WO2015154725A1 - 喹唑啉衍生物及其制备方法和在医药上的应用 - Google Patents

喹唑啉衍生物及其制备方法和在医药上的应用 Download PDF

Info

Publication number
WO2015154725A1
WO2015154725A1 PCT/CN2015/076449 CN2015076449W WO2015154725A1 WO 2015154725 A1 WO2015154725 A1 WO 2015154725A1 CN 2015076449 W CN2015076449 W CN 2015076449W WO 2015154725 A1 WO2015154725 A1 WO 2015154725A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
acid
oxy
added
compound
Prior art date
Application number
PCT/CN2015/076449
Other languages
English (en)
French (fr)
Inventor
李瑶
徐波
陈雷
李升�
刘建余
钱枚琳
Original Assignee
四川海思科制药有限公司
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 四川海思科制药有限公司 filed Critical 四川海思科制药有限公司
Priority to CN201580012570.5A priority Critical patent/CN106068262B/zh
Publication of WO2015154725A1 publication Critical patent/WO2015154725A1/zh

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

Definitions

  • the invention relates to a quinazoline derivative, a preparation method thereof and application in medicine, in particular to a novel quinazoline derivative having EGFR/HER2 double target inhibition effect or a stereoisomer thereof and a hydrate thereof , solvates, metabolites, pharmaceutically acceptable salts, eutectic or prodrugs, pharmaceutical compositions thereof, and their use in medicine.
  • the receptor tyrosine kinase superfamily in cell surface receptors plays an important role in the regulation of cellular signaling by extracellular growth factors.
  • the receptor tyrosine kinase is capable of catalyzing the transfer of a phosphate group from ATP to the tyrosine group of the substrate.
  • these kinases are in an unphosphorylated monomer state, and their kinase domain is in an inactive structure.
  • the receptor When the ligand binds to the extracellular domain of the receptor tyrosine kinase, the receptor undergoes oligomerization, and autophosphorylation increases the catalytic activity of the kinase and forms a binding site for the signaling protein, and the signal protein binds thereto. Thereby a plurality of signal paths are activated. These signaling pathways are linked to each other to regulate cell proliferation, survival, differentiation, function, migration and apoptosis.
  • the cells When the receptor tyrosine kinase is deregulated and abnormally activated, the cells will transform into tumor cells, which have enhanced proliferation, growth and drug resistance, and have strong angiogenic, invasive and metastatic ability (Yarden and Sliwkowski, 2001, Nat Rev Mol Cell Biol, 2, 127-137).
  • Epidermal growth factor (EGF) receptors are a class of receptor tyrosinases of interest.
  • the EGF receptor family comprises four members: EGFR/ErbB1/HER1, ErbB2/HER2/Neu, ErbB3/HER3, and ErbB4/HER4.
  • Normal EGFR-activated signaling pathways regulate cell proliferation, angiogenesis, growth, migration, and adhesion, and play an important role in organogenesis or in the interaction between adult cells and cells (Wu, Zhong et al., 2007, J Thorac Oncol, 2, 430-439). Many molecular times can lead to sustained activation of EGFR kinase activity, constantly triggering a large number of downstream signal transduction pathways, including K-ras activation.
  • the EGFR family is closely related to tumorigenesis.
  • Overexpression or mutation of EGFR can be detected in various cancers such as head and neck cancer, ovarian cancer, bladder cancer, cervical cancer, esophageal cancer, gastric cancer, breast cancer, endometrial cancer, colon cancer, lung cancer and brain tumor, usually indicating This is a poor prognosis.
  • HER2 is considered to be a strong tumor protein, and its mutations are also seen in a variety of tumors, especially breast cancer, lung cancer and colon cancer (Olayioye, Neve et al, 2000, EMBO J, 19, 3159-3167 ).
  • EGFR tyrosine kinase a variety of small molecule inhibitors of EGFR tyrosine kinase have been developed, such as the first generation of gefitinib and erlotinib, which can selectively reversibly inhibit EGFR, thereby preventing tumor growth, especially Tumors with exon 19 deletion (eg del747-750) and exon 21 mutation (eg L858R) (Barker, Gibson et al, 2001, Bioorg Med) Chem Lett, 11, 1911-1914; Wakeling, Guy et al, 2002, Cancer Res, 62, 5749-5754).
  • exon 19 deletion eg del747-750
  • exon 21 mutation eg L858R
  • T790M mutation affects the catalytic domain of the kinase, attenuating the interaction of the targeted inhibitor with the target, rendering the treatment ineffective (Pao and Chmielecki, 2010, Nat Rev Cancer, 10, 760-774).
  • irreversible double inhibitors such as Afatinib, neratinib, and carartinib
  • Afatinib, neratinib, and carartinib have been developed, which are effective at inhibiting EGFR-sensitive mutations at low concentrations (such as del747-750, compared to reversible inhibitors). L858R), and can effectively overcome the resistance caused by T790M.
  • irreversible dual inhibitors do not produce rapid resistance like gefitinib and erlotinib (Ercan, Zejnullahu et al, 2010, Oncogene, 29, 2346-2356; Chmielecki , Foo et al, 2011, Sci Transl Med, 3, 90ra59).
  • CN10679384 describes novel amide derivatives for inhibiting the growth of cancer cells, the structural formula of which is as follows:
  • R 4 , R 5 , R 6 and R 7 are each independently hydrogen, halogen, NC 1-6 alkyl or N-hydroxyacylamino or CC 1-6 alkyltransamido (-NHCOC 1-6 ), hydroxycarbonyl (-COOH), C 1-6 alkoxycarbonyl (-COOC 1-6 ), C 1-6 alkyl, or an alkyl group substituted by a hydroxy group, a C 1-6 dialkylamine or a heterocyclic group
  • R 1 is an aryl or heterocyclic group substituted by 1 to 5 X, or a C 1-6 alkyl group substituted by an aryl group
  • R 2 is hydrogen, a hydroxyl group, a C 1-6 alkoxy group, or It is a substituted heterocyclyl C 1-6 alkoxy or C 1-6 alkoxy group
  • R 3 is hydrogen, -COOH, C 1-6 alkoxycarbonyl group, or an unsubstituted acylamino N- Or N-
  • CN102731485 describes 4-(substituted phenylamino)quinazoline derivatives having the structural formula:
  • R 1 is selected from R 2 is selected from hydrogen or N,N-dimethylaminomethyl;
  • R 3 is selected from methoxyethyl, tetrahydrofuran-3-yl, (S)-tetrahydrofuran-3-yl or (R)-tetrahydrofuran-3-yl
  • each R 4 and R 5 are independently selected from one or more of hydrogen, halogen, C 1 -C 10 alkyl, C 1 -C 10 alkoxy, and halogenated C 1 -C 10 alkyl;
  • WO2011029265 describes quinazoline derivatives having the following structural formula:
  • A is selected from a carbon atom or a nitrogen atom; when A is a carbon atom, R 1 is selected from a hydrogen atom or an alkoxy group, wherein the alkoxy group is further further selected from one or more selected from halogen or alkoxy.
  • R 2 is selected from a cyano group; when A is a nitrogen atom, R 1 is selected from a hydrogen atom or an alkoxy group, wherein the alkoxy group is optionally further selected from one or more selected from the group consisting of halogen Substituted by a substituent of an alkoxy group, R 2 is unsubstituted; R 3 has the following structure: -DTL or -D; D is selected from an aryl or heteroaryl group, wherein the aryl or heteroaryl group is independently Optionally further substituted with one or more substituents selected from halogen, alkyl or trifluoromethyl; T is selected from -(CH 2 )r-, -O(CH 2 )r-, -NH(CH 2 R- or -S(O)r(CH 2 )r-; L is selected from aryl or heteroaryl, wherein the aryl or heteroaryl are each independently optionally further protected by
  • R 6 and R 7 are unsubstituted;
  • R 8 is selected from a hydrogen atom or an alkyl group;
  • R 9 is selected from a hydrogen atom, an alkyl group, an aryl group, a carboxyl group or a carboxyl group.
  • the present invention provides a novel class of compounds which have been shown to exhibit excellent pharmacodynamic activity as EGFR/HER2 dual target inhibitors.
  • the present invention provides a compound of the formula (II 0 ) or a stereoisomer, hydrate, metabolite, solvate, pharmaceutically acceptable salt, co-crystal or prodrug thereof,
  • R 2 is selected from C 1-3 alkyl or 4 to 8 membered heterocyclic group, and the heterocyclic group is further optionally 0 to 2 selected from F, Cl, hydroxy, C 1-3 alkyl or C Substituted with a substituent of 1-3 alkoxy group, and said heterocyclic group contains 1 to 2 hetero atoms selected from N or O;
  • Ring A is selected from
  • R 3 is selected from the group consisting of F, CF 3 , CHF 2 , CH 2 F, Cl, hydroxy or C 1-3 alkyl;
  • R 4 , R 5 and R 6 are each H;
  • R 9 is selected from the group consisting of F, Cl, hydroxy, C 1-3 alkyl or C 2-3 alkynyl;
  • n, m, t are each selected from 0, 1, 2, 3 or 4;
  • R 2 is selected from C 1-3 alkyl, and when m is 0, ring A is not
  • the compound of the invention is selected from the compounds of formula (II):
  • R 2 is selected from methyl, ethyl or
  • Ring A is selected from
  • n 0 or 1
  • R 3 is selected from F
  • n 0 or 1
  • R 4 , R 5 and R 6 are both H;
  • R 9 is selected from F or Cl
  • the compounds of the present invention are selected from, but not limited to:
  • a compound of the invention or a stereoisomer, hydrate, metabolite, solvate, pharmaceutically acceptable salt, eutectic or prodrug thereof, wherein said salt is selected from the group consisting of hydrochloric acid Salt, hydrobromide, sulfate, phosphate, acetate, trifluoroacetate, thiocyanate, maleate, hydroxymaleate, glutarate, methanesulfonate, B Sulfonate, besylate, p-toluenesulfonate, benzoate, salicylate, phenylacetate, cinnamate, lactate, malonate, pivalate, succinic acid Salt, fumarate, malate, mandelate, tartaric acid a salt, gallate, gluconate, laurate, palmitate, pectinate, picrate, citrate or a combination thereof, preferably, the salt is selected from the group consisting of hydrochloride
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising: an effective amount of a compound of the invention, or a stereoisomer, hydrate, metabolite, solvate, pharmaceutically acceptable salt thereof, A eutectic or prodrug, and a pharmaceutically acceptable carrier, diluent, adjuvant, vehicle or excipient.
  • the pharmaceutical composition of the invention further comprises one or more additional therapeutic agents.
  • the other therapeutic agents described in the pharmaceutical compositions of the invention include: cisplatin, carboplatin, oxaliplatin, dacarbazine, Temozolomide, procarbazine, methotrexate, fluorouracil, cytarabine, gemcitabine, mercaptopurine, fludarabine ), vinblastine, vincristine, vinorelbine, paclitaxel, docetaxel, topotecan, irinotecan, etopo Etoposide, trabectedin, dactinomycin, doxorubicin, epirubicin, daunorubicin, mitoxantrone ), bleomycin, mitomycin, ixabepilone, tamoxifen, flutamide, sirolimus, Afatinib (afatinib), alisertib Amuvatinib, apatinib, axitinib,
  • the present invention also provides the compound or a stereoisomer, hydrate, ester, metabolite, solvate, pharmaceutically acceptable salt, co-crystal or prodrug thereof or the pharmaceutical composition as a Use of an EGFR/HER2 receptor tyrosine kinase inhibitor for the preparation of a pharmaceutical formulation, in particular for the preparation of a pharmaceutical formulation for the treatment and/or prophylaxis of hyperproliferative diseases.
  • the hyperproliferative diseases include brain tumors, non-small cell lung cancer, epidermal squamous cell carcinoma, bladder cancer, pancreatic cancer, colon cancer, breast cancer, ovarian cancer, cervical cancer, endometrial cancer, colorectal cancer, Renal cancer, esophageal adenocarcinoma, esophageal squamous cell carcinoma, solid tumor, non-Hodgkin's lymphoma, liver cancer, lung cancer, stomach cancer, skin cancer, thyroid cancer, head and neck cancer, prostate cancer, glioma and nasopharyngeal carcinoma
  • the carbon, hydrogen, oxygen, sulfur, nitrogen or halogen involved in the groups and compounds of the present invention include their isotopic conditions, and the carbon, hydrogen, oxygen, sulfur involved in the groups and compounds of the present invention.
  • the nitrogen is optionally further replaced by one or more of their corresponding isotopes, wherein the carbon isotopes include 12 C, 13 C, and 14 C, and the hydrogen isotopes include ruthenium (H), ruthenium (D, also known as heavy hydrogen), ⁇ (T, also known as super heavy hydrogen), oxygen isotopes include 16 O, 17 O and 18 O, sulfur isotopes include 32 S, 33 S, 34 S and 36 S, nitrogen isotopes include 14 N and 15 N, fluorine The isotopes include 17 F and 19 F, the chlorine isotopes include 35 Cl and 37 Cl, and the bromine isotopes include 79 Br and 81 Br.
  • “Pharmaceutically acceptable salt” or “pharmaceutically acceptable salt thereof” means that the compound of the invention retains the biological effectiveness and properties of the free base, and the free base is reacted with a non-toxic inorganic or organic acid.
  • the inorganic acid include hydrofluoric acid, hydrochloric acid, hydrobromide, sulfate, phosphoric acid, hypochlorous acid, perchloric acid, iodic acid, carbonic acid, nitrous acid, nitric acid, metaboric acid, Boric acid, metasilicate, silicic acid, metaphosphorous acid, metaphosphoric acid, pyrophosphoric acid, hydrosulfuric acid, sulfurous acid, thiosulfuric acid and permanganic acid;
  • the organic acid include formic acid, acetic acid, trifluorocarbon Acetic acid, thiocyanate, maleic acid, hydroxymaleic acid, glutaric acid, methanesulfonic acid,
  • Carrier means a material that does not cause significant irritation to the organism and does not abrogate the biological activity and properties of the administered compound.
  • Excipient means an inert substance that is added to a pharmaceutical composition to facilitate administration of the compound.
  • Non-limiting examples include calcium carbonate, calcium phosphate, sugar, starch, cellulose derivatives (including microcrystalline cellulose), gelatin, vegetable oils, polyethylene glycols, diluents, granulating agents, lubricants, bonding Agent and disintegrant.
  • an “adjuvant” is a non-specific immunopotentiator that, when injected with an antigen or pre-infused into the body, enhances the body's immune response to the antigen or alters the type of immune response.
  • “Diluent” is also called “filler.”
  • the diluted inert substance is added.
  • Prodrug means a compound of the invention that can be metabolized by metabolism in vivo to a biologically active compound, such transformation being effected by hydrolysis of the prodrug in the blood or enzymatic conversion of the parent tissue into the blood tissue.
  • Prodrugs of the invention are prepared by modifying functional groups in the compounds of the invention which can be removed by conventional procedures or in vivo to provide the parent compound.
  • Cyctic crystal refers to a crystal in which an active pharmaceutical ingredient and a eutectic formation are combined by hydrogen bonding or other non-covalent bond, wherein the pure state of API and CCF is solid at room temperature, and each component There is a fixed stoichiometric ratio between them.
  • Eutectic is a multi-component crystal that contains both a binary eutectic formed between two neutral solids and a multi-component eutectic formed from a neutral solid and or a solvate.
  • Non-limiting examples of such "eutectic formers” include alanine, valine, leucine, isoleucine, valine, phenylalanine, tryptophan, methionine, glycine, serine, Threonine, cysteine, tyrosine, asparagine, glutamine, lysine, arginine, histidine, aspartic acid, aspartic acid, glutamic acid, pyroglutamic acid Acid, sulfuric acid, phosphoric acid, nitric acid, hydrobromic acid, acid, formic acid, acetic acid, propionic acid, benzenesulfonic acid, benzoic acid, phenylacetic acid, salicylic acid, alginic acid, anthranilic acid, camphoric acid, citric acid, ethylene Acid, formic acid, fumaric acid, citric acid, gluconic acid, glucuronic acid, glutamic acid, glycolic acid, isethionic acid, lactic acid
  • Stepoisomer refers to isomers resulting from the spatial arrangement of atoms in a molecule, including cis and trans isomers, enantiomers and conformational isomers.
  • heterocyclic group optionally substituted by an alkyl group means that the alkyl group may be, but not necessarily, the description includes the case where the heterocyclic group is substituted by an alkyl group, and the heterocyclic group thereof is not substituted with an alkyl group.
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) or (and) mass spectrometry (MS).
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • the NMR was measured using a (Bruker Avance III 400 and Bruker Avance 300) nuclear magnetic apparatus, and the solvent was deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD). ), the internal standard is tetramethylsilane (TMS).
  • the HPLC was measured using an Agilent 1260 DAD high pressure liquid chromatograph (Zorbax SB-C18100 x 4.6 mm).
  • Thin layer chromatography silica gel plate uses Yantai Yellow Sea HSGF254 or Qingdao GF254 silica gel plate.
  • the specification of silica gel plate used for thin layer chromatography (TLC) is 0.15mm ⁇ 0.20mm.
  • the specification for thin layer chromatography separation and purification is 0.4mm. ⁇ 0.5mm.
  • the known starting materials of the present invention may be synthesized by or according to methods known in the art, or may be purchased from Titan Technology, Anheji Chemical, Shanghai Demer, Chengdu Kelon Chemical, Suiyuan Chemical Technology, and Belling Technology. And other companies.
  • the nitrogen atmosphere means that the reaction flask is connected to a nitrogen balloon having a volume of about 1 L.
  • the hydrogen atmosphere means that the reaction flask is connected to a hydrogen balloon of about 1 L volume.
  • the hydrogenation reaction is usually evacuated, charged with hydrogen, and operated three times.
  • reaction was carried out under a nitrogen atmosphere.
  • the solution means an aqueous solution.
  • reaction temperature is room temperature.
  • the room temperature is an optimum reaction temperature of 20 ° C to 30 ° C.
  • Bn benzyl; Me: methyl; Et: ethyl; Ts: p-toluenesulfonyl; TBS: tert-butyldimethylsilyl; Boc: Tert-butoxycarbonyl; Ac: acetyl.
  • 6-Hydroxy-7-methoxy-3H-quinazolin-4-one 1a (40 g, 208 mmol, Kangmanin) and acetic anhydride (296 mL, 211 mmol) were added to the reaction flask. After stirring, pyridine (67 mL) was added. The mixture was stirred at 100 ° C for 4 hours and then cooled to room temperature. The reaction solution was poured into ice water (500 g), stirred for 5 minutes, and filtered under reduced pressure. The filter cake was washed with water (100 mL ⁇ 2) and petroleum ether (100 mL ⁇ 2), and the filter cake was collected and dried to give a gray solid. 1b (36g, yield: 73%).
  • 3b (51.0 g, 0.135 mol) and 7 M ammonia methanol solution (400 mL) were added to the reaction flask, and stirred at room temperature for 1 hour. After suction filtration, the mixture was washed with methanol (100 mL ⁇ 2), and then filtered and dried to give 3c (23.0 g, yield: 50.0%).
  • N-tert-butoxycarbonylpropenylamine 4a (20 g, 127.39 mmol) and N,N-dimethylformamide (100 mL) were added to the reaction flask, and sodium hydride (60% dispersed in paraffin oil, 7.6) was added. g,191.08 mmol), after completion of the dropwise addition, stirring at 0 ° C for 30 minutes, naturally stirring to room temperature and stirring for 20 minutes, dropwise addition of bromopropyne (30.3 g, 245.78 mmol), and stirring at 0 ° C for 30 minutes, the reaction was carried out.
  • N-fluorobisbenzenesulfonamide (3.77 g, 11.6 mmol) and sodium carbonate (1.85 g, 17.3 mmol) were added to the reaction flask, tetrahydrofuran (33 mL) was added, and the mixture was cooled to -20 ° C, and the above-mentioned 0.1 M (R) was added.
  • 6b (51.1 g, 0.129 mol) and 7 M ammonia methanol solution (250 mL) were added to the reaction flask, and stirred at room temperature for 1 hour. After suction filtration, the mixture was washed with methanol (100 mL ⁇ 2), and the filter cake was collected and dried to give 6c (30 g, yield: 64.2%).
  • Example 2 1-[4-[4-(3-Chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-1 -piperidinyl]propyl-2-en-1-one (Compound 2, Compound 2 is 1-[(3R,4R)-4-[4-(3-chloro-2,4-difluoro-phenylamine) -7-methoxy-quinazolin-6-yl]oxy-3-fluoro-1-piperidinyl]propyl-2-en-1-one and 1-[(3S,4S)-4- [4-(3-Chloro-2,4-difluoro-aniline)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-1-piperidinyl]propyl-2- Mixture of ene-1-one)
  • 2C is N-(3-chloro-2,4-dichloro-phenyl)-6-[[(3R,4R)-3-fluoro-4-piperidine Alkyl]-7-methoxy-quinazolin-4-amine bis(trifluoroacetic acid) and N-(3-chloro-2,4-dichloro-phenyl)-6-[[(3S) , 4S)-3-fluoro-4-piperidinyloxy]-7-methoxy-quinazolin-4-amine bis(trifluoroacetic acid) mixture)
  • 2C is N-(3-chloro-2,4-dichloro-phenyl)-6-[[(3R,4R)-3-fluoro-4-piperidinyl]oxy]-7-methoxy- Quinazoline-4-amine bis(trifluoroacetic acid) and N-(3-chloro-2,4-dichloro-phenyl)-6-[[(3S,4S)-3-fluoro-4-piperidine A mixture of oxy]-7-methoxy-quinazolin-4-amine bis(trifluoroacetic acid).
  • Compound 2 is 1-[(3R,4R)-4-[4-(3-chloro-2,4-difluoro-aniline)-7-methoxy-quinazolin-6-yl]oxy-3 -fluoro-1-piperidinyl]propyl-2-en-1-one and 1-[(3S,4S)-4-[4-(3-chloro-2,4-difluoro-aniline)-7 a mixture of -methoxy-quinazolin-6-yl]oxy-3-fluoro-1-piperidinyl]propyl-2-en-1-one.
  • Example 3 1-[4-[4-(3-Chloro-2,4-difluoro-aniline)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-1- Piperidinyl]propyl-2-en-1-one (Compound 3, Compound 3 is 1-[(3R,4S)-4-[4-(3-chloro-2,4-difluoro-aniline)- 7-Methoxy-quinazolin-6-yl]oxy-3-fluoro-1-piperidinyl]propyl-2-en-1-one and 1-[(3S,4R)-4-[ 4-(3-Chloro-2,4-difluoro-aniline)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-1-piperidinyl]propyl-2-ene a mixture of -1-ketone)
  • 3C (0.65g, 0.10mmol) was dissolved in a mixture of tetrahydrofuran (25mL) and water (4mL), sodium hydrogencarbonate (0.41g, 4.80mmol) was added, and acryloyl chloride (0.10) was added dropwise to 0 °C in an ice bath. g, 0.11 mmol) in tetrahydrofuran (1.5 mL), EtOAc (EtOAc) (EtOAc)EtOAc. The mixture was dried, filtered, evaporated, evaporated,jjjjjjjjjjjjjj :99.70%).
  • 4,5-Difluoro-2-nitro-benzoic acid 4A 25 g, 123 mmol
  • tetrahydrofuran 400 mL
  • benzyl alcohol 29.3 g, 270.6 mmol
  • a 2 M solution of bis(trimethylsilyl)amide in tetrahydrofuran 197 mL, 394 mmol was added dropwise at 0 ° C, and the mixture was reacted at 0 ° C for 1 hour.
  • Step 7 6-Benzyloxy-N-(3-chloro-2,4-difluoro-phenyl)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazoline-4- Ammonia (4H)
  • Step 8 4-(3-Chloro-2,4-difluoro-aniline)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazolin-6-ol (4I)
  • Step 9 4-[4-(3-Chloro-2,4-difluoro-aniline)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazolin-6-yl]oxypipe Butyl-1-carboxylic acid tert-butyl ester (4J)
  • Step 10 N-(3-Chloro-2,4-difluoro-phenyl)-6-(4-piperidinyloxy-7-[(3S)-tetrahydrofuran-3-yl]oxyquinazole Porphyrin-4-amine (4K)
  • Example 5 1-[(3aR,6aS)-5-[4-(3-Chloro-2,4-difluoro-aniline)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quin Oxazolin-6-yl]oxy-3,3a,4,5,6,6a-hexahydro-1H-cyclopenta[c]pyrrol-2-yl]propyl-2-en-1-one (Compound 5)
  • Second step 6-[[(3aR,6aS)-1,2,3,3a,4,5,6,6a-octahydrocyclopenta[c]pyrrole-5-yl]oxy]-N -(3-chloro-2,4-difluoro-phenyl)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazolin-4-ammonium (5C)
  • Example 6 1-[(3aR,6aS)-5-[4-[(3-Chloro-2,4-difluoro-phenyl)amino]-7-methoxy-quinazolin-6-yl Oxy-hexahydrocyclopenta[c]pyrrole-2(1H)-yl]propyl-2-en-1-one (compound 6)
  • Example 7 1-[(3R,4R)-4-[4-(3-Chloro-2,4-difluoro-aniline)-7-methoxy-quinazolin-6-yl]oxy- 3-fluoro-1-piperidinyl]propyl-2-en-1-one maleate (Compound 7)
  • Example 8 1-[(3R,4S)-4-[4-(3-Chloro-2,4-difluoro-aniline)-7-methoxy-quinazolin-6-yl]oxy- 3-fluoro-1-piperidinyl]propyl-2-en-1-one maleate (Compound 8)
  • Example 10 1-[(3R,4S)-4-[4-(3-Chloro-2,4-difluoro-aniline)-7-methoxy-quinazolin-6-yl]oxy- 3-fluoro-1-piperidinyl]propyl-2-en-1-one hydrochloride (Compound 10)
  • the fourth step 7-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)tetrahydro-1H- Azole [3,4-a]pyridine-3(5H)-one (compound 15)
  • the fourth step 7-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)tetrahydro-1H- Azole [3,4-a]pyridine-3(5H)-thione (compound 16)
  • the SRB experiment is to detect the proliferation of cells under the action of drugs. After 72 hours of dosing, the proliferation of the cells was reflected by SRB staining and a 490 nm reading of the microplate reader.
  • the cells were plated in 96-well plates, 8000 cells per H1975 cell line, 10,000 cells per A431 cell line, incubator at 37 ° C, and 5% CO 2 overnight. Before dosing, 6 wells of each cell were fixed by adding 30 ⁇ l of 50% trichloroacetic acid.
  • the compound was formulated as a solution in DMSO at a maximum concentration of 10 ⁇ M, diluted 10 times in a total of ten gradients, and the test compound was diluted with a medium containing 0.1% FBS to a final concentration of 2 times.
  • the compounds of the present invention have a significant inhibitory effect on the proliferation of H1975 (L858R/T790M) and A431 (EGFRwt amplification) cells.
  • mice Male Sprague-Dawley rats (purchased from Vitallihua Experimental Animal Co., Ltd.) 180-240 g, fasting water supply overnight, 3 rats orally administered with 5 mg/kg, and 3 rats intravenously injected with 1 mg/kg.
  • the compound was formulated into a suspension of 0.5 mg ⁇ mL -1 in a 0.5% methylcellulose (MC) solution (containing 0.4% Tween 80), before administration and 30 minutes after administration. Blood was collected at 1, 2, 4, 6, 8, 12, and 24 hours; in the intravenous administration group, the compound was formulated with 10% DMA, 20% Solutol HS-15 (30%, w/v), and 70% physiological saline.
  • a solution of mg x mL -1 was taken before administration and at 5, 15 and 30 minutes and 1, 2, 4, 8, 12 and 24 hours after administration. Heparin anticoagulation.
  • the blood sample was centrifuged at 5,500 rpm for 10 minutes, and plasma was collected and stored at -80 °C.
  • Take 10 ⁇ L of rat plasma at each time point add 500 ⁇ L of acetonitrile solution containing internal standard, mix, vortex for 4 minutes, centrifuge at 3700 rpm for 18 minutes, take 70 ⁇ L of the supernatant and mix with 70 ⁇ L of water, and take 5 ⁇ L of the mixture for LC- MS/MS analysis.
  • the main pharmacokinetic parameters were analyzed by WinNonlin 6.3 software non-compartmental model, and the results of pharmacokinetic evaluation were shown in Table 2.
  • NCI-H1975 cells After 3 days of adaptation in the laboratory environment of nude mice, the right flank was subcutaneously inoculated with H1975 cells 5 ⁇ 10 6 /piece. Tumors were grown to 200-250 mm 3 and randomized into groups of 10 mice each.
  • A431 cells After 3 days of adaptation in the laboratory environment of nude mice, A431 cells were inoculated subcutaneously into the right flank by 5 ⁇ 10 6 /only. Tumors were grown to 200-250 mm 3 and randomized into groups of 10 mice each.
  • NCI-H1975 cell xenograft nude mice The test compound was formulated into two doses of 0.15 mg/ml and 0.5 mg/ml with PEG400/Tween80. Animals were weighed and administered with test compound 1.5 mg/kg and 5 mg/kg, respectively; Afatinib was formulated with PEG400/Tween80 to make 1.2 mg/ml and 4 mg/ml. Animals were weighed and administered with test compounds 12 mg/kg and 40 mg/kg, respectively. The blank control group was administered the same volume of drug-free PEG400/Tween80 solution. The drug was administered once a day for 10 consecutive days.
  • A431 cell transplanted tumor nude mice The test compound was formulated into PEG400/Tween80 at 0.03 mg/ml and 0.1 mg/ml. Animals were weighed and administered with test compound 0.3 mg/kg and 1 mg/kg, respectively; Afatinib was formulated with PEG400/Tween80 to prepare 0.3 mg/ml and 1 mg/ml. Animals were weighed and administered with test compound 3 mg/kg and 10 mg/kg blank control group, respectively, and the same volume of drug-free PEG400/Tween80 solution was administered. The drug was administered once a day for 10 consecutive days.
  • the tumor volume was measured once a week with vernier calipers (calculated formula), and the body weight was weighed.
  • V 1/2 ⁇ long diameter ⁇ short diameter ⁇ short diameter
  • Relative volume (RTV) VT / V0
  • Tumor inhibition rate (%) (CRTV-TRTV) / CRTV ⁇ 100%)
  • V0 and VT are the tumor volume before the start of administration and after the last administration, respectively.
  • CRTV and TRTV were the relative tumor volumes of the blank control group and the experimental group after the last administration. The results are shown in Tables 3 and 4.
  • the compound of the present invention can significantly inhibit the growth of human lung cancer NCI-H1975 containing human epithelial cell carcinoma A431 and EGFR T790M mutations of wild type EGFR, and the doses are only positive control. At the doses of /10 and 1/8, the effect on transplanted tumors of A431 and H1975 was significantly stronger than that of the control drugs.
  • the cells used in this assay were CHO cell lines transfected with hERG cDNA and stably expressing hERG channels.
  • a manual patch clamp system German HEKA EPC-10 signal amplifier and digital conversion system
  • a circular slide with CHO hERG cells grown on it was placed in an electrophysiology recording trough under an inverted microscope.
  • the extracellular fluid was continuously perfused in the recording tank (about 1 ml per minute).
  • the experimental procedure uses conventional whole-cell patch clamp current recording techniques. The cells were clamped at a voltage of -80 mV.
  • the cell clamp voltage was depolarized to +20 mV to activate the hERG potassium channel, and after 5 seconds it was clamped to -50 mV to eliminate inactivation and generate tail current.
  • the tail current peak is used as the value of the hERG current magnitude.
  • Test compounds were tested at concentrations of 30, 10, 3, 1, 0.3, 0.1 and 0.03 ⁇ M for testing. Prior to testing, the stock solutions were first diluted with DMSO to 30, 10, 3, 1, 0.3 and 0.1 mM and diluted to the final ⁇ M concentration with extracellular fluid. The final concentration of DMSO in each concentration of the compound solution was 0.1%. The positive control Cisapride (cisapride) was tested at a concentration of 0.1 [mu]M.
  • test data was analyzed by HEKA Patchmaster, Microsoft Excel and data analysis software provided by Graphpad Prism. The results are shown in Table 5.
  • the compounds of the present invention have weak inhibition on hERG potassium channel and have high safety, especially compounds 9 and 13, which have weaker inhibition on hERG potassium channel and higher safety.

Landscapes

  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

本发明涉及一种喹唑啉衍生物及其制备方法和在医药上的应用,具体而言本发明涉及喹唑啉衍生物或者其立体异构体、水合物、代谢产物、溶剂化物、药学上可接受的盐、共晶或者前药,涉及它们的制备方法,涉及包括其药物组合物以及本发明的化合物药物组合物在医药上的用途,特别作为EGFR/HER2双靶点抑制剂的用途。

Description

喹唑啉衍生物及其制备方法和在医药上的应用 技术领域
本发明涉及一种喹唑啉衍生物及其制备方法和在医药上的应用,具体是一种具有EGFR/HER2双靶点抑制作用的新颖喹唑啉衍生物或其立体异构体、水合物、溶剂化物、代谢产物、药学上可接受的盐、共晶或前药、其药物组合物以及其在医药上的应用。
背景技术
细胞表面受体中的受体酪氨酸激酶超家族通过细胞外生长因子对细胞信号的调节起重要的作用。受体酪氨酸激酶能够催化磷酸基团从ATP转移至底物的酪氨酸基团上。当没有配体激活受体酪氨酸激酶时,这些激酶处于未磷酸化的单体状态,其激酶域呈非活性的结构。当配体与受体酪氨酸激酶的胞外段结合时,受体发生寡聚化,并且自磷酸化,增加激酶的催化活性的同时形成了信号蛋白的结合位点,信号蛋白与其结合,从而激活多条信号通路。这些信号通路相互联系,调控细胞的增殖、生存、分化、功能、迁移和凋亡。当受体酪氨酸激酶失去调控,异常激活时,细胞会发生转化成肿瘤细胞,增殖、生长能力和耐药能力提高,具有较强的成血管能力、侵袭力和转移能力(Yarden和Sliwkowski,2001,Nat Rev Mol Cell Biol,2,127-137)。
表皮生长因子(EGF)受体是一类备受关注的受体酪氨酸酶。EGF受体家族包含4个成员:EGFR/ErbB1/HER1、ErbB2/HER2/Neu、ErbB3/HER3和ErbB4/HER4。正常的EGFR激活的信号通路能够调节细胞的增殖、成血管、生长、迁移和粘附,在器官发生过程或成体的细胞与细胞间的相互作用中起着重要作用(Wu,Zhong等,2007,J Thorac Oncol,2,430-439)。许多分子时间能够导致EGFR激酶活性的持续激活,不断的触发大量的下游信号转导通路,包括K-ras激活。因此EGFR家族与成瘤密切相关。EGFR的过表达或突变在头颈癌、卵巢癌、膀胱癌、宫颈癌、食道癌、胃癌、乳腺癌、内膜癌、结肠癌、肺癌和脑瘤等多种癌症中都能检测到,通常预示这不良的预后。另外,HER2被认为是一种较强的肿瘤蛋白,其突变也在多种肿瘤中可见,尤其是乳腺癌、肺癌和结肠癌等(Olayioye,Neve等,2000,EMBO J,19,3159-3167)。
目前,已开发出多种EGFR酪氨酸激酶小分子抑制剂,例如第一代的吉非替尼和厄罗替尼,能够选择性地可逆地抑制EGFR,从而阻止肿瘤的生长,尤其是含有外显子19缺失(如del747-750)和外显子21突变(如L858R)的肿瘤(Barker,Gibson等,2001,Bioorg Med  Chem Lett,11,1911-1914;Wakeling,Guy等,2002,Cancer Res,62,5749-5754)。然而,很多使用吉非替尼和厄罗替尼的患者会出现获得性耐药,这种获得性耐药有50%-60%是由于外显子20发生的T790M突变造成的。T790M突变会影响激酶催化域,减弱靶向抑制剂与靶点的相互作用,使治疗无效(Pao和Chmielecki,2010,Nat Rev Cancer,10,760-774)。
研究发现,不可逆抑制剂能够与激酶的活性位点中起决定性作用的半胱氨酸残基形成共价结合,成为一种有潜力的抑制T790M突变的策略。而在EGFR家族中,仅EGFR和HER2在相应的位点具有半胱氨酸。因此这种不可逆抑制剂对EGFR和HER2具有高度的特异性(Singh,Dobrusin等,1997,J Med Chem,40,1130-1135;Fry,Bridges等,1998,Proc Natl Acad Sci U S A,95,12022-12027)。已开发出许多不可逆的双抑制剂(如Afatinib、来那替尼和卡拉替尼等),相较于可逆抑制剂,这些药物不但能在低浓度下有效抑制EGFR敏感突变(如del747-750、L858R),而且能够有效克服T790M造成的耐药。值得注意的是,在临床前研究中,不可逆的双抑制剂不像吉非替尼和厄罗替尼产生快速的耐药性(Ercan,Zejnullahu等,2010,Oncogene,29,2346-2356;Chmielecki,Foo等,2011,Sci Transl Med,3,90ra59)。
为了满足临床需求,需要继续研发在能够有效克服T790M突变产生的耐药且安全的EGFR/HER2双靶点抑制剂。
CN10679384描述了用于抑制癌细胞生长的新酰胺衍生物,其结构式如下:
Figure PCTCN2015076449-appb-000001
其中,A是
Figure PCTCN2015076449-appb-000002
R4、R5、R6和R7各自独立为氢、卤素、N-C1-6烷基或N-羟基酰氨基或者C-C1-6烷基反酰氨基(-NHCOC1-6)、羟基羰基(-COOH)、C1-6烷氧基羰基(-COOC1-6)、C1-6烷基,或者为被羟基、C1-6二烷基胺或杂环基团取代的烷基;R1是被1-5个X取代的芳基或杂环基团,或者是被芳基取代的C1-6烷基;R2是氢、羟基、C1-6烷氧基,或者是被C1-6烷氧基或杂环基团取代的C1-6烷氧基;R3是氢、-COOH、C1-6烷氧基羰基,或者是N-未取代的酰氨基或者N-被取代的酰氨基;na和nb各自为0-6的整数;X是氢、卤素、羟基、氰基、硝基、(一卤代、二卤代或三卤代)甲基、巯基、C1-6烷硫基、丙烯酰氨基、C1-6烷基、C1-6烯基、C1-6炔基、C1-6烷氧基、芳氧基、C1-6二烷基氨基,或者为被Z取代的C1-6烷基或C1-6烷氧基,条件是当X的数量为两个或更多个时,X基团可以稠合在一起形成环结构;Y是羟基、C1-6烷基,或者是被Z取代的C1-6烷基,所述C1-6烷基含有1-4 个选自N、O、S、SO和SO2的基团;并且Z是C1-6烷基、芳基或杂环基团,所述芳基基团为C5-12单环或双环芳族基团,所述杂环基团为含有1-4个选自N、O、S、SO和SO2的基团的C5-12单环或双环的芳族或非芳族基团,并且所述芳基和杂环基团是未取代的,或者被选自卤素、羟基、氨基、硝基、氰基、C1-6烷基、C1-6烯基、C1-6炔基、C1-6烷氧基、C1-6单烷基氨基和C1-6二烷基氨基的取代基取代。不认为此专利中具体描述是本发明的一部分。
CN102731485描述了4-(取代苯氨基)喹唑啉衍生物,其结构式如下:
Figure PCTCN2015076449-appb-000003
R1选自
Figure PCTCN2015076449-appb-000004
Figure PCTCN2015076449-appb-000005
R2选自氢或N,N-二甲氨基甲基;R3选自甲氧乙基、四氢呋喃-3-基、(S)-四氢呋喃-3-基或(R)-四氢呋喃-3-基,以及各R4和R5独立地选自一个或多个氢、卤素、C1-C10烷基、C1-C10烷氧基和卤代C1-C10烷基;当且仅当R2为氢,R3为甲氧乙基时,R1不为
Figure PCTCN2015076449-appb-000006
当且仅当R2为N,N-二甲氨基甲基,R3为四氢呋喃-3-基时,R1不为
Figure PCTCN2015076449-appb-000007
不认为此专利中具体描述是本发明的一部分。
WO2011029265描述了喹唑啉衍生物,其结构式如下:
Figure PCTCN2015076449-appb-000008
其中,A选自碳原子或氮原子;当A为碳原子时,R1选自氢原子或烷氧基,其中所述的烷氧基任选进一步被一个或多个选自卤素或烷氧基的取代基所取代,R2选自氰基;当A为氮原子时,R1选自氢原子或烷氧基,其中所述的烷氧基任选进一步被一个或多个选自卤素或烷氧基的取代基所取代,R2无取代;R3具有下列结构:-D-T-L或-D;D选自芳基或杂芳基,其中所述的芳基或杂芳基各自独立地任选进一步被一个或多个选自卤素、烷基或三 氟甲基的取代基所取代;T选自-(CH2)r-、-O(CH2)r-、-NH(CH2)r-或-S(O)r(CH2)r-;L选自芳基或杂芳基,其中所述的芳基或杂芳基各自独立地任选进一步被一个或多个卤素或烷基所取代;R4和R5各自独立地选自氢原子、烷基、烷氧基、羟基、羟烷基、卤素、羰基、氨基、氰基、硝基、羧酸或羧酸酯;B选自碳原子、氧原子或S(O)r基团;当B为碳原子时,R6和R7各自独立地选自氢原子、烷基、烷氧基、羟基、羟烷基、卤素、羰基、氨基、氰基、硝基、羧酸或羧酸酯;当B为氧原子或S(O)r基团时,R6和R7无取代;R8选自氢原子或烷基;R9选自氢原子、烷基、芳基、羧基或羧酸酯;r为0、1或2;且n为1、2、3、4或5。不认为此专利中具体描述是本发明的一部分。
发明内容
本发明提供一类结构新颖的化合物,经过研究表明,本发明化合物作为EGFR/HER2双靶点抑制剂显示出优异的药效活性。
本发明提供了一种通式(II0)所示的化合物或其立体异构体、水合物、代谢产物、溶剂化物、药学上可接受的盐、共晶或前药,
Figure PCTCN2015076449-appb-000009
其中:
R2选:自C1-3烷基或者4至8元杂环基,所述的杂环基任选进一步被0至2个选自F、Cl、羟基、C1-3烷基或者C1-3烷氧基的取代基所取代,且所述的杂环基含有1至2个选自N或者O的杂原子;
环A选自
Figure PCTCN2015076449-appb-000010
R3选自F、CF3、CHF2、CH2F、Cl、羟基或者C1-3烷基;
X1
Figure PCTCN2015076449-appb-000011
或者X1与环A一起形成
Figure PCTCN2015076449-appb-000012
R4、R5和R6各自为H;
R9选自F、Cl、羟基、C1-3烷基或者C2-3炔基;
n、m、t各自选自0、1、2、3或者4;
限制条件是,当n为0,R2选自C1-3烷基,且m为0时,环A不为
Figure PCTCN2015076449-appb-000013
优选地,本发明的化合物选自通式(II)所示的化合物:
Figure PCTCN2015076449-appb-000014
其中:
R2选自甲基、乙基或者
Figure PCTCN2015076449-appb-000015
环A选自
Figure PCTCN2015076449-appb-000016
n为0或1;
R3选自F;
m为0或1;
R4、R5和R6均为H;
R9选自F或者Cl;
t为3;
限制条件是,当n为0,R2选自甲基或乙基,且m为0时,环A不为
Figure PCTCN2015076449-appb-000017
具体而言,本发明涉及的化合物或其立体异构体、水合物、代谢产物、溶剂化物、药学上可接受的盐、共晶或前药选自,但不限于:
Figure PCTCN2015076449-appb-000018
Figure PCTCN2015076449-appb-000019
根据本发明的具体实施方案,本发明的化合物或其立体异构体、水合物、代谢产物、溶剂化物、药学上可接受的盐、共晶或前药,其中所述的盐选自盐酸盐、氢溴酸盐、硫酸盐、磷酸盐、醋酸盐、三氟乙酸盐、硫氰酸盐、马来酸盐、羟基马来酸盐、戊二酸盐、甲磺酸盐、乙磺酸盐、苯磺酸盐、对甲苯磺酸盐、苯甲酸盐、水杨酸盐、苯乙酸盐、肉桂酸盐、乳酸盐、丙二酸盐、特戊酸盐、琥珀酸盐、富马酸盐、苹果酸盐、扁桃酸盐、酒石酸 盐、没食子酸盐、葡萄糖酸盐、月桂酸盐、棕榈酸盐、果胶酸盐、苦味酸盐、柠檬酸盐或者它们的组合,优选的,所述的盐选自盐酸盐、氢溴酸盐、硫酸盐、磷酸盐、醋酸盐、马来酸盐、甲磺酸盐、苯磺酸盐、三氟乙酸盐、对甲苯磺酸盐、苯甲酸盐、水杨酸盐、肉桂酸盐、乳酸盐、丙二酸盐、琥珀酸盐、富马酸盐、苹果酸盐、酒石酸盐、柠檬酸盐或者它们的组合,更优选盐酸盐和马来酸盐。
本发明还提供了一种药物组合物,所述的组合物包括:有效剂量的本发明所述的化合物或其立体异构体、水合物、代谢产物、溶剂化物、药学上可接受的盐、共晶或前药,以及药学上可接受的载体、稀释剂、佐剂、媒介物或赋形剂。
根据本发明的具体实施方案,本发明的药物组合物还进一步包括一种或多种其他治疗剂。
根据本发明的具体实施方案,本发明的药物组合物中所述的其他治疗剂包括:顺铂(cisplatin),卡铂(carboplatin),奥沙利铂(oxaliplatin),达卡巴嗪(dacarbazine),替莫唑胺(temozolomide),丙卡巴肼(procarbazine),甲氨蝶呤(methotrexate),氟尿嘧啶(fluorouracil),阿糖胞苷(cytarabine),吉西他滨(gemcitabine),巯基嘌呤(mercaptopurine),氟达拉滨(fludarabine),长春碱(vinblastine),长春新碱(vincristine),长春瑞滨(vinorelbine),紫杉醇(paclitaxel),多西紫杉醇(docetaxel),拓扑替康(topotecan),伊立替康(irinotecan),依托泊苷(etoposide),曲贝替定(trabectedin),更生霉素(dactinomycin),多柔比星(doxorubicin),表柔比星(epirubicin),道诺霉素(daunorubicin),米托蒽醌(mitoxantrone),博来霉素(bleomycin),丝裂霉素C(mitomycin),伊沙匹隆(ixabepilone),他莫昔芬(tamoxifen),氟他胺(flutamide),西罗莫司(sirolimus),Afatinib(afatinib),alisertib,amuvatinib,阿帕替尼(apatinib),阿西替尼(axitinib),硼替佐米(bortezomib),波舒替尼(bosutinib),布立尼布(brivanib),卡博替尼(cabozantinib),西地尼布(cediranib),crenolanib,克卓替尼(crizotinib),dabrafenib(达拉非尼),dacomitinib,达鲁舍替(danusertib),达沙替尼(dasatinib),多韦替尼(dovitinib),厄洛替尼(erlotinib),foretinib,ganetespib,gefitinib(吉非替尼),依鲁替尼(ibrutinib),埃克替尼(icotinib),伊马替尼(imatinib),iniparib,拉帕替尼(lapatinib),lenvatinib,linifanib,linsitinib,马赛替尼(masitinib),momelotinib,莫替沙尼(motesanib),来那替尼(neratinib),尼罗替尼(nilotinib),niraparib,oprozomib,olaparib,帕唑帕尼(pazopanib),pictilisib,ponatinib,quizartinib,regorafenib,rigosertib,rucaparib,ruxolitinib,塞卡替尼(saracatinib),saridegib,索拉非尼(sorafenib),舒尼替尼(sunitinib),tasocitinib,telatinib,tivantinib,tivozanib,tofacitinib,trametinib,凡德他尼(vandetanib),veliparib,威罗菲尼(vemurafenib),vismodegib,volasertib,阿仑单抗(alemtuzumab),贝伐单抗(bevacizumab),brentuximab vedotin,卡妥索单抗 (catumaxomab),西妥昔单抗(cetuximab),地诺单抗(denosumab),吉妥珠单抗(gemtuzumab),伊匹单抗(ipilimumab),尼妥珠单抗(nimotuzumab),奥法木单抗(ofatumumab),帕尼单抗(panitumumab),利妥昔单抗(rituximab),托西莫单抗(tositumomab),曲妥珠单抗(trastuzumab)或它们的组合。
本发明还提供了所述的化合物或其立体异构体、水合物、酯、代谢产物、溶剂化物、药学上可接受的盐、共晶或前药或者所述的药物组合物在作为一种EGFR/HER2受体酪氨酸激酶抑制剂在制备药物制剂的应用,特别是在用于制备用于治疗和/或预防过度增殖性疾病的药物制剂中的应用。
根据本发明的具体实施方案,本发明的化合物或其立体异构体、水合物、酯、代谢产物、溶剂化物、药学上可接受的盐、共晶或前药或者所述的药物组合物的应用中,所述过度增殖性疾病包括脑瘤、非小细胞肺癌、表皮鳞癌、膀胱癌、胰腺癌、结肠癌、乳腺癌、卵巢癌、子宫颈癌、子宫内膜癌、结直肠癌、肾癌、食管腺癌、食管鳞状细胞癌、实体瘤、非霍奇金淋巴瘤、肝癌、肺癌,胃癌、皮肤癌、甲状腺癌、头颈癌、前列腺癌、神经胶质瘤和鼻咽癌中的一种或多种,优选非小细胞肺癌、乳腺癌、表皮鳞癌、胃癌和结肠癌中的一种或多种。
发明的详细说明
除非有相反的陈述,在说明书和权利要求书中使用的术语具有下述含义。
本发明所述基团和化合物中所涉及的碳、氢、氧、硫、氮或卤素均包括它们的同位素情况,及本发明所述基团和化合物中所涉及的碳、氢、氧、硫或氮任选进一步被一个或多个它们对应的同位素所替代,其中碳的同位素包括12C、13C和14C,氢的同位素包括氕(H)、氘(D,又叫重氢)、氚(T,又叫超重氢),氧的同位素包括16O、17O和18O,硫的同位素包括32S、33S、34S和36S,氮的同位素包括14N和15N,氟的同位素包括17F和19F,氯的同位素包括35Cl和37Cl,溴的同位素包括79Br和81Br。
“药学上可接受的盐”或者“其药学上可接受的盐”是指本发明化合物保持游离碱的生物有效性和特性,且所述的游离碱通过与无毒的无机酸或者有机酸反应获得的盐。所述的无机酸的非限制性实施例包括氢氟酸、盐酸、氢溴酸盐、硫酸盐、磷酸、次氯酸、高氯酸、碘酸、碳酸、亚硝酸、次硝酸、偏硼酸、硼酸、偏硅酸、硅酸、偏亚磷酸、偏磷酸、焦磷酸、氢硫酸、亚硫酸、硫代硫酸和高锰酸;所述的有机酸非限制性实施例包括甲酸、醋酸、三氟乙酸、硫氰酸、马来酸、羟基马来酸、戊二酸、甲磺酸、乙磺酸、苯磺酸、对甲苯磺 酸、苯甲酸、水杨酸、苯乙酸、肉桂酸、乳酸、丙二酸、特戊酸、琥珀酸、富马酸、苹果酸、扁桃酸、酒石酸、没食子酸、葡萄糖酸、月桂酸、棕榈酸、果胶酸、苦味酸和柠檬酸。
“载体”是指不会对生物体产生明显刺激且不会消除所给予化合物的生物活性和特性的材料。
“赋形剂”是指加入到药物组合物中以促进化合物给药的惰性物质。非限制性实施例包括碳酸钙、磷酸钙、糖、淀粉、纤维素衍生物(包括微晶纤维素)、明胶、植物油、聚乙二醇类、稀释剂、成粒剂、润滑剂、粘合剂和崩解剂。
“佐剂”是非特异性免疫增强剂,当与抗原一起注射或预先注入机体时,可增强机体对抗原的免疫应答或改变免疫应答类型。
“稀释剂”也叫“填充剂”。把原药加工成粉剂时,或为了使其便于喷施所加入的进行稀释的惰性物质。如:粘土、高岭土、陶土、滑石粉等。
“前药”是指可经体内代谢转化为具有生物活性的本发明化合物,这样的转化受前体药物在血液中水解或在血液组织中经酶转化为母体结构的影响。本发明的前药通过修饰本发明化合物中的功能基团来制备,该修饰可以通过常规的操作或者在体内被除去,而得到母体化合物。
“共晶”是指活性药物成分和共晶形成物在氢键或其他非共价键的作用下结合而成的晶体,其中API和CCF的纯态在室温下均为固体,并且各组分间存在固定的化学计量比。共晶是一种多组分晶体,既包含两种中性固体之间形成的二元共晶,也包含中性固体与或溶剂化物形成的多元共晶。所述“共晶形成物”的非限定性实例包括丙氨酸、缬氨酸、亮氨酸、异亮氨酸、脯氨酸、苯丙氨酸、色氨酸、蛋氨酸、甘氨酸、丝氨酸、苏氨酸、半胱氨酸、酪氨酸、天冬酰胺、谷氨酰胺、赖氨酸、精氨酸、组氨酸、天冬氨酸、门冬氨酸、谷氨酸、焦谷氨酸、硫酸、磷酸、硝酸、氢溴酸、酸、甲酸、乙酸、丙酸、苯磺酸、苯甲酸、苯乙酸、水杨酸、褐藻酸、氨茴酸、樟脑酸、柠檬酸、乙烯磺酸、蚁酸、富马酸、糠酸、葡萄糖酸、葡萄糖醛酸、谷氨酸、乙醇酸、羟乙磺酸、乳酸、马来酸、苹果酸、扁桃酸、粘液酸、双羟萘酸、泛酸、硬脂酸、琥珀酸、磺胺酸、酒石酸、对甲苯磺酸、丙二酸、2-羟基丙酸、草酸、羟乙酸、葡萄糖醛酸、半乳糖醛酸、枸橼酸、肉桂酸、对甲苯磺酸、甲磺酸、乙磺酸或三氟甲磺酸、氨、异丙基胺、三甲基胺、二乙胺、三乙胺、三丙基胺、二乙醇胺、乙醇胺、二甲基乙醇胺、2-二甲基氨基乙醇、2-二乙基氨基乙醇、二环己基胺、咖啡碱、普鲁卡因、胆碱、甜菜碱、苯明青霉素、乙二胺、葡萄糖胺、甲基葡糖胺、可可碱、三乙醇胺、氨丁三醇、嘌呤、哌嗪、哌啶和N-乙基哌啶。
“立体异构体”是指由分子中原子在空间上排列方式不同所产生的异构体,包括顺反异构体、对映异构体和构象异构体。
“任选”或“任选地”或“选择性的”或“选择性地”是指随后所述的事件或状况可以但未必发生,该描述包括其中发生该事件或状况的情况及其中未发生的情况。例如,“选择性地被烷基取代的杂环基”是指该烷基可以但未必存在,该描述包括其中杂环基被烷基取代的情况,及其中杂环基未被烷基取代的情况。
具体实施方式
以下通过具体实施例详细说明本发明的实施过程和产生的有益效果,旨在帮助阅读者更好地理解本发明的实质和特点,不作为对本案可实施范围的限定。
化合物的结构是通过核磁共振(NMR)或(和)质谱(MS)来确定的。NMR的测定是用(Bruker Avance III 400和Bruker Avance 300)核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6),氘代氯仿(CDCl3),氘代甲醇(CD3OD),内标为四甲基硅烷(TMS)。
MS的测定用(Agilent 6120B(ESI)和Agilent 6120B(APCI))。
HPLC的测定使用安捷伦1260DAD高压液相色谱仪(Zorbax SB-C18100×4.6mm)。
薄层层析硅胶板使用烟台黄海HSGF254或青岛GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm~0.20mm,薄层层析分离纯化产品采用的规格是0.4mm~0.5mm。
柱层析一般使用烟台黄海硅胶200~300目硅胶为载体。
本发明的己知的起始原料可以采用或按照本领域已知的方法来合成,或可购买于泰坦科技、安耐吉化学、上海德默、成都科龙化工、韶远化学科技、百灵威科技等公司。
氮气氛是指反应瓶连接一个约1L容积的氮气气球。
氢气氛是指反应瓶连接一个约1L容积的氢气气球。
氢化反应通常抽真空,充入氢气,反复操作3次。
实施例中无特殊说明,反应在氮气氛下进行。
实施例中无特殊说明,溶液是指水溶液。
实施例中无特殊说明,反应的温度为室温。
室温为最适宜的反应温度,为20℃~30℃。
本文所用的其他符号具有下列意义:
Bn:苄基;Me:甲基;Et:乙基;Ts:对甲苯磺酰基;TBS:叔丁基二甲基硅基;Boc: 叔丁氧羰基;Ac:乙酰基。
中间体1:4-氯-7-甲氧基-喹唑啉-6-醇(1d)
4-chloro-7-methoxy-quinazolin-6-ol
Figure PCTCN2015076449-appb-000020
第一步:(4-羟基-7-甲氧基-喹唑啉-6-基)乙酸酯(1b)
(4-hydroxy-7-methoxy-quinazolin-6-yl)acetate
向反应瓶中加入6-羟基-7-甲氧基-3H-喹唑啉-4-酮1a(40g,208mmol,康满林)和醋酸酐(296mL,211mmol),搅拌均匀后,加入吡啶(67mL),升至100℃搅拌4小时后冷却至室温。将反应液倒入冰水(500g)中,搅拌5分钟后,减压抽滤,依次用水(100mL×2)、石油醚(100mL×2)洗涤滤饼,收集滤饼,烘干得到灰色固体状的1b(36g,产率:73%)。
第二步:(4-氯-7-甲氧基-喹唑啉-6-基)乙酸酯盐酸盐(1c)
(4-chloro-7-methoxy-quinazolin-6-yl)acetate hydrochloride
向反应瓶中加入1b(30g,130mmol)和二氯亚砜(205mL,128mmol),缓慢滴加三氯氧磷(38mL,126mmol)和N,N-二甲基甲酰胺(3mL),搅拌均匀后,升至120℃搅拌4小时,冷却至室温,减压浓缩得残留物。将甲苯加入残留物种,减压浓缩旋除溶剂,重复此步骤两次,减压浓缩得到棕褐色固体状的1c粗品(289g),直接用于下步反应。
第三步:4-氯-7-甲氧基-喹唑啉-6-醇(1d)
4-chloro-7-methoxy-quinazolin-6-ol
向反应瓶中加入1c(325.5g,100mmol)和氨甲醇溶液(7mol/L,560mL),室温搅拌1小时。抽滤,用甲醇(100mL×2)洗涤,收集滤饼,干燥得到灰色固体状的1d(17g,产率:65.4%)。
中间体2:(3S)-3-氟-4-羟基-哌啶-1-甲酸叔丁酯(2b)
tert-butyl(3S)-3-fluoro-4-hydroxy-piperidine-1-carboxylate
Figure PCTCN2015076449-appb-000021
向反应瓶中加入3-氟-4-氧代哌啶-1-甲酸叔丁酯2a(20g,92mmol)和甲醇(70mL),搅拌均匀后,冷却至0℃,分批加入硼氢化钠(7g,184mmol),升至室温反应1小时。减压浓缩,向残留物中加入乙酸乙酯(200mL),饱和氯化铵水溶液(200mL),水(300mL),用乙酸乙酯(300mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,将滤液减压浓缩得到白色固体状的2b(20g,产率:90%)。
快速纯化色谱仪(Biotage,Isolera one,洗脱剂:5%乙酸乙酯/石油醚(v/v)),得到白色固体状的2b-1和2b-3的混合物(8g,产率:40%)和白色固体状的2b-2和2b-4的混合物(10g,产率:50%)。
中间体3:4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-醇(3c)
4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-ol
Figure PCTCN2015076449-appb-000022
第一步:[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]乙酸酯(3b)
[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]acetate
向反应瓶中依次加入1c(34.0g,0.135mol)、2,4二氟-3-氯苯胺(24.3g,0.148mol)和异丙醇(400mL),加热至85℃反应3小时。冷却至室温,抽滤,收集滤饼,干燥得到灰色固体状的3b(51.0g)。
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)。
第二步:4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-醇(3c)
4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-ol
向反应瓶中加入3b(51.0g,0.135mol)和7M的氨甲醇溶液(400mL),室温搅拌1小时。抽滤,用甲醇(100mL×2)洗涤,收集滤饼,干燥得到灰色固体状的3c(23.0g,产率:50.0%)。
MS m/z(ESI):338.0[M+1]。
中间体4:(3aS,6aR)-5-羟基-3,3a,4,5,6,6a-六氢-1H-环戊烷[c]吡咯-2-甲酸叔丁酯(4d)
tertbutyl-(3aS,6aR)-5-hydroxy-3,3a,4,5,6,6a-hexahydro-1H-cyclopenta[c]pyrrole-2-carboxy late
Figure PCTCN2015076449-appb-000023
第一步:N-烯丙基-N-丙-2-炔基氨基甲酸叔丁酯(4b)
tert-butyl N-allyl-N-prop-2-ynyl-carbamate
冰浴下,向反应瓶中加入N-叔丁氧羰基丙烯胺4a(20g,127.39mmoL)和N,N-二甲基甲酰胺(100mL),加入氢化钠(60%分散于石蜡油,7.6g,191.08mmol),滴加完毕后0℃下搅拌30分钟,自然升至室温搅拌20分钟,冰浴下滴加溴丙炔(30.3g,245.78mmol),0℃下搅拌30分钟,将反应液缓慢倒入冰块(200g)中,用乙酸乙酯(100mL×3)萃取,合并有机相,饱和食水(100mL×3)洗涤,无水硫酸钠干燥,过滤,硅胶柱色谱分离提纯(石油醚/乙酸乙酯(v/v)=100:1~50:1)得到黄色液体状的4b(21g,收率:85%)。
MS m/z(ESI):140.1[M+1]。
第二步:(3aS,6aR)-5-氧代-1,3,3a,4,6,6a-六氢环戊烷并[c]吡咯-2-甲酸叔丁酯(4c)
tert-butyl(3aR,6aS)-5-oxo-1,3,3a,4,6,6a-hexahydrocyclopenta[c]pyrrole-2-carboxylate
向反应瓶中依次加入4b(39g,202.2mmoL)、甲醚(275mL)、八羰基二钴(83g,242.7mmoL)和水(82.4mL),加热回流搅拌过夜,减压浓缩,向残留物中加入饱和食水(500mL)、乙酸乙酯(250mL)和1M的盐酸(240mL),室温搅拌5分钟,静置分液,水相用乙酸乙酯(250mL×3)萃取,合并有机相,饱和食水(250mL×2)洗涤,有机相用无水硫酸钠干燥,过滤,减压浓缩,残留物用硅胶柱色谱分离提纯(石油醚/乙酸乙酯(v/v)=3:1~2:1)得到淡黄色固体状的4c(27.6g,收率: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)。
第三步:(3aS,6aR)-5-羟基-3,3a,4,5,6,6a-六氢-1H-环戊烷并[c]吡咯-2-甲酸叔丁酯(4d)
tert-butyl-(3aS,6aR)-5-hydroxy-3,3a,4,5,6,6a-hexahydro-1H-cyclopenta[c]pyrrole-2-carbox ylate
向反应瓶中加入4c(1.5g,6.7mmol),搅拌均匀后,冰浴冷却至0℃,加入硼氢化钠(0.38g,10mmol),加完后0℃反应30分钟。向反应液中滴加饱和氯化铵(20mL),加入乙酸乙酯(50mL),水层用乙酸乙酯萃取(40mL×2),合并有机相,用饱和食水洗涤(40mL×1),有机相用无水硫酸钠干燥,过滤,减压浓缩,硅胶柱色谱分离提纯(石油醚/乙酸乙酯(v/v)=1:1)得到无色油状的4d(1.4g,产率: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)。
中间体5:(3R)-3-氟-4-羟基-哌啶-1-甲酸叔丁酯(5c)
tert-butyl(3R)-3-fluoro-4-hydroxy-piperidine-1-carboxylate
Figure PCTCN2015076449-appb-000024
Figure PCTCN2015076449-appb-000025
第一步:(R)-(5-乙基奎宁-2-基)-(6-甲氧基-4-喹啉基)甲胺三盐酸盐(5b)
(R)-(5-ethylquinuclidin-2-yl)-(6-methoxy-4-quinolyl)methanamine trihydrochloride
向反应瓶中加入氢化奎宁定5a(25g,76.7mmol)和三苯基膦(30g,115mmol),加入四氢呋喃(250mL),冷却至0℃,滴加偶氮二甲酸二异丙酯(26.5g,130.4mmol),0℃反应10分钟,滴加叠氮磷酸二苯酯(36g,130.4mmol),室温反应过夜,分批加入三苯基膦(30g,115mmol),45℃反应4小时,加入水(25g),45℃反应过夜,减压浓缩,加入二氯甲烷(250mL)和2M盐酸(150mL),搅拌20分钟,分出水层,用二氯甲烷(250mL×2)萃取,水层浓缩,用乙酸乙酯(150mL),甲醇(50mL)打浆,用乙酸乙酯(150mL),甲醇(30mL)洗涤,滤出固体,干燥得到白色固体状的5b(26g,产率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)。
第二步:(3R)-3-氟-4-羟基-哌啶-1-甲酸叔丁酯(5c)
tert-butyl(3R)-3-fluoro-4-hydroxy-piperidine-1-carboxylate
取5b(2.03g,4.63mmol)溶于46.3mL的四氢呋喃中,冰水浴冷却,饱和碳酸钠水溶液调至中性,旋蒸至干,用甲苯带水三次,加入46.3mL的四氢呋喃,过滤,加入三氯乙酸(0.75g,4.63mmol),水配置成0.1M的(R)-(5-乙基奎宁-2-基)-(6-甲氧基-4-喹啉基)甲胺单三氯乙酸单水合物的四氢呋喃溶液。向反应瓶中加入N-氟代双苯磺酰胺(3.77g,11.6mmol)和碳酸钠(1.85g,17.3mmol),加入四氢呋喃(33mL),冷却至-20℃,加入上述0.1M的(R)-((1S,2R,4S,5R)-5-乙基奎宁-2-基)(6-甲氧基喹啉-4-基)甲胺单三氯乙酸单水合物的四氢呋喃溶液,-20℃反应10分钟,加入N-叔丁氧羰基-4-哌啶酮(4.63g,23.1mmol),-20℃反应过夜,加入甲基叔丁基醚(50mL),搅拌30分钟,过滤,将滤液减压浓缩,硅胶柱色谱分离提纯(石油醚/乙酸乙酯(v/v)=5:1)得到白色固体状的5c(1.9g,产率74.8%)。
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)。
将5c(1.9g,8.7mmol)溶于甲醇(8mL),冷却至0℃,加硼氢化钠(0.5g,13.0mmol),0℃反应2.5小时,加入饱和氯化铵水溶液(20mL),搅拌20分钟,减压浓缩,加入水(45mL),用乙酸乙酯(25mL×3)萃取,合并有机层,用无水硫酸钠干燥,过滤,将滤液减压浓缩,硅胶柱色谱分离提纯(石油醚/乙酸乙酯(v/v)=7:1),得到白色固体状的5c-1(0.6g,产率30%,ee>99%)和5c-2(0.45g,产率22.5%,ee>99%).
中间体6:4-(3,4二氯-2-氟-苯胺)-7-甲氧基-喹唑啉-6-醇(6c)
4-((3,4-dichloro-2-fluoro-anilino)-7-methoxyquinazolin-6-ol
Figure PCTCN2015076449-appb-000026
第一步:[4-(3,4二氟-2-氯-苯胺)-7-甲氧基-喹唑啉-6-基]乙酸酯(6b)
[4-(3,4-difluoro-2-chloro-anilino)-7-methoxy-quinazolin-6-yl]acetate
向反应瓶中依次加入1c(32.4g,0.129mol)、3,4二氟-2-氯苯胺(32.6g,0.155mol)和异丙醇(250mL),加热至100℃反应4小时。冷却至室温,抽滤,收集滤饼,干燥得到灰色固体状的6b(51.1g),直接用于下步反应。
MS m/z(ESI):396.0[M+1]。
第二步:4-(3,4二氟-2-氯-苯胺)-7-甲氧基-喹唑啉-6-醇(6c)
4-(3,4-dichloro-2-fluoro-anilino)-7-methoxyquinazolin-6-ol
向反应瓶中加入6b(51.1g,0.129mol)和7M的氨甲醇溶液(250mL),室温搅拌1小时。抽滤,用甲醇(100mL×2)洗涤,收集滤饼,干燥得到灰色固体状的6c(30g,产率: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]。
实施例1:1-[4-[[4-(3-氯-2,4-二氟-苯胺基)-7-甲氧基-喹唑啉-6-基]氧基甲基]-1-哌啶基]丙基-2-烯-1-酮(化合物1)
1-[4-[[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxymethyl]-1-piperidyl]prop-2-en-1-one
Figure PCTCN2015076449-appb-000027
第一步:4-[(4-氯-7-甲氧基-喹唑啉-6-基)氧基甲基]哌啶-1-甲酸叔丁酯(1B)
tert-butyl4-[(4-chloro-7-methoxy-quinazolin-6-yl)oxymethyl]piperidine-1-carboxylate
向反应瓶中加入1d(3.0g,14.2mmol)、三苯基膦(5.58g,21.3mmol)和二氯甲烷(50mL),氮气保护,冰浴冷却至0℃,滴加偶氮二甲酸二异丙酯(4.30g,21.3mmol)的二氯甲烷(10mL)溶液,滴加完毕后,升至室温反应30分钟。0℃下缓慢加入4-羟甲基哌啶-1-甲酸叔丁酯(4.58g,21.3mmol),升至室温反应12小时。减压浓缩,残留物用硅胶柱色谱分离提纯(石油醚/乙酸乙酯(v/v)=10:1)得到白色固体状的1B粗品(5.80g),直接用于下步反应。
第二步:4-[[4-(3-氯2,4-二氟苯胺)-7-甲氧基-喹唑啉-6-基]氧甲基]哌啶-1-甲酸叔丁酯(1C)
tert-butyl 4-[[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxymethyl]piperidine-1-carboxylate
向反应瓶中依次加入1B(5.80g,14.2mmol)、2,4-二氯-3-氯苯胺(2.6g,15.6mmol)和异丙醇(30mL),加热至120℃反应2小时,减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1~50:1~30:1)得到白色固体状的1C(3.80g,产率:50.0%)。
MS m/z(ESI):535.1[M+1]。
第三步:N-(3-氯-2,4-二氟-苯基)-7-甲氧基-6-(4-哌啶基甲氧基)喹唑啉-4-胺二(三氟乙酸)盐(1D)
N-(3-chloro-2,4-difluoro-phenyl)-7-methoxy-6-(4-piperidylmethoxy)quinazolin-4-amine ditrifluoroacetic acid
向反应瓶中加入1C(3.80g,0.93mmol)和二氯甲烷(20mL),冰水浴下滴加三氟乙酸(20mL),滴加完毕后撤去冰浴,室温反应2小时。减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=10:1)得到白色固体状的1D(3.30g,产率:68.0%)。
第四步:1-[4-[[4-(3-氯-2,4-二氟-苯胺基)-7-甲氧基-喹唑啉-6-基]氧基甲基]-1-哌啶基]丙基-2-烯-1-酮(化合物1)
1-[4-[[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxymethyl]-1-piperidyl]prop-2-en-1-one
向反应瓶中依次加入1D(3.30g,4.93mmol)、三乙胺(4.1mL)和二氯甲烷(15mL),干冰乙醇浴冷却至-60℃,滴加丙烯酰氯(0.45mL,5.48mmol)的二氯甲烷(2mL)溶液,自然升至室温反应30分钟。向反应体系中加入饱和氯化钠溶液(20mL),用二氯甲烷(20mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1)得到白色固体状的化合物1(0.08g,产率: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)。
实施例2:1-[4-[4-(3-氯-2,4-二氟-苯胺基)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮(化合物2,化合物2是1-[(3R,4R)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮和1-[(3S,4S)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮的混合物)
1-[4-[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-1-piperidyl]prop-2-en-1-one
Figure PCTCN2015076449-appb-000028
Figure PCTCN2015076449-appb-000029
第一步:4-[4-(3-氯-2,4-二氟-苯胺基)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-哌啶-1-甲酸叔丁酯(2B,2B是(3R,4R)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-哌啶-1-甲酸叔丁酯和(3S,4S)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-哌啶-1-甲酸叔丁酯的混合物)
tert-butyl4-[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-pipe ridine-1-carboxylate
向反应瓶中依次加入3c(3.0g,8.96mmol)、二氯甲烷(60mL)、2b-1和2b-3的混合物(3.9g,17.91mmol)和三苯基膦(9.40g,35.90mmol),氮气保护,冷却至-15℃下滴加偶氮二甲酸二叔丁酯(6.20g,26.90mmol)的二氯甲烷(36mL)溶液,滴加完毕后,升至室温反应3小时。减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1)得到黄色固体状的2B(4.1g,产率:85.4%,2B是(3R,4R)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-哌啶-1-甲酸叔丁酯和(3S,4S)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-哌啶-1-甲酸叔丁酯的混合物)。
第二步:N-(3-氯-2,4-二氯-苯基)-6-[(3-氟-4-哌啶基)氧]-7-甲氧基-喹唑啉-4-胺二(三氟乙酸)盐(2C,2C是N-(3-氯-2,4-二氯-苯基)-6-[[(3R,4R)-3-氟-4-哌啶基]氧基]-7-甲氧基-喹唑啉-4-胺二(三氟乙酸)和N-(3-氯-2,4-二氯-苯基)-6-[[(3S,4S)-3-氟-4-哌啶基]氧基]-7-甲氧基-喹唑啉-4-胺二(三氟乙酸)的混合物)
N-(3-chloro-2,4-difluoro-phenyl)-6-[(3-fluoro-4-piperidyl)oxy]-7-methoxy-quinazolin-4-am ine
2C是N-(3-氯-2,4-二氯-苯基)-6-[[(3R,4R)-3-氟-4-哌啶基]氧基]-7-甲氧基-喹唑啉-4-胺二(三氟乙酸)和N-(3-氯-2,4-二氯-苯基)-6-[[(3S,4S)-3-氟-4-哌啶基]氧基]-7-甲氧基-喹唑啉-4-胺二(三氟乙酸)的混合物。
向反应瓶中依次加入2B(4.10g,7.62mmol)和二氯甲烷(17mL),冰水浴冷却下滴加 三氟乙酸(17mL),滴加完毕后自然升至室温反应1小时,减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=20:1)得到白色固体状的2C(3.00g,产率:60.0%)。
MS m/z(ESI):439.0[M+1]。
第三步:1-[4-[4-(3-氯-2,4-二氟-苯胺基)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮(化合物2,化合物2是1-[(3R,4R)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮和1-[(3S,4S)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮的混合物)
1-[4-[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-1-piperidyl]prop-2-en-1-one
化合物2是1-[(3R,4R)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮和1-[(3S,4S)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮的混合物。
将2C(2.30g,5.25mmol)溶于四氢呋喃(90mL)和水(14mL)的混合溶液中,加入碳酸氢钠(1.3g,15.80mmol),冰浴冷却至0℃下滴加丙烯酰氯(0.50g,5.78mmol)的四氢呋喃(7mL)溶液,0℃搅拌15分钟。向反应体系中加入饱和食水(20mL),用二氯甲烷(20mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1)得到白色固体状的化合物2(0.68g,产率:26.4%)。
将化合物2(0.68g)用制备设备和手性柱对手性异构体进行分离(分离条件:手性柱CHIRALPAK AS-H,流动相:A:正己烷(0.1%二乙醇胺)),B:异丙醇,A:B=50:50(v/v),流速:6.0mL/分钟,UV=250nm,柱温:35℃),得到白色固体状的1-[(3R,4R)-4-[4-(3-氯-2,4-二氟-苯胺基)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮(化合物2-1)(200mg,保留时间:40.363min,ee>95%)和1-[(3S,4S)-4-[4-(3-氯-2,4-二氟-苯胺基)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮(化合物2-2)(200mg,保留时间: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)。
实施例3:1-[4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮(化合物3,化合物3是1-[(3R,4S)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮和1-[(3S,4R)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮的混合物)
1-[4-[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-1-piperidyl]prop-2-en-1-one
Figure PCTCN2015076449-appb-000030
第一步:4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-哌啶-1-甲酸叔丁酯(3B,3B是(3R,4S)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-哌啶-1-甲酸叔丁酯和叔(3S,4R)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-哌啶-1-甲酸丁酯的混合物)
tert-butyl4-[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-pipe ridine-1-carboxylate
向反应瓶中依次加入3c(2.0g,6.16mmol,中间体3)、二氯甲烷(40mL)、2b-2和2b-4的混合物(2.7g,12.30mmol)和三苯基膦(6.50g,24.60mmol),氮气保护,冷却至-15℃下缓慢滴加偶氮二甲酸二叔丁酯(4.30g,18.50mmol)的二氯甲烷(24mL)溶液,滴加完毕后,自然升至室温反应3小时。减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=50:1)得到黄色固体状的3B(1.40g,产率:43.8%,3B是(3R,4S)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-哌啶-1-甲酸叔丁酯和(3S,4R)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-哌啶-1-甲酸叔丁酯的混合物)。
第二步:N-(3-氯-2,4-二氯-苯基)-6-[(3-氟-4-哌啶基)氧基]-7-甲氧基-喹唑啉-4-胺二(三 氟乙酸)盐(3C,3C是N-(3-氯-2,4-二氯-苯基)-6-[[(3R,4S)-3-氟-4-哌啶基]氧基]-7-甲氧基-喹唑啉-4-胺二(三氟乙酸)盐和N-(3-氯-2,4-二氯-苯基)-6-[[(3S,4R)-3-氟-4-哌啶基]氧基]-7-甲氧基-喹唑啉-4-胺二(三氟乙酸)盐的混合物)
N-(3-chloro-2,4-difluoro-phenyl)-6-[(3-fluoro-4-piperidyl)oxy]-7-methoxy-quinazolin-4-am ine
向反应瓶中加入3B(1.40g,2.60mmol)和二氯甲烷(7mL),冰水浴冷却下滴加三氟乙酸(7mL),室温反应1小时。减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=20:1)得到白色固体状的3C(0.65g,产率:37.5%,3C是N-(3-氯-2,4-二氯-苯基)-6-[[(3R,4S)-3-氟-4-哌啶基]氧基]-7-甲氧基-喹唑啉-4-胺二(三氟乙酸)盐和N-(3-氯-2,4-二氯-苯基)-6-[[(3S,4R)-3-氟-4-哌啶基]氧基]-7-甲氧基-喹唑啉-4-胺二(三氟乙酸)盐的混合物)。
第三步:1-[4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮(化合物3,化合物3是1-[(3R,4S)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮和1-[(3S,4R)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮的混合物)
1-[4-[4-(3-chloro-2,4-difluoro-anilino)-7-methoxy-quinazolin-6-yl]oxy-3-fluoro-1-piperidyl]prop-2-en-1-one
将3C(0.65g,0.10mmol)溶于四氢呋喃(25mL)和水(4mL)的混合溶液中,加入碳酸氢钠(0.41g,4.80mmol),冰浴冷却至0℃下滴加丙烯酰氯(0.10g,0.11mmol)的四氢呋喃(1.5mL)溶液,0℃反应15分钟,向反应体系中加入饱和食水(20mL),用二氯甲烷(20mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1)得到白色固体状的化合物3(0.18g,产率:36.7%,HPLC:99.70%)。
将化合物3(0.18g)用制备设备和手性柱对手性异构体进行分离(分离条件:手性柱CHIRALPAK AD-H,流动相:A:正己烷(0.1%二乙醇胺),B:异丙醇,A:B=50:50(v/v),流速:6.0mL/分钟,UV=254nm,柱温:35℃),得到白色固体状的化合物3-1(60mg,保留时间:9.158min,ee>99%)和化合物3-2(60mg,保留时间: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)。
实施例4:1-[4-[4-(3-氯-2,4-二氟-苯胺)-7-[(3S)-四氢呋喃-3-基]氧基-喹唑啉-6-基]氧基-1-哌啶基]丙基-2-烯-1-酮(化合物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
Figure PCTCN2015076449-appb-000031
第一步:5-苄氧基-4-氟-2-硝基苯甲酸(4B)
5-benzyloxy-4-fluoro-2-nitro-benzoic acid
向反应瓶中加入4,5-二氟-2-硝基-苯甲酸4A(25g,123mmol)和四氢呋喃(400mL),搅拌均匀后,加入苄醇(29.3g,270.6mmol),冰浴冷却至0℃,滴加2M双(三甲硅基)氨基钠的四氢呋喃溶液(197mL,394mmol),滴加完毕后0℃反应1小时。向反应液中滴加2M的盐酸调节体系pH值至1-2,加入乙酸乙酯(400mL),搅拌后分出有机相,水相用乙酸乙酯萃取(100mL×3),合并有机相,无水硫酸钠干燥,过滤,减压浓缩至剩余乙酸乙 酯约(100mL),0℃静置2小时,减压抽滤,收集滤饼,烘干得到黄色固体状的4B(17g,产率: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)。
第二步:5-苄氧基-4-氟-2-硝基苯甲酸甲酯(4C)
methyl 5-benzyloxy-4-fluoro-2-nitro-benzoate
向反应瓶中加入4B(17g,58.4mmol)和N,N-二甲基甲酰胺(170mL),搅拌均匀后,冰浴冷却,加入碳酸钾(24.2g,175.2mmol)和碘甲烷(24.8g,175.2mmol),加完升至室温反应2小时。将反应液倾入冰水中,用乙酸乙酯萃取(100mL×4),合并有机相,用水洗涤(100mL×1),有机相用无水硫酸钠干燥,过滤,减压浓缩,硅胶柱色谱分离提纯(石油醚/乙酸乙酯(v/v)=10:1~8:1)得到黄色固体状的4C(15.9g,产率89%)。
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)。
第三步:2-氨基-5-苄氧基-4-氟-苯甲酸甲酯(4D)
methyl 2-amino-5-benzyloxy-4-fluoro-benzoate
将4C(18.4g,60.3mmol)溶于异丙醇(294mL)和四氢呋喃(74mL)的混合溶液中,搅拌均匀后,加入铁粉(18.1g,324.1mmol),加热至100℃,加入氯化铵(12g,3.75mmol)的水溶液(15mL),100℃反应7小时。趁热过滤,用乙酸乙酯(100mL×2)洗涤滤饼,将滤液减压浓缩,向残留物种加入乙酸乙酯(400mL),用饱和碳酸氢钠洗涤(50mL×2),有机相用无水硫酸钠干燥,过滤,减压浓缩,硅胶柱色谱分离提纯(石油醚/乙酸乙酯(v/v)=8:1~7:1)得到灰色固体状的4D(15g,产率90.4%)。
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)。
第四步:6-苄氧基-7-氟-3H-喹唑啉-4-酮(4E)
6-benzyloxy-7-fluoro-3H-quinazolin-4-one
向反应瓶中加入4D(15g,54.5mmol)和甲酰胺(50mL),加入N,N-二甲基甲酰胺(0.5mL),加热至180℃反应7小时。反应液冷却至室温,将反应液缓慢倒入水(250mL),搅拌30分钟,过滤,收集滤饼,得到黄色固体状的4E(13g,产率88%)。
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)。
第五步:6-苄氧基-7-[(3S)-四氢呋喃-3-基]氧基-3H-喹唑啉-4-酮(4F)
6-benzyloxy-7-[(3S)-tetrahydrofuran-3-yl]oxy-3H-quinazolin-4-one
向反应瓶中加入(S)-3-羟基四氢呋喃(20mL)和金属钠(2g,88.9mmol),升温至120℃反应1.5小时,加入4E(4g,14.8mmol),升温至150℃反应2.5小时。反应液冷却至室温,将反应液倒入冰水中,用浓盐酸调节体系pH值至中性,加入乙酸乙酯(10mL),过滤,收集滤饼,得到灰色固体状的4F(4.5g,产率90%)。
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)。
第六步:6-苄氧基-4-氯-7-[(3S)-四氢呋喃-3-基]氧基-喹唑啉(4G)
6-benzyloxy-4-chloro-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazoline
向反应瓶中加入4F(2.5g,7.4mmol)和二氯亚砜(25mL),加入DMF(0.5mL),回流反应2.5小时。减压浓缩至干,加入二氯甲烷(50mL)和水(50mL),用氨水调节pH值至9-10,分出有机相,水相用二氯甲烷萃取(30mL×2),合并有机相,依次用水(30mL×1)、饱和食水(30mL×1)洗涤,有机相用无水硫酸钠干燥,过滤,减压浓缩,硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1),得到浅黄色固体状的4G(2g,产率77%)。
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)。
第七步:6-苄氧基-N-(3-氯-2,4-二氟-苯基)-7-[(3S)-四氢呋喃-3-基]氧基-喹唑啉-4-氨(4H)
6-benzyloxy-N-(3-chloro-2,4-difluoro-phenyl)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazolin-4-amine
向反应瓶中加入4G(3g,8.4mmol)和3-氯-2,4-二氟苯胺(2.7g,16.9mmol),加入异丙醇(60mL),加热回流反应3小时。反应液冷却至室温,过滤,收集滤饼,得到淡黄色固体状的4H(3.3g,产率81%)。
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)。
第八步:4-(3-氯-2,4-二氟-苯胺)-7-[(3S)-四氢呋喃-3-基]氧基-喹唑啉-6-醇(4I)
4-(3-chloro-2,4-difluoro-anilino)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazolin-6-ol
向反应瓶中依次加入4H(1.1g,2.28mmol)、三氟醋酸(10mL)和苯甲硫醚(1mL),加热回流反应2.5小时。减压浓缩至干,加入乙醚(15mL),用7M的氨甲醇溶液调节体系pH值至9,减压浓缩,加入水(20mL),过滤,收集滤饼,干燥得到淡黄色固体状的4I(740mg,产率83%)。
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)。
第九步:4-[4-(3-氯-2,4-二氟-苯胺)-7-[(3S)-四氢呋喃-3-基]氧-喹唑啉-6-基]氧基哌啶-1-甲酸叔丁酯(4J)
tert-butyl-4-[4-(3-chloro-2,4-difluoro-anilino)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quinazolin-6-yl]oxypiperidine-1-carboxylate
向反应瓶中依次加入4I(600mg,1.53mmol)、N-Boc-4-羟基哌啶醇(615mg,3.06mmol)和三苯基膦(1.6g,6.12mmol),加入二氯甲烷(12mL),降温至-15℃,滴加偶氮二甲酸二叔丁酯(615mg,3.06mmol)的二氯甲烷溶液(6mL),滴完升至室温反应3小时。减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=50:1→30:1)得到白色固体4J(750mg,产率85%)。
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)。
第十步:N-(3-氯-2,4-二氟-苯基)-6-(4-哌啶基氧基-7-[(3S)-四氢呋喃-3-基]氧基喹唑啉-4-胺(4K)
N-(3-chloro-2,4-difluoro-phenyl)-6-(4-piperidyloxy)-7-[(3S)-tetrahydrofuran-3-yl]oxy-quin azolin-4-amine
向反应瓶中依次加入4J(750mg,1.3mmol)、三氟醋酸(3mL)和二氯甲烷(10mL),滴加完毕后室温反应3小时。减压浓缩,向残留物中加入乙醚(15mL),用7M的氨甲醇溶液调节体系pH值至9,减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=10:1→5:1),向残留物中加入饱和碳酸氢钠溶液(20mL)和氯仿(30mL),分出有机层,水层用氯仿萃取(10mL×3),合并有机相,无水硫酸钠干燥,过滤,将滤液减压浓缩,得到淡黄色固体状的4K(575mg,产率93%)。
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)。
第十一步:1-[4-[4-(3-氯-2,4-二氟-苯胺)-7-[(3S)-四氢呋喃-3-基]氧基喹唑啉-6-基]氧基-1-哌啶基]丙基-2-烯-1-酮(化合物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
向反应瓶中依次加入4K(310mg,0.65mmol)、四氢呋喃(12mL)和碳酸氢钠(164mg,1.95mmol)的水溶液(1.8mL),冰浴冷却至0℃,滴加丙烯酰氯(65mg,0.72mmol)四氢呋喃溶液(1mL),加完0℃反应15分钟,滴加饱和碳酸氢钠水溶液(20mL),加入氯仿(50mL),分出有机层,水层用氯仿萃取(20mL×3),合并有机相,无水硫酸钠干燥,过滤,将滤液减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1→30:1)得到白色固体状的化合物4(207mg,产率: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)。
实施例5:1-[(3aR,6aS)-5-[4-(3-氯-2,4-二氟-苯胺)-7-[(3S)-四氢呋喃-3-基]氧基-喹唑啉-6-基]氧基-3,3a,4,5,6,6a-六氢-1H-环戊烷并[c]吡咯-2-基]丙基-2-烯-1-酮(化合物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
Figure PCTCN2015076449-appb-000032
第一步:(3aR,6aS)-5-[4-(3-氯-2,4-二氟-苯胺)-7-[(3S)-四氢呋喃-3-基]氧基-喹唑啉-6-基]氧基-3,3a,4,5,6,6a-六氢-1H-环戊烷[c]吡咯-2-甲酸叔丁酯(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
向反应瓶中依次加入4I(300mg,0.75mmol),4d(339mg,1.50mmol,中间体4)和三苯基膦(786mg,3.00mmol),加入二氯甲烷(6mL),降温至-15℃下滴加偶氮二甲酸二叔丁酯(518mg,2.25mmol)的二氯甲烷溶液(3mL),滴加完毕后升至室温反应2小时。减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1→50:1)得到白色固体状的5B(353mg,产率: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)。
第二步:6-[[(3aR,6aS)-1,2,3,3a,4,5,6,6a-八氢环戊烷并[c]吡咯-5-基]氧基]-N-(3-氯-2,4-二氟-苯基)-7-[(3S)-四氢呋喃-3-基]氧基-喹唑啉-4-氨(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
向反应瓶中依次加入5B(470mg,0.78mmol),三氟醋酸(2mL)和二氯甲烷(6mL),加完室温反应30分钟。减压浓缩至干,加入乙醚(15mL),用7M的氨甲醇溶液调节体系pH值至 9,减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=10:1~7:1),向残留物中加入饱和碳酸氢钠溶液(20mL)和氯仿(30mL),分出有机层,水层用氯仿萃取(10mL×3),合并有机相,无水硫酸钠干燥,过滤,减压浓缩,得到黄色固体状的5C(340mg,产率: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)。
第三步:1-[(3aR,6aS)-5-[4-(3-氯-2,4-二氟-苯胺)-7-[(3S)-四氢呋喃-3-基]氧基-喹唑啉-6-基]氧基-3,3a,4,5,6,6a-六氢-1H-环戊烷并[c]吡咯-2-基]丙基-2-烯-1-酮(化合物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
向反应瓶中依次加入5C(340mg,0.68mmol)、四氢呋喃(12mL)和碳酸氢钠(171mg,2.04mmol)的水(1.8mL)溶液,冷至0℃,滴加丙烯酰氯(68mg,0.75mmol)的四氢呋喃(1mL)溶液,0℃反应30分钟,向反应体系中滴加饱和碳酸氢钠溶液(10mL),加入二氯甲烷(30mL),分出有机层,水层用二氯甲烷萃取(10mL×2),合并有机相,饱和食水洗涤(20mL×1),无水硫酸钠干燥,过滤,减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1~30:1)得到黄色固体状的化合物5(100mg,产率: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)。
实施例6:1-[(3aR,6aS)-5-[4-[(3-氯-2,4-二氟-苯基)氨基]-7-甲氧基-喹唑啉-6-基]氧基-六氢环戊烷并[c]吡咯-2(1H)-基]丙基-2-烯-1-酮(化合物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
Figure PCTCN2015076449-appb-000033
Figure PCTCN2015076449-appb-000034
第一步:(3aR,6aS)-5-((4-氯-7-甲氧基-喹唑啉-6-基)氧基-六氢环戊烷并[c]吡咯-2(1H)--甲酸叔丁酯(6B)
(3aR,5s,6aS)-tert-butyl 5-((4-chloro-7-methoxyquinazolin-6-yl)oxy)hexahydrocyclopenta[c]pyrrole-2(1H)-carboxylate
向反应瓶中依次加入1d(1.1g,5.2mmol)、4d(1.7g,7.8mmol,中间体4)和三苯基膦(2.1g,7.8mmol),搅拌均匀后,冰水浴冷却至0℃,将偶氮二甲酸二异丙酯(1.6g,7.8mmol)的二氯甲烷(5mL)溶液逐滴加入到体系中,自然升至室温搅拌过夜。减压浓缩,残留物用硅胶柱色谱分离(石油醚/乙酸乙酯(v/v)=3:2→2:3)得到无色油状的6B(2.2g)。
MS m/z(ESI):420.3[M+1]。
第二步:(3aR,6aS)-5-[[4-[(3-氯-2,4-二氟-苯基)氨基]-7-甲氧基-喹唑啉-6-基]氧基]-六氢环戊烷[c]吡咯-2(1H)-甲酸叔丁酯(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
向反应瓶中依次加入6B(4g,9.5mmol)、3-氯-2,4-二氟苯胺(3g,19mmol,毕得)和异丙醇(65mL),升至100℃反应3小时,减压浓缩,残留物用硅胶柱色谱分离(二氯甲烷/甲醇(v/v)=30:1~10:1)得到浅黄色油状的6C(1.7g,产率: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)。
第三步:N-(3-氯-2,4-二氟-苯基)-7-甲氧基-6-[[(3aR,6aS)-八氢环戊烷并[c]吡咯-5-基]氧基]喹唑啉-4-胺.二(三氟乙酸)盐(6D)
N-(3-chloro-2,4-difluorophenyl)-7-methoxy-6-(((3aR,5s,6aS)-octahydrocyclopenta[c]pyrrol-5-yl)oxy)quinazolin-4-amine
冰水浴下向反应瓶中依次加入6C(0.59g,1.1mmol)、二氯甲烷(4.5mL)和三氟乙酸(2.2mL),室温搅拌2小时。减压浓缩,残留物用硅胶柱色谱分离(二氯甲烷/甲醇(v/v)=20:1~5:1)得到浅黄色固体状的6D(0.6g,产率:82%)。
第四步:1-[(3aR,6aS)-5-[[4-[(3-氯-2,4-二氟-苯基)氨基]-7-甲氧基-喹唑啉-6-基]氧基-六氢环戊烷并[c]吡咯-2(1H)-基]丙-2-烯-1-酮(化合物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
向反应瓶中依次6D(0.257g,0.38mmol)、三乙胺(0.116g,1.14mmol)和二氯甲烷(12mL),干冰乙醇浴冷却至-40℃下逐滴加入丙烯酰氯(0.041g,0.46mmol),-40℃搅拌50分钟。将反应液倒入饱和碳酸氢钠溶液(60mL),用氯仿(60mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,减压浓缩,残留物用硅胶柱色谱分离(二氯甲烷/甲醇(v/v)=60:1~30:1)得到无色油状的化合物6(0.06g,产率: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)。
实施例7:1-[(3R,4R)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮马来酸盐(化合物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
Figure PCTCN2015076449-appb-000035
向反应瓶中加入化合物2-1(200mg,0.41mmol)、甲醇(2.0mL)和马来酸(47.1mg,0.41mmol),室温搅拌5小时。过滤得到浅黄色固体状的化合物7(90mg,产率: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。
实施例8:1-[(3R,4S)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮马来酸盐(化合物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
Figure PCTCN2015076449-appb-000036
向反应瓶中加入化合物3-2(170mg,0.35mmol)、乙醇(2.0mL)和马来酸(40.1mg,0.35mmol),室温搅拌5小时。过滤得到浅黄色固体状的化合物8(80mg,产率: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。
实施例9:1-[(3R,4R)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮盐酸盐(化合物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
Figure PCTCN2015076449-appb-000037
向反应瓶中加入化合物2-1(400mg,0.81mmol)、甲醇(2.8mL)和盐酸(0.1mL,1.23mmol)的甲醇(0.5mL)溶液,室温搅拌4小时。减压浓缩得到白色固体状的化合物9(395mg,产率: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)。
实施例10:1-[(3R,4S)-4-[4-(3-氯-2,4-二氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮盐酸盐(化合物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
Figure PCTCN2015076449-appb-000038
向反应瓶中加入化合物3-2(150mg,0.30mmol)、甲醇(0.8mL)和盐酸(16mg,0.45mmol)的甲醇(0.3mL)溶液,室温搅拌5小时。减压浓缩得到白色固体状的化合物10(135mg,产率: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)。
实施例11:1-[(3R,4R)-4-[4-(3,4-二氯-2-氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮(化合物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
Figure PCTCN2015076449-appb-000039
第一步:(3R,4R)4-[4-(3,4-二氯-2-氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-哌啶-1-甲酸叔丁酯(11B)
(3R,4R)-tert-butyl4-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)-3-fluoropiperidine-1-carboxylate
向反应瓶中依次加入6c(2.6g,7.3mmol,中间体6)、二氯甲烷(18mL)、5c-1(0.53g,2.43mmol)和三苯基膦(1.91g,7.3mmol),氮气保护,冷却至-15℃下滴加偶氮二甲酸二叔丁酯(1.27g,7.3mmol)的二氯甲烷(18mL)溶液,滴加完毕后,升至室温反应18小时。减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1)得到黄色固体状的11B(0.2g,产率:15%)。
第二步:N-(3,4-二氯-2-氟-苯基)-6-[[(3R,4R)-3-氟-4-哌啶基]氧基]-7-甲氧基-喹唑啉-4-胺二(三氟乙酸)盐(11C)
N-(3,4-dichloro-2-fluorophenyl)-6-(((3R,4R)-3-fluoropiperidin-4-yl)oxy)-7-methoxyquinaz olin-4-amine(ditrifluoroacetic acid)
向反应瓶中依次加入11B(0.2g,0.35mmol)和二氯甲烷(3mL),冰水浴冷却下滴加三氟乙酸(3mL),滴加完毕后自然升至室温反应1小时,减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=20:1)得到白色固体状的的11C(0.16g,产率:99%)。
第三步:1-[(3R,4R)-4-[4-(3,4-二氯-2-氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮(化合物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
将11C(0.7g,1.04mmol)溶于四氢呋喃(25mL)和水(5mL)的混合溶液中,加入碳酸氢钠(0.44g,5.22mmol),冰浴冷却至-30℃下滴加丙烯酰氯(0.11g,1.14mmol),-15℃搅拌30分钟。向反应体系中加入饱和食水(20mL),用二氯甲烷(35mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,将滤液减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1)得到白色固体状的化合物11(0.12g,产率: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)。
实施例12:1-[(3R,4S)-4-[4-(3,4-二氯-2-氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮(化合物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
Figure PCTCN2015076449-appb-000040
第一步:(3R,4S)-4-[4-(3,4-二氯-2-氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-哌啶-1-甲酸叔丁酯(12B)
(3R,4S)-tert-butyl4-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)-3-fluoropiperidine-1-carboxylate
向反应瓶中依次加入6c(7.8g,21.9mmol,中间体6)、二氯甲烷(60mL)、5c-2(1.6g,7.3mmol)和三苯基膦(5.7g,21.9mmol),氮气保护,冷却至-15℃下滴加偶氮二甲酸二叔丁酯(3.8g,21.9mmol),滴加完毕后,升至室温反应18小时。减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1)得到黄色固体状的12B粗品(4.0g),直接用于下步反应。
第二步:N-(3,4-二氯-2-氟-苯基)-6-[[(3R,4S)-3-氟-4-哌啶基]氧基]-7-甲氧基-喹唑啉-4-胺二(三氟乙酸)盐(12C)
N-(3,4-dichloro-2-fluorophenyl)-6-(((3R,4S)-3-fluoropiperidin-4-yl)oxy)-7-methoxyquinaz olin-4-amine(ditrifluoroacetic acid)
向反应瓶中依次加入12B(4.0g,7.3mmol)和二氯甲烷(10mL),冰水浴冷却下滴加三氟乙酸(10mL),滴加完毕后自然升至室温反应1小时,减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=20:1)得到白色固体状的12C(1.3g,第一步和第二步总收率为27%)。
第三步:1-[(3R,4S)-4-[4-(3,4-二氯-2-氟-苯胺)-7-甲氧基-喹唑啉-6-基]氧基-3-氟-1-哌啶基]丙基-2-烯-1-酮(化合物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
将12C(1.2g,1.8mmol)溶于四氢呋喃(40mL)和水(8mL)的混合溶液中,加入碳酸氢钠(0.8g,9.0mmol),冰浴冷却至-30℃下滴加丙烯酰氯(0.2g,2.2mmol),-15℃搅拌30分钟。向反应体系中加入饱和食水(50mL),用二氯甲烷(50mL×3)萃取,合并有机相,无水硫酸钠干燥,过滤,减压浓缩,残留物用硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1)得到白色固体状的化合物12(0.21g,产率: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)。
实施例13:1-((3R,4R)-4-((4-((3,4-二氯-2-氟)氨基)-7-甲氧喹唑啉-6-基)氧基)-3-氟哌啶-1-基)丙基-2-烯-1-酮盐酸盐(化合物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
Figure PCTCN2015076449-appb-000041
向反应瓶中加入化合物11(80mg,0.16mmol)、甲醇(1mL)和(0.1mL,0.23mmol)盐酸的甲醇溶液(2.3M),室温搅拌4小时,减压浓缩得到白色固体状的化合物13(73mg,产率: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)。
实施例14:1-((3R,4S)-4-((4-((3,4-二氯-2-氟)氨基)-7-甲氧喹唑啉-6-基)氧基)-3-氟哌啶-1-基)丙基-2-烯-1-酮盐酸盐(化合物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
Figure PCTCN2015076449-appb-000042
向反应瓶中加入化合物12(78mg,0.15mmol)、甲醇(1mL)和(0.1ml,0.23mmol)盐酸的 甲醇溶液(2.3M),室温搅拌5小时。浓缩得到白色固体状的化合物14(73mg,产率: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)。
实施例15
7-((4-((3,4-二氯-2-氟苯基)氨基)-7-甲氧基喹唑啉-6-基)氧基)四氢-1H-恶唑并[3,4-a]吡啶-3(5H)-酮(化合物15)
7-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)tetrahydro-1H-oxazolo[3,4-a]pyridin-3(5H)-one
Figure PCTCN2015076449-appb-000043
第一步:1-叔丁基2-甲基4-羟基哌啶-1,2-二甲酸酯(15B)
1-tert-butyl 2-methyl 4-hydroxypiperidine-1,2-dicarboxylate
将1-叔丁基2-甲基4-氧代哌啶-1,2-二甲酸酯15A(6.1g,24mmol)加入反应瓶中,加入甲醇60mL,冰浴冷却,分批加入硼氢化钠(1.4g,36mmol),加完,保温反应30分钟。滴加饱和氯化铵水溶液50mL,减压浓缩,加入乙酸乙酯及水各100mL,分液,水相以乙酸乙酯萃取50mL×2,合并有机相,无水硫酸钠干燥,减压浓缩,硅胶柱色谱分离提纯(石油醚/乙酸乙酯(v/v)=3:1~2:1)得到无色油状物15B(5.8g,产率94%)。
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)。
第二步:1-叔丁基2-甲基4-((4-((3,4-二氯-2-氟苯基)氨基)-7-甲氧基喹唑啉-6-基)氧代)哌啶-1,2-二甲酸酯(15C)
1-tert-butyl 2-methyl 4-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)piperidine-1,2-dicarboxylate
将15B(2.0g,7.71mmol),4-((3,4-二氯-2-氟苯基)氨基)-7-甲氧基喹唑啉-6-醇(1.37g,3.86mmol),三苯基膦(4.05g,15.42mmol),加入反应瓶中,加入二氯甲烷(60mL),降温至-20℃,滴加偶氮二羧酸二叔丁酯(2.66g,11.57mmol)的二氯甲烷溶液10mL,滴完,升至室温反应4小时。减压浓缩,硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1~50:1)得到淡黄色固体15C(1.5g,产率65%)。
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)。
第三步:叔丁基4-((4-((3,4-二氯-2-氟苯基)氨基)-7-甲氧基喹唑啉-6-基)氧代)-2-(羟甲基)哌啶-1-甲酸酯(15D)
tert-butyl 4-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)-2-(hydroxymethyl)piperidine-1-carboxylate
将15C(0.5g,0.8mmol)加入反应瓶中,加入四氢呋喃(4mL),冷却至-78℃,滴加二异丁基氢化铝的甲苯溶液(4mL,1.5mol/L),滴完缓慢升至室温反应4小时。将反应液倾入氢氧化钠水溶液(100mL,1mol/L)和冰的混合物中,加入乙酸乙酯(30mL),分液,水相以乙酸乙酯萃取(20mL×2),合并有机相,无水硫酸钠干燥,过滤,减压浓缩,硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1~20:1)得到黄色油状物15D(0.3g,产率60%)。
MS m/z(ESI):567.1[M+1]。
第四步:7-((4-((3,4-二氯-2-氟苯基)氨基)-7-甲氧基喹唑啉-6-基)氧基)四氢-1H-恶唑[3,4-a]吡啶-3(5H)-酮(化合物15)
7-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)tetrahydro-1H-oxazolo[3,4-a]pyridin-3(5H)-one
将15D(0.25g,0.44mmol)加入反应瓶中,加入N,N-二甲基甲酰胺(2.5mL),冰浴冷却,加入氢化钠(0.07g,1.8mmol),加完,升至室温反应1小时。将反应液倾入冰水中,加入乙酸乙酯(20mL),分液,水层以乙酸乙酯萃取20mL×2,合并有机相,以饱和食盐水 洗涤(20mL×4),无水硫酸钠干燥,过滤,减压浓缩,制备液相分离提纯,得白色固体化合物15(0.044g,产率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)。
实施例16
7-((4-((3,4-二氯-2-氟苯基)氨基)-7-甲氧基喹唑啉-6-基)氧基)四氢-1H-恶唑[3,4-a]吡啶-3(5H)-硫酮(化合物16)
7-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)tetrahydro-1H-oxazolo[3,4-a]pyridin-3(5H)-thione
Figure PCTCN2015076449-appb-000044
第一步:叔丁基-4-羟基-2-(羟甲基)哌啶-1-甲酸酯(16B)
tert-butyl 4-hydroxy-2-(hydroxymethyl)piperidine-1-carboxylate
将15B(1.35g,5.21mmol)加入反应瓶中,加入乙醇(15mL),降温至0℃,加入硼氢化钠(0.99g,26mmol),加完缓慢升至室温反应8小时。将反应液倾入氢氧化钠的水溶液(100mL,1mol/L)中,加入乙酸乙酯(30mL),搅拌分液,分出有机相,水相以乙酸乙酯萃取(30mL×2),合并有机相,无水硫酸钠干燥,减压浓缩,硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=50:1-10:1)得到无色油状物16B(0.7g,产率60%)。
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)。
第二步:2-(羟甲基)哌啶-4-醇三氟乙酸盐(16C)
2-(hydroxymethyl)piperidin-4-ol trifluoroacetic acid salt
将16B(0.26g,1.12mmol)加入反应瓶中,加入二氯甲烷(1.5mL),冰浴冷却,加入三氟醋酸(1.5mL),加完升至室温反应1小时。减压浓缩,未经纯化直接用于下步反应。
MS m/z(ESI):132.1[M+1]。
第三步:7-羟基四氢-1H-噁唑[3,4-a]吡啶-3(5H)-硫酮(16D)
7-hydroxytetrahydro-1H-oxazolo[3,4-a]pyridine-3(5H)-thione
将16C(0.147g,1.12mmol)加入反应瓶中,加入二氯甲烷(3mL),冰浴冷却,加入三乙胺(0.34g,3.36mmol),加入硫羰基二咪唑(0.24g,1.34mmol),加完,升至室温反应2小时。向反应液中加入盐酸水溶液(10mL,1mol/L),搅拌分液,水层以二氯甲烷萃取(5mL×7),合并有机相,无水硫酸钠干燥,过滤,减压浓缩,硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1-70:1)得到黄色油状物16D(0.07g,产率40%)。
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)。
第四步:7-((4-((3,4-二氯-2-氟苯基)氨基)-7-甲氧基喹唑啉-6-基)氧基)四氢-1H-恶唑[3,4-a]吡啶-3(5H)-硫酮(化合物16)
7-((4-((3,4-dichloro-2-fluorophenyl)amino)-7-methoxyquinazolin-6-yl)oxy)tetrahydro-1H-oxazolo[3,4-a]pyridin-3(5H)-thione
将16D(0.18g,1.0mmol),4-((3,4-二氯-2-氟苯基)氨基)-7-甲氧基喹唑啉-6-醇(0.74g,2.0mmol),三苯基膦(1.1g,4.0mmol),加入反应瓶中,加入二氯甲烷(10mL),降温至-20℃,滴加偶氮二羧酸二叔丁酯(0.72g,3.1mmol)的二氯甲烷溶液(5mL),滴完,升至室温反应4小时。减压浓缩,硅胶柱色谱分离提纯(二氯甲烷/甲醇(v/v)=100:1~70:1)得到白色固体化合物16(0.127g,产率24%)。
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)。
生物测试例
生物测试例1:SRB细胞增殖实验
SRB实验是检测细胞在药物作用下的增殖情况。加药72小时后,通过SRB染色及酶标仪490nm读数,反映出细胞的增殖情况。
细胞铺板96孔板,H1975细胞系每空8000个细胞,A431细胞系每空10000个细胞,37℃孵箱,5%CO2培养过夜。加药前,每种细胞取6个孔加入30μl 50%三氯乙酸固定。化合物以DMSO配置成溶液,最高浓度10μM,5倍稀释共十个梯度,用含0.1%FBS的培养基梯度稀释待测化合物,并使其为终浓度2倍。NCI-H1975、A431细胞的96孔细胞培养板培养基换为新鲜含0.1%FBS的培养基(每孔100μl),再加入100μl含2倍终浓度的待测化合物,37℃孵箱,5%CO2培养72小时。孵育结束后,加入50μl 50%TCA,置于4℃冰箱中固定1h。弃液,300μlddH2O洗5次,室温干燥1h或更久。每空加入50μl 0.4%SRB,染色15min。弃染液,用1%乙酸洗6-7次,室温干燥透。用200μl 10mM unbuffered Trisbase(pH=10.5)溶解,平板振荡2h。酶标仪测定490nm吸光度。
表1:SRB细胞增殖实验结果
Figure PCTCN2015076449-appb-000045
结论:本发明化合物具有明显地抑制H1975(L858R/T790M)、A431(EGFRwt扩增)细胞增殖作用。
生物测试例2:药代动力学评价
雄性SD大鼠(购自维通利华实验动物有限公司)180-240g,禁食给水过夜,3只大鼠口服灌胃5mg/kg,3只大鼠静脉注射1mg/kg。口服给药组,化合物以0.5%甲基纤维素(MC)溶液(含0.4%吐温80)配制成0.5mg×mL-1的混悬液,在给药前和在给药后30分钟以及1、2、4、6、8、12和24小时采血;静脉给药组,化合物以10%的DMA,20%Solutol HS-15(30%,w/v)和70%生理盐水配制成0.2mg×mL-1的溶液,在给药前和在给药后5、15和30分钟以及1、2、4、8、12和24小时采血。肝素抗凝。血液样品5500转/分钟离心10分钟,收集血浆,于-80℃保存。取各时间点大鼠血浆10μL,加入含内标的乙腈溶液500μL 混合后,涡旋混合4分钟,3700转/分钟离心18分钟,取上清液70μL与70μL水混合,取混和液5μL进行LC-MS/MS分析。主要药代动力学参数用WinNonlin 6.3软件非房室模型分析,药代动力学评价试验结果见表2。
表2药代动力学评价试验结果
Figure PCTCN2015076449-appb-000046
结论:本发明化合物表现出明显优于对照药物Afatinib的药代动力学特征。
生物测试例3:裸小鼠肿瘤细胞移植瘤实验测定
(1)实验动物:
BALB/cA-nude裸小鼠,4-6周龄,雌性,购自北京维通利华实验动物技术有限公司。合格证号:SCXK(京)2012-0001。
(2)肿瘤细胞移植:
NCI-H1975细胞:裸小鼠实验室环境适应3天后,右肋部皮下接种H1975细胞5×106/只。肿瘤生长至200-250mm3时,随机分组,每组10只小鼠。
A431细胞:裸小鼠实验室环境适应3天后,右肋部皮下接种A431细胞5×106/只。肿瘤生长至200-250mm3时,随机分组,每组10只小鼠。
(3)给药剂量及方法:
NCI-H1975细胞移植瘤裸小鼠:受试化合物以PEG400/Tween80配制成0.15mg/ml和0.5mg/ml两个剂量。动物称量体重后分别灌胃给药受试化合物1.5mg/kg和5mg/kg;Afatinib以PEG400/Tween80配制成1.2mg/ml和4mg/ml。动物称量体重后分别灌胃给药受试化合物12mg/kg和40mg/kg。空白对照组给予相同体积不含药物的PEG400/Tween80溶液。每天一次给药,连续给药10天。
A431细胞移植瘤裸小鼠:受试化合物以PEG400/Tween80配制成0.03mg/ml和0.1mg/ml。动物称量体重后分别灌胃给药受试化合物0.3mg/kg和1mg/kg;Afatinib以PEG400/Tween80配制成0.3mg/ml和1mg/ml。动物称量体重后分别灌胃给药受试化合物3mg/kg和10mg/kg空白对照组给予相同体积不含药物的PEG400/Tween80溶液。每天一次给药,连续给药10天。
(4)移植瘤体积及裸小鼠体重测定
肿瘤细胞接种裸小鼠后每周一、四以游标卡尺各测一次瘤体积(计算公式见后),并称量体重。
(5)数据统计
使用Office Excel软件计算平均值和标准差(SD),SEM(STDEV/SQRT)。组间差异以T-testj检验,P<0.05为显著差异标准
肿瘤体积(V)计算公式为:V=1/2×长径×短径×短径
相对体积(RTV)=VT/V0
抑瘤率(%)=(CRTV-TRTV)/CRTV×100%)
其中V0、VT分别为开始给药前及最后一次给药后的肿瘤体积。CRTV、TRTV分别为次最后给药后空白对照组及实验组的相对肿瘤体积。结果见表3、4。
表3:对A431细胞移植瘤的抑制结果
Figure PCTCN2015076449-appb-000047
表4:对H1975细胞移植瘤的抑制结果
Figure PCTCN2015076449-appb-000048
结论:本发明化合物与对照药物Afatinib相比,更能明显抑制含有EGFR野生型的人上皮细胞癌A431和EGFR T790M突变的人肺癌NCI-H1975的生长,在给药剂量分别仅为阳性对照的1/10、1/8的剂量下,分别对A431、H1975移植瘤效果明显强于对照药物。
生物测试例4:电生理手动膜片钳检测本发明化合物对hERG钾离子通道的作用
本试验所用的细胞为转染有hERG cDNA与稳定表达hERG通道的CHO细胞系。采用手动膜片钳系统(德国HEKA EPC-10信号放大器及数字转换系统)作全细胞电流的记录。表面生长有CHO hERG细胞的圆形玻片被放置于倒置显微镜下的电生理记录槽中。记录槽内以细胞外液作持续灌流(大约每分钟1毫升)。实验过程采用常规全细胞膜片钳电流记录技术。细胞钳制在-80mV的电压下。细胞钳制电压去极化到+20mV以激活hERG钾通道,5秒后再钳制到-50mV以消除失活并产生尾电流。尾电流峰值用作hERG电流大小的数值。上述步骤所记录的hERG钾电流在记录槽内持续的细胞外液灌流下达到稳定后则可以叠加灌流待测试的药物,直到药物对hERG电流的抑制作用达到稳定状态。达到稳定态势以后以细胞外液灌流冲洗直到hERG电流回复到加药物之前的大小。
待测化合物浓度为30,10,3,1,0.3,0.1和0.03μM,来作测试。在试验之前,首先用DMSO稀释成30,10,3,1,0.3和0.1mM的贮备液,再用细胞外液稀释成最终的μM浓度。各浓度化合物溶液中DMSO的最终浓度都为0.1%。阳性对照Cisapride(西沙比利)的测试浓度为0.1μM。
试验数据由HEKA Patchmaster,Microsoft Excel以及Graphpad Prism提供的数据分析软件进行分析,结果如表5。
表5:对hERG钾离子通道的作用结果
化合物编号 hERG IC50(μM)
Afatinib 2.40
9 5.84
13 3.55
14 2.12
结论:本发明化合物对hERG钾离子通道抑制弱,具有较高的安全性,特别是化合物9和13,对hERG钾离子通道抑制更弱,具备更高安全性。

Claims (10)

  1. 一种通式(II0)所示的化合物或其立体异构体、水合物、代谢产物、溶剂化物、药学上可接受的盐、共晶或前药,
    Figure PCTCN2015076449-appb-100001
    其中:
    R2选自C1-3烷基、3至4元碳环基或者4至8元杂环基,所述的杂环基任选进一步被0至2个选自F、Cl、羟基、C1-3烷基或者C1-3烷氧基的取代基所取代,且所述的杂环基含有1至2个选自N或者O的杂原子;
    环A选自
    Figure PCTCN2015076449-appb-100002
    或者
    Figure PCTCN2015076449-appb-100003
    R3选自F、CF3、CHF2、CH2F、Cl、羟基或者C1-3烷基;
    X1为
    Figure PCTCN2015076449-appb-100004
    或者X1与环A一起形成
    Figure PCTCN2015076449-appb-100005
    Figure PCTCN2015076449-appb-100006
    R4、R5和R6各自为H;
    R9选自F、Cl、羟基、C1-3烷基或者C2-3炔基;
    n、m、t各自选自0、1、2、3或者4;
    限制条件是,当n为0,R2选自C1-3烷基,且m为0时,环A不为
    Figure PCTCN2015076449-appb-100007
  2. 根据权利要求1所述的化合物或其立体异构体、水合物、代谢产物、溶剂化物、药学上可接受的盐、共晶或前药,其中所述的化合物选自通式(II)所示的化合物:
    Figure PCTCN2015076449-appb-100008
    其中:
    R2选自甲基、乙基或者
    Figure PCTCN2015076449-appb-100009
    环A选自
    Figure PCTCN2015076449-appb-100010
    或者
    Figure PCTCN2015076449-appb-100011
    n为0或1;
    R3选自F;
    m为0或1;
    R4、R5和R6均为H;
    R9选自F或者Cl;
    t为3;
    限制条件是,当n为0,R2选自甲基或乙基,且m为0时,环A不为
  3. 根据权利要求1所述的化合物或其立体异构体、水合物、代谢产物、溶剂化物、药学上可接受的盐、共晶或前药,其中所述的化合物选自:
    Figure PCTCN2015076449-appb-100013
    Figure PCTCN2015076449-appb-100014
  4. 根据权利要求1中所述的化合物或其立体异构体、水合物、代谢产物、溶剂化物、药学上可接受的盐、共晶或前药,其中其药学上可接受的盐中所述的盐选自盐酸盐、氢溴酸盐、硫酸盐、磷酸盐、醋酸盐、三氟乙酸盐、硫氰酸盐、马来酸盐、羟基马来酸盐、戊二酸盐、甲磺酸盐、乙磺酸盐、苯磺酸盐、对甲苯磺酸盐、苯甲酸盐、水杨酸盐、苯乙酸盐、肉桂酸盐、乳酸盐、丙二酸盐、特戊酸盐、琥珀酸盐、富马酸盐、苹果酸盐、扁桃酸盐、酒石酸盐、没食子酸盐、葡萄糖酸盐、月桂酸盐、棕榈酸盐、果胶酸盐、苦味酸盐、柠檬酸盐或者它们的组合。
  5. 根据权利要求1中所述的化合物或其立体异构体、水合物、代谢产物、溶剂化物、药学上可接受的盐、共晶或前药,其中所述的盐选自盐酸盐、氢溴酸盐、硫酸盐、磷酸盐、醋酸盐、马来酸盐、甲磺酸盐、苯磺酸盐、三氟乙酸盐、对甲苯磺酸盐、苯甲酸盐、水杨酸盐、肉桂酸盐、乳酸盐、丙二酸盐、琥珀酸盐、富马酸盐、苹果酸盐、酒石酸盐、柠檬酸盐或者它们的组合。
  6. 一种药物组合物,所述的组合物包括:有效剂量的根据权利要求1-3中任一项所述的化合物或其立体异构体、水合物、代谢产物、溶剂化物、药学上可接受的盐、共晶或前药,以及药学上可接受的载体、稀释剂、佐剂、媒介物或赋形剂;所述的组合物还可进一步包括一种或多种其他治疗剂。
  7. 根据权利要求6所述的药物组合物,其中所述的其他治疗剂是顺铂(cisplatin),卡铂(carboplatin),奥沙利铂(oxaliplatin),达卡巴嗪(dacarbazine),替莫唑胺(temozolomide),丙卡巴肼(procarbazine),甲氨蝶呤(methotrexate),氟尿嘧啶(fluorouracil),阿糖胞苷(cytarabine),吉西他滨(gemcitabine),巯基嘌呤(mercaptopurine),氟达拉滨(fludarabine),长春碱(vinblastine),长春新碱(vincristine),长春瑞滨(vinorelbine),紫杉醇(paclitaxel),多西紫杉醇 (docetaxel),拓扑替康(topotecan),伊立替康(irinotecan),依托泊苷(etoposide),曲贝替定(trabectedin),更生霉素(dactinomycin),多柔比星(doxorubicin),表柔比星(epirubicin),道诺霉素(daunorubicin),米托蒽醌(mitoxantrone),博来霉素(bleomycin),丝裂霉素C(mitomycin),伊沙匹隆(ixabepilone),他莫昔芬(tamoxifen),氟他胺(flutamide),西罗莫司(sirolimus),Afatinib(afatinib),alisertib,amuvatinib,阿帕替尼(apatinib),阿西替尼(axitinib),硼替佐米(bortezomib),波舒替尼(bosutinib),布立尼布(brivanib),卡博替尼(cabozantinib),西地尼布(cediranib),crenolanib,克卓替尼(crizotinib),达拉非尼(dabrafenib),dacomitinib,达鲁舍替(danusertib),达沙替尼(dasatinib),多韦替尼(dovitinib),厄洛替尼(erlotinib),foretinib,ganetespib,gefitinib(吉非替尼),依鲁替尼(ibrutinib),埃克替尼(icotinib),伊马替尼(imatinib),iniparib,拉帕替尼(lapatinib),lenvatinib,linifanib,linsitinib,马赛替尼(masitinib),momelotinib,莫替沙尼(motesanib),来那替尼(neratinib),尼罗替尼(nilotinib),niraparib,oprozomib,olaparib,帕唑帕尼(pazopanib),pictilisib,ponatinib,quizartinib,regorafenib,rigosertib,rucaparib,ruxolitinib,塞卡替尼(saracatinib),saridegib,索拉非尼(sorafenib),舒尼替尼(sunitinib),tasocitinib,telatinib,tivantinib,tivozanib,tofacitinib,trametinib,凡德他尼(vandetanib),veliparib,威罗菲尼(vemurafenib),vismodegib,volasertib,阿仑单抗(alemtuzumab),贝伐单抗(bevacizumab),brentuximab vedotin,卡妥索单抗(catumaxomab),西妥昔单抗(cetuximab),地诺单抗(denosumab),吉妥珠单抗(gemtuzumab),伊匹单抗(ipilimumab),尼妥珠单抗(nimotuzumab),奥法木单抗(ofatumumab),帕尼单抗(panitumumab),利妥昔单抗(rituximab),托西莫单抗(tositumomab),曲妥珠单抗(trastuzumab)或它们的组合。
  8. 权利要求1-5中任一项所述的化合物或其立体异构体、水合物、酯、溶剂化物、共晶体、代谢产物、药学上可接受的盐或前药或者权利要求6-7中任一项所述的药物组合物在作为EGFR/HER2受体酪氨酸激酶抑制剂在制备药物制剂中的应用,特别是在制备用于治疗和/或预防过度增殖性疾病的药物制剂中的应用。
  9. 一种治疗和/或预防过度增殖性疾病的方法,该方法包括给予个体有效量的权利要求1-5中任一项所述的化合物或其立体异构体、水合物、酯、溶剂化物、共晶体、代谢产物、药学上可接受的盐或前药或者权利要求6-7中任一项所述的药物组合物,其中所述的过度增殖性疾病包括脑瘤、非小细胞肺癌、鳞状上皮细胞、膀胱癌、胰腺癌、结肠癌、乳腺癌、卵巢癌、子宫颈癌、子宫内膜癌、结直肠癌、肾癌、食管腺癌、食管鳞状细胞癌、实体瘤、非霍奇金淋巴瘤、肝癌、肺癌,皮肤癌、甲状腺癌、头颈癌、前列腺癌、神经胶质瘤及鼻咽癌中的一种或多种。
  10. 根据权利要求9所述的方法,其中所述过度增殖性疾病包括非小细胞肺癌、乳腺癌、表皮鳞癌、胃癌及结肠癌中的一种或多种。
PCT/CN2015/076449 2014-04-11 2015-04-13 喹唑啉衍生物及其制备方法和在医药上的应用 WO2015154725A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201580012570.5A CN106068262B (zh) 2014-04-11 2015-04-13 喹唑啉衍生物及其制备方法和在医药上的应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201410145927 2014-04-11
CN201410145927.4 2014-04-11

Publications (1)

Publication Number Publication Date
WO2015154725A1 true WO2015154725A1 (zh) 2015-10-15

Family

ID=54287332

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2015/076449 WO2015154725A1 (zh) 2014-04-11 2015-04-13 喹唑啉衍生物及其制备方法和在医药上的应用

Country Status (2)

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

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105399733A (zh) * 2015-12-03 2016-03-16 中国人民解放军南京军区南京总医院 一种新型喹唑啉衍生物lu1504及其制备方法和应用
WO2018086446A1 (zh) * 2016-11-08 2018-05-17 威尚(上海)生物医药有限公司 具有穿过血脑屏障能力的取代的喹唑啉化合物
CN110054613A (zh) * 2018-05-24 2019-07-26 深圳市塔吉瑞生物医药有限公司 取代的氨基喹唑啉类化合物及其药物组合物及其用途
WO2019196619A1 (zh) * 2018-04-08 2019-10-17 威尚(上海)生物医药有限公司 喹唑啉衍生物盐型晶型及制备方法和应用
CN110343090A (zh) * 2018-04-08 2019-10-18 威尚(上海)生物医药有限公司 喹唑啉衍生物盐型晶型及制备方法和应用
CN110357858A (zh) * 2018-04-09 2019-10-22 威尚(上海)生物医药有限公司 具有穿过血脑屏障能力的5取代二氟哌啶化合物
WO2019233459A1 (zh) * 2018-06-08 2019-12-12 江苏威凯尔医药科技有限公司 人表皮生长因子受体抑制剂及其制备方法和应用
CN110577514A (zh) * 2018-06-08 2019-12-17 江苏威凯尔医药科技有限公司 人表皮生长因子受体抑制剂及其制备方法和应用
WO2020053816A1 (en) * 2018-09-14 2020-03-19 Hanmi Pharmaceutical Co., Ltd. Crystalline forms of a quinazole compound and its hydrochloride salts
WO2020147774A1 (zh) * 2019-01-18 2020-07-23 南京明德新药研发有限公司 喹唑啉衍生物在制备治疗鼻咽癌药物中的应用
CN111763215A (zh) * 2020-07-21 2020-10-13 成都海博为药业有限公司 一种具有含氮杂环结构的化合物及其制备方法和用途
WO2020219904A1 (en) 2019-04-25 2020-10-29 Board Of Regents, The University Of Texas System Heterocyclic inhibitors of tyrosine kinase
JP2021506919A (ja) * 2017-12-19 2021-02-22 メッドシャイン ディスカバリー インコーポレイテッド キナゾリン誘導体およびその使用
WO2021127397A1 (en) * 2019-12-19 2021-06-24 Black Diamond Therapeutics, Inc. Nitrogen heterocyclic compounds and methods of use
CN113278012A (zh) * 2020-02-19 2021-08-20 郑州泰基鸿诺医药股份有限公司 用作激酶抑制剂的化合物及其应用
CN113444074A (zh) * 2020-03-26 2021-09-28 上海中医药大学 一种具有EGFR和Wnt双重抑制作用的化合物及其制备方法和应用

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112125890B (zh) * 2020-09-25 2022-12-06 华东理工大学 一种含异吲哚酮基喹唑啉基羧酸酯类衍生物及其应用
CN114516862B (zh) * 2022-03-17 2024-03-01 河北康泰药业有限公司 一种喹唑啉衍生物及其制备方法和应用
CN114560852B (zh) * 2022-03-17 2023-11-28 河北康泰药业有限公司 喹唑啉衍生物及其制备方法和应用

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005012290A1 (en) * 2003-07-29 2005-02-10 Astrazeneca Ab Piperidyl-quinazoline derivatives as tyrosine kinase inhibitors
CN101535279A (zh) * 2006-09-11 2009-09-16 柯瑞斯公司 含锌结合基的喹唑啉基egfr抑制剂
CN101679384A (zh) * 2007-06-05 2010-03-24 韩美药品株式会社 用于抑制癌细胞生长的新酰胺衍生物
CN102686581A (zh) * 2009-12-21 2012-09-19 张强 新的喹唑啉衍生物

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102020639A (zh) * 2009-09-14 2011-04-20 上海恒瑞医药有限公司 6-氨基喹唑啉或3-氰基喹啉类衍生物、其制备方法及其在医药上的应用
CN102731485B (zh) * 2011-04-02 2016-06-15 齐鲁制药有限公司 4-(取代苯氨基)喹唑啉衍生物及其制备方法、药物组合物和用途

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005012290A1 (en) * 2003-07-29 2005-02-10 Astrazeneca Ab Piperidyl-quinazoline derivatives as tyrosine kinase inhibitors
CN101535279A (zh) * 2006-09-11 2009-09-16 柯瑞斯公司 含锌结合基的喹唑啉基egfr抑制剂
CN101679384A (zh) * 2007-06-05 2010-03-24 韩美药品株式会社 用于抑制癌细胞生长的新酰胺衍生物
CN102686581A (zh) * 2009-12-21 2012-09-19 张强 新的喹唑啉衍生物

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105399733A (zh) * 2015-12-03 2016-03-16 中国人民解放军南京军区南京总医院 一种新型喹唑啉衍生物lu1504及其制备方法和应用
CN108069946B (zh) * 2016-11-08 2020-06-05 威尚(上海)生物医药有限公司 具有穿过血脑屏障能力的取代的喹唑啉化合物
JP2019528318A (ja) * 2016-11-08 2019-10-10 威尚(上海)生物医薬有限公司Weishang (Shanghai) Bio−Pharmaceutical Co., Ltd. 血液脳関門通過能を有する置換キナゾリン化合物
WO2018086446A1 (zh) * 2016-11-08 2018-05-17 威尚(上海)生物医药有限公司 具有穿过血脑屏障能力的取代的喹唑啉化合物
CN108069946A (zh) * 2016-11-08 2018-05-25 威尚(上海)生物医药有限公司 具有穿过血脑屏障能力的取代的喹唑啉化合物
US11168067B2 (en) * 2016-11-08 2021-11-09 Weishang (Shanghai) Bio-Pharmaceutical Co., Ltd. Substituted quinazoline compound having blood-brain barrier penetration capability
JP2021506919A (ja) * 2017-12-19 2021-02-22 メッドシャイン ディスカバリー インコーポレイテッド キナゾリン誘導体およびその使用
EP3733663A4 (en) * 2017-12-19 2021-08-25 Medshine Discovery Inc. QUINAZOLINE DERIVATIVE AND ITS USE
JP7296641B2 (ja) 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 キナゾリン誘導体およびその使用
CN110343090B (zh) * 2018-04-08 2021-06-04 威尚(上海)生物医药有限公司 喹唑啉衍生物盐型晶型及制备方法和应用
CN110343090A (zh) * 2018-04-08 2019-10-18 威尚(上海)生物医药有限公司 喹唑啉衍生物盐型晶型及制备方法和应用
WO2019196619A1 (zh) * 2018-04-08 2019-10-17 威尚(上海)生物医药有限公司 喹唑啉衍生物盐型晶型及制备方法和应用
CN110357858A (zh) * 2018-04-09 2019-10-22 威尚(上海)生物医药有限公司 具有穿过血脑屏障能力的5取代二氟哌啶化合物
CN110357858B (zh) * 2018-04-09 2022-02-18 威尚(上海)生物医药有限公司 具有穿过血脑屏障能力的5取代二氟哌啶化合物
WO2019223671A1 (zh) * 2018-05-24 2019-11-28 深圳市塔吉瑞生物医药有限公司 取代的氨基喹唑啉类化合物及其药物组合物及其用途
CN110054613A (zh) * 2018-05-24 2019-07-26 深圳市塔吉瑞生物医药有限公司 取代的氨基喹唑啉类化合物及其药物组合物及其用途
CN110054613B (zh) * 2018-05-24 2020-11-27 深圳市塔吉瑞生物医药有限公司 取代的氨基喹唑啉类化合物及其药物组合物及其用途
WO2019233459A1 (zh) * 2018-06-08 2019-12-12 江苏威凯尔医药科技有限公司 人表皮生长因子受体抑制剂及其制备方法和应用
CN110577514A (zh) * 2018-06-08 2019-12-17 江苏威凯尔医药科技有限公司 人表皮生长因子受体抑制剂及其制备方法和应用
CN112823153A (zh) * 2018-09-14 2021-05-18 韩美制药有限公司 喹唑啉化合物及其盐酸盐的结晶形式
US11883402B2 (en) 2018-09-14 2024-01-30 Hanmi Pharmaceutical Co., Ltd. Crystalline forms of a quinazoline compound and its hydrochloride salts
WO2020053816A1 (en) * 2018-09-14 2020-03-19 Hanmi Pharmaceutical Co., Ltd. Crystalline forms of a quinazole compound and its hydrochloride salts
WO2020147774A1 (zh) * 2019-01-18 2020-07-23 南京明德新药研发有限公司 喹唑啉衍生物在制备治疗鼻咽癌药物中的应用
CN113286790B (zh) * 2019-01-18 2024-03-15 成都金瑞基业生物科技有限公司 喹唑啉衍生物在制备治疗鼻咽癌药物中的应用
CN113286790A (zh) * 2019-01-18 2021-08-20 南京明德新药研发有限公司 喹唑啉衍生物在制备治疗鼻咽癌药物中的应用
US11365189B2 (en) 2019-04-25 2022-06-21 Board Of Regents, The University Of Texas System Heterocyclic inhibitors of tyrosine kinase
CN114026083A (zh) * 2019-04-25 2022-02-08 德州大学系统董事会 酪氨酸激酶的杂环抑制剂
WO2020219904A1 (en) 2019-04-25 2020-10-29 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
WO2021164793A1 (zh) * 2020-02-19 2021-08-26 郑州同源康医药有限公司 用作激酶抑制剂的化合物及其应用
CN113278012B (zh) * 2020-02-19 2022-07-12 郑州同源康医药有限公司 用作激酶抑制剂的化合物及其应用
CN113278012A (zh) * 2020-02-19 2021-08-20 郑州泰基鸿诺医药股份有限公司 用作激酶抑制剂的化合物及其应用
CN113444074A (zh) * 2020-03-26 2021-09-28 上海中医药大学 一种具有EGFR和Wnt双重抑制作用的化合物及其制备方法和应用
CN113444074B (zh) * 2020-03-26 2022-08-09 上海中医药大学 一种具有EGFR和Wnt双重抑制作用的化合物及其制备方法和应用
CN111763215A (zh) * 2020-07-21 2020-10-13 成都海博为药业有限公司 一种具有含氮杂环结构的化合物及其制备方法和用途
CN111763215B (zh) * 2020-07-21 2021-05-18 成都海博为药业有限公司 一种具有含氮杂环结构的化合物及其制备方法和用途

Also Published As

Publication number Publication date
CN106068262A (zh) 2016-11-02
CN106068262B (zh) 2019-10-29

Similar Documents

Publication Publication Date Title
WO2015154725A1 (zh) 喹唑啉衍生物及其制备方法和在医药上的应用
JP7374960B2 (ja) 縮合環化合物
AU2018347375B2 (en) Formulations comprising 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazab icyclo(3.1.1)heptan-3-yl)pyridin-3-yl)pyrazolo(1,5-a)pyridine-3-carbonitrile
TWI609012B (zh) 氨基喹唑啉類衍生物及其鹽和使用方法
EP2739618B1 (en) Quinazoline compounds as serine/threonine kinase inhibitors
KR20170097651A (ko) 테트라하이드로-피리도[3,4-b]인돌 에스트로겐 수용체 조절제 및 이의 용도
KR20120062839A (ko) 단백질 키나제 억제제로서의 화합물 및 조성물
JP2008543756A (ja) チエノピリミジンおよびチエノピリジンキナーゼモジュレーターを用いるflt3キナーゼの相乗的モジュレーター
JP2018534289A (ja) Egfr阻害剤の塩、結晶形及びその使用
WO2017157792A1 (en) Combinations of copanlisib
CN114302878A (zh) 酪氨酸激酶非受体1(tnk1)抑制剂及其用途
CN106279128B (zh) 环氧乙烷衍生物及其制备方法和在医药上的应用
KR102566089B1 (ko) 치환된 우레아 유도체의 염 및 의약에서 이의 용도
CN103319468B (zh) 取代的螺双环化合物及其使用方法和用途
CN106916112B (zh) 嘧啶衍生物及其制备方法和在医药上的应用
KR20200015595A (ko) 화합물
TWI570116B (zh) 取代的吡唑酮化合物及其使用方法
RU2783706C1 (ru) Соединения с конденсированными кольцами
EP3868759A1 (en) 2,3,5-substituted pyrrolo[2,3-b]pyridines as erbb modulators useful for treating cancer
WO2023150291A2 (en) Heterocyclic compounds and methods of use
WO2024023666A1 (en) Crystalline forms of an akr1c3 dependent kars inhibitor
CN117500803A (zh) 用于治疗具egfr突变的癌症的氨基取代杂环
JP2024521818A (ja) ヘテロ環式化合物およびその使用方法
JP2022506289A (ja) インダゾールキナーゼ阻害剤及びその使用
CN106279045A (zh) 环丙烷衍生物及其制备方法和在医药上的应用

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15776381

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15776381

Country of ref document: EP

Kind code of ref document: A1