- aniline of 2-) -7- methoxy-quinazoline -6- alcohol (6c)
4-(3,4-dichloro-2-fluoro-anilino)-7-methoxyquinazolin-6-ol
6b (51.1g, 0.129mol) and 7M methanolic ammonia solution (250mL) are added into reaction bulb, is stirred at room temperature 1 hour.Suction filtration, is washed with methanol (100mL × 2), is collected filter cake, is dried to obtain 6c (30g, yield of gray solid shape:64.2%).
1H NMR(400MHz,DMSO)δ8.36(s,1H),7.65(s,1H),7.62–7.50(m,2H),7.21(s,1H),4.00–3.92(m,3H)。
19F NMR(400MHz,DMSO)δ-112.23(1F)。
MS m/z(ESI):338.0[M+1]。
Embodiment 1:1- [4- [[4- (the chloro- 2,4- difluoro-anilines bases of 3-) -7- methoxy-quinazoline -6- bases] epoxide methyl] -1- piperidyls] propyl group -2- alkene -1- ketone (compound 1)
1-[4-[[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxymethyl]-1-piperidyl]prop-2-en-1-one
The first step:4- [(the chloro- 7- methoxy-quinazolines -6- bases of 4-) epoxide methyl] piperidines -1- t-butyl formates (1B)
tert-butyl4-[(4-chloro-7-methoxy-quinazolin-6-yl)oxymethyl]piperidine-1-carboxylate
1d (3.0g are added into reaction bulb; 14.2mmol), triphenylphosphine (5.58g; 21.3mmol) with dichloromethane (50mL); nitrogen is protected; ice bath is cooled to 0 DEG C, and dichloromethane (10mL) solution of diisopropyl azodiformate (4.30g, 21.3mmol) is added dropwise; after completion of dropping, reaction 30 minutes is warmed to room temperature.4- hydroxymethyl piperidine -1- t-butyl formates (4.58g, 21.3mmol) are slowly added at 0 DEG C, reaction 12 hours is warmed to room temperature.It is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (petrol ether/ethyl acetate (v/v)=10:1) the 1B crude products (5.80g) of white solid are obtained, the next step is directly used in.
Second step:4- [[4- (3- chlorine 2,4 difluorobenzenes amine) -7- methoxy-quinazoline -6- bases] oxygen methyl] piperidines -1- t-butyl formates (1C)
tert-butyl 4-[[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxymethyl]piperidine-1-carboxylate
1B (5.80g are sequentially added into reaction bulb, 14.2mmol), 2, the chloro- 3- chloroanilines (2.6g of 4- bis-, 15.6mmol) with isopropanol (30mL), 120 DEG C are heated to react 2 hours, it is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1~50:1~30:1) 1C (3.80g, yield of white solid are obtained:50.0%).
MS m/z(ESI):535.1[M+1]。
3rd step:N- (the chloro- 2,4- difluorophenyls of 3-) -7- methoxyl groups -6- (4- piperidyl methyls epoxide) quinazoline -4- amine two (trifluoroacetic acid) salt (1D)
N-(3-chloro-2,4-difluoro-phenyl)-7-methoxy-6-(4-piperidylmethoxy)quinazolin-4-amine ditrifluoroacetic acid
Added into reaction bulb and trifluoroacetic acid (20mL) is added dropwise under 1C (3.80g, 0.93mmol) and dichloromethane (20mL), ice-water bath, ice bath is removed in completion of dropping recession, is reacted at room temperature 2 hours.It is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=10:1) 1D (3.30g, yield of white solid are obtained:68.0%).
4th step:1- [4- [[4- (the chloro- 2,4- difluoro-anilines bases of 3-) -7- methoxy-quinazoline -6- bases] epoxide methyl] -1- piperidyls] propyl group -2- alkene -1- ketone (compound 1)
1-[4-[[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxymethyl]-1-piperidyl]prop-2-en-1-one
1D (3.30g are sequentially added into reaction bulb, 4.93mmol), triethylamine (4.1mL) and dichloromethane (15mL), dry ice ethanol bath is cooled to -60 DEG C, acryloyl chloride (0.45mL is added dropwise, dichloromethane (2mL) solution 5.48mmol), is warmed to room temperature reaction 30 minutes naturally.Saturated nacl aqueous solution (20mL) is added into reaction system, extracted with dichloromethane (20mL × 3), merge organic phase, anhydrous sodium sulfate drying, filtering, it is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1) compound 1 (0.08g, yield of white solid are obtained:3.3%).
MS m/z(ESI):535.1[M+1]。
1H NMR(400MHz,CDCl3)δ8.61(s,1H),8.03(td,2H),7.34(s,1H),7.27(d,1H),7.10–6.97(m,1H),6.58(ddd,1H),6.24(dt,1H),5.66(ddd,1H),5.30(s,0H),4.74(d,1H),4.10–3.81(m,7H),3.51(s,0H),3.15(s,1H),2.72(s,1H),2.25–1.61(m,6H),1.43–1.16(m,4H)。
19F NMR(376MHz,CDCl3):-117.92(1F),-123.77(1F)。
Embodiment 2:1- [4- [4- (3- chloro- 2,4- difluoro-anilines base) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone (compounds 2, compound 2 is 1- [(3R, 4R) -4- [4- (3- chloro- 2,4- difluoro-anilines) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone and 1- [(3S, 4S) -4- [4- (chloro- 2, the 4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone mixture)
1-[4-[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-1-piperidyl]prop-2-en-1-one
The first step:4- [4- (3- chloro- 2,4- difluoro-anilines base) -7- methoxy-quinazoline -6- bases] epoxide -3- fluoro-piperidine -1- t-butyl formates (2B, 2B is (3R, 4R) -4- [4- (3- chloro- 2,4- difluoro-anilines) -7- methoxy-quinazoline -6- bases] epoxide -3- fluoro-piperidine -1- t-butyl formates and (3S, 4S) the mixture of -4- [4- (chloro- 2, the 4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- bases] epoxide -3- fluoro-piperidine -1- t-butyl formates)
tert-butyl4-[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-pipe ridine-1-carboxylate
3c (3.0g are sequentially added into reaction bulb; 8.96mmol), dichloromethane (60mL), 2b-1 and 2b-3 mixture (3.9g; 17.91mmol) with triphenylphosphine (9.40g; 35.90mmol); nitrogen is protected, and is cooled to dichloromethane (36mL) solution that tert-butyl azodicarboxylate (6.20g, 26.90mmol) is added dropwise at -15 DEG C; after completion of dropping, reaction 3 hours is warmed to room temperature.It is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1) 2B (4.1g, yield of yellow solid are obtained:85.4%, 2B is (3R, 4R) -4- [4- (3- chloro- 2,4- difluoro-anilines) -7- methoxy-quinazoline -6- bases] epoxide -3- fluoro-piperidine -1- t-butyl formates and (3S, 4S) the mixture of -4- [4- (chloro- 2, the 4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- bases] epoxide -3- fluoro-piperidine -1- t-butyl formates).
Second step:N- (3- chloro- 2,4- Dichloro-phenyls) two (trifluoroacetic acid) salt (2C of -6- [(the fluoro- 4- piperidyls of 3-) oxygen] -7- methoxy-quinazoline -4- amine, 2C is N- (3- chloro- 2,4- Dichloro-phenyls) -6- [[(3R, 4R) the fluoro- 4- piperidyls of -3-] epoxide] -7- methoxy-quinazoline -4- amine two (trifluoroacetic acid) and N- (3- chloro- 2,4- Dichloro-phenyls) -6- [[the fluoro- 4- piperidyls of (3S, 4S) -3-] epoxide] -7- methoxy-quinazoline -4- amine two (trifluoroacetic acid) mixture)
N-(3-chloro-2,4-difluoro-phenyl)-6-[(3-fluoro-4-piperidyl)oxy]-7-methoxy-quinazolin-4-am ine
2C is N- (3- chloro- 2,4- Dichloro-phenyls) -6- [[(3R, 4R) the fluoro- 4- piperidyls of -3-] epoxide] -7- methoxy-quinazoline -4- amine two (trifluoroacetic acid) and N- (3- chloro- 2,4- Dichloro-phenyls) -6- [[the fluoro- 4- piperidyls of (3S, 4S) -3-] epoxide] -7- methoxy-quinazoline -4- amine two (trifluoroacetic acid) mixture.
2B (4.10g, 7.62mmol) and dichloromethane (17mL) are sequentially added into reaction bulb, ice-water bath cooling is lower to be added dropwise Naturally reaction 1 hour is warmed to room temperature after trifluoroacetic acid (17mL), completion of dropping, is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=20:1) 2C (3.00g, yield of white solid are obtained:60.0%).
MS m/z(ESI):439.0[M+1]。
3rd step:1- [4- [4- (3- chloro- 2,4- difluoro-anilines base) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone (compounds 2, compound 2 is 1- [(3R, 4R) -4- [4- (3- chloro- 2,4- difluoro-anilines) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone and 1- [(3S, 4S) -4- [4- (chloro- 2, the 4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone mixture)
1-[4-[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-1-piperidyl]prop-2-en-1-one
Compound 2 is 1- [(3R, 4R) -4- [4- (3- chloro- 2,4- difluoro-anilines) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone and 1- [(3S, 4S) -4- [4- (the chloro- 2,4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone mixture.
By 2C (2.30g, in the mixed solution for 5.25mmol) being dissolved in tetrahydrofuran (90mL) and water (14mL), add sodium acid carbonate (1.3g, 15.80mmol), ice bath, which is cooled at 0 DEG C, is added dropwise acryloyl chloride (0.50g, tetrahydrofuran (7mL) solution 5.78mmol), 0 DEG C is stirred 15 minutes.Saturation drinking water (20mL) is added into reaction system, is extracted with dichloromethane (20mL × 3), merges organic phase, anhydrous sodium sulfate drying, filtering, is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1) compound 2 (0.68g, yield of white solid are obtained:26.4%).
Compound 2 (0.68g) is separated into (separation condition with Preparation equipment and the chiral isomers of chiral column:Chiral column CHIRALPAK AS-H, mobile phase:A:N-hexane (0.1% diethanol amine)), B:Isopropanol, A:B=50:50 (v/v), flow velocity:6.0mL/ minutes, UV=250nm, column temperature:35 DEG C), obtain the 1- [(3R of white solid, 4R) -4- [4- (3- chloro- 2,4- difluoro-anilines base) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone (compound 2-1) (200mg, retention time:40.363min, ee > 95%) and 1- [(3S, 4S) -4- [4- (3- chloro- 2,4- difluoro-anilines base) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone (compound 2-2) (200mg, retention time:39.280min, ee > 95%).
MS m/z(ESI):493.3[M+1]。
1H NMR(400MHz,CDCl3):δ8.61(s,1H),7.99(dd,1H),7.66(s,1H),7.31(s,1H),7.27(s,0H),7.03(td,1H),6.58(dd,1H),6.25(d,1H),5.71(dd,1H),5.30(s,0H),4.95–4.89(m,1H),4.83–4.77(m,1H),4.67(d,1H),4.41–4.14(m,2H),4.00(s,3H),3.89(s,0H),3.75–3.40(m,2H),3.28(d,1H),2.32–2.06(m,2H),1.95(s,1H)。
19F NMR(400MHz,CDCl3):δ -117.02 (1F), -121.91 (1F), -199.56~-200.97 (1F).
Embodiment 3:1- [4- [4- (3- chloro- 2,4- difluoro-anilines) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone (compounds 3, compound 3 is 1- [(3R, 4S) -4- [4- (3- chloro- 2,4- difluoro-anilines) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone and 1- [(3S, 4R) -4- [4- (chloro- 2, the 4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone mixture)
1-[4-[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-1-piperidyl]prop-2-en-1-one
The first step:4- [4- (3- chloro- 2,4- difluoro-anilines) -7- methoxy-quinazoline -6- bases] epoxide -3- fluoro-piperidine -1- t-butyl formates (3B, 3B is (3R, 4S) -4- [4- (3- chloro- 2,4- difluoro-anilines) -7- methoxy-quinazoline -6- bases] epoxide -3- fluoro-piperidine -1- t-butyl formates and uncle (3S, 4R) the mixture of -4- [4- (chloro- 2, the 4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- bases] epoxide -3- fluoro-piperidine -1- butyl formates)
tert-butyl4-[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-pipe ridine-1-carboxylate
3c (2.0g are sequentially added into reaction bulb; 6.16mmol; intermediate 3), dichloromethane (40mL), 2b-2 and 2b-4 mixture (2.7g; 12.30mmol) with triphenylphosphine (6.50g, 24.60mmol), nitrogen protection; it is cooled at -15 DEG C and tert-butyl azodicarboxylate (4.30g is slowly added dropwise; after dichloromethane (24mL) solution 18.50mmol), completion of dropping, reaction 3 hours is warmed to room temperature naturally.It is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=50:1) 3B (1.40g, yield of yellow solid are obtained:43.8%, 3B is (3R, 4S) -4- [4- (3- chloro- 2,4- difluoro-anilines) -7- methoxy-quinazoline -6- bases] epoxide -3- fluoro-piperidine -1- t-butyl formates and (3S, 4R) the mixture of -4- [4- (chloro- 2, the 4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- bases] epoxide -3- fluoro-piperidine -1- t-butyl formates).
Second step:N- (the chloro- 2,4- Dichloro-phenyls of 3-) -6- [(the fluoro- 4- piperidyls of 3-) epoxide] -7- methoxy-quinazoline -4- amine two (three Fluoroacetic acid) salt (3C, 3C is N- (3- chloro- 2,4- Dichloro-phenyls) -6- [[(3R, 4S) the fluoro- 4- piperidyls of -3-] epoxide] -7- methoxy-quinazoline -4- amine two (trifluoroacetic acid) salt and N- (3- chloro- 2,4- Dichloro-phenyls) -6- [[the fluoro- 4- piperidyls of (3S, 4R) -3-] epoxide] -7- methoxy-quinazoline -4- amine two (trifluoroacetic acid) salt mixture)
N-(3-chloro-2,4-difluoro-phenyl)-6-[(3-fluoro-4-piperidyl)oxy]-7-methoxy-quinazolin-4-am ine
3B (1.40g, 2.60mmol) and dichloromethane (7mL) are added into reaction bulb, ice-water bath cooling is lower to be added dropwise trifluoroacetic acid (7mL), reacts at room temperature 1 hour.It is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=20:1) 3C (0.65g, yield of white solid are obtained:37.5%, 3C is N- (3- chloro- 2,4- Dichloro-phenyls) -6- [[(3R, 4S) the fluoro- 4- piperidyls of -3-] epoxide] -7- methoxy-quinazoline -4- amine two (trifluoroacetic acid) salt and N- (3- chloro- 2,4- Dichloro-phenyls) -6- [[the fluoro- 4- piperidyls of (3S, 4R) -3-] epoxide] -7- methoxy-quinazoline -4- amine two (trifluoroacetic acid) salt mixture).
3rd step:1- [4- [4- (3- chloro- 2,4- difluoro-anilines) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone (compounds 3, compound 3 is 1- [(3R, 4S) -4- [4- (3- chloro- 2,4- difluoro-anilines) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone and 1- [(3S, 4R) -4- [4- (chloro- 2, the 4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone mixture)
1-[4-[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-1-piperidyl]prop-2-en-1-one
By 3C (0.65g, in the mixed solution for 0.10mmol) being dissolved in tetrahydrofuran (25mL) and water (4mL), add sodium acid carbonate (0.41g, 4.80mmol), ice bath, which is cooled at 0 DEG C, is added dropwise acryloyl chloride (0.10g, tetrahydrofuran (1.5mL) solution 0.11mmol), 0 DEG C is reacted 15 minutes, saturation drinking water (20mL) is added into reaction system, extracted with dichloromethane (20mL × 3), merge organic phase, anhydrous sodium sulfate drying, filtering, it is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1) compound 3 (0.18g, yield of white solid are obtained:36.7%, HPLC:99.70%).
Compound 3 (0.18g) is separated into (separation condition with Preparation equipment and the chiral isomers of chiral column:Chiral column CHIRALPAK AD-H, mobile phase:A:N-hexane (0.1% diethanol amine), B:Isopropanol, A:B=50:50 (v/v), flow velocity:6.0mL/ minutes, UV=254nm, column temperature:35 DEG C), obtain compound 3-1 (60mg, retention time of white solid:9.158min, ee > 99%) and compound 3-2 (60mg, retention time:10.662min, ee > 99%).
MS m/z(ESI):493.3[M+1]。
1H NMR(400MHz,CDCl3)δ8.61(s,1H),7.99(dd,1H),7.66(s,1H),7.31(s,1H),7.27(s,0H),7.03(td,1H),6.58(dd,1H),6.25(d,1H),5.71(dd,1H),5.30(s,0H),4.95–4.89(m,1H),4.83–4.77(m,1H),4.67(d,1H),4.41–4.14(m,2H),4.00(s,3H),3.89(s,0H),3.75–3.40(m, 2H),3.28(d,1H),2.32–2.06(m,2H),1.95(s,1H)。
19F NMR(400MHz,CDCl3):δ -117.02 (1F), -121.91 (1F), -199.56~-200.97 (1F).
Embodiment 4:1- [4- [4- (the chloro- 2,4- difluoro-anilines of 3-)-7- [(3S)-tetrahydrofuran-3- bases] epoxide-quinazoline-6- bases] Oxy-1-piperidyl] propyl group-2- alkene-1- ketone (compound 4)
1-[4-[4-(3-chloro-2,4-difluoro-anilino)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazolin-6-yl]ox y-1-piperidyl]prop-2-en-1-one
The first step:The fluoro- 2- nitrobenzoic acids (4B) of 5- benzyloxies -4-
5-benzyloxy-4-fluoro-2-nitro-benzoic acid
4 are added into reaction bulb, the fluoro- 2- Nitro-benzoic acids 4A (25g of 5- bis-, 123mmol) with tetrahydrofuran (400mL), after stirring, benzylalcohol (29.3g, 270.6mmol) is added, ice bath is cooled to 0 DEG C, 0 DEG C is reacted 1 hour after the tetrahydrofuran solution (197mL, 394mmol) of double (trimethylsilyl) Sodamides of dropwise addition 2M, completion of dropping.2M hydrochloric acid regulation system pH value is added dropwise into reaction solution to 1-2, adds ethyl acetate (400mL), organic phase is separated after stirring, aqueous phase is extracted with ethyl acetate (100mL × 3), merges organic phase, anhydrous sodium sulfate drying, filtering, is concentrated under reduced pressure into residual acetic acid second Ester about (100mL), 0 DEG C stands 2 hours, depressurizes suction filtration, collects filter cake, and drying obtains 4B (17g, yield of yellow solid:47.5%).
MS m/z(ESI):292.0[M+1]。
1H NMR(400MHz,DMSO)δ7.70(d,1H),7.39-7.31(m,6H),5.28(s,2H)。
Second step:The fluoro- 2- nitrobenzene methyls (4C) of 5- benzyloxies -4-
methyl 5-benzyloxy-4-fluoro-2-nitro-benzoate
4B (17g are added into reaction bulb, 58.4mmol) and N, dinethylformamide (170mL), after stirring, ice bath is cooled down, and adds potassium carbonate (24.2g, 175.2mmol) and iodomethane (24.8g, 175.2mmol), add and be warmed to room temperature reaction 2 hours.Reaction solution is poured into frozen water, it is extracted with ethyl acetate (100mL × 4), merge organic phase, it is washed with water (100mL × 1), organic phase anhydrous sodium sulfate drying, filtering, is concentrated under reduced pressure, silica gel column chromatography separating-purifying (petrol ether/ethyl acetate (v/v)=10:1~8:1) 4C (15.9g, yield 89%) of yellow solid is obtained.
MS m/z(ESI):306.1[M+1]。
1H NMR(400MHz,CDCl3)δ7.79(d,1H),7.42-7.35(m,5H),7.25(d,1H),5.23(s,2H),3.91(s,3H)。
3rd step:The 2- amino fluoro- methyl benzoates of -5- benzyloxies -4- (4D)
methyl 2-amino-5-benzyloxy-4-fluoro-benzoate
By 4C (18.4g, in the mixed solution for 60.3mmol) being dissolved in isopropanol (294mL) and tetrahydrofuran (74mL), after stirring, add iron powder (18.1g, 324.1mmol), 100 DEG C are heated to, ammonium chloride (12g is added, the aqueous solution (15mL) 3.75mmol), 100 DEG C are reacted 7 hours.Filter while hot, filter cake is washed with ethyl acetate (100mL × 2), filtrate decompression is concentrated, ethyl acetate (400mL) is added to residual species, washed (50mL × 2), organic phase anhydrous sodium sulfate drying, filtered with saturated sodium bicarbonate, it is concentrated under reduced pressure, silica gel column chromatography separating-purifying (petrol ether/ethyl acetate (v/v)=8:1~7:1) 4D (15g, yield 90.4%) of gray solid shape is obtained.
MS m/z(ESI):276.1[M+1]。1H NMR(400MHz,CDCl3)δ7.53(d,1H),7.47–7.28(m,5H),6.42(d,1H),5.03(s,2H),3.85(s,3H)。
4th step:The fluoro- 3H- quinazoline-4-ones (4E) of 6- benzyloxies -7-
6-benzyloxy-7-fluoro-3H-quinazolin-4-one
4D (15g, 54.5mmol) and formamide (50mL) are added into reaction bulb, DMF (0.5mL) is added, 180 DEG C is heated to and reacts 7 hours.Reaction solution is cooled to room temperature, and reaction solution is poured slowly into water (250mL), stirs 30 minutes, and filtering collects filter cake, obtains the 4E (13g, yield 88%) of yellow solid.
MS m/z(ESI):271.1[M+1]。
1H NMR(400MHz,DMSO)δ12.33(brs,1H),8.07(s,1H),7.78(d,1H),7.56(d,1H),7.50(d,2H),7.40(dt,3H),7.24–7.05(m,1H),5.33(s,2H)。
5th step:6- benzyloxies -7- [(3S)-tetrahydrofuran -3- bases] epoxide -3H- quinazoline-4-ones (4F)
6-benzyloxy-7-[(3S)-tetrahydrofuran-3-yl]oxy-3H-quinazolin-4-one
(S) -3- hydroxyl tetrahydrofurans (20mL) and metallic sodium (2g are added into reaction bulb, 88.9mmol), 120 DEG C are warming up to react 1.5 hours, 4E (4g are added, 14.8mmol), 150 DEG C are warming up to react 2.5 hours.Reaction solution is cooled to room temperature, and reaction solution is poured into frozen water, with concentrated hydrochloric acid regulation system pH value to neutrality, adds ethyl acetate (10mL), filtering collects filter cake, obtains the 4F (4.5g, yield 90%) of gray solid shape.
MS m/z(ESI):339.1[M+1]。
1H NMR(400MHz,DMSO)δ7.98(s,1H),7.55(s,1H),7.47(d,2H),7.40(t,2H),7.34(d,1H),7.14(s,1H),5.24(s,2H),3.95(dd,1H),3.87(q,2H),3.77(td,1H),2.31(td,1H),2.06-2.01(m,1H)。
6th step:The chloro- 7- of 6- benzyloxies -4- [(3S)-tetrahydrofuran -3- bases] epoxide-quinazoline (4G)
6-benzyloxy-4-chloro-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazoline
4F (2.5g, 7.4mmol) and thionyl chloride (25mL) are added into reaction bulb, DMF (0.5mL), back flow reaction 2.5 hours is added.It is concentrated under reduced pressure into dry, add dichloromethane (50mL) and water (50mL), pH value is adjusted to 9-10 with ammoniacal liquor, separates organic phase, aqueous phase is extracted (30mL × 2) with dichloromethane, merge organic phase, washed, organic phase anhydrous sodium sulfate drying, filtered with water (30mL × 1), saturation drinking water (30mL × 1) successively, it is concentrated under reduced pressure, silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1) 4G (2g, yield 77%) of pale-yellow solid, is obtained.
MS m/z(ESI):357.0[M+1]。
1H NMR(400MHz,CDCl3)δ8.86(s,1H),7.50(d,3H),7.42(t,2H),7.36(t,1H),7.27(d,1H),5.29(s,2H),5.20–5.07(m,1H),4.14(d,2H),4.06(dd,1H),3.96(td,1H),2.45–2.22(m,2H)。
7th step:6- benzyloxies-N- (the chloro- 2,4- difluorophenyls of 3-) -7- [(3S)-tetrahydrofuran -3- bases] epoxide-quinazoline -4- ammonia (4H)
6-benzyloxy-N-(3-chloro-2,4-difluoro-phenyl)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazolin-4-amine
4G (3g, 8.4mmol) and chloro- 2, the 4- difluoroanilines (2.7g, 16.9mmol) of 3- are added into reaction bulb, isopropanol (60mL), heating reflux reaction 3 hours is added.Reaction solution is cooled to room temperature, filtering, collects filter cake, obtains the 4H (3.3g, yield 81%) of faint yellow solid shape.
MS m/z(ESI):484.1[M+1]。
1H NMR(400MHz,DMSO)δ11.74(s,1H),8.85(s,1H),8.54(s,1H),7.64(dd,1H),7.58–7.34(m,7H),5.36(s,2H),5.26(s,1H),3.97(d,2H),3.90(dd,1H),3.80(dd,1H),2.38(td,1H),2.19–1.94(m,1H)。
8th step:4- (the chloro- 2,4- difluoro-anilines of 3-) -7- [(3S)-tetrahydrofuran -3- bases] epoxide-quinazoline -6- alcohol (4I)
4-(3-chloro-2,4-difluoro-anilino)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazolin-6-ol
4H (1.1g, 2.28mmol), trifluoracetic acid (10mL) and thioanisole (1mL), heating reflux reaction 2.5 hours are sequentially added into reaction bulb.It is concentrated under reduced pressure into dry, ether (15mL) is added, with 7M methanolic ammonia solution regulation system pH value to 9, it is concentrated under reduced pressure, adds water (20mL), filtering, filter cake is collected, the 4I (740mg, yield 83%) of faint yellow solid shape is dried to obtain.
MS m/z(ESI):394.1[M+1]。
1H NMR(400MHz,DMSO)δ9.64(s,1H),9.56(s,1H),8.34(s,1H),7.67(s,1H),7.54(dd,1H),7.38(t,1H),7.16(s,1H),5.24(s,1H),4.04–3.86(m,3H),3.79(dd,1H),2.32(td,1H),2.18–2.05(m,1H)。
9th step:4- [4- (the chloro- 2,4- difluoro-anilines of 3-) -7- [(3S)-tetrahydrofuran -3- bases] oxygen-quinazoline -6- bases] epoxide piperidines -1- t-butyl formates (4J)
tert-butyl-4-[4-(3-chloro-2,4-difluoro-anilino)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazolin-6-yl]oxypiperidine-1-carboxylate
4I (600mg are sequentially added into reaction bulb, 1.53mmol), N-Boc-4- hydroxy piperidines alcohol (615mg, 3.06mmol) with triphenylphosphine (1.6g, 6.12mmol), dichloromethane (12mL) is added, -15 DEG C are cooled to, tert-butyl azodicarboxylate (615mg is added dropwise, dichloromethane solution (6mL) 3.06mmol), drips off and is warmed to room temperature reaction 3 hours.It is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=50:1→30:1) white solid 4J (750mg, yield 85%) is obtained.
MS m/z(ESI):577.0[M+1]。
1H NMR(400MHz,DMSO)δ9.62(s,1H),8.37(s,1H),7.93(s,1H),7.57(td,1H),7.40(t,1H),7.22(s,1H),5.26(s,1H),4.64(d,1H),4.00–3.93(m,1H),3.92–3.74(m,3H),3.61(d,2H),3.30(s,1H),2.32(td,1H),2.12–1.85(m,4H),1.69(d,2H),1.42(s,9H)。
Tenth step:N- (the chloro- 2,4- difluorophenyls of 3-) -6- (4- piperidyl epoxides -7- [(3S)-tetrahydrofuran -3- bases] epoxide quinazoline -4- amine (4K)
N-(3-chloro-2,4-difluoro-phenyl)-6-(4-piperidyloxy)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quin azolin-4-amine
Sequentially add and reacted at room temperature 3 hours after 4J (750mg, 1.3mmol), trifluoracetic acid (3mL) and dichloromethane (10mL), completion of dropping into reaction bulb.It is concentrated under reduced pressure, ether (15mL) is added into residue, with 7M methanolic ammonia solution regulation system pH value to 9, is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=10:1→5:1), saturated sodium bicarbonate solution (20mL) and chloroform (30mL) are added into residue, separate organic layer, water layer is extracted (10mL × 3) with chloroform, merges organic phase, anhydrous sodium sulfate drying, filtering, filtrate decompression is concentrated, the 4K (575mg, yield 93%) of faint yellow solid shape is obtained.
MS m/z(ESI):477.0[M+1]。
1H NMR(400MHz,DMSO)δ9.62(s,1H),8.36(d,1H),7.88(s,1H),7.56(dd,1H),7.40(td,1H),7.20(s,1H),5.25(s,1H),4.65–4.42(m,1H),3.98(dd,1H),3.93–3.85(m,2H),3.80(td,1H),3.06–2.95(m,2H),2.61(dd,2H),2.32(td,1H),2.13–1.89(m,3H),1.58(d,2H),1.23(s,1H)。
11st step:1- [4- [4- (the chloro- 2,4- difluoro-anilines of 3-)-7- [(3S)-tetrahydrofuran-3- bases] epoxide quinazoline-6- bases] Oxy-1-piperidyl] propyl group-2- alkene-1- ketone (compound 4)
1-[4-[4-(3-chloro-2,4-difluoro-anilino)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazolin-6-yl]ox y-1-piperidyl]prop-2-en-1-one
4K (310mg are sequentially added into reaction bulb, 0.65mmol), tetrahydrofuran (12mL) and sodium acid carbonate (164mg, the aqueous solution (1.8mL) 1.95mmol), ice bath is cooled to 0 DEG C, acryloyl chloride (65mg is added dropwise, 0.72mmol) tetrahydrofuran solution (1mL), 0 DEG C is added to react 15 minutes, saturated sodium bicarbonate aqueous solution (20mL) is added dropwise, add chloroform (50mL), separate organic layer, water layer is extracted (20mL × 3) with chloroform, merge organic phase, anhydrous sodium sulfate drying, filtering, filtrate decompression is concentrated, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1→30:1) compound 4 (207mg, yield of white solid are obtained:60%).
MS m/z(ESI):531.0[M+1]。
1H NMR(400MHz,CDCl3)δ8.61(s,1H),8.08(dd,1H),7.51(s,1H),7.25(s,1H),7.04(td,1H),6.60(dd,1H),6.27(d,1H),5.70(d,1H),5.11(s,1H),4.68(s,1H),4.08(d,2H),4.05–3.89(m,2H),3.85-3.76(m,3H),3.59(s,1H),2.36(td,1H),2.28–2.18(m,1H),2.03-1.95(m,3H),1.37-1.22(m,3H)。
Embodiment 5:1- [(3aR, 6aS) -5- [4- (3- chloro- 2,4- difluoro-anilines) -7- [(3S)-tetrahydrofuran -3- bases] epoxide-quinazoline -6- bases] epoxide -3,3a, 4,5,6,6a- hexahydro -1H- pentamethylene simultaneously [c] pyrroles -2- bases] propyl group -2- alkene -1- ketone (compound 5)
1-[(3aR,6aS)-5-[4-(3-chloro-2,4-difluoro-anilino)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazo lin-6-yl]oxy-3,3a,4,5,6,6a-hexahydro-1H-cyclopenta[c]pyrrol-2-yl]prop-2-en-1-one
The first step:(3aR, 6aS) -5- [4- (the chloro- 2,4- difluoro-anilines of 3-) -7- [(3S)-tetrahydrofuran -3- bases] epoxide-quinazoline -6- bases] epoxide -3,3a, 4,5,6,6a- hexahydro -1H- pentamethylene [c] pyrroles -2- t-butyl formates (5B)
tert-butyl-(3aR,6aS)-5-[4-(3-chloro-2,4-difluoro-anilino)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazolin-6-yl]oxy-3,3a,4,5,6,6a-hexahydro-1H-cyclopenta[c]pyrrole-2-carboxylate
4I (300mg are sequentially added into reaction bulb, 0.75mmol), 4d (339mg, 1.50mmol, intermediate 4) and triphenylphosphine (786mg, 3.00mmol), add dichloromethane (6mL), reaction 2 hours is warmed to room temperature after being cooled to the dichloromethane solution (3mL) that tert-butyl azodicarboxylate (518mg, 2.25mmol) is added dropwise at -15 DEG C, completion of dropping.It is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1→50:1) 5B (353mg, yield of white solid are obtained:78%).
MS m/z(ESI):603.0[M+1]。
1H NMR(400MHz,DMSO)δ9.62(s,1H),8.36(s,1H),7.76(s,1H),7.57(dd,1H),7.40(t,1H),7.19(s,1H),5.22(s,1H),5.08(s,1H),3.96(dd,1H),3.88(d,2H),3.83–3.72(m,1H),3.48(td,2H),3.13(d,2H),2.84(s,3H),2.32(dt,1H),2.12-2.01(m,4H),1.97-1.90(m,2H),1.39(s,9H)。
Second step:6- [[(3aR, 6aS) -1,2,3,3a, 4,5,6,6a- octahydros pentamethylene simultaneously [c] pyrroles -5- bases] epoxide]-N- (the chloro- 2,4- difluorophenyls of 3-) -7- [(3S)-tetrahydrofuran -3- bases] epoxide-quinazoline -4- ammonia (5C)
6-[[(3aR,6aS)-1,2,3,3a,4,5,6,6a-octahydrocyclopenta[c]pyrrol-5-yl]oxy]-N-(3-chloro-2,4-dif luoro-phenyl)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazolin-4-amine
5B (470mg, 0.78mmol), trifluoracetic acid (2mL) and dichloromethane (6mL) are sequentially added into reaction bulb, room temperature reaction 30 minutes is added.It is concentrated under reduced pressure into dry, adds ether (15mL), with 7M methanolic ammonia solution regulation system pH value extremely 9, it is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=10:1~7:1), saturated sodium bicarbonate solution (20mL) and chloroform (30mL) are added into residue, separate organic layer, water layer is extracted (10mL × 3) with chloroform, merges organic phase, anhydrous sodium sulfate drying, filtering, it is concentrated under reduced pressure, obtains 5C (340mg, yield of yellow solid:87%).
MS m/z(ESI):503.0[M+1]。
1H NMR(400MHz,DMSO)δ8.36(s,1H),7.91(d,1H),7.56(d,1H),7.41-7.36(m,1H),7.20(s,1H),5.23(s,1H),5.10(d,1H),3.97(dd,1H),3.88(d,2H),3.83–3.73(m,1H),3.24(dd,1H),3.13–2.99(m,2H),2.88(t,3H),2.32(td,1H),2.21–1.96(m,3H),1.87(d,2H)。
3rd step:1- [(3aR, 6aS) -5- [4- (3- chloro- 2,4- difluoro-anilines) -7- [(3S)-tetrahydrofuran -3- bases] epoxide-quinazoline -6- bases] epoxide -3,3a, 4,5,6,6a- hexahydro -1H- pentamethylene simultaneously [c] pyrroles -2- bases] propyl group -2- alkene -1- ketone (compound 5)
1-[(3aR,6aS)-5-[4-(3-chloro-2,4-difluoro-anilino)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazo lin-6-yl]oxy-3,3a,4,5,6,6a-hexahydro-1H-cyclopenta[c]pyrrol-2-yl]prop-2-en-1-one
5C (340mg are sequentially added into reaction bulb, 0.68mmol), tetrahydrofuran (12mL) and sodium acid carbonate (171mg, water (1.8mL) solution 2.04mmol), it is cooled to 0 DEG C, acryloyl chloride (68mg is added dropwise, tetrahydrofuran (1mL) solution 0.75mmol), 0 DEG C is reacted 30 minutes, saturated sodium bicarbonate solution (10mL) is added dropwise into reaction system, add dichloromethane (30mL), separate organic layer, water layer is extracted (10mL × 2) with dichloromethane, merge organic phase, saturation drinking water washs (20mL × 1), anhydrous sodium sulfate drying, filtering, it is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1~30:1) compound 5 (100mg, yield of yellow solid are obtained:27%, HPLC:97%).
MS m/z(ESI):557.0[M+1]。
1H NMR(400MHz,CDCl3)δ8.60(s,1H),7.41(s,1H),7.19(s,1H),7.03(td,1H),6.41(dd,1H),6.32(dd,1H),5.66(dd,1H),5.14–5.01(m,2H),4.08(d,2H),4.03(dd,1H),3.93(td,1H),3.74(ddd,2H),3.52–3.36(m,3H),3.14–3.00(m,1H),3.00–2.85(m,1H),2.33-2.19(m,4H),1.91(ddd,3H)。
Embodiment 6:1- [(3aR, 6aS) -5- [4- [(the chloro- 2,4- difluorophenyls of 3-) amino] -7- methoxy-quinazoline -6- bases] epoxide-hexahydro pentamethylene simultaneously [c] pyrroles -2 (1H)-yl] propyl group -2- alkene -1- ketone (compound 6)
1-((3aR,5s,6aS)-5-((4-((3-chloro-2,4-difluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)hexahydrocyclopenta[c]pyrrol-2(1H)-yl)prop-2-en-1-one
The first step:(3aR, 6aS) -5- ((the chloro- 7- methoxy-quinazolines -6- bases of 4-) epoxide-hexahydro pentamethylene simultaneously [c] pyrroles -2 (1H) -- t-butyl formates (6B)
(3aR,5s,6aS)-tert-butyl 5-((4-chloro-7-methoxyquinazolin-6-yl)oxy)hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate
1d (1.1g are sequentially added into reaction bulb, 5.2mmol), 4d (1.7g, 7.8mmol, intermediate 4) and triphenylphosphine (2.1g, 7.8mmol), after stirring, ice-water bath is cooled to 0 DEG C, diisopropyl azodiformate (1.6g, 7.8mmol) dichloromethane (5mL) solution is added dropwise in system, is warmed to room temperature and is stirred overnight naturally.It is concentrated under reduced pressure, residue separates (petrol ether/ethyl acetate (v/v)=3 with silica gel column chromatography:2→2:3) 6B (2.2g) of colorless oil is obtained.
MS m/z(ESI):420.3[M+1]。
Second step:(3aR, 6aS) -5- [[4- [(the chloro- 2,4- difluorophenyls of 3-) amino] -7- methoxy-quinazoline -6- bases] epoxide]-hexahydro pentamethylene [c] pyrroles -2 (1H)-t-butyl formate (6C)
(3aR,5s,6aS)-tert-butyl 5-((4-((3-chloro-2,4-difluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate
6B (4g are sequentially added into reaction bulb, 9.5mmol), 3- chloro- 2,4- difluoroanilines (3g, 19mmol, finish) and isopropanol (65mL), rise to 100 DEG C to react 3 hours, be concentrated under reduced pressure, residue separates (methylene chloride/methanol (v/v)=30 with silica gel column chromatography:1~10:1) 6C (1.7g, yield of pale yellowish oil are obtained:32.7%).
MS m/z(ESI):547.2[M+1]。
1H NMR(300MHz,CDCl3)δ8.30(s,2H),7.40(m,2H),6.83(m,1H),5.38(m,1H),3.98(s,3H),3.52(m,2H),3.25(m,2H),2.90(m,2H),2.22(m,2H),2.05(m,2H),1.43(s,9H)。
3rd step:N- (the chloro- 2,4- difluorophenyls of 3-) -7- methoxyl groups -6- [[(3aR, 6aS)-octahydro pentamethylene simultaneously [c] pyrroles -5- bases] epoxide] bis- (trifluoroacetic acid) salt (6D) of quinazoline -4- amine
N-(3-chloro-2,4-difluorophenyl)-7-methoxy-6-(((3aR,5s,6aS)-octahydrocyclopenta[c]pyrrol-5-yl)oxy)quinazolin-4-amine
6C (0.59g, 1.1mmol), dichloromethane (4.5mL) and trifluoroacetic acid (2.2mL) are sequentially added under ice-water bath into reaction bulb, is stirred at room temperature 2 hours.It is concentrated under reduced pressure, residue separates (methylene chloride/methanol (v/v)=20 with silica gel column chromatography:1~5:1) 6D (0.6g, yield of pale-yellow solid are obtained:82%).
4th step:1- [(3aR, 6aS) -5- [[4- [(the chloro- 2,4- difluorophenyls of 3-) amino] -7- methoxy-quinazoline -6- bases] epoxide-hexahydro pentamethylene simultaneously [c] pyrroles -2 (1H)-yl] propyl- 2- alkene -1- ketone (compound 6)
1-((3aR,5s,6aS)-5-((4-((3-chloro-2,4-difluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)hexahydrocyclopenta[c]pyrrol-2(1H)-yl)prop-2-en-1-one
6D (the 0.257g successively into reaction bulb, 0.38mmol), triethylamine (0.116g, 1.14mmol) with dichloromethane (12mL), dry ice ethanol bath, which is cooled at -40 DEG C, is added dropwise acryloyl chloride (0.041g, 0.46mmol), -40 DEG C are stirred 50 minutes.Reaction solution is poured into saturated sodium bicarbonate solution (60mL), extracted with chloroform (60mL × 3), merges organic phase, anhydrous sodium sulfate drying, filtering, is concentrated under reduced pressure, residue separates (methylene chloride/methanol (v/v)=60 with silica gel column chromatography:1~30:1) compound 6 (0.06g, yield of colorless oil are obtained:36.6%, HPLC:98.46%).
MS m/z(ESI):501.0[M+1]。
1H NMR(300MHz,CDCl3)δ8.59(s,1H),7.90(m,1H),7.44(s,1H),7.27(d,1H),7.03(m,1H),6.30(m,2H),5.66(m,1H),5.10(m,1H),3.99(s,3H),3.71(m,2H),3.42(m,2H),3.01(m,2H),2.28(m,3H),1.96(m,3H)。
Embodiment 7:1- [(3R, 4R) -4- [4- (the chloro- 2,4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone maleates (compound 7)
1-[(3R,4R)-4-[4-(3-chloro-2,4-difluorophenyl)amino)-7-methoxyquinazolin-6-yl]oxy-3-fluo ropiperidin-1-yl]prop-2-en-1-one maleic acid salt
Compound 2-1 (200mg, 0.41mmol), methanol (2.0mL) and maleic acid (47.1mg, 0.41mmol) are added into reaction bulb, is stirred at room temperature 5 hours.It is filtrated to get compound 7 (90mg, yield of pale-yellow solid:36%).
1H NMR(400MHz,DMSO)δ8.53(s,1H),8.31(s,1H),8.05(s,1H),7.60(td,1H),7.43 (td,1H),7.28(s,1H),6.85(dd,1H),6.21(s,2H),6.15(dd,1H),5.73(dd,1H),4.96–4.74(m,2H),4.03–3.91(m,5H),3.75(s,1H),3.62(s,1H),2.23–2.00(m,1H),1.85–1.73(m,1H)。
19F NMR(400MHz,DMSO)δ-114.82,-188.00,-188.82。
Embodiment 8:1- [(3R, 4S) -4- [4- (the chloro- 2,4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone maleates (compound 8)
1-[(3R,4S)-4-[4-(3-chloro-2,4-difluorophenyl)amino)-7-methoxyquinazolin-6-yl]oxy-3-fluo ropiperidin-1-yl]prop-2-en-1-one maleic acid salt
Compound 3-2 (170mg, 0.35mmol), ethanol (2.0mL) and maleic acid (40.1mg, 0.35mmol) are added into reaction bulb, is stirred at room temperature 5 hours.It is filtrated to get compound 8 (80mg, yield of pale-yellow solid:37.7%).
1H NMR(400MHz,DMSO)δ8.51(s,1H),7.96(s,1H),7.59(td,1H),7.43(td,1H),7.27(s,1H),6.85(d,1H),6.21(s,2H),6.14(dd,1H),5.72(dd,1H),5.09(d,1H),4.88–4.70(m,1H),4.34(s,2H),3.98(s,4H),3.43(dt,3H),3.05(d,1H),2.05(dd,2H)。
19F NMR(400MHz,DMSO)δ-114.62,-114.79,-201.63。
Embodiment 9:1- [(3R, 4R) -4- [4- (the chloro- 2,4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- keto hydrochlorides (compound 9)
1-[(3R,4R)-4-[4-(3-chloro-2,4-difluorophenyl)amino)-7-methoxyquinazolin-6-yl]oxy-3-fluo ropiperidin-1-yl]prop-2-en-1-one hydrochloric acid salt
Methanol (0.5mL) solution of compound 2-1 (400mg, 0.81mmol), methanol (2.8mL) and hydrochloric acid (0.1mL, 1.23mmol) is added into reaction bulb, is stirred at room temperature 4 hours.It is concentrated under reduced pressure to give compound 9 (395mg, yield of white solid:93.4%).
1H NMR(400MHz,DMSO)δ12.16(s,1H),8.91–8.68(m,2H),7.67–7.56(m,1H),7.49(t,1H),7.42(s,1H),6.86(dd,1H),6.14(dd,1H),5.72(dd,1H),5.16(s,1H),4.96–4.64(m,1H),4.03(s,3H),3.82(s,1H),3.63(s,2H),3.47(s,2H),2.24(d,1H),1.68(d,1H)。
19F NMR (400MHz, DMSO) δ -112.89 (1F), -113.91 (1F), -187.52~-188.50 (1F).
Embodiment 10:1- [(3R, 4S) -4- [4- (the chloro- 2,4- difluoro-anilines of 3-) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- keto hydrochlorides (compound 10)
1-[(3R,4S)-4-[4-(3-chloro-2,4-difluorophenyl)amino)-7-methoxyquinazolin-6-yl]oxy-3-fluo ropiperidin-1-yl]prop-2-en-1-one hydrochloric acid salt
Methanol (0.3mL) solution of compound 3-2 (150mg, 0.30mmol), methanol (0.8mL) and hydrochloric acid (16mg, 0.45mmol) is added into reaction bulb, is stirred at room temperature 5 hours.It is concentrated under reduced pressure to give compound 10 (135mg, yield of white solid:84.3%).
1H NMR(400MHz,DMSO)δ10.58(s,1H),8.55(s,1H),8.25(s,1H),7.67–7.19(m,3H),6.97–6.69(m,1H),6.17–6.01(m,1H),5.71(d,1H),5.12(d,1H),5.00–4.86(m,1H),4.38(t,1H),3.99(d,3H),3.69–3.49(m,1H),3.36(dd,3H),3.05(dd,1H),2.05(dt,1H),2.00–1.76(m,1H)。
19F NMR (376MHz, DMSO) δ -114.51 (1F), -114.61 (1F), -201.01~-202.20 (1F).
Embodiment 11:1- [(3R, 4R) -4- [4- (3, the 4- bis- chloro- fluoro- aniline of 2-) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone (compound 11)
1-[(3R,4R)-4-[4-(3,4-dichloro-2-fluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-1- piperidyl]prop-2-en-1-one
The first step:(3R, 4R) 4- [4- (3, the 4- bis- chloro- fluoro- aniline of 2-) -7- methoxy-quinazoline -6- bases] epoxide -3- fluoro-piperidine -1- t-butyl formates (11B)
(3R,4R)-tert-butyl4-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)-3-fluoropiperidine-1-carboxylate
6c (2.6g are sequentially added into reaction bulb; 7.3mmol; intermediate 6), dichloromethane (18mL), 5c-1 (0.53g; 2.43mmol) with triphenylphosphine (1.91g, 7.3mmol), nitrogen protection; it is cooled at -15 DEG C and tert-butyl azodicarboxylate (1.27g is added dropwise; after dichloromethane (18mL) solution 7.3mmol), completion of dropping, reaction 18 hours is warmed to room temperature.It is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1) 11B (0.2g, yield of yellow solid are obtained:15%).
Second step:(trifluoroacetic acid) salt (11C) of N- (3,4- bis- chloro- 2- fluoro-phenyls) -6- [[the fluoro- 4- piperidyls of (3R, 4R) -3-] epoxide] -7- methoxy-quinazoline -4- amine two
N-(3,4-dichloro-2-fluorophenyl)-6-(((3R,4R)-3-fluoropiperidin-4-yl)oxy)-7-methoxyquinaz olin-4-amine(ditrifluoroacetic acid)
11B (0.2g are sequentially added into reaction bulb, 0.35mmol) with dichloromethane (3mL), ice-water bath cooling is lower to be added dropwise trifluoroacetic acid (3mL), naturally reaction 1 hour is warmed to room temperature after completion of dropping, it is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=20:1) 11C (0.16g, yield of white solid are obtained:99%).
3rd step:1- [(3R, 4R) -4- [4- (3, the 4- bis- chloro- fluoro- aniline of 2-) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone (compound 11)
1-((3R,4R)-4-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)-3-fl uoropiperidin-1-yl)prop-2-en-1-one
By 11C (0.7g, in the mixed solution for 1.04mmol) being dissolved in tetrahydrofuran (25mL) and water (5mL), add sodium acid carbonate (0.44g, 5.22mmol), ice bath, which is cooled at -30 DEG C, is added dropwise acryloyl chloride (0.11g, 1.14mmol), -15 DEG C are stirred 30 minutes.Saturation drinking water (20mL) is added into reaction system, extracted with dichloromethane (35mL × 3), merge organic phase, anhydrous sodium sulfate drying, filtering, filtrate decompression is concentrated, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1) compound 11 (0.12g, yield of white solid are obtained:23%, HPLC:94.80%).
MS m/z(ESI):509.0[M+1]。
1H NMR(400MHz,CDCl3):δ1H NMR(400MHz,CDCl3)δ8.69(s,1H),8.35(t,1H),7.43(s,1H),7.32(dd,2H),6.59(dd,1H),6.30(d,1H),5.74(dd,1H),4.74(dd,2H),4.18–3.45(m,7H),2.19(s,1H),1.95(m,1H)。
19F NMR(400MHz,CDCl3):δ -123.08 (1F), -188.63~-189.44 (1F).
Embodiment 12:1- [(3R, 4S) -4- [4- (3, the 4- bis- chloro- fluoro- aniline of 2-) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone (compound 12)
1-((3R,4S)-4-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)-3-fl uoropiperidin-1-yl)prop-2-en-1-one
The first step:(3R, 4S) -4- [4- (3, the 4- bis- chloro- fluoro- aniline of 2-) -7- methoxy-quinazoline -6- bases] epoxide -3- fluoro-piperidine -1- t-butyl formates (12B)
(3R,4S)-tert-butyl4-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)-3-fluoropiperidine-1-carboxylate
6c (7.8g are sequentially added into reaction bulb; 21.9mmol; intermediate 6), dichloromethane (60mL), 5c-2 (1.6g; 7.3mmol) with triphenylphosphine (5.7g, 21.9mmol), nitrogen protection; it is cooled at -15 DEG C and tert-butyl azodicarboxylate (3.8g is added dropwise; 21.9mmol), after completion of dropping, it is warmed to room temperature reaction 18 hours.It is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1) the 12B crude products (4.0g) of yellow solid are obtained, the next step is directly used in.
Second step:(trifluoroacetic acid) salt (12C) of N- (3,4- bis- chloro- 2- fluoro-phenyls) -6- [[the fluoro- 4- piperidyls of (3R, 4S) -3-] epoxide] -7- methoxy-quinazoline -4- amine two
N-(3,4-dichloro-2-fluorophenyl)-6-(((3R,4S)-3-fluoropiperidin-4-yl)oxy)-7-methoxyquinaz olin-4-amine(ditrifluoroacetic acid)
12B (4.0g are sequentially added into reaction bulb, 7.3mmol) with dichloromethane (10mL), ice-water bath cooling is lower to be added dropwise trifluoroacetic acid (10mL), naturally reaction 1 hour is warmed to room temperature after completion of dropping, it is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=20:1) obtaining the 12C of white solid, (27%) 1.3g, the first step and second step total recovery is.
3rd step:1- [(3R, 4S) -4- [4- (3, the 4- bis- chloro- fluoro- aniline of 2-) -7- methoxy-quinazoline -6- bases] the fluoro- 1- piperidyls of epoxide -3-] propyl group -2- alkene -1- ketone (compound 12)
1-((3R,4S)-4-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)-3-fluoropiperidin-1-yl)prop-2-en-1-one
By 12C (1.2g, in the mixed solution for 1.8mmol) being dissolved in tetrahydrofuran (40mL) and water (8mL), add sodium acid carbonate (0.8g, 9.0mmol), ice bath, which is cooled at -30 DEG C, is added dropwise acryloyl chloride (0.2g, 2.2mmol), -15 DEG C are stirred 30 minutes.Saturation drinking water (50mL) is added into reaction system, is extracted with dichloromethane (50mL × 3), merges organic phase, anhydrous sodium sulfate drying, filtering, is concentrated under reduced pressure, residue silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1) compound 12 (0.21g, yield of white solid are obtained:22.8%).
MS m/z(ESI):509.0[M+1]。
1H NMR(400MHz,CDCl3):δ1H NMR(400MHz,CDCl3)δ8.64(s,1H),8.16(t,1H),7.63(s,1H),7.38–7.18(m,2H),6.58(dd,1H),6.26(dd,1H),5.72(dd,1H),5.00–4.57(m,2H),4.40–4.06(m,2H),4.00(s,3H),3.95–3.21(m,2H),2.27–2.10(m,1H),1.96(s,1H)。
19F NMR(400MHz,CDCl3):δ -121.02 (1F), -197.39~-198.96 (1F).
Embodiment 13:1- ((3R, 4R) -4- ((4- ((the chloro- 2- fluorine of 3,4- bis-) amino) -7- methoxy quinazoline -6- bases) epoxide) -3- fluorine resources -1- bases) propyl group -2- alkene -1- keto hydrochlorides (compound 13)
1-((3R,4R)-4-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)-3-fluoropiperidin-1-yl)prop-2-en-1-one Hydrochloric acid salt
(the 80mg of compound 11 is added into reaction bulb, 0.16mmol), methanol (1mL) and (0.1mL, 0.23mmol) the methanol solution (2.3M) of hydrochloric acid, it is stirred at room temperature 4 hours, it is concentrated under reduced pressure to give compound 13 (73mg, yield of white solid:90.4%).
1H NMR(400MHz,DMSO-d6)δ12.19(d,1H),8.96–8.68(m,2H),7.78–7.53(m,2H),7.41(s,1H),6.86(dd,1H),6.14(dd,1H),5.72(dd,1H),5.15(s,1H),4.96–4.57(m,1H),4.36–3.93(m,5H),3.82(d,2H),2.34–2.10(m,1H),1.68(d,1H)。
19F NMR(400MHz,DMSO-d6):δ -111.12 (1F), -187.67~-188.44 (1F).
Embodiment 14:1- ((3R, 4S) -4- ((4- ((the chloro- 2- fluorine of 3,4- bis-) amino) -7- methoxy quinazoline -6- bases) epoxide) -3- fluorine resources -1- bases) propyl group -2- alkene -1- keto hydrochlorides (compound 14)
1-((3R,4S)-4-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)-3-fluoropiperidin-1-yl)prop-2-en-1-one Hydrochloric acid salt
Compound 12 (78mg, 0.15mmol), methanol (1mL) and (0.1ml, 0.23mmol) hydrochloric acid are added into reaction bulb Methanol solution (2.3M), is stirred at room temperature 5 hours.It is concentrated to give compound 14 (73mg, yield of white solid:85%).
1H NMR(400MHz,DMSO)δ12.43(s,1H),8.86(s,1H),7.84–7.49(m,2H),7.39(s,1H),6.80(dd,2H),6.22–6.00(m,2H),5.32–5.02(m,1H),4.72–3.92(m,8H),2.07(s,1H),1.42(ddd,,1H)。
19F NMR(400MHz,DMSO):δ -110.94 (1F), -186.83~-187.43 (1F).
Embodiment 15
7- ((4- ((the chloro- 2- fluorophenyls of 3,4- bis-) amino) -7- methoxyquinazoline hydrochloride -6- bases) epoxide) tetrahydrochysene -1H- oxazoles simultaneously [3,4-a] pyridine -3 (5H) -one (compound 15)
7-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)tetrahydro-1H-oxazolo[3,4-a]pyridin-3(5H)-one
The first step:1- tert-butyl group 2- methyl 4- hydroxy piperidine -1,2- dicarboxylic acid esters (15B)
1-tert-butyl 2-methyl 4-hydroxypiperidine-1,2-dicarboxylate
1- tert-butyl group 2- methyl 4- oxo-piperidine -1,2- dicarboxylic acid esters 15A (6.1g, 24mmol) are added in reaction bulb, methanol 60mL is added, ice bath cooling is added portionwise sodium borohydride (1.4g, 36mmol), added, insulation reaction 30 minutes.Saturated aqueous ammonium chloride 50mL is added dropwise, it is concentrated under reduced pressure, add ethyl acetate and each 100mL of water, divide liquid, aqueous phase extracts 50mL × 2 with ethyl acetate, merges organic phase, anhydrous sodium sulfate drying, it is concentrated under reduced pressure, silica gel column chromatography separating-purifying (petrol ether/ethyl acetate (v/v)=3:1~2:1) colorless oil 15B (5.8g, yield 94%) is obtained.
MS m/z(ESI):160.1[M+1]。
1H NMR(400MHz,CDCl3)δ4.93(d,1H),4.21–3.90(m,1H),3.73(s,3H),3.66(t,1H),2.99(d,1H),2.44(t,1H),1.91(dd,1H),1.60(t,1H),1.46(s,9H)。
Second step:1- tert-butyl group 2- methyl 4- ((4- ((the chloro- 2- fluorophenyls of 3,4- bis-) amino) -7- methoxyquinazoline hydrochloride -6- bases) oxo) piperidines -1,2- dicarboxylic acid esters (15C)
1-tert-butyl 2-methyl 4-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)piperidine-1,2-dicarboxylate
By 15B (2.0g, 7.71mmol), 4- ((3, the chloro- 2- fluorophenyls of 4- bis-) amino) -7- methoxyquinazoline hydrochloride -6- alcohol (1.37g, 3.86mmol), triphenylphosphine (4.05g, 15.42mmol), add in reaction bulb, add dichloromethane (60mL), be cooled to -20 DEG C, azo-2-carboxylic acid's di tert butyl carbonate (2.66g is added dropwise, dichloromethane solution 10mL 11.57mmol), is dripped off, and is warmed to room temperature reaction 4 hours.It is concentrated under reduced pressure, silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1~50:1) faint yellow solid 15C (1.5g, yield 65%) is obtained.
MS m/z(ESI):595.0[M+1]。
1H NMR(400MHz,DMSO)δ9.77(s,1H),8.41(s,1H),7.82(s,1H),7.69–7.54(m,2H),7.20(s,1H),4.92(s,1H),4.67(d,1H),3.93(s,3H),3.81(s,1H),3.52(d,3H),2.68(t,1H),2.02(dd,2H),1.81(t,1H),1.41(d,9H),1.24(s,1H)。
3rd step:Tert-butyl group 4- ((4- ((the chloro- 2- fluorophenyls of 3,4- bis-) amino) -7- methoxyquinazoline hydrochloride -6- bases) oxo) -2- (methylol) piperidines -1- formic acid esters (15D)
tert-butyl 4-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)-2-(hydroxymethyl)piperidine-1-carboxylate
15C (0.5g, 0.8mmol) is added in reaction bulb, tetrahydrofuran (4mL) is added, -78 DEG C are cooled to, the toluene solution (4mL, 1.5mol/L) of diisobutyl aluminium hydride is added dropwise, drips off and is slowly increased to room temperature reaction 4 hours.Reaction solution is poured into sodium hydrate aqueous solution (100mL, 1mol/L) and in the mixture of ice, add ethyl acetate (30mL), divide liquid, aqueous phase is extracted (20mL × 2) with ethyl acetate, merge organic phase, anhydrous sodium sulfate drying, filtering, is concentrated under reduced pressure, silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1~20:1) yellow oil 15D (0.3g, yield 60%) is obtained.
MS m/z(ESI):567.1[M+1]。
4th step:7- ((4- ((the chloro- 2- fluorophenyls of 3,4- bis-) amino) -7- methoxyquinazoline hydrochloride -6- bases) epoxide) tetrahydrochysene -1H- oxazoles [3,4-a] pyridine -3 (5H) -one (compound 15)
7-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)tetrahydro-1H-oxazolo[3,4-a]pyridin-3(5H)-one
15D (0.25g, 0.44mmol) is added in reaction bulb, DMF (2.5mL) is added, ice bath cooling adds sodium hydride (0.07g, 1.8mmol), added, is warmed to room temperature reaction 1 hour.Reaction solution is poured into frozen water, ethyl acetate (20mL) is added, point liquid, water layer extracts 20mL × 2 with ethyl acetate, merges organic phase, with saturated aqueous common salt Wash (20mL × 4), anhydrous sodium sulfate drying, filtering is concentrated under reduced pressure, prepare liquid phase separation purification, obtain compound as white solid 15 (0.044g, yield 20%).
MS m/z(ESI):492.9[M+1]。
1H NMR(400MHz,DMSO)δ9.69(s,1H),8.41(s,1H),7.89(s,1H),7.60(s,2H),7.25(s,1H),5.76(s,1H),4.67(tt,1H),4.40(t,1H),4.06–3.97(m,1H),3.97(s,3H),3.80(dd,1H),3.08(td,1H),2.36(d,1H),2.21(d,1H),1.47(q,2H)。
Embodiment 16
7- ((4- ((the chloro- 2- fluorophenyls of 3,4- bis-) amino) -7- methoxyquinazoline hydrochloride -6- bases) epoxide) tetrahydrochysene -1H- oxazoles [3,4-a] pyridine -3 (5H)-thioketones (compound 16)
7-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)tetrahydro-1H-oxazolo[3,4-a]pyridin-3(5H)-thione
The first step:Tertiary butyl-4-hydroxy -2- (methylol) piperidines -1- formic acid esters (16B)
tert-butyl 4-hydroxy-2-(hydroxymethyl)piperidine-1-carboxylate
15B (1.35g, 5.21mmol) is added in reaction bulb, ethanol (15mL) is added, 0 DEG C is cooled to, sodium borohydride (0.99g, 26mmol) is added, adds and is slowly increased to room temperature reaction 8 hours.Reaction solution is poured into the aqueous solution (100mL of sodium hydroxide, in 1mol/L), add ethyl acetate (30mL), stirring point liquid, separates organic phase, aqueous phase is extracted (30mL × 2) with ethyl acetate, merge organic phase, anhydrous sodium sulfate drying, is concentrated under reduced pressure, silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=50:1-10:1) colorless oil 16B (0.7g, yield 60%) is obtained.
MS m/z(ESI):254.1[M+23]。
1H NMR(400MHz,CDCl3)δ4.45(dd,1H),4.15–4.00(m,1H),3.96–3.77(m,2H),3.73 (dd,1H),3.62(dd,1H),2.92(t,1H),2.16–1.76(m,3H),1.59–1.20(m,9H)。
Second step:2- (methylol) piperidines -4- alcohol trifluoroacetate (16C)
2-(hydroxymethyl)piperidin-4-ol trifluoroacetic acid salt
16B (0.26g, 1.12mmol) is added in reaction bulb, dichloromethane (1.5mL) is added, ice bath cooling adds trifluoracetic acid (1.5mL), adds and be warmed to room temperature reaction 1 hour.It is concentrated under reduced pressure, it is not purified to be directly used in the next step.
MS m/z(ESI):132.1[M+1]。
3rd step:7- hydroxy tetrahydro -1H- oxazoles [3,4-a] pyridine -3 (5H)-thioketones (16D)
7-hydroxytetrahydro-1H-oxazolo[3,4-a]pyridine-3(5H)-thione
By 16C (0.147g, 1.12mmol) add in reaction bulb, add dichloromethane (3mL), ice bath is cooled down, and adds triethylamine (0.34g, 3.36mmol), add thiocarbonyldiimidazole (0.24g, 1.34mmol), add, be warmed to room temperature reaction 2 hours.Aqueous hydrochloric acid solution (10mL is added into reaction solution, 1mol/L), stirring point liquid, water layer is extracted (5mL × 7) with dichloromethane, merge organic phase, anhydrous sodium sulfate drying, filtering, it is concentrated under reduced pressure, silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1-70:1) yellow oil 16D (0.07g, yield 40%) is obtained.
MS m/z(ESI):174.1[M+1]。
1H NMR(400MHz,CDCl3)δ4.65(t,1H),4.40–4.25(m,3H),4.12–4.01(m,1H),3.59–3.46(m,1H),2.05(ddd,1H),1.86–1.74(m,2H),1.68–1.55(m,1H)。
4th step:7- ((4- ((the chloro- 2- fluorophenyls of 3,4- bis-) amino) -7- methoxyquinazoline hydrochloride -6- bases) epoxide) tetrahydrochysene -1H- oxazoles [3,4-a] pyridine -3 (5H)-thioketones (compound 16)
7-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)tetrahydro-1H-oxazolo[3,4-a]pyridin-3(5H)-thione
By 16D (0.18g, 1.0mmol), 4- ((3, the chloro- 2- fluorophenyls of 4- bis-) amino) -7- methoxyquinazoline hydrochloride -6- alcohol (0.74g, 2.0mmol), triphenylphosphine (1.1g, 4.0mmol), add in reaction bulb, add dichloromethane (10mL), be cooled to -20 DEG C, azo-2-carboxylic acid's di tert butyl carbonate (0.72g is added dropwise, dichloromethane solution (5mL) 3.1mmol), is dripped off, and is warmed to room temperature reaction 4 hours.It is concentrated under reduced pressure, silica gel column chromatography separating-purifying (methylene chloride/methanol (v/v)=100:1~70:1) compound as white solid 16 (0.127g, yield 24%) is obtained.
MS m/z(ESI):509.0[M+1]。
1H NMR(400MHz,DMSO)δ9.63(s,1H),8.41(s,1H),7.89(s,1H),7.69–7.50(m,2H),7.26(s,1H),4.80–4.56(m,2H),4.41–4.13(m,3H),3.95(s,3H),3.36–3.24(m,1H),2.45(d,1H),2.31(d,1H),1.57(d,2H)。
Biological test example
Biological test example 1:SRB cell proliferation experiments
SRB experiments are the proliferative conditions for detecting cell under medicine effect.After dosing 72 hours, by SRB dyeing and ELIASA 490nm readings, reflect the proliferative conditions of cell.
The orifice plate of plating cells 96, H1975 cell lines are per empty 8000 cells, and A431 cell lines are per empty 10000 cells, 37 DEG C of incubators, 5%CO2Overnight incubation.Before dosing, every kind of cell takes 6 holes to add the trichloroacetic acids of 30 μ l 50% and fixed.Compound is configured to solution with DMSO, 10 μM of maximum concentration, 5 times of dilutions totally ten gradients, with the culture medium gradient dilution testing compound containing 0.1%FBS, and is 2 times of final concentration.96 porocyte culture plates culture mediums of NCI-H1975, A431 cell are changed to the fresh culture medium (per μ l of hole 100) containing 0.1%FBS, add testing compounds of the 100 μ l containing 2 times of final concentrations, 37 DEG C of incubators, 5%CO2Culture 72 hours.After incubation terminates, 50 μ l 50%TCA are added, is placed in 4 DEG C of refrigerators and fixes 1h.Abandon liquid, 300 μ lddH2O is washed 5 times, drying at room temperature 1h or more long.50 μ l 0.4%SRB are added per empty, 15min is dyed.Dye liquor is abandoned, is washed 6-7 times with 1% acetic acid, drying at room temperature is saturating.Dissolved with 200 μ l 10mM unbuffered Trisbase (pH=10.5), Oscillating Flat 2h.ELIASA determines 490nm absorbances.
Table 1:SRB cell proliferation experiment results
Conclusion:The compounds of this invention, which has, significantly suppresses H1975 (L858R/T790M), A431 (EGFRwt amplifications) cel l proliferation.
Biological test example 2:Pharmacokinetic Evaluation
Male SD rat (purchased from the magnificent experimental animal Co., Ltd of dimension tonneau) 180-240g, fasting feedwater is stayed overnight, 3 Oral Administration in Rats gavages 5mg/kg, 3 rat intravenous injection 1mg/kg.Oral administration group, compound is configured to 0.5mg × mL with 0.5% methylcellulose (MC) solution (containing 0.4% Tween 80)-1Suspension, before administration and upon administration 30 minutes and 1,2,4,6,8, blood samplings in 12 and 24 hours;Intravenously administrable group, compound is with 10% DMA, and 20%Solutol HS-15 (30%, w/v) and 70% normal saline are into 0.2mg × mL-1Solution, before administration and upon administration 5,15 and 30 minutes and 1,2,4,8, blood samplings in 12 and 24 hours.Anticoagulant heparin.5500 revs/min of blood sample is centrifuged 10 minutes, collects blood plasma, in -80 DEG C of preservations.Each μ L of time point rat plasma 10 are taken, the μ L of acetonitrile solution 500 of containing the internal standard are added After mixing, vortex mixed 4 minutes, 3700 revs/min centrifuge 18 minutes, take the μ L of supernatant 70 to be mixed with 70 μ L water, take the μ L of mixed liquor 5 to carry out LC-MS/MS analyses.Main pharmacokinetic parameter is analyzed with the non-compartment model of the softwares of WinNonlin 6.3, and Pharmacokinetic Evaluation result of the test is shown in Table 2.
The Pharmacokinetic Evaluation result of the test of table 2
Conclusion:The compounds of this invention shows to be substantially better than control drug Afatinib Pharmacokinetic Characteristics.
Biological test example 3:Nude mouse tumor cell transplantation knurl measuring
(1) experimental animal:
BALB/cA-nude nude mouses, 4-6 week old, female, purchased from Beijing Vital River Experimental Animals Technology Co., Ltd..Quality certification number:SCXK (capital) 2012-0001.
(2) tumor cell transplantation:
NCI-H1975 cells:After nude mouse laboratory environment is adapted to 3 days, right flank subcutaneous vaccination H1975 cells 5 × 106/ only.Tumour growth is to 200-250mm3When, random packet, every group of 10 mouse.
A431 cells:After nude mouse laboratory environment is adapted to 3 days, right flank subcutaneous vaccination A431 cells 5 × 106/ only.Tumour growth is to 200-250mm3When, random packet, every group of 10 mouse.
(3) dosage and method:
NCI-H1975 cell transplantation knurl nude mouses:Test-compound is configured to two dosage of 0.15mg/ml and 0.5mg/ml with PEG400/Tween80.Animal distinguishes gastric infusion test-compound 1.5mg/kg and 5mg/kg after weighing in;Afatinib is configured to 1.2mg/ml and 4mg/ml with PEG400/Tween80.Animal distinguishes gastric infusion test-compound 12mg/kg and 40mg/kg after weighing in.Blank control group gives the PEG400/Tween80 solution of same volume not drug containing.It is administered once a day, successive administration 10 days.
A431 cell transplantation knurl nude mouses:Test-compound is configured to 0.03mg/ml and 0.1mg/ml with PEG400/Tween80.Animal distinguishes gastric infusion test-compound 0.3mg/kg and 1mg/kg after weighing in;Afatinib is configured to 0.3mg/ml and 1mg/ml with PEG400/Tween80.Animal weigh in after respectively gastric infusion test-compound 3mg/kg and 10mg/kg blank control group give the PEG400/Tween80 solution of same volume not drug containing.It is administered once a day, successive administration 10 days.
(4) transplantable tumor volume and nude mouse body weight determination
After tumor cell inoculation nude mouse on every Mondays, four a knurl volume (calculation formula is seen below) is respectively surveyed with slide measure, and weigh in.
(5) data statistics
Average value and standard deviation (SD), SEM (STDEV/SQRT) are calculated using Office Excel softwares.Group difference is examined with T-testj, P<0.05 is significant difference standard
Gross tumor volume (V) calculation formula is:V=1/2 × major diameter × minor axis × minor axis
Relative volume (RTV)=VT/V0
Tumour inhibiting rate (%)=(CRTV-TRTV)/CRTV × 100%)
Wherein V0, VT are respectively to start the gross tumor volume before administration and after last time administration.CRTV, TRTV are respectively the relative tumour volume of blank control group and experimental group after time last administration.It the results are shown in Table 3,4.
Table 3:To the histamine result of A431 cell transplantation knurls
Table 4:To the histamine result of H1975 cell transplantation knurls
Conclusion:The compounds of this invention is compared with control drug Afatinib, it more can substantially suppress the human lung cancer NCI-H1975 of human epithelial cells cancer A431 and EGFR the T790M mutation containing EGFR wild types growth, in the case where dosage is only respectively 1/10,1/8 dosage of positive control, control drug is significantly stronger than to A431, H1975 transplantable tumor effect respectively.
Biological test example 4:Effect of the manual patch-clamp detection the compounds of this invention of electro physiology to hERG potassium-channels
This experiment cell used has hERG cDNA and stable expression hERG passages Chinese hamster ovary celI system for transfection.Make the record of full cell currents using manual Patch Clamp System (German HEKA EPC-10 signal amplifiers and digital switching system).The circular slide that superficial growth has CHO hERG cells is placed in the electrophysiological recording groove under inverted microscope.Continuous perfusion (about per minute 1 milliliter) is made with extracellular fluid in track.Experimentation uses conventional whole-cell patch-clamp electric current recording technique.Cell is clamped down under -80mV voltage.The depolarising of cell Clamping voltages clamps down on -50mV to eliminate inactivation and produce tail current again to+20mV to activate hERG potassium channels, after 5 seconds.Tail current peak value is used as the numerical value of hERG size of current.The hERG potassium currents that above-mentioned steps are recorded extracellular fluid perfusion lasting in track can then be superimposed perfusion medicine to be tested after being issued to stabilization, until medicine reaches stable state to the inhibitory action of hERG electric currents.Reach the size of stable situation later with extracellular fluid perfusion wash before hERG current reverts to dosing thing.
Testing compound concentration is 30,10,3,1,0.3,0.1 and 0.03 μM, to test.Before the test, 30,10,3,1,0.3 and 0.1mM stock solution are diluted to DMSO first, then is diluted to extracellular fluid final μM concentration.DMSO ultimate density is all 0.1% in each concentration compound solution.Positive control Cisapride (Cisapride) test concentrations are 0.1 μM.
Test data is by HEKA Patchmaster, and the DAS that Microsoft Excel and Graphpad Prism are provided is analyzed, as a result such as table 5.
Table 5:To the exercising result of hERG potassium-channels
Compound number hERG IC<sub>50</sub>(μM)
Afatinib 2.40
9 5.84
13 3.55
14 2.12
Conclusion:The compounds of this invention suppresses weak to hERG potassium-channels, with higher security, particularly compound 9 and 13, suppresses weaker to hERG potassium-channels, possesses greater security.

Claims (10)

  1. A kind of formula (II0) shown in compound or its stereoisomer, hydrate, metabolite, solvate, pharmaceutically acceptable salt, eutectic or prodrug,
    Wherein:
    R2Selected from C1-3Alkyl, 3 to 4 yuan of carbocylic radicals or 4 to 8 circle heterocycles bases, described heterocyclic radical are optionally further selected from F, Cl, hydroxyl, C by 0 to 21-3Alkyl or C1-3The substituent of alkoxy is replaced, and described heterocyclic radical contains 1 to 2 hetero atom for being selected from N or O;
    Ring A is selected fromOr
    R3Selected from F, CF3、CHF2、CH2F, Cl, hydroxyl or C1-3Alkyl;
    X1 isOr X1Formed together with ring AOr
    R4、R5And R6Respectively H;
    R9Selected from F, Cl, hydroxyl, C1-3Alkyl or C2-3Alkynyl;
    N, m, t each are selected from 0,1,2,3 or 4;
    Restrictive condition is, when n is 0, R2Selected from C1-3Alkyl, and m be 0 when, ring A is not
  2. Compound according to claim 1 or its stereoisomer, hydrate, metabolite, solvate, pharmaceutically acceptable salt, eutectic or prodrug, wherein described compound is selected from the compound shown in logical formula (II):
    Wherein:
    R2Selected from methyl, ethyl or
    Ring A is selected fromOr
    N is 0 or 1;
    R3Selected from F;
    M is 0 or 1;
    R4、R5And R6It is H;
    R9Selected from F or Cl;
    T is 3;
    Restrictive condition is, when n is 0, R2Selected from methyl or ethyl, and m, when being 0, ring A is not
  3. Compound according to claim 1 or its stereoisomer, hydrate, metabolite, solvate, pharmaceutically acceptable salt, eutectic or prodrug, wherein described compound is selected from:
  4. Compound or its stereoisomer according to claim 1, hydrate, metabolite, solvate, pharmaceutically acceptable salt, eutectic or prodrug, salt wherein described in its pharmaceutically acceptable salt is selected from hydrochloride, hydrobromate, sulfate, phosphate, acetate, trifluoroacetate, rhodanate, maleate, hydroxymaleic acid salt, glutarate, mesylate, esilate, benzene sulfonate, tosilate, benzoate, salicylate, phenylacetate, cinnamate, lactate, malonate, pivalate, succinate, fumarate, malate, mandelate, tartrate, gallate, gluconate, laruate, palmitate, pectate, picrate, citrate or combinations thereof.
  5. Compound or its stereoisomer, hydrate, metabolite, solvate, pharmaceutically acceptable salt, eutectic or prodrug according to claim 1, wherein described salt is selected from hydrochloride, hydrobromate, sulfate, phosphate, acetate, maleate, mesylate, benzene sulfonate, trifluoroacetate, tosilate, benzoate, salicylate, cinnamate, lactate, malonate, succinate, fumarate, malate, tartrate, citrate or combinations thereof.
  6. A kind of pharmaceutical composition, described composition includes:The compound according to any one of claim 1-3 or its stereoisomer, hydrate, metabolite, solvate, pharmaceutically acceptable salt, eutectic or prodrug of effective dose, and pharmaceutically acceptable carrier, diluent, adjuvant, medium or excipient;Described composition can also further comprise one or more other therapeutic agents.
  7. Pharmaceutical composition according to claim 6, wherein described other therapeutic agents are cis-platinum (cisplatin), carboplatin (carboplatin), oxaliplatin (oxaliplatin), Dacarbazine (dacarbazine), Temozolomide (temozolomide), procarbazine (procarbazine), methotrexate (MTX) (methotrexate), fluorouracil (fluorouracil), cytarabine (cytarabine), gemcitabine (gemcitabine), purinethol (mercaptopurine), fludarabine (fludarabine), vincaleukoblastinum (vinblastine), vincristine (vincristine), vinorelbine (vinorelbine), taxol (paclitaxel), Docetaxel (docetaxel),TPT (topotecan),Irinotecan (irinotecan),Etoposide (etoposide),ET-743 (trabectedin),Dactinomycin D (dactinomycin),Doxorubicin (doxorubicin),Epirubicin (epirubicin),Daunomycin (daunorubicin),Mitoxantrone (mitoxantrone),Bleomycin (bleomycin),Mitomycin C (mitomycin),Ipsapirone (ixabepilone),TAM (tamoxifen),Flutamide (flutamide),Sirolimus (sirolimus),Afatinib(afatinib),alisertib,amuvatinib,A Pa replaces Buddhist nun (apatinib),Axitinib (axitinib),Bortezomib (bortezomib),SKI-606 (bosutinib),Bu Linibu (brivanib),Card is rich to replace Buddhist nun (cabozantinib),AZD2171 (cediranib),crenolanib,Ke Zhuo replaces Buddhist nun (crizotinib),Da Lafeini (dabrafenib),dacomitinib,Da Lushe replaces (danusertib),Dasatinib (dasatinib),Many Weis replace Buddhist nun (dovitinib),Tarceva (erlotinib),foretinib,ganetespib,Gefitinib (Gefitinib),Buddhist nun (ibrutinib) is replaced according to Shandong,Conmana (icotinib),Imatinib (imatinib),iniparib,Lapatinib (lapatinib),lenvatinib,linifanib,linsitinib,Masitinib (masitinib),momelotinib,Not for husky Buddhist nun (motesanib),HKI-272 (neratinib),Nilotinib (nilotinib),niraparib,oprozomib,olaparib,Pazopanib (pazopanib),pictilisib,ponatinib,quizartinib,regorafenib,rigosertib,rucaparib,ruxolitinib,Saracatinib (saracatinib),saridegib,Sorafenib (sorafenib),Sutent (sunitinib),tasocitinib,telatinib,tivantinib,tivozanib,tofacitinib,trametinib,ZD6474 (vandetanib),veliparib,Wei Luofeini (vemurafenib),vismodegib,volasertib,Alemtuzumab (alemtuzumab),Bevacizumab (bevacizumab),brentuximab vedotin,Catumaxomab (catumaxomab),Cetuximab (cetuximab),Ground promise monoclonal antibody (denosumab),Lucky trastuzumab (gemtuzumab),Her monoclonal antibody (ipilimumab),Buddhist nun's trastuzumab (nimotuzumab),Difficult to understand (ofatumumab),Victibix (panitumumab),Rituximab (rituximab),Tositumomab (tositumomab),Herceptin (trastuzumab) or combinations thereof.
  8. The pharmaceutical composition any one of compound or its stereoisomer, hydrate, ester, solvate, eutectic, metabolite, pharmaceutically acceptable salt or prodrug or claim 6-7 any one of claim 1-5 is particularly preparing the application in being used to treat and/or prevent the pharmaceutical preparation of excess proliferative disease in the application as EGFR/HER2 receptor tyrosine kinase inhibitors in pharmaceutical preparation is prepared.
  9. A kind of method treated and/or prevent excess proliferative disease, this method includes giving the compound or its stereoisomer any one of the claim 1-5 of individual effective dose, hydrate, ester, solvate, eutectic, metabolite, pharmaceutically acceptable salt or prodrug or the pharmaceutical composition any one of claim 6-7, wherein described excess proliferative disease includes brain tumor, non-small cell lung cancer, squamous cell, carcinoma of urinary bladder, cancer of pancreas, colon cancer, breast cancer, oophoroma, cervix cancer, carcinoma of endometrium, colorectal cancer, kidney, adenocarcinoma of esophagus, esophageal squamous cell carcinoma, solid tumor, NHL, liver cancer, lung cancer, cutaneum carcinoma, thyroid cancer, head and neck cancer, prostate cancer, one or more in glioma and nasopharyngeal carcinoma.
  10. Method according to claim 9, wherein the excess proliferative disease includes the one or more in non-small cell lung cancer, breast cancer, epidermis squamous carcinoma, stomach cancer and colon cancer.
CN201580012570.5A 2014-04-11 2015-04-13 Quinazoline derivant and preparation method thereof and application in medicine Expired - Fee Related CN106068262B (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN201410145927 2014-04-11
CN2014101459274 2014-04-11
PCT/CN2015/076449 WO2015154725A1 (en) 2014-04-11 2015-04-13 Quinazoline derivative and preparation method and use in medicine thereof

Publications (2)

Publication Number Publication Date
CN106068262A true CN106068262A (en) 2016-11-02
CN106068262B CN106068262B (en) 2019-10-29

Family

ID=54287332

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201580012570.5A Expired - Fee Related CN106068262B (en) 2014-04-11 2015-04-13 Quinazoline derivant and preparation method thereof and application in medicine

Country Status (2)

Country Link
CN (1) CN106068262B (en)
WO (1) WO2015154725A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112125890A (en) * 2020-09-25 2020-12-25 华东理工大学 Isoindolinone-based quinazoline-based carboxylic ester derivative and application thereof
CN114516862A (en) * 2022-03-17 2022-05-20 李明 Quinazoline derivative and preparation method and application thereof
CN114560852A (en) * 2022-03-17 2022-05-31 李明 Quinazoline derivative and preparation method and application thereof

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105399733B (en) * 2015-12-03 2018-04-24 中国人民解放军南京军区南京总医院 A kind of novel quinazoline quinoline derivant LU1504 and its preparation method and application
CN108069946B (en) * 2016-11-08 2020-06-05 威尚(上海)生物医药有限公司 Substituted quinazoline compounds having the ability to cross the blood brain barrier
JP7296641B2 (en) * 2017-12-19 2023-06-23 チェンドゥ・ジンルイ・ファウンデーション・バイオテック・カンパニー・リミテッド Quinazoline derivatives and uses thereof
WO2019196619A1 (en) * 2018-04-08 2019-10-17 威尚(上海)生物医药有限公司 Quinazoline derivative salt-form crystal form, and preparation method therefor and application thereof
CN110343090B (en) * 2018-04-08 2021-06-04 威尚(上海)生物医药有限公司 Quinazoline derivative salt crystal form, preparation method and application
CN110357858B (en) * 2018-04-09 2022-02-18 威尚(上海)生物医药有限公司 5-substituted difluoropiperidine compounds having blood-brain barrier crossing capability
CN110054613B (en) * 2018-05-24 2020-11-27 深圳市塔吉瑞生物医药有限公司 Substituted aminoquinazoline compound, pharmaceutical composition and application thereof
WO2019233459A1 (en) * 2018-06-08 2019-12-12 江苏威凯尔医药科技有限公司 Human epidermal growth factor receptor inhibitor, preparation method therefor and use thereof
CN110577514B (en) * 2018-06-08 2022-07-05 江苏威凯尔医药科技有限公司 Human epidermal growth factor receptor inhibitor and preparation method and application thereof
WO2020053816A1 (en) * 2018-09-14 2020-03-19 Hanmi Pharmaceutical Co., Ltd. Crystalline forms of a quinazole compound and its hydrochloride salts
CN113286790B (en) * 2019-01-18 2024-03-15 成都金瑞基业生物科技有限公司 Application of quinazoline derivative in preparation of medicine for treating nasopharyngeal carcinoma
US11365189B2 (en) 2019-04-25 2022-06-21 Board Of Regents, The University Of Texas System Heterocyclic inhibitors of tyrosine kinase
WO2021127397A1 (en) * 2019-12-19 2021-06-24 Black Diamond Therapeutics, Inc. Nitrogen heterocyclic compounds and methods of use
CN113278012B (en) * 2020-02-19 2022-07-12 郑州同源康医药有限公司 Compounds useful as kinase inhibitors and uses thereof
CN113444074B (en) * 2020-03-26 2022-08-09 上海中医药大学 Compound with EGFR (epidermal growth factor receptor) and Wnt dual inhibition effects as well as preparation method and application thereof
CN111763215B (en) * 2020-07-21 2021-05-18 成都海博为药业有限公司 Compound with nitrogen-containing heterocyclic structure and preparation method and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101679384A (en) * 2007-06-05 2010-03-24 韩美药品株式会社 Novel amide derivative for inhibiting the growth of cancer cells
WO2011029265A1 (en) * 2009-09-14 2011-03-17 江苏恒瑞医药股份有限公司 6-amino quinazoline or 3-cyano quinoline derivatives, preparation methods and pharmaceutical uses thereof
CN102731485A (en) * 2011-04-02 2012-10-17 齐鲁制药有限公司 4-(substituted phenylamino)quinazoline derivative, its preparation method, pharmaceutical composition and application

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1660479A1 (en) * 2003-07-29 2006-05-31 Astrazeneca AB Piperidyl-quinazoline derivatives as tyrosine kinase inhibitors
WO2008033749A2 (en) * 2006-09-11 2008-03-20 Curis, Inc. Quinazoline based egfr inhibitors containing a zinc binding moiety
CN105037333A (en) * 2009-12-21 2015-11-11 张强 Novel quinazoline derivatives

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101679384A (en) * 2007-06-05 2010-03-24 韩美药品株式会社 Novel amide derivative for inhibiting the growth of cancer cells
WO2011029265A1 (en) * 2009-09-14 2011-03-17 江苏恒瑞医药股份有限公司 6-amino quinazoline or 3-cyano quinoline derivatives, preparation methods and pharmaceutical uses thereof
CN102731485A (en) * 2011-04-02 2012-10-17 齐鲁制药有限公司 4-(substituted phenylamino)quinazoline derivative, its preparation method, pharmaceutical composition and application

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
仇缀百: "《药物设计学(第1版)》", 30 November 1999, 高等教育出版社 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112125890A (en) * 2020-09-25 2020-12-25 华东理工大学 Isoindolinone-based quinazoline-based carboxylic ester derivative and application thereof
CN112125890B (en) * 2020-09-25 2022-12-06 华东理工大学 Isoindolinone-based quinazoline-based carboxylic ester derivative and application thereof
CN114516862A (en) * 2022-03-17 2022-05-20 李明 Quinazoline derivative and preparation method and application thereof
CN114560852A (en) * 2022-03-17 2022-05-31 李明 Quinazoline derivative and preparation method and application thereof
CN114516862B (en) * 2022-03-17 2024-03-01 河北康泰药业有限公司 Quinazoline derivative and preparation method and application thereof

Also Published As

Publication number Publication date
WO2015154725A1 (en) 2015-10-15
CN106068262B (en) 2019-10-29

Similar Documents

Publication Publication Date Title
CN106068262B (en) Quinazoline derivant and preparation method thereof and application in medicine
JP7374960B2 (en) fused ring compound
TWI609012B (en) Aminoquinazoline derivatives and their salts and methods of use thereof
CN103874689B (en) The combination of AKT inhibitor compound and Wei Luofeini and using method
CN1681508B (en) Novel inhibitors of kinases
KR20170097651A (en) TETRAHYDRO-PYRIDO[3,4-b]INDOLE ESTROGEN RECEPTOR MODULATORS AND USES THEREOF
CA2853390A1 (en) Anticancer pyridopyrazines via the inhibition of fgfr kinases
TWI557126B (en) Serine/threonine kinase inhibitors
WO2021079302A1 (en) Pharmaceutical combination of prmt5 inhibitors
CN103140484A (en) Cyclic amine azaheterocyclic carboxamides
CN106279128B (en) Epoxyethane derivative and preparation method thereof and application in medicine
CN105384695B (en) Pyrimidine derivative, preparation method and medical application thereof
CN106916112B (en) Pyrimidine derivative, preparation method and medical application thereof
CN103319468B (en) The spiral shell dicyclic compound replaced and using method and purposes
TWI570116B (en) Substituted pyrazolone compounds and methods of use
RU2783706C1 (en) Compounds with condensed rings
WO2023239945A1 (en) Btological activities of 5-(2-(4-(4-fluoro-2-methyl-1h-indol-5- yloxy)-6-methoxyquinolin-7-yloxy)ethyl)-5-azaspiro[2.4]-heptan- 7-ol crystalline, phosphoric acid salt and its enantiomers
KR20220128590A (en) Novel pyrimidine derivative showing growth inhibition of cancer cell
CN106279045A (en) Cyclopropane derivative and preparation method thereof and in application pharmaceutically
WO2009117352A1 (en) Cancer treatment 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
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20190726

Address after: 17 No. 856099, Tibet autonomous region, city Zedang town Sanxiang Road

Applicant after: HAISCO PHARMACEUTICAL Group Inc.

Address before: Gabriel Road on both sides of the Strait science and Technology Park in Wenjiang District of Chengdu City, Sichuan Province, No. 136 611130

Applicant before: Sichuan Haisco Pharmaceutical Co.,Ltd.

GR01 Patent grant
GR01 Patent grant
CF01 Termination of patent right due to non-payment of annual fee
CF01 Termination of patent right due to non-payment of annual fee

Granted publication date: 20191029